1
|
Sweeney A, Langley A, Xavierselvan M, Shethia RT, Solomon P, Arora A, Mallidi S. Vascular regional analysis unveils differential responses to anti-angiogenic therapy in pancreatic xenografts through macroscopic photoacoustic imaging. Theranostics 2025; 15:2649-2671. [PMID: 39990229 PMCID: PMC11840746 DOI: 10.7150/thno.99361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/17/2024] [Indexed: 02/25/2025] Open
Abstract
Background: Amongst the various imaging techniques that provide surrogate tumor radiographic indications to aid in planning, monitoring, and predicting outcomes of therapy, ultrasound-guided photoacoustic imaging (US-PAI) is a promising non-ionizing modality based on endogenous blood (hemoglobin) and blood oxygen saturation (StO₂) contrast. Adaptation of US-PAI to the clinical realm requires macroscopic system configurations for adequate depth visualization. Methods: Here we present a vascular regional analysis (VRA) methodology of obtaining areas of low and high vessel density regions within the tumor (LVD and HVD respectively) by frequency domain filtering of macroscopic PA images. In this work, we evaluated the various vascular and oxygenation profiles of different murine xenografts of pancreatic cancer (AsPC-1, MIA PaCa-2, and BxPC-3) that have varying levels of angiogenic potentials and investigated the effects of receptor tyrosine kinase inhibitor (sunitinib) on the tumor microvessel density and StO₂. Results: The administration of sunitinib resulted in transient deoxygenation and reduction in vessel density within 72 h in two (AsPC-1 and MIA PaCa-2) of the three tumor types. Utilizing VRA, the regional change in StO2 (∆StO2) revealed the preferential targeting of sunitinib in LVD regions in only the AsPC-1 tumors. We also identified the presence of vascular normalization (validated through immunohistochemistry) in the sunitinib treated AsPC-1 tumors at day 8 post-treatment where a significant increases in HVD ∆StO2 (~20%) were seen following the 72-hour time point, indicative of improved vessel flow and functionality. Treated AsPC-1 vasculature displayed increased maturity and functionality compared to non-treated tumors on day 8, while these same metrics showed no conclusive evidence of vascular normalization in MIA PaCa-2 or BxPC-3 tumors. Conclusion: Overall, VRA as a tool to monitor treatment response allowed us to identify time points of vascular remodeling, highlighting its ability to provide insights into the tumor microenvironment for sunitinib treatment and other anti-angiogenic therapies.
Collapse
Affiliation(s)
- Allison Sweeney
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Andrew Langley
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Marvin Xavierselvan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Ronak T. Shethia
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Patrick Solomon
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Aayush Arora
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Srivalleesha Mallidi
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
2
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
3
|
Sweeney A, Xavierselvan M, Langley A, Solomon P, Arora A, Mallidi S. Vascular regional analysis unveils differential responses to anti-angiogenic therapy in pancreatic xenografts through macroscopic photoacoustic imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.595784. [PMID: 38854042 PMCID: PMC11160648 DOI: 10.1101/2024.05.27.595784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy and the third leading cause of cancer deaths in the U.S. Despite major innovations in imaging technologies, there are limited surrogate radiographic indicators to aid in therapy planning and monitoring. Amongst the various imaging techniques Ultrasound-guided photoacoustic imaging (US-PAI) is a promising modality based on endogenous blood (hemoglobin) and blood oxygen saturation (StO 2 ) contrast to monitor response to anti-angiogenic therapies. Adaptation of US-PAI to the clinical realm requires macroscopic configurations for adequate depth visualization, illuminating the need for surrogate radiographic markers, including the tumoral microvessel density (MVD). In this work, subcutaneous xenografts with PC cell lines AsPC-1 and MIA-PaCa-2 were used to investigate the effects of receptor tyrosine kinase inhibitor (sunitinib) treatment on MVD and StO 2 . Through histological correlation, we have shown that regions of high and low vascular density (HVD and LVD) can be identified through frequency domain filtering of macroscopic PA images which could not be garnered from purely global analysis. We utilized vascular regional analysis (VRA) of treatment-induced StO 2 and total hemoglobin (HbT) changes. VRA as a tool to monitor treatment response allowed us to identify potential timepoints of vascular remodeling, highlighting its ability to provide insights into the TME not only for sunitinib treatment but also other anti-angiogenic therapies.
Collapse
|
4
|
Sarfaty E, Khajoueinejad N, Zewde MG, Yu AT, Cohen NA. Surgical management of pancreatic ductal adenocarcinoma: a narrative review. Transl Gastroenterol Hepatol 2023; 8:39. [PMID: 38021357 PMCID: PMC10643215 DOI: 10.21037/tgh-23-27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/21/2023] [Indexed: 12/01/2023] Open
Abstract
Background and Objective Pancreatic ductal adenocarcinoma (PDAC) is the third-leading cause of cancer-related death in the United States and is projected to become the second-leading cause of cancer-related death by 2030. Despite advances in systemic and radiation therapy, for patients with surgically resectable PDAC, complete surgical resection is the only potentially curative treatment option. The conduct of a safe, technically excellent pancreatectomy is essential to achieve optimal perioperative outcomes and long-term survival. In this narrative review, evidence from large, well-executed studies and clinical trials examining the technical aspects of pancreatectomy is reviewed. Methods A search was conducted in PubMed, Medline, and Cochrane Review databases to identify English-language randomized clinical trials, meta-analyses, and systematic reviews assessing surgical aspects of pancreatectomy for PDAC published between 2010 to 2023. Key Content and Findings We identified retrospective and prospective studies evaluating the technical aspects of surgery for PDAC. In this review, we evaluate data on surgical techniques of pancreatectomy for PDAC, including the role of minimally invasive techniques, extent of lymphadenectomy, reconstruction options after pancreatoduodenectomy, and the role of surgical drainage. Conclusions Surgical resection has a critical role in the treatment of operable PDAC. While pancreatic cancer surgery is an active area of research, conducting a technically excellent surgical resection maintains paramount importance for both oncological and perioperative outcomes. In this review, we summarize the latest evidence on surgical technique for operable PDAC.
Collapse
Affiliation(s)
- Elad Sarfaty
- Division of Surgical Oncology, Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nazanin Khajoueinejad
- Division of Surgical Oncology, Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Makda G. Zewde
- Division of Surgical Oncology, Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allen T. Yu
- Division of Surgical Oncology, Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
5
|
Xu Y, Chen Y, Han F, Wu J, Zhang Y. Neoadjuvant therapy vs. upfront surgery for resectable pancreatic cancer: An update on a systematic review and meta-analysis. Biosci Trends 2021; 15:365-373. [PMID: 34759120 DOI: 10.5582/bst.2021.01459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The effectiveness of neoadjuvant therapy (NAT) remains controversial in the treatment of pancreatic cancer (PC). Therefore, this meta-analysis aimed to investigate the clinical differences between NAT and upfront surgery (US) in resectable pancreatic cancer (RPC). Eligible studies were retrieved from PubMed, Embase, and Cochrane Library. The endpoints assessed were R0 resection rate, pathological T stage < 2 rate, positive lymph node rate, and overall survival. A total of 4,588 potentially relevant studies were identified, and 13 studies were included in this study. In patients with RPC, this meta-analysis showed that NAT presented an increased R0 resection rate, pathological T stage < 2 rate, and a remarkably reduced positive lymph node rate compared to US. However, patients receiving NAT did not result in a significantly increased overall survival. These findings supported the application of NAT, especially as a patient selection strategy, in the management of RPC. Additional large clinical studies are needed to determine whether NAT is superior to US.
Collapse
Affiliation(s)
- Youyao Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.,Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yizhen Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.,Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Fang Han
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jia Wu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yuhua Zhang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Patel SH, Katz MHG, Ahmad SA. The Landmark Series: Preoperative Therapy for Pancreatic Cancer. Ann Surg Oncol 2021; 28:4104-4129. [PMID: 34047859 DOI: 10.1245/s10434-021-10075-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
The surgical treatment of pancreas ductal adenocarcinoma (PDAC) is plagued by high rates of distant recurrences despite complete resection, highlighting the importance of systemic therapy. Historically, patients with PDAC have been treated with postoperative therapy, but this sequencing strategy can be associated with the inability to complete therapy due to perioperative complications and prolonged recovery. In addition, a subset of patients progress early, irrespective of whether surgery is performed, highlighting the systemic nature of this disease. Preoperative therapy has increasingly been utilized in clinical practice, but studies examining its benefits are limited. In this Landmark Series, we will review seminal studies for resectable and borderline resectable PDAC.
Collapse
Affiliation(s)
- Sameer H Patel
- Department of Surgery and the Division of Surgical Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Matthew H G Katz
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Syed A Ahmad
- Department of Surgery and the Division of Surgical Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
7
|
Hwang SH, Park MS. [Radiologic Evaluation for Resectability of Pancreatic Adenocarcinoma]. TAEHAN YONGSANG UIHAKHOE CHI 2021; 82:315-334. [PMID: 36238739 PMCID: PMC9431945 DOI: 10.3348/jksr.2021.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/25/2022]
Abstract
Imaging studies play an important role in the detection, diagnosis, assessment of resectability, staging, and determination of patient-tailored treatment options for pancreatic adenocarcinoma. Recently, for patients diagnosed with borderline resectable or locally advanced pancreatic cancers, it is recommended to consider curative-intent surgery following neoadjuvant or palliative therapy, if possible. This review covers how to interpret imaging tests and what to consider when assessing resectability, diagnosing distant metastasis, and re-assessing the resectability of pancreatic cancer after neoadjuvant or palliative therapy.
Collapse
|
8
|
González IA, Kang LI, Williams GA, Liu J, DeNardo DG, Hawkins WG, Chatterjee D. Tumor-insular Complex in Neoadjuvant Treated Pancreatic Ductal Adenocarcinoma Is Associated With Higher Residual Tumor. Am J Surg Pathol 2020; 44:817-825. [PMID: 32091434 PMCID: PMC7225071 DOI: 10.1097/pas.0000000000001454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC) plays a vital role in treatment response, and therefore, patient survival. We and others have observed an intimate association of neoplastic ductal cells with non-neoplastic islet cells, recapitulating the ductoinsular complex. We define this phenomenon as tumor-insular complex (TIC). Herein, we describe the clinicopathologic characteristics of TIC in neoadjuvant treated PDAC cases for the first time. We retrospectively reviewed the pathology of 105 cases of neoadjuvant treated PDAC resected at our institution. TIC was noted in 35 cases (33.3%), the mean tumor bed size was 2.7±1.0 cm, mean percentage of residual tumor 40±28% and mean Residual Tumor Index (RTI) (an index previously established as a prognostic parameter by our group) was 1.1±1.0. TIC was significantly associated with perineural invasion (P=0.001), higher tumor bed size (P=0.007), percentage of residual tumor (P=0.009), RTI (P=0.001), ypT stage (P=0.045), and poor treatment response, grouped by a previously established criteria (P=0.010). Using our prior binary reported prognostic cutoff for RTI of ≤0.35 and >0.35, TIC was associated with a RTI >0.35 (P=0.002). Moreover, patients who did not receive neoadjuvant radiation were associated with a higher frequency of TIC (P=0.003). In this cohort, RTI but not TIC was also shown to be a significant independent prognosticator for recurrence-free survival and overall survival on multivariate analysis. In conclusion, TIC is significantly associated with a more aggressive neoplasm which shows a poor treatment response. Further studies will be needed to better understand the tumor biology of TICs.
Collapse
Affiliation(s)
- Iván A. González
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Liang-I Kang
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Gregory A. Williams
- Division of HPB and GI surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Jingxia Liu
- Division of Public Health, Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - David G. DeNardo
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - William G. Hawkins
- Division of HPB and GI surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Deyali Chatterjee
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
9
|
Kamarajah SK, Bundred JR, Boyle C, Oo J, Pandanaboyana S, Loveday B. Impact of neoadjuvant therapy on post-operative pancreatic fistula: a systematic review and meta-analysis. ANZ J Surg 2020; 90:2201-2210. [PMID: 32418344 DOI: 10.1111/ans.15885] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The use of neoadjuvant therapy (NAT) for pancreatic cancer is increasing, although its impact on post-operative pancreatic fistula (POPF) is variably reported. This systematic review and meta-analysis aimed to assess the impact of NAT on POPF. METHODS A systematic literature search until October 2019 identified studies reporting POPF following NAT (radiotherapy, chemotherapy or chemoradiotherapy) versus upfront resection. The primary outcome was overall POPF. Secondary outcomes included grade B/C POPF, delayed gastric emptying (DGE), post-operative pancreatic haemorrhage (PPH) and overall and major complications. RESULTS The search identified 24 studies: pancreatoduodenectomy (PD), 19 studies (n = 19 416) and distal pancreatectomy (DP), five studies (n = 477). Local staging was reported in 17 studies, with borderline resectable and locally advanced disease comprising 6% (0-100%) and 1% (0-33%) of the population, respectively. For PD, any NAT was significantly associated with lower rates of overall POPF (OR: 0.57, P < 0.001) and grade B/C POPF (OR: 0.55, P < 0.001). In DP, NAT was not associated with significantly lower rates of overall or grade B/C POPF. CONCLUSION NAT is associated with significantly lower rates of POPF after PD but not after DP. Further studies are required to determine whether NAT should be added to POPF risk calculators.
Collapse
Affiliation(s)
- Sivesh K Kamarajah
- Department of HPB and Transplant Surgery, The Freeman Hospital, Newcastle upon Tyne, Tyne and Wear, UK.,Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, Newcastle, UK
| | - James R Bundred
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Charles Boyle
- Department of HPB and Transplant Surgery, The Freeman Hospital, Newcastle upon Tyne, Tyne and Wear, UK
| | - June Oo
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sanjay Pandanaboyana
- Department of HPB and Transplant Surgery, The Freeman Hospital, Newcastle upon Tyne, Tyne and Wear, UK
| | - Benjamin Loveday
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Surgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Surgery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Okubo S, Kojima M, Matsuda Y, Hioki M, Shimizu Y, Toyama H, Morinaga S, Gotohda N, Uesaka K, Ishii G, Mino-Kenudson M, Takahashi S. Area of residual tumor (ART) can predict prognosis after post neoadjuvant therapy resection for pancreatic ductal adenocarcinoma. Sci Rep 2019; 9:17145. [PMID: 31748528 PMCID: PMC6868132 DOI: 10.1038/s41598-019-53801-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/05/2019] [Indexed: 12/17/2022] Open
Abstract
An increasing number of patients with pancreatic ductal adenocarcinoma (PDAC) have undergone resection after neoadjuvant therapy (NAT). We have reported Area of Residual Tumor (ART) as a useful pathological assessment method to predict patient outcomes after post NAT resection in various cancer types. The aim of this study was to assess the prognostic performance of ART in PDAC resected after NAT. Sixty-three patients with PDAC after post NAT resection were analyzed. The viable residual tumor area was outlined and the measurement of ART was performed using morphometric software. The results were compared with those of the College of American Pathologist (CAP) regression grading. Of 63 cases, 39 (62%) patients received chemoradiation therapy and 24 (38%) received chemotherapy only. The median value of ART was 163 mm2. Large ART with 220 mm2 as the cut-off was significantly associated with lymphatic invasion, vascular invasion and perineural invasion, while CAP regression grading was not associated with any clinicopathological features. By multivariate analysis, large ART (≥220 mm2) was an independent predictor of shorter relapse free survival. Together with our previous reports, an ART-based pathological assessment may become a useful method to predict patient outcomes after post NAT resection across various cancer types.
Collapse
Affiliation(s)
- Satoshi Okubo
- Division of pathology, Research Center for Innovative Oncology, National Cancer Center Hospital East, Chiba, Japan
- Department of Hepatobiliary and Pancreatic surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Motohiro Kojima
- Division of pathology, Research Center for Innovative Oncology, National Cancer Center Hospital East, Chiba, Japan.
| | - Yoko Matsuda
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Sakae-cho, Itabashi-ku, Tokyo, Japan
| | - Masayoshi Hioki
- Department of Gastroenterological Surgery, Fukuyama City Hospital, Okayama, Japan
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Aichi, Japan
| | - Hirochika Toyama
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Soichiro Morinaga
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Naoto Gotohda
- Department of Hepatobiliary and Pancreatic surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Katsuhiko Uesaka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Genichiro Ishii
- Division of pathology, Research Center for Innovative Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shinichiro Takahashi
- Department of Hepatobiliary and Pancreatic surgery, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
11
|
Liu F, Ma J, Wang K, Li Z, Jiang Q, Chen H, Li W, Xia J. High expression of PDE4D correlates with poor prognosis and clinical progression in pancreaticductal adenocarcinoma. J Cancer 2019; 10:6252-6260. [PMID: 31772658 PMCID: PMC6856734 DOI: 10.7150/jca.35443] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/26/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Phosphodiesterase 4D (PDE4D) has recently been reported as an oncogene in various types of human cancers. However, the expression and significance of PDE4D in pancreatic ductal adenocarcinoma (PDAC) have not been elucidated. Methods: Immunohistochemistry (IHC) was used to examine the expression of PDE4D in 104 clinicopathologically characterized PDAC cases. PDE4D expression in paired tumor tissues and adjacent noncancerous tissues were detected by western blotting and real time qRT-PCR. The correlation of PDE4D expression levels with clinicopathological features and prognosis in patients were analyzed by univariate and multivariate methods. Effect of PDE4D on pancreatic cancer cells was detected by cell migration and invasion assays. Results: We found that PDE4D was significantly up-regulated in PDAC tumor tissues compared to those paired adjacent noncancerous tissues at both protein and mRNA levels. High level of PDE4D was significantly associated with clinical stage (P = 0.004), T classification (P = 0.003), lymph node metastasis (P = 0.022) and liver metastasis (P = 0.038). Patients with higher levels of PDE4D had shorter overall survival time contrast with those with lower PDE4D expression (P = 0.002). Multivariate analysis indicated that PDE4D may be an independent prognostic factor for PDAC. PDE4D depletion significantly suppressed β-catenin and Snail expression as well as the migration and invasion abilities of pancreatic cancer cells. Conclusions: Our study reveals that PDE4D up-regulated in PDAC was closely associated with poor prognosis of PDAC patients and multiple aggressive clinicopathological characteristics. PDE4D could be a useful prognostic biomarker and therapeutic target for PDAC.
Collapse
Affiliation(s)
- Fude Liu
- Department of General Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Jieyi Ma
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Kebing Wang
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhi Li
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Qingping Jiang
- Department of Pathology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Hui Chen
- Department of Pathology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Wen Li
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jintang Xia
- Department of General Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| |
Collapse
|
12
|
Diagnostic and therapeutic recommendations in pancreatic ductal adenocarcinoma. Recommendations of the Working Group of the Polish Pancreatic Club. GASTROENTEROLOGY REVIEW 2019; 14:1-18. [PMID: 30944673 PMCID: PMC6444110 DOI: 10.5114/pg.2019.83422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 02/15/2019] [Indexed: 02/07/2023]
Abstract
These recommendations refer to the current management in pancreatic ductal adenocarcinoma (PDAC), a neoplasia characterised by an aggressive course and extremely poor prognosis. The recommendations regard diagnosis, surgical, adjuvant and palliative treatment, with consideration given to endoscopic and surgical methods. A vast majority of the statements are based on data obtained in clinical studies and experts' recommendations on PDAC management, including the following guidelines: International Association of Pancreatology/European Pancreatic Club (IAP/EPC), American Society of Clinical Oncology (ASCO), European Society for Medical Oncology (ESMO), National Comprehensive Cancer Network (NCCN) and Polish Society of Gastroenterology (PSG) and The National Institute for Health and Care Excellence (NICE). All recommendations were voted on by members of the Working Group of the Polish Pancreatic Club. Results of the voting and brief comments are provided with each recommendation.
Collapse
|
13
|
Song E, Segarra D, Latifi K, Leuthold S, Belinc D, Pena L, Malafa MP, Frakes JM, Hoffe SE. Recognition of Tumor Invasion of a Pancreatic Head Biliary Stent During Stereotactic Body Radiation Therapy. Pract Radiat Oncol 2019; 9:132-135. [PMID: 30708132 DOI: 10.1016/j.prro.2019.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 11/26/2022]
Affiliation(s)
- Ethan Song
- University of South Florida Health Morsani College of Medicine, Tampa, Florida.
| | - Daniel Segarra
- University of South Florida Health Morsani College of Medicine, Tampa, Florida
| | - Kujtim Latifi
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Susan Leuthold
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Dalila Belinc
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Luis Pena
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Mokenge P Malafa
- Department of Surgical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Jessica M Frakes
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Sarah E Hoffe
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
14
|
Ren X, Wei X, Ding Y, Qi F, Zhang Y, Hu X, Qin C, Li X. Comparison of neoadjuvant therapy and upfront surgery in resectable pancreatic cancer: a meta-analysis and systematic review. Onco Targets Ther 2019; 12:733-744. [PMID: 30774360 PMCID: PMC6348975 DOI: 10.2147/ott.s190810] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective The role of neoadjuvant therapy (NAT) in resectable pancreatic cancer (RPC) remains controversial. Therefore, this meta-analysis was performed to compare the clinical differences between NAT and upfront surgery in RPC. Materials and methods A systematic literature search was performed in PubMed, Embase, Web of Science, and the Cochrane Register of Controlled Trials databases. Only patients with RPC who underwent tumor resection and received adjuvant or neoadjuvant treatment were enrolled. The OR or HR and 95% CIs were calculated employing fixed-effects or random-effects models. The HR and its 95% CI were extracted from each article that provided survival curve. Publication bias was estimated using funnel plots and Egger’s regression test. Results In total, eleven studies were included with 9,386 patients. Of these patients, 2,508 (26.7%) received NAT. For patients with RPC, NAT resulted in an increased R0 resection rate (OR=1.89; 95% CI=1.26–2.83) and a reduced positive lymph node rate (OR=0.34; 95% CI=0.31–0.37) compared with upfront surgery. Nevertheless, patients receiving NAT did not exhibit a significantly increased overall survival (OS) time (HR=0.91; 95% CI=0.79–1.05). Conclusion In patients with RPC, R0 resection rate and positive lymph node rate after NAT were superior to those of patients with upfront surgery. The NAT group exhibited no significant effect on OS time when compared with the upfront surgery group. However, this conclusion requires more clinical evidence to improve its credibility.
Collapse
Affiliation(s)
- Xiaohan Ren
- Department of First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Xiyi Wei
- Department of First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Yichao Ding
- Department of First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Feng Qi
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China,
| | - Yundi Zhang
- Department of First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Xin Hu
- Department of First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China,
| | - Xiao Li
- Department of Urology, Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China,
| |
Collapse
|
15
|
|
16
|
Sielaff CM, Mousa SA. Status and future directions in the management of pancreatic cancer: potential impact of nanotechnology. J Cancer Res Clin Oncol 2018; 144:1205-1217. [PMID: 29721665 DOI: 10.1007/s00432-018-2651-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at a late stage, has limited treatments, and patients have poor survival rates. It currently ranks as the seventh leading cause of cancer deaths globally and has increasing rates of diagnosis. Improved PDAC treatment requires the development of innovative, effective, and economical therapeutic drugs. The late stage diagnosis limits options for surgical resection, and traditional PDAC chemotherapeutics correlate with increased organ and hematologic toxicity. In addition, PDAC tumor tissue is dense and highly resistant to many traditional chemotherapeutic applications, making the disease difficult to treat and impeding options for palliative care. New developments in nanotechnology may offer innovative options for targeted PDAC therapeutic drug delivery. Nanotechnology can be implemented using multimodality methods that offer increased opportunities for earlier diagnosis, precision enhanced imaging, targeted long-term tumor surveillance, and controlled drug delivery, as well as improved palliative care and patient comfort. Nanoscale delivery methods have demonstrated the capacity to infiltrate the dense, fibrous tumor tissue associated with PDAC, increasing delivery and effectiveness of chemotherapeutic agents and reducing toxicity through the loading of multiple drug therapies on a single nano delivery vehicle. This review presents an overview of nanoscale drug delivery systems and multimodality carriers at the forefront of new PDAC treatments.
Collapse
Affiliation(s)
- Catherine M Sielaff
- Department of Toxicology, School of Pharmacy, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, 12144, USA.
| |
Collapse
|
17
|
Chu M, Wang T, Sun A, Chen Y. Nimesulide inhibits proliferation and induces apoptosis of pancreatic cancer cells by enhancing expression of PTEN. Exp Ther Med 2018; 16:370-376. [PMID: 29896263 DOI: 10.3892/etm.2018.6191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/05/2018] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer-associated cases of mortality worldwide. Prostaglandin-endoperoxide synthase 2 (COX-2) is considered a therapeutic target for prevention of pancreatic cancer. Nimesulide, a selective COX-2 inhibitor, can induce cell apoptosis, resulting in an anti-cancer effect. However, the mechanism underlying this effect remains to be elucidated. The present study aimed to evaluate the effects of nimesulide on proliferation of PANC-1 cells using an MTT assay. Apoptosis was evaluated by DNA laddering and Annexin V-fluorescein isothiocyanate/propidium iodide-stained flow cytometry. Furthermore, western blot analysis was used to elucidate the mechanism underlying nimesulide treatment in PANC-1 cells. It was determined that proliferation of PANC-1 cells was inhibited by nimesulide in a dose-dependent manner. Nimesulide promoted apoptosis of PANC-1 cells. Western blot analysis demonstrated that nimesulide increased expression of cleaved caspase-3 and apoptosis regulator Bax (Bcl-2 associated protein X), and decreased the expression of pro-caspase-3 and apoptosis regulator Bcl-2 (B-cell lymphoma 2). Furthermore, nimesulide enhanced expression of phosphatase and tensin homolog (PTEN), and decreased the expression level of COX-2 and vascular endothelial growth factor. In summary, the results of the present study demonstrated that nimesulide could induce apoptosis and inhibit growth of PANC-1 cells by enhancing the expression of PTEN, which indicates the potential of nimesulide to prevent tumor angiogenesis.
Collapse
Affiliation(s)
- Meifen Chu
- College of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Tongtong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Aihua Sun
- College of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Yu Chen
- College of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
18
|
Pecorelli N, Capretti G, Sandini M, Damascelli A, Cristel G, De Cobelli F, Gianotti L, Zerbi A, Braga M. Impact of Sarcopenic Obesity on Failure to Rescue from Major Complications Following Pancreaticoduodenectomy for Cancer: Results from a Multicenter Study. Ann Surg Oncol 2018; 25:308-317. [PMID: 29116490 DOI: 10.1245/s10434-017-6216-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Failure to rescue (FTR) is a quality-of-care indicator in pancreatic surgery, but may also identify patients who may not tolerate major postoperative complications despite being treated with best available care. Previous studies found that high visceral adipose tissue-to-skeletal muscle ratio is associated with poor outcomes following pancreaticoduodenectomy (PD). The aim of the study is to assess the impact of sarcopenic obesity on occurrence of FTR from major complications in cancer patients undergoing PD. METHODS Prospectively collected data from three high-volume hospitals were reviewed. Total abdominal muscle area (TAMA) and visceral fat area (VFA) were assessed at preoperative staging computed tomography scan. Sarcopenic obesity was defined as high VFA/TAMA ratio. FTR was defined as postoperative mortality following major complication. RESULTS 120 patients with major complications were included. FTR occurred in 23 (19.2%) patients. The "seminal" complications leading to FTR were pancreatic or biliary fistula-related sepsis (n = 14), postoperative pancreatic fistula (POPF)-related hemorrhage (n = 5), and duodenojejunal anastomosis leak-related sepsis (n = 1). On univariate analysis, older age [odds ratio (OR) 3.5, p = 0.034], American Society of Anesthesiologists (ASA) score 3+ (OR 4.2, p = 0.005), cardiovascular disease (OR 3.3, p = 0.013), low serum albumin (OR 2.6, p = 0.042), sarcopenic obesity (OR 4.2, p = 0.009), POPF (OR 3.1, p = 0.027), and cardiorespiratory complications (OR 3.7, p = 0.011) were significantly associated with FTR. On multivariate analysis, sarcopenic obesity [OR 5.7, 95% confidence interval (CI) 1.6-20.7, p = 0.008], ASA score 3+ (OR 4.1, 95% CI 1.2-14.3, p = 0.025), and pancreatic fistula (OR 3.2, 95% CI 1.0-10.2, p = 0.045) were independently associated with FTR. CONCLUSION Sarcopenic obesity, low preoperative physical status, and occurrence of pancreatic fistula are associated with significantly higher risk of FTR from major complications after PD.
Collapse
Affiliation(s)
- Nicolò Pecorelli
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy.
| | - Giovanni Capretti
- Pancreatic Surgery Unit, Humanitas University, Humanitas Research Hospital, Rozzano, Italy
| | - Marta Sandini
- Unit of Hepato-biliary-pancreatic Surgery, School of Medicine and Surgery, Milano-Bicocca University, San Gerardo Hospital, Monza, Italy
| | - Anna Damascelli
- Department of Radiology, Vita-Salute San Raffaele University Hospital, Milan, Italy
| | - Giulia Cristel
- Department of Radiology, Vita-Salute San Raffaele University Hospital, Milan, Italy
| | - Francesco De Cobelli
- Department of Radiology, Vita-Salute San Raffaele University Hospital, Milan, Italy
| | - Luca Gianotti
- Unit of Hepato-biliary-pancreatic Surgery, School of Medicine and Surgery, Milano-Bicocca University, San Gerardo Hospital, Monza, Italy
| | - Alessandro Zerbi
- Pancreatic Surgery Unit, Humanitas University, Humanitas Research Hospital, Rozzano, Italy
| | - Marco Braga
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
19
|
Rahman SH, Urquhart R, Molinari M. Neoadjuvant therapy for resectable pancreatic cancer. World J Gastrointest Oncol 2017; 9:457-465. [PMID: 29290916 PMCID: PMC5740086 DOI: 10.4251/wjgo.v9.i12.457] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 08/24/2017] [Accepted: 09/16/2017] [Indexed: 02/05/2023] Open
Abstract
The use of neoadjuvant therapies has played a major role for borderline resectable and locally advanced pancreatic cancers (PCs). For this group of patients, preoperative chemotherapy or chemoradiation has increased the likelihood of surgery with negative resection margins and overall survival. On the other hand, for patients with resectable PC, the main rationale for neoadjuvant therapy is that the overall survival with current strategies is unsatisfactory. There is a consensus that we need new treatments to improve the overall survival and quality of life of patients with PC. However, without strong scientific evidence supporting the theoretical advantages of neoadjuvant therapies, these potential benefits might turn out not to be worth the risk of tumors progression while waiting for surgery. The focus of this paper is to provide the readers an overview of the most recent evidence on this subject.
Collapse
Affiliation(s)
| | - Robin Urquhart
- Department of Surgery, Dalhousie University, Halifax B3H 2Y9, Nova Scotia, Canada
| | - Michele Molinari
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| |
Collapse
|
20
|
Miyake K, Mori R, Homma Y, Matsuyama R, Okayama A, Murakami T, Hirano H, Endo I. MZB1 in borderline resectable pancreatic cancer resected after neoadjuvant chemoradiotherapy. J Surg Res 2017; 220:391-401. [PMID: 29180208 DOI: 10.1016/j.jss.2017.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/08/2017] [Accepted: 07/03/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND A high accumulation of CD8+ tumor-infiltrating lymphocytes (TILs) induced by neoadjuvant chemoradiotherapy (NACRT) is associated with a favorable prognosis in patients with pancreatic ductal adenocarcinoma (PDAC). However, the correlation between a high accumulation of CD8+ TILs and a favorable prognosis has yet to be fully clarified. The aim of this study was to determine predictive markers of a high accumulation of CD8+ TILs, with a favorable prognosis, using proteomic analysis. MATERIALS AND METHODS We studied 72 resected borderline resectable PDAC patients treated with NACRT between April 2009 and March 2014. Three matched pairs of high CD8+ TIL patients with a favorable prognosis and low CD8+ TIL patients with a poor prognosis were selected. Shotgun proteomics of the stroma and cancerous lesion was performed using formalin-fixed, paraffin-embedded tissue. Validation of the identified proteins was performed using immunohistochemical staining. Relationships between the identified proteins and TILs and clinical outcomes were assessed. RESULTS Marginal zone B- and B1-cell-specific protein (MZB1) was detected in the tumor stroma. MZB1 expression was positively correlated with a high accumulation of CD8+ TILs. High stromal MZB1 expression also correlated with disease-free and overall survival. In a subgroup analysis of CD8+ expression, there was a significant association between stromal MZB1 expression and disease-free and overall survival in the high CD8+ TIL group. CONCLUSIONS MZB1 is a potential marker of a high accumulation of CD8+ TILs in borderline resectable PDACs resected after NACRT. Combination of CD8+ TILs with MZB1 may be a new biomarker of resected cases after NACRT.
Collapse
Affiliation(s)
- Kentaro Miyake
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryutaro Mori
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuki Homma
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akiko Okayama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Takashi Murakami
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
21
|
Impact of Intraoperative Re-resection to Achieve R0 Status on Survival in Patients With Pancreatic Cancer: A Single-center Experience With 483 Patients. Ann Surg 2017; 265:1219-1225. [PMID: 27280512 DOI: 10.1097/sla.0000000000001808] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The aim of this study was to test the hypothesis that intraoperative frozen section (FS) and re-resection results to achieve R0 status are associated with different long-term outcomes in pancreatic cancer patients. BACKGROUND Recent data have challenged the survival benefit of additional resection in patients with pancreatic cancer in case of positive FS to achieve clear pathological section (PS). METHODS Patients who underwent surgery for exocrine pancreatic malignancy with curative intent were identified from a prospective database. Data were stratified by resection margin (group I: FS-R0 → PS-R0; group II: FS-R1 → PS-R0; group III: FS-R1 → PS-R1). Associations with survival were analyzed by univariate and multivariate analyses. RESULTS A total of 483 patients met the inclusion criteria. Of these, 61 patients were excluded due to R2 or Rx status. Three hundred seventeen (75%) patients were allocated to margin group I, 32 (8%) to group II, and 73 (17%) to group III. Median overall survival in group I, II, and III was 29, 36, and 12 months (P < 0.001). There was no significant difference in survival between patients in Group I and II (P = 0.849), whereas patients in group III had significantly poorer outcome than group I (P < 0.001) and II (P = 0.039). The prognostic value of margin group status was confirmed on multivariate analysis (hazard ratio = 1.694, 95% confidence interval 1.175-2.442). CONCLUSIONS FS analysis with intraoperative re-resection should be performed routinely in patients undergoing pancreatic cancer surgery with the aim to achieve a R0 resection.
Collapse
|
22
|
Vendrely V, Peuchant E, Buscail E, Moranvillier I, Rousseau B, Bedel A, Brillac A, de Verneuil H, Moreau-Gaudry F, Dabernat S. Resveratrol and capsaicin used together as food complements reduce tumor growth and rescue full efficiency of low dose gemcitabine in a pancreatic cancer model. Cancer Lett 2017; 390:91-102. [PMID: 28089829 DOI: 10.1016/j.canlet.2017.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/16/2016] [Accepted: 01/08/2017] [Indexed: 12/23/2022]
Abstract
Pancreatic adenocarcinoma, highly resistant to all current anti-cancer treatments, necessitates new approaches promoting cell death. We hypothesized that combined actions of several Bioactive Food Components (BFCs) might provide specific lethal effect towards tumor cells, sparing healthy cells. Human tumor pancreatic cell lines were tested in vitro for sensitivity to resveratrol, capsaicin, piceatannol, and sulforaphane cytotoxic effects. Combination of two or three components showed striking synergetic effect with gemcitabine in vitro. Each BFC used alone did not affect pancreatic tumor growth in a preclinical in vivo model, whereas couples of BFCs had anti-tumor activity. In addition, tumor toxicity was similar using gemcitabine alone or a combination of BFCs and two thirds of gemcitabine dose. Moreover, BFCs enhanced fibrotic response as compared to gemcitabine treatment alone. Reactive oxygen species (ROS) and apoptosis increases were observed, while cell cycle was very mildly affected. This study raises the possibility to use BFCs as beneficial food complements in the therapy of pancreatic adenocarcinoma, especially for patients unable to receive full doses of chemotherapy.
Collapse
Affiliation(s)
| | - Evelyne Peuchant
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - Etienne Buscail
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | | | | | - Aurélie Bedel
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - Aurélia Brillac
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France
| | - Hubert de Verneuil
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - François Moreau-Gaudry
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - Sandrine Dabernat
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France.
| |
Collapse
|
23
|
Birhanu G, Javar HA, Seyedjafari E, Zandi-Karimi A. Nanotechnology for delivery of gemcitabine to treat pancreatic cancer. Biomed Pharmacother 2017; 88:635-643. [PMID: 28142120 DOI: 10.1016/j.biopha.2017.01.071] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/02/2017] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most deadly and quickly fatal human cancers with a 5-year mortality rate close to 100%. Its prognosis is very poor, mainly because of its hostile biological behavior and late onset of symptoms for clinical diagnosis; these bring limitations on therapeutic interventions. Factors contributing for the difficulties in treating PC include: high rate of drug resistance, fast metastasis to different organs, poor prognosis and relapse of the tumor after therapy. After being approved by US FDA 1997, Gemcitabine (Gem) is the first line and the gold standard drug for all stages of advanced PC till now. However, its efficacy is unsatisfactory, mainly due to; its chemical instability and poor cellular uptake, resulting in an extremely short half-life and low bioavailability. To solve this drawbacks and increase the therapeutic outcome important progress has been achieved in the field of nanotechnology and offers a promising and effective alternative. This review mainly focus on the most commonly investigated nanoparticle (NP) delivery systems of Gem for PC treatment and the latest progresses achieved. Novel nanocarriers with better tumor targeting efficiencies and maximum treatment outcome to treat this deadly due are given much attention.
Collapse
Affiliation(s)
- Gebremariam Birhanu
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, International Campus (TUMS-IC), Tehran, Iran; School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Hamid Akbari Javar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | - Ali Zandi-Karimi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
24
|
Cloyd JM, Katz MHG, Prakash L, Varadhachary GR, Wolff RA, Shroff RT, Javle M, Fogelman D, Overman M, Crane CH, Koay EJ, Das P, Krishnan S, Minsky BD, Lee JH, Bhutani MS, Weston B, Ross W, Bhosale P, Tamm EP, Wang H, Maitra A, Kim MP, Aloia TA, Vauthey JN, Fleming JB, Abbruzzese JL, Pisters PWT, Evans DB, Lee JE. Preoperative Therapy and Pancreatoduodenectomy for Pancreatic Ductal Adenocarcinoma: a 25-Year Single-Institution Experience. J Gastrointest Surg 2017; 21:164-174. [PMID: 27778257 DOI: 10.1007/s11605-016-3265-1] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 08/24/2016] [Indexed: 01/31/2023]
Abstract
BACKGROUND The purpose of this study was to evaluate a single-institution experience with delivery of preoperative therapy to patients with pancreatic ductal adenocarcinoma (PDAC) prior to pancreatoduodenectomy (PD). METHODS Consecutive patients (622) with PDAC who underwent PD following chemotherapy and/or chemoradiation between 1990 and 2014 were retrospectively reviewed. Preoperative treatment regimens, clinicopathologic characteristics, operative details, and long-term outcomes in four successive time periods (1990-1999, 2000-2004, 2005-2009, 2010-2014) were evaluated and compared. RESULTS The average number of patients per year who underwent PD following preoperative therapy as well as the proportion of operations performed for borderline resectable and locally advanced (BR/LA) tumors increased over time. The use of induction systemic chemotherapy, as well as postoperative adjuvant chemotherapy, also increased over time. Throughout the study period, the mean EBL decreased while R0 margin rates and vascular resection rates increased overall. Despite the increase in BR/LA resections, locoregional recurrence (LR) rates remained similar over time, and overall survival (OS) improved significantly (median 24.1, 28.1, 37.3, 43.4 months, respectively, p < 0.0001). CONCLUSIONS Despite increases in case complexity, relatively low rates of LR have been maintained while significant improvements in OS have been observed. Further improvements in patient outcomes will likely require disruptive advances in systemic therapy.
Collapse
Affiliation(s)
- Jordan M Cloyd
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA
| | - Laura Prakash
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA
| | - Gauri R Varadhachary
- Department of Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert A Wolff
- Department of Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rachna T Shroff
- Department of Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Milind Javle
- Department of Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Fogelman
- Department of Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Overman
- Department of Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher H Crane
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruce D Minsky
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey H Lee
- Department of Gasteroenterology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manoop S Bhutani
- Department of Gasteroenterology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian Weston
- Department of Gasteroenterology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Ross
- Department of Gasteroenterology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priya Bhosale
- Department of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eric P Tamm
- Department of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huamin Wang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P Kim
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA
| | - Thomas A Aloia
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA
| | - Jean-Nicholas Vauthey
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA
| | - Jason B Fleming
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA
| | - James L Abbruzzese
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA
| | | | - Douglas B Evans
- Division of Surgical Oncology, Department of Surgery, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1484, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Anderson JD, Wan W, Kaplan BJ, Myers J, Fields EC. Changing paradigm in pancreatic cancer: from adjuvant to neoadjuvant chemoradiation. J Gastrointest Oncol 2016; 7:1004-1010. [PMID: 28078125 DOI: 10.21037/jgo.2016.11.09] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Historically, management of pancreatic cancer has been determined based on whether the tumor was amenable to resection and all patients deemed resectable received curative intent surgery followed by adjuvant therapy with chemotherapy (CT) ± RT. However, patients who undergo resection with microscopic (R1) positive margins have inferior rates of survival. The purpose of this study is to identify patients who have undergone pancreatectomy for pancreatic cancer, determine the surgical margins, types of adjuvant therapies given and patterns of failure. Our hypothesis was that in patients who have surgery without pre-operative therapy, there is a high rate of R1 resections and subsequent local recurrence, despite adjuvant therapy. METHODS Seventy-one patients with curative resections for pancreatic cancer between 2003 and 2015 were reviewed. Tumor stage, margin status, distance to closest margin, receipt of adjuvant therapy and length of survival were collected. Patients were divided into two groups based on whether they received adjuvant CT + RT (n=37) or CT alone (n=37). Patients were further divided based on whether resection was R1 (n=29) or R0 (n=42). Wilcoxon survival tests and Cox proportional hazards regression models were performed to determine the effects of CT + RT vs. CT alone, stratified by surgical margin status. RESULTS Of the 29 patients (39%) who had R1, 15 received CT + RT and 14 received only CT. Patients who received CT + RT experienced a significantly longer period of PFS (13 vs. 7.5 mos, P=0.03) than patients who received CT alone. However, there was no significant difference found in time to death post cancer resection between CT + RT vs. CT alone (P=0.73). Of the 42 patients with R0, 21 received CT + RT and 21 received CT. There was a trend towards increase in PFS in patients treated with CT + RT (25 vs. 17 months, P=0.05), but there was no significant increase in time to death compared to patients treated with CT alone (P=0.53. Of the 36 patients with CT + RT, 21 had R0 and 15 had R1. Patients with R0 were more likely to have longer PFS (25 vs. 13 months, P=0.06), but there was no significant difference in time to death compared to patients with CT alone (P=0.68). CONCLUSIONS After curative resection, the addition of RT to CT improves PFS in both R0 and R1 settings. However, patients with R1 have significantly worse PFS and OS compared to patients with R0 and even aggressive adjuvant therapy does not make up for the difference. The paradigm has shifted and now for patients with resectable pancreatic cancers we recommend neoadjuvant CT + RT to improve RT targeting and treatment response assessment and most importantly, improve chances of obtaining R0.
Collapse
Affiliation(s)
- Justin D Anderson
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Wen Wan
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Brian J Kaplan
- Department of Surgical Oncology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jennifer Myers
- Department of Hematology Oncology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Emma C Fields
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
26
|
Virk RK, Gamez R, Mehrotra S, Atieh M, Barkan GA, Wojcik EM, Pambuccian SE. Variation of cytopathologists' use of the indeterminate diagnostic categories "atypical" and "suspicious for malignancy" in the cytologic diagnosis of solid pancreatic lesions on endoscopic ultrasound-guided fine-needle aspirates. Diagn Cytopathol 2016; 45:3-13. [PMID: 27873469 DOI: 10.1002/dc.23565] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/04/2016] [Indexed: 12/17/2022]
Abstract
Indeterminate cytologic diagnoses in endoscopic ultrasound guided fine needle aspiration biopsy (EUS-FNA) of solid pancreatic lesions include the diagnostic categories "atypical" (ATY) and "suspicious for malignancy" (SUSP), which are used at variable rates and are associated with variable underlying risk of malignancy. The aim of this study was to determine individual cytopathologists' rates of indeterminate diagnoses in EUS-FNA of solid pancreatic lesions and their relationship to cytopathologists' experience and volume of pancreatic EUS-FNA examined, as well as the potential impact of departmental consensus review on indeterminate diagnoses. DESIGN The diagnostic rates of ATY and SUSP and their underlying risk of malignancy were calculated for six cytopathologists who diagnosed 1,114 of 1,225 EUS-FNA of solid pancreatic lesions from 1/1/2001 to 9/15/2014, and were then compared for the periods before and after the implementation of departmental consensus review during 2009. RESULTS The six cytopathologists diagnosed 10% of cases as indeterminate; 82 (7.4%) as "atypical" and 29 (2.6%) as "suspicious". The individual cytopathologists' indeterminate diagnosis rates varied twofold (6.67-12.80%) and did not correlate with their experience, total or annual volume of EUS-FNAs. Of the 56/99 (56.57%) cases with follow-up, the underlying rate of malignancy was 47% (35/75; for "atypical" and 87.5% (21/24); for "suspicious"). The underlying rates of malignancy were 33-67% for "atypical" and 80-100% for "suspicious" diagnoses made by individual cytopathologists. The rate of indeterminate diagnoses decreased from 11.55 to 7.88% after the implementation of departmental consensus review. CONCLUSION Individual cytopathologists' rates of indeterminate diagnoses and their significance vary; however, consensus review is helpful in reducing these rates. Diagn. Cytopathol. 2017;45:3-13. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Renu K Virk
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Roberto Gamez
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Swati Mehrotra
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Mohammed Atieh
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Güliz A Barkan
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Eva M Wojcik
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Stefan E Pambuccian
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| |
Collapse
|
27
|
Crocenzi T, Cottam B, Newell P, Wolf RF, Hansen PD, Hammill C, Solhjem MC, To YY, Greathouse A, Tormoen G, Jutric Z, Young K, Bahjat KS, Gough MJ, Crittenden MR. A hypofractionated radiation regimen avoids the lymphopenia associated with neoadjuvant chemoradiation therapy of borderline resectable and locally advanced pancreatic adenocarcinoma. J Immunother Cancer 2016; 4:45. [PMID: 27532020 PMCID: PMC4986363 DOI: 10.1186/s40425-016-0149-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/18/2016] [Indexed: 12/13/2022] Open
Abstract
Background Preclinical studies have shown synergy between radiation therapy and immunotherapy. However, in almost all preclinical models, radiation is delivered in single doses or short courses of high doses (hypofractionated radiation). By contrast in most clinical settings, radiation is delivered as standard small daily fractions of 1.8-2 Gy to achieve total doses of 50–54 Gy (fractionated radiation). We do not yet know the optimal dose and scheduling of radiation for combination with chemotherapy and immunotherapy. Methods To address this, we analyzed the effect of neoadjuvant standard fractionated and hypofractionated chemoradiation on immune cells in patients with locally advanced and borderline resectable pancreatic adenocarcinoma. Results We found that standard fractionated chemoradiation resulted in a significant and extended loss of lymphocytes that was not explained by a lack of homeostatic cytokines or response to cytokines. By contrast, treatment with hypofractionated radiation therapy avoided the loss of lymphocytes associated with conventional fractionation. Conclusion Hypofractionated neoadjuvant chemoradiation is associated with reduced systemic loss of T cells. Trial registration ClinicalTrials.gov NCT01342224, April 21, 2011; NCT01903083, July 2, 2013. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0149-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Todd Crocenzi
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Benjamin Cottam
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Pippa Newell
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA ; The Oregon Clinic, Portland, OR 97213 USA ; Providence Hepatobiliary and Pancreatic Cancer Program, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Ronald F Wolf
- The Oregon Clinic, Portland, OR 97213 USA ; Providence Hepatobiliary and Pancreatic Cancer Program, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Paul D Hansen
- The Oregon Clinic, Portland, OR 97213 USA ; Providence Hepatobiliary and Pancreatic Cancer Program, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Chet Hammill
- The Oregon Clinic, Portland, OR 97213 USA ; Providence Hepatobiliary and Pancreatic Cancer Program, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | | | - Yue-Yun To
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Amy Greathouse
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Garth Tormoen
- Oregon Health and Sciences University, Sam Jackson Parkway, Portland, OR USA
| | - Zeljka Jutric
- Providence Hepatobiliary and Pancreatic Cancer Program, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Kristina Young
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA ; The Oregon Clinic, Portland, OR 97213 USA
| | - Keith S Bahjat
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR 97213 USA ; The Oregon Clinic, Portland, OR 97213 USA
| |
Collapse
|
28
|
Pecorelli N, Nobile S, Partelli S, Cardinali L, Crippa S, Balzano G, Beretta L, Falconi M. Enhanced recovery pathways in pancreatic surgery: State of the art. World J Gastroenterol 2016; 22:6456-6468. [PMID: 27605881 PMCID: PMC4968126 DOI: 10.3748/wjg.v22.i28.6456] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/21/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic surgery is being offered to an increasing number of patients every year. Although postoperative outcomes have significantly improved in the last decades, even in high-volume centers patients still experience significant postoperative morbidity and full recovery after surgery takes longer than we think. In recent years, enhanced recovery pathways incorporating a large number of evidence-based perioperative interventions have proved to be beneficial in terms of improved postoperative outcomes, and accelerated patient recovery in the context of gastrointestinal, genitourinary and orthopedic surgery. The role of these pathways for pancreatic surgery is still unclear as high-quality randomized controlled trials are lacking. To date, non-randomized studies have shown that care pathways for pancreaticoduodenectomy and distal pancreatectomy are safe with no difference in postoperative morbidity, leading to early discharge and no increase in hospital readmissions. Hospital costs are reduced due to better organization of care and resource utilization. However, further research is needed to clarify the effect of enhanced recovery pathways on patient recovery and post-discharge outcomes following pancreatic resection. Future studies should be prospective and follow recent recommendations for the design and reporting of enhanced recovery pathways.
Collapse
|
29
|
Xia BT, Habib DA, Dhar VK, Levinsky NC, Kim Y, Hanseman DJ, Sutton JM, Wilson GC, Smith M, Choe KA, Sussman JJ, Ahmad SA, Abbott DE. Early Recurrence and Omission of Adjuvant Therapy after Pancreaticoduodenectomy Argue against a Surgery-First Approach. Ann Surg Oncol 2016; 23:4156-4164. [PMID: 27459987 DOI: 10.1245/s10434-016-5457-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Sequencing therapy for patients with periampullary malignancy is controversial. Clinical trial data report high rates of adjuvant therapy completion, though contemporary, real-world rates remain incomplete. We sought to identify patients who failed to receive adjuvant therapy and those at risk for early recurrence (ER) who might benefit most from neoadjuvant therapy (NT). METHODS We retrospectively reviewed medical records of 201 patients who underwent pancreaticoduodenectomy for periampullary malignancies between 1999 and 2015; patients receiving NT were excluded. Univariate and multivariate analyses were performed to identify predictors of failure to receive adjuvant therapy and ER (within 6 months) as the primary end points. RESULTS The median age at the time of surgery was 65.5 years (interquartile range 57-74 years). The majority of tumors were pancreatic ductal adenocarcinoma (76.6 %), and 71.6 % of patients received adjuvant therapy after resection. Univariate predictors of failure to undergo adjuvant therapy were advanced age, age-adjusted Charlson comorbidity index, operative transfusion, reoperation, length of stay, and 30- to 90-day readmissions (all p < 0.05). Advanced age, specifically among patients >70 years, persisted as a significant preoperative predictor on multivariate analysis (p < 0.01). Patients who failed to receive adjuvant therapy and/or developed ER had significantly worse overall survival rates compared to all other patients (27.8 vs. 9.7 months; p < 0.01). CONCLUSIONS Approximately one-third of surgery-first patients undergoing pancreaticoduodenectomy at our institution did not receive adjuvant therapy and/or demonstrated ER. This substantial subset of patients may particularly benefit from NT, ensuring completion of multimodal therapy and/or avoiding futile surgical intervention.
Collapse
Affiliation(s)
- Brent T Xia
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - David A Habib
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Vikrom K Dhar
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Nick C Levinsky
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Young Kim
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Dennis J Hanseman
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Jeffrey M Sutton
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Gregory C Wilson
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Milton Smith
- Department of Medicine, Division of Gastroenterology, University of Cincinnati, Cincinnati, OH, USA
| | - Kyuran Ann Choe
- Department of Radiology, University of Cincinnati, Cincinnati, OH, USA
| | - Jeffrey J Sussman
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Syed A Ahmad
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Daniel E Abbott
- Department of Surgery, Division of Surgical Oncology, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
30
|
Bergquist JR, Puig CA, Shubert CR, Groeschl RT, Habermann EB, Kendrick ML, Nagorney DM, Smoot RL, Farnell MB, Truty MJ. Carbohydrate Antigen 19-9 Elevation in Anatomically Resectable, Early Stage Pancreatic Cancer Is Independently Associated with Decreased Overall Survival and an Indication for Neoadjuvant Therapy: A National Cancer Database Study. J Am Coll Surg 2016; 223:52-65. [DOI: 10.1016/j.jamcollsurg.2016.02.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 02/07/2023]
|
31
|
Christians KK, Heimler JW, George B, Ritch PS, Erickson BA, Johnston F, Tolat PP, Foley WD, Evans DB, Tsai S. Survival of patients with resectable pancreatic cancer who received neoadjuvant therapy. Surgery 2016; 159:893-900. [PMID: 26602840 DOI: 10.1016/j.surg.2015.09.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/20/2015] [Accepted: 09/21/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Enthusiasm for neoadjuvant therapy is growing from the emerging consensus that pancreatic cancer is a systemic disease at the time of diagnosis. Those who remain in favor of upfront surgery often cite the lack of reported data to support alternative treatment sequencing. We therefore report the results of all patients treated outside of a clinical trial under the direction of a multidisciplinary pancreatic cancer working group. METHODS We reviewed all patients with resectable pancreatic cancer treated with neoadjuvant therapy (NeoTx) from 2009 to 2013; we excluded those patients treated on prospective clinical trials as they will be the subject of subsequent reports. Data regarding demographics, NeoTx, operative outcomes, pathology, and survival data were abstracted from a prospective database. RESULTS NeoTx was initiated in 69 patients; median age was 65 years (interquartile range [IQR]: 11) and median carbohydrate antigen 19-9 at diagnosis was 96.5 (IQR 210). NeoTx consisted of chemotherapy alone (n = 10, 14%), chemotherapy and radiation (chemoradiation, n = 53, 77%), or both (n = 6, 9%). Median carbohydrate antigen 19-9 after NeoTx was 39 (IQR 104) corresponding to a median decrease of 60%. Operative resection was completed in 60 (87%) of the 69 patients. At restaging after NeoTx, 5 (7%) of 69 patients were not considered candidates for surgery because of the development of metastatic disease (n = 4) or an inadequate performance status (n = 1). At the time of surgery, 4 (6%) of 64 patients had metastatic disease found at laparoscopy. Of the 60 patients who underwent surgical resection, a complete pathologic response was observed in 2 (3%) patients; 20 (33%) had positive lymph nodes, and the median number of positive lymph nodes was 2 (IQR 3). R0 resections were achieved in 58 (97%) of the 60 patients. Additional postoperative adjuvant therapy was administered to 37 (62%) of the 60 patients. Median survival of all 69 patients was 31.5 months; 44.9 months for the 60 patients who completed all NeoTx and resection compared with 8.1 months for the 9 patients who were not resected (log rank P < .001). CONCLUSION NeoTx for resectable pancreatic cancer was associated with a median overall survival of 32 months; something not reported for patients treated with surgery first if based on intent-to-treat analysis. Treatment sequencing may provide an oncologic benefit beyond that of the selection bias afforded surgery after a period of induction therapy.
Collapse
Affiliation(s)
- Kathleen K Christians
- Department of Surgery, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI.
| | - Jonathan W Heimler
- Department of Surgery, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Ben George
- Department of Medical Oncology, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Paul S Ritch
- Department of Medical Oncology, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Beth A Erickson
- Department of Radiation Oncology, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Fabian Johnston
- Department of Surgery, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Parag P Tolat
- Department of Diagnostic Radiology, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - William D Foley
- Department of Diagnostic Radiology, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Douglas B Evans
- Department of Surgery, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Susan Tsai
- Department of Surgery, Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
32
|
Zeiderman MR, Morgan DE, Christein JD, Grizzle WE, McMasters KM, McNally LR. Acidic pH-targeted chitosan capped mesoporous silica coated gold nanorods facilitate detection of pancreatic tumors via multispectral optoacoustic tomography. ACS Biomater Sci Eng 2016. [PMID: 28626793 DOI: 10.1021/acsbiomaterials.6b00111] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We present a cancer nanomedicine based on acidic pH targeted gold nanorods designed for multispectral optoacoustic tomography (MSOT). We have designed gold nanorods coated with mesoporous silica and subsequently capped with chitosan (CMGs). We have conjugated pH-sensitive variant 7 pHLIP peptide to the CMGs (V7-CMG) to provide targeting specificity to the acidic tumor microenvironment. In vitro, treatment of S2VP10 and MiaPaca2 cells with V7-CMG containing gemcitabine resulted in significantly greater cytotoxicity with 97% and 96.5% cell death, respectively than gemcitabine alone 60% and 76% death at pH 6.5 (S2VP10 pH 6.5 p=0.009; MiaPaca2 pH 6.5 p=0.0197). In vivo, the V7-CMGs provided the contrast and targeting specificity necessary for MSOT of retroperitoneal orthotopic pancreatic tumors. In the in vivo S2VP10 model, the V7-CMG particle preferentially accumulated within the tumor at 17.1 MSOT a.u. signal compared with 0.7 MSOT a.u. in untargeted CMG control in tumor (P = 0.0002). Similarly, V7-CMG signal was 9.34 MSOT a.u. in the S2013 model compared with untargeted CMG signal at 0.15 MSOT a.u. (P = 0.0004). The pH-sensitivity of the targeting pHLIP peptide and chitosan coating makes the particles suitable for simultaneous in vivo tumor imaging and drug delivery.
Collapse
Affiliation(s)
| | - Desiree E Morgan
- University of Alabama at Birmingham, School of Medicine, Birmingham, AL, 35294, USA
| | - John D Christein
- University of Alabama at Birmingham, School of Medicine, Birmingham, AL, 35294, USA
| | - William E Grizzle
- University of Alabama at Birmingham, School of Medicine, Birmingham, AL, 35294, USA
| | - Kelly M McMasters
- University of Louisville, School of Medicine, Louisville KY, 40202, USA
| | - Lacey R McNally
- University of Louisville, School of Medicine, Louisville KY, 40202, USA
| |
Collapse
|
33
|
Tai CJ, Huang MT, Wu CH, Wang CK, Tai CJ, Chang CC, Hsieh CI, Chang YJ, Wu CJ, Kuo LJ, Wei PL, Chen RJ, Chiou HY. Combination of Two Targeted Medications (Bevacizumab Plus Cetuximab) Improve the Therapeutic Response of Pancreatic Carcinoma. Medicine (Baltimore) 2016; 95:e3259. [PMID: 27082562 PMCID: PMC4839806 DOI: 10.1097/md.0000000000003259] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The objective of this study is to evaluate the efficacy and safety profiles of the targeted medications, bevacizumab and cetuximab, in combination with cytostatic drugs in patients with locally advanced or metastatic pancreatic cancer. In this retrospective phase 2 study, a total of 59 patients with pancreatic cancer were recruited and received conventional (gemcitabine, cisplatin, and fluorouracil) or targeted regimen (conventional plus bevacizumab and cetuximab for the first cycle) in 2-week intervals for four cycles. The primary end-point for this study was the overall response rate. Secondary end-points were progression-free survival and the safety profiles of the combined therapy. The median time-to-progression and overall survival were 3 and 7 months, respectively, in the conventional treatment group as well as 11 and 13 months, respectively, in the targeted medications treatment group. The most common adverse events in both treatment groups were nausea and vomiting. Moderate (Grade 2) nausea and vomiting were more common in the conventional group than the targeted group but severe (Grade 3) nausea and vomiting were more common in the targeted group. Bevacizumab and cetuximab in combination with gemcitabine, cisplatin, and fluorouracil may help lengthen overall survival up to six months for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Cheng-Jeng Tai
- From the Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital (C-JT, C-KW, C-CC, C-IH); Department of Internal Medicine (C-JT, C-KW, C-IH); Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University (M-TH, L-JK, P-LW, RJC); Division of General Surgery, Department of Surgery, Shuang Ho Hospital (M-TH, C-HW); Center of Excellence for Cancer Research, Taipei Medical University (C-HW); Department of Surgery, Taipei Medical University-Shuang Ho Hospital (C-HW); Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University (C-KW, C-JT); Department of Chinese Medicine (C-KW, C-JT); Traditional Herbal Medicine Research Center (C-KW, C-JT); Division of Gastroenterology, Department of Internal Medicine (C-CC); Cancer Research Center, Taipei Medical University Hospital (Y-JC); Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University (Y-JC, L-JK, P-LW); Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei (Y-JC, L-JK, P-LW, R-JC); Department of Food Science (C-JW); Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung (C-JW); Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University (P-LW); and School of Public Health, College of Public Health and Nutrition, Taipei Medical University, Taipei, Taiwan (H-YC)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lee JC, Ahn S, Paik KH, Kim HW, Kang J, Kim J, Hwang JH. Clinical impact of neoadjuvant treatment in resectable pancreatic cancer: a systematic review and meta-analysis protocol. BMJ Open 2016; 6:e010491. [PMID: 27016245 PMCID: PMC4809107 DOI: 10.1136/bmjopen-2015-010491] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 02/15/2016] [Accepted: 03/01/2016] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Although the only curative strategy for pancreatic cancer is surgical resection, up to 85% of patients relapse after surgery. The efficacy of neoadjuvant treatment in resectable pancreatic cancer (RPC) remains unclear and there is no systematic review focusing fully on this issue. Recently, two prospective trials of neoadjuvant treatment in RPC were terminated early because of slow recruiting and existing randomised controlled trials (RCTs) have too small sample sizes. Therefore, to overcome probable biases, it would be more reasonable to include both RCTs and non-randomised studies (NRSs) with selected criteria. This review aims to investigate the effect of neoadjuvant chemotherapy (CTx) and chemoradiation therapy (CRT) in RPC using RCTs and specific NRSs. METHOD AND ANALYSIS This systematic review will include conventional RCTs as group I, and quasi-randomised controlled trials, non-randomised controlled trials and prospective cohort studies as group II. Two groups will be assessed and analysed separately. Comprehensive literature search will use Medline, Embase, Cochrane library and Scopus databases. Additionally, we will search references from relevant studies and abstracts from major conferences. Two authors will independently identify, screen, include studies, extract data and assess the risk of bias. Discrepancies will be resolved by consensus with another author. An independent methodologist will categorise and assess NRSs to minimise heterogeneity. In each study group, meta-analysis will be conducted using a random-effect model and statistical heterogeneity will be evaluated using I(2)-statistics. Publication bias will be visualised with contour-enhanced funnel plots and analysed with Egger's test. In group I, cumulative meta-analysis will be considered because the CTx regimen and CRT protocol have changed. The quality of evidence will be summarised using the GRADE (Grading of Recommendations Assessment, Development and Evaluation) approach. ETHICS AND DISSEMINATION This review does not use primary data, and formal ethical approval is not required. Findings will be disseminated through peer-reviewed journals and committee conferences. TRIAL REGISTRATION NUMBER CRD42015023820.
Collapse
Affiliation(s)
- Jong-chan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Soyeon Ahn
- Department of Biostatistics, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyu-hyun Paik
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyoung Woo Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jingu Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
35
|
Byun S, Shin SH, Lee E, Lee J, Lee SY, Farrand L, Jung SK, Cho YY, Um SJ, Sin HS, Kwon YJ, Zhang C, Tsang BK, Bode AM, Lee HJ, Lee KW, Dong Z. The retinoic acid derivative, ABPN, inhibits pancreatic cancer through induction of Nrdp1. Carcinogenesis 2015; 36:1580-9. [PMID: 26464195 DOI: 10.1093/carcin/bgv148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 10/07/2015] [Indexed: 12/20/2022] Open
Abstract
Combination chemotherapy for the treatment of pancreatic cancer commonly employs gemcitabine with an EGFR inhibitor such as erlotinib. Here, we show that the retinoic acid derivative, ABPN, exhibits more potent anticancer effects than erlotinib, while exhibiting less toxicity toward noncancerous human control cells. Low micromolar concentrations of ABPN induced apoptosis in BxPC3 and HPAC pancreatic cancer cell lines, concomitant with a reduction in phosphorylated EGFR as well as decreased ErbB3, Met and BRUCE protein levels. The degradation of ErbB3 is a result of proteasomal degradation, possibly due to the ABPN-dependent upregulation of Nrdp1. Administration of ABPN showed significant reductions in tumor size when tested using a mouse xenograft model, with higher potency than erlotinib at the same concentration. Analysis of the tumors demonstrated that ABPN treatment suppressed ErbB3 and Met and induced Nrdp1 in vivo. The data suggest that ABPN may be more suitable in combination chemotherapy with gemcitabine than the more widely used EGFR inhibitor, erlotinib.
Collapse
Affiliation(s)
- Sanguine Byun
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA, Department of Agricultural Biotechnology, Seoul National University, Advanced Institutes of Convergence Technology, Seoul National University, Seoul 151-742, Republic of Korea
| | - Seung Ho Shin
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA, Program in Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN 55454, USA
| | - Eunjung Lee
- Department of Agricultural Biotechnology, Seoul National University, Traditional Alcoholic Beverage Research Team, Korea Food Research Institute, Seongnam, Republic of Korea
| | - Jihoon Lee
- Department of Agricultural Biotechnology, Seoul National University
| | - Sung-Young Lee
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lee Farrand
- Department of Agricultural Biotechnology, Seoul National University, Yuhan Research Institute, Yuhan Corporation, Giheung-gu, Yongin-si 416-1, Republic of Korea
| | - Sung Keun Jung
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Yong-Yeon Cho
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Soo-Jong Um
- Department of Bioscience and Biotechnology/Institute of Bioscience, BK21 Graduate Program, Sejong University, Seoul, Republic of Korea, CheBiGen Inc, BioPark infrastructure BI. Business, 452-79 Jang-Dong, Jeonju, Jeollabukdo, Republic of Korea
| | - Hong-Sig Sin
- CheBiGen Inc, BioPark infrastructure BI. Business, 452-79 Jang-Dong, Jeonju, Jeollabukdo, Republic of Korea
| | - Youn-Ja Kwon
- CheBiGen Inc, BioPark infrastructure BI. Business, 452-79 Jang-Dong, Jeonju, Jeollabukdo, Republic of Korea
| | - Chengjuan Zhang
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA, Department of Molecular Pathology, The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, China and
| | - Benjamin K Tsang
- Department of Agricultural Biotechnology, Seoul National University, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Ann M Bode
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Hyong Joo Lee
- Department of Agricultural Biotechnology, Seoul National University, Advanced Institutes of Convergence Technology, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Advanced Institutes of Convergence Technology, Seoul National University, Seoul 151-742, Republic of Korea
| | - Zigang Dong
- Department of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA,
| |
Collapse
|
36
|
Sho M, Akahori T, Tanaka T, Kinoshita S, Nagai M, Tamamoto T, Ohbayashi C, Hasegawa M, Kichikawa K, Nakajima Y. Importance of resectability status in neoadjuvant treatment for pancreatic cancer. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2015; 22:563-70. [PMID: 25921623 DOI: 10.1002/jhbp.258] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/29/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Much attention has been paid to neoadjuvant treatment (NAT) as a new strategy especially for borderline resectable pancreatic cancer (BRPC). However, the optimal indication of NAT remains undetermined. METHODS We analyzed 248 patients with pancreatic cancer (PC). One hundred resectable tumors were classified as R group. Sixty-nine tumors with venous involvement were classified as BR-P group, while 31 tumors with arterial involvement were classified as BR-A group. Ninety-nine patients received NAT. Furthermore, 48 unresectable locally advanced PC served as controls (LAPC group). Among them, 11 patients received adjuvant surgery afterwards (Ad-surg group). RESULTS The overall median survival time in the R, BR-P and BR-A groups was 45.3, 24.8 and 16.8 months. In the R and BR-P groups, patients treated with NAT had a better prognosis than those without. In contrast, NAT had no impact on prognosis in the BR-A group. Patients treated with NAT in the BR-P, but not BR-A group, had a better prognosis than patients in the LAPC group. Furthermore, patients in the Ad-surg group had a significantly better prognosis than patients in the BR-A group. CONCLUSIONS Borderline resectable pancreatic cancer with venous involvement, but without arterial involvement, may be a good indication for NAT. Our data highlight the importance of preoperative resectability assessment to evaluate the indication and efficacy of NAT.
Collapse
Affiliation(s)
- Masayuki Sho
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Takahiro Akahori
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Toshihiro Tanaka
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | - Shoichi Kinoshita
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Minako Nagai
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Tetsuro Tamamoto
- Department of Radiation Oncology, Nara Medical University, Kashihara, Nara, Japan
| | - Chiho Ohbayashi
- Department of Diagnostic Pathology, Nara Medical University, Kashihara, Nara, Japan
| | - Masatoshi Hasegawa
- Department of Radiation Oncology, Nara Medical University, Kashihara, Nara, Japan
| | - Kimihiko Kichikawa
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | | |
Collapse
|
37
|
He Y, Zheng R, Li D, Zeng H, Zhang S, Chen W. Pancreatic cancer incidence and mortality patterns in China, 2011. Chin J Cancer Res 2015; 27:29-37. [PMID: 25717223 DOI: 10.3978/j.issn.1000-9604.2015.02.05] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 01/15/2015] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The National Central Cancer Registry (NCCR) collected population-based cancer registration data in 2011 from all cancer registries in China. The incidence and mortality rates for pancreatic cancer were compiled and pancreatic cancer incident new cases and deaths were estimated. METHODS A total of 234 cancer registries submitted cancer data to NCCR. Data from 177 cancer registries were qualified and compiled for cancer statistics in 2011. Pancreatic cancer cases were extracted and analyzed from the national database. The pooled data were stratified by area (urban/rural), gender and age group (0, 1-4, 5-9, 10-14…85+). Pancreatic cancer incident cases and deaths were estimated using age-specific rates and national population in 2010. The national census in 2000 and Segi's population were used for age-standardized rates. RESULTS All 177 cancer registries (77 in urban and 100 in rural areas) covered 175,310,169 populations (98,341,507 in urban and 76,968,662 in rural areas). The morphology verified pancreatic cancer cases (MV%) accounting for 40.52% and 4.33% of pancreatic cancer incident cases were identified through death certifications only (DCO%) with mortality to incidence ratio (M/I) of 0.91. The estimated number of newly diagnosed pancreatic cancer cases and deaths were 80,344 and 72,723 in 2011, respectively. The crude incidence rate was 5.96/100,000 (males 6.57/100,000, females 5.32/100,000). The age-standardized incidence rates by Chinese standard population (ASIRC) and by world standard population (ASIRW) were 4.27/100,000 and 4.23/100,000 respectively, ranking 10(th) among all cancers. Pancreatic cancer incidence rate and ASIRC were 7.03/100,000 and 4.94/100,000 in urban areas whereas they were 4.84/100,000 and 3.56/100,000 in rural areas. The incidence rate of pancreatic cancer of 33 cancer registries increased from 3.24/100,000 in 2003 to 3.59/100,000 in 2011 with an annual percentage change (APC) of 1.44. The pancreatic cancer mortality rate was 5.40/100,000 (males 5.88/100,000, females 4.89/100,000), ranking 6(th) among all cancers. The age-standardized mortality rates by Chinese standard population (ASMRC) and by world standard population (ASMRW) were 3.81/100 000 and 3.79/100 000. The pancreatic cancer mortality and ASMRC were 6.47/100,000 and 4.48/100,000 in urban areas, and 4.27/100,000 and 3.08/100,000 in rural areas, respectively. The mortality rates of pancreatic cancer showed an approximately 1.14-fold increase, from 2.85/100,000 in 2003 to 3.26/100,000 in 2011, with an APC of 1.68. CONCLUSIONS The burden of pancreatic cancer is increasing in China. Identification of high-risk population and adequate treatment and prevention are important.
Collapse
Affiliation(s)
- Yutong He
- 1 Cancer Institute, the Fourth Hospital of Hebei Medical University/the Tumor Hospital of Hebei Province, Shijiazhuang 050011, China ; 2 National Office for Cancer Prevention and Control, National Cancer Center, Beijing 100021, China
| | - Rongshou Zheng
- 1 Cancer Institute, the Fourth Hospital of Hebei Medical University/the Tumor Hospital of Hebei Province, Shijiazhuang 050011, China ; 2 National Office for Cancer Prevention and Control, National Cancer Center, Beijing 100021, China
| | - Daojuan Li
- 1 Cancer Institute, the Fourth Hospital of Hebei Medical University/the Tumor Hospital of Hebei Province, Shijiazhuang 050011, China ; 2 National Office for Cancer Prevention and Control, National Cancer Center, Beijing 100021, China
| | - Hongmei Zeng
- 1 Cancer Institute, the Fourth Hospital of Hebei Medical University/the Tumor Hospital of Hebei Province, Shijiazhuang 050011, China ; 2 National Office for Cancer Prevention and Control, National Cancer Center, Beijing 100021, China
| | - Siwei Zhang
- 1 Cancer Institute, the Fourth Hospital of Hebei Medical University/the Tumor Hospital of Hebei Province, Shijiazhuang 050011, China ; 2 National Office for Cancer Prevention and Control, National Cancer Center, Beijing 100021, China
| | - Wanqing Chen
- 1 Cancer Institute, the Fourth Hospital of Hebei Medical University/the Tumor Hospital of Hebei Province, Shijiazhuang 050011, China ; 2 National Office for Cancer Prevention and Control, National Cancer Center, Beijing 100021, China
| |
Collapse
|