51
|
Li Z, Zhuo Q, Li B, Liu M, Chen C, Shi Y, Xu W, Liu W, Ji S, Yu X, Xu X. Feasibility of laparoscopic versus open pancreatoduodenectomy following neoadjuvant chemotherapy for borderline resectable pancreatic cancer: a retrospective cohort study. World J Surg Oncol 2024; 22:1. [PMID: 38169384 PMCID: PMC10759588 DOI: 10.1186/s12957-023-03277-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND There is no evidence supporting the feasibility of laparoscopic pancreaticoduodenectomy (LPD) compared to open pancreatoduodenectomy (OPD) following neoadjuvant chemotherapy (NACT) for pancreatic ductal adenocarcinoma (PDAC). METHODS The clinical data of consecutive patients with borderline resectable PDAC who received NACT and underwent either LPD or OPD between January 2020 and December 2022 at Fudan University Shanghai Cancer Center was prospectively collected and retrospectively analyzed. RESULTS The analysis included 57 patients in the OPD group and 20 in the LPD group. Following NACT, the LPD group exhibited a higher median CA19-9 decrease rate compared to the OPD group (85.3% vs. 66.9%, P = 0.042). Furthermore, 3 anatomically borderline PDACs in the LPD group and 5 in the OPD group were downstaged into resectable status (30.0% vs. 12.3%, P = 0.069). According to RECIST criteria, 51 (66.2%) patients in the entire cohort were evaluated as having stable disease. The median operation time for the LPD group was longer than the OPD group (419 vs. 325 min, P < 0.001), while the venous resection rate was 35.0% vs. 43.9%, respectively (P = 0.489). There was no difference in the number of retrieved lymph nodes, with a median number of 18.5 in the LPD group and 22 in the OPD group, and the R1 margin rate (15.0% vs. 12.3%) was also comparable. The incidence of Clavien-Dindo complications (35.0% vs. 66.7%, P = 0.018) was lower in the LPD group compared to the OPD group. Multivariable regression analysis revealed that a tumor diameter > 3 cm before NACT (HR 2.185) and poor tumor differentiation (HR 1.805) were independent risk factors for recurrence-free survival, and a decrease rate of CA19-9 > 70% (OR 0.309) was a protective factor for early tumor recurrence and overall survival. CONCLUSIONS LPD for PDAC following NACT is feasible and oncologically equivalent to OPD. Effective control of CA19-9 levels is beneficial in reducing early tumor recurrence and improving overall survival.
Collapse
Affiliation(s)
- Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Qifeng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Borui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Mengqi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yihua Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
52
|
Yang HK, Park MS, Han K, Eom G, Chung YE, Choi JY, Bang S, Kang CM, Seong J, Kim MJ. Risk Stratification of Pancreatic Ductal Adenocarcinoma Patients Undergoing Curative-Intent Surgery after Neoadjuvant Therapy. Cancer Res Treat 2024; 56:247-258. [PMID: 37605535 PMCID: PMC10789942 DOI: 10.4143/crt.2023.586] [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: 04/17/2023] [Accepted: 08/21/2023] [Indexed: 08/23/2023] Open
Abstract
PURPOSE Clinical prognostic criteria using preoperative factors were not developed for post-neoadjuvant therapy (NAT) surgery of pancreatic ductal adenocarcinoma (PDAC). We aimed to identify preoperative factors associated with overall survival (OS) in PDAC patients who underwent post-NAT curative-intent surgery and develop risk stratification criteria. MATERIALS AND METHODS Consecutive PDAC patients who underwent post-NAT curative-intent surgeries between 2007 and 2020 were retrospectively analyzed. Demographic, laboratory, surgical, and histopathologic variables were collected. Baseline, preoperative, and interval changes of computed tomography (CT) findings proposed by the Society of Abdominal Radiology and the American Pancreatic Association were analyzed. Cox proportional hazard analysis was used to select preoperative variables associated with OS. We developed risk stratification criteria composed of the significant preoperative variables, i.e., post-NAT response criteria. We compared the discrimination performance of post-NAT response criteria with that of post-NAT pathological (yp) American Joint Cancer Committee TNM staging system. RESULTS One hundred forty-five PDAC patients were included. Stable or increased tumor size on CT (hazard ratio [HR], 2.58; 95% confidence interval [CI], 1.58 to 4.21; p < 0.001) and elevated preoperative carbohydrate antigen 19-9 (CA19-9) level (HR, 1.98; 95% CI, 1.11 to 3.55; p=0.021) were independent factors of OS. The OS of the patient groups stratified by post-NAT response criteria which combined changes in tumor size and CA19-9 showed significant difference (p < 0.001). Such stratification was comparable to ypTNM staging in discrimination performance (difference of C-index, 0.068; 95% CI, -0.012 to 0.142). CONCLUSION "Any degree of decrease in tumor size on CT" and CA19-9 normalization or staying normal were independent favorable factors of OS. The combination of the two factors discriminated OS comparably to ypTNM staging.
Collapse
Affiliation(s)
- Hyun Kyung Yang
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mi-Suk Park
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Sciences and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Geonsik Eom
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Eun Chung
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jin-Young Choi
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seungmin Bang
- Department of Internal Medicine, Severance Hospital, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Moo Kang
- Department of Surgery, Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jinsil Seong
- Department of Radiation Oncology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Myeong-Jin Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
53
|
Akahori T, Terai T, Nagai M, Nakamura K, Kohara Y, Yasuda S, Matsuo Y, Doi S, Sakata T, Sho M. Total neoadjuvant therapy improves survival of patients with borderline resectable pancreatic cancer with arterial involvement. Ann Gastroenterol Surg 2024; 8:151-162. [PMID: 38250684 PMCID: PMC10797818 DOI: 10.1002/ags3.12726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 01/23/2024] Open
Abstract
Aim This study aimed to evaluate the prognostic impact of total neoadjuvant therapy (TNT) for borderline resectable pancreatic cancer with arterial involvement (BR-A) pancreatic cancer. Methods We analyzed 81 patients initially diagnosed as BR-A who received initial treatments between 2007 and 2021. Among them, 18 patients who received upfront surgery were classified as the UFS group, while 30 patients who were treated with neoadjuvant chemoradiotherapy were classified as the NACRT group. Furthermore, 33 patients who planned to receive a combination treatment of over 6 months of systemic chemotherapies followed by chemoradiotherapy before surgery were classified as the TNT group. Results There were no significant differences in the patients' backgrounds between the three groups at the time of initial treatment. The resection rates of the UFS, NACRT, and TNT groups were 89%, 77%, and 67%, respectively. NACRT had no impact on the prognosis compared to upfront surgery. In sharp contrast, the TNT group had a significantly better prognosis compared to the other groups, especially after pancreatic resection. Multivariate analysis demonstrated that TNT and resection were independent prognostic factors for the patients of BR-A. Conclusion TNT can be a promising therapeutic strategy for patients with BR-A.
Collapse
Affiliation(s)
| | - Taichi Terai
- Department of SurgeryNara Medical UniversityNaraJapan
| | - Minako Nagai
- Department of SurgeryNara Medical UniversityNaraJapan
| | - Kota Nakamura
- Department of SurgeryNara Medical UniversityNaraJapan
| | | | | | - Yasuko Matsuo
- Department of SurgeryNara Medical UniversityNaraJapan
| | - Shunsuke Doi
- Department of SurgeryNara Medical UniversityNaraJapan
| | | | - Masayuki Sho
- Department of SurgeryNara Medical UniversityNaraJapan
| |
Collapse
|
54
|
Ikuta S, Aihara T, Nakajima T, Yamanaka N. Predicting Pathological Response to Preoperative Chemotherapy in Pancreatic Ductal Adenocarcinoma Using Post-Chemotherapy Computed Tomography Radiomics. Cureus 2024; 16:e52193. [PMID: 38348011 PMCID: PMC10859726 DOI: 10.7759/cureus.52193] [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: 01/12/2024] [Indexed: 02/15/2024] Open
Abstract
INTRODUCTION Assessing the response to preoperative treatment in pancreatic cancer provides valuable information for guiding subsequent treatment strategies. The present study aims to develop and validate a computed tomography (CT) radiomics-based machine learning (ML) model for predicting pathological response (PR) to preoperative chemotherapy in pancreatic ductal adenocarcinoma (PDAC). METHODS Retrospective data were analyzed from 86 PDAC patients undergoing neoadjuvant or conversion chemotherapy followed by surgical resection from January 2018 to May 2023. The cohort was randomly divided into training (70%, n = 60) and testing (30%, n = 26) sets. Favorable PR was defined as Evans grade IIb or greater. Radiomic features were extracted from post-chemotherapy CT images, and dimensionality reduction was performed using the least absolute shrinkage and selection operator (LASSO) logistic regression. Four ML classifiers (Light Gradient Boosting Machine (LGBM), Random Forest, AdaBoost, and Quadratic Discriminant Analysis) were evaluated for predicting a favorable PR. Model performance was primarily assessed using the area under the receiver operating characteristic curve (AUC), Brier score, and decision curve analysis. RESULTS Forty-one (47.7%) patients had a favorable PR. LASSO analysis on the training set identified five radiomic features. The LGBM model demonstrated the best performance, with a training AUC of 0.902 and a testing AUC of 0.923. It also exhibited the lowest Brier scores, both in training (0.136) and testing (0.135). Decision curve analysis further confirmed its clinical potential. CONCLUSION The CT radiomics-based ML model exhibited promising performance in predicting PR in PDAC after neoadjuvant/conversion chemotherapy. This suggests clinical utility in optimizing surgical candidates and timing of surgery, leading to personalized treatment strategies.
Collapse
|
55
|
Shao Z, Chen X, Qiu H, Xu M, Wen X, Chen Z, Liu Z, Ding X, Zhang L. CircNEK6 promotes the progression of pancreatic ductal adenocarcinoma through targeting miR-503/CCND1 axis. Transl Oncol 2024; 39:101810. [PMID: 37871516 PMCID: PMC10622713 DOI: 10.1016/j.tranon.2023.101810] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/16/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
PURPOSE The present study aimed to reveal the function and underlying molecular mechanism of circRNA NIMA related kinase 6 (circNEK6) in promoting the progression of pancreatic ductal adenocarcinoma (PDAC). METHODS The differentially expressed circRNAs in three paired PDAC tissues and adjacent tissues were identified by RNA sequencing. CircNEK6 was screened out to further explore its relationship with the prognosis of PDAC patients. The target microRNAs and mRNAs of circNEK6 were analyzed through online databases and detected by quantitative real-time polymerase chain reaction. Cell counting kit-8 assay, clone formation assay, transwell assay, flow cytometry and western blot were used to explore the function of circNEK6 on the biological behaviors of PDAC cells. The in vivo antitumor effect of circNEK6 silencing on PDAC was investigated by nude mouse xenograft models. RESULTS 203 differentially expressed circRNAs including circNEK6 were identified between paired PDAC tissues and adjacent tissues, and the expression level of circNEK6 was negatively correlated with the prognosis of PDAC patients. The results of in vitro experiments showed that knockdown of circNEK6 repressed the proliferation, migration and invasion, but induced the apoptosis of PDAC cells. Moreover, circNEK6 silencing inhibited tumor growth and prolonged the survival time of PDAC-bearing mice. Mechanistically, miR-503/cyclin D1 (CCND1) axis was predicted and confirmed as the target of circNEK6. CONCLUSIONS CircNEK6 serves as a competing endogenous RNA of CCND1 by absorbing miR-503, which might be treated as a novel and potential target for PDAC treatment.
Collapse
Affiliation(s)
- Zhiying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xueting Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Hui Qiu
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, Jiangsu 221000, China
| | - Muchen Xu
- Department of Radiation Oncology, Dushu Lake Hospital Affilated to Soochow University, Medical Center of Soochow University, Suzhou, Jiangsu 215000, China
| | - Xin Wen
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, Jiangsu 221000, China
| | - Ziqin Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Zhengyang Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xin Ding
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, Jiangsu 221000, China.
| | - Longzhen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, Jiangsu 221000, China.
| |
Collapse
|
56
|
Vitello D, Talamonti MS. Modern Treatment Strategies for Borderline Resectable Pancreatic Cancer. Cancer Treat Res 2024; 192:67-88. [PMID: 39212916 DOI: 10.1007/978-3-031-61238-1_4] [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: 09/04/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to be a daunting clinical challenge. In 2023, it is estimated that 64,000 people will be newly diagnosed with PDAC and 51,000 people will die from PDAC. By 2030, PDAC is predicted to be the second leading cause of cancer-related death, second only to lung cancer (Siegel et al in, CA Cancer J Clin 73(1):17-48, 2023). It is a disease characterized by its late presentation, rapid demise thereafter, and, until recently, relatively ineffective systemic therapies. Despite this grim prognosis, appreciable progress has been made in the identification of patients with localized disease, who may be candidates for potentially curative resections, and in the understanding of the technical nuances and efficacy of aggressive surgical procedures. Currently, the overall 5-year survival rate is 15-25% for patients who undergo resection and receive adjuvant chemotherapy with or without chemoradiation therapy.
Collapse
Affiliation(s)
- Dominic Vitello
- Feinberg School of Medicine, Department of Surgery, Northwestern University, Evanston, IL, USA
| | - Mark S Talamonti
- University of Chicago Pritzker School of Medicine, Chicago, IL, USA.
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA.
| |
Collapse
|
57
|
Leonhardt CS, Hank T, Pils D, Gustorff C, Sahora K, Schindl M, Verbeke CS, Strobel O, Klaiber U. Prognostic impact of resection margin status on survival after neoadjuvant treatment for pancreatic cancer: systematic review and meta-analysis. Int J Surg 2024; 110:453-463. [PMID: 38315795 PMCID: PMC10793837 DOI: 10.1097/js9.0000000000000792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/10/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND A greater than 1 mm tumour-free resection margin (R0 >1 mm) is a prognostic factor in upfront-resected pancreatic ductal adenocarcinoma. After neoadjuvant treatment (NAT); however, the prognostic impact of resection margin (R) status remains controversial. METHODS Randomised and non-randomised studies assessing the association of R status and survival in resected pancreatic ductal adenocarcinoma after NAT were sought by systematic searches of MEDLINE, Web of Science and CENTRAL. Hazard ratios (HR) and their corresponding 95% CI were collected to generate log HR using the inverse-variance method. Random-effects meta-analyses were performed and the results presented as weighted HR. Sensitivity and meta-regression analyses were conducted to account for different surgical procedures and varying length of follow-up, respectively. RESULTS Twenty-two studies with a total of 4929 patients were included. Based on univariable data, R0 greater than 1 mm was significantly associated with prolonged overall survival (OS) (HR 1.76, 95% CI 1.57-1.97; P<0.00001) and disease-free survival (DFS) (HR 1.66, 95% CI 1.39-1.97; P<0.00001). Using adjusted data, R0 greater than 1 mm was significantly associated with prolonged OS (HR 1.65, 95% CI 1.39-1.97; P<0.00001) and DFS (HR 1.76, 95% CI 1.30-2.39; P=0.0003). Results for R1 direct were comparable in the entire cohort; however, no prognostic impact was detected in sensitivity analysis including only partial pancreatoduodenectomies. CONCLUSION After NAT, a tumour-free margin greater than 1 mm is independently associated with improved OS as well as DFS in patients undergoing surgical resection for pancreatic cancer.
Collapse
Affiliation(s)
- Carl-Stephan Leonhardt
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Hank
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Charlotte Gustorff
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Klaus Sahora
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Martin Schindl
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Caroline S. Verbeke
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oliver Strobel
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Ulla Klaiber
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
58
|
Wahler IL, Damanakis A, Große Hokamp N, Bruns C, Schmidt T. Therapy of Locally Advanced and Oligometastatic Pancreatic Adenocarcinoma. Cancers (Basel) 2023; 15:5881. [PMID: 38136425 PMCID: PMC10741431 DOI: 10.3390/cancers15245881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic adenocarcinoma is a lethal disease, and surgical resection remains the only curative treatment option. Unfortunately, upon primary diagnosis, only 15-20% of all patients with pancreatic ductal adenocarcinoma (PDAC) have localized disease that is eligible for operation. The remainder of patients either have borderline resectable or locally advanced disease or present with distant metastasis. In this review, we present a comprehensive overview regarding the current strategies and future directions in the multimodal therapy of locally advanced and oligometastasized pancreatic adenocarcinoma and discuss the benefit of surgery following neoadjuvant therapy in these patients.
Collapse
Affiliation(s)
- Isabell Luisa Wahler
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| | - Alexander Damanakis
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| | - Nils Große Hokamp
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital of Cologne, 50923 Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| | - Thomas Schmidt
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| |
Collapse
|
59
|
Nicolais LM, Mohamed A, Macgillivray D, Verdini N, Inhorn R, Dugan M, Hayward CM, Fitzgerald TL. Improved Overall Survival of Patients with Pancreatic Cancer through Multiagent Chemotherapy and Increased Rates of Surgical Resection. Am Surg 2023; 89:5964-5971. [PMID: 37295019 DOI: 10.1177/00031348221148350] [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] [Indexed: 06/11/2023]
Abstract
BACKGROUND Seminal trials have demonstrated improved survival in pancreatic adenocarcinoma with novel multiagent chemotherapy regimens. To understand the clinical ramifications of this paradigm shift, we reviewed our institutional experience. METHODS This retrospective cohort study utilized a prospective database at a single institution to study all patients diagnosed with and treated for pancreatic adenocarcinoma between 2000 and 2020. RESULTS 1,572 patients were included of which 36% were diagnosed before (Era 1) and 64% after (Era 2) 2011. Survival improved in Era 2 (Median survival 10 vs 8 months, HR .79; P < .001). The survival advantage for Era 2 was primarily seen in patients with high-risk disease (12 vs10 months, HR .71; P < .001). A similar trend was noted for patients undergoing surgical resection (26 vs 21 months, HR .80; P = .081) and with imminently resectable tumors (19 vs 15 months, HR .88; P = .4); however, this was not statistically significant. There was no survival advantage for patients with stage IV disease (4 vs 4 months). Patients in Era 2 were more likely to undergo surgery (OR 2.78; CI 2.00-3.92, P < .001). This increase was driven primarily by increased surgical resection for those with high-risk disease (42 vs 20%, OR 3.74; P < .001). DISCUSSION/CONCLUSIONS This single institutional study showed improved survival after the shift to novel chemotherapy regimens. This was driven by improved survival for patients with high-risk disease and may be due to more effective eradication of microscopic metastatic disease with adjuvant chemotherapy and increased resection rates.
Collapse
Affiliation(s)
- Laura M Nicolais
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
- Graduate School of Biomedical Sciences, Tufts University Clinical and Translational Science Graduate Program, Boston, MA, USA
| | | | - Dougald Macgillivray
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Nicholas Verdini
- Tufts University School of Medicine, Washington St, Boston, MA, USA
- Boston Medical Center, Boston, MA, USA
| | - Roger Inhorn
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Matthew Dugan
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Cynthia M Hayward
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Timothy L Fitzgerald
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| |
Collapse
|
60
|
Terai T, Nagai M, Nakamura K, Kohara Y, Yasuda S, Matsuo Y, Doi S, Sakata T, Sho M. Combination of carbohydrate antigen 19-9 level and tumor size after neoadjuvant chemoradiation therapy may predict early recurrence of resectable pancreatic ductal adenocarcinoma. Pancreatology 2023; 23:970-977. [PMID: 37914628 DOI: 10.1016/j.pan.2023.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Although the overall survival rate of patients with resectable pancreatic cancer has gradually improved, some patients relapse early and have a poor prognosis. This study aimed to identify the preoperative risk factors for early recurrence after neoadjuvant chemoradiotherapy in patients with resectable pancreatic cancer. METHODS This study analyzed patients who underwent pancreatectomy after receiving neoadjuvant chemoradiotherapy for resectable pancreatic cancer between January 2009 and June 2021 and excluded those with borderline resectable and unresectable pancreatic cancers. Early recurrence was defined as recurrence within 6 months after surgery. RESULTS This study included 203 patients, of whom 22 experienced early recurrence. The median survival time of patients with early recurrence was 18.3 months, which was significantly worse than that of patients with late recurrence (44.0 months, p < 0.001) or no recurrence (not reached, p < 0.001). Logistic regression analysis revealed that a carbohydrate antigen 19-9 level of >100 units/mL and a T status of ≥T2 after neoadjuvant chemoradiotherapy were independent predictive risk factors for early recurrence. The median recurrence-free survival time of patients with two risk factors was 9.7 months and significantly worse than that of those with either risk factors (20.5 months, p = 0.024) and those with no risk factor (26.2 months, p < 0.001). CONCLUSIONS A combination of a high-level carbohydrate antigen 19-9 and a T status of ≥T2 after neoadjuvant chemoradiotherapy are predictors of early recurrence and may be helpful for selecting patients who require a stronger preoperative treatment.
Collapse
Affiliation(s)
- Taichi Terai
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Minako Nagai
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Kota Nakamura
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Yuichiro Kohara
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Satoshi Yasuda
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Yasuko Matsuo
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Shunsuke Doi
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Takeshi Sakata
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan.
| |
Collapse
|
61
|
Karbhari A, Mosessian S, Trivedi KH, Valla F, Jacobson M, Truty MJ, Patnam NG, Simeone DM, Zan E, Brennan T, Chen H, Kuo PH, Herrmann K, Goenka AH. Gallium-68-labeled fibroblast activation protein inhibitor-46 PET in patients with resectable or borderline resectable pancreatic ductal adenocarcinoma: A phase 2, multicenter, single arm, open label non-randomized study protocol. PLoS One 2023; 18:e0294564. [PMID: 38011131 PMCID: PMC10681241 DOI: 10.1371/journal.pone.0294564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/20/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease prone to widespread metastatic dissemination and characterized by a desmoplastic stroma that contributes to poor outcomes. Fibroblast activation protein (FAP)-expressing Cancer-Associated Fibroblasts (CAFs) are crucial components of the tumor stroma, influencing carcinogenesis, fibrosis, tumor growth, metastases, and treatment resistance. Non-invasive tools to profile CAF identity and function are essential for overcoming CAF-mediated therapy resistance, developing innovative targeted therapies, and improved patient outcomes. We present the design of a multicenter phase 2 study (clinicaltrials.gov identifier NCT05262855) of [68Ga]FAPI-46 PET to image FAP-expressing CAFs in resectable or borderline resectable PDAC. METHODS We will enroll up to 60 adult treatment-naïve patients with confirmed PDAC. These patients will be eligible for curative surgical resection, either without prior treatment (Cohort 1) or after neoadjuvant therapy (NAT) (Cohort 2). A baseline PET scan will be conducted from the vertex to mid-thighs approximately 15 minutes after administering 5 mCi (±2) of [68Ga]FAPI-46 intravenously. Cohort 2 patients will undergo an additional PET after completing NAT but before surgery. Histopathology and FAP immunohistochemistry (IHC) of initial diagnostic biopsy and resected tumor samples will serve as the truth standards. Primary objective is to assess the sensitivity, specificity, and accuracy of [68Ga]FAPI-46 PET for detecting FAP-expressing CAFs. Secondary objectives will assess predictive values and safety profile validation. Exploratory objectives are comparison of diagnostic performance of [68Ga]FAPI-46 PET to standard-of-care imaging, and comparison of pre- versus post-NAT [68Ga]FAPI-46 PET in Cohort 2. CONCLUSION To facilitate the clinical translation of [68Ga]FAPI-46 in PDAC, the current study seeks to implement a coherent strategy to mitigate risks and increase the probability of meeting FDA requirements and stakeholder expectations. The findings from this study could potentially serve as a foundation for a New Drug Application to the FDA. TRIAL REGISTRATION @ClinicalTrials.gov identifier NCT05262855.
Collapse
Affiliation(s)
- Aashna Karbhari
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sherly Mosessian
- Clinical Development, Sofie Biosciences, Dulles, Virginia, United States of America
| | - Kamaxi H. Trivedi
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Frank Valla
- Radiopharmaceutical and Contract Manufacturing, Sofie Biosciences, Dulles, Virginia, United States of America
| | - Mark Jacobson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Mark J. Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Nandakumar G. Patnam
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Diane M. Simeone
- Departments of Surgery and Pathology, NYU Langone Health, New York, New York, United States of America
| | - Elcin Zan
- Department of Radiology, Weill Cornell Medicine, New York, New York, United States of America
| | - Tracy Brennan
- Discovery Life Sciences, Newtown, Pennsylvania, United States of America
| | - Hongli Chen
- Discovery Life Sciences, Newtown, Pennsylvania, United States of America
| | - Phillip H. Kuo
- Departments of Medical Imaging, Medicine and Biomedical Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ajit H. Goenka
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
62
|
Farnes I, Kleive D, Verbeke CS, Aabakken L, Issa-Epe A, Småstuen MC, Fosby BV, Dueland S, Line PD, Labori KJ. Resection rates and intention-to-treat outcomes in borderline and locally advanced pancreatic cancer: real-world data from a population-based, prospective cohort study (NORPACT-2). BJS Open 2023; 7:zrad137. [PMID: 38155512 PMCID: PMC10755199 DOI: 10.1093/bjsopen/zrad137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Systemic chemotherapy is the initial treatment strategy for borderline resectable and locally advanced pancreatic cancer to facilitate curative resection. The aim of this study was to investigate the resection rates and overall survival in patients with borderline resectable pancreatic cancer and locally advanced pancreatic cancer. METHODS Consecutive patients with borderline resectable pancreatic cancer/locally advanced pancreatic cancer discussed by Oslo University Hospital multidisciplinary team between 2018 and 2020, serving a population of 3.1 million within a geographically defined area in south-eastern Norway, were included in this prospective Norwegian Pancreatic Cancer Trial-2 study, according to intention-to-treat principles. The total number of patients with pancreatic cancer was sought from the Cancer Registry of Norway. RESULTS A total of 1178 patients were diagnosed with pancreatic cancer, of whom 618 were referred to Oslo University Hospital. After multidisciplinary team evaluation, 230 patients were considered to have borderline resectable pancreatic cancer/locally advanced pancreatic cancer. The final study group consisted of 188 patients (borderline resectable pancreatic cancer n = 96, locally advanced pancreatic cancer n = 92) who were fit to receive primary chemotherapy. Resection rates were 46.9% (45 of 96) for borderline resectable pancreatic cancer and 13% (12 of 92) for locally advanced pancreatic cancer (P <0.001). Median overall survival was 14.6 months (borderline resectable pancreatic cancer 16.4 months; locally advanced pancreatic cancer 13.7 months, (P = 0.2)). Adjusted for immortal time bias, median overall survival for patients undergoing resection versus only chemotherapy was 24.4 months versus 10.1 months (P <0.001) for borderline resectable pancreatic cancer and 28.4 months versus 12.6 months for locally advanced pancreatic cancer (P = 0.001). CONCLUSION Resection rates and survival in patients with borderline resectable pancreatic cancer and locally advanced pancreatic cancer treated at a high-volume centre in a universal healthcare system compare well with those treated at international expert centres.Registration number: NCT04423731 (http://www.clinicaltrials.gov).
Collapse
Affiliation(s)
- Ingvild Farnes
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dyre Kleive
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Caroline S Verbeke
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Lars Aabakken
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Aart Issa-Epe
- Department of Radiology, Oslo University Hospital, Oslo, Norway
| | | | - Bjarte V Fosby
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Svein Dueland
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Pål-Dag Line
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Knut J Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
63
|
Shrikhande SV, Kunte AR, Chopde AN, Chaudhari VA, Bhandare MS. Big data and RCT's in surgical oncology: Impact on improving hepatopancreatobiliary cancer surgical care on the global stage. J Surg Oncol 2023; 128:1003-1010. [PMID: 37818909 DOI: 10.1002/jso.27467] [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: 09/08/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023]
Abstract
Randomized controlled clinical trials (RCTs) are at the heart of "evidence-based" medicine. Conducting well-designed RCTs for surgical procedures is often challenged by inadequate recruitment accrual, blinding, or standardization of the surgical procedure, as well as lack of funding and evolution of the treatment strategy during the many years over which such trials are conducted. In addition, most clinical trials are performed in academic high-volume centers with highly selected patients, which may not necessarily reflect a "real-world" practice setting. Large databases provide easy and inexpensive access to data on a large and diverse patient population at a variety of treatment centers. Furthermore, large database studies provide the opportunity to answer questions that would be impossible or very arduous to answer using RCTs, including questions regarding health policy efficacy, trends in surgical practice, access to health care, the impact of hospital volume, and adherence to practice guidelines, as well as research questions regarding rare disease, infrequent surgical outcomes, and specific subpopulations. Prospective data registries may also allow for quality benchmarking and auditing. There are several high-quality RCTs providing evidence to support current practices in hepatopancreatobiliary (HPB) oncology. Evidence from big data bridges the gap in several instances where RCTs are lacking. In this article, we review the evidence from RCTs and big data in HPB oncology identify the existing lacunae, and discuss the future directions of research in HPB oncology.
Collapse
Affiliation(s)
- Shailesh V Shrikhande
- Gastrointestinal and Hepato-Pancreato-Biliary Service, Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Aditya R Kunte
- Gastrointestinal and Hepato-Pancreato-Biliary Service, Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit N Chopde
- Gastrointestinal and Hepato-Pancreato-Biliary Service, Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vikram A Chaudhari
- Gastrointestinal and Hepato-Pancreato-Biliary Service, Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Manish S Bhandare
- Gastrointestinal and Hepato-Pancreato-Biliary Service, Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
64
|
Okada K, Kimura K, Yamashita Y, Shibuya K, Matsumoto I, Satoi S, Yoshida K, Kodera Y, Akahori T, Hirono S, Eguchi H, Asakuma M, Tani M, Hatano E, Ikoma H, Ohira G, Hayashi H, Wan K, Shimokawa T, Kawai M, Yamaue H. Efficacy and safety of neoadjuvant nab-paclitaxel plus gemcitabine therapy in patients with borderline resectable pancreatic cancer: A multicenter single-arm phase II study (NAC-GA trial). Ann Gastroenterol Surg 2023; 7:997-1008. [PMID: 37927936 PMCID: PMC10623952 DOI: 10.1002/ags3.12712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 11/07/2023] Open
Abstract
Background Nab-paclitaxel plus gemcitabine is a standard treatment for metastatic/locally advanced pancreatic cancer. The effectiveness of neoadjuvant therapy with nab-paclitaxel plus gemcitabine (GnP-NAT) in patients with borderline resectable pancreatic cancer (BRPC) remains unclear. Patients and Methods This single-arm phase II trial included 61 patients with BRPC that were treated with two cycles of GnP-NAT, (nab-paclitaxel 125 mg/m2 and gemcitabine 1000 mg/m2), on days 1, 8, and 15 over a 4-week period, which comprised one cycle. The primary endpoint was overall survival time. In the absence of disease progression, patients underwent planned pancreatectomy. Results Median overall survival, the primary endpoint, was 25.2 months, and the median recurrence-free survival was 12.3 months. The overall rate of grade 3/4 events was 73.8%. One patient, who had a history of radiation therapy for past esophageal cancer, died from exacerbation via pneumonia. The overall resection rate was 73.8% (n = 45), and the R0 resection rate was 63.9% (n = 39). Overall, postoperative complications were found in 19 patients (42%) with 24 events, and nine patients (20%) with nine events ≥ grade IIIa, based on Dindo's classification. Conclusions This protocol treatment is thought to be a feasible, safe, and promising treatment regimen, but we caution against its use in patients with a history of interstitial lung disease and/or prior pulmonary irradiation. The survival data from this study suggest the need for further investigations of GnP-NAT efficacy in patients with BRPC, as well as prospective evaluation of adverse events. Clinical Trial Registration UMIN Clinical Trials Registry, UMIN000024154 and ClinicalTrials.gov, NCT02926183.
Collapse
Affiliation(s)
- Ken‐ichi Okada
- Second Department of SurgeryWakayama Medical UniversityWakayamaJapan
| | - Kenjiro Kimura
- Department of Hepato‐Biliary‐Pancreatic SurgeryOsaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Yo‐Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kazuto Shibuya
- Department of Surgery and Science, Faculty of Medicine, Academic AssemblyUniversity of ToyamaToyamaJapan
| | - Ippei Matsumoto
- Department of SurgeryKindai University Faculty of MedicineOsaka‐SayamaJapan
| | - Sohei Satoi
- Department of SurgeryKansai Medical UniversityHirakataJapan
| | - Kazuhiro Yoshida
- Department of Surgical OncologyGifu University Graduate School of MedicineGifuJapan
| | - Yasuhiro Kodera
- Department of Gastroenterological SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Seiko Hirono
- Second Department of SurgeryWakayama Medical UniversityWakayamaJapan
- Division of Hepato‐Biliary‐Pancreatic Surgery, Department of Gastroenterological SurgeryHyogo Medical UniversityNishinomiyaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversitySuitaJapan
| | - Mitsuhiro Asakuma
- Department of General and Gastroenterological SurgeryOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Masaji Tani
- Department of SurgeryShiga University of Medical ScienceŌtsuJapan
| | - Etsuro Hatano
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Go Ohira
- Department of Hepato‐Biliary‐Pancreatic SurgeryOsaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Ke Wan
- Clinical Study Support CenterWakayama Medical UniversityWakayamaJapan
| | - Toshio Shimokawa
- Clinical Study Support CenterWakayama Medical UniversityWakayamaJapan
| | - Manabu Kawai
- Second Department of SurgeryWakayama Medical UniversityWakayamaJapan
| | - Hiroki Yamaue
- Department of Cancer ImmunologyWakayama Medical UniversityWakayamaJapan
| | | |
Collapse
|
65
|
Yu W, Li M, Xia J. Identification of immunogenic cell death‑related prognostic signatures in pancreatic cancer. Oncol Lett 2023; 26:473. [PMID: 37809045 PMCID: PMC10551861 DOI: 10.3892/ol.2023.14061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/22/2023] [Indexed: 10/10/2023] Open
Abstract
Targeting immunogenic cell death (ICD) may enable the response of pancreatic cancer to immune checkpoint inhibitors (ICIs). The aim of the present study was to elucidate the role of ICD-related genes in pancreatic cancer. Utilizing the k-means method, consensus clustering was employed to effectively group patients with pancreatic cancer. Subsequently, a set of differentially expressed genes was identified between the two subtypes related to ICD, facilitating the execution of a comprehensive enrichment analysis. Furthermore, the construction of an ICD-related prognostic signature (IRPS) was accomplished through LASSO Cox regression, thereby enabling the assessment of responses to both chemotherapy and immunotherapy. In addition, the biological functionality of 5'-nucleotidase ecto (NT5E) was elucidated through experimental investigations. Patients characterized as the ICD high subtype experienced a comparatively shorter overall survival. This subtype exhibited a noteworthy correlation with HLA families and immune checkpoint molecules, underscoring its immunological significance. Subsequently, patients with elevated IRPS risk scores displayed resistance towards immunotherapy interventions. Of note, synergistic downregulation of NT5E in combination with Gemcitabine was observed to significantly induce tumor cell apoptosis, emphasizing its potential therapeutic value. Leveraging ICD-related genes, a novel classification system was meticulously devised to comprehensively evaluate both the clinical outcomes and therapeutic responses of patients diagnosed with pancreatic cancer.
Collapse
Affiliation(s)
- Wenjing Yu
- Department of Laboratory Medicine, The 13th People's Hospital of Chongqing, Chongqing 400053, P.R. China
| | - Mei Li
- Department of Laboratory Medicine, The 13th People's Hospital of Chongqing, Chongqing 400053, P.R. China
| | - Jing Xia
- Department of Laboratory Medicine, The 13th People's Hospital of Chongqing, Chongqing 400053, P.R. China
| |
Collapse
|
66
|
Alfano MS, Garnier J, Palen A, Ewald J, Piana G, Poizat F, Mitry E, Delpero JR, Turrini O. Peak Risk of Recurrence Occurs during the First Two Years after a Pancreatectomy in Patients Receiving Neoadjuvant FOLFIRINOX. Cancers (Basel) 2023; 15:5151. [PMID: 37958326 PMCID: PMC10649429 DOI: 10.3390/cancers15215151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/07/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
No codified/systematic surveillance program exists for borderline/locally advanced pancreatic ductal carcinoma treated with neoadjuvant FOLFIRINOX and a secondary resection. This study aimed to determine the trend of recurrence in patients who were managed using such a treatment strategy. From 2010, 101 patients received FOLFIRINOX and underwent a pancreatectomy, in a minimum follow-up of 5 years. Seventy-one patients (70%, R group) were diagnosed with recurrence after a median follow-up of 11 months postsurgery. In the multivariable analysis, patients in the R-group had a higher rate of weight loss (p = 0.018), higher carbohydrate antigen (CA 19-9) serum levels at diagnosis (p = 0.012), T3/T4 stage (p = 0.017), and positive lymph nodes (p < 0.01) compared to patients who did not experience recurrence. The risk of recurrence in patients with T1/T2 N0 R0 was the lowest (19%), and all recurrences occurred during the first two postoperative years. The peak risk of recurrence for the entire population was observed during the first two postoperative years. The probability of survival decreased until the second year and rebounded to 100% permanently, after the ninth postoperative year. Close monitoring is needed at reduced intervals during the first 2 years following a pancreatectomy and should be extended to later than 5 years for those with unfavorable pathological results.
Collapse
Affiliation(s)
- Marie-Sophie Alfano
- Department of Surgical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France; (M.-S.A.)
| | - Jonathan Garnier
- Department of Surgical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France; (M.-S.A.)
| | - Anaïs Palen
- Department of Surgical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France; (M.-S.A.)
| | - Jacques Ewald
- Department of Surgical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France; (M.-S.A.)
| | - Gilles Piana
- Department of Radiology, Institut Paoli-Calmettes, 13009 Marseille, France
| | - Flora Poizat
- Department of Pathology, Institut Paoli-Calmettes, 13009 Marseille, France
| | - Emmanuel Mitry
- Department of Oncology, Institut Paoli-Calmettes, 13009 Marseille, France
| | - Jean-Robert Delpero
- Department of Surgical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France; (M.-S.A.)
- Faculté de Médecine, Aix-Marseille University, 13005 Marseille, France
| | - Olivier Turrini
- Department of Surgical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France; (M.-S.A.)
- Faculté de Médecine, Aix-Marseille University, 13005 Marseille, France
| |
Collapse
|
67
|
Dong X, Wang K, Yang H, Cheng R, Li Y, Hou Y, Chang J, Yuan L. The Nomogram predicting the overall survival of patients with pancreatic cancer treated with radiotherapy: a study based on the SEER database and a Chinese cohort. Front Endocrinol (Lausanne) 2023; 14:1266318. [PMID: 37955009 PMCID: PMC10634587 DOI: 10.3389/fendo.2023.1266318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023] Open
Abstract
Objective Patients with pancreatic cancer (PC) have a poor prognosis. Radiotherapy (RT) is a standard palliative treatment in clinical practice, and there is no effective clinical prediction model to predict the prognosis of PC patients receiving radiotherapy. This study aimed to analyze PC's clinical characteristics, find the factors affecting PC patients' prognosis, and construct a visual Nomogram to predict overall survival (OS). Methods SEER*Stat software was used to collect clinical data from the Surveillance, Epidemiology, and End Results (SEER) database of 3570 patients treated with RT. At the same time, the relevant clinical data of 115 patients were collected from the Affiliated Cancer Hospital of Zhengzhou University. The SEER database data were randomly divided into the training and internal validation cohorts in a 7:3 ratio, with all patients at The Affiliated Cancer Hospital of Zhengzhou University as the external validation cohort. The lasso regression was used to screen the relevant variables. All non-zero variables were included in the multivariate analysis. Multivariate Cox proportional risk regression analysis was used to determine the independent prognostic factors. The Kaplan-Meier(K-M) method was used to plot the survival curves for different treatments (surgery, RT, chemotherapy, and combination therapy) and calculate the median OS. The Nomogram was constructed to predict the survival rates at 1, 3, and 5 years, and the time-dependent receiver operating characteristic curves (ROC) were plotted with the calculated curves. Calculate the area under the curve (AUC), the Bootstrap method was used to plot the calibration curve, and the clinical efficacy of the prediction model was evaluated using decision curve analysis (DCA). Results The median OS was 25.0, 18.0, 11.0, and 4.0 months in the surgery combined with chemoradiotherapy (SCRT), surgery combined with radiotherapy, chemoradiotherapy (CRT), and RT alone cohorts, respectively. Multivariate Cox regression analysis showed that age, N stage, M stage, chemotherapy, surgery, lymph node surgery, and Grade were independent prognostic factors for patients. Nomogram models were constructed to predict patients' OS. 1-, 3-, and 5-year Time-dependent ROC curves were plotted, and AUC values were calculated. The results suggested that the AUCs were 0.77, 0.79, and 0.79 for the training cohort, 0.79, 0.82, and 0.81 for the internal validation cohort, and 0.73, 0.93, and 0.88 for the external validation cohort. The calibration curves Show that the model prediction probability is in high agreement with the actual observation probability, and the DCA curve shows a high net return. Conclusion SCRT significantly improves the OS of PC patients. We developed and validated a Nomogram to predict the OS of PC patients receiving RT.
Collapse
Affiliation(s)
- Xiaotao Dong
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kunlun Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Yang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruilan Cheng
- Department of Hematology and Oncology, Shenzhen Children’s Hospital Affiliated to China Medical University, Shenzhen, China
| | - Yan Li
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanqi Hou
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiali Chang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ling Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
68
|
Fayad E, Altalhi SA, Abualnaja MM, Alrohaimi AH, Elsaid FG, Abu Almaaty AH, Saleem RM, Bazuhair MA, Ahmed Maghrabi AH, Beshay BY, Zaki I. Novel Acrylate-Based Derivatives: Design, Synthesis, Antiproliferative Screening, and Docking Study as Potential Combretastatin Analogues. ACS OMEGA 2023; 8:38394-38405. [PMID: 37867686 PMCID: PMC10586439 DOI: 10.1021/acsomega.3c05051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023]
Abstract
A variety of 3-(4-chlorophenyl) acrylic acids 4a,b and 3-(4-chlorophenyl)acrylate esters 5a-i were synthesized and structurally proven by spectroscopic studies such as IR, 1H NMR, and 13C NMR as well as mass spectrometry. All substances were investigated for their antiproliferative efficacy against the MDA-MB-231 cell line. Among these, acrylic acid compound 4b demonstrated the most potent cytotoxic effect with an IC50 value of 3.24 ± 0.13 μM, as compared to CA-4 (IC50 = 1.27 ± 09 μM). Additionally, acrylic acid molecule 4b displayed an inhibitory effect against β-tubulin polymerization with a percentage inhibition of 80.07%. Furthermore, compound 4b was found to produce considerable cell cycle arrest at the G2/M stage and cellular death, as demonstrated by FACS analysis. In addition, the in vivo antitumor screening of the sodium salt of acrylic acid 4b was carried out, and the results have shown that the tested molecule showed a significant decrease in viable EAC count and EAC volume, accompanied by a considerable increase in the life span prolongation, if compared to the positive control group. Furthermore, molecular modeling studies were performed to understand how the highly efficient chemicals 4b and 5e interact with the colchicine-binding region on tubulin. This work aims to shed light on the reasons behind their exceptional cytotoxicity and their better capacity to inhibit tubulin in comparison to CA-4.
Collapse
Affiliation(s)
- Eman Fayad
- Department
of Biotechnology, College of Sciences, Taif
University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Sarah Awwadh Altalhi
- Department
of Biotechnology, College of Sciences, Taif
University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Matokah M. Abualnaja
- Department
of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah
Al Mukarrama 24230, Saudi Arabia
| | - Abdulmohsen H. Alrohaimi
- Department
of Pharmacy Practice, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Fahmy G. Elsaid
- Biology
Department, College of Science, King Khalid
University, P.O.Box 960, Asir, Abha 61421, Saudi Arabia
| | - Ali H. Abu Almaaty
- Zoology
Department, Faculty of Science Port Said
University, Port Said 42526, Egypt
| | - Rasha Mohammed Saleem
- Department
of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha 65431, Saudi Arabia
| | - Mohammed A. Bazuhair
- Department
of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ali Hassan Ahmed Maghrabi
- Department
of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Botros Y. Beshay
- Pharmaceutical
Sciences (Pharmaceutical Chemistry) Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime
Transport, Alexandria 21913, Egypt
| | - Islam Zaki
- Pharmaceutical
Organic Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
| |
Collapse
|
69
|
Pedrazzoli S. Currently Debated Topics on Surgical Treatment of Pancreatic Ductal Adenocarcinoma: A Narrative Review on Surgical Treatment of Borderline Resectable, Locally Advanced, and Synchronous or Metachronous Oligometastatic Tumor. J Clin Med 2023; 12:6461. [PMID: 37892599 PMCID: PMC10607532 DOI: 10.3390/jcm12206461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Previously considered inoperable patients (borderline resectable, locally advanced, synchronous oligometastatic or metachronous pancreatic adenocarcinoma (PDAC)) are starting to become resectable thanks to advances in chemo/radiotherapy and the reduction in operative mortality. METHODS This narrative review presents a chosen literature selection, giving a picture of the current state of treatment of these patients. RESULTS Neoadjuvant therapy (NAT) is generally recognized as the treatment of choice before surgery. However, despite the increased efficacy, the best pathological response is still limited to 10.9-27.9% of patients. There are still limited data on the selection of possible NAT responders and how to diagnose non-responders early. Multidetector computed tomography has high sensitivity and low specificity in evaluating resectability after NAT, limiting the resection rate of resectable patients. Ca 19-9 and Positron emission tomography are giving promising results. The prediction of early recurrence after a radical resection of synchronous or metachronous metastatic PDAC, thus identifying patients with poor prognosis and saving them from a resection of little benefit, is still ongoing, although some promising data are available. CONCLUSION In conclusion, high-level evidence demonstrating the benefit of the surgical treatment of such patients is still lacking and should not be performed outside of high-volume centers with interdisciplinary teams of surgeons and oncologists.
Collapse
|
70
|
Pinelli D, Micalef A, Merelli B, Trezzi R, Amaduzzi A, Agnesi S, Guizzetti M, Camagni S, Fedele V, Colledan M. Pancreatic ductal adenocarcinoma complete regression after preoperative chemotherapy: Surgical results in a small series. Cancer Treat Res Commun 2023; 37:100770. [PMID: 37837717 DOI: 10.1016/j.ctarc.2023.100770] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/30/2023] [Accepted: 10/07/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) becomes a systemic disease from an early stage. Complete surgical resection remains the only validated and potentially curative treatment; disappointingly only 20% of patients present with a resectable tumour. Although a complete pathological regression (pCR) after the preoperative chemotherapy could intuitively lead to better outcomes and prolonged survival some reports highlighted significant rates of recurrence. CASES PRESENTATION We describe three cases of pCR following preoperative chemotherapy for PDAC. The first two cases received neoadjuvant mFOLFIRINOX and PAX-G scheme for borderline resectable PDAC. Recurrence appeared 9 and 12 months after surgery. Although both patients started adjuvant therapy straight after the diagnosis of recurrence, the disease rapidly progressed and led them to death 12 and 15 months after surgery. The third case was characterized by germline BRCA2 mutation. The patient presented with PDAC of the body, intrapancreatic biliary stenosis and suspected peritoneal metastasis. One year later, after first and second-line chemotherapy, she underwent explorative laparoscopy and total spleno-pancreatectomy without evidence of viable tumour cells in the surgical specimen. At six months she is recurrence-free. CONCLUSIONS Very few reports describe a complete pathological response following preoperative chemotherapy in pancreatic cancer. We observed three cases in the last three years with disappointing oncological results. Further investigations are needed to predict PDAC prognosis in pCR after chemotherapy.
Collapse
Affiliation(s)
- Domenico Pinelli
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy
| | - Andrea Micalef
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy; Università degli Studi di Milano, Milano, Italy.
| | - Barbara Merelli
- Unit of Medical Oncology, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy
| | - Rosangela Trezzi
- Unit of Pathology, ASST-Papa Giovanni XXIII, Piazza OMS 1, 24127, Bergamo, Italy
| | - Annalisa Amaduzzi
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy
| | - Stefano Agnesi
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy
| | - Michela Guizzetti
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy
| | - Stefania Camagni
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy
| | - Veronica Fedele
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy; Università degli Studi di Milano, Milano, Italy
| | - Michele Colledan
- Department of Organ Failure and Transplantation, ASST-Papa Giovanni XXIII, Piazza OMS, 1, 24127, Bergamo, Italy; University of Bicocca, Milano, Italy
| |
Collapse
|
71
|
Sindayigaya R, Barat M, Tzedakis S, Dautry R, Dohan A, Belle A, Coriat R, Soyer P, Fuks D, Marchese U. Modified Appleby procedure for locally advanced pancreatic carcinoma: A primer for the radiologist. Diagn Interv Imaging 2023; 104:455-464. [PMID: 37301694 DOI: 10.1016/j.diii.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most prevalent pancreatic neoplasm accounting for more than 90% of pancreatic malignancies. Surgical resection with adequate lymphadenectomy remains the only available curative strategy for patients with PDAC. Despite improvements in both chemotherapy regimen and surgical care, body/neck PDAC still conveys a poor prognosis because of the vicinity of major vascular structures, including celiac trunk, which favors insidious disease spread at the time of diagnosis. Body/neck PDAC involving the celiac trunk is considered locally advanced PDAC in most guidelines and therefore not eligible for upfront resection. However, a more aggressive surgical approach (i.e., distal pancreatectomy with splenectomy and en-bloc celiac trunk resection [DP-CAR]) was recently proposed to offer hope for cure in selected patients with locally advanced body/neck PDAC responsive to induction therapy at the cost of higher morbidity. The so-called "modified Appleby procedure" is highly demanding and requires optimal preoperative staging as well as appropriate patient preparation for surgery (i.e., preoperative arterial embolization). Herein, we review current evidence regarding DP-CAR indications and outcomes as well as the critical role of diagnostic and interventional radiology in patient preparation before DP-CAR, and early identification and management of DP-CAR complications.
Collapse
Affiliation(s)
- Rémy Sindayigaya
- Department of Digestive, Pancreatic, Hepato-biliary and Endocrine Surgery, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014, Paris, France; Université Paris Cité, Faculté de Médecine, 75006 Paris, France.
| | - Maxime Barat
- Université Paris Cité, Faculté de Médecine, 75006 Paris, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Stylianos Tzedakis
- Department of Digestive, Pancreatic, Hepato-biliary and Endocrine Surgery, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014, Paris, France; Université Paris Cité, Faculté de Médecine, 75006 Paris, France
| | - Raphael Dautry
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Anthony Dohan
- Université Paris Cité, Faculté de Médecine, 75006 Paris, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Arthur Belle
- Department of Gastroenterology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Romain Coriat
- Université Paris Cité, Faculté de Médecine, 75006 Paris, France; Department of Gastroenterology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Philippe Soyer
- Université Paris Cité, Faculté de Médecine, 75006 Paris, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - David Fuks
- Department of Digestive, Pancreatic, Hepato-biliary and Endocrine Surgery, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014, Paris, France; Université Paris Cité, Faculté de Médecine, 75006 Paris, France
| | - Ugo Marchese
- Department of Digestive, Pancreatic, Hepato-biliary and Endocrine Surgery, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014, Paris, France; Université Paris Cité, Faculté de Médecine, 75006 Paris, France
| |
Collapse
|
72
|
Hayasaki A, Mizuno S, Nagata M, Kaluba B, Maeda K, Shinkai T, Ito T, Gyoten K, Fujii T, Iizawa Y, Tanemura A, Murata Y, Kuriyama N, Isaji S, Kishiwada M. Extrapancreatic extension is a better adverse prognostic factor than tumor size in patients with localized pancreatic ductal adenocarcinoma treated with chemoradiotherapy - comparison of T category between the American Joint Committee on Cancer and Japan Pancreas Society. HPB (Oxford) 2023; 25:1268-1277. [PMID: 37419780 DOI: 10.1016/j.hpb.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/17/2023] [Accepted: 06/22/2023] [Indexed: 07/09/2023]
Abstract
BACKGROUND T category classification for pancreatic ductal adenocarcinoma (PDAC) in the Classification of Pancreatic Cancer by the Japan Pancreas Society (JPS) is quite different from that of the American Joint Committee on Cancer (AJCC). The JPS classification focuses on extrapancreatic extension, while the AJCC focuses mainly on tumor size. This study aimed at identifying prognostic factors in PDAC patients undergoing chemoradiotherapy (CRT) by comparing the differences of T categories in these two classifications. METHODS This retrospective study involved 344 PDAC patients who underwent CRT from 2005 to 2019 and their T-category variables were re-evaluated on computed tomography (CT) images. Disease-specific survival (DSS) was compared based on the JPS and AJCC T categories, while multivariate analysis was performed to identify prognostic factors. RESULTS Based on the AJCC, 5-year DSS of T3 was better than those of T1 and T2 (57.1% vs. 47.7% and 37.4%). In multivariate analysis, performance status, CEA, the involvement of superior mesenteric vein and superior mesenteric artery, the JPS stage before CRT, and regimen of chemotherapy were identified as independent prognostic factors. CONCLUSIONS In localized PDAC patients treated with chemoradiotherapy, extrapancreatic extension, as while as biological, conditional and therapeutic factors, is a better prognostic factor than tumor size.
Collapse
Affiliation(s)
- Aoi Hayasaki
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan.
| | - Shugo Mizuno
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | | | - Benson Kaluba
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Koki Maeda
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Toru Shinkai
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Takahiro Ito
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Kazuyuki Gyoten
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Takehiro Fujii
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Yusuke Iizawa
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Akihiro Tanemura
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Yasuhiro Murata
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Naohisa Kuriyama
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Shuji Isaji
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| | - Masashi Kishiwada
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Mie University, Tsu, Mie, Japan
| |
Collapse
|
73
|
Wu VS, Elshami M, Stitzel HJ, Lee JJ, Hue JJ, Kyasaram RK, Hardacre JM, Ammori JB, Winter JM, Selfridge JE, Mohamed A, Chakrabarti S, Bajor D, Mahipal A, Ocuin LM. Why the Treatment Sequence Matters: Interplay Between Chemotherapy Cycles Received, Cumulative Dose Intensity, and Survival in Resected Early-stage Pancreas Cancer. Ann Surg 2023; 278:e677-e684. [PMID: 37071769 DOI: 10.1097/sla.0000000000005830] [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: 04/20/2023]
Abstract
OBJECTIVE To define the optimal threshold of perioperative chemotherapy completion and relative dose intensity (RDI) for patients with resected pancreatic ductal adenocarcinoma (PDAC). BACKGROUND Many patients who undergo pancreatectomy for PDAC fail to initiate or complete recommended perioperative chemotherapy. The association between the amount of perioperative chemotherapy received and overall survival (OS) is not well-defined. METHODS Single-institution analysis of 225 patients who underwent pancreatectomy for stage I/II PDAC (2010-2021). Associations between OS, chemotherapy cycles completed, and RDI were analyzed. RESULTS Regardless of treatment sequence, completion of ≥67% of recommended cycles was associated with improved OS compared with no chemotherapy [median OS: 34.5 vs 18.1 months; hazard ratio (HR): 0.43; 95% CI: 0.25-0.74] and <67% of cycles (median OS: 17.9 months; HR: 0.39; 95% CI: 0.24-0.64). A near-linear relationship existed between cycles completed and the RDI received (β = 0.82). A median RDI of 56% corresponded to the completion of 67% of cycles. Receipt of ≥56% RDI was associated with improved OS compared with no chemotherapy (median OS: 35.5 vs 18.1 months; HR: 0.44; 95% CI: 0.23-0.84) and <56% RDI (median OS: 27.2 months; HR: 0.44; 95% CI: 0.20-0.96). Neoadjuvant chemotherapy is associated with increased odds of receiving ≥67% of recommended cycles (odds ratio: 2.94; 95% CI: 1.45-6.26) and ≥56% RDI (odds ratio: 4.47; 95% CI: 1.72-12.50). CONCLUSIONS Patients with PDAC who received ≥67% of recommended chemotherapy cycles or ≥56% cumulative RDI had improved OS. Neoadjuvant therapy was associated with increased odds of receiving ≥67% of cycles and ≥56% cumulative RDI and should be considered in all patients with resectable PDAC.
Collapse
Affiliation(s)
- Victoria S Wu
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Mohamedraed Elshami
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Henry J Stitzel
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Jonathan J Lee
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Jonathan J Hue
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Ravi K Kyasaram
- Department of Cancer Informatics, University Hospitals Cleveland Medical Center/Seidman Cancer Center, Cleveland, OH
| | - Jeffrey M Hardacre
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - John B Ammori
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Jordan M Winter
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Jennifer Eva Selfridge
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Amr Mohamed
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Sakti Chakrabarti
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - David Bajor
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Amit Mahipal
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Lee M Ocuin
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH
| |
Collapse
|
74
|
de Carvalho LFA, Gryspeerdt F, Rashidian N, Van Hove K, Maertens L, Ribeiro S, Hoorens A, Berrevoet F. Predictive factors for survival in borderline resectable and locally advanced pancreatic cancer: are these really two different entities? BMC Surg 2023; 23:296. [PMID: 37775737 PMCID: PMC10541717 DOI: 10.1186/s12893-023-02200-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND The treatment of borderline resectable (BR) and locally advanced (LA) pancreatic ductal adenocarcinoma (PDAC) has evolved with a wider application of neoadjuvant chemotherapy (NACHT). The aim of this study was to identify predictive factors for survival in BR and LA PDAC. METHODS Clinicopathologic data of patients with BR and LA PDAC who underwent surgical exploration between January 2011 and June 2021 were retrospectively collected. Survival from the date of surgery was estimated using the Kaplan-Meier method. Simple and multiple Cox proportional hazards models were fitted to identify factors associated with survival. Surgical resection was analyzed in combination with the involvement of lymph nodes as this last was only known after a formal resection. RESULTS Ninety patients were surgically explored (BR: 45, LA: 45), of which 51 (57%) were resected (BR: 31, LA: 20). NACHT was administered to 43 patients with FOLFIRINOX being the most frequent regimen applied (33/43, 77%). Major complications (Clavien-Dindo grade III and IV) occurred in 7.8% of patients and 90-day mortality rate was 3.3%. The median overall survival since surgery was 16 months (95% CI 12-20) in the group which underwent surgical resection and 10 months (95% CI 7-13) in the group with an unresectable tumor (p=0.001). Cox proportional hazards models showed significantly lower mortality hazard for surgical resection compared to no surgical resection, even after adjusting for National Comprehensive Cancer Network (NCCN) classification and administration of NACHT [surgical resection with involved lymph nodes vs no surgical resection (cHR 0.49; 95% CI 0.29-0.82; p=0.007)]. There was no significant difference in survival between patients with BR and LA disease (cHR= 1.01; 95% CI 0.63-1.62; p=0.98). CONCLUSIONS Surgical resection is the only predictor of survival in patients with BR and LA PDAC, regardless of their initial classification as BR or LA. Our results suggest that surgery should not be denied to patients with LA PDAC a priori. Prospective studies including patients from the moment of diagnosis are required to identify biologic and molecular markers which may allow a better selection of patients who will benefit from surgery.
Collapse
Affiliation(s)
- Luís Filipe Abreu de Carvalho
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| | - Filip Gryspeerdt
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Niki Rashidian
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Kobe Van Hove
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Lambertine Maertens
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Suzane Ribeiro
- Department of Gastroenterology, Division of Digestive Oncology, Ghent University Hospital, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Frederik Berrevoet
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| |
Collapse
|
75
|
Liang Y, Cui J, Ding F, Zou Y, Guo H, Man Q, Chang S, Gao S, Hao J. A new staging system for postoperative prognostication in pancreatic ductal adenocarcinoma. iScience 2023; 26:107589. [PMID: 37664604 PMCID: PMC10469961 DOI: 10.1016/j.isci.2023.107589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/09/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
The current TNM staging system for pancreatic ductal adenocarcinoma (PDAC) has revised the definitions of T and N categories as well as stage groups. However, studies validating these modifications have yielded inconsistent results. The existing TNM staging system in prognostic prediction remains unsatisfactory. The prognosis of PDAC is closely associated with pathological and biological factors. Herein, we propose a new staging system incorporating distant metastasis, postoperative serum levels of CA19-9 and CEA, tumor size, lymph node metastasis, lymphovascular involvement, and perineural invasion to enhance the accuracy of prognosis assessment. The proposed staging system exhibited a strong correlation with both overall survival and recurrence-free survival, effectively stratifying survival into five distinct tiers. Additionally, it had favorable discrimination and calibration. Thus, the proposed staging system demonstrates superior prognostic performance compared to the TNM staging system, and can serve as a valuable complementary tool to address the limitations of TNM staging in prognostication.
Collapse
Affiliation(s)
- Yuexiang Liang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center For Cancer, Tianjin 30060, China
- Department of Gastrointestinal Oncology Surgery, Center of Cancer Prevention and Therapy, the First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Jingli Cui
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center For Cancer, Tianjin 30060, China
- Department of General Surgery, Weifang People’s Hospital, Weifang 261044, China
| | - Fanghui Ding
- Department of General Surgery, the First Hospital of Lanzhou University, Lanzhou 730013, China
| | - Yiping Zou
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center For Cancer, Tianjin 30060, China
| | - Hanhan Guo
- Department of Gastrointestinal Oncology Surgery, Center of Cancer Prevention and Therapy, the First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Quan Man
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center For Cancer, Tianjin 30060, China
| | - Shaofei Chang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center For Cancer, Tianjin 30060, China
- Department of Gastrointestinal Pancreatic Surgery, Shanxi Provincial People’s Hospital, Taiyuan 030012, China
| | - Song Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center For Cancer, Tianjin 30060, China
| | - Jihui Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center For Cancer, Tianjin 30060, China
| |
Collapse
|
76
|
Nicolais LM, Caron M, Verdini N, Fitzgerald TL. Pathologic Tumor Regression is Associated With Improved Survival in Pancreatic Cancer. Am Surg 2023; 89:3778-3783. [PMID: 37227766 DOI: 10.1177/00031348231175120] [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: 05/26/2023]
Abstract
OBJECTIVES The advent of effective chemotherapy regimens has increased the use of neoadjuvant multiagent chemotherapy in pancreatic cancer. However, the effect of tumor downstaging with neoadjuvant treatment on survival is unclear. METHODS Retrospective study included all resected patients with pancreatic adenocarcinoma who underwent neoadjuvant chemotherapy with FOLFIRINOX or gemcitabine/Abraxane. Downstaging was quantified using (1) difference between presenting AJCC clinical and final pathologic stage and (2) College of American Pathologists (CAP) Tumor Regression Grading Schema. RESULTS Eighty-seven patients met inclusion criteria. FOLFIRINOX was the most common regimen, 63.2% vs 21.8%. Change in regimen occurred in 15% of patients. Downstaging based on a difference in AJCC stage group occurred in only 4.6%. In contrast, 45.2% were classified as downstaged by the CAP Tumor Regression of 0-2. Downstaging was similar for FOLFIRINOX gemcitabine/Abraxane (64.7 vs 53.6, P = .12) using the CAP criteria. On univariate analysis, treatment regimen (gemcitabine/Abraxane vs FOLFIRINOX, median survival 27 vs 29 mo; HR 1.57, P = .2) had similar survival. Downstaging by the AJCC stage was not associated with improved survival (HR 1.51, P = .4). However, there was a survival benefit for those downstaged by the CAP Tumor Regression Grading Schema, the median survival of 41 mo vs 25 mo; HR 3.05, P = .009. Improved survival 3.32 (1.35-8.16), P = .009) was maintained on multivariate analysis. CONCLUSION Survival is significantly improved in those downstaged, as assessed by the CAP Tumor Regression Schema. Downstaging is an important prognostic variable that can help with joint decision making for clinicians and patients.
Collapse
Affiliation(s)
- Laura M Nicolais
- Division of Surgical Oncology, Tufts University School of Medicine-Maine Medical Center, Portland, ME, USA
- Tufts University Clinical and Translational Science Graduate Program, Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Molly Caron
- Division of Surgical Oncology, Tufts University School of Medicine-Maine Medical Center, Portland, ME, USA
| | - Nicholas Verdini
- Tufts University Clinical and Translational Science Graduate Program, Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Timothy L Fitzgerald
- Division of Surgical Oncology, Tufts University School of Medicine-Maine Medical Center, Portland, ME, USA
| |
Collapse
|
77
|
Umezawa R, Mizuma M, Nakagawa K, Yamamoto T, Takahashi N, Suzuki Y, Kishida K, Omata S, Unno M, Jingu K. Clinical impact of multimodal treatment including chemoradiotherapy, conversion surgery and postoperative chemotherapy for borderline resectable and unresectable locally advanced pancreatic cancer without disease progression after gemcitabine plus nab-paclitaxel. Pancreatology 2023; 23:650-656. [PMID: 37453848 DOI: 10.1016/j.pan.2023.07.002] [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: 12/13/2022] [Revised: 05/31/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND The purpose of this study was to investigate treatment outcomes of chemoradiotherapy (CRT) using S-1 with or without conversion surgery after gemcitabine plus nab-paclitaxel (GnP) for borderline resectable (BR) and unresectable locally advanced (UR-LA) pancreatic cancer. METHODS From 2016 to 2020, patients without disease progression after GnP for BR or UR-LA pancreatic cancer underwent CRT with S-1. If distant metastasis was not detected after CRT, conversion surgery and oral administration of S-1 as postoperative adjuvant chemotherapy for at least 6 months was performed. RESULTS Forty patients were included in the present study. The median number of cycles of GnP was 6. Surgery was performed after CRT in 25 patients. The median progression-free survival (PFS) and overall survival (OS) periods from the start of radiotherapy were 24.6 and 27.4 months, respectively. The OS periods from the start of radiotherapy in patients who underwent conversion surgery and those who did not undergo conversion surgery were 41.3 and 16.8 months, respectively. The PFS periods from the start of radiotherapy in patients who underwent surgery and those who did not undergo surgery were 28.3 and 8.6 months, respectively. Patients who were able to receive S-1 after conversion surgery for more than 6 months had better OS than those who were not (p = 0.039), although there was no significant difference of PFS (p = 0.365). CONCLUSIONS In BR/UR-LA pancreatic cancer without disease progression after GnP, multimodal treatment including CRT, conversion surgery and the scheduled postoperative chemotherapy may be effective.
Collapse
Affiliation(s)
- Rei Umezawa
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Masamichi Mizuma
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takaya Yamamoto
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriyoshi Takahashi
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Suzuki
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keita Kishida
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - So Omata
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keiichi Jingu
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
78
|
Nagai M, Nakamura K, Terai T, Kohara Y, Yasuda S, Matsuo Y, Doi S, Sakata T, Sho M. Significance of multiple tumor markers measurements in conversion surgery for unresectable locally advanced pancreatic cancer. Pancreatology 2023; 23:721-728. [PMID: 37328387 DOI: 10.1016/j.pan.2023.06.001] [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: 03/31/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND This study aimed to evaluate the significance of multiple tumor markers (TMs) measurements in determining the indications for conversion surgery (CS) in the management of unresectable locally advanced pancreatic cancer (UR-LAPC). METHODS A total of 103 patients with UR-LAPC, treated between 2008 and June 2021, were enrolled in this study. Three TMs, including carbohydrate antigen 19-9 (CA19-9), carcinoembryonic antigen (CEA), and Duke pancreatic monoclonal antigen type 2 (DUPAN-2), were measured. RESULTS Twenty-five patients (24%) underwent CS. The median preoperative treatment period was 9.5 months. The median survival time (MST) from the initial treatment for patients with CS was significantly longer than that for patients without surgery (34.6 vs. 18.9 months, P < 0.001). The number of elevated TMs before CS was one in five patients and two in five patients, while 15 patients had normal levels of all three TMs. Notably, the MST from the initial treatment for patients with all three preoperative normal TMs levels was favorable for 70.5 months. In contrast, patients with one or two preoperatively elevated TMs levels had a significantly worse prognosis (25.4 and 21.0 months, respectively, P < 0.001). Furthermore, the relapse-free survival of patients with three preoperative normal TMs levels was significantly longer than those with one or two elevated TMs levels (21.9 vs. 11.3 or 3.0 months, respectively, P < 0.001). Non-normal values of all TMs before CS were identified as independent poor prognostic factors. CONCLUSIONS Simultaneous measurement and assessment of the three TMs levels may help determine the surgical indications for UR-LAPC after systemic anticancer treatment.
Collapse
Affiliation(s)
- Minako Nagai
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Kota Nakamura
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Taichi Terai
- Department of Surgery, Nara Medical University, Nara, Japan
| | | | - Satoshi Yasuda
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Yasuko Matsuo
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Shunsuke Doi
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Takeshi Sakata
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Nara, Japan.
| |
Collapse
|
79
|
Sinnamon AJ, Mehta R, Saeed S, Lauwers GY, Palm RF, Frakes JM, Hoffe SE, Baldonado JJ, Fontaine JP, Pimiento JM. Induction Fluorouracil-Based Chemotherapy and PET-Adapted Consolidation Chemoradiation with Esophagectomy for High-Risk Gastroesophageal Adenocarcinoma. Cancers (Basel) 2023; 15:4375. [PMID: 37686650 PMCID: PMC10486661 DOI: 10.3390/cancers15174375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Background: Neoadjuvant chemoradiation with esophagectomy is standard management for locally advanced esophageal adenocarcinoma. Induction chemotherapy with a tailored approach to chemoradiation based on metabolic response to therapy on PET was explored as an alternative strategy in the CALGB 80803 trial. We sought to describe real-world institutional experience implementing this approach outside of a clinical trial. Methods: Patients who were treated with induction fluorouracil-leucovorin-oxaliplatin (FOLFOX) or fluorouracil-leucovorin-oxaliplatin-docetaxel (FLOT) with tailored chemoradiation based on PET response and subsequent esophagectomy were identified from a prospectively maintained database. Primary outcomes were pathologic complete response (pCR) and overall survival (OS) following completion of all therapy. Results: There were 35 patients who completed induction chemotherapy, chemoradiation, and esophagectomy. Thirty-three completed restaging PET following induction chemotherapy with metabolic response seen in 76% (n = 25/33). The pCR rate was 31% (n = 11/35) and the ypN0 rate was 71% (n = 25/35). Among the patients who demonstrated metabolic response to induction FOLFOX/FLOT and subsequently continued fluorouracil-based chemoradiation, the pCR rate was 39% (n = 9/23). The rate of pathologically negative lymph nodes in this group was high (n = 19/23, 83%) with 100% R0 resection rate (n = 23/23). With the median follow-up of 43 months, the median OS was not reached for this group and was significantly longer than the OS for the remainder of the cohort (p = 0.027, p = 0.046 adjusted for clinical stage). Conclusions: Induction FOLFOX/FLOT chemotherapy with evaluation of sensitivity via metabolic response and tailored chemoradiation seems to lead to high pCR and ypN0 rates in high-risk patients with adenocarcinoma of the esophagus and GE junction. This approach in clinical practice seems to recapitulate encouraging results in clinical trials.
Collapse
Affiliation(s)
- Andrew J Sinnamon
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Rutika Mehta
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Samir Saeed
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Gregory Y Lauwers
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Russell F Palm
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jessica M Frakes
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Sarah E Hoffe
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jobelle J Baldonado
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jacques P Fontaine
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jose M Pimiento
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
80
|
Oba A, Del Chiaro M, Fujii T, Okano K, Stoop TF, Wu YHA, Maekawa A, Yoshida Y, Hashimoto D, Sugawara T, Inoue Y, Tanabe M, Sho M, Sasaki T, Takahashi Y, Matsumoto I, Sasahira N, Nagakawa Y, Satoi S, Schulick RD, Yoon YS, He J, Jang JY, Wolfgang CL, Hackert T, Besselink MG, Takaori K, Takeyama Y. "Conversion surgery" for locally advanced pancreatic cancer: A position paper by the study group at the joint meeting of the International Association of Pancreatology (IAP) & Japan Pancreas Society (JPS) 2022. Pancreatology 2023; 23:712-720. [PMID: 37336669 DOI: 10.1016/j.pan.2023.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/10/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Locally advanced pancreatic cancer (LAPC), which progresses locally and surrounds major vessels, has historically been deemed unresectable. Surgery alone failed to provide curative resection and improve overall survival. With the advancements in treatment, reports have shown favorable results in LAPC after undergoing successful chemotherapy therapy or chemoradiation therapy followed by surgical resection, so-called "conversion surgery", at experienced high-volume centers. However, recognizing significant regional and institutional disparities in the management of LAPC, an international consensus meeting on conversion surgery for LAPC was held during the Joint Congress of the 26th Meeting of the International Association of Pancreatology (IAP) and the 53rd Annual Meeting of Japan Pancreas Society (JPS) in Kyoto in July 2022. During the meeting, presenters reported the current best multidisciplinary practices for LAPC, including preoperative modalities, best systemic treatment regimens and durations, procedures of conversion surgery with or without vascular resections, biomarkers, and genetic studies. It was unanimously agreed among the experts in this meeting that "cancer biology is surpassing locoregional anatomical resectability" in the era of effective multiagent treatment. The biology of pancreatic cancer has yet to be further elucidated, and we believe it is essential to improve the treatment outcomes of LAPC patients through continued efforts from each institution and more international collaboration. This article summarizes the agreement during the discussion amongst the experts in the meeting. We hope that this will serve as a foundation for future international collaboration and recommendations for future guidelines.
Collapse
Affiliation(s)
- Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Keiichi Okano
- Department of Gastroenterological Surgery, Kagawa University School of Medicine, Kagawa, Japan
| | - Thomas F Stoop
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Y H Andrew Wu
- Department Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aya Maekawa
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuta Yoshida
- Department of Surgery, Kindai University, Osaka, Japan
| | | | - Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | | | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Sohei Satoi
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA; Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Richard D Schulick
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jin He
- Department Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Germany; Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Germany
| | - Marc G Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | | |
Collapse
|
81
|
Jain AJ, Maxwell JE, Katz MHG, Snyder RA. Surgical Considerations for Neoadjuvant Therapy for Pancreatic Adenocarcinoma. Cancers (Basel) 2023; 15:4174. [PMID: 37627202 PMCID: PMC10453019 DOI: 10.3390/cancers15164174] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a challenging disease process with a 5-year survival rate of only 11%. Neoadjuvant therapy in patients with localized pancreatic cancer has multiple theoretical benefits, including improved patient selection for surgery, early delivery of systemic therapy, and assessment of response to therapy. Herein, we review key surgical considerations when selecting patients for neoadjuvant therapy and curative-intent resection. Accurate determination of resectability at diagnosis is critical and should be based on not only anatomic criteria but also biologic and clinical criteria to determine optimal treatment sequencing. Borderline resectable or locally advanced pancreatic cancer is best treated with neoadjuvant therapy and resection, including vascular resection and reconstruction when appropriate. Lastly, providing nutritional, prehabilitation, and supportive care interventions to improve patient fitness prior to surgical intervention and adequately address the adverse effects of therapy is critical.
Collapse
Affiliation(s)
| | | | | | - Rebecca A. Snyder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.J.)
| |
Collapse
|
82
|
Lintern N, Smith AM, Jayne DG, Khaled YS. Photodynamic Stromal Depletion in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:4135. [PMID: 37627163 PMCID: PMC10453210 DOI: 10.3390/cancers15164135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest solid malignancies, with a five-year survival of less than 10%. The resistance of the disease and the associated lack of therapeutic response is attributed primarily to its dense, fibrotic stroma, which acts as a barrier to drug perfusion and permits tumour survival and invasion. As clinical trials of chemotherapy (CT), radiotherapy (RT), and targeted agents have not been successful, improving the survival rate in unresectable PDAC remains an urgent clinical need. Photodynamic stromal depletion (PSD) is a recent approach that uses visible or near-infrared light to destroy the desmoplastic tissue. Preclinical evidence suggests this can resensitise tumour cells to subsequent therapies whilst averting the tumorigenic effects of tumour-stromal cell interactions. So far, the pre-clinical studies have suggested that PDT can successfully mediate the destruction of various stromal elements without increasing the aggressiveness of the tumour. However, the complexity of this interplay, including the combined tumour promoting and suppressing effects, poses unknowns for the clinical application of photodynamic stromal depletion in PDAC.
Collapse
Affiliation(s)
- Nicole Lintern
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Andrew M. Smith
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| | - David G. Jayne
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Yazan S. Khaled
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
83
|
Li J, Du J, Li Y, Meng M, Hang J, Shi H. A nomogram based on CT texture features to predict the response of patients with advanced pancreatic cancer treated with chemotherapy. BMC Gastroenterol 2023; 23:274. [PMID: 37563572 PMCID: PMC10416463 DOI: 10.1186/s12876-023-02902-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE This study aimed to evaluate the predictive value of computed tomography (CT) texture features in the treatment response of patients with advanced pancreatic cancer (APC) receiving palliative chemotherapy. METHODS This study enrolled 84 patients with APC treated with first-line chemotherapy and conducted texture analysis on primary pancreatic tumors. 59 patients and 25 were randomly assigned to the training and validation cohorts at a ratio of 7:3. The treatment response to chemotherapy was evaluated according to the Response Evaluation Criteria in Solid Tumors (RECIST1.1). The patients were divided into progressive and non-progressive groups. The least absolute shrinkage selection operator (LASSO) was applied for feature selection in the training cohort and a radiomics signature (RS) was calculated. A nomogram was developed based on a multivariate logistic regression model incorporating the RS and carbohydrate antigen 19-9 (CA19-9), and was internally validated using the C-index and calibration plot. We performed the decision curve analysis (DCA) and clinical impact curve analysis to reflect the clinical utility of the nomogram. The nomogram was further externally confirmed in the validation cohort. RESULTS The multivariate logistic regression analysis indicated that the RS and CA19-9 were independent predictors (P < 0.05), and a trend was found for chemotherapy between progressive and non-progressive groups. The nomogram incorporating RS, CA19-9 and chemotherapy showed favorable discriminative ability in the training (C-index = 0.802) and validation (C-index = 0.920) cohorts. The nomogram demonstrated favorable clinical utility. CONCLUSION The RS of significant texture features was significantly associated with the early treatment effect of patients with APC treated with chemotherapy. Based on the RS, CA19-9 and chemotherapy, the nomogram provided a promising way to predict chemotherapeutic effects for APC patients.
Collapse
Affiliation(s)
- Jingjing Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Jiadi Du
- Department of Computer Science, Missouri University of Science and Technology, Rolla, MO, U.S
| | - Yuying Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Mingzhu Meng
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Junjie Hang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 518116, Shenzhen, China.
- Department of Oncology, the Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, China.
| | - Haifeng Shi
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China.
| |
Collapse
|
84
|
Kitano Y, Inoue Y, Takeda T, Oba A, Ono Y, Sato T, Ito H, Ozaka M, Sasaki T, Sasahira N, Baba H, Takahashi Y. Clinical Efficacy of Neoadjuvant Chemotherapy with Gemcitabine plus S-1 for Resectable Pancreatic Ductal Adenocarcinoma Compared with Upfront Surgery. Ann Surg Oncol 2023; 30:5093-5102. [PMID: 37140750 DOI: 10.1245/s10434-023-13534-z] [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: 10/10/2022] [Accepted: 04/03/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND The efficacy of neoadjuvant chemotherapy with gemcitabine plus S-1 (NAC-GS) in the prognosis of patients with resectable pancreatic ductal adenocarcinoma (PDAC) has been reported. NAC-GS is now assumed to be a standard regimen for resectable PDAC in Japan. However, the reason for this improvement in prognosis remains unclear. METHODS In 2019, we introduced NAC-GS for resectable PDAC. From 2015 to 2021, 340 patients were diagnosed with resectable PDAC (anatomical and biological [carbohydrate antigen (CA) 19-9 < 500 U/mL]) and were divided according to the treatment period (upfront surgery [UPS] group, 2015-2019, n = 241; NAC-GS group, 2019-2021, n = 80). We used "intention-to-treat" analysis to compare the clinical outcomes of NAC-GS to those of UPS. RESULTS Of the 80 patients with NAC-GS, 75 (93.8%) completed two cycles of NAC-GS, and the resection rate of the NAC-GS group was comparable to that of the UPS group (92.5 vs. 91.3%, P = 0.73). The R0 resection rate was significantly higher in the NAC-GS group than in the UPS group (91.3 vs. 82.6%, P = 0.04), even though the surgical burden was smaller. Progression-free survival tended to be better (hazard ratio [HR] = 0.70, P = 0.06), and overall survival was significantly better in the NAC-GS group than in the UPS group (HR 0.55, P = 0.02). CONCLUSIONS NAC-GS provided improvements in microscopic invasion leading to a high R0 rate and smooth administration and completion of adjuvant therapy, which might lead to an improved prognosis in patients with resectable PDAC.
Collapse
Affiliation(s)
- Yuki Kitano
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan.
| | - Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Yoshihiro Ono
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Takafumi Sato
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Hiromichi Ito
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| |
Collapse
|
85
|
Mukherjee S, Korfiatis P, Khasawneh H, Rajamohan N, Patra A, Suman G, Singh A, Thakkar J, Patnam NG, Trivedi KH, Karbhari A, Chari ST, Truty MJ, Halfdanarson TR, Bolan CW, Sandrasegaran K, Majumder S, Goenka AH. Bounding box-based 3D AI model for user-guided volumetric segmentation of pancreatic ductal adenocarcinoma on standard-of-care CTs. Pancreatology 2023; 23:522-529. [PMID: 37296006 PMCID: PMC10676442 DOI: 10.1016/j.pan.2023.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVES To develop a bounding-box-based 3D convolutional neural network (CNN) for user-guided volumetric pancreas ductal adenocarcinoma (PDA) segmentation. METHODS Reference segmentations were obtained on CTs (2006-2020) of treatment-naïve PDA. Images were algorithmically cropped using a tumor-centered bounding box for training a 3D nnUNet-based-CNN. Three radiologists independently segmented tumors on test subset, which were combined with reference segmentations using STAPLE to derive composite segmentations. Generalizability was evaluated on Cancer Imaging Archive (TCIA) (n = 41) and Medical Segmentation Decathlon (MSD) (n = 152) datasets. RESULTS Total 1151 patients [667 males; age:65.3 ± 10.2 years; T1:34, T2:477, T3:237, T4:403; mean (range) tumor diameter:4.34 (1.1-12.6)-cm] were randomly divided between training/validation (n = 921) and test subsets (n = 230; 75% from other institutions). Model had a high DSC (mean ± SD) against reference segmentations (0.84 ± 0.06), which was comparable to its DSC against composite segmentations (0.84 ± 0.11, p = 0.52). Model-predicted versus reference tumor volumes were comparable (mean ± SD) (29.1 ± 42.2-cc versus 27.1 ± 32.9-cc, p = 0.69, CCC = 0.93). Inter-reader variability was high (mean DSC 0.69 ± 0.16), especially for smaller and isodense tumors. Conversely, model's high performance was comparable between tumor stages, volumes and densities (p > 0.05). Model was resilient to different tumor locations, status of pancreatic/biliary ducts, pancreatic atrophy, CT vendors and slice thicknesses, as well as to the epicenter and dimensions of the bounding-box (p > 0.05). Performance was generalizable on MSD (DSC:0.82 ± 0.06) and TCIA datasets (DSC:0.84 ± 0.08). CONCLUSION A computationally efficient bounding box-based AI model developed on a large and diverse dataset shows high accuracy, generalizability, and robustness to clinically encountered variations for user-guided volumetric PDA segmentation including for small and isodense tumors. CLINICAL RELEVANCE AI-driven bounding box-based user-guided PDA segmentation offers a discovery tool for image-based multi-omics models for applications such as risk-stratification, treatment response assessment, and prognostication, which are urgently needed to customize treatment strategies to the unique biological profile of each patient's tumor.
Collapse
Affiliation(s)
- Sovanlal Mukherjee
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Panagiotis Korfiatis
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Naveen Rajamohan
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Anurima Patra
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Garima Suman
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Aparna Singh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Jay Thakkar
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Nandakumar G Patnam
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Kamaxi H Trivedi
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Aashna Karbhari
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Mark J Truty
- Department of Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | | | - Candice W Bolan
- Department of Radiology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA.
| | - Kumar Sandrasegaran
- Department of Radiology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA.
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
86
|
Wang M, Hu Q, Huang J, Zhang F, Yao Z, Shao S, Zhao X, Liang T. In Situ Formed ROS-Responsive Hydrogel with STING Agonist and Gemcitabine to Intensify Immunotherapy against Pancreatic Ductal Adenocarcinoma. Adv Healthc Mater 2023; 12:e2203264. [PMID: 36971070 DOI: 10.1002/adhm.202203264] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/22/2023] [Indexed: 03/29/2023]
Abstract
Immunotherapy, the most revolutionary anticancer strategy, faces major obstacles in yielding desirable outcomes in pancreatic ductal adenocarcinoma (PDAC) due to the highly immunosuppressive tumor microenvironment (TME). Meanwhile, when used alone, the traditional first-line chemotherapeutic agent gemcitabine (GEM) in PDAC treatment is also insufficient to achieve lasting efficacy. In this study, a reactive oxygen species degradable hydrogel system, denoted as GEM-STING@Gel, is engineered to codeliver gemcitabine and the stimulator of interferon genes (STING) agonist DMXAA (5,6-dimethylxanthenone-4-acetic acid) into the tumor site. In this work, the strategy addresses the major challenges of current immunotherapies with a facile platform, which can synergistically activate innate immunity and promote the cytotoxic T lymphocytes infiltration at the tumor site, thereby modulating the immunosuppressive TME. Further, the efficient therapeutic potency of the immunotherapy is confirmed in an orthotopic postsurgical model, unleashing the translational potential to prevent tumor recurrence after surgical resection. This study underscores the advantages of this integrative strategy that combines chemotherapy, immunotherapy, and biomaterial-based hydrogel, including improved therapeutic efficacy, operational convenience, and superior biosafety.
Collapse
Affiliation(s)
- Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Qida Hu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Junming Huang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Fu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhuo Yao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shiyi Shao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Xinyu Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310003, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 311121, China
| |
Collapse
|
87
|
Racu ML, Bernardi D, Chaouche A, Zindy E, Navez J, Loi P, Maris C, Closset J, Van Laethem JL, Decaestecker C, Salmon I, D’Haene N. SMAD4 Positive Pancreatic Ductal Adenocarcinomas Are Associated with Better Outcomes in Patients Receiving FOLFIRINOX-Based Neoadjuvant Therapy. Cancers (Basel) 2023; 15:3765. [PMID: 37568581 PMCID: PMC10417261 DOI: 10.3390/cancers15153765] [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: 06/28/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND SMAD4 is inactivated in 50-55% of pancreatic ductal adenocarcinomas (PDACs). SMAD4 loss of expression has been described as a negative prognostic factor in PDAC associated with an increased rate of metastasis and resistance to therapy. However, the impact of SMAD4 inactivation in patients receiving neoadjuvant therapy (NAT) is not well characterized. The aim of our study was to investigate whether SMAD4 status is a prognostic and predictive factor in patients receiving NAT. METHODS We retrospectively analyzed 59 patients from a single center who underwent surgical resection for primary PDAC after NAT. SMAD4 nuclear expression was assessed by immunohistochemistry, and its relationship to clinicopathologic variables and survival parameters was evaluated. Interaction testing was performed between SMAD4 status and the type of NAT. RESULTS 49.15% of patients presented loss of SMAD4. SMAD4 loss was associated with a higher positive lymph node ratio (p = 0.03), shorter progression-free survival (PFS) (p = 0.02), and metastasis-free survival (MFS) (p = 0.02), but it was not an independent prognostic biomarker in multivariate analysis. Interaction tests demonstrated that patients with SMAD4-positive tumors receiving FOLFIRINOX-based NAT showed the best outcome. CONCLUSION This study highlights the potential prognostic and predictive role of SMAD4 status in PDAC patients receiving FOLFIRINOX-based NAT.
Collapse
Affiliation(s)
- Marie-Lucie Racu
- Departement of Pathology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (D.B.); (A.C.); (C.M.); (I.S.); (N.D.)
| | - Dana Bernardi
- Departement of Pathology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (D.B.); (A.C.); (C.M.); (I.S.); (N.D.)
| | - Aniss Chaouche
- Departement of Pathology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (D.B.); (A.C.); (C.M.); (I.S.); (N.D.)
| | - Egor Zindy
- Digital Image Analysis in Pathology (DIAPath), Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium; (E.Z.); (C.D.)
- Laboratory of Image Synthesis and Analysis (LISA), Brussels School of Engineering/École Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Julie Navez
- Department of Hepato-Biliary-Pancreatic Surgery, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (J.N.); (P.L.); (J.C.); (J.-L.V.L.)
| | - Patrizia Loi
- Department of Hepato-Biliary-Pancreatic Surgery, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (J.N.); (P.L.); (J.C.); (J.-L.V.L.)
| | - Calliope Maris
- Departement of Pathology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (D.B.); (A.C.); (C.M.); (I.S.); (N.D.)
| | - Jean Closset
- Department of Hepato-Biliary-Pancreatic Surgery, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (J.N.); (P.L.); (J.C.); (J.-L.V.L.)
| | - Jean-Luc Van Laethem
- Department of Hepato-Biliary-Pancreatic Surgery, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (J.N.); (P.L.); (J.C.); (J.-L.V.L.)
| | - Christine Decaestecker
- Digital Image Analysis in Pathology (DIAPath), Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium; (E.Z.); (C.D.)
- Laboratory of Image Synthesis and Analysis (LISA), Brussels School of Engineering/École Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Isabelle Salmon
- Departement of Pathology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (D.B.); (A.C.); (C.M.); (I.S.); (N.D.)
- Digital Image Analysis in Pathology (DIAPath), Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium; (E.Z.); (C.D.)
| | - Nicky D’Haene
- Departement of Pathology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium; (D.B.); (A.C.); (C.M.); (I.S.); (N.D.)
| |
Collapse
|
88
|
Gudmundsdottir H, Yonkus JA, Alva-Ruiz R, Kendrick ML, Smoot RL, Warner SG, Starlinger P, Thiels CA, Nagorney DM, Cleary SP, Grotz TE, Truty MJ. Yield of Staging Laparoscopy for Pancreatic Cancer in the Modern Era: Analysis of More than 1,000 Consecutive Patients. J Am Coll Surg 2023; 237:49-57. [PMID: 37026837 PMCID: PMC10262988 DOI: 10.1097/xcs.0000000000000704] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Accurate staging prior to resection of pancreatic ductal adenocarcinoma (PDAC) is imperative to avoid unnecessary operative morbidity and oncologic futility in patients with occult intra-abdominal distant metastases. We aimed to determine the diagnostic yield of staging laparoscopy (SL) and to identify factors associated with increased risk of positive laparoscopy (PL) in the modern era. STUDY DESIGN Patients with radiographically localized PDAC who underwent SL from 2017 to 2021 were retrospectively reviewed. The yield of SL was defined as the proportion of patients with PL, including gross metastases and/or positive peritoneal cytology. Factors associated with PL were assessed using univariate analysis and multivariable logistic regression. RESULTS Of 1,004 patients who underwent SL, 180 (18%) had PL due to gross metastases (n = 140) and/or positive cytology (n = 96). Patients who had neoadjuvant chemotherapy prior to laparoscopy had lower rates of PL (14% vs 22%, p = 0.002). When the analysis was restricted to chemo-naive patients who had concurrent peritoneal lavage performed, 95 of 419 patients (23%) had PL. In multivariable analysis, PL was associated with younger (<60) age, indeterminate extrapancreatic lesions on preoperative imaging, body/tail tumor location, larger tumor size, and elevated serum CA 19-9 (all p < 0.05). Among patients with no indeterminate extrapancreatic lesions on preoperative imaging, the rate of PL ranged from 1.6% in patients with no risk factors to 42% in young patients with large body/tail tumors and elevated serum CA 19-9. CONCLUSIONS The rate of PL in patients with PDAC remains high in the modern era. SL with peritoneal lavage should be considered for the majority of patients prior to resection, specifically those with high-risk features, and ideally prior to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Rory L Smoot
- From the Department of Surgery, Mayo Clinic, Rochester, MN
| | | | | | | | | | - Sean P Cleary
- From the Department of Surgery, Mayo Clinic, Rochester, MN
| | - Travis E Grotz
- From the Department of Surgery, Mayo Clinic, Rochester, MN
| | - Mark J Truty
- From the Department of Surgery, Mayo Clinic, Rochester, MN
| |
Collapse
|
89
|
Fogliati A, Fiorentini G, Alva-Ruiz R, Abdelrahman AM, Zironda A, Lynch IT, Smoot RL, Starlinger PP, Cleary SP, Kendrick ML, Truty MJ. Technical Outcomes of Porto-Mesenteric Venous Reconstruction in Pancreatic Resection Using Autologous Left Renal Vein Graft as Conduit. J Am Coll Surg 2023; 237:58-67. [PMID: 37158471 DOI: 10.1097/xcs.0000000000000744] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
BACKGROUND Portal or superior mesenteric vein (PV-SMV) resection and reconstruction is sometimes required during pancreatic tumor resection. In patients requiring segmental venous resection with interposition grafting, the left renal vein (LRV) is an accessible autologous solution. However, long-term patency outcomes of the LRV as an interposition conduit in this setting have not been analyzed. STUDY DESIGN We conducted a retrospective analysis of patients undergoing pancreatic resection with PV-SMV reconstruction using LRV between 2002 and 2022. The primary outcome was PV-SMV patency at last follow-up, assessed with postoperative CT scans and analyzed using Kaplan-Meier survival methods that account for variation in follow-up duration. Development of any postoperative acute kidney injury within 7 days of surgery and morbidity were secondary outcomes. RESULTS The study cohort includes 65 patients who underwent LRV harvest; 60 (92%) ultimately underwent successful reconstruction with harvested LRV graft. Kaplan-Meier 2-year estimated patency rate of the LRV graft was 88%, with no cases of complete occlusion. Six (10%) patients experienced graft stenosis. Nine of 61 (15%) patients experienced grade II or III acute kidney injury, 6 of 9 returning to normal renal function before discharge. No difference in median serum creatinine was observed at baseline, 6 and 12 months from surgery. LRV remnant thrombosis was seen in 7 of 65 (11%) patients. Only 3 of 61 (5%) patients had persistent acute kidney injury caused by complications unrelated to LRV harvesting. CONCLUSIONS Autologous LRV graft was a reliable conduit for segmental PV-SMV reconstruction, resulting in a high patency rate and marginal impact on renal function. LRV harvest is a safe and potentially ideal surgical option for PV-SMV reconstruction in pancreatic surgery.
Collapse
Affiliation(s)
- Alessandro Fogliati
- From the Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Omiya K, Oba A, Inoue Y, Kobayashi K, Wu YHA, Ono Y, Sato T, Sasaki T, Ozaka M, Sasahira N, Ito H, Saiura A, Takahashi Y. Serum DUPAN-2 could be an Alternative Biological Marker for CA19-9 Nonsecretors with Pancreatic Cancer. Ann Surg 2023; 277:e1278-e1283. [PMID: 35081567 DOI: 10.1097/sla.0000000000005395] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study investigates the use of serum DUPAN-2 in predicting the PC progression in CA19-9 nonsecretors. BACKGROUND Although we previously reported that serum CA19-9 >500U/ mL is a poor prognostic factor and an indication for enhanced neoadjuvant treatment, there is not a biomarker surrogate that equivalently predicts prognosis for CA19-9 nonsecretors. METHODS We evaluated consecutive PC patients who underwent pancreatectomy from 2005 to 2019. All patients were categorized as either nonsecretor or secretor (CA19-9 ≤ or >2.0U/mL). RESULTS Of the 984 resected PC patients, 94 (9.6%) were nonsecretors and 890 (90.4%) were secretors. The baseline characteristics were not statistically different between the 2 groups except for the level of DUPAN-2 (720 vs. 100U/mL, P < 0.001). Survival curves after resection were similar between the 2 groups (29.4 months vs. 31.3 months, P = 0.900). Survival curves of patients with DUPAN-2 >2000U/mL in the nonsecretors and patients with CA19-9 >500U/mL in the secretors were nearly equivalent as well (hazard ratio 2.08 vs. 1.89). In the multivariate analysis, DUPAN-2 >2000U/mL (hazard ratio 2.53, P = 0.010) was identified as independent prognostic factor after resection. CONCLUSION DUPAN-2 >2000U/mL in CA19-9 nonsecretors can be an unfavorable factor that corresponds to CA19-9 >500U/mL in CA19-9 secretors which is an indicator for enhanced neoadjuvant treatment. The current results shed light on the subset of nonsecretors with poor prognosis that were traditionally categorized in a group with a more favorable prognosis group.
Collapse
Affiliation(s)
- Kojiro Omiya
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kosuke Kobayashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Y H Andrew Wu
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yoshihiro Ono
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takafumi Sato
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Sasaki
- Department of Gastroenterological medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; and
| | - Masato Ozaka
- Department of Gastroenterological medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; and
| | - Naoki Sasahira
- Department of Gastroenterological medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; and
| | - Hiromichi Ito
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
91
|
Wang G, Zhang W, Luan X, Wang Z, Liu J, Xu X, Zhang J, Xu B, Lu S, Wang R, Ma G. The role of 18F-FDG PET in predicting the pathological response and prognosis to unresectable HCC patients treated with lenvatinib and PD-1 inhibitors as a conversion therapy. Front Immunol 2023; 14:1151967. [PMID: 37215117 PMCID: PMC10196479 DOI: 10.3389/fimmu.2023.1151967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Purpose To investigate the diagnostic value of 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET), as an imaging biomarker, for predicting pathological response and prognosis of unresectable hepatocellular carcinoma (HCC) patients treated with Lenvatinib and programmed cell death protein 1 (PD-1) inhibitors as a conversion therapy. Methods A total of 28 unresectable HCC patients with BCLC stage B or C were treated with Lenvatinib and PD-1 inhibitors before surgery. The 18F-FDG PET/CT scans were acquired before pre- (scan-1) and post-conversion therapy (scan-2). The maximum standardized uptake value (SUVmax), TLR (tumor-to-normal liver standardized uptake value ratio), and the percentages of post-treatment changes in metabolic parameters (ΔSUVmax [%] and ΔTLR [%]) were calculated. Major pathological response (MPR) was identified based on the residual viable tumor in the resected primary tumor specimen (≤10%). Differences in the progression-free survival (PFS) and overall survival (OS) stratified by ΔTLR were examined by the Kaplan-Meier method. Results 11 (11/28, 39.3%) patients were considered as MPR responders and 17 (17/28, 60.7%) patients as non-MPR responders after conversion therapy. ΔSUVmax (-70.0 [-78.8, -48.8] vs. -21.7 [-38.8, 5.7], respectively; P<0.001) and ΔTLR (-67.6 [-78.1, -56.8] vs. -18.6 [-27.9, 4.0], respectively; P<0.001) were reduced in the responder group than those in the non-responder group. According to the results of the receiver operating characteristic curve analysis, ΔTLR showed an excellent predictive value for the MPR of primary HCC lesions (area under curve=0.989, with the optimal diagnostic threshold of -46.15). When using ΔTLR of -21.36% as a threshold, patients with ΔTLR-based metabolic response had superior PFS (log-rank test, P=0.001) and OS (log-rank test, P=0.016) compared with those without ΔTLR-based metabolic response. Conclusion 18F-FDG PET is a valuable tool for predicting pathological response and prognosis of unresectable HCC patients treated by Lenvatinib combined with PD-1 as a conversion therapy.
Collapse
Affiliation(s)
- Guanyun Wang
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenwen Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese People's Liberation Army (PLA) General Hospital/Institute of Hepatobiliary Surgery of Chinese People's Liberation Army/Key Laboratory of Digital Hepetobiliary Surgery, People's Liberation Army, Beijing, China
| | - Xiaohui Luan
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School, Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Zhanbo Wang
- Department of Pathology, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiajin Liu
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaodan Xu
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jinming Zhang
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Baixuan Xu
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shichun Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese People's Liberation Army (PLA) General Hospital/Institute of Hepatobiliary Surgery of Chinese People's Liberation Army/Key Laboratory of Digital Hepetobiliary Surgery, People's Liberation Army, Beijing, China
| | - Ruimin Wang
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Guangyu Ma
- Department of Nuclear Medicine, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
92
|
Coveler AL, Pillarisetty VG, Koh WJ, Zhen DB, Park JO, King GG, Sham JG, Hannan LM, Mann GN, Baker KK, Redman MW, Swanson PE, Chiorean EG, Whiting SH. Perioperative Chemotherapy and Chemoradiotherapy for Patients With Resectable and Borderline Resectable Pancreatic Adenocarcinoma. Pancreas 2023; 52:e282-e287. [PMID: 37782886 DOI: 10.1097/mpa.0000000000002253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDA) is the third most common cause of cancer death in the United States. Most patients who undergo resection develop recurrence. Standard treatment confers a median overall survival (OS) of 24 months. Exposure to alternate regimens may prevent chemoresistance. This study evaluated multiagent perioperative therapy for potentially resectable PDA patients to improve OS. METHODS A single center, phase 2, trial of patients with resectable or borderline resectable PDA. Patients received neoadjuvant therapy with induction chemotherapy (gemcitabine, docetaxel, capecitabine) for 3 cycles, chemoradiation (intensity-modulated radiation therapy with capecitabine and oxaliplatin) followed by surgery, and 2 months of adjuvant gemcitabine and oxaliplatin and 2 months of gemcitabine. The primary endpoint was OS. The secondary endpoint was recurrence-free survival (RFS). RESULTS Thirty-two eligible patients were enrolled. Twenty-two patients underwent surgical resection. After a median follow-up of 56.8 months, mOS was 31.6 months (95% confidence interval [CI], 14.2-58.1) for all patients, 58.1 months (95% CI, 31.6 to NR) for those who completed surgery. The mRFS was 31.3 months (95% CI, 12.5 to NR). CONCLUSIONS Perioperative therapy with GTX, chemoradiotherapy, and adjuvant GemOx/Gem resulted in promising survival of 58 months for patients who underwent resection and may represent another treatment option for PDA.
Collapse
Affiliation(s)
| | | | - Wui-Jin Koh
- National Comprehensive Cancer Network (NCCN), Plymouth Meeting, PA
| | | | - James O Park
- Department of Surgery, University of Washington, Seattle, WA
| | | | - Jonathan G Sham
- Department of Surgery, University of Washington, Seattle, WA
| | - Lindsay M Hannan
- From the Department of Medicine, University of Washington, Seattle, WA
| | - Gary N Mann
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | | | - Paul E Swanson
- Department of Pathology, University of Washington, Seattle, WA
| | | | | |
Collapse
|
93
|
Peng M, Ying Y, Zhang Z, Liu L, Wang W. Reshaping the Pancreatic Cancer Microenvironment at Different Stages with Chemotherapy. Cancers (Basel) 2023; 15:cancers15092448. [PMID: 37173915 PMCID: PMC10177210 DOI: 10.3390/cancers15092448] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/09/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
The dynamic tumor microenvironment, especially the immune microenvironment, during the natural progression and/or chemotherapy treatment is a critical frontier in understanding the effects of chemotherapy on pancreatic cancer. Non-stratified pancreatic cancer patients always receive chemotherapeutic strategies, including neoadjuvant chemotherapy and adjuvant chemotherapy, predominantly according to their physical conditions and different disease stages. An increasing number of studies demonstrate that the pancreatic cancer tumor microenvironment could be reshaped by chemotherapy, an outcome caused by immunogenic cell death, selection and/or education of preponderant tumor clones, adaptive gene mutations, and induction of cytokines/chemokines. These outcomes could in turn impact the efficacy of chemotherapy, making it range from synergetic to resistant and even tumor-promoting. Under chemotherapeutic impact, the metastatic micro-structures in the primary tumor may be built to leak tumor cells into the lymph or blood vasculature, and micro-metastatic/recurrent niches rich in immunosuppressive cells may be recruited by cytokines and chemokines, which provide housing conditions for these circling tumor cells. An in-depth understanding of how chemotherapy reshapes the tumor microenvironment may lead to new therapeutic strategies to block its adverse tumor-promoting effects and prolong survival. In this review, reshaped pancreatic cancer tumor microenvironments due to chemotherapy were reflected mainly in immune cells, pancreatic cancer cells, and cancer-associated fibroblast cells, quantitatively, functionally, and spatially. Additionally, small molecule kinases and immune checkpoints participating in this remodeling process caused by chemotherapy are suggested to be blocked reasonably to synergize with chemotherapy.
Collapse
Affiliation(s)
- Maozhen Peng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Ying
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
94
|
Guggenberger KV, Bley TA, Held S, Keller R, Flemming S, Wiegering A, Germer CT, Kimmel B, Kunzmann V, Hartlapp I, Anger F. Predictive value of computed tomography on surgical resectability in locally advanced pancreatic cancer treated with multiagent induction chemotherapy: Results from a prospective, multicentre phase 2 trial (NEOLAP-AIO-PAK-0113). Eur J Radiol 2023; 163:110834. [PMID: 37080059 DOI: 10.1016/j.ejrad.2023.110834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE To assess the role of current imaging-based resectability criteria and the degree of radiological downsizing in locally advanced pancreatic adenocarcinoma (LAPC) after multiagent induction chemotherapy (ICT) in multicentre, open-label, randomized phase 2 trial. METHOD LAPC patients were prospectively treated with multiagent ICT followed by surgical exploration within the NEOLAP trial. All patients underwent CT scan at baseline and after ICT to assess resectability status according to national comprehensive cancer network guidelines (NCCN) criteria and response evaluation criteria in solid tumors (RECIST) at the local study center and retrospectively in a central review. Imaging results were compared in terms of local and central staging, downsizing and pathological resection status. RESULTS 83 patients were evaluable for central review of baseline and restaging imaging results. Downstaging by central review was rarely seen after multiagent ICT (7.7%), whereas tumor downsizing was documented frequently (any downsizing 90.4%, downsizing to partial response (PR) according to RECIST: 26.5%). Patients with any downsizing showed no significant different R0 resection rate (37.3%) as patients that fulfilled the criteria of PR (40.9%). The sensitivity of any downsizing for predicting R0 resection was 97% with a negative predictive value (NPV) of 0.88. ROC-analysis revealed that tumor downsizing was a predictor of R0 resection (AUC 0.647, p = 0.028) with a best cut-off value of 22.5% downsizing yielding a sensitivity of 65% and a specificity of 61%. CONCLUSIONS Imaging-based tumor downsizing and not downstaging can guide the selection of patients with a realistic chance of R0-resection in LAPC after multi-agent ICT.
Collapse
Affiliation(s)
- K V Guggenberger
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Wuerzburg, Germany.
| | - T A Bley
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - S Held
- Department of Biometrics, ClinAssess GmbH, Leverkusen, Germany
| | - R Keller
- Clinical Research, AIO Studien gGmbH, Berlin, Germany
| | - S Flemming
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - A Wiegering
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - C T Germer
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - B Kimmel
- Department of Internal Medicine II, Medical Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - V Kunzmann
- Department of Internal Medicine II, Medical Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - I Hartlapp
- Department of Internal Medicine II, Medical Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - F Anger
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
95
|
de Jong TL, Koopman D, van der Worp CAJ, Stevens H, Vuijk FA, Vahrmeijer AL, Mieog JSD, de Groot JWB, Meijssen MAC, Nieuwenhuijs VB, de Geus-Oei LF, Jager PL, Patijn GA. Added value of digital FDG-PET/CT in disease staging and restaging in patients with resectable or borderline resectable pancreatic cancer. Surg Oncol 2023; 47:101909. [PMID: 36739788 DOI: 10.1016/j.suronc.2023.101909] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/09/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND We studied the added value of digital FDG-PET/CT in disease staging and restaging compared to the standard work-up with contrast enhanced CT (ceCT) and CA19-9 in patients with resectable or borderline resectable pancreatic cancer who received neo-adjuvant therapy. Primary endpoints were tumor response compared to ceCT and CA19.9 as well as the ability to detect distant metastatic disease. METHODS 35 patients were included in this dual-center prospective study. FDG-PET using digital photon counting technology combined with CT scans were acquired before (T1) and after neo-adjuvant therapy (T2). Patients were staged and restaged based on standard protocol with ceCT and CA 19.9, while all PET/CT scans were stored securely and not included in clinical decision making. After the pancreatic resection, an expert team retrospectively assessed the CT tumor diameter, CA19-9, tumor FDG-uptake, and appearance of metastatic disease of all patients for both time points. RESULTS CA19-9 levels, CT tumor diameter, and tumor FDG-uptake on PET significantly decreased from T1 to T2 (p = 0.017, p = 0.001, and p < 0.0001). The change in FDG-uptake values showed a strong positive correlation with the change in CT tumor diameter and change in CA19-9 (R = 0.75 and R = 0.73, respectively). In addition, small-volume liver lesions were detected on digital PET/CT in 5/35 patients (14%), 4 of which were pathology confirmed at laparotomy. Only one of these five cases was detected on baseline staging ceCT (3%). CONCLUSION We found that adding digital PET/CT strengthens restaging after neo-adjuvant therapy based on the observed strong correlation with ceCT tumor diameter and Ca19.9. Also, digital PET/CT was found to detect occult metastatic disease not visualized on ceCT, that would have resulted in altered disease staging and therapeutic strategy in a substantial proportion of patients.
Collapse
Affiliation(s)
- Tonke L de Jong
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Daniëlle Koopman
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | | | - Henk Stevens
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Floris A Vuijk
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Maarten A C Meijssen
- Department of Gastroenterology and Hepatology, Isala Hospital, Zwolle, the Netherlands
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands
| | - Pieter L Jager
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Gijs A Patijn
- Department of Surgery, Isala Hospital, Zwolle, the Netherlands.
| |
Collapse
|
96
|
Noda Y, Mizuno N, Kawai N, Ando T, Kawaguchi M, Nagata S, Fujimoto K, Nakamura F, Kaga T, Ishihara T, Hyodo F, Kato H, Kambadakone AR, Matsuo M. Determination of arterial invasion in pancreatic ductal adenocarcinoma: what is the best diagnostic criterion on CT? Eur Radiol 2023; 33:3617-3626. [PMID: 36897348 DOI: 10.1007/s00330-023-09521-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/09/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023]
Abstract
OBJECTIVES To investigate the diagnostic performance and interobserver variability in the determination of arterial invasion in pancreatic ductal adenocarcinoma (PDAC) and determine the best CT imaging criterion. METHODS We retrospectively evaluated 128 patients with PDAC (73 men and 55 women) who underwent preoperative contrast-enhanced CT. Five board-certified radiologists (expert) and four fellows (non-expert]) independently assessed the arterial invasion (celiac, superior mesenteric, splenic, and common hepatic arteries) using a 6-point score: 1, no tumor contact; 2, hazy attenuation ≤ 180°; 3, hazy attenuation > 180°; 4, solid soft tissue contact ≤ 180°; 5, solid soft tissue contact > 180°; and 6, contour irregularity. ROC analysis was performed to evaluate the diagnostic performance and determine the best diagnostic criterion for arterial invasion, with pathological or surgical findings as references. Interobserver variability was assessed using Fleiss's ĸ statistics. RESULTS Among the 128 patients, 35.2% (n = 45/128) received neoadjuvant treatment (NTx). Solid soft tissue contact ≤ 180° was the best diagnostic criterion for arterial invasion as defined by the Youden Index both in patients who did and did not receive NTx (sensitivity, 100% vs. 100%; specificity, 90% vs. 93%; and AUC, 0.96 vs. 0.98, respectively). Interobserver variability among the non-expert was not inferior to that among the expert (ĸ = 0.61 vs 0.61; p = .39 and ĸ = 0.59 vs 0.51; p < .001 in patients treated with and without NTx, respectively). CONCLUSIONS Solid soft tissue contact ≤ 180° was the best diagnostic criterion for the determination of arterial invasion in PDAC. Considerable interobserver variability was seen among the radiologists. KEY POINTS • Solid soft tissue contact ≤ 180° was the best diagnostic criterion for the determination of arterial invasion in pancreatic ductal adenocarcinoma. • Interobserver agreement among non-expert radiologists was almost comparable to that among expert radiologists.
Collapse
Affiliation(s)
- Yoshifumi Noda
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Nozomi Mizuno
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Nobuyuki Kawai
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Tomohiro Ando
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Masaya Kawaguchi
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Shoma Nagata
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Keita Fujimoto
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Fumihiko Nakamura
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Tetsuro Kaga
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takuma Ishihara
- Innovative and Clinical Research Promotion Center, Gifu University Hospital, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Fuminori Hyodo
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
- Institute for Advanced Study, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Hiroki Kato
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Avinash R Kambadakone
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, White 270, Boston, MA, 02114, USA
| | - Masayuki Matsuo
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
97
|
Alkhasawneh A, Rashid T, Mohammed I, Elhaddad B, Al-Balas H, Virarkar M, Awad Z, Baskovich B, Gopinath A. The prognostic significance of duodenal wall invasion in pancreatic adenocarcinoma. World J Surg Oncol 2023; 21:79. [PMID: 36872330 PMCID: PMC9987094 DOI: 10.1186/s12957-023-02962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/23/2023] [Indexed: 03/07/2023] Open
Abstract
OBJECTIVE The most recent edition of the American Joint Committee on Cancer Staging Manual (AJCC, 8th edition) relies only on tumor size for staging resectable pancreatic adenocarcinoma, and the presence of duodenal wall invasion (DWI) no longer has an impact on staging. However, very few studies have evaluated its significance. In this study, we aim to evaluate the prognostic significance of DWI in pancreatic adenocarcinoma. METHODS We reviewed 97 consecutive internal cases of resected pancreatic head ductal adenocarcinoma, and clinicopathologic parameters were recorded. All cases were staged according to the 8th edition of AJCC, and the patients were divided into two groups based on the presence or absence of DWI. RESULTS Out of our 97 cases, 53 patients had DWI (55%). In univariate analysis, DWI was significantly associated with lymphovascular invasion and lymph node metastasis (AJCC 8th edition pN stage). In univariate analysis of overall survival, age > 60, absence of DWI, and African American race were associated with worse overall survival. In multivariate analysis, age > 60, absence of DWI, and African American race were associated with worse progression-free survival and overall survival. CONCLUSION Although DWI is associated with lymph node metastasis, it is not associated with inferior disease-free/overall survival.
Collapse
Affiliation(s)
| | - Tasnuva Rashid
- UF Health Jacksonville, 655 W 8th st, Jacksonville, FL, USA
| | | | - Basma Elhaddad
- UF Health Jacksonville, 655 W 8th st, Jacksonville, FL, USA
| | | | - Mayur Virarkar
- UF Health Jacksonville, 655 W 8th st, Jacksonville, FL, USA
| | - Ziad Awad
- UF Health Jacksonville, 655 W 8th st, Jacksonville, FL, USA. .,University of Florida College of Medicine, Jacksonville, 653 West 8th Street, Jacksonville, FL, 32209, USA.
| | | | - Arun Gopinath
- UF Health Jacksonville, 655 W 8th st, Jacksonville, FL, USA
| |
Collapse
|
98
|
Kelly BN, Nicolais L, Mohamed A, Fitzgerald TL. Contemporary Treatment Paradigms are Associated with Improved Survival in Pancreatic Cancer. Am Surg 2023:31348231157897. [PMID: 36872555 DOI: 10.1177/00031348231157897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
INTRODUCTION Over the last decade, a paradigm shift has been made in treating pancreatic cancer. Starting in 2011, several trials demonstrated a survival advantage for multiagent chemotherapy (MAC). However, the implication for survival at the population level remains unclear. METHODS A retrospective study of the National Cancer Database from 2006 to 2019 was conducted. Patients treated from 2006 to 2010 were classified as "Era 1", and those treated from 2011 to 2019 as "Era 2." RESULTS A total of 316,393 patients with pancreatic adenocarcinoma were identified, with 87,742 treated in Era 1 and 228,651 in Era 2. Survival increased from Era 1 to Era 2 in all patients and sub-analyses; surgical (18.7 vs 24.6 months, HR .85, 95% CI 0.82-.88, P < .001), imminently resectable (Stage IA and IB, 12.2 vs 14.8 months, HR .90, 95% CI 0.86-.95, P < .001), high-risk (Stage IIA, IIB, and III, 9.6 vs 11.6 months, HR .82, 95% CI 0.79-.85, P < .001), and Stage IV (3.5 vs 3.9 months, HR .86, 95% CI 0.84-.89, P < .001). Survival was decreased for those who were African American (P = .031), on Medicaid (P < .001), or in the lowest quartile of annual income (P < .001). Surgery rates decreased from 20.5% in Era 1 to 19.8% in Era 2 (P < .001). DISCUSSION Adoption of MAC regimens at a population level correlates with improved pancreatic cancer survival. Unfortunately, socioeconomic factors are associated with an unequal benefit from new treatment regimens, and underuse of surgery for resectable neoplasms persists.
Collapse
Affiliation(s)
- Bridget N Kelly
- 115985University of New England College of Osteopathic Medicine, Biddeford, ME, USA
| | - Laura Nicolais
- Division of Surgical Oncology, 92602Maine Medical Center, Portland, ME, USA
| | - Abdimajid Mohamed
- Division of Surgical Oncology, 92602Maine Medical Center, Portland, ME, USA
| | | |
Collapse
|
99
|
Hill CS, Herman JM. The Current Role of Radiation in Pancreatic Cancer and Future Directions. Clin Colorectal Cancer 2023; 22:12-23. [PMID: 36804206 DOI: 10.1016/j.clcc.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/15/2022] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
Survival outcomes for localized pancreatic adenocarcinoma remains poor. Multimodality therapeutic regimens are critical to maximizing survival outcomes for these patients, which includes the use of systemic therapy, surgery, and radiation. In this review, the evolution of radiation techniques are discussed with a focus on modern techniques such as intensity modulated radiation and stereotactic body radiation therapy. However, the current role of radiation within the most common clinical scenarios for pancreatic cancer in the neoadjuvant, definitive, and adjuvant settings continues to be highly debated. The role of radiation in these settings is reviewed in the context of historical and modern clinical studies. In addition, emerging concepts including dose-escalated radiation, magnetic resonance-guided radiation therapy, and particle therapy are discussed to promote an understanding of how such concepts may change the role of radiation in the future.
Collapse
Affiliation(s)
- Colin S Hill
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY.
| | - Joseph M Herman
- Radiation Medicine, Zucker School of Medicine at Hofstra/Northwell, New York, NY
| |
Collapse
|
100
|
Sugawara T, Rodriguez Franco S, Sherman S, Kirsch MJ, Colborn K, Ishida J, Grandi S, Al-Musawi MH, Gleisner A, Schulick RD, Del Chiaro M. Association of Adjuvant Chemotherapy in Patients With Resected Pancreatic Adenocarcinoma After Multiagent Neoadjuvant Chemotherapy. JAMA Oncol 2023; 9:316-323. [PMID: 36480190 PMCID: PMC9857517 DOI: 10.1001/jamaoncol.2022.5808] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/13/2022] [Indexed: 12/13/2022]
Abstract
Importance The total number of patients with pancreatic ductal adenocarcinoma (PDAC) who receive neoadjuvant chemotherapy (NAC) is increasing. However, the added role of adjuvant chemotherapy (AC) in these patients remains unknown. Objective To evaluate the association of AC with overall survival (OS) in patients with PDAC who received multiagent NAC followed by curative-intent surgery. Design, Setting, and Participants This retrospective, matched-cohort study used data from the National Cancer Database and included patients with PDAC diagnosed between 2010 and 2018. The study included patients at least 18 years of age who received multiagent NAC followed by surgical resection and had available records of the pathological findings. Patients were excluded if they had clinical or pathological stage IV disease or died within 90 days of their operation. Exposures All included patients received NAC and underwent resection for primary PDAC. Some patients received adjuvant chemotherapy. Main Outcomes and Measures The main outcome was the OS of patients who received AC (AC group) vs those who did not (non-AC group). Interactions between pathological findings and AC were investigated in separate multivariable Cox regression models. Results In total, 1132 patients (mean [SD] age, 63.5 [9.4] years; 577 [50.1%] male; 970 [85.7%] White) were included, 640 patients in the non-AC group and 492 patients in the AC group. After being matched by propensity score according to demographic and pathological characteristics, 444 patients remained in each group. The multivariable Cox regression model adjusted for all covariates revealed an association between AC and improved survival (hazard ratio, 0.71; 95% CI, 0.59-0.85; P < .001). Subgroup interaction analysis revealed that AC was significantly associated with better OS (26.6 vs 21.2 months; P = .002), but the benefit varied by age, pathological T category, and tumor differentiation. Of note, AC was associated with better survival in patients with any pathological N category and positive margin status. Conclusions and Relevance In this cohort study, AC following multiagent NAC and resection in patients with PDAC was associated with significant survival benefit compared with that in patients who did not receive AC. These findings suggest that patients with aggressive tumors may benefit from AC to achieve prolonged survival, even after multiagent NAC and curative-intent resection.
Collapse
Affiliation(s)
- Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Salvador Rodriguez Franco
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora
| | - Samantha Sherman
- Department of Surgery, Parkview Hospital Randallia, Fort Wayne, Indiana
| | - Michael J. Kirsch
- Department of Surgery, University of Colorado School of Medicine, Aurora
| | - Kathryn Colborn
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora
- Surgical Outcomes and Applied Research Program, University of Colorado School of Medicine, Aurora
| | - Jun Ishida
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora
| | - Samuele Grandi
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora
| | - Mohammed H. Al-Musawi
- Clinical Trials Office, Department of Surgery, University of Colorado School of Medicine, Aurora
| | - Ana Gleisner
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora
| | - Richard D. Schulick
- Department of Surgery, University of Colorado School of Medicine, Aurora
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora
| |
Collapse
|