1
|
Zhang X, Lan R, Liu Y, Pillarisetty VG, Li D, Zhao CL, Sarkar SA, Liu W, Hanna I, Gupta M, Hajdu C, Melamed J, Shusterman M, Widmer J, Allendorf J, Liu YZ. Complement activation in tumor microenvironment after neoadjuvant therapy and its impact on pancreatic cancer outcomes. NPJ Precis Oncol 2025; 9:58. [PMID: 40032924 PMCID: PMC11876354 DOI: 10.1038/s41698-025-00848-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
Neoadjuvant therapy (NAT) is increasingly being used for pancreatic ductal adenocarcinoma (PDAC). This study investigates how NAT differentially impacts PDAC's carcinoma cells and the tumor microenvironment (TME). Spatial transcriptomics was used to compare gene expression profiles in carcinoma cells and the TME of 23 NAT-treated versus 13 NAT-naïve PDACs. Findings were validated by single-nucleus RNA sequencing (snRNA-seq) analysis. NAT induces apoptosis and inhibits proliferation of carcinoma cells and coordinately upregulates multiple complement genes (C1R, C1S, C3, C4B and C7) within the TME. Higher TME complement expression following NAT is associated with increased immunomodulatory and neurotrophic cancer-associated fibroblasts (CAFs); more CD4+ T cells; reduced immune exhaustion gene expression, and improved overall survival. snRNA-seq analysis demonstrates C3 complement is mainly upregulated in CAFs. These findings suggest that local complement dynamics could serve as a novel biomarker for prognosis, evaluating treatment response, and guiding therapeutic strategies in NAT-treated PDAC patients.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA.
| | - Ruoxin Lan
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yongjun Liu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Danting Li
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Chaohui L Zhao
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Suparna A Sarkar
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Weiguo Liu
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Iman Hanna
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Mala Gupta
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Cristina Hajdu
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Jonathan Melamed
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Michael Shusterman
- Department of Oncology, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Jessica Widmer
- Department of Gastroenterology, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - John Allendorf
- Department of Surgery, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.
| |
Collapse
|
2
|
Huynh AS, Cohen AS, Doligalski M, Casagni TJ, Moberg VE, Huang X, Morse J, Abrahams D, Lloyd MC, Centeno BA, Baldwin MK, McLaughlin ML, Vagner J, Morse DL. Intraoperative Guidance of Pancreatic Cancer Resection Using a Toll-like Receptor 2-Targeted Fluorescence Molecular Imaging Agent. CANCER RESEARCH COMMUNICATIONS 2024; 4:2877-2887. [PMID: 39320054 PMCID: PMC11536076 DOI: 10.1158/2767-9764.crc-24-0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/01/2024] [Accepted: 09/23/2024] [Indexed: 09/26/2024]
Abstract
To increase the achievement of negative R0 surgical margins and increase the low survival rates of pancreatic cancer, improvements in assessing tumor margins during surgical resections are needed. This can be accomplished by using pancreatic cancer-targeted fluorescence molecular imaging agents to intraoperatively detect tumor margins in real time. Because Toll-like receptor 2 (TLR2) is broadly expressed among many cancer types including pancreatic adenocarcinomas, a high-affinity TLR2-targeted fluorescence molecular imaging agent (TLR2L-800) was developed. We investigate the potential for increased survival by employing real-time intraoperative tumor detection in a preclinical orthotopic human pancreatic xenograft tumor model using TLR2L-800. Three cohorts of nude mice bearing orthotopic human pancreatic xenograft tumors were intravenously injected with TLR2L-800. At 24 hours postinjection, one cohort underwent in vivo fluorescence-guided surgical removal of tumors using a real-time fluorescence imaging platform, a second cohort underwent visible light surgery (VLS), and a third cohort did not undergo surgery. A fourth, nontumor-bearing cohort was administered TLR2L-800 with no surgery. At 41 days postsurgery, the survival rates were 53% for the fluorescence-guided surgery (FGS) group and 0% for both the VLS and the tumor-bearing no-surgery group. The overall 200-day survival rate of 35% for the FGS group was significant compared with 0% for the VLS group (P value = 0.0018). This study demonstrates the potential of increasing disease-free survival for patients with pancreatic cancer by increasing the attainment of R0 margins using a novel tumor-targeted lipopeptide ligand-based fluorescence molecular imaging agent, TLR2L-800, during real-time FGS. SIGNIFICANCE Human TLR2 is broadly expressed among pancreatic adenocarcinomas, and the highly specific TLR2L-800 fluorescence molecular imaging agent has potential for use in fluorescence-guided surgery to increase R0 margins and improve patient survival.
Collapse
Affiliation(s)
- Amanda S. Huynh
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Allison S. Cohen
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Michael Doligalski
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Todd J. Casagni
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Valerie E. Moberg
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Xuan Huang
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jennifer Morse
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Dominique Abrahams
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mark C. Lloyd
- Analytic Microscopy Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Barbara A. Centeno
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Margaret K. Baldwin
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mark L. McLaughlin
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia
| | - Josef Vagner
- BIO5 Institute, University of Arizona, Tucson, Arizona
| | - David L. Morse
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida
- Department of Physics, University of South Florida, Tampa, Florida
- Department of Medical Engineering, University of South Florida, Tampa, Florida
| |
Collapse
|
3
|
van Eijck CWF, Ju J, van 't Land FR, Verheij M, Li Y, Stubbs A, Doukas M, Lila K, Heij LR, Wiltberger G, Alonso L, Malats N, Groot Koerkamp B, Vietsch EE, van Eijck CHJ. The tumor immune microenvironment in resected treatment-naive pancreatic cancer patients with long-term survival. Pancreatology 2024; 24:1057-1065. [PMID: 39218754 DOI: 10.1016/j.pan.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide. Presently, only a fraction of patients undergo successful surgical resection, the most effective treatment. Enhancing treatment strategies necessitates a deep comprehension of the factors underlying extended survival after surgical resection in patients. METHODS This study aims to identify the important factors of PDAC patients' long-term survival with metatranscriptomics and multiplex immunofluorescence (IF) staining analyses. Specifically, differences in tumor immune microenvironment (TIME) were investigated between treatment-naïve PDAC short-term survivors (STS, overall survival <6 months) and long-term survivors (LTS, overall survival >5 years). RESULTS As a result, we detected 589 over-expressed genes, including HOXB9, CDA, and HOXB8, and 507 under-expressed genes, including AMY2B, SCARA5, and SLC2A2 in LTS. Most of the Reactome overbiological pathways enriched in our data were over-expressed in LTS, such as RHO GTPase Effectors and Cell Cycle Checkpoints. Eleven microbiomes significantly differed between LTS and STS, including Sphingopyxis and Capnocytophaga. Importantly, we demonstrate that the TIME profile with an increased abundance of memory B cells and the reduction of M0 and pro-tumoral M2 macrophages are associated with a good prognosis in PDAC. CONCLUSIONS In this study, we delved into the TIME with metatranscriptomics and IF staining analyses to understand the prerequisite of prolonged survival in PDAC patients. In LTS, several biological pathways were overexpressed, and specific microbiomes were identified. Furthermore, apparent differences in driven immune factors were found that provide valuable insights into developing new treatment strategies.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| | - Jie Ju
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Freek R van 't Land
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Maaike Verheij
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Andrew Stubbs
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Michael Doukas
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Karishma Lila
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Lara R Heij
- Institute of Pathology, Medical Center University Duisburg-Essen, Essen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Germany; Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Georg Wiltberger
- Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Lola Alonso
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Bas Groot Koerkamp
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
4
|
Alomari A, Althunibat I, Obri MS, Curran J, Aldroubi B, Davis W, Pompa R. A Case of Metastatic Seminoma Mimicking a Primary Pancreatic Tumor. Cureus 2024; 16:e70329. [PMID: 39463534 PMCID: PMC11512812 DOI: 10.7759/cureus.70329] [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] [Accepted: 09/27/2024] [Indexed: 10/29/2024] Open
Abstract
Metastatic seminoma to the pancreas is exceedingly rare, with few reported cases in medical literature. We present a case of a 66-year-old male, six years post-remission from testicular seminoma, who presented with obstructive jaundice and a pancreatic mass mimicking primary malignancy. Diagnostic workup including endoscopic ultrasound-guided biopsy confirmed metastatic seminoma. He underwent successful treatment with four cycles of cisplatin and etoposide, achieving complete remission. This case underscores the diagnostic challenge of pancreatic metastases and emphasizes the role of biopsy in guiding appropriate management. Awareness of such presentations is crucial for timely intervention and improved patient outcomes.
Collapse
Affiliation(s)
- Ahmad Alomari
- Internal Medicine, Henry Ford Health System, Detroit, USA
| | | | - Mark S Obri
- Gastroenterology and Hepatology, Henry Ford Health System, Detroit, USA
| | - John Curran
- Internal Medicine, Henry Ford Health System, Detroit, USA
| | | | - William Davis
- Gastroenterology and Hepatology, Henry Ford Health System, Detroit, USA
| | - Robert Pompa
- Gastroenterology and Hepatology, Henry Ford Health System, Detroit, USA
| |
Collapse
|
5
|
Hajibandeh S, Hajibandeh S, Evans D, Athwal TS. Meta-analysis of pancreatic re-resection for locally recurrent pancreatic cancer following index pancreatectomy. Ann Hepatobiliary Pancreat Surg 2024; 28:315-324. [PMID: 38802115 PMCID: PMC11341876 DOI: 10.14701/ahbps.24-041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
The role of surgical resection in patients with recurrent pancreatic cancer is unclear. We aimed to evaluate the survival outcomes of pancreatic re-resection for locally recurrent pancreatic cancer following index pancreatectomy. A literature search was carried out in CENTRAL, EMBASE, MEDLINE, CINAHL, and Web of Science. Proportion meta-analysis model was constructed to quantify 1 to 5-year survival after pancreatic re-resection for locally recurrent pancreatic cancer. Random-effects modelling was applied to calculate pooled outcome data. Fifteen retrospective studies were included, reporting a total of 250 patients who underwent pancreatic re-resection for locally recurrent pancreatic cancer following their index pancreatectomy. Pancreatic re-resection was associated with 1-year survival 70.6% (95% confidence interval [CI], 65.0-76.2), 2-year survival 38.8% (95% CI, 28.6-49.0), 3-year survival 20.2% (95% CI, 13.8-26.7), and 5-year survival 9.2% (95% CI, 5.5-12.8). The between-study heterogeneity was insignificant in all outcome syntheses. Repeat pancreatectomy for local recurrence of pancreatic cancer in the remnant pancreas following the index pancreatectomy is associated with acceptable overall patient survival. We recommend selective re-resection of such recurrences in younger patients with favorable tumor size and location. Our findings may encourage more robust studies to be conducted in this context to provide stronger evidence.
Collapse
Affiliation(s)
- Shahin Hajibandeh
- Department of Hepatobiliary and Pancreatic Surgery, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Shahab Hajibandeh
- Department of Hepatobiliary and Pancreatic Surgery, Swansea Bay University Health Board, Swansea, UK
| | - Daisy Evans
- Department of Hepatobiliary and Pancreatic Surgery, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Tejinderjit S. Athwal
- Department of Hepatobiliary and Pancreatic Surgery, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| |
Collapse
|
6
|
van Oosten AF, Groot VP, Dorland G, Burkhart RA, Wolfgang CL, van Santvoort HC, He J, Molenaar IQ, Daamen LA. Dynamics of Serum CA19-9 in Patients Undergoing Pancreatic Cancer Resection. Ann Surg 2024; 279:493-500. [PMID: 37389896 DOI: 10.1097/sla.0000000000005977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
BACKGROUND Carbohydrate antigen (CA) 19-9 is an established perioperative prognostic biomarker for pancreatic ductal adenocarcinoma (PDAC). However, it is unclear how CA19-9 monitoring should be used during postoperative surveillance to detect recurrence and to guide the initiation of recurrence-focused therapy. OBJECTIVE This study aimed to elucidate the value of CA19-9 as a diagnostic biomarker for disease recurrence in patients who underwent PDAC resection. METHODS Serum CA19-9 levels at diagnosis, after surgery, and during postoperative follow-up were analyzed in patients who underwent PDAC resection. All patients with at least two postoperative follow-up CA19-9 measurements before recurrence were included. Patients deemed to be nonsecretors of CA19-9 were excluded. The relative increase in postoperative CA19-9 was calculated for each patient by dividing the maximum postoperative CA19-9 value by the first postoperative value. Receiver operating characteristic analysis was performed to identify the optimal threshold for the relative increase in CA19-9 levels to identify recurrence in the training set using Youden's index. The performance of this cutoff was validated in a test set by calculating the area under the curve (AUC) and was compared to the performance of the optimal cutoff for postoperative CA19-9 measurements as a continuous value. In addition, sensitivity, specificity, and predictive values were assessed. RESULTS In total, 271 patients were included, of whom 208 (77%) developed recurrence. Receiver operating characteristic analysis demonstrated that a relative increase in postoperative serum CA19-9 of 2.6× was predictive of recurrence, with 58% sensitivity, 83% specificity, 95% positive predictive value, and 28% negative predictive value. The AUC for a 2.6× relative increase in the CA19-9 level was 0.719 in the training set and 0.663 in the test set. The AUC of postoperative CA19-9 as a continuous value (optimal threshold, 52) was 0.671 in the training set. In the training set, the detection of a 2.6-fold increase in CA19-9 preceded the detection of recurrence by a mean difference of 7 months ( P <0.001) and in the test set by 10 months ( P <0.001). CONCLUSIONS A relative increase in the postoperative serum CA19-9 level of 2.6-fold is a stronger predictive marker for recurrence than a continuous CA19-9 cutoff. A relative CA19-9 increase can precede the detection of recurrence on imaging for up to 7 to 10 months. Therefore, CA19-9 dynamics can be used as a biomarker to guide the initiation of recurrence-focused treatment.
Collapse
Affiliation(s)
- A Floortje van Oosten
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital, Nieuwegein, The Netherlands
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vincent P Groot
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Galina Dorland
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital, Nieuwegein, The Netherlands
| | - R A Burkhart
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Hjalmar C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Jin He
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Lois A Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital, Nieuwegein, The Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht Cancer Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
7
|
Kaslow SR, Sacks GD, Berman RS, Lee AY, Correa-Gallego C. Natural History of Stage IV Pancreatic Cancer. Identifying Survival Benchmarks for Curative-intent Resection in Patients With Synchronous Liver-only Metastases. Ann Surg 2023; 278:e798-e804. [PMID: 36353987 DOI: 10.1097/sla.0000000000005753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To evaluate long-term oncologic outcomes of patients with stage IV pancreatic ductal adenocarcinoma and to identify survival benchmarks for comparison when considering resection in these patients. BACKGROUND Highly selected cohorts of patients with liver-oligometastatic pancreas cancer have reported prolonged survival after resection. The long-term impact of surgery in this setting remains undefined because of a lack of appropriate control groups. METHODS We identified patients with clinical stage IV pancreatic ductal adenocarcinoma with synchronous liver metastases within our cancer registry. We estimated overall survival (OS) among various patient subgroups using the Kaplan-Meier method. To mitigate immortal time bias, we analyzed long-term outcomes of patients who survived beyond 12 months (landmark time) from diagnosis. RESULTS We identified 241 patients. Median OS was 7 months (95% CI, 5-9), both overall and for patients with liver-only metastasis (n=144). Ninety patients (38% of liver only; 40% of whole cohort) survived at least 12 months; those who received chemotherapy in this subgroup had a median OS of 26 months (95% CI, 17-39). Of these patients, those with resectable or borderline resectable primary tumors and resectable liver-only metastasis (n=9, 4%) had a median OS of 39 months (95% CI, 13-NR). CONCLUSIONS The 4% of our cohort that were potentially eligible for surgery experienced a prolonged survival compared with all-comers with stage IV disease. Oncologic outcomes of patients undergoing resection of metastatic pancreas cancer should be assessed in the context of the expected survival of patients potentially eligible for surgery and not relative to all patients with stage IV disease.
Collapse
Affiliation(s)
- Sarah R Kaslow
- Department of Surgery, New York University Grossman School of Medicine, New York City, NY
| | - Greg D Sacks
- Department of Surgery, New York University Grossman School of Medicine, New York City, NY
| | - Russell S Berman
- Department of Surgery, New York University Grossman School of Medicine, New York City, NY
| | - Ann Y Lee
- Department of Surgery, New York University Grossman School of Medicine, New York City, NY
| | - Camilo Correa-Gallego
- Department of Surgery, New York University Grossman School of Medicine, New York City, NY
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY
| |
Collapse
|
8
|
van Oosten AF, Daamen LA, Groot VP, Biesma NC, Habib JR, van Goor IWJM, Kinny-Köster B, Burkhart RA, Wolfgang CL, van Santvoort HC, He J, Molenaar IQ. Predicting post-recurrence survival for patients with pancreatic cancer recurrence after primary resection: A Bi-institutional validated risk classification. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:106910. [PMID: 37173152 DOI: 10.1016/j.ejso.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/07/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Over 80% of patients will develop disease recurrence after radical resection of pancreatic ductal adenocarcinoma (PDAC). This study aims to develop and validate a clinical risk score predicting post-recurrence survival (PRS) at time of recurrence. METHODS All patients who had recurrence after undergoing pancreatectomy for PDAC at the Johns Hopkins Hospital or at the Regional Academic Cancer Center Utrecht during the study period were included. Cox proportional hazard model was used to develop the risk model. Performance of the final model was assessed in a test set after internal validation. RESULTS Of 718 resected PDAC patients, 72% had recurrence after a median follow-up of 32 months. The median overall survival was 21 months and the median PRS was 9 months. Prognostic factors associated with shorter PRS were age (hazard ratio [HR] 1.02; 95% confidence interval [95%CI] 1.00-1.04), multiple-site recurrence (HR 1.57; 95%CI 1.08-2.28), and symptoms at time of recurrence (HR 2.33; 95%CI 1.59-3.41). Recurrence-free survival longer than 12 months (HR 0.55; 95%CI 0.36-0.83), FOLFIRINOX and gemcitabine-based adjuvant chemotherapy (HR 0.45; 95%CI 0.25-0.81; HR 0.58; 95%CI 0.26-0.93, respectively) were associated with a longer PRS. The resulting risk score had a good predictive accuracy (C-index: 0.73). CONCLUSION This study developed a clinical risk score based on an international cohort that predicts PRS in patients who underwent surgical resection for PDAC. This risk score will become available on www.evidencio.com and can help clinicians with patient counseling on prognosis.
Collapse
Affiliation(s)
- A Floortje van Oosten
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands; Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lois A Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands; Division of Imaging and Oncology, University Medical Center Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - Vincent P Groot
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | - Nanske C Biesma
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | - Joseph R Habib
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Iris W J M van Goor
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | - Benedict Kinny-Köster
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Surgery, New York University Langone Medical Center, New York City, NY, USA
| | - Richard A Burkhart
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher L Wolfgang
- Department of Surgery, New York University Langone Medical Center, New York City, NY, USA
| | - Hjalmar C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | - Jin He
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands.
| |
Collapse
|
9
|
Ohm H, Abdel-Rahman O. Impact of Patient Characteristics on the Outcomes of Patients with Gastrointestinal Cancers Treated with Immune Checkpoint Inhibitors. Curr Oncol 2023; 30:786-802. [PMID: 36661709 PMCID: PMC9858132 DOI: 10.3390/curroncol30010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Gastrointestinal (GI) cancers are a group of malignancies that globally account for a significant portion of cancer incidence and cancer-related death. Survival outcomes for esophageal, gastric, pancreatic, and hepatobiliary cancers remain poor, but new treatment paradigms are emerging with the advent of immune checkpoint inhibitor (ICI) therapy. This review characterizes patient-related prognostic factors that influence the response to ICI therapy. We performed an analysis of the landmark randomized clinical trials in esophageal, gastric, colorectal, hepatocellular, pancreatic, and biliary tract cancers in terms of patient demographic factors. A literature review of smaller retrospective studies investigating patient-related factors was completed. The immunological bases for these associations were further explored. The key predictive factors identified include age, sex, performance status, geography, body mass index, sarcopenia, gut microbiome, various biochemical factors, and disease distribution.
Collapse
Affiliation(s)
- Hyejee Ohm
- Department of Medicine, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Omar Abdel-Rahman
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
10
|
Asakura Y, Toyama H, Ishida J, Asari S, Terai S, Shirakawa S, Yamashita H, Shimizu T, Ogura Y, Matsumoto I, Gon H, Tsugawa D, Komatsu S, Kuramitsu K, Yanagimoto H, Kido M, Ajiki T, Fukumoto T. Clinicopathological variables and risk factors for lung recurrence after resection of pancreatic ductal adenocarcinoma. Asian J Surg 2023; 46:207-212. [PMID: 35370072 DOI: 10.1016/j.asjsur.2022.03.043] [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: 08/01/2021] [Revised: 02/02/2022] [Accepted: 03/17/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a high recurrence rate even after curative resection. Lung recurrence may have better outcomes than other recurrences. However, its detailed clinicopathological features are unclear. We investigated the clinicopathological features and risk factors for lung recurrence after pancreatectomy for PDAC. METHODS The study included 161 patients with potentially and borderline resectable PDAC who had undergone R0 or R1 pancreatectomy between January 2008 and December 2016. We retrospectively examined the prognosis and predictors for lung recurrence after curative resection. RESULTS Seventeen patients (10.6%) had isolated lung recurrence. The median overall and recurrence-free survivals were 38.0 and 16.1 months, respectively. In multivariate analysis, para-aortic lymph node (PALN) metastasis (p = 0.006) and female sex (p = 0.027) were independent factors for lung recurrence. CONCLUSION Lung recurrence had a better prognosis than other recurrences. PALN metastasis and female sex are independent risk factors for lung recurrence after curative resection for PDAC.
Collapse
Affiliation(s)
- Yu Asakura
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Hirochika Toyama
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Jun Ishida
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Sadaki Asari
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Sachio Terai
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Sachiyo Shirakawa
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Hironori Yamashita
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Takashi Shimizu
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Yuta Ogura
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Ippei Matsumoto
- Department of Surgery, Kindai University Faculty of Medicine, 377-2, Ohno-higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Hidetoshi Gon
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Daisuke Tsugawa
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Shohei Komatsu
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Kaori Kuramitsu
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Hiroaki Yanagimoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Masahiro Kido
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Tetsuo Ajiki
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Takumi Fukumoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| |
Collapse
|
11
|
Conde D, Rey C, Pardo M, Recaman A, Sabogal Olarte JC. Hepatic artery lymph node relevance in periampullary tumors: A retrospective analysis of survival outcomes. Front Surg 2022; 9:963855. [PMID: 36561573 PMCID: PMC9763566 DOI: 10.3389/fsurg.2022.963855] [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: 06/08/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Background The Periampullary area comprehends a heterogeneous and complex structure with different histological tissues. Surgical standards include the peripancreatic regional lymphadenectomy, and during pancreatoduodenectomy (PD) the hepatic artery lymph node HALN(8a) is dissected. We aimed to describe the prognostic significance of the HALN(8a) lymph node metastasis in terms of disease-free survival (DFS) and overall survival (OS) in a specific cohort of patients in limited economic and social conditions. Methods A retrospective study was conducted based on a prospective database from the HPB department of patients who underwent pancreaticoduodenectomy (PD) due to periampullary tumors during 2014-2021. Overall survival (OS) and disease-free survival (DFS) were estimated to be associated with positive HALN(8a) using Kaplan-Meier analysis. Log Rank test and Cox proportional hazards regression analysis was used. Results 111 patients were included, 55,4% female. The most frequent pathology was ductal adenocarcinoma (60.3%). The positive rate of the HALN(8a) node was 21.62%. The Median OS time was 25.5 months, and the median DFS time was 13,8 months. Positive HLAN(8a) node, the cutoff of lymph node ratio resection (LNRR), and vascular invasion showed a strong association with OS. (CoxRegression p = 0.03 HR 0.5, p 0.003 HR = 1.8, p = 0.02 HR 0.4 CI 95%). In terms of DFS, lymph node ratio cutoff, tumoral size, and vascular invasion showed a statistically significant association with the outcome (p = 0.008, HR = 1.5; p = 0.04 HR = 2.1; p = 0.02 HR = 0.4 CI 95%). Conclusion In this series of PD, OS was reduced in patients with HALN(8a) compromise in patients with pancreatic cancer, however without statistical significance in DFS. In multivariate analysis, lymph node status remains an independent predictor of OS and DFS. Further studies are needed.
Collapse
Affiliation(s)
- Danny Conde
- Department of Hepatobiliary and Pancreatic Surgery, Hospital Universitario Mayor Méderi, Bogotá, Colombia
| | - Carlos Rey
- School of Medicine, Universidad el Rosario, Bogotá, Colombia
| | - Manuel Pardo
- School of Medicine, Universidad el Rosario, Bogotá, Colombia
| | - Andrea Recaman
- School of Medicine, Universidad el Rosario, Bogotá, Colombia
| | - Juan Carlos Sabogal Olarte
- Chief and Chairman of Hepatobiliary and Pancreatic Surgery Department, Hospital Universitario Mayor Méderi, Bogotá, Colombia
| |
Collapse
|
12
|
van Goor IWJM, Daamen LA, Besselink MG, Bruynzeel AME, Busch OR, Cirkel GA, Groot Koerkamp B, Haj Mohammed N, Heerkens HD, van Laarhoven HWM, Meijer GJ, Nuyttens J, van Santvoort HC, van Tienhoven G, Verkooijen HM, Wilmink JW, Molenaar IQ, Intven MPW. A nationwide randomized controlled trial on additional treatment for isolated local pancreatic cancer recurrence using stereotactic body radiation therapy (ARCADE). Trials 2022; 23:913. [PMID: 36307892 PMCID: PMC9617359 DOI: 10.1186/s13063-022-06829-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/06/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Disease recurrence is the main cause of mortality after resection of pancreatic ductal adenocarcinoma (PDAC). In 20-30% of resected patients, isolated local PDAC recurrence occurs. Retrospective studies have suggested that stereotactic body radiation therapy (SBRT) might lead to improved local control in these patients, potentially having a beneficial effect on both survival and quality of life. The "nationwide randomized controlled trial on additional treatment for isolated local pancreatic cancer recurrence using stereotactic body radiation therapy" (ARCADE) will investigate the value of SBRT in addition to standard of care in patients with isolated local PDAC recurrence compared to standard of care alone, regarding both survival and quality of life outcomes. METHODS The ARCADE trial is nested within a prospective cohort (Dutch Pancreatic Cancer Project; PACAP) according to the 'Trials within Cohorts' design. All PACAP participants with isolated local PDAC recurrence after primary resection who provided informed consent for being randomized in future studies are eligible. Patients will be randomized for local therapy (5 fractions of 8 Gy SBRT) in addition to standard of care or standard of care alone. In total, 174 patients will be included. The main study endpoint is survival after recurrence. The most important secondary endpoint is quality of life. DISCUSSION It is hypothesized that additional SBRT, compared to standard of care alone, improves survival and quality of life in patients with isolated local recurrence after PDAC resection. TRIAL REGISTRATION ClinicalTrials.gov registration NCT04881487 . Registered on May 11, 2021.
Collapse
Affiliation(s)
- I W J M van Goor
- Department of Surgery, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands.
- Department of Radiation Oncology, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands.
| | - L A Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - M G Besselink
- Department of Surgery, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - A M E Bruynzeel
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Center, location Vrije Universiteit, Amsterdam, the Netherlands
| | - O R Busch
- Department of Surgery, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - G A Cirkel
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - B Groot Koerkamp
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - N Haj Mohammed
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - H D Heerkens
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - H W M van Laarhoven
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center, location University of Amsterdam, Amsterdam, the Netherlands
| | - G J Meijer
- Department of Radiation Oncology, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - J Nuyttens
- Department of Radiation Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - H C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - G van Tienhoven
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - H M Verkooijen
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - J W Wilmink
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center, location University of Amsterdam, Amsterdam, the Netherlands
| | - I Q Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - M P W Intven
- Department of Radiation Oncology, Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
13
|
Villano AM, O’Halloran E, Goel N, Ruth K, Barrak D, Lefton M, Reddy SS. Total neoadjuvant therapy is associated with improved overall survival and pathologic response in pancreatic adenocarcinoma. J Surg Oncol 2022; 126:502-512. [PMID: 35476892 PMCID: PMC9340441 DOI: 10.1002/jso.26906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Few studies have evaluated outcomes of total neoadjuvant therapy (TNT) compared with single modality neoadjuvant therapy (SMNT) or surgery first (SF) for pancreatic ductal adenocarcinoma (PDAC). METHODS A single-institution retrospective review of PDAC patients who underwent pancreatectomy was conducted (1993-2019). Overall survival (OS) estimates from diagnosis and from surgery were determined using Kaplan-Meier methods; Cox proportional hazards models adjusted for covariates. RESULTS Surgery was performed upfront (SF) in 168 (46.9%), while 111 (31.0%) had chemotherapy or chemoradiation before resection (SMNT), and 79 (22.1%) underwent TNT (chemotherapy and chemoradiation). Resection margins were more frequently R0 in the TNT group (86.1%) compared with SMNT (64.0%) and SF (72%) (p < 0.001). Complete pathologic response was more common in the TNT group (10.1%) compared with SMNT (3.6%) or SF (0.6%) (p = 0.001), resulting in prolonged survival (median OS = 100.2 months). TNT patients demonstrated longer median OS from surgery (33.6 months) compared with SF (19.1 months) and SMNT (17.4 months) (p = 0.010), which persisted after controlling for covariates. CONCLUSIONS TNT is associated with more frequent complete pathologic response, a higher rate of margin negative resection, and prolonged OS as compared with SF or SMNT. Additional studies to identify subgroups that derive the greatest benefit are warranted.
Collapse
Affiliation(s)
- Anthony M. Villano
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Eileen O’Halloran
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Neha Goel
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Karen Ruth
- Department of Biostatistics, Fox Chase Cancer Center, Philadelphia, PA
| | - Dany Barrak
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Max Lefton
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Sanjay S. Reddy
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| |
Collapse
|
14
|
Kubo H, Ohgi K, Sugiura T, Ashida R, Yamada M, Otsuka S, Yamazaki K, Todaka A, Sasaki K, Uesaka K. The Association Between Neoadjuvant Therapy and Pathological Outcomes in Pancreatic Cancer Patients After Resection: Prognostic Significance of Microscopic Venous Invasion. Ann Surg Oncol 2022; 29:4992-5002. [PMID: 35368218 DOI: 10.1245/s10434-022-11628-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/04/2022] [Indexed: 12/17/2023]
Abstract
BACKGROUND The impact of neoadjuvant therapy (NAT) on pathological outcomes, including microscopic venous invasion (MVI), remains unclear in pancreatic cancer. METHODS A total of 456 patients who underwent pancreatectomy for resectable and borderline resectable pancreatic cancer between July 2012 and February 2020 were retrospectively reviewed. Patients were divided into two groups: patients with NAT (n = 120, 26%) and those without NAT (n = 336, 74%). Clinicopathological factors, survival outcomes and recurrence patterns were analyzed. RESULTS Regarding pathological findings, the proportion of MVI was significantly lower in patients with NAT than in those without NAT (43% vs 62%, P = 0.001). The 5-year survival rate in patients with NAT was significantly better than that in those without NAT (54% vs 45%, P = 0.030). A multivariate analysis showed that MVI was an independent prognostic factor for the overall survival (OS) (hazard ratio 2.86, P = 0.003) in patients who underwent NAT. MVI was an independent risk factor for liver recurrence (odds ratio [OR] 2.38, P = 0.016) and multiple-site recurrence (OR 1.92, P = 0.027) according to a multivariate analysis. The OS in patients with liver recurrence was significantly worse than that in patients with other recurrence patterns (vs lymph node, P = 0.047; vs local, P < 0.001; vs lung, P < 0.001). The absence of NAT was a significant risk factor for MVI (OR 1.93, P = 0.007). CONCLUSION MVI was a crucial prognostic factor associated with liver and multiple-site recurrence in pancreatic cancer patients with NAT. MVI may be reduced by NAT, which may contribute to the improvement of survival in pancreatic cancer patients.
Collapse
Affiliation(s)
- Hidemasa Kubo
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhisa Ohgi
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan.
| | - Teiichi Sugiura
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Ryo Ashida
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Mihoko Yamada
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Shimpei Otsuka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Akiko Todaka
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Keiko Sasaki
- Division of Diagnostic Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhiko Uesaka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
15
|
Bahardoust M, Abyazi MA, Emami SA, Ghadimi P, Khodabandeh M, Mahmoudi F, Hosseinzadeh R, Heiat M, Agah S. Predictors of survival rate in patients with pancreatic cancer: A multi-center analytical study in Iran. Cancer Rep (Hoboken) 2022; 5:e1547. [PMID: 34494396 PMCID: PMC9351653 DOI: 10.1002/cnr2.1547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is among the deadliest cancers of the gastrointestinal tract worldwide and a growing global health concern. AIM This study was aimed to evaluate the survival rate and prognostic factors of survival in patients with PC. METHODS In this retrospective cohort study, the records of 556 patients with PC registered in the hospital cancer registration system from September 2007 to September 2020 were evaluated. In this regard, demographic data, tumor characteristics, received treatments, and patients' final status were analyzed. Kaplan-Meier and Cox's regression were used for univariate and multivariate analyses, respectively. RESULTS The 5-year survival rate was found to be 4.3%. The median survival time was 12.4 ± 6.6 months. Univariate analysis showed that age, BMI (kg/m2 ), blood transfusions, differentiation, tumor stage, tumor size, number of involved lymph nodes, lymph node ratio (LNR), and type of treatment received were significantly associated with patient survival (p < .05). Multivariate Cox regression indicated that the age ≥60 years [Hazard Ratio (HR) = 1.25, 95% confidence interval (CI) = 1.03-1.49], BMI <18 (kg/m2 ; HR = 1.56, 95% CI = 1.13-2.14), poor differentiation (HR = 2.12, 95% CI = 1.75-2.49), tumor size >2.5 cm (HR = 4.61, 95% CI = 3.30-6.78), metastasis presence (HR = 1.97, 95% CI = 1.49-2.60), more than two involved lymph nodes (HR = 1.52, 95% CI = 1.31-1.77), LNR <0.2 (HR = 0.56, 95% CI = 0.36-0.77), and adjuvant therapy with surgery and chemotherapy (HR = 0.44, 95% CI = 0.28-0.61) are the most important prognostic factors of survival in patients with PC (p < .05). CONCLUSIONS This study showed that the survival rate of patients with pancreatic cancer varies based on the characteristics of the tumor and the type of treatment received.
Collapse
Affiliation(s)
- Mansour Bahardoust
- Baqiyatallah Research Center for Gastroenterology and Liver DiseasesBaqiyatallah University of Medical SciencesTehranIran
- Department of EpidemiologySchool of Public Health, Shahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Ali Abyazi
- Baqiyatallah Research Center for Gastroenterology and Liver DiseasesBaqiyatallah University of Medical SciencesTehranIran
| | - Sayed Ali Emami
- Heart Failure Research CenterIsfahan Cardiovascular Research Institute, Cardiovascular Research Institute, Isfahan University of Medical SciencesIsfahanIran
| | - Parmida Ghadimi
- Faculty of MedicineIran University of Medical SciencesTehranIran
| | - Mehrdad Khodabandeh
- Department of Physical Medicine and RehabilitationIran University of Medical SciencesTehranIran
| | - Farhad Mahmoudi
- Medical Students Research CenterIsfahan University of Medical SciencesIsfahanIran
| | - Ramin Hosseinzadeh
- Baqiyatallah Research Center for Gastroenterology and Liver DiseasesBaqiyatallah University of Medical SciencesTehranIran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver DiseasesBaqiyatallah University of Medical SciencesTehranIran
| | - Shahram Agah
- Colorectal Research CenterIran University of Medical SciencesTehranIran
| |
Collapse
|
16
|
Risk Factors of Early Liver Metastasis for Pancreatic Ductal Adenocarcinoma after Radical Resection. Gastroenterol Res Pract 2022; 2022:8061879. [PMID: 35693325 PMCID: PMC9177337 DOI: 10.1155/2022/8061879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/26/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Background Liver metastasis arises in many postoperative patients with PDAC, occurring in the early stage appears to lead to a very poor prognosis. Objective We aimed to analyze the risk factors for early liver metastasis after radical resection for patients with pancreatic ductal adenocarcinoma (PDAC) and to indicate the poor prognosis of early liver metastasis. Methods Patients who underwent pancreatectomy for PDAC at the Ningbo Medical Centre Lihuili Hospital between January 2015 and June 2021 were included. The exclusion criteria were death within 30 days after the operation, complications with other malignancies, and a positive final resection margin (R1). Liver metastasis and its occurrence time were recorded, and risk factors for early (≤6 months) liver metastasis were analyzed by logistic regression models. The prognosis of patients with early liver metastasis and different recurrence patterns was analyzed by Kaplan–Meier curves and the log-rank test. Results From the identified cohort of 184 patients, 172 patients were included for further analysis. 55 patients developed early liver metastasis within 6 months after the operation. Univariate analysis showed that CA125 ≥ 30 IU/ml, tumor size ≥ 4 cm, poor tumor differentiation, and portal vein/superior mesenteric vein (PV/SMV) reconstruction were risk factors, and multivariate analysis showed that poor tumor differentiation and PV/SMV reconstruction were independent risk factors for early liver metastasis. The prognosis of liver metastasis was the worst among the different recurrence patterns. Early liver metastasis indicates a poor prognosis in patients with PDAC. Conclusions Poor differentiation and PV/SMV reconstruction are independent risk factors for early liver metastasis in patients with PDAC, and early liver metastasis indicates a poor prognosis.
Collapse
|
17
|
Dang C, Wang M, Zhu F, Qin T, Qin R. Controlling nutritional status (CONUT) score-based nomogram to predict overall survival of patients with pancreatic cancer undergoing radical surgery. Asian J Surg 2022; 45:1237-1245. [PMID: 34493426 DOI: 10.1016/j.asjsur.2021.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/30/2021] [Accepted: 08/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND /Objective: As a new immune-nutritional marker, the controlled nutritional status (CONUT) score has been reported to predict the prognosis of cancer patients. We aimed to elucidate the prognostic value of preoperative CONUT score in pancreatic cancer patients undergoing radical surgery, and to construct a nomogram based on CONUT score to predict individual survival. METHODS Preoperative CONUT scores were calculated prospectively in 382 patients with pancreatic cancer who underwent radical surgery. Evaluated the relationship between CONUT score and pancreatic cancer prognosis. Cox proportional hazard models were used to determine predictors of survival and a new nomogram was established to predict pancreatic cancer overall survival (OS). RESULTS The area under curve of CONUT score was higher than other immune-nutritional indexes. The OS of the high-CONUT group were significantly lower than that of low-CONUT group. Multivariate analysis showed that CONUT score, gender, AJCC stage, complications and reoperation were independent prognostic factors for OS. Nomogram based on these variables has better discriminant ability in predicting survival compared with other traditional staging systems. CONCLUSIONS Preoperative CONUT score is an effective independent predictor of OS in pancreatic cancer patients undergoing radical surgery. This new CONUT based nomogram provides accurate, individualized survival prediction for pancreatic cancer.
Collapse
Affiliation(s)
- Chao Dang
- Department of Pancreatic-Biliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Min Wang
- Department of Pancreatic-Biliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Feng Zhu
- Department of Pancreatic-Biliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Tingting Qin
- Department of Pancreatic-Biliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| | - Renyi Qin
- Department of Pancreatic-Biliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| |
Collapse
|
18
|
Strobel O, Lorenz P, Hinz U, Gaida M, König AK, Hank T, Niesen W, Kaiser JÖR, Al-Saeedi M, Bergmann F, Springfeld C, Berchtold C, Diener MK, Schneider M, Mehrabi A, Müller-Stich BP, Hackert T, Jager D, Büchler MW. Actual Five-year Survival After Upfront Resection for Pancreatic Ductal Adenocarcinoma: Who Beats the Odds? Ann Surg 2022; 275:962-971. [PMID: 32649469 DOI: 10.1097/sla.0000000000004147] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To determine actual five-year survival (5YS) rates associated with a strategy of upfront surgery and adjuvant therapy in pancreatic ductal adenocarcinoma (PDAC). BACKGROUND The rate of actual 5YS in PDAC remains controversial. Available data is restricted to cohorts acquired over several decades and series of resection after patient selection by neoadjuvant therapy. METHODS All patients undergoing upfront resection for resectable and borderline-resectable PDAC from 10/2001 to 12/2011 were identified from a prospective database. Actual overall survival was assessed after a follow-up of at least 5 years. Uni- and multivariable logistic regression analyses were performed. RESULTS Median survival of 937 patients was 22.1 months. The actual 5YS rate was 17.0% (n = 159) including 89 (9.5%) patients without evidence of disease >5 years after resection. 5YS rates in patients with or without adjuvanttherapy were 18.8% vs. 12.2%, respectively. Tumorgrading, number of positive lymph nodes, a context of intraductal papillary mucinous neoplasia, and vascular resections were independently associated with 5YS. Patient-related parameters and CA 19-9 levels were associated with observed survival up to 3 years, but lost relevance thereafter. The extent of lymph node involvement was the strongest predictor of 5YS. Patients with pN0R0 had a 5YS rate of 38.2%. in patients with exclusively favorable factors the observed 5YS rate was above 50%. CONCLUSIONS This is the largest series of long-term survivors with histologically confirmed PDAC. With upfront resection and adjuvant therapy an actual overall 5YS rate of 18.8% can be expected. in favorable subgroups actual 5YS is above 50%.
Collapse
Affiliation(s)
- Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Lorenz
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulf Hinz
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias Gaida
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Anna-Katharina König
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Hank
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Willem Niesen
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - J Ö Rg Kaiser
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Mohammed Al-Saeedi
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Frank Bergmann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Pathology, Klinikum Darmstadt GmbH, Darmstadt, Germany
| | - Christoph Springfeld
- National Center for Tumor Diseases, Department of Medical Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christoph Berchtold
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus K Diener
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Arianeb Mehrabi
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Beat P Müller-Stich
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Dirk Jager
- National Center for Tumor Diseases, Department of Medical Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
19
|
Malvi D, Vasuri F, Maloberti T, Sanza V, De Leo A, Fornelli A, Masetti M, Benini C, Lombardi R, Offi MF, Di Marco M, Ravaioli M, Fiorino S, Franceschi E, Brandes AA, Jovine E, D’Errico A, Tallini G, de Biase D. Molecular Characterization of Pancreatic Ductal Adenocarcinoma Using a Next-Generation Sequencing Custom-Designed Multigene Panel. Diagnostics (Basel) 2022; 12:1058. [PMID: 35626213 PMCID: PMC9139796 DOI: 10.3390/diagnostics12051058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023] Open
Abstract
Despite the efforts made in the management of PDAC, the 5-year relative survival rate of pancreatic ductal adenocarcinoma (PDAC) still remains very low (10%). To date, precision oncology is far from being ready to be applied in cases of PDAC, although studies exploring the molecular and genetic alterations have been conducted, and the genomic landscape of PDAC has been characterized. This study aimed to apply a next-generation sequencing (NGS) laboratory-developed multigene panel to PDAC samples to find molecular alterations that could be associated with histopathological features and clinical outcomes. A total of 68 PDACs were characterized by using a laboratory-developed multigene NGS panel. KRAS and TP53 mutations were the more frequent alterations in 75.0% and 44.6% of cases, respectively. In the majority (58.7%) of specimens, more than one mutation was detected, mainly in KRAS and TP53 genes. KRAS mutation was significantly associated with a shorter time in tumor recurrence compared with KRAS wild-type tumors. Intriguingly, KRAS wild-type cases had a better short-term prognosis despite the lymph node status. In conclusion, our work highlights that the combination of KRAS mutation with the age of the patient and the lymph node status may help in predicting the outcome in PDAC patients.
Collapse
Affiliation(s)
- Deborah Malvi
- Pathology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (D.M.); (F.V.); (A.D.)
| | - Francesco Vasuri
- Pathology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (D.M.); (F.V.); (A.D.)
| | - Thais Maloberti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna-Molecular Diagnostic Unit, Azienda USL di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (G.T.)
- Division of Molecular Pathology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Viviana Sanza
- Division of Molecular Pathology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Antonio De Leo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna-Molecular Diagnostic Unit, Azienda USL di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (G.T.)
- Division of Molecular Pathology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Adele Fornelli
- Anatomic Pathology Unit, Azienda USL, Maggiore Hospital, 40133 Bologna, Italy;
| | - Michele Masetti
- Department of General Surgery, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Maggiore Hospital, 40133 Bologna, Italy; (M.M.); (C.B.); (R.L.); (M.F.O.); (E.J.)
| | - Claudia Benini
- Department of General Surgery, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Maggiore Hospital, 40133 Bologna, Italy; (M.M.); (C.B.); (R.L.); (M.F.O.); (E.J.)
| | - Raffaele Lombardi
- Department of General Surgery, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Maggiore Hospital, 40133 Bologna, Italy; (M.M.); (C.B.); (R.L.); (M.F.O.); (E.J.)
| | - Maria Fortuna Offi
- Department of General Surgery, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Maggiore Hospital, 40133 Bologna, Italy; (M.M.); (C.B.); (R.L.); (M.F.O.); (E.J.)
| | - Mariacristina Di Marco
- Division of Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, 40138 Bologna, Italy
| | - Matteo Ravaioli
- Department of General Surgery and Transplantation, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Sirio Fiorino
- Internal Medicine Unit, Budrio Hospital, Budrio (Bologna), Azienda USL di Bologna, 40054 Bologna, Italy;
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (E.F.); (A.A.B.)
| | - Alba A. Brandes
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (E.F.); (A.A.B.)
| | - Elio Jovine
- Department of General Surgery, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Maggiore Hospital, 40133 Bologna, Italy; (M.M.); (C.B.); (R.L.); (M.F.O.); (E.J.)
| | - Antonietta D’Errico
- Pathology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (D.M.); (F.V.); (A.D.)
| | - Giovanni Tallini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna-Molecular Diagnostic Unit, Azienda USL di Bologna, 40138 Bologna, Italy; (T.M.); (A.D.L.); (G.T.)
- Division of Molecular Pathology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Dario de Biase
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
20
|
Sun L, Shang H, Wu Y, Xin X. Hypericin-mediated photodynamic therapy enhances gemcitabine induced Capan-2 cell apoptosis via inhibiting NADPH level. J Pharm Pharmacol 2022; 74:596-604. [PMID: 34089613 DOI: 10.1093/jpp/rgab073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/29/2021] [Indexed: 11/14/2022]
Abstract
OBJECTIVES The combination of gemcitabine (Gem) and hypericin (HY) enhances the apoptosis of Capan-2 cells, providing a promising option for the treatment of pancreatic cancer. Our study further explored the cytotoxic mechanism of HY combined with chemotherapy drugs on pancreatic cancer. METHODS The proliferation rate of the cells assayed with the MTT method. The ROS (reactive oxygen species) levels of each treatment were evaluated by DCFH-DA oxidisation methods. The activity of glutathione reductase and the levels of reduced glutathione (GSH) and oxidised glutathione (GSSG) were assessed using assay kits. The expression levels of apoptosis-related proteins were analysed by western blotting. KEY FINDINGS The activity of glucose-6-phosphate dehydrogenase (G6PDH), a key enzyme of the pentose phosphate pathway, significantly decreased in Gem + HY groups, however, the ROS level enhanced accompanying with GSH depleting, mitochondrial membrane depolarisation and cytochrome C release. Gem + HY inhibits the expression of Bcl-2 but stimulates Bax level, triggering caspase activation and PARP cleavage and thus promoted apoptosis of Capan-2 cells. CONCLUSIONS We demonstrated that Gem combined HY-PDT could inhibit the proliferation of Capan-2 cells and induce cell apoptosis. HY-PDT combined with Gem had a great potential on pancreatic cancer treatment clinically.
Collapse
Affiliation(s)
- Liyun Sun
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Huoli Shang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuzhen Wu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Xiujuan Xin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
21
|
Thomas RJ, Bartee E. The use of oncolytic virotherapy in the neoadjuvant setting. J Immunother Cancer 2022; 10:jitc-2021-004462. [PMID: 35414592 PMCID: PMC9006794 DOI: 10.1136/jitc-2021-004462] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
Surgical removal of tumors remains a front-line therapy for many types of cancer. However, this treatment often fails to eradicate disease due to either recurrence of the original tumor or development of distant micrometastases. To address these challenges, patients are often given non-curative treatments presurgery with the intent of improving surgical outcomes. These treatments, collectively known as neoadjuvant therapies, have traditionally focused on the presurgical use of chemotherapeutics. Recently, however, a variety of immunotherapies have also been identified as potentially effective in the neoadjuvant setting. One of these immunotherapies is oncolytic virotherapy, whose clinical use has exploded with the Food and Drug Administration approval of Talimogene Laherparepvec. This review summarizes both the preclinical and clinical literature examining the use of oncolytic virotherapy in the neoadjuvant setting for different types of cancers and discusses some of the major questions that still need to be addressed in order for this unique use of immunotherapy to become clinically viable.
Collapse
Affiliation(s)
- Raquela J Thomas
- Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Eric Bartee
- Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
22
|
Genetic Alterations Predict Long-Term Survival in Ductal Adenocarcinoma of the Pancreatic Head. Cancers (Basel) 2022; 14:cancers14030850. [PMID: 35159117 PMCID: PMC8833892 DOI: 10.3390/cancers14030850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/28/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Pancreatic cancer is notorious for its poor prognosis. However, rare long-term survivors of pancreatic cancer exist. The aim of this study was to characterize the molecular profile of pancreatic cancer long-term survivors, to improve the stratification and management of pancreatic cancer patients in the future. Thirty-nine pancreatic cancer patients including short-term and long-term survivors were evaluated thoroughly. Their molecular profile was analyzed using panel next generation sequencing. As a result, patients with mutations commonly found in pancreatic cancer (KRAS G12D mutations and/or TP53 nonsense and splice site mutations) showed significantly worse survival. In contrast, long-term survivors of pancreatic cancer did not show the above-mentioned mutations but did show rare mutations of KRAS (Q61H/D57N). In conclusion, long-term survivors of pancreatic cancer do have a distinct molecular profile. Further studies using larger patient cohorts are warranted to confirm these results and possibly unravel rare potential targets for targeted therapy in pancreatic cancer. Abstract Background: Survival of patients with adenocarcinoma of the pancreas (PDAC) is poor and has remained almost unchanged over the past decades. The genomic landscape of PDAC has been characterized in recent years. The aim of this study was to identify a genetic profile as a possible predictor of prolonged survival in order to tailor therapy for PDAC patients. Methods: Panel next generation sequencing (NGS) and immunohistochemistry (IHC) were performed on paraffin-embedded tumor tissues from curatively treated PDAC patients. Tumor slides were re-evaluated with a focus on the histomorphology. Patients were subgrouped according to short and long overall (<4 years/>4 years) and disease-free (<2 years/>2 years) survival. Results: Thirty-nine patients were included in the study. Clinicopathological staging variables as well as the histomorphological subgroups were homogenously distributed between short- and long-term overall and disease-free survivors. In survival analysis, patients with the KRAS G12D mutation and patients with TP53 nonsense and splice-site mutations had a significantly worse overall survival (OS) and disease-free survival (DFS). Patients with long-term OS and DFS showed no KRAS G12D, no TP53 nonsense or splice-site mutations. Rare Q61H/D57N KRAS mutations were only found in long-term survivors. The allele frequency rate of KRAS and TP53 mutations in tumor cells was significantly higher in short-term disease-free survivors and overall survivors, respectively. Conclusions: NGS of PDAC revealed significant differences in survival outcome in a patient collective with homogenously distributed clinicopathological variables. Further multi-institutional studies are warranted to identify more long-term survivors to detect genetic differences suitable for targeted therapy.
Collapse
|
23
|
Li X, Wan Y, Lou J, Xu L, Shi A, Yang L, Fan Y, Yang J, Huang J, Wu Y, Niu T. Preoperative recurrence prediction in pancreatic ductal adenocarcinoma after radical resection using radiomics of diagnostic computed tomography. EClinicalMedicine 2022; 43:101215. [PMID: 34927034 PMCID: PMC8649647 DOI: 10.1016/j.eclinm.2021.101215] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The high recurrence rate after radical resection of pancreatic ductal adenocarcinoma (PDAC) leads to its poor prognosis. We aimed to develop a model to preoperatively predict the risk of recurrence based on computed tomography (CT) radiomics and multiple clinical parameters. METHODS Datasets were retrospectively collected and analysed of 220 PDAC patients who underwent contrast-enhanced computed tomography (CE-CT) and received radical resection at 3 institutions in China between 2013 and 2017, with 153 from one institution as a training set, the remaining 67 as a validation set. For each patient, CT radiomics features were extracted from intratumoral and peritumoral regions to establish intratumoral, peritumoral and combined radiomics models using artificial neural network (ANN) algorithm. By incorporating clinical factors, radiomics-clinical nomograms were finally built by multivariable logistic regression analysis to predict 1- and 2-year recurrence risk. FINDINGS The developed radiomics model integrating intratumoral and peritumoral radiomics features was superior to the conventionally constructed model merely using intratumoral radiomics features. Further, radiomics-clinical nomograms outperformed other models in predicting 1-year recurrence with an area under the receiver operating characteristic curve (AUROC) of 0.916 (95%CI, 0.860-0.955) in the training set and 0.764 (95%CI, 0.644-0.859) in the validation set, and 2-year recurrence with an AUROC of 0.872 (95%CI: 0.809-0.921) in the training set and 0.773 (95%CI, 0.654-0.866) in the validation set. INTERPRETATION This study has developed and externally validated a radiomics-clinical nomogram integrating intra- and peritumoral CT radiomics signature as well as clinical factors to predict the recurrence risk of PDAC after radical resection, which will facilitate optimized and individualized treatment strategies. FUNDING This work was supported by the National Key R&D Program of China [grant number: 2018YFE0114800], the General Program of National Natural Science Foundation of China [grant number: 81772562, 2017; 81871351, 2018], the Fundamental Research Funds for the Central Universities [grant number: 2021FZZX005-08], and Zhejiang Provincial Key Projects of Technology Research [grant number: WKJ-ZJ-2033].
Collapse
Affiliation(s)
- Xiawei Li
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yidong Wan
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianyao Lou
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Xu
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Aiguang Shi
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Litao Yang
- Department of Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Yiqun Fan
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jing Yang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junjie Huang
- Department of Surgery, Changxing People's Hospital, Huzhou, Zhejiang, China
| | - Yulian Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianye Niu
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Lei C, Hou Y, Chen J. Specificity protein 1-activated bone marrow stromal cell antigen 2 accelerates pancreatic cancer cell proliferation and migration. Exp Ther Med 2021; 22:1459. [PMID: 34737799 PMCID: PMC8561758 DOI: 10.3892/etm.2021.10894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022] Open
Abstract
Bone marrow stromal cell antigen 2 (BST2) has been reported to act as an oncogene in the tumorigenesis of numerous types of cancer. Bioinformatics analysis has predicted the binding interaction between BST2 and specificity protein 1 (SP1) and the involvement of SP1 in pancreatic cancer. Therefore, the present study set out to verify this interaction and determine how it may affect pancreatic cancer progression. Normal human pancreatic duct epithelial cells (HPDE6-C7) and pancreatic cancer cell lines (SW1990, BxPC3, PANC1 and PSN-1) were selected for western blotting and reverse transcription-quantitative PCR detection of BST2 expression. Colony formation, Cell Counting Kit-8 and wound healing assays were performed to detect the proliferative and migratory abilities of PANC1 cells following transfection with small interfering RNA against BST2. The expression of proliferation and migration markers were assayed using western blotting. Chromatin immunoprecipitation and luciferase reporter assays were employed to verify the bioinformatics prediction of BST2-SP1 binding. PANC1 cell proliferation and migration were analyzed following BST2 knockdown and SP1 overexpression. In comparison with HPDE6-C7 cells, all four pancreatic cancer cell lines were found to exhibit increased BST2 expression levels to varying degrees, with the highest levels observed in PANC1 cells. BST2 knockdown inhibited PANC1 cell colony formation, proliferation and migration. Additionally, SP1 was shown to bind to the BST2 promoter and could promote PANC1 cell proliferation and migration when overexpressed. However, BST2 knockdown rescued SP1 overexpression-induced PANC1 cell colony formation, proliferation and migration. In conclusion, activation of BST2 by the transcription factor SP1 was shown to accelerate pancreatic cancer cell proliferation and migration, suggesting that BST2 and SP1 may be plausible therapeutic targets in targeted therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Chun Lei
- Department of General Surgery, Tongling People's Hospital, Tongling, Anhui 244009, P.R. China.,Department of General Surgery, Tongling People's Hospital Affiliated to Wannan Medical College, Tongling, Anhui 244009, P.R. China.,Department of General Surgery, Tongling Branch of the First Affiliated Hospital of University of Science and Technology of China, Tongling, Anhui 244009, P.R. China
| | - Yafeng Hou
- Department of General Surgery, Tongling People's Hospital, Tongling, Anhui 244009, P.R. China.,Department of General Surgery, Tongling People's Hospital Affiliated to Wannan Medical College, Tongling, Anhui 244009, P.R. China.,Department of General Surgery, Tongling Branch of the First Affiliated Hospital of University of Science and Technology of China, Tongling, Anhui 244009, P.R. China
| | - Jiong Chen
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, Anhui 230001, P.R. China
| |
Collapse
|
25
|
Kim YJ, Cheon YK, Lee TY, Chang SH, Yu MH. Longstanding postoperative fluid collection influences recurrence of pancreatic malignancy. Korean J Intern Med 2021; 36:1338-1346. [PMID: 34147058 PMCID: PMC8588986 DOI: 10.3904/kjim.2021.056] [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] [Received: 01/26/2021] [Accepted: 03/19/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS Postoperative abdominal fluid collection (PAFC) is a frequent complication of pancreatobiliary cancer surgery. The effects of the existence and duration of PAFC are not well known. This study aimed to assess the effects of PAFC on patient prognosis after surgery for pancreatobiliary adenocarcinoma and the association of longstanding PAFC with the recurrence of pancreatic cancer. METHODS We retrospectively analyzed the data of 194 consecutive patients with pancreatobiliary adenocarcinoma who underwent curative operations from August 2005 to December 2019. The presence of PAFC was assessed using computed tomography within a week of surgery; PAFC lasting > 4 weeks was defined as longstanding PAFC. RESULTS Among 194 patients, PAFC occurred in 165 (85.1%), and 74 of these had longstanding PAFC. The recurrence rate of pancreatobiliary adenocarcinoma was significantly higher in patients with longstanding PAFC than in patients with non-longstanding PAFC (p = 0.025). Recurrence was also significantly associated with high T stage (T3, T4; p = 0.040), lymph node involvement (p < 0.001), perineural invasion (p < 0.006), and non-receipt of adjuvant chemotherapy (p = 0.025). Longstanding PAFC was significantly associated with the recurrence of pancreatic adenocarcinoma (p = 0.016). However, cancer-specific survival was related to neither the presence nor the duration of PAFC. CONCLUSION The presence of longstanding PAFC was associated with the recurrence of pancreatic adenocarcinoma. However, a larger prospective study is necessary to confirm the findings.
Collapse
Affiliation(s)
- Young Jung Kim
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul,
Korea
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju,
Korea
| | - Young Koog Cheon
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul,
Korea
| | - Tae Yoon Lee
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul,
Korea
| | - Seong-Hwan Chang
- Department of Surgery, Konkuk University School of Medicine, Seoul,
Korea
| | - Mi-Hye Yu
- Department of Radiology, Konkuk University School of Medicine, Seoul,
Korea
| |
Collapse
|
26
|
Minimally Invasive Versus Open Radical Antegrade Modular Pancreatosplenectomy: A Meta-Analysis. World J Surg 2021; 46:235-245. [PMID: 34609574 DOI: 10.1007/s00268-021-06328-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Radical antegrade modular pancreatosplenectomy (RAMPS) was introduced to improve the tangential resection margin rates and N1 node clearance following resection of malignancies of the pancreatic body and tail. Owing to its technical complexity, minimally invasive RAMPS (MI-RAMPS) has only been reported by a few centers worldwide. We performed this meta-analysis to compare both short- and long-term outcomes between open RAMPS (O-RAMPS) and minimally invasive RAMPS (MI-RAMPS). METHODS A systematic search of the electronic databases PubMed, Medline (via PubMed), Cochrane Register of Controlled Trials (CENTRAL), EMBASE, Scopus and Web of Science was performed to identify eligible studies published in the English language regardless of study design. The outcomes of interest were operation time, estimated blood loss, transfusion rates, overall complications, Grade B/C post-operative pancreatic fistula (POPF) rates, post-pancreatectomy hemorrhage (PPH), delayed gastric emptying (DGE), length of stay (LOS), R0 resection rates, lymph node (LN) yield and overall survival (OS). RESULTS Five non-randomized studies comprising of a total 229 patients (89 MI-RAMPS, 140 O-RAMPS) were included for analysis. Intra-operative blood loss was observed to be significantly reduced in MI-RAMPS as compared to O-RAMPS (MD -256.16, P < 0.001), while LN yield was higher in O-RAMPS as compared to MI-RAMPS (MD -2.73, P = 0.02). There were no statistically significant differences observed for the other perioperative, oncologic and survival outcomes. CONCLUSIONS This meta-analysis provides early evidence to suggest that MI-RAMPS may produce comparable short- and long-term outcomes to O-RAMPS, when undertaken by appropriately skilled surgeons in well-selected patients. Further large-scale prospective studies are required to corroborate these findings.
Collapse
|
27
|
Mathy RM, Fritz F, Mayer P, Klauss M, Grenacher L, Stiller W, Kauczor HU, Skornitzke S. Iodine concentration and tissue attenuation in dual-energy contrast-enhanced CT as a potential quantitative parameter in early detection of local pancreatic carcinoma recurrence after surgical resection. Eur J Radiol 2021; 143:109944. [PMID: 34482176 DOI: 10.1016/j.ejrad.2021.109944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/08/2021] [Accepted: 08/26/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE Due to the difficult differentiation from non-specific postoperative soft tissue formation (PSF), early diagnosis of pancreatic carcinoma recurrence remains challenging. Thus, we investigated the diagnostic potential of dual-energy (DE) contrast-enhanced CT. METHOD After potentially curative pancreatic carcinoma resection, 31 consecutive patients with PSF were examined via DE perfusion CT, acquiring 34 images (80 kVp/140 kVp) every 1.5 s, as the initial purpose of this study was evaluating CT-Perfusion. Corresponding time points of arterial, pancreatic, and early venous phase were calculated from bolus trigger times in prior conventional CT. Iodine and 120 kVp-equivalent images were calculated. Regions of interest were placed in each soft tissue formation. Diagnosis of local recurrence was confirmed by regular follow-up or histopathology. RESULTS Final diagnosis was local recurrence in 17 patients and non-specific PSF in 14 patients. Iodine concentrations in early venous phase were significantly higher in recurrent carcinoma than in non-specific PSF (1.47 mg/ml vs. 0.96 mg/ml, p = 0.007). In earlier contrast phases iodine concentrations tended to be higher, but not significantly. CT numbers in recurrent carcinoma in 120 kVp-equivalent images in venous phase were significantly higher, too (74HU vs 47HU, p = 0.002). ROC-curve analysis for iodine concentrations in early venous phase suggests a cut-off value of ≥ 1.55 mg/ml for local recurrence (AUC = 0.78, specificity = 1.0, sensitivity = 0.53) and for CT numbers in 120kVp-equivalent images a cut-off value of ≥ 57HU (AUC = 0.82, specificity = 0.82, sensitivity = 0.71). CONCLUSION In difficult cases, measuring iodine concentrations or CT numbers in PSF in (early) venous phase DECT could be a valuable additional parameter for differentiating local recurrence from non-specific PSF.
Collapse
Affiliation(s)
- René Michael Mathy
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| | - Franziska Fritz
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; Radiology Darmstadt, Fachärztezentrum am Klinikum Darmstadt, Grafenstraße 13, 64283 Darmstadt, Germany.
| | - Philipp Mayer
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| | - Miriam Klauss
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| | - Lars Grenacher
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; Conradia Radiology & Medical Prevention, Conradia Radiologie München, Augustenstraße 115, 80798 Munich, Germany.
| | - Wolfram Stiller
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| | - Hans-Ulrich Kauczor
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| | - Stephan Skornitzke
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| |
Collapse
|
28
|
CT Radiomics-Based Preoperative Survival Prediction in Patients With Pancreatic Ductal Adenocarcinoma. AJR Am J Roentgenol 2021; 217:1104-1112. [PMID: 34467768 DOI: 10.2214/ajr.20.23490] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE. Pancreatic ductal adenocarcinoma (PDAC) is often a lethal malignancy with limited preoperative predictors of long-term survival. The purpose of this study was to evaluate the prognostic utility of preoperative CT radiomics features in predicting postoperative survival of patients with PDAC. MATERIALS AND METHODS. A total of 153 patients with surgically resected PDAC who underwent preoperative CT between 2011 and 2017 were retrospectively identified. Demographic, clinical, and survival information was collected from the medical records. Survival time after the surgical resection was used to stratify patients into a low-risk group (survival time > 3 years) and a high-risk group (survival time < 1 year). The 3D volume of the whole pancreatic tumor and background pancreas were manually segmented. A total of 478 radiomics features were extracted from tumors and 11 extra features were computed from pancreas boundaries. The 10 most relevant features were selected by feature reduction. Survival analysis was performed on the basis of clinical parameters both with and without the addition of the selected features. Survival status and time were estimated by a random survival forest algorithm. Concordance index (C-index) was used to evaluate performance of the survival prediction model. RESULTS. The mean age of patients with PDAC was 67 ± 11 (SD) years. The mean tumor size was 3.31 ± 2.55 cm. The 10 most relevant radiomics features showed 82.2% accuracy in the classification of high-risk versus low-risk groups. The C-index of survival prediction with clinical parameters alone was 0.6785. The addition of CT radiomics features improved the C-index to 0.7414. CONCLUSION. Addition of CT radiomics features to standard clinical factors improves survival prediction in patients with PDAC.
Collapse
|
29
|
Carbon ion radiotherapy as definitive treatment in locally recurrent pancreatic cancer. Strahlenther Onkol 2021; 198:378-387. [PMID: 34351449 PMCID: PMC8940823 DOI: 10.1007/s00066-021-01827-9] [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: 12/30/2020] [Accepted: 07/05/2021] [Indexed: 11/22/2022]
Abstract
Purpose Data on management of locally recurrent pancreatic cancer (LRPC) after primary resection are limited. Recently, surprisingly high overall survival rates were reported after irradiation with carbon ions. Here, we report on our clinical experience using carbon ion radiotherapy as definitive treatment in LRPC at the Heidelberg Ion-Beam Therapy Center (HIT). Methods Between 2015 and 2019, we treated 13 patients with LRPC with carbon ions with a median total dose of 48 Gy (RBE) in 12 fractions using an active raster-scanning technique at a rotating gantry. No concomitant chemotherapy was administered. Overall survival, local control, and toxicity rates were evaluated 18 months after the last patient finished radiotherapy. Results With a median follow-up time of 9.5 months, one patient is still alive (8%). Median OS was 12.7 months. Ten patients (77%) developed distant metastases. Additionally, one local recurrence (8%) and two regional tumor recurrences (15%) were observed. The estimated 1‑year local control and locoregional control rates were 87.5% and 75%, respectively. During radiotherapy, we registered one gastrointestinal bleeding CTCAE grade III (8%) due to gastritis. The bleeding was sufficiently managed with conservative therapy. No further higher-grade acute or late toxicities were observed. Conclusion We demonstrate high local control rates in a rare cohort of LRPC patients treated with carbon ion radiotherapy. The observed median overall survival rate was not improved compared to historical in-house data using photon radiotherapy. This is likely due to a high rate of distant tumor progression, highlighting the necessity of additional chemotherapy.
Collapse
|
30
|
Belfiori G, Crippa S, Francesca A, Pagnanelli M, Tamburrino D, Gasparini G, Partelli S, Andreasi V, Rubini C, Zamboni G, Falconi M. Long-Term Survivors after Upfront Resection for Pancreatic Ductal Adenocarcinoma: An Actual 5-Year Analysis of Disease-Specific and Post-Recurrence Survival. Ann Surg Oncol 2021; 28:8249-8260. [PMID: 34258720 DOI: 10.1245/s10434-021-10401-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Data on long-term actual survival in patients with surgically resected pancreatic ductal adenocarcinoma (PDAC) are limited. The aim of this study was to evaluate the actual 5-year disease-specific survival (DSS) and post-recurrence survival (PRS) in patients who underwent pancreatectomy for PDAC. METHODS Data from patients who underwent upfront surgical resection for PDAC between 2009 and 2014 were analyzed. Exclusion criteria included PDAC arising in the background of an intraductal papillary mucinous neoplasm and patients undergoing neoadjuvant therapy. All alive patients had a minimum follow-up of 60 months. Independent predictors of PRS, DSS, and survival > 5 years were searched. RESULTS Of the 176 patients included in this study, 48 (27%) were alive at 5 years, but only 20 (11%) had no recurrence. Median PRS was 12 months. In the 154 patients after disease recurrence, independent predictors of shorter PRS were total pancreatectomy, G3 tumors, early recurrence (< 12 months from surgery), and no treatment at recurrence. Median DSS was 36 months. Independent predictors of DSS were CA19-9 at diagnosis > 200 U/mL, total pancreatectomy, N + status, G3 tumors and perineural invasion. Only the absence of perineural invasion was a favorable independent predictor of survival > 5 years. CONCLUSION More than one-quarter of patients who underwent upfront surgery for PDAC were alive after 5 years, although only 11% of the initial cohort were cancer-free. Long-term survival can also be achieved in tumors with more favorable biology in an upfront setting followed by adjuvant chemotherapy.
Collapse
Affiliation(s)
- Giulio Belfiori
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Crippa
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Aleotti Francesca
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Michele Pagnanelli
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Domenico Tamburrino
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Gasparini
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Partelli
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Andreasi
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Corrado Rubini
- Department of Pathology, Università Politecnica delle Marche, Ospedali Riuniti, Ancona, Italy
| | - Giuseppe Zamboni
- Department of Pathology, Ospedale Sacro Cuore-Don Calabria, Negrar, Italy
| | - Massimo Falconi
- Division of Pancreatic Surgery, Department of Surgery, Pancreas Translational and Clinical Research Center, Università Vita-Salute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
31
|
Fujii Y, Kamachi H, Matsuzawa F, Mizukami T, Kobayashi N, Fukai M, Taketomi A. Early administration of amatuximab, a chimeric high-affinity anti-mesothelin monoclonal antibody, suppresses liver metastasis of mesothelin-expressing pancreatic cancer cells and enhances gemcitabine sensitivity in a xenograft mouse model. Invest New Drugs 2021; 39:1256-1266. [PMID: 33905019 DOI: 10.1007/s10637-021-01118-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/15/2021] [Indexed: 01/11/2023]
Abstract
Amatuximab is a promising therapeutic antibody targeting mesothelin, a 40-kDa glycoprotein that is highly expressed in pancreatic cancer. We investigated the effectiveness of early amatuximab treatment, imitating an adjuvant chemotherapy setting, and combination therapy with amatuximab and gemcitabine in liver metastasis of pancreatic cancer. Liver metastasis mouse models were established in 8-week-old male BALB/c nu/nu mice using the hemisplenic injection method. Tridaily amatuximab monotherapy or combination with gemcitabine was administered to the liver metastasis mouse model before metastatic lesions had formed huge masses. Gaussia luciferase-transfected AsPC-1 was used as a mesothelin-overexpressing pancreatic cancer cell line. The amount of liver metastases and the serum luciferase activity were significantly lower in the treatment groups than those in the control IgG group. Notably, the anti-tumor activity of gemcitabine was synergically enhanced by combination therapy with amatuximab. Furthermore, western blotting revealed that the high expression of phosphorylated c-Met and AKT in liver metastatic lesions treated with gemcitabine monotherapy was canceled by its combination with amatuximab. This result indicated that the observed synergic therapeutic effect may have occurred as a result of the inhibitory effect of amatuximab on the phosphorylation of c-Met and AKT, which were promoted by exposure to GEM. In conclusion, our study revealed that early administration of amatuximab alone or in combination with GEM significantly suppressed the liver metastases of mesothelin-expressing pancreatic cancer cells. A phase II clinical trial of amatuximab as part of an adjuvant chemotherapy regimen for resected pancreatic cancer is expected.
Collapse
Affiliation(s)
- Yuki Fujii
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, N15W7, Kitaku, Sapporo, Hokkaido, 060-8638, Japan
| | - Hirofumi Kamachi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, N15W7, Kitaku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Fumihiko Matsuzawa
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, N15W7, Kitaku, Sapporo, Hokkaido, 060-8638, Japan
| | - Tatsuzo Mizukami
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, N15W7, Kitaku, Sapporo, Hokkaido, 060-8638, Japan
| | - Nozomi Kobayashi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, N15W7, Kitaku, Sapporo, Hokkaido, 060-8638, Japan
| | - Moto Fukai
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, N15W7, Kitaku, Sapporo, Hokkaido, 060-8638, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, N15W7, Kitaku, Sapporo, Hokkaido, 060-8638, Japan
| |
Collapse
|
32
|
Lee DW, Kim HS, Han I. Actual long-term survival after resection of stage III soft tissue sarcoma. BMC Cancer 2021; 21:21. [PMID: 33402132 PMCID: PMC7786893 DOI: 10.1186/s12885-020-07730-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022] Open
Abstract
Background Actuarial survival based on the Kaplan–Meier method can overestimate actual long-term survival, especially among those with factors of poor prognosis. Patients with American Joint Committee on Cancer stage III soft tissue sarcoma (STS) represent a subset with a high risk of STS-specific mortality. Therefore, we aimed to characterize the clinicopathological characteristics associated with actual long-term survival in patients with stage III STS. Methods We retrospectively reviewed 116 patients who underwent surgical resection for stage III STS with curative intent between March 2000 and December 2013. Long-term survivors (n = 61), defined as those who survived beyond 5 years, were compared with short-term survivors (n = 36), who died of STS within 5 years. Results Multivariate logistic regression analyses showed that a tumor size < 10 cm [odds ratio (OR) 3.95, p = 0.047], histological grade of 2 (OR 8.12, p = 0.004), and American Society of Anesthesiologists (ASA) score of 1 (OR 11.25, p = 0.001) were independently associated with actual 5-year survival. However, 66% of the long-term survivors exhibited factors of poor prognosis: 36% had a tumor size > 10 cm and 48% had a histological grade of 3. Leiomyosarcoma (3 of 10) was negatively associated with actual long-term survival. Conclusions Actual 5-year survival after resection of stage III STS was associated with tumor size, histological grade, and ASA score. However, majority of the actual 5-year survivors exhibit factors of poor prognosis, suggesting that aggressive treatment should be offered for a chance of long-term survival in these patients.
Collapse
Affiliation(s)
- Do Weon Lee
- Department of Orthopaedic Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Han-Soo Kim
- Department of Orthopaedic Surgery, Seoul National University Hospital, Seoul, South Korea.,Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Ilkyu Han
- Department of Orthopaedic Surgery, Seoul National University Hospital, Seoul, South Korea. .,Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
33
|
Sekigami Y, Michelakos T, Fernandez-Del Castillo C, Kontos F, Qadan M, Wo JY, Harrison J, Deshpande V, Catalano O, Lillemoe KD, Hong TS, Ferrone CR. Intraoperative Radiation Mitigates the Effect of Microscopically Positive Tumor Margins on Survival Among Pancreatic Adenocarcinoma Patients Treated with Neoadjuvant FOLFIRINOX and Chemoradiation. Ann Surg Oncol 2021; 28:4592-4601. [PMID: 33393047 DOI: 10.1245/s10434-020-09444-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/14/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Microscopically positive margins (R1) negatively impact survival in pancreatic ductal adenocarcinoma (PDAC). For patients with close/positive margins, intraoperative radiotherapy (IORT) can improve local control. The prognostic impact of an R1 resection in patients who receive total neoadjuvant therapy (TNT; FOLFIRINOX with chemoradiation) and IORT is unknown. METHODS Clinicopathologic data were retrospectively collected for borderline/locally advanced (BR/LA) PDAC patients who received TNT and underwent resection between 2011 and 2019. Disease-free (DFS) and overall survival (OS) measured from time of diagnosis were compared between groups. RESULTS Two hundred one patients received TNT and were resected, with a median DFS and OS of 24 months and 47 months, respectively. Eighty-eight patients (44%) received IORT; of these, 69 (78%) underwent an R0 and 19 (22%) an R1 resection. There was no significant difference in clinicopathologic factors between the IORT and no-IORT groups, except for resectability status (LA: IORT 69%, no-IORT 53%, p = 0.021) and surgeons' concern for a positive/close margin. R1 resection was associated with worse DFS and OS in the no-IORT population. However, among patients who received IORT, there was no difference in DFS (R0: 29 months, IQR 14-47 vs R1: 20 months, IQR 15-28; p = 0.114) or OS (R0: 48 months, IQR 25-not reached vs R1: 37 months, IQR 30-47; p = 0.307) between patients who underwent R0 vs R1 resection. In multivariate analysis, within the IORT group, R1 resection was not associated with DFS or OS. CONCLUSION IORT may mitigate the adverse effect of an R1 resection on DFS and OS in BR/LA PDAC patients receiving TNT.
Collapse
Affiliation(s)
- Yurie Sekigami
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA
| | - Theodoros Michelakos
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA
| | - Carlos Fernandez-Del Castillo
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA
| | - Filippos Kontos
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jon Harrison
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Onofrio Catalano
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA.,Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith D Lillemoe
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Wang 460, 15 Parkman Street, Boston, MA, 02114, USA.
| |
Collapse
|
34
|
Important CT and histopathological findings for recurrence and overall survival in patients with pancreatic ductal adenocarcinoma who underwent surgery after neoadjuvant FOLFIRINOX. Eur Radiol 2020; 31:3616-3626. [PMID: 33201279 DOI: 10.1007/s00330-020-07489-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/10/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES To investigate important factors for recurrence-free survival (RFS) and overall survival (OS) in patients with pancreatic ductal adenocarcinoma (PDA) who underwent surgery after neoadjuvant FOLFIRINOX using CT and histopathological findings. MATERIALS AND METHODS Sixty-nine patients with PDA who underwent surgery after neoadjuvant FOLFIRINOX were retrospectively included. All patients underwent baseline and first follow-up CT. Two reviewers assessed the CT findings and resectability based on the NCCN guideline. They graded extrapancreatic perineural invasion (EPNI) using a 3-point scale focused on 5 routes. Clinical and histopathological results, such as T- and N-stage, tumor regression grade (TRG) using the College of American Pathology (CAP) grading system, and resection status, were also investigated. Kaplan-Meier methods were used for RFS and OS. The Cox proportional hazard model and logistic regression model were used to identify significant predictive factors. RESULTS There were 57 patients (82.6%) without residual tumors (R0) and 12 patients (17.4%) with residual tumors (R1 or R2). The median RFS was 13 months (range 0~22 months). For RFS, EPNI on baseline CT (hazard ratio (HR) 2.53, 95% confidence interval (CI) 1.116-5.733, p = 0.026) and TRG (HR 1.76, 95% CI 1.000-3.076, p = 0.046) were important predictors of early recurrence. The mean OS was 48 months (range 11~35 months). For OS, TRG (HR 1.05, 95% CI 1.251-6.559, p = 0.013) was a significant factor. However, there were no independent predictors for residual tumors according to the CT findings. CONCLUSION EPNI on baseline CT and TRG were important prognostic factors for tumor recurrence. In addition, TRG was also an important prognostic factor for OS. KEY POINTS • CT and histopathological findings are helpful for predicting early recurrence and poor survival. • EPNI on baseline CT (HR 2.53, p = 0.026) is an important predictor of early recurrence. • The TRG is an important prognostic factor for early recurrence (HR 1.76, p = 0.046) and poor survival (HR 1.05, p = 0.013).
Collapse
|
35
|
Kim JR, Kim H, Kwon W, Jang JY, Kim SW. Pattern of local recurrence after curative resection in pancreatic ductal adenocarcinoma according to the initial location of the tumor. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2020; 28:105-114. [PMID: 33084211 DOI: 10.1002/jhbp.854] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND/PURPOSE The aim of the present study was to identify the types of recurrence in pancreatic ductal adenocarcinoma (PDAC) and discover the frequent location of the local recurrence. METHODS This study included 361 patients with PDAC who underwent curative-intent surgery between 2007-2014. RESULTS Among 361 patients, 75.1% (n = 271) developed recurrence during the follow-up period. The 5-year overall survival rate of recurred patients was 8.3%. The patterns of recurrence were classified as local (17.7%), systemic (62.0%), and loco-systemic (20.3%). According to the preoperative tumor locations, patients with uncinate and head cancer showed higher rates of local recurrence than those with body and tail cancer (47.8% vs 17.2%, P < .001). When comparing uncinate and head cancer only, patients with uncinate cancer had much more frequent local recurrence around the superior mesenteric artery/vein (M zone) than around the hepatoduodenal ligament/common hepatic artery (H zone). Patients with head cancer had a higher rate of local recurrence in the H zone (H zone vs M zone; 53.5% vs 81.4% in uncinate cancer, P = .001; 66.7% vs 44.4% in head cancer, P = .056). CONCLUSION Discovering the patterns of recurrence and frequent locations of recurrence may assist in local control as well as in the development of a customized individual approach for each patient.
Collapse
Affiliation(s)
- Jae Ri Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Department of Surgery, Pusan National University Hospital, Busan, South Korea
| | - Hongbeom Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Sun-Whe Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, South Korea
| |
Collapse
|
36
|
Xie H, Liu J, Yin J, Ogden JR, Mahipal A, McWilliams RR, Truty MJ, Bekaii‐Saab TS, Petersen GM, Jatoi A, Hubbard JM, Ma WW. Role of Surgery and Perioperative Therapy in Older Patients with Resectable Pancreatic Ductal Adenocarcinoma. Oncologist 2020; 25:e1681-e1690. [PMID: 32663355 PMCID: PMC7648330 DOI: 10.1634/theoncologist.2020-0086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND It is unclear whether results from recent trials of resectable pancreatic ductal adenocarcinoma (PDAC) are generalizable to older patients, who are underrepresented. We aimed to evaluate outcomes of surgery and of neoadjuvant and adjuvant therapy in older patients with resectable PDAC. PATIENTS AND METHODS We included patients aged ≥65 years with upfront resectable PDAC from a prospectively maintained pancreatic cancer registry from 2007 to 2016. Patients were stratified into ages 65-75 and 75+ years. Overall survival (OS) was assessed in treatment comparisons: (A) surgery (n = 636) versus nonsurgical (n = 178), (B) neoadjuvant therapy (n = 139) versus upfront surgery (n = 497), and (C) adjuvant therapy (n = 379) versus surgery alone (n = 118). We compared neoadjuvant (n = 139) versus adjuvant therapy (n = 379) in an exploratory analysis. RESULTS Nine hundred and three patients had a median age of 73.7 (range, 65-96.6) years. Median OS was 26.6 versus 11.9 months (adjusted hazard ratio [HRadj ], 0.4; 95% confidence interval [CI], 0.31-0.52; p < .001) in Comparison A groups, 30.7 versus 25.8 months (HRadj , 0.69; 95% CI, 0.49-0.96; p = .03) in Comparison B groups, and 26.9 versus 17.4 months (HRadj , 0.62; 95% CI, 0.44-0.88; p = .008) in Comparison C groups, respectively. OS benefit in these treatment comparisons was present in age group 75+ with HRadj 0.24 (95% CI, 0.16-0.36; p < .001) in Comparison A and HRadj 0.52 (95% CI, 0.27-1; p = .049) in Comparison B, but not in Comparison C with HRadj 0.68 (95% CI, 0.43-1.08; p = .1). Statistically comparable median OS of patients who received neoadjuvant or adjuvant therapy stratified by age groups was observed. CONCLUSION Older patients with resectable PDAC who received surgery, neoadjuvant therapy, or adjuvant therapy appeared to have improved survival outcomes compared with those who did not receive such treatment. IMPLICATIONS FOR PRACTICE Older patients with resectable pancreatic ductal adenocarcinoma (PDAC) in general are underrepresented in large clinical trials and less well studied in terms of the role of surgery, neoadjuvant therapy, and adjuvant therapy. This study collected data on older patients with resectable PDAC from a prospectively maintained single-institutional pancreatic cancer registry of a tertiary referral center from 2007 to 2016. It was found that, with multidisciplinary evaluation, older patients with resectable PDAC who received surgery, neoadjuvant therapy, or adjuvant therapy appeared to have improved survival outcomes compared with those who did not receive such treatment. These results are of substantial importance to practitioners who treat older patients, who are traditionally underrepresented in most clinical trials.
Collapse
Affiliation(s)
- Hao Xie
- Divisions of Medical Oncology, Mayo ClinicRochesterMinnesotaUSA
| | - Junjia Liu
- Albert Einstein College of MedicineBronxNew YorkUSA
| | - Jun Yin
- Divisions of Biomedical Statistics and Informatics, Mayo ClinicRochesterMinnesotaUSA
| | - John R. Ogden
- Divisions of Internal Medicine, Mayo ClinicRochesterMinnesotaUSA
| | - Amit Mahipal
- Divisions of Medical Oncology, Mayo ClinicRochesterMinnesotaUSA
| | | | - Mark J. Truty
- Divisions of Hepatobiliary and Pancreas Surgery, Mayo ClinicRochesterMinnesotaUSA
| | | | | | - Aminah Jatoi
- Divisions of Medical Oncology, Mayo ClinicRochesterMinnesotaUSA
| | | | - Wen Wee Ma
- Divisions of Medical Oncology, Mayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
37
|
Bengtsson A, Andersson R, Ansari D. The actual 5-year survivors of pancreatic ductal adenocarcinoma based on real-world data. Sci Rep 2020; 10:16425. [PMID: 33009477 PMCID: PMC7532215 DOI: 10.1038/s41598-020-73525-y] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/17/2020] [Indexed: 12/14/2022] Open
Abstract
Survival data for pancreatic cancer are usually based on actuarial calculations and actual long-term survival rates are rarely reported. Here we use population-level data from the Surveillance, Epidemiology, and End Results program for patients with microscopically confirmed pancreatic ductal adenocarcinoma diagnosed from 1975 to 2011. A total of 84,275 patients with at least 5 years of follow-up were evaluated (follow-up cutoff date: December 31, 2016). Actual 5-year survival for pancreatic cancer increased from 0.9% in 1975 to 4.2% in 2011 in patients of all stages (p < 0.001), while in surgically resected patients, it rose from 1.5% to 17.4% (p < 0.001). In non-resected patients, the actual 5-year survival remained unchanged over the same time period (0.8% vs 0.9%; p = 0.121). Multivariable analysis of surgically resected patients diagnosed in the recent time era (2004-2011) showed that age, gender, grade, tumour size, TNM-stage and chemotherapy were significant independent predictors of actual 5-year survival, while age, grade and TNM-stage were significant independent predictors in non-resected patients. However, unfavourable clinicopathological factors did not preclude long-term survival. Collectively, our findings indicate that actual 5-year survival for pancreatic cancer is still below 5% despite improvement of survival for the subset of patients undergoing surgical resection.
Collapse
Affiliation(s)
- Axel Bengtsson
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | - Roland Andersson
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | - Daniel Ansari
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, 221 85, Lund, Sweden.
| |
Collapse
|
38
|
Lee J, Lee JC, Kim HW, Kim J, Hwang JH. Postoperative muscle mass restoration as a prognostic factor in patients with resected pancreatic cancer. PLoS One 2020; 15:e0238649. [PMID: 32936836 PMCID: PMC7494072 DOI: 10.1371/journal.pone.0238649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/20/2020] [Indexed: 01/06/2023] Open
Abstract
Background Recent studies have found that muscle depletion may be a prognostic predictor in patients with pancreatic cancer (PC). However, in these studies, limited data were used to assess the relationship between the serial change in body composition and outcomes after PC resection. Hence, we evaluated the changes in body composition during the perioperative period in patients with PC and their association with the overall survival (OS). Methods A total of 89 patients with PC who received surgery with curative intent between 2006 and 2015 were included in this study retrospectively. These patients underwent serial computed tomography (CT) scans: preoperatively, immediately after surgery (4 weeks), and 12 and 24 weeks after resection. The muscle and visceral fat areas were measured at the third lumbar vertebra level on cross-sectional CT images using sliceOmatic V5.0 program (TomoVision, Canada). The body composition ratio was determined by dividing the post-resection body composition at each point (4, 12, and 24 weeks) by the pre-resection body composition. Patients were divided into two groups—higher and lower groups—based on this body composition ratio (skeletal muscle mass ratio [SMR], visceral fat mass ratio [VFR]). The OS was compared between the two groups using the log-rank test. Results The median age of patients was 63 (27–84) years, and the baseline body mass index was 23.0 (17.0–35.8) kg/m2. In the comparison of the SMR, there was no significant difference in the OS between the two groups at 4 and 12 weeks (4 weeks, P = 0.488; 12 weeks, P = 0.397). However, the higher group showed a longer OS than the lower group at 24 weeks (39 vs. 20 months, P = 0.008). Similarly, in the VFR, there was no significant difference in the OS between the two groups at 4 and 12 weeks (4 weeks, P = 0.732; 12 weeks, P = 0.060). However, the OS was longer in the higher group at 24 weeks (35 vs. 22 months, P = 0.023). When we analyzed the effect of muscle restoration at 24 weeks after resection on the OS by gender, there was no significant difference between the OS and SMR in the male group (P = 0.213), but a significant difference was noted in the female group (P = 0.002). In the multivariate Cox regression analysis, the SMR at 24 weeks after resection was significantly associated with the OS (P = 0.023) but not VFR at 24 weeks. In 69 patients without recurrence at 6 months, the SMR at 24 weeks was related to longer OS but without statistical significance (P = 0.07). Conclusions This study suggests that the restoration of muscle mass at 24 weeks after resection may be an independent prognostic factor for survival in patients with resected PC.
Collapse
Affiliation(s)
- Jongchan Lee
- Department of Internal Medicine, College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jong-chan Lee
- Department of Internal Medicine, College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyoung Woo Kim
- Department of Internal Medicine, College of Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Jaihwan Kim
- Department of Internal Medicine, College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- * E-mail:
| |
Collapse
|
39
|
Gillies RJ, Schabath MB. Radiomics Improves Cancer Screening and Early Detection. Cancer Epidemiol Biomarkers Prev 2020; 29:2556-2567. [PMID: 32917666 DOI: 10.1158/1055-9965.epi-20-0075] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/18/2020] [Accepted: 08/31/2020] [Indexed: 11/16/2022] Open
Abstract
Imaging is a key technology in the early detection of cancers, including X-ray mammography, low-dose CT for lung cancer, or optical imaging for skin, esophageal, or colorectal cancers. Historically, imaging information in early detection schema was assessed qualitatively. However, the last decade has seen increased development of computerized tools that convert images into quantitative mineable data (radiomics), and their subsequent analyses with artificial intelligence (AI). These tools are improving diagnostic accuracy of early lesions to define risk and classify malignant/aggressive from benign/indolent disease. The first section of this review will briefly describe the various imaging modalities and their use as primary or secondary screens in an early detection pipeline. The second section will describe specific use cases to illustrate the breadth of imaging modalities as well as the benefits of quantitative image analytics. These will include optical (skin cancer), X-ray CT (pancreatic and lung cancer), X-ray mammography (breast cancer), multiparametric MRI (breast and prostate cancer), PET (pancreatic cancer), and ultrasound elastography (liver cancer). Finally, we will discuss the inexorable improvements in radiomics to build more robust classifier models and the significant limitations to this development, including access to well-annotated databases, and biological descriptors of the imaged feature data.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. .,Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
40
|
Radiographic patterns of first disease recurrence after neoadjuvant therapy and surgery for patients with resectable and borderline resectable pancreatic cancer. Surgery 2020; 168:440-447. [DOI: 10.1016/j.surg.2020.04.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 12/16/2022]
|
41
|
Honselmann KC, Pergolini I, Castillo CFD, Deshpande V, Ting D, Taylor MS, Bolm L, Qadan M, Wellner U, Sandini M, Bausch D, Warshaw AL, Lillemoe KD, Keck T, Ferrone CR. Timing But Not Patterns of Recurrence Is Different Between Node-negative and Node-positive Resected Pancreatic Cancer. Ann Surg 2020; 272:357-365. [PMID: 32675550 PMCID: PMC6639153 DOI: 10.1097/sla.0000000000003123] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Our aim was to evaluate recurrence patterns of surgically resected PDAC patients with negative (pN0) or positive (pN1) lymph nodes. SUMMARY BACKGROUND DATA Pancreatic ductal adenocarcinoma (PDAC) is predicted to become the second leading cause of cancer death by 2030. This is mostly due to early local and distant metastasis, even after surgical resection. Knowledge about patterns of recurrence in different patient populations could offer new therapeutic avenues. METHODS Clinicopathologic data were collected for 546 patients who underwent resection of their PDAC between 2005 and 2016 from 2 tertiary university centers. Patients were divided into an upfront resection group (n = 394) and a neoadjuvant group (n = 152). RESULTS Tumor recurrence was significantly less common in pN0 patients as compared with pN1 patients, (upfront surgery: 55% vs. 77%, P < 0.001 and 64% vs. 78%, P = 0.040 in the neoadjuvant group). In addition, time to recurrence was significantly longer in pN0 versus pN1 patients in the upfront resected patients (median 16 mo pN0 vs. 10 mo pN1 P < 0.001), and the neoadjuvant group (pN0 21 mo vs. 11 mo pN1, P < 0.001). Of the patients who recurred, 62% presented with distant metastases (63% of pN0 and 62% of pN1, P = 0.553), 24% with local disease (27% of pN0 and 23% of pN1, P = 0.672) and 14% with synchronous local and distant disease (10% of pN0 and 15% of pN1, P = 0.292). Similarly, there was no difference in recurrence patterns between pN0 and pN1 in the neoadjuvant group, in which 68% recurred with distant metastases (76% of pN0 and 64% of pN1, P = 0.326) and 18% recurred with local disease (pN0: 22% and pN1: 15%, P = 0.435). CONCLUSION Time to recurrence was significantly longer for pN0 patients. However, patterns of recurrence for pN0 vs. pN1 patients were identical. Lymph node status was predictive of time to recurrence, but not location of recurrence.
Collapse
Affiliation(s)
- Kim C Honselmann
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Ilaria Pergolini
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Carlos Fernandez-Del Castillo
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Vikram Deshpande
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - David Ting
- MGH Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Martin S Taylor
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Louisa Bolm
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Motaz Qadan
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ulrich Wellner
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Marta Sandini
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Dirk Bausch
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Andrew L Warshaw
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Keith D Lillemoe
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Tobias Keck
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Cristina R Ferrone
- Department of Gastrointestinal Surgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
42
|
Nweke EE, Brand M. Downregulation of the let-7 family of microRNAs may promote insulin receptor/insulin-like growth factor signalling pathways in pancreatic ductal adenocarcinoma. Oncol Lett 2020; 20:2613-2620. [PMID: 32782579 PMCID: PMC7400736 DOI: 10.3892/ol.2020.11854] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer type characterized by dysregulated cell signalling pathways and resistance to treatment. The insulin-like growth factor (IGF) signalling pathway has been identified to have a role in tumour progression and therapy resistance. However, its regulatory roles in PDAC have remained to be fully elucidated. In the present study, dysregulated microRNAs (miRNAs) in PDAC were explored with a focus on those that may be involved in regulating the insulin/IGF signalling pathway. A total of 208 patients were recruited, comprising 112 patients with PDAC, 50 patients with chronic pancreatitis (CP) and 46 subjects as a control group (CG). miRNA-specific quantitative PCR assays were used to measure 300 candidate miRNAs. The Student's t-test was applied to compare miRNA regulation between cancer patients and controls with a false discovery rate correction using Bonferroni-type comparison procedures. The DIANA-mirPath v.3 tool and HMDD v3.0 were used to identify miRNA-mRNA interactions within specific pathways. In patients with PDAC, 42 miRNAs were significantly upregulated and 42 were downregulated compared to the CG (P<0.01). In the PDAC vs. CP analysis, 16 significantly (P<0.01) upregulated and 16 downregulated miRNAs were identified. Of note, members of the let-7 family of miRNAs were downregulated and were indicated to target several components of the insulin receptor (INSR)/IGF pathway, including receptors and binding proteins, for upregulation and thus, may enable the activation of the pathway. Downregulation of the let-7 family may help promote the INSR/IGF pathway in PDAC. It may thus be an effective target for the development of INSR/IGF pathway-specific treatment strategies.
Collapse
Affiliation(s)
- Ekene Emmanuel Nweke
- Department of Surgery, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg 2193, South Africa
| | - Martin Brand
- School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg 2193, South Africa.,Department of Surgery, Steve Biko Academic Hospital and The University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
43
|
Yanagimoto H, Satoi S, Yamamoto T, Yamaki S, Hirooka S, Kotsuka M, Ryota H, Ishida M, Matsui Y, Sekimoto M. Benefits of Conversion Surgery after Multimodal Treatment for Unresectable Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2020; 12:cancers12061428. [PMID: 32486418 PMCID: PMC7352934 DOI: 10.3390/cancers12061428] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Traditionally, the treatment options for unresectable locally advanced (UR-LA) and metastatic (UR-M) pancreatic ductal adenocarcinoma (PDAC) are palliative chemotherapy or chemoradiotherapy. The benefits of surgery for such patients remains unknown. The present study investigated clinical outcomes of patients undergoing conversion surgery (CS) after chemo(radiation)therapy for initially UR-PDAC. METHODS We recruited patients with UR-PDAC who underwent chemo(radiation)therapy for initially UR-PDAC between April 2006 and September 2017. We analyzed resectability of CS, predictive parameters for overall survival, and early recurrence (within six months). RESULTS A total of 468 patients (108 with UR-LA and 360 with UR-M PDAC) were enrolled in this study, of whom, 17 (15.7%) with UR-LA and 15 (4.2%) with UR-M underwent CS. The median survival time (MST) and five-year survival of patients who underwent CS was 37.2 months and 34%, respectively; significantly better than non-resected patients (nine months and 1%, respectively, p < 0.0001). MST did not differ according to UR-LA or UR-M (50.5 vs. 29.0 months, respectively, p = 0.53). Early recurrence after CS occurred in eight patients (18.8%). Lymph node metastasis, positive washing cytology, large tumor size (>35 mm), and lack of postoperative adjuvant chemotherapy were statistically significant predictive factors for early recurrence. Moreover, the site of pancreatic lesion and administration of postoperative adjuvant chemotherapy were statistically significant prognostic factors for overall survival in the patients undergoing CS. CONCLUSION Conversion surgery offers benefits in terms of increase survival for initially UR-PDAC for patients who responded favorably to chemo(radiation)therapy when combined with postoperative adjuvant chemotherapy.
Collapse
Affiliation(s)
- Hiroaki Yanagimoto
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| | - Sohei Satoi
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
- Correspondence: ; Tel.: +81-72-804-0101; Fax: +81-72-804-2578
| | - Tomohisa Yamamoto
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| | - So Yamaki
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| | - Satoshi Hirooka
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| | - Masaya Kotsuka
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| | - Hironori Ryota
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| | - Mitsuaki Ishida
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Hirakata 573-1010, Japan;
| | - Yoichi Matsui
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| | - Mitsugu Sekimoto
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.Y.); (T.Y.); (S.Y.); (S.H.); (M.K.); (H.R.); (Y.M.); (M.S.)
| |
Collapse
|
44
|
Ren H, Wu CR, Aimaiti S, Wang CF. Development and validation of a novel nomogram for predicting the prognosis of patients with resected pancreatic adenocarcinoma. Oncol Lett 2020; 19:4093-4105. [PMID: 32382348 PMCID: PMC7202273 DOI: 10.3892/ol.2020.11495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
The survival prediction for patients with resected pancreatic adenocarcinoma by using the Tumor-Node-Metastasis (TNM) staging system remains limited. A nomogram is a efficient tool that can be used to predict the outcome of patients with various types of malignancy. The present study aimed to develop and validate a nomogram for patients with resected pancreatic adenocarcinoma. A total of 368 patients (258 in the training set and 110 in the validation set) who underwent pancreatic adenocarcinoma resection at the China National Cancer Center between January 2008 and October 2018 were included in the present study. The nomogram was established according to the results from Cox multivariate analysis, which was validated by discrimination and calibration. The area under the receiver operating characteristic curve (AUC) was determined to assess the accuracy of survival predictions. The results from multivariate analysis in the training set demonstrated that blood transfusion, T-stage, N-stage, tumor grade, capsule invasion, carbohydrate antigen 199, neutrophil percentage and adjuvant therapy were independent prognostic factors for overall survival (OS; all P<0.05). Subsequently, a nomogram predicting the 1-year, 3-year and 5-year OS rates, with favorable calibration, was established based on the independent prognostic factors. The concordance indices of the nomogram were higher compared with the TNM staging system in both training and validation sets. Furthermore, a clear risk stratification system based on the nomogram was used to classify patients into the three following groups: Low-risk group (≤168), moderate-risk group (168–255) and high-risk group (>255). The risk stratification system demonstrated an improved ability in predicting the 1-year, 3-year and 5-year OS rates compared with the TNM system (AUC, 0.758, 0.709 and 0.672 vs. AUC, 0.614, 0.604 and 0.568; all P<0.05). The present study developed and validated a nomogram for patients with resected pancreatic adenocarcinoma by including additional independent prognostic factors, including tumor marker, immune index, surgical information, pathological data and adjuvant therapy. Taken together, the results from the present study indicated an improved performance of the nomogram in predicting the prognosis of patients with resected pancreatic adenocarcinoma compared with the TNM staging system.
Collapse
Affiliation(s)
- Hu Ren
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Chao-Rui Wu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Saderbieke Aimaiti
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Cheng-Feng Wang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| |
Collapse
|
45
|
Huang H, Zou Y, Zhang H, Li X, Li Y, Deng X, Sun H, Guo Z, Ao L. A qualitative transcriptional prognostic signature for patients with stage I-II pancreatic ductal adenocarcinoma. Transl Res 2020; 219:30-44. [PMID: 32119844 DOI: 10.1016/j.trsl.2020.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/14/2020] [Accepted: 02/10/2020] [Indexed: 02/04/2023]
Abstract
Accurately prognostic evaluation of patients with stage I-II pancreatic ductal adenocarcinoma (PDAC) is of importance to treatment decision and patient management. Most previously reported prognostic signatures were based on risk scores summarized from quantitative expression measurements of signature genes, which are susceptible to experimental batch effects and impractical for clinical applications. Based on the within-sample relative expression orderings of genes, we developed a robust qualitative transcriptional prognostic signature, consisting of 64 gene pairs (64-GPS), to predict the overall survival (OS) of 161 stage I-II PDAC patients in the training dataset who were treated with surgery only. Samples were classified into the high-risk group when at least 25 of 64 gene pairs suggested it was at high risk. The signature was successfully validated in 324 samples from 6 independent datasets produced by different laboratories. All samples in the low-risk group had significantly better OS than samples in the high-risk group. Multivariate Cox regression analyses showed that the 64-GPS remained significantly associated with the OS of patients after adjusting available clinical factors. Transcriptomic analysis of the 2 prognostic subgroups showed that the differential expression signals were highly reproducible in all datasets, whereas the differences between samples grouped by the TNM staging system were weak and irreproducible. The epigenomic analysis showed that the epigenetic alternations may cause consistently transcriptional changes between the 2 different prognostic groups. The genomic analysis revealed that mutation‑induced disturbances in several key genes, such as LRMDA, MAPK10, and CREBBP, might lead to poor prognosis for PDAC patients. Conclusively, the 64-GPS can robustly predict the prognosis of patients with stage I-II PDAC, which provides theoretical basis for clinical individualized treatment.
Collapse
Affiliation(s)
- Haiyan Huang
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yi Zou
- Department of Automation and Key Laboratory of China MOE for System Control and Information Processing, Shanghai Jiao Tong University, Shanghai, China
| | - Huarong Zhang
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiang Li
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yawei Li
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xusheng Deng
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huaqin Sun
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zheng Guo
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Key Laboratory of Medical Bioinformatics, Fujian Province, Fuzhou, China
| | - Lu Ao
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Key Laboratory of Medical Bioinformatics, Fujian Province, Fuzhou, China.
| |
Collapse
|
46
|
De Jesus-Acosta A, Narang A, Mauro L, Herman J, Jaffee EM, Laheru DA. Carcinoma of the Pancreas. ABELOFF'S CLINICAL ONCOLOGY 2020:1342-1360.e7. [DOI: 10.1016/b978-0-323-47674-4.00078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
47
|
Rho SY, Lee SG, Park M, Lee J, Lee SH, Hwang HK, Lee MJ, Paik YK, Lee WJ, Kang CM. Developing a preoperative serum metabolome-based recurrence-predicting nomogram for patients with resected pancreatic ductal adenocarcinoma. Sci Rep 2019; 9:18634. [PMID: 31819109 PMCID: PMC6901525 DOI: 10.1038/s41598-019-55016-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
We investigated the potential application of preoperative serum metabolomes in predicting recurrence in patients with resected pancreatic cancer. From November 2012 to June 2014, patients who underwent potentially curative pancreatectomy for pancreatic ductal adenocarcinoma were examined. Among 57 patients, 32 were men; 42 had pancreatic head cancers. The 57 patients could be clearly categorized into two main clusters using 178 preoperative serum metabolomes. Patients within cluster 2 showed earlier tumor recurrence, compared with those within cluster 1 (p = 0.034). A nomogram was developed for predicting the probability of early disease-free survival in patients with resected pancreatic cancer. Preoperative cancer antigen (CA) 19-9 levels and serum metabolomes PC.aa.C38_4, PC.ae.C42_5, and PC.ae.C38_6 were the most powerful preoperative clinical variables with which to predict 6-month and 1-year cancer recurrence-free survival after radical pancreatectomy, with a Harrell's concordance index of 0.823 (95% CI: 0.750-0.891) and integrated area under the curve of 0.816 (95% CI: 0.736-0.893). Patients with resected pancreatic cancer could be categorized according to their different metabolomes to predict early cancer recurrence. Preoperative detectable parameters, serum CA 19-9, PC.aa.C38_4, PC.ae.C42_5, and PC.ae.C38_6 were the most powerful predictors of early recurrence of pancreatic cancer.
Collapse
Affiliation(s)
- Seoung Yoon Rho
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Pancreatobiliary Cancer Center, Severance Hospital, Seoul, Korea
| | - Sang-Guk Lee
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Minsu Park
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinae Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Hwan Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ho Kyoung Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Pancreatobiliary Cancer Center, Severance Hospital, Seoul, Korea
| | - Min Jung Lee
- Yonsei Proteome Research Center and ‡Department of Integrated OMICS for Biomedical Science and Department of Biochemistry, Yonsei University College of Life Science and Biotechnology, Seoul, Korea
| | - Young-Ki Paik
- Yonsei Proteome Research Center and ‡Department of Integrated OMICS for Biomedical Science and Department of Biochemistry, Yonsei University College of Life Science and Biotechnology, Seoul, Korea
| | - Woo Jung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Pancreatobiliary Cancer Center, Severance Hospital, Seoul, Korea
| | - Chang Moo Kang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea.
- Yonsei Pancreatobiliary Cancer Center, Severance Hospital, Seoul, Korea.
| |
Collapse
|
48
|
Ren B, Liu X, Suriawinata AA. Pancreatic Ductal Adenocarcinoma and Its Precursor Lesions: Histopathology, Cytopathology, and Molecular Pathology. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:9-21. [PMID: 30558727 DOI: 10.1016/j.ajpath.2018.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 10/10/2018] [Accepted: 10/23/2018] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma is one of the most aggressive malignant neoplasms with poor outcomes. At the time of diagnosis, the disease is usually at an advanced stage and only a minority is eligible for surgical resection. To improve the prognosis, it is essential to diagnose and treat the disease in an early stage before its progression into an invasive disease. This article reviews clinical features, histopathology, cytopathology, and molecular alterations of pancreatic ductal adenocarcinoma and its precursors. Moreover, we review a recently updated two-tier classification system for precursor lesions, new findings in premalignant cystic neoplasms, and recently updated staging criteria for invasive carcinoma based on the Cancer Staging Manual, eighth edition, from the American Joint Committee on Cancer. Finally, we discuss the potential clinical applications of the rapidly growing molecular and genetic information of pancreatic cancer and its precursors.
Collapse
Affiliation(s)
- Bing Ren
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Xiaoying Liu
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Arief A Suriawinata
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
49
|
Mehtsun WT, Hashimoto DA, Ferrone CR. Status of 5-Year Survivors of the Whipple Procedure for Pancreatic Adenocarcinoma. Adv Surg 2019; 53:253-269. [PMID: 31327451 DOI: 10.1016/j.yasu.2019.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Affiliation(s)
- Winta T Mehtsun
- Department of Surgery, Massachusetts General Hospital, 15 Parkman Street, WAC 460, Boston, MA 02114, USA
| | - Daniel A Hashimoto
- Department of Surgery, Massachusetts General Hospital, 15 Parkman Street, WAC 460, Boston, MA 02114, USA
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, 15 Parkman Street, WAC 460, Boston, MA 02114, USA.
| |
Collapse
|
50
|
Byun Y, Han Y, Kang JS, Choi YJ, Kim H, Kwon W, Kim SW, Oh DY, Lee SH, Ryu JK, Kim YT, Jang JY. Role of surgical resection in the era of FOLFIRINOX for advanced pancreatic cancer. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2019; 26:416-425. [PMID: 31218836 DOI: 10.1002/jhbp.648] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The introduction of FOLFIRINOX regimen greatly changed the treatment for advanced pancreatic cancers. However, detailed studies on the clinical effects and factors affecting the prognosis are insufficient. We performed this study to evaluate the effects of FOLFIRINOX and the surgical resection in advanced pancreatic cancer. METHODS Three hundred and thirty-seven patients with advanced pancreatic cancer who initially received FOLFIRINOX, from January 2011 to December 2017, were retrospectively reviewed. Patients were evaluated according to the National Comprehensive Cancer Network guideline, responses after four to six cycles of FOLFIRINOX were re-evaluated according to the response evaluation criteria in solid tumors, and further treatment was decided in the multidisciplinary meeting. RESULTS Sixty-seven (19.9%) patients had borderline resectable pancreatic cancer, 135 (40.1%) locally advanced pancreatic cancer, and 135 (40.1%) metastatic pancreatic cancer. The median survival period was significantly longer in the surgical group than in the nonsurgical group in each clinical stage, even in metastatic pancreatic cancer (32 vs. 14, P = 0.012). In multivariate analysis, metastatic status at diagnosis, progressive disease after FOLFIRINOX, surgical resection, and declined CA19-9 after FOLFIRINOX were significant prognostic factors. CONCLUSIONS Surgical treatment greatly affects survival outcomes in advanced pancreatic cancer treated with FOLFIRINOX. Further studies on the optimal indication of operation and the protocol are needed.
Collapse
Affiliation(s)
- Yoonhyeong Byun
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Youngmin Han
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Jae Seung Kang
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Yoo Jin Choi
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hongbeom Kim
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Wooil Kwon
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Sun-Whe Kim
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sang Hyub Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Ji Kon Ryu
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Yong-Tae Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin-Young Jang
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|