1
|
Citterio D, Droz Dit Busset M, Sposito C, Mazzola M, Grandi S, Zironda A, Leoncini G, Simonotti N, Battiston C, Flores M, Ferrari G, Mazzaferro V. Prediction of early recurrence as a marker of surgical futility in pancreatic adenocarcinoma. Surg Oncol 2025; 59:102208. [PMID: 40086295 DOI: 10.1016/j.suronc.2025.102208] [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: 11/13/2024] [Revised: 02/06/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Long-term survival after resection for pancreatic ductal adenocarcinoma (PDAC) is impaired by very high recurrence rates. When recurrence occurs within 6 months (early recurrence: ER) the benefit of surgery is equivalent to palliative chemotherapy in unresectable patients. Therefore, ER is a surrogate of surgical futility in PDAC. MATERIALS AND METHODS To investigate predictive factors of ER and its impact on survival, a training and a validation cohort of prospectively collected patients who underwent surgery for resectable or borderline-resectable PDAC were analyzed in two independent Pancreas Units during the same period. Logistic regression model on the training cohort identified independent predictors of ER, used to build a prognostic risk-score then tested on the validation cohort. RESULTS Out of 176 patients in the training cohort, 21.6 % experienced ER, with significant impact on survival (OS: 9.7 months vs. 32.7 months for ER vs. late/no recurrence, respectively). At multivariable analysis, three independent risk factors for ER were identified: Ca19.9 > 100 U/mL, G3 tumors and lack of adjuvant chemotherapy. Based on such features the derived ER-score stratified three prognostic classes at incremental risk of ER (12 %, 35 % and 53 %) with different OS (31.1, 19.7 and 9.3 months, respectively, p < 0.001). The ER predictive score was then tested on a validation cohort of 242 patients, 22.3 % of whom underwent ER. Despite significant differences in tumor-related features, the score was able to discriminate among the predicted ER-risk classes (15 %, 27 % and 53 %, respectively) and forecast significantly different OS (5.8, 19 and 31.1 months, p > 0.001). The discriminative capability of the score in the two cohorts was similar (training AUC = 0.72 vs. validation AUC = 0.68, p = 0.28). CONCLUSION An externally validated clinical score, able to identify three prognostic classes at incremental risk of developing ER after resection of PDAC is provided. In patients at high risk of ER, prediction of surgical futility may help in decision-making.
Collapse
Affiliation(s)
- Davide Citterio
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Michele Droz Dit Busset
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Carlo Sposito
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Michele Mazzola
- ASST Grande Ospedale Metropolitano Niguarda, Division of Minimally-invasive Surgical Oncology, Milan, Italy
| | - Samuele Grandi
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Andrea Zironda
- ASST Grande Ospedale Metropolitano Niguarda, Division of Minimally-invasive Surgical Oncology, Milan, Italy
| | - Giuseppe Leoncini
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Nicolò Simonotti
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Carlo Battiston
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Maria Flores
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Giovanni Ferrari
- ASST Grande Ospedale Metropolitano Niguarda, Division of Minimally-invasive Surgical Oncology, Milan, Italy
| | - Vincenzo Mazzaferro
- HPB Surgery, Liver Transplantation and Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy.
| |
Collapse
|
2
|
Kishiwada M, Mizuno S, Hayasaki A, Kaluba B, Fujii T, Noguchi D, Ito T, Iizawa Y, Tanemura A, Murata Y, Kuriyama N. Impact of Surgical Resection After Induction Gemcitabine Plus S-1-Based Chemoradiotherapy in Patients with Locally Advanced Pancreatic Ductal Adenocarcinoma: A Focus on UR-LA Cases. Cancers (Basel) 2025; 17:1048. [PMID: 40149381 DOI: 10.3390/cancers17061048] [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: 01/24/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background: This study aimed to assess the safety and efficacy of gemcitabine plus S-1-based chemoradiotherapy (GS-CRT) among patients with locally advanced pancreatic ductal adenocarcinoma (PDAC), especially among those with unresectable locally advanced (UR-LA) cases. Methods: A total of 351 consecutive PDAC patients were enrolled and prognostic predictors of disease-specific survival (DSS) were identified. Results: The treatment completion rate was 98.9% and Grade 3 or higher adverse events occurred in 181 cases (51.6%). Among 319 re-evaluated patients, pancreatectomy was performed in 184 (57.7%). Based on resectability, the 5-year DSS rates for the entire cohort were 39.6% (R), 43.8% (BR-PV), 21.2% (BR-A) and 13.3% (UR-LA), while the predictors of DSS were performance status (PS), hemoglobin (Hb) level, celiac artery (CA) involvement of ≥180 degrees and JPS 8th T category. In the resected cases, the predictors of DSS were preoperative PS, preoperative CA19-9 level, preoperative JPS-T factor, degree of histological response and adjuvant chemotherapy. In UR-LA resected patients, preoperative prognostic nutritional index (PNI), absence of pathological venous invasion and adjuvant chemotherapy were predictors of DSS. Conclusions: Even though Grade 3 or higher adverse events were encountered in about half of the cases, they were uneventfully managed. Therefore, GS-CRT is safe and highly tolerable with potential to improve patients' prognosis. Preoperative PS, CA19-9 levels and histological response are important prognostic factors, as well as adjuvant therapy. In UR-LA patients, prognostic nutritional index (PNI) and adjuvant chemotherapy were important for curative intent surgery.
Collapse
Affiliation(s)
- Masashi Kishiwada
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Shugo Mizuno
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Aoi Hayasaki
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Benson Kaluba
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Takehiro Fujii
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Daisuke Noguchi
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Takahiro Ito
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Yusuke Iizawa
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Akihiro Tanemura
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Yasuhiro Murata
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Naohisa Kuriyama
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| |
Collapse
|
3
|
Ito R, Yoshioka R, Yanagisawa N, Ishii S, Sugitani J, Furuya R, Fujisawa M, Imamura H, Mise Y, Isayama H, Saiura A. Survival Analysis of Conversion Surgery in Borderline Resectable and Locally Advanced Unresectable Pancreatic Ductal Adenocarcinoma Addressing Selection and Immortal Time Bias: A Retrospective Single-Center Study. Ann Surg Oncol 2024; 31:8744-8755. [PMID: 39361176 DOI: 10.1245/s10434-024-16203-x] [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: 05/23/2024] [Accepted: 09/03/2024] [Indexed: 11/10/2024]
Abstract
BACKGROUND The purpose of this study was to provide a detailed evaluation of the oncological advantages of surgery following neoadjuvant chemotherapy (NAC) for patients with borderline resectable (BR) or unresectable (UR) pancreatic ductal adenocarcinoma (PDAC), with a focus on minimizing biases. Recently, NAC has become the standard care for BR or UR locally advanced (UR-LA) PDAC, however, many studies have assessed survival benefits and favorable variables without consideration for biases, particularly immortal time bias. PATIENTS AND METHODS This study included patients diagnosed with BR or UR-LA PDAC at Juntendo University Hospital from 2019 to 2022. To mitigate bias, we applied methods such as propensity score matching (PSM), time-dependent covariate Cox proportional hazard regression analysis (TDC), landmark analysis, and multivariable Cox proportional hazards regression model. RESULTS The study analyzed 124 patients, dividing them into a surgery group (n = 57) and a chemotherapy-only group (n = 67). After PSM, there were 21 matched pairs. Survival analysis using TDC analysis showed that the surgery group had significantly better overall survival compared with the chemotherapy-only group in both the entire cohort and the matched pairs. Cox regression analysis of the entire cohort also revealed a similar superiority of surgery, while the landmark analysis showed varying results depending on the landmark setting. CONCLUSIONS After careful adjustment for selection and immortal time biases, surgery following NAC appears to significantly extend survival in patients with BR or UR PDAC.
Collapse
Affiliation(s)
- Ryota Ito
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryuji Yoshioka
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naotake Yanagisawa
- Medical Technology Innovation Centre, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigeto Ishii
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jun Sugitani
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryoji Furuya
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Fujisawa
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Imamura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshihiro Mise
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Isayama
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
4
|
Shi S, Ye L, Jin K, Yu X, Guo D, Wu W. The complement C3a/C3aR pathway is associated with treatment resistance to gemcitabine-based neoadjuvant therapy in pancreatic cancer. Comput Struct Biotechnol J 2024; 23:3634-3650. [PMID: 39469671 PMCID: PMC11513484 DOI: 10.1016/j.csbj.2024.09.032] [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: 06/30/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
Gemcitabine is a standard first-line drug for pancreatic cancer chemotherapy. Nevertheless, gemcitabine resistance is common and significantly limits its therapeutic efficacy, impeding advancements in pancreatic cancer treatment. In this study, through a comprehensive analysis of gemcitabine-resistant cell lines and patient samples, 39 gemcitabine resistance-associated risk genes were identified, and two distinct gemcitabine response-related phenotypes were delineated. Through a combination of bioinformatics analysis and in vivo and in vitro experiments, we identified the C3a/C3aR signaling pathway as a pivotal player in the development of gemcitabine resistance in pancreatic cancer. We found that activation of the C3a/C3aR signaling pathway promoted the proliferation, migration and gemcitabine resistance of pancreatic cancer cells, while the C3aR antagonist SB290157 effectively counteracted these effects by impeding the activation of the C3a/C3aR pathway. Our study reveals the fundamental role of complement C3a in the progression of pancreatic cancer, suggesting that complement C3a may serve as a promising biomarker in pancreatic cancer.
Collapse
Affiliation(s)
- Saimeng 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
| | - Longyun Ye
- 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
| | - Kaizhou Jin
- 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
| | - Duancheng Guo
- 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
| | - Weiding Wu
- 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
|
5
|
Zhao K, Zhang J, Zhou L, Sun Z. Scutellaria baicalensis and its flavonoids in the treatment of digestive system tumors. Front Pharmacol 2024; 15:1483785. [PMID: 39654621 PMCID: PMC11625591 DOI: 10.3389/fphar.2024.1483785] [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: 08/20/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Scutellaria baicalensis has been used for the treatment of digestive system disorders for thousands of years in China and other regions. Modern research have revealed its therapeutic efforts in digestive system tumors. Thus, to review the updated progress of S. baicalensis and its main flavonoids in the treatment of digestive system tumors in the past 10 years, this article summarized the therapeutic effect and molecular mechanisms of S. baicalensis and its 5 flavonoids on tumors in oral cavity, esophagus, stomach, colon, liver, pancreas by inhibiting tumor cell proliferation, inducing autophagy, stimulating immune response, and increasing drug sensitivity. In conclusion, S. baicalensis and its flavonoids could be applied to treat digestive system tumors with different type of methods.
Collapse
Affiliation(s)
- Kangning Zhao
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinlong Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Sun
- The Second Gastroenterology Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
6
|
Son J, Lee W, Lee JB, Hong K, Sung MK, Park Y, Jun E, Song KB, Hwang DW, Lee JH, Kim SC. Prognostic model for resected borderline and locally advanced pancreatic cancer after neoadjuvant chemotherapy. Int J Surg 2024; 110:7080-7087. [PMID: 38976903 PMCID: PMC11573055 DOI: 10.1097/js9.0000000000001927] [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: 03/25/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The current AJCC 8th has been reported to have a poor ability to predict the prognosis in patients with resected borderline resectable pancreatic cancer and locally advanced pancreatic cancer following neoadjuvant chemotherapy. This study aimed to develop an improved prognostic model by incorporating pathology and parameters of biologic response (BR). MATERIALS AND METHODS A retrospective cohort study was conducted including patients who underwent curative-intent surgery following chemotherapy. The authors developed a modified ypT staging system and incorporated the BR, involving normalization of carbohydrate antigen 19-9 and reduction in the maximum standardized uptake value simultaneously after chemotherapy. The prognostic performance of the current pathologic system, modified pathologic system, and newly developed system incorporating pathology and BR were compared. RESULTS In this study, 171 patients underwent surgery following chemotherapy. The modified T stage, which unified ypT2 and ypT3, demonstrated improved prognostic performance than the current staging system [area under the curve (AUC): 0.706 vs. 0.661]. Biologic unresponsiveness was an independent prognostic factor for worse survival (hazard ratio 2.31, 95% CI: 1.50-3.55, P <0.001). The modified pathology with BR system demonstrated the highest discriminative ability in predicting 5-year overall survival than the current pathologic system (AUC: 0.785 vs. 0.661, P =0.010) and modified pathologic staging system (AUC: 0.785 vs. 0.706, P =0.002). CONCLUSIONS The prognostic model, incorporating modified ypT staging and elevated carbohydrate antigen 19-9 levels and maximum standardized uptake value simultaneously, demonstrated improved results in predicting oncologic outcomes for patients who underwent surgery following neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Jimin Son
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Woohyung Lee
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Jung Bok Lee
- Department of Clinical Epidemiology and Biostatistics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Kwangpyo Hong
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Min Kyu Sung
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Yejong Park
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Eunsung Jun
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Ki Byung Song
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Dae Wook Hwang
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Jae Hoon Lee
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| | - Song Cheol Kim
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, University of Ulsan College of Medicine, Asan Medical Center
| |
Collapse
|
7
|
Okano N, Kawai M, Ueno M, Yu X, Inoue Y, Takahashi S, Wang W, Takahashi H, Okamura Y, Morinaga S, Matsumoto I, Shimizu Y, Yoshida K, Yamamoto T, Ohtsuka M, Inokawa Y, Nara S, Tamura J, Shinoda S, Yamamoto K, Yamaue H, Furuse J. Outcomes of patients with initially unresectable pancreatic cancer who underwent conversion surgery after FOLFIRINOX or gemcitabine plus nab-paclitaxel chemotherapy: A multicenter retrospective cohort study (PC-CURE-1). JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:816-829. [PMID: 39150050 PMCID: PMC11589395 DOI: 10.1002/jhbp.12066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
BACKGROUND The efficacy and safety of conversion surgery (CS) after FOLFIRINOX or gemcitabine plus nab-paclitaxel (GnP) chemotherapy in patients with initially unresectable pancreatic cancer (PC) remains unclear. METHODS This multicenter retrospective cohort study enrolled patients, between 2014 and 2018, with initially locally advanced or metastatic PC who were considered candidates for CS following FOLFIRINOX or GnP chemotherapy. They were classified into surgery (207 patients [194 resection and 13 exploratory laparotomy only]) and continued chemotherapy (10 patients, control) groups. The primary endpoint was overall survival (OS) from the day of diagnosis of potentially curative resection on imaging studies, with an expected hazard ratio (HR) of 0.7. RESULTS OS in the surgery group was longer than that in the control group (HR, 0.47; 95% confidence interval [CI]: 0.24-0.93). The median OS was 34.4 (95% CI: 27.9-43.4) and 19.8 (95% CI: 14.9-31.1) months in the surgery and control groups, respectively. The Clavien-Dindo grade ≥ IIIa postoperative complication and in-hospital mortality rates were 19.6% and 0.5%, respectively. Multivariate analysis revealed that preoperative chemotherapy duration was not associated with OS. CONCLUSIONS CS, following a favorable response to FOLFIRINOX or GnP chemotherapy, improved initially unresectable PC prognosis (specifically, OS), regardless of the chemotherapy duration.
Collapse
Affiliation(s)
- Naohiro Okano
- Department of Medical OncologyKyorin University Faculty of MedicineTokyoJapan
| | - Manabu Kawai
- Second Department of SurgeryWakayama Medical University School of MedicineWakayamaJapan
| | - Makoto Ueno
- Department of GastroenterologyKanagawa Cancer CenterYokohamaJapan
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic SurgeryCancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Shinichiro Takahashi
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Wenquan Wang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Hidenori Takahashi
- Department of Gastroenterological SurgeryOsaka International Cancer InstituteOsakaJapan
| | - Yukiyasu Okamura
- Division of Hepato‐Biliary‐Pancreatic SurgeryShizuoka Cancer Center HospitalShizuokaJapan
| | - Soichiro Morinaga
- Department of Hepato‐Biliary and Pancreatic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Ippei Matsumoto
- Department of SurgeryKindai University Faculty of MedicineOsakasayamaJapan
| | - Yasuhiro Shimizu
- Department of Gastroenterological SurgeryAichi Cancer Center HospitalNagoyaJapan
| | - Kazuhiro Yoshida
- Department of Surgical OncologyGifu University Graduate School of MedicineGifuJapan
| | | | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Yoshikuni Inokawa
- Department of Gastroenterological SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | - Satoshi Nara
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center HospitalTokyoJapan
| | - Jun Tamura
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Satoru Shinoda
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Kouji Yamamoto
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Hiroki Yamaue
- Second Department of SurgeryWakayama Medical University School of MedicineWakayamaJapan
| | - Junji Furuse
- Department of Medical OncologyKyorin University Faculty of MedicineTokyoJapan
- Department of GastroenterologyKanagawa Cancer CenterYokohamaJapan
| | | |
Collapse
|
8
|
Bao QR, Ventin M, Dell'Atti L, Tripepi M, Frigerio I, Butturini G, Crimì F, Scarpa M, Pucciarelli S, Ferrone CR, Spolverato G. Impact of neoadjuvant chemoradiotherapy on pathologic response in pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Pancreatology 2024; 24:1107-1114. [PMID: 39353845 DOI: 10.1016/j.pan.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND The impact of chemoradiotherapy on pathologic response, resection margin, and survival benefit is still debated. The aim of this study was to compare the rate of pathologic complete response (pCR) in surgical resection following neoadjuvant chemotherapy vs. chemoradiotherapy, and secondarily, to compare the rate of R0 resection and Overall Survival (OS). METHODS A systematic review on MEDLINE/PubMed, Embase, Cochrane, Web of Science and Google Scholar was conducted for studies published between 2012 and 2024 (PROSPERO CRD42022341467). All studies reporting clinical outcomes of patients with Pancreatic ductal adenocarcinoma (PDAC) following neoadjuvant therapy were considered eligible for inclusion. A meta-analysis comparing the rate of pCR, R0 resection rate, and 3-year OS following Chemotherapy vs chemoradiotherapy in patients was performed. The overall quality of evidence was evaluated using a GRADE approach. RESULTS Out of 5194 potentially relevant studies, 29 studies were considered eligible for full-text assessment, and 11 studies were included in the systematic review and in the meta-analysis. Of these, five were retrospective single-center, five retrospective multi-center studies, and one was a phase II multi-center RCT. Overall, 1830 Chemotherapy patients and 2299 Chemoradiotherapy patients were included in the meta-analysis. A statistically significant increased rate of pCR and R0 resections were found in chemoradiotherapy patients (OR 3.58, 95 % CI 2.47-5.18, p ≤ 0.00001) (OR 1.49, 95 % CI 1.17-1.90, p = 0.001), whereas 3-year OS (OR 1.07, 95 % CI 0.84-1.36, p = 0.6) did not differ significantly. CONCLUSIONS Chemoradiotherapy may have a positive impact on pathologic response and R0 resection rate, whereas a survival benefit was not reported.
Collapse
Affiliation(s)
- Quoc Riccardo Bao
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, Padova, Italy
| | - Marco Ventin
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lorenzo Dell'Atti
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, Padova, Italy
| | - Marzia Tripepi
- Pancreatic Surgery, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Isabella Frigerio
- Pancreatic Surgery, Pederzoli Hospital, Peschiera del Garda, Verona, Italy; Collegium Medicum, University of Social Sciences, Łodz, Poland.
| | - Giovanni Butturini
- Pancreatic Surgery, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Filippo Crimì
- Institute of Radiology, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Marco Scarpa
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, Padova, Italy
| | - Salvatore Pucciarelli
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, Padova, Italy
| | - Cristina R Ferrone
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gaya Spolverato
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, Padova, Italy
| |
Collapse
|
9
|
Lim AHW, Zobel J, Bills M, Hsieh W, Crouch B, Joshi R, Thomson JE, Neo E, Kuan LL, Safaeian R, Tse E, Rayner CK, Ruszkiewicz A, Singhal N, Bartholomeusz D, Nguyen NQ. The Impact of Combined Chemotherapy and Intra-Tumoural Injection of Phosphorus-32 Microparticles on Vascularity in Locally Advanced Pancreatic Carcinoma. Cancers (Basel) 2024; 16:3412. [PMID: 39410031 PMCID: PMC11475738 DOI: 10.3390/cancers16193412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Poor intra-tumoural vascularity contributes to a lack of response to chemotherapy in pancreatic cancers. Preliminary data suggest that the addition of endoscopic ultrasound (EUS)-guided intra-tumoural injection of phosphorus-32 (32P) microparticles to standard chemotherapy is potentially beneficial in locally advanced pancreatic cancer (LAPC). We aimed to assess changes in pancreatic tumour vascularity following 32P implantation, using contrast-enhanced EUS (CE-EUS). METHODS This was a prospective single-centre trial from January 2022 to 2024 of patients with unresectable, non-metastatic LAPC undergoing standard FOLFIRINOX chemotherapy and 32P implantation. We performed CE-EUS pre-implantation after two chemotherapy cycles and 4 and 12 weeks after implantation. Time-intensity curves were analysed for 90 s after IV contrast bolus to ascertain peak intensity and intensity gain. RESULTS A total of 20 patients underwent 32P implantation, with 15 completing 12-week follow-up. The technical success of 32P implantation was 100%. The median primary tumour size reduced from 32 mm (IQR 27.5-38.75) pre-implantation to 24 mm (IQR 16-26) 12 weeks post-implantation (p < 0.001). Five patients (25%) had tumour downstaging, and four underwent resections. The baseline (pre-implantation, post-chemotherapy) median intensity gain of contrast enhancement within the tumour was 32.15 (IQR 18.08-54.35). This increased to 46.85 (IQR 35.05-76.6; p = 0.007) and 66.3 (IQR 54.7-76.3; p = 0.001) at 4 weeks and 12 weeks post-implantation, respectively. Over a median follow-up of 11.2 months (IQR 7.8-12.8), 15/20 (75%) of patients remained alive, with 3/20 (15%) demonstrating local disease progression. Overall survival was not significantly different between patients with or without an increased intensity of 10 a.u. or more at 12 weeks post-implantation. CONCLUSION This is the first clinical study to demonstrate treatment-induced increased vascularity within pancreatic primary tumours, which followed 32P implantation and FOLFIRINOX chemotherapy. Larger comparative trials are warranted.
Collapse
Affiliation(s)
- Amanda Huoy Wen Lim
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| | - Joshua Zobel
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
| | - Madison Bills
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - William Hsieh
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - Benjamin Crouch
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - Rohit Joshi
- Medical Oncology, Lyell McEwin Hospital, Adelaide, SA 5112, Australia;
| | - John-Edwin Thomson
- Department of Hepatobiliary Surgery, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (J.-E.T.); (E.N.)
| | - EuLing Neo
- Department of Hepatobiliary Surgery, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (J.-E.T.); (E.N.)
| | - Li Lian Kuan
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital, Adelaide, SA 5011, Australia;
| | - Romina Safaeian
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| | - Christopher K. Rayner
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| | - Andrew Ruszkiewicz
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
- Surgical Pathology, SA Pathology, Adelaide, SA 5000, Australia
- Centre of Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia
| | - Nimit Singhal
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
- Oncology, Royal Adelaide Hospital, Adelaide, SA 5081, Australia
| | - Dylan Bartholomeusz
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - Nam Quoc Nguyen
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| |
Collapse
|
10
|
Barenboim A, Mercer D, Sahnan K, Gaffan A, Goren O, Halperin S, Brazowski E, Pelles Avraham S, Klausner JM, Lubezky N. The Relationship between Treatment Response and Overall Survival in Borderline, Non-Resectable and Resectable Pancreatic Cancer Patients Treated with Neoadjuvant FOLFIRINOX. J Clin Med 2024; 13:5206. [PMID: 39274419 PMCID: PMC11396552 DOI: 10.3390/jcm13175206] [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: 07/28/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Background: The National Comprehensive Cancer Network (NCCN)-recommended treatment for patients with borderline-resectable pancreatic cancer (BRPC) and locally advanced pancreatic cancer (LAPC) involves a combination of neoadjuvant FOLFIRINOX chemotherapy and the curative surgical resection of the tumor. This study seeks to identify the clinical, radiological, laboratory, and pathologic predictors that can anticipate the oncological outcomes of patients. Methods: In this study, we conducted a retrospective analysis of patients who had undergone curative surgical resection for BRPC, LAPC, or resectable disease with high-risk features after receiving neoadjuvant FOLFIRINOX at two institutions. We evaluated by means of multivariate analysis whether clinical and laboratory response, tumor markers, radiological response, and pathologic tumor response grade correlated with overall survival (OS) and disease-free survival (DFS). Results: The study enrolled a total of 70 patients with BRPC, LAPC, and resectable disease with high-risk features who underwent resection after neoadjuvant FOLFIRINOX. Age above 65 years and fewer than nine cycles of chemotherapy (OR 4.2; 95% CI 1.4-12.0; p-value 0.007); locally advanced tumors after neoadjuvant treatment (NAT) (OR 7.0; 95% CI 1.9-25.7; p-value 0.003); and lymph node disease and histological tumor regression grade 2 and 3 (OR 4.3; 95% CI 0.9-19.2; p-value 0.05) were risk factors linked to adverse OS and DFS. The median OS and DFS were 33 (22-43.9) months and 16.5 (11.3-21.6) months, respectively. Conclusions: Classification as a LA tumor after NAT was the only preoperative radiological factor that predicted adverse survival in patients undergoing curative surgery after NAT. Other clinical, biochemical, and radiological measures of response were not found to predict OS. Patient age, the cumulative administration of more than eight cycles of chemotherapy, and a significant pathological response were associated with better OS. The results of this study are important for treatment decision-making and prognostication in patients with BRPC and LAPC.
Collapse
Affiliation(s)
- Alex Barenboim
- Department of Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Diego Mercer
- Department of Radiology, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| | | | - Alex Gaffan
- Department of Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Or Goren
- Institute of Anesthesiology, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Sharon Halperin
- Institute of Oncology, Sheba Medical Center, Ramat Gan 5262000, Israel
| | - Eli Brazowski
- Institute of Pathology, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Sharon Pelles Avraham
- Institute of Oncology, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Joseph M Klausner
- Department of Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Nir Lubezky
- Department of HPB and Transplant Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
11
|
Ma H, Esfahani SA, Krishna S, Ataeinia B, Zhou IY, Rotile NJ, Weigand-Whittier J, Boice AT, Liss AS, Tanabe KK, Caravan P. Allysine-Targeted Molecular MRI Enables Early Prediction of Chemotherapy Response in Pancreatic Cancer. Cancer Res 2024; 84:2549-2560. [PMID: 38759082 PMCID: PMC11293968 DOI: 10.1158/0008-5472.can-23-3548] [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: 11/13/2023] [Revised: 02/20/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Neoadjuvant therapy is routinely used in pancreatic ductal adenocarcinoma (PDAC), but not all tumors respond to this treatment. Current clinical imaging techniques are not able to precisely evaluate and predict the response to neoadjuvant therapies over several weeks. A strong fibrotic reaction is a hallmark of a positive response, and during fibrogenesis, allysine residues are formed on collagen proteins by the action of lysyl oxidases. Here, we report the application of an allysine-targeted molecular MRI probe, MnL3, to provide an early, noninvasive assessment of treatment response in PDAC. Allysine increased 2- to 3-fold after one dose of neoadjuvant therapy with FOLFIRINOX in sensitive human PDAC xenografts in mice. Molecular MRI with MnL3 could specifically detect and quantify fibrogenesis in PDAC xenografts. Comparing the MnL3 signal before and 3 days after one dose of FOLFIRINOX predicted subsequent treatment response. The MnL3 tumor signal increased by 70% from day 0 to day 3 in mice that responded to subsequent doses of FOLFIRINOX, whereas no signal increase was observed in FOLFIRINOX-resistant tumors. This study indicates the promise of allysine-targeted molecular MRI as a noninvasive tool to predict chemotherapy outcomes. Significance: Allysine-targeted molecular MRI can quantify fibrogenesis in pancreatic tumors and predict response to chemotherapy, which could guide rapid clinical management decisions by differentiating responders from nonresponders after treatment initiation.
Collapse
Affiliation(s)
- Hua Ma
- Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Shadi A. Esfahani
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts 02129, United States
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Shriya Krishna
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Bahar Ataeinia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts 02129, United States
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Iris Y. Zhou
- Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Nicholas J. Rotile
- Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Jonah Weigand-Whittier
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Avery T. Boice
- Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Andrew S. Liss
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, Massachusetts, 02114, United States
| | - Kenneth K. Tanabe
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Peter Caravan
- Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts 02129, United States
| |
Collapse
|
12
|
Zhang Z, Guo S, Cheng C, Cao K, Jiang H, Jin G, Zuo C. Integrated 68 Ga-FAPI-04 PET/MR in Pancreatic Cancer : Prediction of Tumor Response and Tumor Resectability After Neoadjuvant Therapy. Clin Nucl Med 2024; 49:715-721. [PMID: 38914015 DOI: 10.1097/rlu.0000000000005300] [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: 06/26/2024]
Abstract
PURPOSE This study aimed to investigate the value of 68 Ga-fibroblast activation protein inhibitor (FAPI) PET/MR semiquantitative parameters in the prediction of tumor response and resectability after neoadjuvant therapy in patients with pancreatic cancer. PATIENTS AND METHODS This study was performed retrospectively in patients with borderline resectable or locally advanced pancreatic cancer who underwent 68 Ga-FAPI PET/MRI from June 2020 to June 2022. The SUV max , SUV mean , SUV peak , uptake tumor volume (UTV), and total lesion FAP expression (TLF) of the primary tumor were recorded. The target-to-background ratios (TBRs) of the primary tumor to normal tissue muscle (TBR muscle ) and blood (TBR blood ) were also calculated. In addition, the minimum apparent diffusion coefficient value of the tumor was measured. After 3-4 cycles of gemcitabine + nab-paclitaxel chemotherapy, patients were divided into responders and nonresponders groups according to RECIST criteria (v.1.1). They were also divided into resectable and unresectable groups according to the surgical outcome. The variables were compared separately between groups. RESULTS A total of 18 patients who met the criteria were included in this study. The UTV and TLF were significantly higher in nonresponders than in responders ( P < 0.05). The SUV max , SUV mean , and TBR muscle were significantly higher in unresectable patients than in resectable ones ( P < 0.05). Receiver operating characteristic curve analysis identified UTV (area under the curve [AUC] = 0.840, P = 0.015) and TLF (AUC = 0.877, P = 0.007) as significant predictors for the response to gemcitabine + nab-paclitaxel chemotherapy, with cutoff values of 25.05 and 167.38, respectively. In addition, SUV max (AUC = 0.838, P = 0.016), SUV mean (AUC = 0.812, P = 0.026), and TBR muscle (AUC = 0.787, P = 0.041) were significant predictors of the resectability post-NCT, with cutoff values of 14.0, 6.0, and 13.9, respectively. According to logistic regression analysis, TLF was found to be significantly associated with tumor response ( P = 0.032) and was an independent predictor of tumor response ( P = 0.032). In addition, apparent diffusion coefficient value was an independent predictor of tumor resectability ( P = 0.043). CONCLUSIONS This pilot study demonstrates the value of 68 Ga-FAPI PET/MR for the prediction of tumor response and resectability after neoadjuvant therapy. It may aid in individualized patient management by guiding the treatment regimens.
Collapse
Affiliation(s)
- Zeyu Zhang
- From the Departments of Nuclear Medicine
| | | | - Chao Cheng
- From the Departments of Nuclear Medicine
| | | | - Hui Jiang
- Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Gang Jin
- Hepatobiliary Pancreatic Surgery
| | | |
Collapse
|
13
|
Qin C, Li T, Lin C, Zhao B, Li Z, Zhao Y, Wang W. The systematic role of pancreatic cancer exosomes: distant communication, liquid biopsy and future therapy. Cancer Cell Int 2024; 24:264. [PMID: 39054529 PMCID: PMC11271018 DOI: 10.1186/s12935-024-03456-5] [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: 05/02/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal diseases worldwide. Cancer-derived exosomes, benefiting from the protective role of the lipid membrane, exhibit remarkable stability in the circulatory system. These exosomes, released by tumor microenvironment, contain various biomolecules such as proteins, RNAs, and lipids that plays a pivotal role in mediating distant communication between the local pancreatic tumor and other organs or tissues. They facilitate the transfer of oncogenic factors to distant sites, contributing to the compromised body immune system, distant metastasis, diabetes, cachexia, and promoting a microenvironment conducive to tumor growth and metastasis in pancreatic cancer patients. Beyond their intrinsic roles, circulating exosomes in peripheral blood can be detected to facilitate accurate liquid biopsy. This approach offers a novel and promising method for the diagnosis and management of pancreatic cancer. Consequently, circulating exosomes are not only crucial mediators of systemic cell-cell communication during pancreatic cancer progression but also hold great potential as precise tools for pancreatic cancer management and treatment. Exosome-based liquid biopsy and therapy represent promising advancements in the diagnosis and treatment of pancreatic cancer. Exosomes can serve as drug delivery vehicles, enhancing the targeting and efficacy of anticancer treatments, modulating the immune system, and facilitating gene editing to suppress tumor growth. Ongoing research focuses on biomarker identification, drug delivery systems, and clinical trials to validate the safety and efficacy of exosome-based therapies, offering new possibilities for early diagnosis and precision treatment in pancreatic cancer. Leveraging the therapeutic potential of exosomes, including their ability to deliver targeted drugs and modulate immune responses, opens new avenues for innovative treatment strategies.
Collapse
Affiliation(s)
- Cheng Qin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianyu Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Lin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bangbo Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeru Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yutong Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weibin Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Badheeb AM, Awad MA, Al Masad AG, Alyami MS, Fagihi MA, Al Walani M, Alkarak S, Al Bahili HM, Alatawi A, Nagi NM, Madbouly AR, Abu Bakar A, Ahmed F, Badheeb M. Pancreatic Cancer: A Retrospective Study From the Najran Region of Saudi Arabia. Cureus 2024; 16:e65685. [PMID: 39205701 PMCID: PMC11357715 DOI: 10.7759/cureus.65685] [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: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Despite advances in treatment, pancreatic cancer frequently has a low survival rate due to its advanced-stage diagnosis. Treatment focuses on prolonging survival and maintaining quality of life. This study investigates the characteristics associated with survival in advanced pancreatic cancer patients treated at a single academic cancer center in Najran, Saudi Arabia. METHOD A retrospective chart review study covering the period January 1, 2015, and December 31, 2023, involved 80 adult patients with pathologically confirmed pancreatic cancer (ductal adenocarcinoma) at King Khalid Hospital in Najran, Saudi Arabia. Clinicopathological characteristics, therapy, response, and survival outcomes were all gathered and analyzed. The chi-squared test, Kaplan-Meier, and Cox proportional hazards method with hazard ratios (HR) and 95% confidence intervals (CI) were used for statistical analysis. RESULT The mean age was 65.7±14.1 years and 54 (67.5%) cases were male. The main symptom was abdominal pain (n=54, 67.5%), while jaundice was presented in 17 (21.2%) of cases. The baseline serum carbohydrate antigen 19-9 (CA 19-9) level varied among cases, with 35 (43.8%) having normal levels. The majority of cases (n=59, 73.8%) had distant metastases at the initial presentation, while 12 cases (15%) had localized disease (resectable), and 22 (27.5%) were locally advanced at the first presentation. The most commonly reported pathologic grade was poorly differentiated ductal adenocarcinoma in 39 (48.8%). FOLFIRINOX was used as first-line chemotherapy in 54 (67.5%) cases, while gemcitabine alone was used in 15 (18.8%) cases. First-line chemotherapy resulted in progressive disease in 30 (37.5%), stable disease in 30 (37.5%), and partial response in 14 (17.5%). With a mean follow-up time of 14.8±8.6 months, 57 (71.2%) were dead, where the main cause of death was disease progression (n=51, 89.5%). The median overall survival was 13.5 months, with a 12-month survival rate of 56% and a 36-month survival rate of 17%. The median cancer-specific survival was 16 months (95% CI: 13-22 months). The 12-month median cancer-specific survival was 61% (95% CI: 51-73%), and the 36-month median cancer-specific survival was 19% (95% CI: 10-34%). In univariate analysis, initial metastasis presentation (HR: 35.46; 95% CI: 4.90-256.83, p<0.001), poor Eastern Cooperative Oncology Group Performance Status (ECOG-PS) (3-4) (HR: 2.34; 95% CI:1.34-4.09, p=0.003), and presence of multiple metastases (HR: 1.33; 95% CI: 1.09-1.62, p=0.004) were associated with worsened survival. Patients who received the first chemotherapy were associated with better survival (HR: 0.53; 95% CI: 0.29-0.98, p=0.043). Furthermore, the response rate in patients who received FOLFIRINOX was better than that of those who received gemcitabine alone, which was statistically significant (p=0.002). CONCLUSION Our study showed that initial metastatic presentation, poor ECOG-PS, and the occurrence of numerous metastases were all linked with poor survival of patients with pancreatic adenocarcinoma. Additionally, FOLFIRINOX as a first-line treatment showed better survival rates than gemcitabine alone. Raising awareness among healthcare providers on the alarming signs of pancreatic cancer and the introduction of personalized oncology might improve the outcome of this fatal malignancy.
Collapse
Affiliation(s)
- Ahmed M Badheeb
- Oncology, King Khalid Hospital-Oncology Center, Najran, SAU
- Medicine, Hadhramaut University, Mukalla, YEM
| | | | - Ali G Al Masad
- Gastroenterology and Hepatology, King Khalid Hospital, Najran, SAU
| | | | | | - Mugahed Al Walani
- Gastroenterology and Advanced Endoscopy, King Khalid Hospital, Najran, SAU
| | | | - Hamad M Al Bahili
- Hepatobiliary Surgery, Prince Sultan Military Medical City, Riyadh, SAU
| | | | | | - Ahmed R Madbouly
- Gastroenterology and Hepatology, King Khalid Hospital, Najran, SAU
| | | | | | - Mohamed Badheeb
- Internal Medicine, Yale New Haven Health, Bridgeport Hospital, Bridgeport, USA
| |
Collapse
|
15
|
Saúde-Conde R, El Ghali B, Navez J, Bouchart C, Van Laethem JL. Total Neoadjuvant Therapy in Localized Pancreatic Cancer: Is More Better? Cancers (Basel) 2024; 16:2423. [PMID: 39001485 PMCID: PMC11240662 DOI: 10.3390/cancers16132423] [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: 05/15/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in oncology due to its advanced stage upon diagnosis and limited treatment options. Surgical resection, the primary curative approach, often results in poor long-term survival rates, leading to the exploration of alternative strategies like neoadjuvant therapy (NAT) and total neoadjuvant therapy (TNT). While NAT aims to enhance resectability and overall survival, there appears to be potential for improvement, prompting consideration of alternative neoadjuvant strategies integrating full-dose chemotherapy (CT) and radiotherapy (RT) in TNT approaches. TNT integrates chemotherapy and radiotherapy prior to surgery, potentially improving margin-negative resection rates and enabling curative resection for locally advanced cases. The lingering question: is more always better? This article categorizes TNT strategies into six main groups based on radiotherapy (RT) techniques: (1) conventional chemoradiotherapy (CRT), (2) the Dutch PREOPANC approach, (3) hypofractionated ablative intensity-modulated radiotherapy (HFA-IMRT), and stereotactic body radiotherapy (SBRT) techniques, which further divide into (4) non-ablative SBRT, (5) nearly ablative SBRT, and (6) adaptive ablative SBRT. A comprehensive analysis of the literature on TNT is provided for both borderline resectable pancreatic cancer (BRPC) and locally advanced pancreatic cancer (LAPC), with detailed sections for each.
Collapse
Affiliation(s)
- Rita Saúde-Conde
- Digestive Oncology Department, Hôpitaux Universitaires de Bruxelles (HUB), Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Benjelloun El Ghali
- Department of Radiation Oncology, Hôpitaux Universitaires de Bruxelles (HUB), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (B.E.G.); (C.B.)
| | - Julie Navez
- Department of Abdominal Surgery and Transplantation, Hôpitaux Universitaires de Bruxelles (HUB), Hopital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Christelle Bouchart
- Department of Radiation Oncology, Hôpitaux Universitaires de Bruxelles (HUB), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (B.E.G.); (C.B.)
| | - Jean-Luc Van Laethem
- Digestive Oncology Department, Hôpitaux Universitaires de Bruxelles (HUB), Université Libre de Bruxelles, 1070 Brussels, Belgium;
| |
Collapse
|
16
|
Ozaki M, Kageyama K, Kimura K, Eguchi S, Yamamoto A, Tanaka R, Nota T, Yonezawa H, Nishiofuku H, Sakai Y, Tani N, Jogo A, Terai M, Sato T, Ishizawa T, Miki Y. A rat-based preclinical platform facilitating transcatheter hepatic arterial infusion in immunodeficient rats with liver xenografts of patient-derived pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:10529. [PMID: 38719893 PMCID: PMC11079078 DOI: 10.1038/s41598-024-61142-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
Liver metastases from pancreatic ductal adenocarcinoma (PDAC) are highly fatal. A rat-based patient-derived tumor xenograft (PDX) model is available for transcatheter therapy. This study aimed to create an immunodeficient rat model with liver xenografts of patient-derived primary PDAC and evaluate efficacy of hepatic arterial infusion chemotherapy with cisplatin in this model. Three patient-derived PDACs were transplanted into the livers of 21 rats each (totally, 63 rats), randomly assigned into hepatic arterial infusion, systemic venous infusion, and control groups (n = 7 each) four weeks post-implantation. Computed tomography evaluated tumor volumes before and four weeks after treatment. Post-euthanasia, resected tumor specimens underwent histopathological examination. A liver-implanted PDAC PDX rat model was established in all 63 rats, with first CT identifying all tumors. Four weeks post-treatment, arterial infusion groups exhibited significantly smaller tumor volumes than controls for all three tumors on second CT. Xenograft tumors histologically maintained adenocarcinoma features compared to original patient tumors. Ki67 expression was significantly lower in arterial infusion groups than in the other two for the three tumors, indicating reduced tumor growth in PDX rats. A liver-implanted PDAC PDX rat model was established as a rat-based preclinical platform. Arterial cisplatin infusion chemotherapy represents a potential therapy for PDAC liver metastasis.
Collapse
Affiliation(s)
- Masanori Ozaki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Ken Kageyama
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan.
| | - Kenjiro Kimura
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Shinpei Eguchi
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Akira Yamamoto
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Ryota Tanaka
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Takehito Nota
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Hiroki Yonezawa
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Hideyuki Nishiofuku
- Department of Diagnostic and Interventional Radiology, Nara Medical University, 840 Shijocho, Kashihara, Nara, 6348521, Japan
| | - Yuki Sakai
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Naoki Tani
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Atsushi Jogo
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Mizue Terai
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, 1024 Curtis Building, Philadelphia, PA, 19107, USA
| | - Takami Sato
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, 1024 Curtis Building, Philadelphia, PA, 19107, USA
| | - Takeaki Ishizawa
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Yukio Miki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| |
Collapse
|
17
|
van Eijck CWF, Sabroso-Lasa S, Strijk GJ, Mustafa DAM, Fellah A, Koerkamp BG, Malats N, van Eijck CHJ. A liquid biomarker signature of inflammatory proteins accurately predicts early pancreatic cancer progression during FOLFIRINOX chemotherapy. Neoplasia 2024; 49:100975. [PMID: 38335839 PMCID: PMC10873733 DOI: 10.1016/j.neo.2024.100975] [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: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is often treated with FOLFIRINOX, a chemotherapy associated with high toxicity rates and variable efficacy. Therefore, it is crucial to identify patients at risk of early progression during treatment. This study aims to explore the potential of a multi-omics biomarker for predicting early PDAC progression by employing an in-depth mathematical modeling approach. METHODS Blood samples were collected from 58 PDAC patients undergoing FOLFIRINOX before and after the first cycle. These samples underwent gene (GEP) and inflammatory protein expression profiling (IPEP). We explored the predictive potential of exclusively IPEP through Stepwise (Backward) Multivariate Logistic Regression modeling. Additionally, we integrated GEP and IPEP using Bayesian Kernel Regression modeling, aiming to enhance predictive performance. Ultimately, the FOLFIRINOX IPEP (FFX-IPEP) signature was developed. RESULTS Our findings revealed that proteins exhibited superior predictive accuracy than genes. Consequently, the FFX-IPEP signature consisted of six proteins: AMN, BANK1, IL1RL2, ITGB6, MYO9B, and PRSS8. The signature effectively identified patients transitioning from disease control to progression early during FOLFIRINOX, achieving remarkable predictive accuracy with an AUC of 0.89 in an independent test set. Importantly, the FFX-IPEP signature outperformed the conventional CA19-9 tumor marker. CONCLUSIONS Our six-protein FFX-IPEP signature holds solid potential as a liquid biomarker for the early prediction of PDAC progression during toxic FOLFIRINOX chemotherapy. Further validation in an external cohort is crucial to confirm the utility of the FFX-IPEP signature. Future studies should expand to predict progression under different chemotherapies to enhance the guidance of personalized treatment selection in PDAC.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.
| | - Sergio Sabroso-Lasa
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), Madrid, Spain
| | - Gaby J Strijk
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dana A M Mustafa
- Department of Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Amine Fellah
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), Madrid, Spain
| | - Casper H J van Eijck
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.
| |
Collapse
|
18
|
Stoop TF, Theijse RT, Seelen LWF, Groot Koerkamp B, van Eijck CHJ, Wolfgang CL, van Tienhoven G, van Santvoort HC, Molenaar IQ, Wilmink JW, Del Chiaro M, Katz MHG, Hackert T, Besselink MG. Preoperative chemotherapy, radiotherapy and surgical decision-making in patients with borderline resectable and locally advanced pancreatic cancer. Nat Rev Gastroenterol Hepatol 2024; 21:101-124. [PMID: 38036745 DOI: 10.1038/s41575-023-00856-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/05/2023] [Indexed: 12/02/2023]
Abstract
Surgical resection combined with systemic chemotherapy is the cornerstone of treatment for patients with localized pancreatic cancer. Upfront surgery is considered suboptimal in cases with extensive vascular involvement, which can be classified as either borderline resectable pancreatic cancer or locally advanced pancreatic cancer. In these patients, FOLFIRINOX or gemcitabine plus nab-paclitaxel chemotherapy is currently used as preoperative chemotherapy and is eventually combined with radiotherapy. Thus, more patients might reach 5-year overall survival. Patient selection for chemotherapy, radiotherapy and subsequent surgery is based on anatomical, biological and conditional parameters. Current guidelines and clinical practices vary considerably regarding preoperative chemotherapy and radiotherapy, response evaluation, and indications for surgery. In this Review, we provide an overview of the clinical evidence regarding disease staging, preoperative therapy, response evaluation and surgery in patients with borderline resectable pancreatic cancer or locally advanced pancreatic cancer. In addition, a clinical work-up is proposed based on the available evidence and guidelines. We identify knowledge gaps and outline a proposed research agenda.
Collapse
Affiliation(s)
- Thomas F Stoop
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam, Netherlands
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Rutger T Theijse
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Leonard W F Seelen
- Department of Surgery, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht and St. Antonius Hospital Nieuwegein, Utrecht, Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Christopher L Wolfgang
- Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York City, NY, USA
| | - Geertjan van Tienhoven
- Cancer Center Amsterdam, Amsterdam, Netherlands
- Amsterdam UMC, location University of Amsterdam, Department of Radiation Oncology, Amsterdam, Netherlands
| | - Hjalmar C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht and St. Antonius Hospital Nieuwegein, Utrecht, Netherlands
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht and St. Antonius Hospital Nieuwegein, Utrecht, Netherlands
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Amsterdam, Netherlands
- Amsterdam UMC, location University of Amsterdam, Department of Medical Oncology, Amsterdam, Netherlands
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Marc G Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands.
- Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
19
|
van Schelt AS, Gottwald LM, Wassenaar NPM, Runge JH, Sinkus R, Stoker J, Nederveen AJ, Schrauben EM. Single Breath-Hold MR Elastography for Fast Biomechanical Probing of Pancreatic Stiffness. J Magn Reson Imaging 2024; 59:688-698. [PMID: 37194646 DOI: 10.1002/jmri.28773] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) stromal disposition is thought to influence chemotherapy efficacy and increase tissue stiffness, which could be quantified noninvasively via MR elastography (MRE). Current methods cause position-based errors in pancreas location over time, hampering accuracy. It would be beneficial to have a single breath-hold acquisition. PURPOSE To develop and test a single breath-hold three-dimensional MRE technique utilizing prospective undersampling and a compressed sensing reconstruction (CS-MRE). STUDY TYPE Prospective. POPULATION A total of 30 healthy volunteers (HV) (31 ± 9 years; 33% male) and five patients with PDAC (69 ± 5 years; 80% male). FIELD STRENGTH/SEQUENCE 3-T, GRE Ristretto MRE. ASSESSMENT First, optimization of multi breath-hold MRE was done in 10 HV using four combinations of vibration frequency, number of measured wave-phase offsets, and TE and looking at MRE quality measures in the pancreas head. Second, viscoelastic parameters delineated in the pancreas head or tumor of CS-MRE were compared against (I) 2D and (II) 3D four breath-hold acquisitions in HV (N = 20) and PDAC patients. Intrasession repeatability was assessed for CS-MRE in a subgroup of healthy volunteers (N = 15). STATISTICAL TESTS Tests include repeated measures analysis of variance (ANOVA), Bland-Altman analysis, and coefficients of variation (CoVs). A P-value <.05 was considered statistically significant. RESULTS Optimization of the four breath-hold acquisitions resulted in 40 Hz vibration frequency, five wave-phases, and echo time (TE) = 6.9 msec as the preferred method (4BH-MRE). CS-MRE quantitative results did not differ from 4BH-MRE. Shear wave speed (SWS) and phase angle differed significantly between HV and PDAC patients using 4BH-MRE or CS-MRE. The limits of agreement for SWS were [-0.09, 0.10] m/second and the within-subject CoV was 4.8% for CS-MRE. DATA CONCLUSION CS-MRE might allow a single breath-hold MRE acquisition with comparable SWS and phase angle as 4BH-MRE, and it may still enable to differentiate between HV and PDAC. LEVEL OF EVIDENCE 2 Technical Efficacy Stage: 2.
Collapse
Affiliation(s)
- Anne-Sophie van Schelt
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Lukas M Gottwald
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nienke P M Wassenaar
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jurgen H Runge
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ralph Sinkus
- Imaging Sciences and Biomedical Engineering, Kings College London, London, UK
- Department of Radiology, Université de Paris, Paris, France
| | - Jaap Stoker
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Endocrinology, Amsterdam Gastroenterology, Amsterdam, The Netherlands
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eric M Schrauben
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Tavakkoli A, Beauchamp A, Prasad T, Zhu H, Singal AG, Elmunzer BJ, Kubiliun NM, Kwon RS, Hughes AE, Pruitt SL. Accessibility to ERCP-performing hospitals among patients with pancreatic cancer living in SEER regions. Cancer Med 2024; 13:e7020. [PMID: 38400670 PMCID: PMC10891451 DOI: 10.1002/cam4.7020] [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: 10/12/2023] [Revised: 01/15/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND AND AIMS The two most common interventions used to treat painless jaundice from pancreatic cancer are endoscopic retrograde cholangiopancreatography (ERCP) and percutaneous transhepatic biliary drainage (PTBD). Our study aimed to characterize the geographic distribution of ERCP-performing hospitals among patients with pancreatic cancer in the United States and the association between geographic accessibility to ERCP-performing hospitals and biliary interventions patients receive. METHODS This is a retrospective cohort study using the Surveillance, Epidemiology, and End Results (SEER)-Medicare database for pancreatic cancer from 2005 to 2013. Multilevel models were used to examine the association between accessibility to ERCP hospitals within a 30- and 45-min drive from the patient's residential ZIP Code and the receipt of ERCP treatment. A two-step floating catchment area model was used to calculate the measure of accessibility based on the distribution across SEER regions. RESULTS 7464 and 782 patients underwent ERCP and PTBD, respectively, over the study period. There were 808 hospitals in which 8246 patients diagnosed with pancreatic cancer in SEER regions from 2005 to 2013 received a procedure. Patients with high accessibility within both 30- and 45-min drive to an ERCP-performing hospital were more likely to receive an ERCP (30-min adjusted odds ratio [aOR]: 1.53, 95% confidence interval [CI]: 1.17-2.01; 45-min aOR: 1.31, 95% CI: 1.01-1.70). Furthermore, in the adjusted model, Black patients (vs. White) and patients with stage IV disease were less likely to receive ERCP than PTBD. CONCLUSIONS Patients with pancreatic cancer and high accessibility to an ERCP-performing hospital were more likely to receive ERCP. Disparities in the receipt of ERCP persisted for Black patients regardless of their access to ERCP-performing hospitals.
Collapse
Affiliation(s)
- Anna Tavakkoli
- Division of Gastroenterology & Liver Diseases, Department of MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Alaina Beauchamp
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Tanushree Prasad
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Hong Zhu
- Department of Public Health SciencesUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Amit G. Singal
- Division of Gastroenterology & Liver Diseases, Department of MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical CenterDallasTexasUSA
| | - B. Joseph Elmunzer
- Division of Gastroenterology and Hepatology, Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Nisa M. Kubiliun
- Division of Gastroenterology & Liver Diseases, Department of MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Richard S. Kwon
- Division of Gastroenterology, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Amy E. Hughes
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical CenterDallasTexasUSA
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Sandi L. Pruitt
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical CenterDallasTexasUSA
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
21
|
Rompen IF, Habib JR, Wolfgang CL, Javed AA. Anatomical and Biological Considerations to Determine Resectability in Pancreatic Cancer. Cancers (Basel) 2024; 16:489. [PMID: 38339242 PMCID: PMC10854859 DOI: 10.3390/cancers16030489] [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: 11/14/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains associated with poor outcomes with a 5-year survival of 12% across all stages of the disease. These poor outcomes are driven by a delay in diagnosis and an early propensity for systemic dissemination of the disease. Recently, aggressive surgical approaches involving complex vascular resections and reconstructions have become more common, thus allowing more locally advanced tumors to be resected. Unfortunately, however, even after the completion of surgery and systemic therapy, approximately 40% of patients experience early recurrence of disease. To determine resectability, many institutions utilize anatomical staging systems based on the presence and extent of vascular involvement of major abdominal vessels around the pancreas. However, these classification systems are based on anatomical considerations only and do not factor in the burden of systemic disease. By integrating the biological criteria, we possibly could avoid futile resections often associated with significant morbidity. Especially patients with anatomically resectable disease who have a heavy burden of radiologically undetected systemic disease most likely do not derive a survival benefit from resection. On the contrary, we could offer complex resections to those who have locally advanced or oligometastatic disease but have favorable systemic biology and are most likely to benefit from resection. This review summarizes the current literature on defining anatomical and biological resectability in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Ingmar F. Rompen
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Joseph R. Habib
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
| | - Christopher L. Wolfgang
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
| | - Ammar A. Javed
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
22
|
Yan X, Fu X, Gui Y, Chen X, Cheng Y, Dai M, Wang W, Xiao M, Tan L, Zhang J, Shao Y, Wang H, Chang X, Lv K. Development and validation of a nomogram model based on pretreatment ultrasound and contrast-enhanced ultrasound to predict the efficacy of neoadjuvant chemotherapy in patients with borderline resectable or locally advanced pancreatic cancer. Cancer Imaging 2024; 24:13. [PMID: 38245789 PMCID: PMC10800053 DOI: 10.1186/s40644-024-00662-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
OBJECTIVES To develop a nomogram using pretreatment ultrasound (US) and contrast-enhanced ultrasound (CEUS) to predict the clinical response of neoadjuvant chemotherapy (NAC) in patients with borderline resectable pancreatic cancer (BRPC) or locally advanced pancreatic cancer (LAPC). METHODS A total of 111 patients with pancreatic ductal adenocarcinoma (PDAC) treated with NAC between October 2017 and February 2022 were retrospectively enrolled. The patients were randomly divided (7:3) into training and validation cohorts. The pretreatment US and CEUS features were reviewed. Univariate and multivariate logistic regression analyses were used to determine the independent predictors of clinical response in the training cohort. Then a prediction nomogram model based on the independent predictors was constructed. The area under the curve (AUC), calibration plot, C-index and decision curve analysis (DCA) were used to assess the nomogram's performance, calibration, discrimination and clinical benefit. RESULTS The multivariate logistic regression analysis showed that the taller-than-wide shape in the longitudinal plane (odds ratio [OR]:0.20, p = 0.01), time from injection of contrast agent to peak enhancement (OR:3.64; p = 0.05) and Peaktumor/ Peaknormal (OR:1.51; p = 0.03) were independent predictors of clinical response to NAC. The predictive nomogram developed based on the above imaging features showed AUCs were 0.852 and 0.854 in the primary and validation cohorts, respectively. Good calibration was achieved in the training datasets, with C-index of 0.852. DCA verified the clinical usefulness of the nomogram. CONCLUSIONS The nomogram based on pretreatment US and CEUS can effectively predict the clinical response of NAC in patients with BRPC and LAPC; it may help guide personalized treatment.
Collapse
Affiliation(s)
- Xiaoyi Yan
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xianshui Fu
- Department of Ultrasound, No.304 Hospital of Chinese PLA, Beijing, 100037, China
| | - Yang Gui
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xueqi Chen
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weibin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Mengsu Xiao
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Tan
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jing Zhang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuming Shao
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Huanyu Wang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ke Lv
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
23
|
van Eijck CWF, Strijk G, Vietsch EE, van der Sijde F, Verheij M, Mustafa DAM, Vink M, Aerts JGJV, van Eijck CHJ, Willemsen M. FOLFIRINOX chemotherapy modulates the peripheral immune landscape in pancreatic cancer: Implications for combination therapies and early response prediction. Eur J Cancer 2024; 196:113440. [PMID: 37988843 DOI: 10.1016/j.ejca.2023.113440] [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: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND FOLFIRINOX chemotherapy has improved outcomes for pancreatic cancer patients, but poor long-term survival outcomes and high toxicity remain challenges. This study investigates the impact of FOLFIRINOX on plasma proteins and peripheral immune cells to guide immune-based combination therapies and, ideally, to identify a potential biomarker to predict early disease progression during FOLFIRINOX. METHODS Blood samples were collected from 86 pancreatic cancer patients before and two weeks after the first FOLFIRINOX cycle and subjected to comprehensive immune cell and proteome profiling. Principal Component Analysis and Linear Mixed Effect Regression models were used for data analysis. FOLFIRINOX efficacy was radiologically evaluated after the fourth cycle. RESULTS One cycle of FOLFIRINOX diminished tumour-cell-related pathways and enhanced pathways related to immune activation, illustrated by an increase in pro-inflammatory IL-18, IL-15, and TNFRSF4. Similarly, FOLFIRINOX promoted the activation of CD4 + and CD8 + T cells, the proliferation of NK(T), and the activation of antigen-presenting cells. Furthermore, high pre-treatment levels of VEGFA and PRDX3 and an elevation in FCRL3 levels after one cycle predicted early progression under FOLFIRINOX. Finally, patients with progressive disease exhibited high levels of inhibitory markers on B cells and CD8 + T cells, while responding patients exhibited high levels of activation markers on CD4 + and CD8 + T cell subsets. CONCLUSION FOLFIRINOX has immunomodulatory effects, providing a foundation for clinical trials exploring immune-based combination therapies that harness the immune system to treat pancreatic cancer. In addition, several plasma proteins hold potential as circulating predictive biomarkers for early prediction of FOLFIRINOX response in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Gaby Strijk
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Fleur van der Sijde
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Maaike Verheij
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Madelief Vink
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Joachim G J V Aerts
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Marcella Willemsen
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
24
|
Pourali G, Donyadideh G, Mehrabadi S, Hamid F, Hassanian SM, Ferns GA, Khazaei M, Avan A. Clinical practice guidelines for interventional treatment of pancreatic cancer. RECENT ADVANCES IN NANOCARRIERS FOR PANCREATIC CANCER THERAPY 2024:345-373. [DOI: 10.1016/b978-0-443-19142-8.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
25
|
Kanda T, Wakiya T, Ishido K, Kimura N, Nagase H, Yoshida E, Nakagawa J, Matsuzaka M, Niioka T, Sasaki Y, Hakamada K. Noninvasive Computed Tomography-Based Deep Learning Model Predicts In Vitro Chemosensitivity Assay Results in Pancreatic Cancer. Pancreas 2024; 53:e55-e61. [PMID: 38019604 DOI: 10.1097/mpa.0000000000002270] [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] [Indexed: 12/01/2023]
Abstract
OBJECTIVES We aimed to predict in vitro chemosensitivity assay results from computed tomography (CT) images by applying deep learning (DL) to optimize chemotherapy for pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS Preoperative enhanced abdominal CT images and the histoculture drug response assay (HDRA) results were collected from 33 PDAC patients undergoing surgery. Deep learning was performed using CT images of both the HDRA-positive and HDRA-negative groups. We trimmed small patches from the entire tumor area. We established various prediction labels for HDRA results with 5-fluorouracil (FU), gemcitabine (GEM), and paclitaxel (PTX). We built a predictive model using a residual convolutional neural network and used 3-fold cross-validation. RESULTS Of the 33 patients, effective response to FU, GEM, and PTX by HDRA was observed in 19 (57.6%), 11 (33.3%), and 23 (88.5%) patients, respectively. The average accuracy and the area under the receiver operating characteristic curve (AUC) of the model for predicting the effective response to FU were 93.4% and 0.979, respectively. In the prediction of GEM, the models demonstrated high accuracy (92.8%) and AUC (0.969). Likewise, the model for predicting response to PTX had a high performance (accuracy, 95.9%; AUC, 0.979). CONCLUSIONS Our CT patch-based DL model exhibited high predictive performance in projecting HDRA results. Our study suggests that the DL approach could possibly provide a noninvasive means for the optimization of chemotherapy.
Collapse
Affiliation(s)
- Taishu Kanda
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Taiichi Wakiya
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Keinosuke Ishido
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Norihisa Kimura
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Hayato Nagase
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Eri Yoshida
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | | | | | | | - Yoshihiro Sasaki
- Medical Informatics, Hirosaki University Hospital, Hirosaki, Japan
| | - Kenichi Hakamada
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| |
Collapse
|
26
|
Dang HX, Saha D, Jayasinghe R, Zhao S, Coonrod E, Mudd J, Goedegebuure S, Fields R, Ding L, Maher C. Single-cell transcriptomics reveals long noncoding RNAs associated with tumor biology and the microenvironment in pancreatic cancer. NAR Cancer 2023; 5:zcad055. [PMID: 38023733 PMCID: PMC10664695 DOI: 10.1093/narcan/zcad055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly heterogeneous and lethal. Long noncoding RNAs (lncRNAs) are an important class of genes regulating tumorigenesis and progression. Prior bulk transcriptomic studies in PDAC have revealed the dysregulation of lncRNAs but lack single-cell resolution to distinguish lncRNAs in tumor-intrinsic biology and the tumor microenvironment (TME). We analyzed single-cell transcriptome data from 73 multiregion samples in 21 PDAC patients to evaluate lncRNAs associated with intratumoral heterogeneity and the TME in PDAC. We found 111 cell-specific lncRNAs that reflected tumor, immune and stromal cell contributions, associated with outcomes, and validated across orthogonal datasets. Single-cell analysis of tumor cells revealed lncRNAs associated with TP53 mutations and FOLFIRINOX treatment that were obscured in bulk tumor analysis. Lastly, tumor subcluster analysis revealed widespread intratumor heterogeneity and intratumoral lncRNAs associated with cancer hallmarks and tumor processes such as angiogenesis, epithelial-mesenchymal transition, metabolism and immune signaling. Intratumoral subclusters and lncRNAs were validated across six datasets and showed clinically relevant associations with patient outcomes. Our study provides the first comprehensive assessment of the lncRNA landscape in PDAC using single-cell transcriptomic data and can serve as a resource, PDACLncDB (accessible at https://www.maherlab.com/pdaclncdb-overview), to guide future functional studies.
Collapse
Affiliation(s)
- Ha X Dang
- Department of Internal Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO 63108, USA
| | - Debanjan Saha
- Department of Internal Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- MD–PhD Program, Washington University in St Louis, St Louis, MO 63110, USA
| | - Reyka Jayasinghe
- Department of Internal Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Sidi Zhao
- Department of Internal Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Emily Coonrod
- Department of Internal Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Jacqueline Mudd
- Department of Surgery, Washington University in St Louis, St Louis, MO 63110, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University in St Louis, St Louis, MO 63110, USA
| | - Ryan Fields
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO 63110, USA
- Department of Surgery, Washington University in St Louis, St Louis, MO 63110, USA
| | - Li Ding
- Department of Internal Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO 63110, USA
| | - Christopher A Maher
- Department of Internal Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO 63108, USA
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO 63130, USA
| |
Collapse
|
27
|
Shi S, Guo D, Ye L, Li T, Fei Q, Lin M, Yu X, Jin K, Wu W. Knockdown of TACC3 inhibits tumor cell proliferation and increases chemosensitivity in pancreatic cancer. Cell Death Dis 2023; 14:778. [PMID: 38012214 PMCID: PMC10682013 DOI: 10.1038/s41419-023-06313-x] [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: 07/09/2023] [Revised: 11/06/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant digestive tract tumor with limited clinical treatments. Transforming acidic coiled-coil-containing protein 3 (TACC3) is a component of the centrosome axis and a member of the TACC family, which affect mitosis and regulate chromosome stability and are involved in tumor development and progression. However, the role of TACC3 in PDAC remains elusive. In this study, by exploiting the TCGA database, we found that high TACC3 expression in PDAC is associated with poor prognosis. shRNA-mediated TACC3 knockdown caused S phase arrest of the cell cycle and inhibited proliferation in PDAC cell lines. Through RNA sequencing and protein co-immunoprecipitation combined with mass spectrometry, KIF11 was identified as a protein that interacts with TACC3. TACC3 stabilizes and regulates KIF11 protein expression levels in PDAC cells through physical interaction. Knockdown of TACC3 or KIF11 resulted in abnormal spindle formation during cell division both in vitro and in vivo. Pharmacological inhibition of TACC3 or KIF11 can suppress tumor cell proliferation and promote apoptosis. Our studies further demonstrated that high expression of TACC3 and KIF11 mediated the resistance of PDAC to gemcitabine, and deficiency of TACC3 or KIF11 increased the sensitivity of PDAC cells to chemotherapy. In conclusion, our study reveals the fundamental role of TACC3 expression in PDAC cell proliferation and chemoresistance, suggesting that TACC3 can be used as a molecular marker to evaluate the prognosis of PDAC.
Collapse
Affiliation(s)
- Saimeng 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
| | - Duancheng Guo
- 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
| | - Longyun Ye
- 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
| | - Tianjiao 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
| | - Qinglin Fei
- 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
| | - Mengxiong Lin
- 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.
| | - Kaizhou Jin
- 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.
| | - Weiding Wu
- 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
|
28
|
Talbot A, Talbot T, Shaughnessy E, Glass A, Das A, Watanabe Y, Cheng D, Johansson M, Rao S, Yusoff I, Tang C, White R, Dean A. Overall survival and treatment modalities in pancreatic adenocarcinoma: a retrospective analysis of a single centre in Western Australia. J Gastrointest Oncol 2023; 14:2221-2228. [PMID: 37969823 PMCID: PMC10643600 DOI: 10.21037/jgo-23-488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 11/17/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of approximately 10.7% in Australia. It is becoming an increasingly common cause of cancer mortality. The therapeutic model for PDAC remains limited, especially for those with metastatic disease on presentation. Methods We completed a retrospective cohort study including all patients with PDAC presenting between April 2008 and October 2021 to St. John of God Subiaco Hospital in Western Australia. Overall survival (OS) was calculated via Kaplan-Meier method. Results We identified 251 patients treated for PDAC. Of these, 134 patients (53%) had resectable, borderline resectable or locally advanced (LA) disease at diagnosis and 117 patients (47%) had metastatic disease. The median age of all patients was 66 years (range, 25-87 years). OS in PDAC was 26 months [95% confidence interval (CI): 23-30]. In the non-metastatic group OS was 34 months (95% CI: 30-39). In the metastatic group OS was 19 months (95% CI: 14-22). Treatment modalities varied between patients. Overall 123 patients were treated with chemotherapy alone, 55 patients had chemoradiotherapy, 34 patients had chemotherapy and surgery and 37 had tri-modality treatment including chemotherapy, surgery and radiotherapy. Two patients received cyberknife radiation alone. Conclusions This retrospective study shows a significant prolonged survival for PDAC patients. Further studies are needed to validate second- and third-line regimens in PDAC.
Collapse
Affiliation(s)
- Alice Talbot
- Department of Oncology, St. John of God Hospital, Subiaco, WA, Australia
| | - Thomas Talbot
- Department of Oncology, St. John of God Hospital, Subiaco, WA, Australia
| | - Emma Shaughnessy
- Department of Oncology, St. John of God Hospital, Subiaco, WA, Australia
| | - Aisling Glass
- Department of Oncology, St. John of God Hospital, Subiaco, WA, Australia
| | - Adarsh Das
- Department of Oncology, St. John of God Hospital, Subiaco, WA, Australia
| | - Yuki Watanabe
- Department of Surgery, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Daniel Cheng
- Department of Gastroenterology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Mikael Johansson
- Department of Surgery, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Samarth Rao
- Department of Gastroenterology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Ian Yusoff
- Department of Gastroenterology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Colin Tang
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Rohen White
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Andrew Dean
- Department of Oncology, St. John of God Hospital, Subiaco, WA, Australia
| |
Collapse
|
29
|
Wang L, Fu D, Weng S, Xu H, Liu L, Guo C, Ren Y, Liu Z, Han X. Genome-scale CRISPR-Cas9 screening stratifies pancreatic cancer with distinct outcomes and immunotherapeutic efficacy. Cell Signal 2023; 110:110811. [PMID: 37468054 DOI: 10.1016/j.cellsig.2023.110811] [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: 04/05/2023] [Revised: 07/02/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
Pancreatic cancer (PC) was featured by dramatic heterogeneity and dismal outcomes. An ideal classification strategy capable of achieving risk stratification and individualized treatment is urgently needed to significantly improve prognosis. In this study, using the 105 prognostic cancer essential genes identified by genome-scale CRISPR-Cas9 screening and univariate Cox analysis, we established and verified three heterogeneous subtypes via non-negative matrix factorization (NMF) and nearest template prediction (NTP) algorithms in the TCGA-PAAD cohort (176 samples) and four multi-center cohorts (233 samples), respectively. Among them, C1 with the worst prognosis was enriched in immune-related pathways, possessed superior infiltration abundance of immune cells and immune checkpoint molecules expression, and might be most sensitive to immunotherapy. C3, owing a moderate prognosis, might be featured by proliferative biological function, and despite its highest immunogenicity, the defects in antigen processing and presentation ability coupled with barren immune environment render it ineffective for immunotherapy, while it had potential sensitivity to paclitaxel and methotrexate. Besides, C2 harbored the best prognosis and was characterized by metabolism-related functions. These results could deepen our understanding of PC molecular heterogeneity and provide a trustworthy reference for prognostic stratification management and precision medicine in clinical practice.
Collapse
Affiliation(s)
- Libo Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Deshuang Fu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China; Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| |
Collapse
|
30
|
Saito K, Michihata N, Hamada T, Jo T, Matsui H, Fushimi K, Nakai Y, Yasunaga H, Fujishiro M. Gemcitabine plus nab-paclitaxel for pancreatic cancer and interstitial lung disease: A nationwide longitudinal study. Cancer Sci 2023; 114:3996-4005. [PMID: 37547944 PMCID: PMC10551588 DOI: 10.1111/cas.15910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/30/2023] [Accepted: 07/09/2023] [Indexed: 08/08/2023] Open
Abstract
Interstitial lung disease (ILD) is an adverse event associated with gemcitabine administration. Gemcitabine plus nab-paclitaxel, which is now a first-line chemotherapy regimen for pancreatic cancer (PC), may increase the risk of ILD; however, large-scale clinical data on this are limited. Thus, this study aimed to elucidate the incidence and risk factors of ILD in patients with PC receiving gemcitabine plus nab-paclitaxel. Through the Diagnosis Procedure Combination database, a Japanese nationwide inpatient database with outpatient data, we identified consecutive patients with PC who received gemcitabine-based chemotherapy between July 2010 and March 2019 at 205 hospitals. Competing-risk analysis was used to examine the cumulative incidence and risk factors of ILD. Among the 6163 patients who received gemcitabine plus nab-paclitaxel, we documented 168 patients (2.7%) who developed ILD with cumulative incidence rates (95% confidence intervals [CIs]) of 2.0% (1.6%-2.4%), 2.7% (2.2%-3.1%), and 3.1% (2.6%-3.6%) at 3, 6, and 12 months, respectively. Compared with patients with PC who received gemcitabine monotherapy, those who received gemcitabine plus nab-paclitaxel had an adjusted subdistribution hazard ratio (SHR) for ILD of 1.93 (95% CI: 1.51-2.47). Older age was associated with a high risk of ILD in patients receiving gemcitabine plus nab-paclitaxel (adjusted SHR comparing ≥75 to ≤74 years, 1.61; 95% CI: 1.16-2.24). In conclusion, this study demonstrated the clinical course of gemcitabine plus nab-paclitaxel-associated ILD in patients with PC. When gemcitabine plus nab-paclitaxel is administered to elderly patients with PC, symptoms associated with ILD must be monitored.
Collapse
Affiliation(s)
- Kei Saito
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Division of Gastroenterology and Hepatology, Department of Internal MedicineNihon University School of MedicineTokyoJapan
| | - Nobuaki Michihata
- Department of Health Services Research, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Tsuyoshi Hamada
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Hepato‐Biliary‐Pancreatic MedicineThe Cancer Institute Hospital of Japanese Foundation for Cancer ResearchTokyoJapan
| | - Taisuke Jo
- Department of Health Services Research, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Respiratory Medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroki Matsui
- Department of Clinical Epidemiology and Health Economics, School of Public HealthThe University of TokyoTokyoJapan
| | - Kiyohide Fushimi
- Department of Health Informatics and Policy, Graduate School of MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Yousuke Nakai
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Endoscopy and Endoscopic SurgeryThe University of Tokyo HospitalTokyoJapan
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public HealthThe University of TokyoTokyoJapan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
31
|
Wang K, Hua X, Fu X, Hao Z, Jiao A, Li S. Petite Integration Factor 1 knockdown enhances gemcitabine sensitivity in pancreatic cancer cells via increasing DNA damage. J Appl Toxicol 2023; 43:1522-1532. [PMID: 37183367 DOI: 10.1002/jat.4494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/18/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
Chemoresistance is still a vital obstacle in various tumors chemotherapy. This study aimed to explore the role of Petite Integration Factor 1 (PIF1) in the sensitivity of gemcitabine response to pancreatic cancer cells. Gene Expression Profiling Interactive Analysis (GEPIA) database was employed for evaluating the level of PIF1 in pancreatic cancer tissues and normal tissues. The mRNA level of PIF1 was detected via reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis. The relative protein expression of PIF1, cleaved caspase-3, and phosphorylated histone H2Ax (γH2Ax) was assessed through western blot. Cell viability and apoptosis were assessed via Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. Moreover, lactate dehydrogenase (LDH) release and caspase-3 activity were determined via the corresponding LDH Cytotoxicity Assay Kit and caspase-3 colorimetric assay kit. PIF1 expression was upregulated in pancreatic cancer tissues and cells. Knockdown of PIF1 exhibited the repressive impact on the viability of AsPC-1 and PANC-1 cells. PIF1 knockdown enhanced LDH release and apoptosis in both AsPC-1 and PANC-1 cells. PIF1 downregulation could augment the sensitivity of gemcitabine in pancreatic cancer cells, as evidenced by lower cell viability and higher LDH release and apoptosis rate after knocking down PIF1 in gemcitabine-treated pancreatic cancer cells relative to pancreatic cancer cells treated with gemcitabine alone. Moreover, PIF1 knockdown increased γH2Ax protein expression and DNA damage, and gemcitabine treatment-induced DNA damage in AsPC-1 and PANC-1 cells was exacerbated by PIF1 silencing. Furthermore, gemcitabine treatment-caused increase of DNA damage was alleviated by PIF1 overexpression; whereas, this effect of PIF1 upregulation was reversed by thymidine, a DNA synthesis inhibitor. In addition, the decreased gemcitabine sensitivity response to pancreatic cancer cells caused by PIF1 upregulation was also hindered by thymidine treatment. In conclusion, PIF1 silencing enhanced gemcitabine sensitivity response to pancreatic cancer cells through aggrandizing DNA damage.
Collapse
Affiliation(s)
- Kun Wang
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangdong Hua
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xibo Fu
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhiqiang Hao
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ao Jiao
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Siyuan Li
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
32
|
Zhou B, Zhang SR, Chen G, Chen P. Developments and challenges in neoadjuvant therapy for locally advanced pancreatic cancer. World J Gastroenterol 2023; 29:5094-5103. [PMID: 37744290 PMCID: PMC10514760 DOI: 10.3748/wjg.v29.i35.5094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/19/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a significant public health challenge and is currently the fourth leading cause of cancer-related mortality in developed countries. Despite advances in cancer treatment, the 5-year survival rate for patients with PDAC remains less than 5%. In recent years, neoadjuvant therapy (NAT) has emerged as a promising treatment option for many cancer types, including locally advanced PDAC, with the potential to improve patient outcomes. To analyze the role of NAT in the setting of locally advanced PDAC over the past decade, a systematic literature search was conducted using PubMed and Web of Science. The results suggest that NAT may reduce the local mass size, promote tumor downstaging, and increase the likelihood of resection. These findings are supported by the latest evidence-based medical literature and the clinical experience of our center. Despite the potential benefits of NAT, there are still challenges that need to be addressed. One such challenge is the lack of consensus on the optimal timing and duration of NAT. Improved criteria for patient selection are needed to further identify PDAC patients likely to respond to NAT. In conclusion, NAT has emerged as a promising treatment option for locally advanced PDAC. However, further research is needed to optimize its use and to better understand the role of NAT in the management of this challenging disease. With continued advances in cancer treatment, there is hope of improving the outcomes of patients with PDAC in the future.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Shi-Ran Zhang
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Geng Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ping Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
33
|
Su YY, Chao YJ, Wang CJ, Liao TK, Su PJ, Huang CJ, Chiang NJ, Yu YT, Tsai HM, Chen LT, Shan YS. The experience of neoadjuvant chemotherapy versus upfront surgery in resectable pancreatic cancer: a cross sectional study. Int J Surg 2023; 109:2614-2623. [PMID: 37300888 PMCID: PMC10498854 DOI: 10.1097/js9.0000000000000495] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Upfront resection (UR) followed by adjuvant chemotherapy remains the standard treatment for resectable pancreatic cancer. There is increasing evidence suggesting favourable outcomes toward neoadjuvant chemotherapy (NAC) followed by surgery. METHODS All clinical staging with resectable pancreatic cancer patients treated at a tertiary medical centre from 2013 to 2020 were identified. The baseline characteristics, treatment course, surgery outcome and survival results of UR or NAC were compared. RESULTS Finally, in 159 resectable patients, 46 patients (29%) underwent NAC and 113 patients (71%) received UR. In NAC, 11 patients (24%) did not receive resection, 4 (36.4%) for comorbidity, 2 (18.2%) for patient refusal and 2 (18.2%) for disease progression. In UR, 13 patients (12%) were unresectable intraoperatively; 6 (46.2%) for locally advanced and 5 (38.5%) for distant metastasis. Overall, 97% of patients in NAC and 58% of patients in UR completed adjuvant chemotherapy. As of data cut-off, 24 patients (69%) in NAC and 42 patients (29%) in UR were still tumour free. The median recurrence-free survival in NAC, UR with adjuvant chemotherapy and without adjuvant chemotherapy were 31.3 months (95% CI, 14.4-not estimable), 10.6 months (95% CI, 9.0-14.3) and 8.5 months (95% CI, 5.8-11.8), P =0.036; and the median overall survival in each group were not reached (95% CI, 29.7-not estimable), 25.9 months (95% CI, 21.1-40.5) and 21.7 months (12.0-32.8), P =0.0053. Based on initial clinical staging, the median overall survival of NAC was not significantly different from UR with a tumour less than or equal to 2 cm, P =0.29. NAC patients had a higher R0 resection rate (83% versus 53%), lower recurrence rate (31% versus 71%) and harvested median number lymph node (23 versus 15). CONCLUSION This study demonstrates that NAC is superior to UR in resectable pancreatic cancer with better survival.
Collapse
Affiliation(s)
- Yung-Yeh Su
- Departments of Oncology
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University
- National Institute of Cancer Research, National Health Research Institute, Tainan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | - Chih-Jung Wang
- Surgery
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University
| | - Ting-Kai Liao
- Surgery
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University
| | | | - Chien-Jui Huang
- Internal Medicine
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institute, Tainan
- Department of Oncology, Taipei Veterans General Hospital, Taipei
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
| | | | - Hong-Ming Tsai
- Medical Imaging, National Cheng-Kung University Hospital
| | - Li-Tzong Chen
- Departments of Oncology
- National Institute of Cancer Research, National Health Research Institute, Tainan
- Department of Internal Medicine, Kaohsiung Medical University Hospital
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yan-Shen Shan
- Surgery
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University
| |
Collapse
|
34
|
Zhu Z, Tang H, Ying J, Cheng Y, Wang X, Wang Y, Bai C. Efficacy and safety of gemcitabine plus S-1 vs. gemcitabine plus nab-paclitaxel in treatment-naïve advanced pancreatic ductal adenocarcinoma. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0189. [PMID: 37646237 PMCID: PMC10618946 DOI: 10.20892/j.issn.2095-3941.2023.0189] [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: 05/27/2023] [Accepted: 08/03/2023] [Indexed: 09/01/2023] Open
Abstract
OBJECTIVE Gemcitabine plus nab-paclitaxel (GnP) is the standard first-line therapy for advanced pancreatic ductal adenocarcinoma (PDAC). S-1, an oral fluoropyrimidine derivative, as compared with gemcitabine, is non-inferior in terms of overall survival (OS) and is associated with lower hematologic toxicity. Accordingly, S-1 is a convenient oral alternative treatment for advanced PDAC. This study was aimed at comparing the efficacy and safety of gemcitabine plus S-1 (GS) vs. GnP as first-line chemotherapy for advanced PDAC. METHODS Patients with advanced PDAC who received first-line GS or GnP at the Peking Union Medical College Hospital between March 2011 and November 2022 were evaluated. RESULTS A total of 300 patients were assessed, of whom 84 received GS and 216 received GnP. The chemotherapy completion rate was higher with GS than GnP (50.0% vs. 30.3%, P = 0.0028). The objective response rate (ORR) was slightly higher (14.3% vs. 9.7%, P = 0.35), and the median OS was significantly longer (17.9 months vs. 13.3 months, P = 0.0078), in the GS group than the GnP group. However, the median progression-free survival (PFS) did not significantly differ between groups. Leukopenia risk was significantly lower in the GS group than the GnP group (14.9% vs. 28.1%, P = 0.049). CONCLUSIONS As first-line chemotherapy for advanced PDAC, the GS regimen led to a significantly longer OS than the GnP regimen. The PFS, ORR, and incidence of severe adverse events were comparable between the GS and GnP groups.
Collapse
Affiliation(s)
- Zhou Zhu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Hui Tang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jinrong Ying
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiang Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
35
|
Eshmuminov D, Aminjonov B, Palm RF, Malleo G, Schmocker RK, Abdallah R, Yoo C, Shaib WL, Schneider MA, Rangelova E, Choi YJ, Kim H, Rose JB, Patel S, Wilson GC, Maloney S, Timmermann L, Sahora K, Rössler F, Lopez-Lopez V, Boyer E, Maggino L, Malinka T, Park JY, Katz MHG, Prakash L, Ahmad SA, Helton S, Jang JY, Hoffe SE, Salvia R, Taieb J, He J, Clavien PA, Held U, Lehmann K. FOLFIRINOX or Gemcitabine-based Chemotherapy for Borderline Resectable and Locally Advanced Pancreatic Cancer: A Multi-institutional, Patient-Level, Meta-analysis and Systematic Review. Ann Surg Oncol 2023; 30:4417-4428. [PMID: 37020094 PMCID: PMC10250524 DOI: 10.1245/s10434-023-13353-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/01/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Pancreatic cancer often presents as locally advanced (LAPC) or borderline resectable (BRPC). Neoadjuvant systemic therapy is recommended as initial treatment. It is currently unclear what chemotherapy should be preferred for patients with BRPC or LAPC. METHODS We performed a systematic review and multi-institutional meta-analysis of patient-level data regarding the use of initial systemic therapy for BRPC and LAPC. Outcomes were reported separately for tumor entity and by chemotherapy regimen including FOLFIRINOX (FIO) or gemcitabine-based. RESULTS A total of 23 studies comprising 2930 patients were analyzed for overall survival (OS) calculated from the beginning of systemic treatment. OS for patients with BRPC was 22.0 months with FIO, 16.9 months with gemcitabine/nab-paclitaxel (Gem/nab), 21.6 months with gemcitabine/cisplatin or oxaliplatin or docetaxel or capecitabine (GemX), and 10 months with gemcitabine monotherapy (Gem-mono) (p < 0.0001). In patients with LAPC, OS also was higher with FIO (17.1 months) compared with Gem/nab (12.5 months), GemX (12.3 months), and Gem-mono (9.4 months; p < 0.0001). This difference was driven by the patients who did not undergo surgery, where FIO was superior to other regimens. The resection rates for patients with BRPC were 0.55 for gemcitabine-based chemotherapy and 0.53 with FIO. In patients with LAPC, resection rates were 0.19 with Gemcitabine and 0.28 with FIO. In resected patients, OS for patients with BRPC was 32.9 months with FIO and not different compared to Gem/nab, (28.6 months, p = 0.285), GemX (38.8 months, p = 0.1), or Gem-mono (23.1 months, p = 0.083). A similar trend was observed in resected patients converted from LAPC. CONCLUSIONS In patients with BRPC or LAPC, primary treatment with FOLFIRINOX compared with Gemcitabine-based chemotherapy appears to provide a survival benefit for patients that are ultimately unresectable. For patients that undergo surgical resection, outcomes are similar between GEM+ and FOLFIRINOX when delivered in the neoadjuvant setting.
Collapse
Affiliation(s)
- Dilmurodjon Eshmuminov
- Department of Surgery and Transplantation, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Botirjon Aminjonov
- Department of Surgery and Transplantation, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Russell F Palm
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Giuseppe Malleo
- Unit of General and Pancreatic Surgery. Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
| | - Ryan K Schmocker
- Department of Surgery, The Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Raëf Abdallah
- Hepatogastroenterology and Gastrointestinal Oncology Department, Hôpital Européen Georges-Pompidou, AGEO (Association des Gastro-Enterologues Oncologues), Université de Paris, SIRIC CARPEM, Paris, France
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Walid L Shaib
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Marcel André Schneider
- Department of Surgery and Transplantation, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Elena Rangelova
- Department of Upper Gastrointestinal Diseases, Karolinska University Hospital and Department of Clinical Science, Intervention, and Technology (CLINTEC) at Karolinska Institute, Stockholm, Sweden
- Department of Surgery, The Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yoo Jin Choi
- Department of Surgery, Seoul National University College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul, 110-744, Korea
| | - Hongbeom Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul, 110-744, Korea
| | - J Bart Rose
- Division of Surgical Oncology, Pancreatobiliary Disease Center at UAB, The University of Alabama at Birmingham, Birmingham, USA
| | - Sameer Patel
- Division of Surgical Oncology, Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Gregory C Wilson
- Division of Surgical Oncology, Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Sarah Maloney
- Department of Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Lea Timmermann
- Department of Surgery, Charité - Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Klaus Sahora
- Departments of Surgery and Comprehensive Cancer Center, University of Vienna, Medical University of Vienna, Vienna, Austria
| | - Fabian Rössler
- Department of Surgery and Transplantation, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Víctor Lopez-Lopez
- Department of General, Visceral and Transplantation Surgery, Clinic and University Hospital Virgen de la Arrixaca, IMIB-ARRIXACA, Murcia, Spain
| | - Emanuel Boyer
- University of South Florida School of Medicine, Tampa, FL, USA
| | - Laura Maggino
- Unit of General and Pancreatic Surgery. Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
| | - Thomas Malinka
- Department of Surgery, Charité - Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | | | - Laura Prakash
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Syed A Ahmad
- Division of Surgical Oncology, Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Scott Helton
- Section of General, Thoracic and Vascular Surgery, Department of Surgery, Virginia Mason Medical Center, Seattle, WA, USA
| | - Jin-Young Jang
- Department of Surgery, Seoul National University College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul, 110-744, Korea
| | - Sarah E Hoffe
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Roberto Salvia
- Unit of General and Pancreatic Surgery. Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
| | - Julien Taieb
- Hepatogastroenterology and Gastrointestinal Oncology Department, Hôpital Européen Georges-Pompidou, AGEO (Association des Gastro-Enterologues Oncologues), Université de Paris, SIRIC CARPEM, Paris, France
| | - Jin He
- Department of Surgery, The Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Pierre-Alain Clavien
- Department of Surgery and Transplantation, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Ulrike Held
- Department of Biostatistics at Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, 8001, Zurich, Switzerland
| | - Kuno Lehmann
- Department of Surgery and Transplantation, University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
36
|
Eshmuminov D, Aminjonov B, Palm RF, Lehmann K. ASO Author Reflections: Chemotherapy Regimen in Borderline Resectable and Locally Advanced Pancreatic Cancer-Resection Cuts the Deal. Ann Surg Oncol 2023; 30:4429-4430. [PMID: 37074518 PMCID: PMC10250261 DOI: 10.1245/s10434-023-13484-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 04/20/2023]
Affiliation(s)
- Dilmurodjon Eshmuminov
- Department of Surgery and Transplantation, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Botirjon Aminjonov
- Department of Surgery and Transplantation, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Russell F Palm
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kuno Lehmann
- Department of Surgery and Transplantation, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Cassese G, Han HS, Yoon YS, Lee JS, Lee B, Cubisino A, Panaro F, Troisi RI. Role of neoadjuvant therapy for nonmetastatic pancreatic cancer: Current evidence and future perspectives. World J Gastrointest Oncol 2023; 15:911-924. [PMID: 37389109 PMCID: PMC10302990 DOI: 10.4251/wjgo.v15.i6.911] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/17/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is one of the most common and lethal human cancers worldwide. Surgery followed by adjuvant chemotherapy offers the best chance of a long-term survival for patients with PDAC, although only approximately 20% of the patients have resectable tumors when diagnosed. Neoadjuvant chemotherapy (NACT) is recommended for borderline resectable pancreatic cancer. Several studies have investigated the role of NACT in treating resectable tumors based on the recent advances in PDAC biology, as NACT provides the potential benefit of selecting patients with favorable tumor biology and controls potential micro-metastases in high-risk patients with resectable PDAC. In such challenging cases, new potential tools, such as ct-DNA and molecular targeted therapy, are emerging as novel therapeutic options that may improve old paradigms. This review aims to summarize the current evidence regarding the role of NACT in treating non-metastatic pancreatic cancer while focusing on future perspectives in light of recent evidence.
Collapse
Affiliation(s)
- Gianluca Cassese
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive HPB Surgery and Transplantation Service, Federico II University Hospital, Naples 80131, Italy
| | - Ho-Seong Han
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Jun Suh Lee
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Boram Lee
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Antonio Cubisino
- Department of HPB Surgery and Transplantation, Beaujon Hospital, Clichy 92110, France
| | - Fabrizio Panaro
- Department of Digestive Surgery and Liver Transplantation, CHU Montpellier, Montpellier 34100, France
| | - Roberto Ivan Troisi
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive HPB Surgery and Transplantation Service, Federico II University Hospital, Naples 80131, Italy
| |
Collapse
|
38
|
Chen EY, Kardosh A, Nabavizadeh N, Foster B, Mayo SC, Billingsley KG, Gilbert EW, Lanciault C, Grossberg A, Bensch KG, Maynard E, Anderson EC, Sheppard BC, Thomas CR, Lopez CD, Vaccaro GM. Phase 2 study of preoperative chemotherapy with nab-paclitaxel and gemcitabine followed by chemoradiation for borderline resectable or node-positive pancreatic ductal adenocarcinoma. Cancer Med 2023; 12:12986-12995. [PMID: 37132281 PMCID: PMC10315770 DOI: 10.1002/cam4.5971] [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: 12/28/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Neoadjuvant treatment with nab-paclitaxel and gemcitabine for potentially operable pancreatic adenocarcinoma has not been well studied in a prospective interventional trial and could down-stage tumors to achieve negative surgical margins. METHODS A single-arm, open-label phase 2 trial (NCT02427841) enrolled patients with pancreatic adenocarcinoma deemed to be borderline resectable or clinically node-positive from March 17, 2016 to October 5, 2019. Patients received preoperative gemcitabine 1000 mg/m2 and nab-paclitaxel 125 mg/m2 on Days 1, 8, 15, every 28 days for two cycles followed by chemoradiation with 50.4 Gy intensity-modulated radiation over 28 fractions with concurrent fluoropyrimidine chemotherapy. After definitive resection, patients received four additional cycles of gemcitabine and nab-paclitaxel. The primary endpoint was R0 resection rate. Other endpoints included treatment completion rate, resection rate, radiographic response rate, survival, and adverse events. RESULTS Nineteen patients were enrolled, with the majority having head of pancreas primary tumors, both arterial and venous vasculature involvement, and clinically positive nodes on imaging. Among them, 11 (58%) underwent definitive resection and eight of 19 (42%) achieved R0 resection. Disease progression and functional decline were primary reasons for deferring surgical resection after neoadjuvant treatment. Pathologic near-complete response was observed in two of 11 (18%) resection specimens. Among the 19 patients, the 12-month progression-free survival was 58%, and 12-month overall survival was 79%. Common adverse events were alopecia, nausea, vomiting, fatigue, myalgia, peripheral neuropathy, rash, and neutropenia. CONCLUSION Gemcitabine and nab-paclitaxel followed by long-course chemoradiation represents a feasible neoadjuvant treatment strategy for borderline resectable or node-positive pancreatic cancer.
Collapse
Affiliation(s)
- Emerson Y. Chen
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Adel Kardosh
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Nima Nabavizadeh
- Department of Radiation MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Bryan Foster
- Department of Diagnostic RadiologyOregon Health & Science UniversityPortlandOregonUSA
| | - Skye C. Mayo
- Division of Surgical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | | | - Erin W. Gilbert
- Division of Gastrointestinal and General SurgeryOregon Health & Science UniversityPortlandOregonUSA
| | | | - Aaron Grossberg
- Department of Radiation MedicineOregon Health & Science UniversityPortlandOregonUSA
| | | | | | - Eric C. Anderson
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Brett C. Sheppard
- Division of Gastrointestinal and General SurgeryOregon Health & Science UniversityPortlandOregonUSA
| | - Charles R. Thomas
- Department of Radiation MedicineOregon Health & Science UniversityPortlandOregonUSA
- Radiation OncologyGeisel School of Medicine at Dartmouth and Dartmouth Cancer CenterNew HampshireLebanonUSA
| | - Charles D. Lopez
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Gina M. Vaccaro
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
- Providence Cancer InstitutePortlandOregonUSA
| | | |
Collapse
|
39
|
Hackner D, Hobbs M, Merkel S, Krautz C, Weber GF, Grützmann R, Brunner M. Impact of Aspirin Intake on Postoperative Survival after Primary Pancreatic Resection of Pancreatic Ductal Adenocarcinoma-A Single-Center Evaluation. Biomedicines 2023; 11:biomedicines11051466. [PMID: 37239137 DOI: 10.3390/biomedicines11051466] [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: 04/17/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: The intake of aspirin (ASS) has been demonstrated to have a relevant impact on the pathogenesis, incidence and outcome in different solid gastrointestinal tumors. However, data on the effect of ASS on the short-term outcome and the long-term survival in patients with pancreatic carcinoma are still limited. (2) Methods: A total of 213 patients who underwent primary resection of PDAC at the University Hospital of Erlangen from January 2000 to December 2018 were included in this retrospective single-center study in total. Patients were stratified according to the aspirin intake into three groups: continuous aspirin intake (cASS), perioperatively interrupted aspirin intake (iASS) and no aspirin intake (no ASS) at the timepoint of surgery. The postoperative outcome as well as long-term survival were compared between the groups. (3) Results: There were no differences regarding postoperative morbidity (iASS: 54% vs. cASS: 53% vs. no ASS: 64%, p = 0.448) and in-hospital mortality (iASS: 4% vs. cASS: 10% vs. no ASS: 3%, p = 0.198) between the groups. The overall survival (OS) and disease-free survival (DFS) did not differ in the groups when comparing the ASS-intake status (OS: iASS 17.8 months vs. cASS 19.6 months vs. no ASS 21.6 months, p = 0.489; DFS: iASS 14.0 months vs. cASS 18.3 months vs. no ASS 14.7 months, p = 0.957). Multivariate analysis revealed that age (hazard ratio (HR) 2.2, p < 0.001), lymph node-positive status (HR 2.0, p < 0.001), R status 1 or 2 (HR 2.8, p < 0.001) and differentiation with a grading of 3 (HR 1.7, p = 0.005) were significant independent prognostic factors regarding the OS. Moreover, age (HR 1.5, p = 0.040), lymph node-positive status (HR 1.8, p = 0.002) and high-grade (G3) carcinomas (HR 1.5, p = 0.037) could be identified as independent prognostic parameters for DFS. (4) Conclusions: In patients undergoing primary surgery for curative resection of pancreatic carcinoma, the perioperative intake of ASS had no significant impact on postoperative outcome, overall and disease-free survival.
Collapse
Affiliation(s)
- Danilo Hackner
- Department of General and Visceral Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Mirianna Hobbs
- Department of General and Visceral Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Susanne Merkel
- Department of General and Visceral Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christian Krautz
- Department of General and Visceral Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Georg F Weber
- Department of General and Visceral Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Robert Grützmann
- Department of General and Visceral Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Maximilian Brunner
- Department of General and Visceral Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
40
|
Mirzayeh Fashami F, Levine M, Xie F, Blackhouse G, Tarride JE. Olaparib versus Placebo in Maintenance Treatment of Germline BRCA-Mutated Metastatic Pancreatic Cancer: A Cost-Utility Analysis from the Canadian Public Payer's Perspective. Curr Oncol 2023; 30:4688-4699. [PMID: 37232812 DOI: 10.3390/curroncol30050354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/11/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Pancreatic cancer has an annual incidence of 2/10,000 in Canada, with a one-year mortality rate greater than 80%. In the absence of a cost-effectiveness analysis in Canada, this study's objective was to assess the cost-effectiveness of olaparib versus a placebo in adult patients with deleterious or suspected deleterious BRCA metastatic pancreatic adenocarcinoma, who did not show any progression for at least 16 weeks with first-line platinum-based chemotherapy. A partitioned survival model with a 5-year time horizon was adopted to estimate the costs and effectiveness. All of the costs were extracted from the public payer's available resources, effectiveness data were obtained from the POLO trial, and Canadian studies were used for utility inputs. Probabilistic sensitivity analyses and scenario analyses were performed. The total costs of olaparib and the placebo over five years were CAD 179,477 and CAD 68,569, with overall quality-adjusted life-years (QALYs) of 1.70 and 1.36, respectively. The incremental cost-effectiveness ratio (ICER) of the olaparib group compared with the placebo was CAD 329,517 per QALY. With a commonly cited willingness to pay (WTP) threshold of CAD 50,000 per QALY, the drug does not achieve acceptable cost-effectiveness mainly due to the high price of the medication and insufficient impact on the overall survival of patients with metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Fatemeh Mirzayeh Fashami
- Health Research Methodology Graduate Program, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
| | - Mitchell Levine
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Center for Health Economics and Policy Analysis (CHEPA), McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Programs for Assessment of Technology in Health (PATH), The Research Institute of St. Joe's Hamilton, St. Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON L8N 4A6, Canada
| | - Feng Xie
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Center for Health Economics and Policy Analysis (CHEPA), McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
| | - Gordon Blackhouse
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Programs for Assessment of Technology in Health (PATH), The Research Institute of St. Joe's Hamilton, St. Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON L8N 4A6, Canada
| | - Jean-Eric Tarride
- Center for Health Economics and Policy Analysis (CHEPA), McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Programs for Assessment of Technology in Health (PATH), The Research Institute of St. Joe's Hamilton, St. Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON L8N 4A6, Canada
- McMaster Chair in Health Technology Management, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
41
|
Springfeld C, Ferrone CR, Katz MHG, Philip PA, Hong TS, Hackert T, Büchler MW, Neoptolemos J. Neoadjuvant therapy for pancreatic cancer. Nat Rev Clin Oncol 2023; 20:318-337. [PMID: 36932224 DOI: 10.1038/s41571-023-00746-1] [Citation(s) in RCA: 155] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/19/2023]
Abstract
Patients with localized pancreatic ductal adenocarcinoma (PDAC) are best treated with surgical resection of the primary tumour and systemic chemotherapy, which provides considerably longer overall survival (OS) durations than either modality alone. Regardless, most patients will have disease relapse owing to micrometastatic disease. Although currently a matter of some debate, considerable research interest has been focused on the role of neoadjuvant therapy for all forms of resectable PDAC. Whilst adjuvant combination chemotherapy remains the standard of care for patients with resectable PDAC, neoadjuvant chemotherapy seems to improve OS without necessarily increasing the resection rate in those with borderline-resectable disease. Furthermore, around 20% of patients with unresectable non-metastatic PDAC might undergo resection following 4-6 months of induction combination chemotherapy with or without radiotherapy, even in the absence of a clear radiological response, leading to improved OS outcomes in this group. Distinct molecular and biological responses to different types of therapies need to be better understood in order to enable the optimal sequencing of specific treatment modalities to further improve OS. In this Review, we describe current treatment strategies for the various clinical stages of PDAC and discuss developments that are likely to determine the optimal sequence of multimodality therapies by integrating the fundamental clinical and molecular features of the cancer.
Collapse
Affiliation(s)
- Christoph Springfeld
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip A Philip
- Wayne State University School of Medicine, Department of Oncology, Henry Ford Cancer Institute, Detroit, MI, USA
| | - Theodore S Hong
- Research and Scientific Affairs, Gastrointestinal Service Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - John Neoptolemos
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
42
|
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: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/09/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
The dynamic tumor microenvironment, especially the immune microenvironment, during the natural progression and/or chemotherapy treatment is a critical frontier in understanding the effects of chemotherapy on pancreatic cancer. Non-stratified pancreatic cancer patients always receive chemotherapeutic strategies, including neoadjuvant chemotherapy and adjuvant chemotherapy, predominantly according to their physical conditions and different disease stages. An increasing number of studies demonstrate that the pancreatic cancer tumor microenvironment could be reshaped by chemotherapy, an outcome caused by immunogenic cell death, selection and/or education of preponderant tumor clones, adaptive gene mutations, and induction of cytokines/chemokines. These outcomes could in turn impact the efficacy of chemotherapy, making it range from synergetic to resistant and even tumor-promoting. Under chemotherapeutic impact, the metastatic micro-structures in the primary tumor may be built to leak tumor cells into the lymph or blood vasculature, and micro-metastatic/recurrent niches rich in immunosuppressive cells may be recruited by cytokines and chemokines, which provide housing conditions for these circling tumor cells. An in-depth understanding of how chemotherapy reshapes the tumor microenvironment may lead to new therapeutic strategies to block its adverse tumor-promoting effects and prolong survival. In this review, reshaped pancreatic cancer tumor microenvironments due to chemotherapy were reflected mainly in immune cells, pancreatic cancer cells, and cancer-associated fibroblast cells, quantitatively, functionally, and spatially. Additionally, small molecule kinases and immune checkpoints participating in this remodeling process caused by chemotherapy are suggested to be blocked reasonably to synergize with chemotherapy.
Collapse
Affiliation(s)
- Maozhen Peng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- 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
|
43
|
Nevler A, Khalilieh S, Lavu H, Bowne W, Yeo CJ. Hypercapnic Tissue Gene Expression and Survival in Early-Stage Pancreatic Ductal Adenocarcinoma. J Am Coll Surg 2023; 236:913-922. [PMID: 36728372 DOI: 10.1097/xcs.0000000000000552] [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: 02/03/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal cancer. Hypercapnic tumor microenvironments were previously shown to promote cancer chemoresistance. In this study, we aimed to investigate the impact of tissue hypercapnia on PDAC prognosis. STUDY DESIGN PDAC cancer-cell lines were cultured in normocapnic (5% CO 2 ) and hypercapnic conditions (10% CO 2 ). RNA was extracted, and whole-exome transcriptome was sequenced. Differentially expressed genes were identified and used to construct a "hypercapnic gene set." PDAC transcriptomic patient data from the Tumor Cancer Genome Atlas was used to calculate single-sample gene set enrichment scores based on each patient's tissue expression of the hypercapnic gene set. Tissue hypercapnic scores (HSs) in PDAC patients (TMN stages Ia-IIb) were determined and correlated with clinicopathological parameters and overall survival. RESULTS A cohort of 135 resected stage I-II PDAC patients were assessed in this study. The average age was 65 ± 11.0 years, and the male:female ratio was 74:61. Median overall survival was 19.5 ± 1.4 months. High HSs were associated with increased tumor stage (p < 0.05) and higher lymph-node ratio (p < 0.05). In active smokers, high HS also correlated with smoking pack-years (p < 0.05). Cox regression analysis revealed high HS to be an independent prognostic factor for overall survival (hazard ratio [HR] 2.66, p = 0.004), along with lymph-node ratio (HR 4.2, p = 0.002) and age at diagnosis (HR 2.63, p = 0.01). CONCLUSIONS The pancreatic tumor microenvironment plays an integral role in tumor aggressiveness, and our previous in vitro data suggest that hypercapnia promotes an aggressive, more resistant phenotype. Herein, we show that in early-stage pancreatic cancer, hypercapnic tissue signatures corresponded with a worse overall survival.
Collapse
Affiliation(s)
- Avinoam Nevler
- From the Jefferson Pancreas, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA
| | | | | | | | | |
Collapse
|
44
|
Huang J, Wang M, Zhang F, Shao S, Yao Z, Zhao X, Hu Q, Liang T. An Ionic Liquid Ablation Agent for Local Ablation and Immune Activation in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206756. [PMID: 36698308 PMCID: PMC10074093 DOI: 10.1002/advs.202206756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Indexed: 06/17/2023]
Abstract
Pancreatic ductal adenocarcinoma rapidly acquires resistance to chemotherapy, remaining a fatal disease. Immunotherapy is one of the breakthroughs in cancer treatment, which includes immune checkpoint inhibitors, chimeric antigen receptor T-cell immunotherapy, and neoantigen vaccines. However, immunotherapy has not achieved satisfactory results in the treatment of pancreatic cancer. Immunogenic death comprises proinflammatory cell death, which provides a way to enhance tumor immunogenicity and promote an immune response in solid tumors. Herein, an ionic liquid ablation agent (LAA), synthesized from choline and geranic acid, which triggers necrosis-induced immunotherapy by remodeling an immunosuppressive "cold" tumor to an immune activated "hot" tumor is described. The results indicate that LAA-treated tumor cells can enhance immunogenicity, inducing dendritic cell maturation, macrophage M1 polarization, and cytotoxic T lymphocyte infiltration. The results of the present study provide a novel strategy for solid tumor immunotherapy.
Collapse
Affiliation(s)
- Junming Huang
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
| | - Meng Wang
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
| | - Fu Zhang
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
| | - Shiyi Shao
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhou310003P. R. China
| | - Zhuo Yao
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
| | - Xinyu Zhao
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
| | - Qida Hu
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhou310003P. R. China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic SurgeryFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310006P. R. China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhou310003P. R. China
- Innovation Center for the Study of Pancreatic DiseasesHangzhou310003P. R. China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic DiseasesHangzhou310003P. R. China
- Cancer CenterZhejiang UniversityHangzhou310058P. R. China
- Research Center for Healthcare Data ScienceZhejiang LabHangzhou310003P. R. China
| |
Collapse
|
45
|
van Eijck CWF, de Koning W, van der Sijde F, Moskie M, Groot Koerkamp B, Homs MYV, van der Burg SH, van Eijck CHJ, Mustafa DAM. A multigene circulating biomarker to predict the lack of FOLFIRINOX response after a single cycle in patients with pancreatic ductal adenocarcinoma. Eur J Cancer 2023; 181:119-134. [PMID: 36652890 DOI: 10.1016/j.ejca.2022.12.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022]
Abstract
INTRODUCTION 5-fluorouracil, folinic acid, irinotecan and oxaliplatin (FOLFIRINOX) is promising in treating patients with pancreatic ductal adenocarcinoma. However, many patients and physicians are reluctant to start FOLFIRINOX due to its high toxicity and limited clinical response rates. In this study, we investigated the effect of a single FOLFIRINOX cycle, in combination with a granulocyte colony-stimulating factor, on the blood immune transcriptome of patients with pancreatic ductal adenocarcinoma. We aimed to identify an early circulating biomarker to predict the lack of FOLFIRINOX response. METHODS Blood samples of 68 patients from all disease stages, who received at least four FOLFIRINOX cycles, were collected at baseline and after the first cycle. The response to treatment was radiologically evaluated following the Response Evaluation Criteria in Solid Tumours criteria 1.1. Targeted immune-gene expression profiling (GEP) was performed using NanoString technologies. To predict the lack of FOLFIRINOX response, we developed a FOLFIRINOX delta GEP (FFX-ΔGEP) score. RESULTS A single FOLFIRINOX cycle significantly altered 395 genes, correlating to 30 significant alterations in relative immune cell abundances and pathway activities. The eight-gene (BID, FOXP3, KIR3DL1, MAF, PDGFRB, RRAD, SIGLEC1 and TGFB2) FFX-ΔGEP score predicted the lack of FOLFIRINOX response with a leave-one-out cross-validated area under the curve (95% confidence interval) of 0.87 (0.60-0.98), thereby outperforming the predictiveness of absolute and proportional Δcarbohydrate antigen19-9 values. CONCLUSIONS A single FOLFIRINOX cycle, combined with granulocyte colony-stimulating factor, alters the peripheral immune transcriptome indisputably. Our novel FFX-ΔGEP is, to our knowledge, the first multigene early circulating biomarker that predicts the lack of FOLFIRINOX response after one cycle. Validation in a larger independent patient cohort is crucial before clinical implementation.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Department of Surgery, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands; Department of Pathology Unit of Tumour Immuno-Pathology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| | - Willem de Koning
- Department of Pathology Unit of Tumour Immuno-Pathology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands; Department of Pathology Unit of Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| | - Fleur van der Sijde
- Department of Surgery, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands; Department of Pathology Unit of Tumour Immuno-Pathology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| | - Miranda Moskie
- Department of Surgery, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| | - Marjolein Y V Homs
- Department of Medical Oncology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands; Department of Pathology Unit of Tumour Immuno-Pathology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| | - Dana A M Mustafa
- Department of Pathology Unit of Tumour Immuno-Pathology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
46
|
Newhook TE, Vreeland TJ, Griffin JF, Tidwell RSS, Prakash LR, Koay EJ, Ludmir EB, Smaglo BG, Pant S, Overman M, Wolff RA, Ikoma N, Maxwell J, Kim MP, Lee JE, Katz MHG, Tzeng CWD. Prognosis Associated With CA19-9 Response Dynamics and Normalization During Neoadjuvant Therapy in Resected Pancreatic Adenocarcinoma. Ann Surg 2023; 277:484-490. [PMID: 36649067 DOI: 10.1097/sla.0000000000005184] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To characterize associations between carbohydrate antigen 19-9 (CA19-9) dynamics during neoadjuvant therapy (NT) and survival for patients with pancreatic ductal adenocarcinoma (PDAC). BACKGROUND Although normalization of CA19-9 during NT is associated with improved outcomes following PDAC resection, we hypothesize that CA19-9 dynamics during NT can improve prognostication. METHODS Characteristics for patients with PDAC undergoing NT (July 2011-October 2018) with ≥3 CA19-9 results (bilirubin<2mg/dL) were collected and grouped by CA19-9 dynamics. Nonproducers (<1 U/ml) were excluded, and normal was ≤35 U/ml. Postresection survival was compared among groups. RESULTS Of 431 patients, 166 had eligible CA19-9 values. Median baseline CA19-9 was 98 U/ml. Overall 2-year postresection recurrence-free survival (RFS) and overall survival (OS) were 37% and 63%, respectively. Patients with normalization (53%) had improved 2-year RFS (47% vs. 28%, P = 0.01) and OS (75% vs. 49%, P = 0.01). CA19-9 dynamics during NT were analyzed by shape, direction, and normalization creating response types ("A-B-C-D-E"). Type A was "Always" decreasing to normalization, B "Bidirectional" with eventual normalization, C "Consistently" normal, D any "Decrease" without normalization, and E "Elevating" without normalization. Types A and B responses were associated with the longest postresection 2-year RFS (51% and 56%) and OS (75% and 92%, respectively) whereas Types D and E had the worst outcomes. After adjusting for node-positivity, perineural invasion, and margin-positivity, CA19-9 response types were independently associated with both RFS and OS, and predicted outcomes are better than CA19-9 normalization alone (likelihood ratio test RFS P < 0.001, OS P = 0.01). CONCLUSIONS This novel A-B-C-D-E classification of CA19-9 dynamics during NT was associated with postresection outcomes more precisely than CA19-9 normalization alone.
Collapse
Affiliation(s)
- Timothy E Newhook
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Rebecca S S Tidwell
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Laura R Prakash
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Eugene J Koay
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ethan B Ludmir
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brandon G Smaglo
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shubham Pant
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael Overman
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert A Wolff
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jessica Maxwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
47
|
Dai WF, Habbous S, Saluja R, Beca JM, Raphael M, Arias J, Gavura S, Earle CC, Biagi JJ, Coburn N, Chan KKW. Comparative Effectiveness of FOLFIRINOX Versus Gemcitabine and Nab-paclitaxel in Initially Unresectable Locally Advanced Pancreatic Cancer: A Population-based Study to Assess Subsequent Surgical Resection and Overall Survival. Clin Oncol (R Coll Radiol) 2023; 35:e303-e311. [PMID: 36863956 DOI: 10.1016/j.clon.2023.02.008] [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: 01/20/2022] [Revised: 12/22/2022] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
AIMS First-line FOLFIRINOX (FOLinic acid, Fluorouracil, IRINotecan, and OXaliplatin) and gemcitabine plus nab-paclitaxel (GnP) have been publicly funded for patients with unresectable locally advanced pancreatic cancer (uLAPC) in Ontario, Canada. We examined the overall survival and surgical resection rate after first-line FOLFIRINOX or GnP and determined the association between resection and overall survival in patients with uLAPC. MATERIALS AND METHODS We conducted a retrospective population-based study including patients with uLAPC who received first-line treatment FOLFIRINOX or GnP from April 2015 to March 2019. The cohort was linked to administrative databases to ascertain demographic and clinical characteristics. Propensity score methods were used to balance differences between FOLFIRINOX and GnP. The Kaplan-Meier method was used to calculate overall survival. Cox regression was used to determine the association between receipt of treatment and overall survival, adjusting for time-dependent surgical resections. RESULTS We identified 723 patients with uLAPC (mean age = 65.8, 43.5% female) who received FOLFIRINOX (55.2%) or GnP (44.8%). The median overall survival and 1-year overall survival probability were higher for FOLFIRINOX (13.7 months, 54.6%) than for GnP (8.7 months, 34.0%). Post-chemotherapy surgical resection occurred in 89 (12.3%) patients (FOLFIRINOX: 74 [18.5%] versus GnP: 15 [4.6%]), with no difference in survival since surgery between FOLFIRINOX and GnP (P = 0.29). After adjusting time-dependent post-treatment surgical resection, FOLFIRINOX (inverse probability treatment weighting hazard ratio 0.72, 95% confidence interval 0.61, 0.84) was independently associated with improved overall survival. CONCLUSIONS In this real-world population-based study of patients with uLAPC, FOLFIRINOX was associated with improved survival and higher resection rates. FOLFIRINOX was associated with improved survival in patients with uLAPC after accounting for the effect of post-chemotherapy surgical resection, suggesting the benefit of FOLFIRINOX was not solely due to improving resectability.
Collapse
Affiliation(s)
- W F Dai
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Applied Research in Cancer Control, Toronto, Onatario, Canada
| | - S Habbous
- Ontario Health (Cancer Care Ontario), Toronto, Ontario, Canada
| | - R Saluja
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - J M Beca
- Canadian Centre for Applied Research in Cancer Control, Toronto, Onatario, Canada; Ontario Health (Cancer Care Ontario), Toronto, Ontario, Canada
| | - M Raphael
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - J Arias
- Ontario Health (Cancer Care Ontario), Toronto, Ontario, Canada
| | - S Gavura
- Ontario Health (Cancer Care Ontario), Toronto, Ontario, Canada
| | - C C Earle
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; ICES, Toronto, Ontario, Canada
| | - J J Biagi
- Cancer Centre of Southeastern Ontario, Kingston, Ontario, Canada
| | - N Coburn
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - K K W Chan
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Applied Research in Cancer Control, Toronto, Onatario, Canada; Ontario Health (Cancer Care Ontario), Toronto, Ontario, Canada; Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.
| |
Collapse
|
48
|
Thalji SZ, Kamgar M, George B, Aldakkak M, Christians KK, Clarke CN, Erickson BA, Hall WA, Tolat PP, Smith ZL, Evans DB, Tsai S. CA19-9 Response to First-Line Neoadjuvant FOLFIRINOX and Second-Line Gemcitabine/Nab-Paclitaxel for Patients with Operable Pancreatic Cancer. Ann Surg Oncol 2023; 30:3013-3021. [PMID: 36788189 DOI: 10.1245/s10434-022-13055-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/22/2022] [Indexed: 02/16/2023]
Abstract
BACKGROUND Response to second-line (2L) neoadjuvant therapy for operable pancreatic cancer (PC) is understudied. This study examined carbohydrate antigen 19-9 (CA19-9) response to first-line (1L) and 2L chemotherapy. METHODS The study identified patients with operable PC and elevated CA19-9 (≥ 35 U/mL with total bilirubin < 2 mg/dL) who received 1L FOLFIRINOX (FFX). The patients were restaged after 2 months and based on response, received additional FFX or gemcitabine/nab-paclitaxel (GnP) as part of total neoadjuvant therapy. Response was defined as a decrease in tumor size on computed tomography (CT) imaging or a decline in CA19-9 of 50% or more and preserved performance status. RESULTS For operable PC with an elevated CA19-9, 108 patients received 1L FFX. After 2 months of chemotherapy, the decision was made to continue FFX (FFX ≥ FFX) for 76 (70%) of the 108 patients and switch to GnP (FFX ≥ GnP)) for 32 (30%) of the patients. Of the 32 FFX ≥ GnP patients, 27 had no evidence of radiographic or biochemical (CA19-9) response to 1L FFX. Of these 27 patients, 26 (96%) demonstrated a response to 2L GnP. After 4 months of chemotherapy, 62 (82%) of the 76 FFX ≥ FFX patients had a CA19-9 response compared with 31 (97%) of the 32 FFX ≥ GnP patients (p = 0.04). CONCLUSIONS Lack of biochemical response to 2 months of 1L FFX may identify a subgroup of patients with a very high rate of response to 2L GnP, emphasizing the importance of assessing treatment response at 2-month intervals.
Collapse
Affiliation(s)
- Sam Z Thalji
- Department of Surgery, Division of Surgical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mandana Kamgar
- Department of Medicine, Division of Medical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ben George
- Department of Medicine, Division of Medical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mohammed Aldakkak
- Department of Surgery, Division of Surgical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kathleen K Christians
- Department of Surgery, Division of Surgical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Callisia N Clarke
- Department of Surgery, Division of Surgical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Beth A Erickson
- Department of Radiation Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - William A Hall
- Department of Radiation Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Parag P Tolat
- Department of Radiology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zachary L Smith
- Department of Medicine, Division of Gastroenterology and Hepatology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Douglas B Evans
- Department of Surgery, Division of Surgical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Susan Tsai
- Department of Surgery, Division of Surgical Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
49
|
Yang XY, Lu YF, Xu JX, Du YZ, Yu RS. Recent Advances in Well-Designed Therapeutic Nanosystems for the Pancreatic Ductal Adenocarcinoma Treatment Dilemma. Molecules 2023; 28:molecules28031506. [PMID: 36771172 PMCID: PMC9920782 DOI: 10.3390/molecules28031506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with an extremely poor prognosis and low survival rate. Due to its inconspicuous symptoms, PDAC is difficult to diagnose early. Most patients are diagnosed in the middle and late stages, losing the opportunity for surgery. Chemotherapy is the main treatment in clinical practice and improves the survival of patients to some extent. However, the improved prognosis is associated with higher side effects, and the overall prognosis is far from satisfactory. In addition to resistance to chemotherapy, PDAC is significantly resistant to targeted therapy and immunotherapy. The failure of multiple treatment modalities indicates great dilemmas in treating PDAC, including high molecular heterogeneity, high drug resistance, an immunosuppressive microenvironment, and a dense matrix. Nanomedicine shows great potential to overcome the therapeutic barriers of PDAC. Through the careful design and rational modification of nanomaterials, multifunctional intelligent nanosystems can be obtained. These nanosystems can adapt to the environment's needs and compensate for conventional treatments' shortcomings. This review is focused on recent advances in the use of well-designed nanosystems in different therapeutic modalities to overcome the PDAC treatment dilemma, including a variety of novel therapeutic modalities. Finally, these nanosystems' bottlenecks in treating PDAC and the prospect of future clinical translation are briefly discussed.
Collapse
Affiliation(s)
- Xiao-Yan Yang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Yuan-Fei Lu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Jian-Xia Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, 318 Chaowang Road, Hangzhou 310005, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
- Correspondence: (Y.-Z.D.); (R.-S.Y.); Tel.: +86-571-88208435 (Y.-Z.D.); +86-571-87783925 (R.-S.Y.)
| | - Ri-Sheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
- Correspondence: (Y.-Z.D.); (R.-S.Y.); Tel.: +86-571-88208435 (Y.-Z.D.); +86-571-87783925 (R.-S.Y.)
| |
Collapse
|
50
|
Walpole I, Lee B, Shapiro J, Thomson B, Lipton L, Ananda S, Usatoff V, Mclachlan SA, Knowles B, Fox A, Wong R, Cooray P, Burge M, Clarke K, Pattison S, Nikfarjam M, Tebbutt N, Harris M, Nagrial A, Zielinski R, Chee CE, Gibbs P. Use and outcomes from neoadjuvant chemotherapy in borderline resectable pancreatic ductal adenocarcinoma in an Australasian population. Asia Pac J Clin Oncol 2023; 19:214-225. [PMID: 35831999 DOI: 10.1111/ajco.13807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/08/2022] [Accepted: 06/13/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND Use of neoadjuvant (NA) chemotherapy is recommended when pancreatic ductal adenocarcinoma (PDAC) is borderline resectable METHOD: A retrospective analysis of consecutive patients with localized PDAC between January 2016 and March 2019 within the Australasian Pancreatic Cancer Registry (PURPLE, Pancreatic cancer: Understanding Routine Practice and Lifting End results) was performed. Clinicopathological characteristics, treatment, and outcome were analyzed. Overall survival (OS) comparison was performed using log-rank model and Kaplan-Meier analysis. RESULTS The PURPLE database included 754 cases with localised PDAC, including 148 (20%) cases with borderline resectable pancreatic cancer (BRPC). Of the 148 BRPC patients, 44 (30%) underwent immediate surgery, 80 (54%) received NA chemotherapy, and 24 (16%) were inoperable. The median age of NA therapy patients was 63 years and FOLFIRINOX (53%) was more often used as NA therapy than gemcitabine/nab-paclitaxel (31%). Patients who received FOLFIRINOX were younger than those who received gemcitabine/nab-paclitaxel (60 years vs. 67 years, p = .01). Surgery was performed in 54% (43 of 80) of BRPC patients receiving NA chemotherapy, with 53% (16 of 30) achieving R0 resections. BRPC patients undergoing surgery had a median OS of 30 months, and 38% (9 of 24) achieved R0 resection. NA chemotherapy patients had a median OS of 20 months, improving to 24 months versus 10 months for patients receiving FOLFIRINOX compared to gemcitabine/nab-paclitaxel (Hazard Ratio (HR) .3, p < .0001). CONCLUSIONS NA chemotherapy use in BRPC is increasing in Australia. One half of patients receiving NA chemotherapy proceed to curative resection, with 53% achieving R0 resections. Patients receiving Infusional 5-flurouracil, Irinotecan and Oxaliplatin (FOLIRINOX) had increased survival than gemcitabine/nab-paclitaxel. Treatment strategies are being explored in the MASTERPLAN and DYNAMIC-Pancreas trials.
Collapse
Affiliation(s)
- Imogen Walpole
- Department of Medical Oncology, Northern Hospital, Victoria, Australia
| | - Belinda Lee
- Department of Medical Oncology, Northern Hospital, Victoria, Australia
- Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Faculty of Medicine & Health Sciences, Faculty fo Medicine University of Melbourne, Victoria, Australia
| | - Jeremy Shapiro
- Department of Medical Oncology, Cabrini Health, Malvern, Victoria, Australia
- Faculty of Medicine & Health Sciences, Monash University, Victoria, Australia
| | - Benjamin Thomson
- Department of Surgery, University of Melbourne, Royal Melbourne Hospital, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - Lara Lipton
- Department of Medical Oncology, Cabrini Health, Malvern, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
- Department of Medical Oncology, Western Health, Victoria, Australia
| | - Sumitra Ananda
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
- Department of Medical Oncology, Western Health, Victoria, Australia
| | - Val Usatoff
- Department of Medical Oncology, Cabrini Health, Malvern, Victoria, Australia
- Department of Medical Oncology, Western Health, Victoria, Australia
| | - Sue-Ann Mclachlan
- Faculty of Medicine & Health Sciences, Faculty fo Medicine University of Melbourne, Victoria, Australia
- Department of Medical Oncology, St Vincent's Hospital, Victoria, Australia
| | - Brett Knowles
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
- Department of Medical Oncology, St Vincent's Hospital, Victoria, Australia
| | - Adrian Fox
- Department of Medical Oncology, St Vincent's Hospital, Victoria, Australia
- Department of Medical Oncology, Eastern Health, Victoria, Australia
| | - Rachel Wong
- Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Faculty of Medicine & Health Sciences, Monash University, Victoria, Australia
- Department of Medical Oncology, Eastern Health, Victoria, Australia
- Department of Medical Oncology, Epworth Hospital, Victoria, Australia
| | - Prasad Cooray
- Department of Medical Oncology, Knox Private Hospital, Victoria, Australia
| | - Matthew Burge
- Department of Medical Oncology, Royal Brisbane Hospital, Queensland, Australia
| | - Kate Clarke
- Department of Medical Oncology, Wellington Hospital, Wellington, New Zealand
| | - Sharon Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Mehrdad Nikfarjam
- Faculty of Medicine & Health Sciences, Faculty fo Medicine University of Melbourne, Victoria, Australia
- Department of Medical Oncology, Austin Health, Victoria, Australia
- Department of Surgery, Warringal Private Hospital, Victoria, Australia
| | - Niall Tebbutt
- Department of Medical Oncology, Austin Health, Victoria, Australia
| | - Marion Harris
- Department of Medical Oncology, Monash Medical Centre, Victoria, Australia
| | - Adnan Nagrial
- Department of Medical Oncology, Westmead Hospital, New South Wales, Australia
| | - Rob Zielinski
- Department of Medical Oncology, Orange Hospital, New South Wales, Australia
- Department of Medical Oncology, Dubbo Base Hospital, New South Wales, Australia
- Department of Medical Oncology, Bathurst Base Hospital, New South Wales, Australia
| | - Cheng Ean Chee
- Department of Medical Oncology, National University Cancer Institute, Singapore
| | - Peter Gibbs
- Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Faculty of Medicine & Health Sciences, Faculty fo Medicine University of Melbourne, Victoria, Australia
| |
Collapse
|