1
|
Cai W, Zhu Y, Teng Z, Li D, Cong R, Chen Z, Ma X, Zhao X. Extracellular volume-based scoring system for tracking tumor progression in pancreatic cancer patients receiving intraoperative radiotherapy. Insights Imaging 2024; 15:116. [PMID: 38735009 PMCID: PMC11089023 DOI: 10.1186/s13244-024-01689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/03/2024] [Indexed: 05/13/2024] Open
Abstract
OBJECTIVES To investigate the value of extracellular volume (ECV) derived from portal-venous phase (PVP) in predicting prognosis in locally advanced pancreatic cancer (LAPC) patients receiving intraoperative radiotherapy (IORT) with initial stable disease (SD) and to construct a risk-scoring system based on ECV and clinical-radiological features. MATERIALS AND METHODS One hundred and three patients with LAPC who received IORT demonstrating SD were enrolled and underwent multiphasic contrast-enhanced CT (CECT) before and after IORT. ECV maps were generated from unenhanced and PVP CT images. Clinical and CT imaging features were analyzed. The independent predictors of progression-free survival (PFS) determined by multivariate Cox regression model were used to construct the risk-scoring system. Time-dependent receiver operating characteristic (ROC) curve analysis and the Kaplan-Meier method were used to evaluate the predictive performance of the scoring system. RESULTS Multivariable analysis revealed that ECV, rim-enhancement, peripancreatic fat infiltration, and carbohydrate antigen 19-9 (CA19-9) response were significant predictors of PFS (all p < 0.05). Time-dependent ROC of the risk-scoring system showed a satisfactory predictive performance for disease progression with area under the curve (AUC) all above 0.70. High-risk patients (risk score ≥ 2) progress significantly faster than low-risk patients (risk score < 2) (p < 0.001). CONCLUSION ECV derived from PVP of conventional CECT was an independent predictor for progression in LAPC patients assessed as SD after IORT. The scoring system integrating ECV, radiological features, and CA19-9 response can be used as a practical tool for stratifying prognosis in these patients, assisting clinicians in developing an appropriate treatment approach. CRITICAL RELEVANCE STATEMENT The scoring system integrating ECV fraction, radiological features, and CA19-9 response can track tumor progression in patients with LAPC receiving IORT, aiding clinicians in choosing individual treatment strategies and improving their prognosis. KEY POINTS Predicting the progression of LAPC in patients receiving IORT is important. Our ECV-based scoring system can risk stratifying patients with initial SD. Appropriate prognostication can assist clinicians in developing appropriate treatment approaches.
Collapse
Affiliation(s)
- Wei Cai
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ze Teng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dengfeng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong Cong
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaowei Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Buss EJ, Kachnic LA, Horowitz DP. Radiotherapy for locally advanced pancreatic ductal adenocarcinoma. Semin Oncol 2021; 48:106-110. [PMID: 33771355 DOI: 10.1053/j.seminoncol.2021.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 01/20/2023]
Abstract
Locally advanced, unresectable pancreatic ductal adenocarcinoma has a poor prognosis with a median overall survival of 10-16 months. It is defined by tumor involvement of neighboring blood vessels that precludes resection. Standard doses of conventionally fractionated radiation have had little effect on overall survival in this setting, although they are associated with improved progression-free survival and time off chemotherapy. Evolving radiotherapy techniques have allowed for higher, ablative doses of radiotherapy to target tumor while also respecting normal tissue constraints of neighboring radiosensitive structures in the gastrointestinal tract. Moreover, advancements in image guidance, organ motion management, and the use of adaptive planning have enabled safe delivery of higher, ablative doses of radiation. This has resulted in improved survival. This review will summarize the expanding role of radiotherapy in the management of locally advanced, unresectable pancreatic cancer.
Collapse
|
3
|
Han X, Zhang WH, Wang WQ, Yu XJ, Liu L. Cancer-associated fibroblasts in therapeutic resistance of pancreatic cancer: Present situation, predicaments, and perspectives. Biochim Biophys Acta Rev Cancer 2020; 1874:188444. [PMID: 33031899 DOI: 10.1016/j.bbcan.2020.188444] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is highly lethal, and the most effective treatment is curative resection followed by chemotherapy. Unfortunately, chemoresistance is an extremely common occurrence, and novel treatment modalities, such as immunotherapy and molecular targeted therapy, have shown limited success in clinical practice. Pancreatic cancer is characterized by an abundant stromal compartment. Cancer-associated fibroblasts (CAFs) and the extracellular matrix they deposit account for a large portion of the pancreatic tumor stroma. CAFs interact directly and indirectly with pancreatic cancer cells and can compromise the effects of, and even promote tumorigenic responses to, various treatment approaches. To eliminate these adverse effects, CAFs depletion strategies were developed. Instead of the anticipated antitumor effects of CAFs depletion, more aggressive tumor phenotypes were occasionally observed. The failure of universal stromal depletion led to the investigation of CAFs heterogeneity that forms the foundation for stromal remodeling and normalization. This review analyzes the role of CAFs in therapeutic resistance of pancreatic cancer and discusses potential CAFs-targeting strategies basing on the diverse biological functions of CAFs, thus to improve the outcome of pancreatic cancer treatment.
Collapse
Affiliation(s)
- Xuan Han
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wu-Hu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Yu Z, Hong Z, Zhang Q, Lin LC, Shahnazi K, Wu X, Lu J, Jiang G, Wang Z. Proton and carbon ion radiation therapy for locally advanced pancreatic cancer: A phase I dose escalation study. Pancreatology 2020; 20:470-476. [PMID: 32033896 DOI: 10.1016/j.pan.2020.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/25/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To determine the maximum tolerated dose (MTD) of proton and carbon ion radiation therapy (PCRT) for locally advanced pancreatic cancer (LAPC). METHODS A single-institution, phase I dose escalation study was performed. The proton dose of 50.4 GyE in 28 fractions was delivered to clinical target volume, and carbon ion as a boost dose to gross tumor volume escalated from 12 GyE to 18 GyE with 3 GyE per fraction in 3 dose levels. The dose limiting toxicity (DLT) was defined as any treatment-related grade (G)3 or higher of non-hematological toxicity. The MTD was exceeded if ≥2 patients in a dose level developed DLT. RESULTS From May 2015 to July 2016, ten patients were enrolled, 3 in dose level 1, 4 in dose level 2, and 3 in dose level 3. With a median follow-up of 17.4 months, no patient developed a DLT, and the acute G1-2 of gastrointestinal (GI) and hepatic toxicity occurred in 40% of patients, and G1 of GI late toxicity, in 30%. The median overall survival was 17.3 months. CONCLUSION Higher than 50.4 GyE could be given by PCRT with slight toxicity and good tolerance for LAPC, and the tumor control and survival had been improved, but not significantly. Better outcome may be achieved using carbon ion radiation therapy with higher biological equivalent dose.
Collapse
Affiliation(s)
- Zhan Yu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Zhengshan Hong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Qing Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Lien-Chun Lin
- Department of Radiation Physics, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Kambiz Shahnazi
- Department of Radiation Physics, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Xiaodong Wu
- Department of Radiation Physics, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jiade Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Guoliang Jiang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China; Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zheng Wang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China; Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.
| |
Collapse
|
5
|
Li W, Wang X, Wang Z, Zhang T, Cai F, Tang P, Meng J, Du H, Wang H, Li M, Li S. The role of seed implantation in patients with unresectable pancreatic carcinoma after relief of obstructive jaundice using ERCP. Brachytherapy 2020; 19:97-103. [PMID: 31564517 DOI: 10.1016/j.brachy.2019.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/27/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE The purpose of the study was to investigate the role of iodine-125 seed implantation, guided by endoscopic ultrasound (EUS) and/or percutanous ultrasound, in patients with unresectable pancreatic carcinoma after relief of obstructive jaundice using endoscopic retrograde cholangiopancreatography (ERCP). METHODS AND MATERIALS A total of 101 patients with obstructive jaundice due to unresectable pancreatic carcinoma were enrolled between January 2010 and December 2017 in this retrospective study. Of these patients, 50 underwent implantation of iodine-125 seeds under EUS and/or percutaneous ultrasound guidance after receiving a stent via ERCP (treatment group), and 51 received a stent via ERCP without undergoing seed implantation (control group). The clinical data and therapeutic outcomes of these patients were analyzed. RESULTS Compared with the control group, the treatment group obtained significant relief of abdominal pain at the 1-week, 1-month, and 3-month followup (p < 0.05), with a significantly lower visual analog scale pain score (p < 0.05). The treatment group obtained a longer median survival (8.8 vs. 6.5 months, p = 0.02), longer median duration of stent patency (10.8 ± 1.4 vs. 6.9 ± 0.8 months, p = 0.02), and prolonged average time to gastric outlet obstruction (6.8 ± 1.6 vs. 5.3 ± 1.3 months, p = 0.02). Differences between liver function and appetite for the two groups were not significant (p > 0.05 and p = 0.59, respectively). CONCLUSIONS Iodine-125 seed implantation after relief of obstructive jaundice via ERCP prolongs survival, biliary stent patency, and time to gastric outlet obstruction and improves patient quality of life by relieving pancreatic pain in patients with unresectable pancreatic carcinoma.
Collapse
Affiliation(s)
- Wen Li
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Xiangdong Wang
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Zikai Wang
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Tiantian Zhang
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Fengchun Cai
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Ping Tang
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Jiangyun Meng
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Hong Du
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Hongbin Wang
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Mingyang Li
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China
| | - Shuling Li
- Department of Gastroenterology and Hepatology, The Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
6
|
Yang Y, Tao X, Li CB, Wang CM. MicroRNA-494 acts as a tumor suppressor in pancreatic cancer, inhibiting epithelial-mesenchymal transition, migration and invasion by binding to SDC1. Int J Oncol 2018; 53:1204-1214. [PMID: 29956739 DOI: 10.3892/ijo.2018.4445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/14/2018] [Indexed: 11/06/2022] Open
Abstract
Pancreatic cancer (PC) is the fourth most common cause of cancer‑related mortality in the industrialized world. Emerging evidence indicates that a variety of microRNAs (miRNAs or miRs) are involved in the development of PC. The aim of the present study was to elucidate the mechanisms through which miR‑494 affects the epithelial‑mesenchymal transition (EMT) and invasion of PC cells by binding to syndecan 1 (SDC1). PC tissues and pancreatitis tissues were collected, and the regulatory effects of miR‑494 on SDC1 were validated using bioinformatics analysis and a dual‑luciferase report gene assay. The cell line with the highest SDC1 expression was selected for use in the following experiments. The role of miR‑494 in EMT was assessed by measuring the expression of SDC1, E‑cadherin and vimentin. Cell proliferation was assessed using a cell counting kit (CCK)‑8 assay, migration was measured using a scratch test, invasion was assessed with a Transwell assay and apoptosis was detected by flow cytometry. Finally, a xenograft tumor model was constructed in nude mice to observe tumor growth in vivo. We found that SDC1 protein expression was significantly higher in the PC tissues. SDC1 was verified as a target gene of miR‑494. The SW1990 cell line was selected for use in further experiments as it had the lowest miR‑494 expression and the highest SDC1 expression. Our results also demonstrated that miR‑494 overexpression and SDC1 silencing significantly decreased the mRNA and protein expression of SDC1 and vimentin in SW1990 cells, while it increased E‑cadherin expression and apoptosis, and inhibited cell growth, migration, invasion and tumor growth. On the whole, the findings of this study demonstrated that miR‑494 is able to downregulate SDC1 expression, thereby inhibiting the progression of PC. These findings reveal a novel mechanism through which miR‑494 affects the development of PC and may thus provide a basis for the application of miR‑494 in pancreatic oncology.
Collapse
Affiliation(s)
- Ying Yang
- Department of General Surgery, Τhe First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiong Tao
- Department of General Surgery, Τhe First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Chun-Bo Li
- Department of General Surgery, Τhe First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Chang-Miao Wang
- Department of General Surgery, Τhe First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
7
|
Hu Y, Qi E, Liu F, Lu Y, Tan S, Sun Y, Han Z, Liang P, Yu X. The application of a three-dimensional visualized seed planning and navigation system in 125I seed implantation for pancreatic cancer. Onco Targets Ther 2018; 11:619-627. [PMID: 29440912 PMCID: PMC5798554 DOI: 10.2147/ott.s141245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objectives To evaluate the effectiveness of iodine-125 (125I) seed implantation for pancreatic cancer (PC), and preliminarily evaluate the clinical value of a self-developed three-dimensional (3D) visualized seed planning and navigation system in 125I seed implantation for treatment of PC. Patients and methods Our team retrospectively reviewed 25 PC patients who underwent 125I seed implantation between December 2010 and November 2016. The patients were divided into two groups: 3D visualization preoperative planning group (12 patients, 13 lesions) and two-dimensional (2D) regular group (13 patients, 14 lesions). We compared and analyzed the parameters of the two groups, such as number of needle insertions, one-time treatment success rate, proportion of added seeds, local control rate, rate of complications, rate of pain relief, and the survival rate and risk factors of the two groups. There was no significant difference in clinical data of the two groups. Results 125I seed implantation was performed successfully in all PC patients, with no occurrence of serious complications during and after the procedure. The one-time treatment success rate of 3D group (80%) was higher than that of 2D group (45.5%) (P<0.05), and the proportion of added seed number of 3D group was lower than that of 2D group (P<0.05). The local control rate of 3D group (76.9%) was higher than that of 2D group (35.7%) (P<0.05). The survival rate of 3D group was significantly higher than that of 2D group (P=0.026), and the median survival of 2D group vs 3D group was 5.00 vs 10.80 months. The median survival of all 25 patients was 7.10 months (95% confidence interval: 4.43-9.77). The rate of pain relief was 77.8% (7/9) in 2D group and 88.9% (8/9) in 3D group. Conclusion Ultrasound-guided, 3D visualized seed planning and navigation system assisted 125I seed implantation is a safe and effective method for the treatment of PC, with a prolonged survival of patients and better local control of tumor.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China.,Medical Department, First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Erpeng Qi
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| | - Fangyi Liu
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| | - Yuhan Lu
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| | - Shuilian Tan
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| | - Ya Sun
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| | - Zhiyu Han
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| | - Ping Liang
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| | - Xiaoling Yu
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, China
| |
Collapse
|
8
|
Wang L, Li P, Hu W, Xia Y, Hu C, Liu L, Jiang X. CD44 +CD24 + subset of PANC-1 cells exhibits radiation resistance via decreased levels of reactive oxygen species. Oncol Lett 2017; 14:1341-1346. [PMID: 28789349 PMCID: PMC5529798 DOI: 10.3892/ol.2017.6301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 02/28/2017] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence has suggested that pancreatic adenocarcinoma is sustained by pancreatic cancer stem cells. The present study aimed to investigate the expression patterns of the pancreatic cancer stem cell surface markers cluster of differentiation CD44 and CD24 in a pancreatic adenocarcinoma cell line, and to investigate the possible mechanisms for their radiation resistance. Flow cytometry was used to analyze the expression patterns of CD44 and CD24 in the pancreatic adenocarcinoma PANC-1 cell line. In addition, a multi-target click model was used to fit cell survival curves and determine the sensitizer enhancement ratio. The apoptosis and cycle distribution of the four cell subsets was determined using flow cytometry, and the level of reactive oxygen species (ROS) was determined using the 2',7'-dichlorofluorescin diacetate probe. The present results identified that the ratios of CD44+ and CD24+ in the sorted PANC-1 cell line were 92.0 and 4.7%, respectively. Prior to radiation, no statistically significant differences were observed among the four groups. Following treatment with 6 MV of X-rays, the rate of apoptosis was decreased in the CD44+CD24+ group compared with other subsets. The percentage of G0/G1 cells was highest in the CD44+CD24+ group compared with the three other groups, which exhibited increased radiosensitivity. In addition, the level of ROS in the CD44+CD24+ group was reduced compared with the other groups. In summary, the results of the present study indicated that CD44+CD24+ exhibited stem cell properties. The lower level of ROS and apoptosis in CD44+CD24+ cells may contribute to their resistance to radiation in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Lei Wang
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Pengping Li
- Department of Bioinformatics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Hu
- Department of Hepatobiliary Surgery, Lianyungang First People's Hospital, Lianyungang, Jiangsu 222002, P.R. China
| | - Youyou Xia
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Chenxi Hu
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Liang Liu
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Xiaodong Jiang
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| |
Collapse
|
9
|
Qiu Z, Chu Y, Xu B, Wang Q, Jiang M, Li X, Wang G, Yu P, Liu G, Wang H, Kang H, Liu J, Zhang Y, Jin JP, Wu K, Liang J. Increased expression of calponin 2 is a positive prognostic factor in pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:56428-56442. [PMID: 28915602 PMCID: PMC5593573 DOI: 10.18632/oncotarget.17701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/10/2017] [Indexed: 02/06/2023] Open
Abstract
Calponin 2 plays an important role in regulating actin cytoskeleton, which is critical for cell division and migration. Previous studies have demonstrated that calponin 2 inhibits prostate cancer cell proliferation and metastasis. However, the role of calponin 2 in pancreatic tumor growth, metastasis and patient survival remains unclear. Here, we demonstrate that the level of calponin 2 is a positive prognostic factor for patients with pancreatic ductal adenocarcinoma (PDAC). Patients with high calponin 2 expression in the tumor presented less lymph node metastasis and longer survival. Knockdown of calponin 2 facilitated pancreatic cancer cell proliferation and metastasis. Further experiments suggested that PI3K/AKT, NF-κB, Vimentin, Fibronectin, Snail and Slug were upregulated and E-cadherin was downregulated after calponin 2 was knocked down, implicating altered functions in PDAC proliferation and metastasis. In addition, we verified that calponin 2 functioned through inhibiting PI3K/AKT and NF-κB pathways. Our study suggests that the upregulation of calponin 2 in PDAC correlates to lower malignancy and presents a novel target for the development of new treatment.
Collapse
Affiliation(s)
- Zhaoyan Qiu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Yi Chu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bing Xu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qian Wang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Mingzuo Jiang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaowei Li
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Pengfei Yu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Guoxiao Liu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Hua Wang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Huijie Kang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiayu Liu
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Yu Zhang
- Department of Cardiovascular Surgery, General Hospital of Lanzhou Military Area Command, Lanzhou, China
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jie Liang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
10
|
Chung SY, Chang JS, Lee BM, Kim KH, Lee KJ, Seong J. Dose escalation in locally advanced pancreatic cancer patients receiving chemoradiotherapy. Radiother Oncol 2017; 123:438-445. [PMID: 28464997 DOI: 10.1016/j.radonc.2017.04.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 03/30/2017] [Accepted: 04/05/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE To investigate whether radiotherapy (RT) dose escalation would improve treatment outcomes without increasing severe toxicity in locally advanced pancreatic cancer patients. METHODS From 2005 to 2015, 497 locally advanced pancreatic cancer patients who received neoadjuvant or definitive chemoradiotherapy (CCRT) were included. Patients were divided according to the total dose (TD). Overall survival (OS), progression-free survival (PFS), local failure-free rate (LFFR), distant failure-free rate (DFFR), and toxicity rates were compared between <61Gy (n=345) and ≥61Gy groups (n=152). Additionally, propensity score matching was performed. RESULTS At a median follow-up of 19.3months (range, 4.8-128.5months), the 1-year OS, PFS, LFFR, and DFFR were significantly higher in the ≥61Gy group. After multivariate analysis, a TD of ≥61Gy remained a significant favorable factor for OS (p=0.019), PFS (p=0.001), LFFR (p=0.004), and DFFR (p=0.008). After propensity score matching, the ≥61Gy group still showed higher OS, PFS, and LFFR, but not DFFR (p=0.205). The acute and late toxicity rates showed no significant difference between the two groups. CONCLUSION Patients who received a higher RT dose showed not only improved PFS and LFFR, but also improved OS without an increase in severe toxicity. Dose-escalated CCRT can be a favorable treatment option in locally advanced pancreatic cancer patients.
Collapse
Affiliation(s)
- Seung Yeun Chung
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jee Suk Chang
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Min Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyong Joo Lee
- Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinsil Seong
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Nanog Predicts Poor Prognosis in Human Pancreatic Cancer and Is Downregulated by QingyihuaJi Formula in Pancreatic Cancer Stem Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:7028289. [PMID: 27829864 PMCID: PMC5086502 DOI: 10.1155/2016/7028289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/07/2016] [Accepted: 08/11/2016] [Indexed: 12/15/2022]
Abstract
Qingyihuaji formula (QYHJ), confirmed efficacious in a series of clinical trials, has been applied to human pancreatic carcinoma treatment in Shanghai Cancer Center for years. Recent evidence highlighted that pluripotent stem cells transcription factor Nanog plays a pivotal role in carcinogenesis. However, there is little published information regarding the underlying clinical significance and mechanisms of transcription factor Nanog in pancreatic cancer. In this study, our results indicated that Nanog is overexpressed in human pancreatic cancer stem cells and downregulated by QYHJ, which may contribute to explain the clinical effectiveness of QYHJ and provide advanced pancreatic cancer patients with a new therapeutic option, supporting our hypothesis that the degradation pathway is another mechanism by which QYHJ affects Nanog expression.
Collapse
|
12
|
Li D, Fu Z, Chen R, Zhao X, Zhou Y, Zeng B, Yu M, Zhou Q, Lin Q, Gao W, Ye H, Zhou J, Li Z, Liu Y, Chen R. Inhibition of glutamine metabolism counteracts pancreatic cancer stem cell features and sensitizes cells to radiotherapy. Oncotarget 2016; 6:31151-63. [PMID: 26439804 PMCID: PMC4741594 DOI: 10.18632/oncotarget.5150] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 08/21/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) cells utilize a novel non-canonical pathway of glutamine metabolism that is essential for tumor growth and redox balance. Inhibition of this metabolic pathway in PDAC can potentially synergize with therapies that increase intracellular reactive oxygen species (ROS) such as radiation. Here, we evaluated the dependence of pancreatic cancer stem cells (PCSCs) on this non-canonical glutamine metabolism pathway and researched whether inhibiting this pathway can enhance radiosensitivity of PCSCs. We showed that glutamine deprivation significantly inhibited self-renewal, decreased expression of stemness-related genes, increased intracellular ROS, and induced apoptosis in PCSCs. These effects were countered by oxaloacetate, but not α-ketoglutarate. Knockdown of glutamic-oxaloacetic transaminase dramatically impaired PCSCs properties, while glutamate dehydrogenase knockdown had a limited effect, suggesting a dependence of PCSCs on non-canonical glutamine metabolism. Additionally, glutamine deprivation significantly increased radiation-induced ROS and sensitized PCSCs to fractionated radiation. Moreover, transaminase inhibitors effectively enhanced ROS generation, promoted radiation sensitivity, and attenuated tumor growth in nude mice following radiation exposure. Our findings reveal that inhibiting the non-canonical pathway of glutamine metabolism enhances the PCSC radiosensitivity and may be an effective adjunct in cancer radiotherapy.
Collapse
Affiliation(s)
- Doudou Li
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiqiang Fu
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruiwan Chen
- Department of Radiotherapy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohui Zhao
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Zhou
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of General Surgery, Guangdong General Hospital, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bing Zeng
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Yu
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of General Surgery, Guangdong General Hospital, Guangzhou, China
| | - Quanbo Zhou
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing Lin
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenchao Gao
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huilin Ye
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiajia Zhou
- Department of Pediatric Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yimin Liu
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rufu Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Chadha AS, Skinner HD, Gunther JR, Munsell MF, Das P, Minsky BD, Delclos ME, Chatterjee D, Wang H, Clemons M, George G, Singh PK, Katz MH, Fleming JB, Javle MM, Wolff RA, Varadhachary GR, Crane CH, Krishnan S. Phase I Trial of Consolidative Radiotherapy with Concurrent Bevacizumab, Erlotinib and Capecitabine for Unresectable Pancreatic Cancer. PLoS One 2016; 11:e0156910. [PMID: 27336466 PMCID: PMC4919049 DOI: 10.1371/journal.pone.0156910] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/20/2016] [Indexed: 01/06/2023] Open
Abstract
Purpose To determine the safety, tolerability and maximum tolerated dose (MTD) of addition of erlotinib to bevacizumab and capecitabine-based definitive chemoradiation (CRT) in unresectable pancreatic cancer. Methods Seventeen patients with CT-staged, biopsy-proven unresectable pancreatic cancer were enrolled between 3/2008 and 10/2010. Prior chemotherapy was permitted. Two patients each were enrolled at dose levels (DLs) 1–4 and 9 patients at DL 5. All patients received 50.4 Gy (GTV only) in 28 fractions with concurrent capecitabine, bevacizumab and erlotinib. Dose of each drug was escalated in 5 DLs using the continual reassessment method. Bevacizumab was escalated from 5mg/Kg q2weeks (DLs 1–4) to 10mg/Kg q2weeks (DL 5); daily erlotinib from 100mg/day (DLs 1–2) to 150 mg/Kg (DLs 3–5); and capecitabine from 400mg/m2 twice daily on days of radiation (DL 1) to 650mg/m2 (DLs 2–3) to 825 mg/m2 (DLs 4–5). Reassessment for potential resection was performed 6–8 weeks later. Results Sixteen patients received gemcitabine-based chemotherapy prior to CRT. With a median clinical follow-up of 10 months, no grade 3 toxicities were observed in DLs 1–4. Three (33%) patients at DL 5 developed a grade 3 acute toxicity (2 diarrhea, 1 rash). No grade 4 or 5 toxicities were seen. DL 4 was selected as the MTD; therefore, the recommended doses in combination with radiation are: bevacizumab, 5mg/Kg q2weeks; erlotinib, 150 mg/Kg daily; and capecitabine, 825mg/m2 BID. Median survival was 17.4 months. Of the five patients who underwent resection, 4 were originally deemed locally advanced and 1 was borderline resectable. Three patients had excellent pathological response (2 complete response and 20% viable tumor) at surgery, and the 2 patients with complete response are still alive at 61 and 67 months of follow up with no local or distant failures. Conclusions This chemoradiation regimen at the recommended dose levels is safe and tolerable for patients with unresectable pancreatic cancer and merits further evaluation.
Collapse
Affiliation(s)
- Awalpreet S. Chadha
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Heath D. Skinner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jillian R. Gunther
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Mark F. Munsell
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bruce D. Minsky
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Marc E. Delclos
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Deyali Chatterjee
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Marilyn Clemons
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Geena George
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Pankaj K. Singh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Matthew H. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jason B. Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Milind M. Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gauri R. Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Christopher H. Crane
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
14
|
Abstract
The outcomes for treatment of pancreatic cancer have not improved dramatically in many decades. However, the recent promising results with combination chemotherapy regimens for metastatic disease increase optimism for future treatments. With greater control of overt or occult metastatic disease, there will likely be an expanding role for local treatment modalities, especially given that nearly a third of pancreatic cancer patients have locally destructive disease without distant metastatic disease at the time of death. Technical advances have allowed for the safe delivery of dose-escalated radiation therapy, which can then be combined with chemotherapy, targeted agents, immunotherapy, and nanoparticulate drug delivery techniques to produce novel and improved synergistic effects. Here we discuss recent advances and future directions for multimodality therapy in pancreatic cancer.
Collapse
|
15
|
Oh SY, Edwards A, Mandelson MT, Lin B, Dorer R, Helton WS, Kozarek RA, Picozzi VJ. Rare long-term survivors of pancreatic adenocarcinoma without curative resection. World J Gastroenterol 2015; 21:13574-13581. [PMID: 26730170 PMCID: PMC4690188 DOI: 10.3748/wjg.v21.i48.13574] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/11/2015] [Accepted: 09/28/2015] [Indexed: 02/06/2023] Open
Abstract
Long-term outcome data in pancreatic adenocarcinoma are predominantly based on surgical series, as resection is currently considered essential for long-term survival. In contrast, five-year survival in non-resected patients has rarely been reported. In this report, we examined the incidence and natural history of ≥ 5-year survivors with non-resected pancreatic adenocarcinoma. All patients with pancreatic adenocarcinoma who received oncologic therapy alone without surgery at our institution between 1995 and 2009 were identified. Non-resected ≥ 5-year survivors represented 2% (11/544) of all non-resected patients undergoing treatment for pancreatic adenocarcinoma, and 11% (11/98) of ≥ 5-year survivors. Nine patients had localized tumor and 2 metastatic disease at initial diagnosis. Disease progression occurred in 6 patients, and the local tumor bed was the most common site of progression. Six patients suffered from significant morbidities including recurrent cholangitis, second malignancy, malnutrition and bowel perforation. A rare subset of patients with pancreatic cancer achieve long-term survival without resection. Despite prolonged survival, morbidities unrelated to the primary cancer were frequently encountered and a close follow-up is warranted in these patients. Factors such as tumor biology and host immunity may play a key role in disease progression and survival.
Collapse
|
16
|
Krishnan S, Chadha AS, Suh Y, Chen HC, Rao A, Das P, Minsky BD, Mahmood U, Delclos ME, Sawakuchi GO, Beddar S, Katz MH, Fleming JB, Javle MM, Varadhachary GR, Wolff RA, Crane CH. Focal Radiation Therapy Dose Escalation Improves Overall Survival in Locally Advanced Pancreatic Cancer Patients Receiving Induction Chemotherapy and Consolidative Chemoradiation. Int J Radiat Oncol Biol Phys 2015; 94:755-65. [PMID: 26972648 DOI: 10.1016/j.ijrobp.2015.12.003] [Citation(s) in RCA: 265] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 11/16/2015] [Accepted: 12/04/2015] [Indexed: 12/18/2022]
Abstract
PURPOSE To review outcomes of locally advanced pancreatic cancer (LAPC) patients treated with dose-escalated intensity modulated radiation therapy (IMRT) with curative intent. METHODS AND MATERIALS A total of 200 patients with LAPC were treated with induction chemotherapy followed by chemoradiation between 2006 and 2014. Of these, 47 (24%) having tumors >1 cm from the luminal organs were selected for dose-escalated IMRT (biologically effective dose [BED] >70 Gy) using a simultaneous integrated boost technique, inspiration breath hold, and computed tomographic image guidance. Fractionation was optimized for coverage of gross tumor and luminal organ sparing. A 2- to 5-mm margin around the gross tumor volume was treated using a simultaneous integrated boost with a microscopic dose. Overall survival (OS), recurrence-free survival (RFS), local-regional and distant RFS, and time to local-regional and distant recurrence, calculated from start of chemoradiation, were the outcomes of interest. RESULTS Median radiation dose was 50.4 Gy (BED = 59.47 Gy) with a concurrent capecitabine-based (86%) regimen. Patients who received BED >70 Gy had a superior OS (17.8 vs 15.0 months, P=.03), which was preserved throughout the follow-up period, with estimated OS rates at 2 years of 36% versus 19% and at 3 years of 31% versus 9% along with improved local-regional RFS (10.2 vs 6.2 months, P=.05) as compared with those receiving BED ≤70 Gy. Degree of gross tumor volume coverage did not seem to affect outcomes. No additional toxicity was observed in the high-dose group. Higher dose (BED) was the only predictor of improved OS on multivariate analysis. CONCLUSION Radiation dose escalation during consolidative chemoradiation therapy after induction chemotherapy for LAPC patients improves OS and local-regional RFS.
Collapse
Affiliation(s)
- Sunil Krishnan
- Department of Radiation Oncology, The University of Texas, Houston, Texas.
| | - Awalpreet S Chadha
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Yelin Suh
- Department of Radiation Physics, The University of Texas, Houston, Texas
| | - Hsiang-Chun Chen
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas
| | - Arvind Rao
- Department of Bioinformatics and Computational Biology, MD Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Bruce D Minsky
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Usama Mahmood
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Marc E Delclos
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Gabriel O Sawakuchi
- Department of Radiation Physics, The University of Texas, Houston, Texas; Graduate School of Biomedical Sciences, The University of Texas, Houston, Texas
| | - Sam Beddar
- Department of Radiation Physics, The University of Texas, Houston, Texas
| | - Matthew H Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Milind M Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gauri R Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
17
|
Chavalitdhamrong D, DiMaio CJ, Siersema PD, Wagh MS. Technical advances in endoscopic ultrasound-guided fiducial placement for the treatment of pancreatic cancer. Endosc Int Open 2015; 3:E373-E377. [PMID: 26355267 PMCID: PMC4554510 DOI: 10.1055/s-0034-1392274] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/26/2015] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy has an important role in the treatment of locally advanced or metastatic pancreatic cancer and can be used alone or in conjunction with surgery and/or systemic chemotherapy. Because of the challenge of delivering an accurate and optimal radiation dose, image-guided radiation therapy can be used to improve targeting. Fiducial markers can be placed in the tumor and used for localization in patients undergoing image-guided radiation therapy. The safety and feasibility of endoscopic ultrasound (EUS)-guided placement of fiducials has been assessed and reported for the management of pancreatic cancer. We herein review the technique, efficacy, and safety profile of EUS-guided fiducial placement. In addition, we highlight recent advances and technological upgrades in EUS-guided fiducial delivery systems for pancreatic cancer most relevant to practicing gastroenterologists and interventional endoscopists.
Collapse
Affiliation(s)
| | - Christopher J. DiMaio
- Division of Gastroenterology, Mount Sinai School of Medicine, New York, New York, United States
| | - Peter D. Siersema
- Department of Gastroenterology, University Medical Center Utrecht, The Netherlands
| | - Mihir S. Wagh
- Division of Gastroenterology, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
18
|
Chadha AS, Kocak-Uzel E, Das P, Minsky BD, Delclos ME, Mahmood U, Guha S, Ahmad M, Varadhachary GR, Javle M, Katz MH, Fleming JB, Wolff RA, Crane CH, Krishnan S. Paraneoplastic thrombocytosis independently predicts poor prognosis in patients with locally advanced pancreatic cancer. Acta Oncol 2015; 54:971-978. [PMID: 25608822 DOI: 10.3109/0284186x.2014.1000466] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/10/2014] [Indexed: 11/13/2022]
Abstract
BACKGROUND AND AIMS Platelets are believed to promote tumor growth and metastasis but their prognostic role in locally advanced pancreatic cancer (LAPC) remains largely unknown. We assessed whether pretreatment platelet counts independently predict survival outcomes in patients with LAPC treated with chemoradiation (CRT). METHODS We retrospectively reviewed the MD Anderson pancreatic cancer database and identified 199 patients with LAPC treated with CRT between 2006 and 2012. Induction chemotherapy was used prior to consolidative CRT in 177 (89%) patients. Median radiation dose was 50.4 Gy. Concurrent radiosensitizers were gemcitabine-based (13%) or capecitabine-based (84%) regimens. Actuarial univariate and multivariate statistical methods were used to determine significant prognostic factors for overall survival (OS) and progression-free survival (PFS) calculated from the start of treatment. RESULTS Median follow-up was 9.9 months. Median OS and PFS durations were 17.7 and 10.7 months, respectively. On univariate analysis, platelet count > 300 K/μl, KPS ≤ 80, ≥ 5% weight loss and pretreatment CA19-9 above the median were associated with inferior OS or PFS. Median OS was lower in patients with platelet count > 300 K/μl compared to patients with platelet count ≤ 300 K/μl (10.2 vs. 19 months; p = 0.0002). Corresponding median PFS times were 7.8 months and 11.1 months (p = 0.004), respectively. On multivariate analysis, platelet count > 300 K/μl (p = 0.012), ≥ 5% weight loss (p = 0.002) and elevated pretreatment CA19-9 (p = 0.005) were independent prognostic factors for OS. Platelet count > 300 K/μl (p = 0.03) and KPS ≤ 80 (p = 0.05) independently predicted PFS. CONCLUSIONS Our analysis suggests that pretreatment thrombocytosis independently predicts inferior OS and PFS in LAPC.
Collapse
Affiliation(s)
- Awalpreet S Chadha
- Department of Radiation Oncology, The University of Texas M D Anderson Cancer Center , Houston, Texas , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Buckel L, Savariar EN, Crisp JL, Jones KA, Hicks AM, Scanderbeg DJ, Nguyen QT, Sicklick JK, Lowy AM, Tsien RY, Advani SJ. Tumor radiosensitization by monomethyl auristatin E: mechanism of action and targeted delivery. Cancer Res 2015; 75:1376-1387. [PMID: 25681274 DOI: 10.1158/0008-5472.can-14-1931] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 02/04/2015] [Indexed: 12/19/2022]
Abstract
Intrinsic tumor resistance to radiotherapy limits the efficacy of ionizing radiation (IR). Sensitizing cancer cells specifically to IR would improve tumor control and decrease normal tissue toxicity. The development of tumor-targeting technologies allows for developing potent radiosensitizing drugs. We hypothesized that the anti-tubulin agent monomethyl auristatin E (MMAE), a component of a clinically approved antibody-directed conjugate, could function as a potent radiosensitizer and be selectively delivered to tumors using an activatable cell-penetrating peptide targeting matrix metalloproteinases and RGD-binding integrins (ACPP-cRGD-MMAE). We evaluated the ability of MMAE to radiosensitize both established cancer cells and a low-passage cultured human pancreatic tumor cell line using clonogenic and DNA damage assays. MMAE sensitized colorectal and pancreatic cancer cells to IR in a schedule- and dose-dependent manner, correlating with mitotic arrest. Radiosensitization was evidenced by decreased clonogenic survival and increased DNA double-strand breaks in irradiated cells treated with MMAE. MMAE in combination with IR resulted in increased DNA damage signaling and activation of CHK1. To test a therapeutic strategy of MMAE and IR, PANC-1 or HCT-116 murine tumor xenografts were treated with nontargeted free MMAE or tumor-targeted MMAE (ACPP-cRGD-MMAE). While free MMAE in combination with IR resulted in tumor growth delay, tumor-targeted ACPP-cRGD-MMAE with IR produced a more robust and significantly prolonged tumor regression in xenograft models. Our studies identify MMAE as a potent radiosensitizer. Importantly, MMAE radiosensitization can be localized to tumors by targeted activatable cell-penetrating peptides.
Collapse
Affiliation(s)
- Lisa Buckel
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | - Angel M Hicks
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | | | - Roger Y Tsien
- Department of Pharmacology.,Howard Hughes Medical Institute
| | - Sunil J Advani
- Department of Radiation Medicine and Applied Sciences.,Center for Advanced Radiotherapy Technologies University of California San Diego
| |
Collapse
|
20
|
Wang Z, Ren ZG, Ma NY, Zhao JD, Zhang Z, Ma XJ, Long J, Xu J, Jiang GL. Intensity modulated radiotherapy for locally advanced and metastatic pancreatic cancer: a mono-institutional retrospective analysis. Radiat Oncol 2015; 10:14. [PMID: 25575617 PMCID: PMC4296685 DOI: 10.1186/s13014-014-0312-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 12/17/2014] [Indexed: 12/12/2022] Open
Abstract
Background To evaluate the role of intensity modulated radiotherapy (IMRT) for locally advanced pancreatic cancer (LAPC) and metastatic pancreatic cancer (MPC), and the prognostic factors in the setting of multidisciplinary approach strategies. Methods 63 patients with LAPC and MPC receiving IMRT in our institution were retrospectively identified. Information on patient baseline, treatment characteristics and overall survival (OS) time were collected. Data of pain relief and toxicity were evaluated. Univariate and multivariate analyses were conducted to investigate the prognostic factors. Results All patients received IMRT with a median dose of 46.0 Gy. The median OS for LAPC and MPC patients were 15.7 months and 8.0 months, respectively (p = 0.029). Symptomatic improvements were observed in the 44 patients with abdominal/back pain after radiotherapy (RT) or concurrent chemoradiotherapy (CCRT), particularly in those with severe pain. Only 13.9% and 14.8% cases presented Grade ≥ 3 hematologic toxicities in RT and CCRT group, while no cases developed Grade ≥ 3 non-hematologic toxicities in both groups. Multivariate analysis indicated that tumors located in pancreas body/tail (HR 0.28, p = 0.008), pretreatment CA19-9 < 1000 U/mL (HR 0.36, p = 0.029) and concurrent chemotherapy (HR 0.37, p = 0.016) were independent favorable predictors for OS. Conclusions CCRT further improved OS for LAPC and MPC with acceptable toxicities, and use of RT markedly alleviated pain. Tumors located in pancreas body/tail, pretreatment CA19-9 level of < 1000 U/mL and CCRT were associated with better OS. However, regional intra-arterial chemotherapy did not show any survival benefit in our study.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Zhi-Gang Ren
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Ning-Yi Ma
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Jian-Dong Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Xue-Jun Ma
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Jin Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China.
| | - Guo-Liang Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032, China. .,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, 4365 Kangxin Road, Shanghai, 201321, China.
| |
Collapse
|
21
|
Segmentation precision of abdominal anatomy for MRI-based radiotherapy. Med Dosim 2014; 39:212-7. [PMID: 24726701 DOI: 10.1016/j.meddos.2014.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 12/20/2013] [Accepted: 02/10/2014] [Indexed: 01/24/2023]
Abstract
The limited soft tissue visualization provided by computed tomography, the standard imaging modality for radiotherapy treatment planning and daily localization, has motivated studies on the use of magnetic resonance imaging (MRI) for better characterization of treatment sites, such as the prostate and head and neck. However, no studies have been conducted on MRI-based segmentation for the abdomen, a site that could greatly benefit from enhanced soft tissue targeting. We investigated the interobserver and intraobserver precision in segmentation of abdominal organs on MR images for treatment planning and localization. Manual segmentation of 8 abdominal organs was performed by 3 independent observers on MR images acquired from 14 healthy subjects. Observers repeated segmentation 4 separate times for each image set. Interobserver and intraobserver contouring precision was assessed by computing 3-dimensional overlap (Dice coefficient [DC]) and distance to agreement (Hausdorff distance [HD]) of segmented organs. The mean and standard deviation of intraobserver and interobserver DC and HD values were DC(intraobserver) = 0.89 ± 0.12, HD(intraobserver) = 3.6mm ± 1.5, DC(interobserver) = 0.89 ± 0.15, and HD(interobserver) = 3.2mm ± 1.4. Overall, metrics indicated good interobserver/intraobserver precision (mean DC > 0.7, mean HD < 4mm). Results suggest that MRI offers good segmentation precision for abdominal sites. These findings support the utility of MRI for abdominal planning and localization, as emerging MRI technologies, techniques, and onboard imaging devices are beginning to enable MRI-based radiotherapy.
Collapse
|
22
|
Li L, Hao X, Qin J, Tang W, He F, Smith A, Zhang M, Simeone DM, Qiao XT, Chen ZN, Lawrence TS, Xu L. Antibody against CD44s inhibits pancreatic tumor initiation and postradiation recurrence in mice. Gastroenterology 2014; 146:1108-18. [PMID: 24397969 PMCID: PMC3982149 DOI: 10.1053/j.gastro.2013.12.035] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 12/23/2013] [Accepted: 12/27/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS CD44s is a surface marker of tumor-initiating cells (TICs); high tumor levels correlate with metastasis and recurrence, as well as poor outcomes for patients. Monoclonal antibodies against CD44s might eliminate TICs with minimal toxicity. This strategy is unclear for treatment of pancreatic cancer, and little is known about how anti-CD44s affect pancreatic cancer initiation or recurrence after radiotherapy. METHODS One hundred ninety-two pairs of human pancreatic adenocarcinoma and adjacent nontumor pancreatic tissues were collected from patients undergoing surgery. We measured CD44s levels in tissue samples and pancreatic cancer cell lines by immunohistochemistry, real-time polymerase chain reaction, and immunoblot; levels were correlated with patient survival times. We studied the effects of anti-CD44s in mice with human pancreatic tumor xenografts and used flow cytometry to determine the effects on TICs. Changes in CD44s signaling were examined by real-time polymerase chain reaction, immunoblot, reporter assay, and in vitro tumorsphere formation assays. RESULTS Levels of CD44s were significantly higher in pancreatic cancer than adjacent nontumor tissues. Patients whose tumors expressed high levels of CD44s had a median survival of 10 months compared with >43 months for those with low levels. Anti-CD44s reduced growth, metastasis, and postradiation recurrence of pancreatic xenograft tumors in mice. The antibody reduced the number of TICs in cultured pancreatic cancer cells and xenograft tumors, as well as their tumorigenicity. In cultured pancreatic cancer cell lines, anti-CD44s down-regulated the stem cell self-renewal genes Nanog, Sox-2, and Rex-1 and inhibited signal transducer and activator of transcription 3-mediated cell proliferation and survival signaling. CONCLUSIONS The TIC marker CD44s is up-regulated in human pancreatic tumors and associated with patient survival time. CD44s is required for initiation, growth, metastasis, and postradiation recurrence of xenograft tumors in mice. Anti-CD44s eliminated bulk tumor cells as well as TICs from the tumors. Strategies to target CD44s cab be developed to block pancreatic tumor formation and post-radiotherapy recurrence in patients.
Collapse
Affiliation(s)
- Ling Li
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan; Cell Engineering Research Centre and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Xinbao Hao
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan; Department of Hematology/Oncology, Hainan Medical College Hospital, Haikou, Hainan, P.R. China
| | - Jun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Wenhua Tang
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan; Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, Kansas
| | - Fengtian He
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Amber Smith
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, Kansas
| | - Min Zhang
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Diane M Simeone
- Department of Surgery, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Xiaotan T Qiao
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Zhi-Nan Chen
- Cell Engineering Research Centre and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, P.R. China.
| | - Theodore S Lawrence
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Liang Xu
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan; Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, Kansas.
| |
Collapse
|
23
|
Wang H, Wang J, Jiang Y, Li J, Tian S, Ran W, Xiu D, Gao Y. The investigation of 125I seed implantation as a salvage modality for unresectable pancreatic carcinoma. J Exp Clin Cancer Res 2013; 32:106. [PMID: 24370348 PMCID: PMC3892087 DOI: 10.1186/1756-9966-32-106] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 12/24/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND To assess the efficacy of intraoperative ultrasound-guided implantation of 125I seeds for the treatment of unresectable pancreatic carcinoma, and analyze the associated prognostic factors. METHODS Twenty-eight patients with pancreatic carcinoma who underwent laparotomy and were considered to have unresectable tumors were included in this study. Nine patients were pathologically diagnosed with Stage II disease, and nineteen patients with Stage III disease. Twenty-eight patients received intraoperative ultrasound-guided 125I seed implantation and received a D90 (at least 90% of the tumor volume received the reference dose) ranging from 60 to 163 Gy, with a median of 120 Gy. Seven patients received an additional 35-50 Gy external beam radiotherapy after seed implantation, and ten patients received two to ten cycles of chemotherapy. Overall survival of the patients was calculated and prognostic factors were evaluated. RESULTS Of the patients, 94.1% (16/17) achieved good to medium pain relief. The tumor response rate was 78.6% (22/28), and local control was achieved in 85.7% (24/28) of patients. The 1-, 2- and 3-year survival rates were 30%, 11% and 4%, and the median survival was 10.1 months (95% CI: 9.0-10.9). Analysis using the Cox proportional hazards model suggested that patients younger than 60 years and patients who received a D90 higher than 110 Gy may survive for a longer period. CONCLUSIONS I seed implantation provides a safe and effective method to relieve pain, control local tumor growth and, to some extent, prolong the survival of patients with stage II and III pancreatic disease, without additional complications. Age and accumulated dose may be factors predictive of a favorable outcome for patients with unresectable pancreatic carcinoma treated with 125I seeds. These findings need to be validated by conducting further studies with larger cohorts.
Collapse
Affiliation(s)
- Hao Wang
- Department of Radiation Oncology, Peking University 3rd Hospital, Huan-yuan North Road 49th, Beijing 100083, P. R. China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University 3rd Hospital, Huan-yuan North Road 49th, Beijing 100083, P. R. China
| | - Yuliang Jiang
- Department of Radiation Oncology, Peking University 3rd Hospital, Huan-yuan North Road 49th, Beijing 100083, P. R. China
| | - Jinna Li
- Department of Radiation Oncology, Peking University 3rd Hospital, Huan-yuan North Road 49th, Beijing 100083, P. R. China
| | - Suqing Tian
- Department of Radiation Oncology, Peking University 3rd Hospital, Huan-yuan North Road 49th, Beijing 100083, P. R. China
| | - Weiqiang Ran
- Department of Ultrasound, Peking University 3rd Hospital, Beijing 100083, P. R. China
| | - Dianrong Xiu
- Department of Surgery, Peking University 3rd Hospital, Beijing 100083, P. R. China
| | - Yang Gao
- Department of Radiation Oncology, Peking University 3rd Hospital, Huan-yuan North Road 49th, Beijing 100083, P. R. China
| |
Collapse
|
24
|
Draganov PV, Chavalitdhamrong D, Wagh MS. Evaluation of a new endoscopic ultrasound-guided multi-fiducial delivery system: a prospective non-survival study in a live porcine model. Dig Endosc 2013; 25:615-621. [PMID: 23489989 DOI: 10.1111/den.12046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/09/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Endoscopic ultrasound (EUS) is an excellent tool for implanting fiducials for gastrointestinal malignancies, but a dedicated delivery system for EUS-guided fiducial placement is not available. The aim of the present study was to evaluate the performance characteristics of a new dedicated EUS-guided multi-fiducial delivery system. METHODS This was a prospective live porcine study using prototype 22-G EUS needles preloaded with four fiducials. Primary outcomes were technical success and accuracy of fiducial placement. Secondary outcomes were ease of passage of delivery system, ease of deployment, EUS visualization of the needle, visual appearance of fiducials on EUS, fluoroscopy and computed tomography (CT) scans, time taken for fiducial placement and adverse events. Outcomes were scored on a predetermined 5-point Likert scale (1 = best, 5 = worst). RESULTS Fiducial deployment was successful in 43 out of 45 needles (95.6%). All 172 fiducials (43 needles, four fiducials each)were successfully deployed on target. Fiducial placement accuracy score was 1.30 ± 0.64. All needles passed with ease (score 1.0 ± 0.0). Ease of fiducial deployment score was 1.85 ± 1.02. Mean scores for EUS visualization of the needle and fiducial were 1.02 ± 0.15 and 2.10 ± 0.91, respectively. Fluoroscopic and CT visualization of fiducials was excellent. Time for placement of four fiducials was <1 min (0.86 ± 0.50 min). No adverse events were seen. CONCLUSION The new EUS-guided multi-fiducial delivery system provided quick, easy, and accurate fiducial deployment without adverse events.
Collapse
Affiliation(s)
- Peter V Draganov
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, USA
| | | | | |
Collapse
|
25
|
Shin EJ, Khashab M. The role of endoscopy in the treatment, management, and personalization of pancreatic cancer. Curr Probl Cancer 2013; 37:293-300. [PMID: 24331185 DOI: 10.1016/j.currproblcancer.2013.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
26
|
Wei F, Liu Y, Guo Y, Xiang A, Wang G, Xue X, Lu Z. miR-99b-targeted mTOR induction contributes to irradiation resistance in pancreatic cancer. Mol Cancer 2013; 12:81. [PMID: 23886294 PMCID: PMC3726417 DOI: 10.1186/1476-4598-12-81] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/22/2013] [Indexed: 02/08/2023] Open
Abstract
Background Radiation exerts direct antitumor effects and is widely used in clinics, but the efficacy is severely compromised by tumor resistance. Therefore uncovering the mechanism of radioresistance might promote the development of new strategies to overcome radioresistance by manipulating activity of the key molecules. Methods Immunohistochemistry were used to find whether mTOR were over-activated in radioresistant patients’ biopsies. Then Western blot, real-time PCR and transfection were used to find whether radiotherapy regulates the expression and activity of mTOR by modulating its targeting microRNA in human pancreatic cancer cell lines PANC-1, Capan-2 and BxPC-3. Finally efficacy of radiation combined with mTOR dual inhibitor AZD8055 was assessed in vitro and in vivo. Results Ionizing radiation promoted mTOR expression and activation in pancreatic cancer cells through reducing miR-99b expression, which negatively regulated mTOR. Novel mTOR inhibitor, AZD8055 (10 nM, 100 nM, 500 nM) synergistically promoted radiation (0–10 Gy) induced cell growth inhibition and apoptosis. In human pancreatic cancer xenografts, fractionated radiation combined with AZD8055 treatment further increased the anti-tumor effect, the tumor volume was shrinked to 278 mm3 after combination treatment for 3 weeks compared with single radiation (678 mm3) or AZD8055 (708 mm3) treatment (P < 0.01). Conclusions Our data provide a rationale for overcoming radio-resistance by combined with mTOR inhibitor AZD8055 in pancreatic cancer therapy.
Collapse
|
27
|
Golden DW, Novak CJ, Minsky BD, Liauw SL. Radiation dose ≥54 Gy and CA 19-9 response are associated with improved survival for unresectable, non-metastatic pancreatic cancer treated with chemoradiation. Radiat Oncol 2012; 7:156. [PMID: 22974515 PMCID: PMC3527337 DOI: 10.1186/1748-717x-7-156] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 09/08/2012] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Unresectable pancreatic cancer (UPC) has low survival. With improving staging techniques and systemic therapy, local control in patients without metastatic disease may have increasing importance. We investigated whether the radiation dose used in chemoradiation (CRT) as definitive treatment for UPC and the CA 19-9 response to therapy have an impact on overall survival (OS). METHODS From 1997-2009 46 patients were treated with CRT for non-metastatic UPC. Median prescribed RT dose was 54 Gy (range 50.4-59.4 Gy). All patients received concurrent chemotherapy (41: 5-fluorouracil, 5: other) and 24 received adjuvant chemotherapy. RESULTS 41 patients were inoperable due to T4 disease and 5 patients with T3 disease were medically inoperable. Five patients did not complete CRT due to progressive disease or treatment-related toxicity (median RT dose 43.2 Gy). Overall, 42 patients were dead of disease at the time of last follow-up. The median and 12 month OS were 8.8 months and 35%, respectively. By univariate analysis, minimum CA 19-9 post-CRT <90 U/mL was favorably associated with OS (12.3 versus 8.8 months, p = 0.012). Radiotherapy dose ≥54 Gy trended towards improved OS (11.3 versus 6.8 months, p = 0.089). By multivariable analysis, a delivered RT dose of ≥54 Gy (HR 0.47, p = 0.028) and minimum CA 19-9 post-CRT of <90 U/mL (HR 0.35, p = 0.008) were associated with OS. CONCLUSIONS CRT as definitive treatment for UPC had low survival. However, our retrospective data suggest that patients treated to ≥54 Gy or observed to have a minimum post-CRT CA 19-9 <90 U/mL had improved likelihood of long-term survival.
Collapse
Affiliation(s)
- Daniel W Golden
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
28
|
Personalized medicine in pancreatic cancer: prognosis and potential implications for therapy. J Gastrointest Surg 2012; 16:1651-2. [PMID: 22744639 PMCID: PMC3556913 DOI: 10.1007/s11605-012-1943-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 06/15/2012] [Indexed: 01/31/2023]
|
29
|
K-Ras mutation-mediated IGF-1-induced feedback ERK activation contributes to the rapalog resistance in pancreatic ductal adenocarcinomas. Cancer Lett 2012; 322:58-69. [PMID: 22342683 DOI: 10.1016/j.canlet.2012.02.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 02/06/2012] [Accepted: 02/07/2012] [Indexed: 12/25/2022]
Abstract
Mammalian target of rapamycin complex 1 (mTORC1) is frequently activated in human cancers; however, clinical trials of rapalog (the mTORC1 inhibitors) have shown that pancreatic ductal adenocarcinomas (PDACs) resist to the treatment. Rapalog treatment activated the extracellular signal-regulated kinase (ERK) pathway in K-Ras mt PDAC cells. K-Ras knockdown abolished the insulin-like growth factor-1 (IGF-1)-induced ERK pathway in the K-Ras mt PDAC cells and enhanced the therapeutic efficacy of everolimus in treating K-Ras mt PDAC cells-derived mouse xenografts. The results indicate that targeting of K-Ras mutation may lead to the development of therapies that overcome rapalog resistance in PDAC.
Collapse
|
30
|
Zeng H, Yu H, Lu L, Jain D, Kidd MS, Saif MW, Chanock SJ, Hartge P, Risch HA. Genetic effects and modifiers of radiotherapy and chemotherapy on survival in pancreatic cancer. Pancreas 2011; 40:657-63. [PMID: 21487324 PMCID: PMC3116071 DOI: 10.1097/mpa.0b013e31821268d1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Germ-line genetic variation may affect clinical outcomes of cancer patients. We applied a candidate-gene approach to evaluate the effect of putative markers on survival of patients with pancreatic cancer. We also examined gene-radiotherapy and gene-chemotherapy interactions, aiming to explain interindividual differences in treatment outcomes. METHODS In total, 211 patients with pancreatic cancer were recruited in a population-based study. Sixty-four candidate genes associated with cancer survival or treatment response were selected from existing publications. Genotype information was obtained from a previous genome-wide association study data set. The main effects of genetic variation and gene-specific treatment interactions on overall survival were examined by proportional hazards regression models. RESULTS Fourteen genes showed evidence of association with pancreatic cancer survival. Among these, rs1760217, located at the DPYD gene; rs17091162 at SERPINA3; and rs2231164 at ABCG2 had the lowest P of 10(-4.60), 0.0013, and 0.0023, respectively. We also observed that 2 genes, RRM1 and IQGAP2, had significant interactions with radiotherapy in association with survival, and 2 others, TYMS and MET, showed evidence of interaction with 5-fluorouracil and erlotinib, respectively. CONCLUSIONS Our study suggested significant associations between germ-line genetic polymorphisms and overall survival in pancreatic cancer, as well as survival interactions between various genes and radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Hongmei Zeng
- Department of Epidemiology and Public Health, School of Public Health and School of Medicine, Yale University, New Haven, Connecticut, USA
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, China
| | - Herbert Yu
- Department of Epidemiology and Public Health, School of Public Health and School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Lingeng Lu
- Department of Epidemiology and Public Health, School of Public Health and School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Dhanpat Jain
- Department of Pathology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Mark S. Kidd
- Department of Surgery, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - M. Wasif Saif
- Division of Hematology/Oncology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Stephen J. Chanock
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | | | - Harvey A. Risch
- Department of Epidemiology and Public Health, School of Public Health and School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|