1
|
Yuan H, Zhang Y, Liu F, Wu Y, Huang X, Liu X, Jiang L, Xiao B, Zhu Y, Chen Q, Wu P, Jiang K. Exploring the biological mechanism and clinical value of perineural invasion in pancreatic cancer. Cancer Lett 2025; 613:217515. [PMID: 39892698 DOI: 10.1016/j.canlet.2025.217515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Pancreatic cancer (PC) is an extremely aggressive malignancy, with a 5-year survival rate of only 13 %. Perineural invasion (PNI) is a hallmark pathological feature of PC and is observed in almost all cases. Accordingly, PC ranks highly among solid tumors in terms of PNI incidence. The interaction between PC and the nervous system plays a pivotal role in tumor growth and metastasis. In PC, PNI is a key driver of local tumor progression, distant metastasis, and poor prognosis. Clarification of tumor-nerve crosstalk and the underlying molecular mechanisms is needed to facilitate the development of new therapeutic strategies to slow PC progression and alleviate PNI-associated symptoms. In this review, we present a comprehensive overview of the manifestations and characteristics of PNI in PC, summarize the molecular networks that regulate PNI, examine the relationship between PNI and the tumor microenvironment, and discuss the current research challenges and future directions in this critical area.
Collapse
Affiliation(s)
- Hao Yuan
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yufeng Zhang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Fengyuan Liu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yang Wu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xumin Huang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xinjian Liu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Luyang Jiang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Bin Xiao
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yi Zhu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China; Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | - Qun Chen
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| | - Pengfei Wu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| | - Kuirong Jiang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Hua R, Yao HF, Song ZY, Yu F, Che ZY, Gao XF, Huo YM, Liu W, Sun YW, Yang MW, Yang JY, Zhang S, Zhang JF. Evaluation of a new scoring system for assessing nerve invasion in resected pancreatic cancer: A single-center retrospective analysis. Cancer Lett 2024; 603:217213. [PMID: 39244006 DOI: 10.1016/j.canlet.2024.217213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Nerve invasion (NI) is a characteristic feature of pancreatic cancer. Traditional dichotomous statements on the presence of NI are unreasonable because almost all cases exhibit NI when sufficient pathological sections are examined. The critical implications of NI in pancreatic cancer highlight the need for a more effective criterion. This study included 511 patients, who were categorized into a training group and a testing group at a ratio of 7:3. According to the traditional definition, NI was observed in 91.2 % of patients using five pathological slides in our study. The prevalence of NI increased as more pathological slides were used. The criterion of 'two points of intraneural (endoneural) invasion in the case of four pathological slides' has the highest receiver operating characteristic (ROC) score. Based on this new criterion, NI was proved to be an independent prognostic factor for overall survival (OS) and disease-free survival (DFS) and was also correlated with tumor recurrence (P = 0.004). Interestingly, gemcitabine-based chemotherapy regimen is an independent favorable factor for patients with high NI. In the high NI group, patients who received a gemcitabine-based regimen exhibited a better prognosis than those who did not receive the gemcitabine-based regimen for OS (P = 0.000) and DFS (P = 0.001). In conclusion, this study establishes assessment criteria to evaluate the severity of NI in order to predict patient outcomes.
Collapse
Affiliation(s)
- Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Hong-Fei Yao
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Zi-Yu Song
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Feng Yu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Zhao-Yu Che
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiao-Fang Gao
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Wei Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Yong-Wei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Min-Wei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Jun-Feng Zhang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation, Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, PR China.
| |
Collapse
|
3
|
Kim HS, Choi YH, Lee JS, Jo IH, Ko SW, Paik KH, Choi HH, Lee HH, Lim YS, Paik CN, Lee IS, Chang JH. Characteristics of Early Pancreatic Cancer: Comparison between Stage 1A and Stage 1B Pancreatic Cancer in Multicenter Clinical Data Warehouse Study. Cancers (Basel) 2024; 16:944. [PMID: 38473306 DOI: 10.3390/cancers16050944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Little is known about the characteristics of early pancreatic cancer. We aimed to identify the characteristics, clues for early detection, and prognostic factors for early pancreatic cancer by analyzing a large number of patients with stage 1 pancreatic cancer. METHODS A clinical data warehouse that includes databases of all the medical records of eight academic institutions was used to select and analyze patients with pancreatic cancer that had been diagnosed from January 2010 to May 2023. RESULTS In total, 257 stage 1 pancreatic cancer patients were included. There were 134 men (52%), and the average age was 67.2 ± 9.9 years. Compared to patients with stage 1B pancreatic cancer (2-4 cm), patients with stage 1A pancreatic cancer (≤2 cm) had more tumors in the body and tail than in the head (p = 0.028), more new-onset diabetes and less old diabetes (p = 0.010), less jaundice (p = 0.020), more follow-up of IPMN (intraductal papillary mucinous neoplasm, p = 0.029), and more histories of acute pancreatitis (p = 0.013). The pathological findings showed that stage 1A pancreatic cancer involved more IPMNs (p < 0.001) and lower pancreatic intraepithelial neoplasia (p = 0.004). IPMN was present in all 13 pancreatic tumors that were smaller than 1 cm. In multivariate analysis, positive resection margin (odds ratio [OR] 1.536, p = 0.040), venous invasion (OR 1.710, p = 0.010), and perineural invasion (OR 1.968, p = 0.002) were found to be risk factors affecting disease-free survival, while old diabetes (odds ratio [OS] 1.981, p = 0.003) and perineural invasion (OR 2.270, p = 0.003) were found to be risk factors affecting overall survival. CONCLUSIONS IPMN is closely associated with early pancreatic cancer and may provide an opportunity for early detection. The presence of perineural invasion was a crucial prognostic factor for both overall and disease-free survival in patients with stage 1 pancreatic cancer.
Collapse
Affiliation(s)
- Hyo Suk Kim
- Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon 14647, Republic of Korea
| | - Young Hoon Choi
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jae Sin Lee
- Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon 21432, Republic of Korea
| | - Ik Hyun Jo
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon 16247, Republic of Korea
| | - Sung Woo Ko
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
| | - Kyu Hyun Paik
- Department of Internal Medicine, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943, Republic of Korea
| | - Hyun Ho Choi
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu 11765, Republic of Korea
| | - Han Hee Lee
- Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
| | - Yeon Soo Lim
- Department of Radiology, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon 14647, Republic of Korea
| | - Chang Nyol Paik
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon 16247, Republic of Korea
| | - In Seok Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jae Hyuck Chang
- Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon 14647, Republic of Korea
| |
Collapse
|
4
|
Chaudhari V, Ramaswamy A, Srinivas S, Agarwal A, Seshadri RA, Talwar V, Bhargava P, Goel S, Kayal S, Rebala P, Prajapati B, Parikh D, Kothari J, Ch RM, Kadamapuzha JM, Kapoor D, Chaudhary A, Goel V, Singh S, Ghosh J, Lavingia V, Gupta A, Sekar A, Misra S, Vishnoi JR, Soni S, Varshney VK, Bairwa S, Bhandare M, Shrikhande SV, Ostwal V. Practice Patterns and Survival in Patients with Resected Pancreatic Ductal Adenocarcinomas (PDAC) - Results from the Multicentre Indian Pancreatic & Periampullary Adenocarcinoma Project (MIPPAP) Study. J Gastrointest Cancer 2023; 54:1338-1346. [PMID: 37273074 DOI: 10.1007/s12029-023-00936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND There is limited data from India with regard to presentation, practice patterns and survivals in resected pancreatic ductal adenocarcinomas (PDACs). METHODS The Multicentre Indian Pancreatic & Periampullary Adenocarcinoma Project (MIPPAP) included data from 8 major academic institutions across India and presents the outcomes in upfront resected PDACs from January 2015 to June 2019. RESULTS Of 288 patients, R0 resection was achieved in 81% and adjuvant therapy was administered in 75% of patients. With a median follow-up of 42 months (95% CI: 39-45), median DFS for the entire cohort was 39 months (95% CI: 25.4-52.5), and median overall survival (OS) was 45 months (95% CI: 32.3-57.7). A separate analysis was done in which patients were divided into 3 groups: (a) those with stage I and absent PNI (SI&PNI-), (b) those with either stage II/III OR presence of PNI (SII/III/PNI+), and (c) those with stage II/III AND presence of PNI (SII/III&PNI+). The DFS was significantly lesser in patients with SII/III&PNI+ (median 25, 95% CI: 14.1-35.9 months), compared to SII/III/PNI + (median 40, 95% CI: 24-55 months) and SI&PNI- (median, not reached) (p = 0.036)). CONCLUSIONS The MIPPAP study shows that resectable PDACs in India have survivals at par with previously published data. Adjuvant therapy was administered in 75% patients. Adjuvant radiotherapy does not seem to add to survival after R0 resection.
Collapse
Affiliation(s)
- Vikram Chaudhari
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Anant Ramaswamy
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sujay Srinivas
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Ajit Agarwal
- Balco Medical Centre Raipur India, Uparwara, Raipur, India
| | | | - Vineet Talwar
- Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
| | - Prabhat Bhargava
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Shaifali Goel
- Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
| | - Smita Kayal
- Jawaharlal Institute of Post Graduate Medical Education and Research, Pondicherry, India
| | | | | | | | | | - Ramesh M Ch
- Lakeshore Hospital & Research Center, Kochi, Kerala, India
| | | | | | | | - Varun Goel
- Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
| | - Shivendra Singh
- Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
| | | | | | - Amit Gupta
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Anbarasan Sekar
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sanjeev Misra
- All India Institute of Medical Sciences, Jodhpur, India
| | | | - Subhash Soni
- All India Institute of Medical Sciences, Jodhpur, India
| | | | | | - Manish Bhandare
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | | | - Vikas Ostwal
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India.
| |
Collapse
|
5
|
Mangieri CW, Valenzuela CD, Solsky IB, Erali RA, Pardee T, Lima CMSR, Howerton R, Clark CJ, Shen P. Comparison of survival for metastatic pancreatic cancer patients treated with CPI-613 versus resected borderline-resectable pancreatic cancer patients. J Surg Oncol 2023; 128:844-850. [PMID: 37341164 DOI: 10.1002/jso.27365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION Treatment of advanced pancreatic adenocarcinoma remains suboptimal. Therapeutic agents with a novel mechanism of action are desperately needed; one such novel agent is CPI-613 targets. We here analyze the outcomes of 20 metastatic pancreatic cancer patients treated with CPI-613 and FOLFIRINOX in our institution and evaluate their outcomes to borderline-resectable patients treated with curative surgery. METHODS A post hoc analysis was performed of the phase I CPI-613 trial data (NCT03504423) comparing survival outcomes to borderline-resectable cases treated with curative resection at the same institution. Survival was measured by overall survival (OS) for all study cases and disease-free survival (DFS) for resected cases with progression-free survival for CPI-613 cases. RESULTS There were 20 patients in the CPI-613 cohort and 60 patients in the surgical cohort. Median follow-up times were 441 and 517 days for CPI-613 and resected cases, respectively. There was no difference in survival times between CPI-613 and resected cases with a mean OS of 1.8 versus 1.9 year (p = 0.779) and mean PFS/DFS of 1.4 versus 1.7 years (p = 0.512). There was also no difference in 3-year survival rates for OS (hazard ratio [HR] = 1.063, 95% confidence interval [CI] 0.302-3.744, p = 0.925) or DFS/PFS (HR = 1.462, 95% CI 0.285-7.505, p = 0.648). CONCLUSION The first study to evaluate the survival between metastatic patients treated with CPI-613 versus borderline-resectable cases undergoing curative resection. Analysis revealed no significant differences in survival outcomes between the cohorts. Study results are suggestive that there may be potential utility with the addition of CPI-613 to potentially resectable pancreatic adenocarcinoma, although additional research with more comparable study groups are required.
Collapse
Affiliation(s)
- Christopher W Mangieri
- Atrium Wake Forest Baptist Medical Center, Division of Surgical Oncology, Winston-Salem, North Carolina, USA
| | - Cristian D Valenzuela
- Atrium Wake Forest Baptist Medical Center, Division of Surgical Oncology, Winston-Salem, North Carolina, USA
| | - Ian B Solsky
- Atrium Wake Forest Baptist Medical Center, Division of Surgical Oncology, Winston-Salem, North Carolina, USA
| | - Richard A Erali
- Atrium Wake Forest Baptist Medical Center, Division of Surgical Oncology, Winston-Salem, North Carolina, USA
| | - Timothy Pardee
- Atrium Wake Forest Baptist Medical Center, Division of Hematology and Oncology, Winston-Salem, North Carolina, USA
| | - Caio Max S Rocha Lima
- Atrium Wake Forest Baptist Medical Center, Division of Hematology and Oncology, Winston-Salem, North Carolina, USA
| | - Russell Howerton
- Atrium Wake Forest Baptist Medical Center, Division of Surgical Oncology, Winston-Salem, North Carolina, USA
| | - Clancy J Clark
- Atrium Wake Forest Baptist Medical Center, Division of Surgical Oncology, Winston-Salem, North Carolina, USA
| | - Perry Shen
- Atrium Wake Forest Baptist Medical Center, Division of Surgical Oncology, Winston-Salem, North Carolina, USA
| |
Collapse
|
6
|
Bahmad HF, Gogola S, Rejzer M, Stoyanov K, Gomez AS, Valencia AK, Cummings A, Skerry T, Alloush F, Aljamal AA, Deb A, Alghamdi S, Poppiti R. Unraveling the Mysteries of Perineural Invasion in Benign and Malignant Conditions. Curr Oncol 2023; 30:8948-8972. [PMID: 37887547 PMCID: PMC10605475 DOI: 10.3390/curroncol30100647] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023] Open
Abstract
Perineural invasion (PNI) is defined as the dissemination of neoplastic cells within the perineural space. PNI can be a strong indicator of malignancy and is linked to poor prognosis and adverse outcomes in various malignant neoplasms; nevertheless, it can also be seen in benign pathologic conditions. In this review article, we discuss various signaling pathways and neurotrophic factors implicated in the development and progression of PNI. We also describe the methodology, benefits, and limitations of different in vitro, ex vivo, and in vivo models of PNI. The spectrum of presentation for PNI can range from diffuse spread within large nerves ("named" nerves) all the way through localized spread into unnamed microscopic nerves. Therefore, the clinical significance of PNI is related to its extent rather than its mere presence or absence. In this article, we discuss the guidelines for the identification and quantification of PNI in different malignant neoplasms based on the College of American Pathologists (CAP) and World Health Organization (WHO) recommendations. We also describe benign pathologic conditions and neoplasms demonstrating PNI and potential mimics of PNI. Finally, we explore avenues for the future development of targeted therapy options via modulation of signaling pathways involved in PNI.
Collapse
Affiliation(s)
- Hisham F. Bahmad
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
| | - Samantha Gogola
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.G.); (M.R.); (K.S.); (A.S.G.); (A.-K.V.); (A.C.); (T.S.)
| | - Michael Rejzer
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.G.); (M.R.); (K.S.); (A.S.G.); (A.-K.V.); (A.C.); (T.S.)
| | - Kalin Stoyanov
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.G.); (M.R.); (K.S.); (A.S.G.); (A.-K.V.); (A.C.); (T.S.)
| | - Aaron S. Gomez
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.G.); (M.R.); (K.S.); (A.S.G.); (A.-K.V.); (A.C.); (T.S.)
| | - Ann-Katrin Valencia
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.G.); (M.R.); (K.S.); (A.S.G.); (A.-K.V.); (A.C.); (T.S.)
| | - Adonicah Cummings
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.G.); (M.R.); (K.S.); (A.S.G.); (A.-K.V.); (A.C.); (T.S.)
| | - Timothy Skerry
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.G.); (M.R.); (K.S.); (A.S.G.); (A.-K.V.); (A.C.); (T.S.)
| | - Ferial Alloush
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
| | - Abed A. Aljamal
- Department of Medicine, Division of Hematology Oncology, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Arunima Deb
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
| | - Sarah Alghamdi
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
- Department of Pathology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Robert Poppiti
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
- Department of Pathology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
7
|
Li K, Zhao F, Guo Y, Wu Q, Luo S, Zhang J, Li H, Hu S, Wu B, Lin G, Qiu H, Niu B, Sun X, Xu L, Lu J, Du X, Wang Z, Wang X, Kang L, Wang Z, Wang Q, Liu Q, Xiao Y. Interaction analysis of high-risk pathological features on adjuvant chemotherapy survival benefit in stage II colon cancer patients: a multi-center, retrospective study. BMC Cancer 2023; 23:797. [PMID: 37718392 PMCID: PMC10506231 DOI: 10.1186/s12885-023-11196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 07/20/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND We aimed to analyze the benefit of adjuvant chemotherapy in high-risk stage II colon cancer patients and the impact of high-risk factors on the prognostic effect of adjuvant chemotherapy. METHODS This study is a multi-center, retrospective study, A total of 931 patients with stage II colon cancer who underwent curative surgery in 8 tertiary hospitals in China between 2016 and 2017 were enrolled in the study. Cox proportional hazard model was used to assess the risk factors of disease-free survival (DFS) and overall survival (OS) and to test the multiplicative interaction of pathological factors and adjuvant chemotherapy (ACT). The additive interaction was presented using the relative excess risk due to interaction (RERI). The Subpopulation Treatment Effect Pattern Plot (STEPP) was utilized to assess the interaction of continuous variables on the ACT effect. RESULTS A total of 931 stage II colon cancer patients were enrolled in this study, the median age was 63 years old (interquartile range: 54-72 years) and 565 (60.7%) patients were male. Younger patients (median age, 58 years vs 65 years; P < 0.001) and patients with the following high-risk features, such as T4 tumors (30.8% vs 7.8%; P < 0.001), grade 3 lesions (36.0% vs 22.7%; P < 0.001), lymphovascular invasion (22.1% vs 6.8%; P < 0.001) and perineural invasion (19.4% vs 13.6%; P = 0.031) were more likely to receive ACT. Patients with perineural invasion showed a worse OS and marginally worse DFS (hazardous ratio [HR] 2.166, 95% confidence interval [CI] 1.282-3.660, P = 0.004; HR 1.583, 95% CI 0.985-2.545, P = 0.058, respectively). Computing the interaction on a multiplicative and additive scale revealed that there was a significant interaction between PNI and ACT in terms of DFS (HR for multiplicative interaction 0.196, p = 0.038; RERI, -1.996; 95%CI, -3.600 to -0.392) and OS (HR for multiplicative interaction 0.112, p = 0.042; RERI, -2.842; 95%CI, -4.959 to -0.725). CONCLUSIONS Perineural invasion had prognostic value, and it could also influence the effect of ACT after curative surgery. However, other high-risk features showed no implication of efficacy for ACT in our study. TRIAL REGISTRATION This study is registered on ClinicalTrials.gov, NCT03794193 (04/01/2019).
Collapse
Affiliation(s)
- Kexuan Li
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Fuqiang Zhao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yuchen Guo
- Department of Gastrointestinal Surgery, General Surgery Center, The First Hospital of Jilin University, No.1 Xin Min Avenue, Chaoyang District, Changchun, 130021, China
| | - Qingbin Wu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China
| | - Shuangling Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, No.26 Yuan Cun Er Heng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, No.26 Yuan Cun Er Heng Road, Tianhe District, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, No.26 Yuan Cun Er Heng Road, Tianhe District, Guangzhou, 510655, China
| | - Junling Zhang
- Department of Gastrointestinal Surgery, Peking University First Hospital, No.8 Xi Shi Ku Street, Xicheng District, Beijing, 100034, China
| | - Heli Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
| | - Shidong Hu
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, No.28 Fu Xing Road, Haidian District, Beijing, 100853, China
| | - Bin Wu
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Guole Lin
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Huizhong Qiu
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Beizhan Niu
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Xiyu Sun
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Lai Xu
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Junyang Lu
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China
| | - Xiaohui Du
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, No.28 Fu Xing Road, Haidian District, Beijing, 100853, China.
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Xin Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, No.8 Xi Shi Ku Street, Xicheng District, Beijing, 100034, China.
| | - Liang Kang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, No.26 Yuan Cun Er Heng Road, Tianhe District, Guangzhou, 510655, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, No.26 Yuan Cun Er Heng Road, Tianhe District, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, No.26 Yuan Cun Er Heng Road, Tianhe District, Guangzhou, 510655, China.
| | - Ziqiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China.
| | - Quan Wang
- Department of Gastrointestinal Surgery, General Surgery Center, The First Hospital of Jilin University, No.1 Xin Min Avenue, Chaoyang District, Changchun, 130021, China.
| | - Qian Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Yi Xiao
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan Road, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
8
|
Mulder BGS, Koller M, Duiker EW, Sarasqueta AF, Burggraaf J, Meijer VED, Vahrmeijer AL, Hoogwater FJH, Bonsing BA, van Dam GM, Mieog JSD, Pranger BK. Intraoperative Molecular Fluorescence Imaging of Pancreatic Cancer by Targeting Vascular Endothelial Growth Factor: A Multicenter Feasibility Dose-Escalation Study. J Nucl Med 2023; 64:82-89. [PMID: 35680414 PMCID: PMC9841260 DOI: 10.2967/jnumed.121.263773] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 01/28/2023] Open
Abstract
Tumor visualization with near-infrared fluorescence (NIRF) imaging might aid exploration and resection of pancreatic cancer by visualizing the tumor in real time. Conjugation of the near-infrared fluorophore IRDye800CW to the monoclonal antibody bevacizumab enables targeting of vascular endothelial growth factor A. The aim of this study was to determine whether intraoperative tumor-specific imaging of pancreatic cancer with the fluorescent tracer bevacizumab-800CW is feasible and safe. Methods: In this multicenter dose-escalation phase I trial, patients in whom pancreatic ductal adenocarcinoma (PDAC) was suspected were administered bevacizumab-800CW (4.5, 10, or 25 mg) 3 d before surgery. Safety monitoring encompassed allergic or anaphylactic reactions and serious adverse events attributed to bevacizumab-800CW. Intraoperative NIRF imaging was performed immediately after laparotomy, just before and after resection of the specimen. Postoperatively, fluorescence signals on the axial slices and formalin-fixed paraffin-embedded tissue blocks from the resected specimens were correlated with histology. Subsequently, tumor-to-background ratios (TBR) were calculated. Results: Ten patients with clinically suspected PDAC were enrolled in the study. Four of the resected specimens were confirmed PDACs; other malignancies were distal cholangiocarcinoma, ampullary carcinoma, and neuroendocrine tumors. No serious adverse events were related to bevacizumab-800CW. In vivo tumor visualization with NIRF imaging differed per tumor type and was nonconclusive. Ex vivo TBRs were 1.3, 1.5, and 2.5 for the 4.5-, 10-, and 25-mg groups, respectively. Conclusion: NIRF-guided surgery in patients with suspected PDAC using bevacizumab-IRDye800CW is feasible and safe. However, suboptimal TBRs were obtained because no clear distinction between pancreatic cancer from normal or inflamed pancreatic tissue was achieved. Therefore, a more tumor-specific tracer than bevacizumab-IRDye800CW for PDAC is preferred.
Collapse
Affiliation(s)
| | - Marjory Koller
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Evelien W Duiker
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Vincent E de Meijer
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | - Frederik J H Hoogwater
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Gooitzen M van Dam
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- AxelaRx/TRACER Europe BV, Groningen, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Bobby K Pranger
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands;
| |
Collapse
|
9
|
Ma Y, Chen J, Yao X, Li Z, Li W, Wang H, Zhu J. Patterns and prognostic predictive value of perineural invasion in esophageal squamous cell carcinoma. BMC Cancer 2022; 22:1287. [PMID: 36482313 PMCID: PMC9733208 DOI: 10.1186/s12885-022-10386-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The pathological phenotype of perineural invasion (PNI) in squamous cell carcinoma (ESCC) is prevalent but highly heterogeneous. METHODS Postoperative specimens from all patients with ESCC at Shaanxi Provincial People's Hospital were evaluated for PNI using haematoxylin and eosin (H&E) staining and S100 immunohistochemistry (IHC). We determined the correlation between PNI status and clinical outcomes. RESULTS Among 349 ESCC cases, PNI was identified in 127 patients (36.3%), and four subtypes of PNI were identified in our study. Correlation analysis confirmed that PNI was related to tumour invasion depth (pT stage) and lymph node status (pN stage) (P < 0.05). Multivariate analysis showed that PNI (P = 0.001) was an independent factor affecting disease-free survival (DFS) in ESCC, and a similar result was found for overall survival (OS) (P = 0.017). Further analysis revealed that PNI status was a prognostic factor of DFS (P < 0.001) and OS (P = 0.003) exclusively in pN-negative patients. We also found that patients with the PNI-a subtype had better DFS (P = 0.002) and OS (P = 0.002) than patients with the other three subtypes (PNI-b, c, d). CONCLUSION The pathological phenotypes of PNI are diverse, and the identification of PNI subtypes has important clinical guiding value.
Collapse
Affiliation(s)
- Yu Ma
- grid.440288.20000 0004 1758 0451Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, 710068 People’s Republic of China
| | - Jie Chen
- grid.440288.20000 0004 1758 0451Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, 710068 People’s Republic of China
| | - Xi Yao
- grid.440288.20000 0004 1758 0451Department of Anesthesiology, Shaanxi Provincial People’s Hospital, Xi’an, 710068 People’s Republic of China
| | - Zhenzhen Li
- grid.440288.20000 0004 1758 0451Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, 710068 People’s Republic of China
| | - Wensheng Li
- grid.440288.20000 0004 1758 0451Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, 710068 People’s Republic of China
| | - Hongtao Wang
- grid.440288.20000 0004 1758 0451Department of Thoracic Surgery, Shaanxi Provincial People’s Hospital, Xi’an, 710068 People’s Republic of China
| | - Jianfei Zhu
- grid.440288.20000 0004 1758 0451Department of Thoracic Surgery, Shaanxi Provincial People’s Hospital, Xi’an, 710068 People’s Republic of China
| |
Collapse
|
10
|
Kiemen AL, Braxton AM, Grahn MP, Han KS, Babu JM, Reichel R, Jiang AC, Kim B, Hsu J, Amoa F, Reddy S, Hong SM, Cornish TC, Thompson ED, Huang P, Wood LD, Hruban RH, Wirtz D, Wu PH. CODA: quantitative 3D reconstruction of large tissues at cellular resolution. Nat Methods 2022; 19:1490-1499. [PMID: 36280719 PMCID: PMC10500590 DOI: 10.1038/s41592-022-01650-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/14/2022] [Indexed: 12/15/2022]
Abstract
A central challenge in biology is obtaining high-content, high-resolution information while analyzing tissue samples at volumes relevant to disease progression. We address this here with CODA, a method to reconstruct exceptionally large (up to multicentimeter cubed) tissues at subcellular resolution using serially sectioned hematoxylin and eosin-stained tissue sections. Here we demonstrate CODA's ability to reconstruct three-dimensional (3D) distinct microanatomical structures in pancreas, skin, lung and liver tissues. CODA allows creation of readily quantifiable tissue volumes amenable to biological research. As a testbed, we assess the microanatomy of the human pancreas during tumorigenesis within the branching pancreatic ductal system, labeling ten distinct structures to examine heterogeneity and structural transformation during neoplastic progression. We show that pancreatic precancerous lesions develop into distinct 3D morphological phenotypes and that pancreatic cancer tends to spread far from the bulk tumor along collagen fibers that are highly aligned to the 3D curves of ductal, lobular, vascular and neural structures. Thus, CODA establishes a means to transform broadly the structural study of human diseases through exploration of exhaustively labeled 3D microarchitecture.
Collapse
Affiliation(s)
- Ashley L Kiemen
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alicia M Braxton
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mia P Grahn
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Kyu Sang Han
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Jaanvi Mahesh Babu
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rebecca Reichel
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ann C Jiang
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Bridgette Kim
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Jocelyn Hsu
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Falone Amoa
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sashank Reddy
- Department of Plastic and Reconstructive Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Toby C Cornish
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Elizabeth D Thompson
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peng Huang
- Department of Biostatistics, The Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D Wood
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, MD, USA.
- Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD, USA.
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.
- Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
11
|
Taniuchi K, Ueno M, Yokose T, Sakaguchi M, Yoshioka R, Ogasawara M, Kosaki T, Naganuma S, Furihata M. Upregulation of PODXL and ITGB1 in pancreatic cancer tissues preoperatively obtained by EUS-FNAB correlates with unfavorable prognosis of postoperative pancreatic cancer patients. PLoS One 2022; 17:e0265172. [PMID: 35275973 PMCID: PMC8916642 DOI: 10.1371/journal.pone.0265172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 02/24/2022] [Indexed: 11/19/2022] Open
Abstract
The upregulation of PODXL and ITGB1 in surgically resected pancreatic cancer tissues is correlated with an unfavorable postoperative prognosis. The aim of this study was to investigate whether PODXL and ITGB1 are useful preoperative markers for the prognosis of postoperative pancreatic cancer patients in comparison with the TNM staging system. Immunohistochemistry was performed using anti-PODXL and anti-ITGB1 antibodies on 24 pancreatic cancer tissue samples preoperatively obtained by endoscopic ultrasound-guided fine-needle aspiration biopsy. Cox proportional hazards regression analysis was performed to investigate if the UICC TNM stage and upregulation of PODXL and ITGB1 were correlated with postoperative overall survival rates. Univariate analysis revealed that PODXL, TNM stage, lymphatic invasion and the combination of PODXL with ITGB1 are correlated with postoperative survival. Multivariate analysis demonstrated TNM stage and the combination of PODXL with ITGB1 to be correlated with postoperative survival, and the combination of PODXL with ITGB1 most accurately predicted the postoperative outcomes of pancreatic cancer patients before resection. Therefore, upregulation of PODXL and ITGB1 may indicate preoperative neoadjuvant therapy for pancreatic cancer patients by accurately predicting the postoperative prognosis.
Collapse
Affiliation(s)
- Keisuke Taniuchi
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Masahiko Sakaguchi
- Faculty of Information and Communication Engineering, Osaka Electro-Communication University, Osaka, Japan
| | - Reiko Yoshioka
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Mitsunari Ogasawara
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Takuhiro Kosaki
- Department of Endoscopic Diagnostics and Therapeutics, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Seiji Naganuma
- Department of Pathology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Mutsuo Furihata
- Department of Pathology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| |
Collapse
|
12
|
CD74 promotes perineural invasion of cancer cells and mediates neuroplasticity via the AKT/EGR-1/GDNF axis in pancreatic ductal adenocarcinoma. Cancer Lett 2021; 508:47-58. [PMID: 33766751 DOI: 10.1016/j.canlet.2021.03.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 01/06/2023]
Abstract
Perineural invasion (PNI) is a common feature of pancreatic ductal adenocarcinoma (PDAC) and is one of the important causes of local recurrence in resected pancreatic cancer, but the molecular mechanism remains largely unexplored. Here, we used immunohistochemistry staining to determine the expression of CD74. Then the in vivo PNI model, in vitro neuroplasticity assay, cell proliferation assay, wound healing and Transwell-based invasion assay were performed to examine the function of CD74 in pancreatic cancer cell lines. ChIP assay and Luciferase reporter assay were used to illustrate the mechanism underlying CD74 induced GDNF expression. We confirmed that the expression level of CD74 was an independent predictor of PNI and poor prognosis for PDAC. Moreover, we found that upregulation of CD74 on PDAC enhanced its migration and invasive capabilities and potentiated the secretion of neurotrophic factor GDNF to promote the neuroplasticity. Mechanistically, CD74 promoted GDNF production via the AKT/EGR-1/GDNF axis in PDAC. Taken together, our findings suggest a supportive role of CD74 in the PNI of PDAC, and deepen our understanding of how cancer cells promote neuroplasticity in the microenvironment of PDAC.
Collapse
|
13
|
Important CT and histopathological findings for recurrence and overall survival in patients with pancreatic ductal adenocarcinoma who underwent surgery after neoadjuvant FOLFIRINOX. Eur Radiol 2020; 31:3616-3626. [PMID: 33201279 DOI: 10.1007/s00330-020-07489-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/10/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES To investigate important factors for recurrence-free survival (RFS) and overall survival (OS) in patients with pancreatic ductal adenocarcinoma (PDA) who underwent surgery after neoadjuvant FOLFIRINOX using CT and histopathological findings. MATERIALS AND METHODS Sixty-nine patients with PDA who underwent surgery after neoadjuvant FOLFIRINOX were retrospectively included. All patients underwent baseline and first follow-up CT. Two reviewers assessed the CT findings and resectability based on the NCCN guideline. They graded extrapancreatic perineural invasion (EPNI) using a 3-point scale focused on 5 routes. Clinical and histopathological results, such as T- and N-stage, tumor regression grade (TRG) using the College of American Pathology (CAP) grading system, and resection status, were also investigated. Kaplan-Meier methods were used for RFS and OS. The Cox proportional hazard model and logistic regression model were used to identify significant predictive factors. RESULTS There were 57 patients (82.6%) without residual tumors (R0) and 12 patients (17.4%) with residual tumors (R1 or R2). The median RFS was 13 months (range 0~22 months). For RFS, EPNI on baseline CT (hazard ratio (HR) 2.53, 95% confidence interval (CI) 1.116-5.733, p = 0.026) and TRG (HR 1.76, 95% CI 1.000-3.076, p = 0.046) were important predictors of early recurrence. The mean OS was 48 months (range 11~35 months). For OS, TRG (HR 1.05, 95% CI 1.251-6.559, p = 0.013) was a significant factor. However, there were no independent predictors for residual tumors according to the CT findings. CONCLUSION EPNI on baseline CT and TRG were important prognostic factors for tumor recurrence. In addition, TRG was also an important prognostic factor for OS. KEY POINTS • CT and histopathological findings are helpful for predicting early recurrence and poor survival. • EPNI on baseline CT (HR 2.53, p = 0.026) is an important predictor of early recurrence. • The TRG is an important prognostic factor for early recurrence (HR 1.76, p = 0.046) and poor survival (HR 1.05, p = 0.013).
Collapse
|
14
|
Zhang J, Fu X, Liu D, Yang M, Yang J, Huo Y, Liu W, Hua R, Sun Y, Wang J. Molecular markers associated with perineural invasion in pancreatic ductal adenocarcinoma. Oncol Lett 2020; 20:5. [PMID: 32774479 PMCID: PMC7405440 DOI: 10.3892/ol.2020.11866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Perineural invasion (PNI) is a prominent characteristic of pancreatic ductal adenocarcinoma (PDAC). PNI is associated with tumor progression, local recurrence and neuropathic pain; therefore, the identification of biomarkers associated with PNI may be beneficial in assessing the prognosis for patients with PDAC. Using an in vivo model of PNI, five pancreatic cancer cell lines (PANC-1, CFPAC-1, CAPAN-2, SW1990 and ASPC-1) were divided into two groups: High-(comprising PANC-1, CFPAC-1 and CAPAN-2) and low PNI (comprising SW1990 and ASPC-1). Differentially expressed genes (DEGs) between the two groups were identified using the GSE26088 dataset, and were regarded as PNI-associated genes. A total of 445 DEGs associated with PNI (fold change >1.5 or <0.66; P<0.05) were identified, which included 176 up- and 269 downregulated genes. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and function annotation were performed, and the NetworkAnalyst database was used for protein-protein interaction network analysis to identify hub genes. A total of 20 hub genes (gene degree, ≥6) were identified. PNI was associated with the function ‘chemokine signaling pathway’. The DEGs and hub genes were validated using the GSE102238 dataset and clinical tissue microarrays. Fibroblast growth factor 2 (FGF2) and catenin α 2 were demonstrated to be associated with PNI using the GSE102238 dataset. Furthermore, clinical tissue microarray analysis demonstrated that FGF2 was associated with PNI and poor prognosis. The present study provided a potential method for the reliable identification of PNI-associated genes, although further investigation is required to validate these results.
Collapse
Affiliation(s)
- Junfeng Zhang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Xueliang Fu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Dejun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jianyu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yanmiao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Wei Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jian Wang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
15
|
Zhai S, Huo Z, Ying X, Jin J, Wang Y, Lu X, Deng X. A Nomogram for Individual Prediction of Poor Prognosis After Radical Surgery in Patients with Primary Pancreatic Duct Adenocarcinoma. Med Sci Monit 2020; 26:e918882. [PMID: 32088726 PMCID: PMC7051101 DOI: 10.12659/msm.918882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Pancreatic cancer is a highly malignant tumor characterized by poor prognosis. TNM stage cannot always provide accurate prediction of prognosis, which is vital for individualized treatment. Therefore, a novel way to identify patients with poor prognosis after radical surgery is urgently needed. Material/Methods The nomogram was established based on a discovery cohort that included 554 patients with PDAC who had received radical surgery from 2012 to 2016. The clinicopathological data were collected. Poor prognosis was evaluated using 25 features, in which appropriate features for a prediction model were identified. A prediction model incorporating the selected features was established. The discriminative capacity was assessed by C-index, calibration by calibration plot, and clinical usefulness by decision curve. The bootstrapping approach was used to perform internal validation. Results Characteristics included in the nomogram were coronary artery disease and stroke history, elevated CA125, AJCC stage >II, R0 resection, operating time >6 h, poor differentiation, nerve invasion, length of stay >30 days, and postoperative complications. A C-index of 0.713 indicated good discrimination of the prediction model, and the calibration curve showed acceptable calibration. Survival analysis showed that this model had better discriminative capacity than the AJCC staging system and could distinguish relatively good prognosis from poor prognosis in patients at stage II (especially IIa) and IV. Conclusions Our study presents a valid and practical model to predict prognosis of pancreatic cancer patients, which contributes to individualized therapy by assisting surgeons to predict poor prognosis in patients who received radical surgery.
Collapse
Affiliation(s)
- Shuyu Zhai
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Zhen Huo
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Xiayang Ying
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Jiabin Jin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Yue Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Xiongxiong Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Xiaxing Deng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
16
|
Yang J, Jiang Y, He R, Liu W, Yang M, Tao L, Fu X, Shen Y, Li J, Liu D, Huo Y, Zhang J, Hua R, Zhang Z, Sun Y. DKK2 Impairs Tumor Immunity Infiltration and Correlates with Poor Prognosis in Pancreatic Ductal Adenocarcinoma. J Immunol Res 2019; 2019:8656282. [PMID: 31583260 PMCID: PMC6754880 DOI: 10.1155/2019/8656282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/09/2019] [Indexed: 12/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most devastating cancer types despite the improvement of modern medicine. In our present study, we found that dickkopf-related protein 2 (DKK2) shares a higher expression in PDAC compared with adjacent pancreas tissue in tissue microarray. In addition, an elevated expression of DKK2 predicts poorer prognosis of patients and positively correlated with poor tumor differentiation. Multivariate Cox regression analysis was also performed and confirmed that the expression of DKK2 is an independent prognostic factor in PDAC. A high expression of DKK2 correlates with cell migration and epithelial mesenchymal transition based on gene set enrichment analysis (GSEA) while knockdown of DKK2 in PDAC cells resulted in impaired cellular migration. Furthermore, GSEA predicts negative correlation between tumor immunity invasion and DKK2 expression. We then confirmed these results and demonstrated that a higher expression of DKK2 imparts the recruitment of CD8+ T cells. Our work suggested that DKK2 imparts tumor immune evasion and is associated with poor prognosis in pancreatic ductal adenocarcinoma.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Staging
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Prognosis
- Survival Analysis
- Wnt Signaling Pathway
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Jianyu Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongsheng Jiang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruizhe He
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingye Tao
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xueliang Fu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Shen
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Li
- Department of Hepatobiliary Pancreas Surgery, Shanghai East Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Dejun Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanmiao Huo
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junfeng Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Hua
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Long Y, Yao DS, Wei YS, Wei CH, Chen XY. Prognostic significance of perineural invasion in vulvar squamous cell carcinoma. Cancer Manag Res 2019; 11:4461-4469. [PMID: 31191008 PMCID: PMC6525828 DOI: 10.2147/cmar.s198047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 04/10/2019] [Indexed: 12/31/2022] Open
Abstract
Background: Perineural invasion (PNI) is closely associated with poor survival in several types of malignant tumours, but whether this is true in vulvar squamous cell carcinoma (VSCC) is unclear. The aims of this study were to determine the prognostic significance of PNI in patients with VSCC. Patients and methods: We retrospectively analysed clinico-pathological data on 105 patients with VSCC (stages IB-IV) treated surgically at our medical center between 2005 and 2015. Results: PNI was detected in 30 (28.6%) patients, and it was significantly associated with well-known clinical risk factors: large tumour size, depth of invasion, lymphatic vascular space invasion (LVSI), and intra- or extra-nodal spread. Significantly greater proportions of patients with PNI received adjuvant therapy after surgery (P=0.001) or showed local recurrence (P=0.002). Multivariable analysis indicated that risk factors for disease-free survival were tumour size (HR 3.02, 95%CI 1.75-7.75), LVSI (HR 4.82, 95%CI 1.36-17.07), depth of invasion (HR 3.11, 95%CI 1.50-6.44), lymph node metastasis (HR 3.15, 95%CI 1.14-8.96) and positive or close surgical margins (HR 4.86, 95%CI 1.67-14.19). The latter three variables were also risk factors for overall survival. PNI was associated with significantly shorter disease-free survival (DFS) (P=0.020) and overall survival (OS) (P=0.017) based on the log-rank test. Among patients who received adjuvant treatment, Kaplan-Meier curves indicated no significant differences between PNI-positive or -negative subgroups in disease-free survival (P=0.085) or overall survival (P=0.061). Based on multivariable analysis of all patients, PNI was not a significant risk factor for either type of survival . Conclusion: PNI in VSCC is associated with significantly shorter disease-free and overall survival, though it appears to be a weak independent predictor of worse prognosis. Combining PNI with other risk factors may be useful for predicting whether postoperative adjuvant therapy will be needed.
Collapse
Affiliation(s)
- Ying Long
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guang Xi Medical University, Nanning, People's Republic of China
| | - De-Sheng Yao
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guang Xi Medical University, Nanning, People's Republic of China
| | - You-Sheng Wei
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guang Xi Medical University, Nanning, People's Republic of China
| | - Chang-Hong Wei
- Department of Pathology, Affiliated Tumor Hospital of Guang Xi Medical University, Nanning, People's Republic of China
| | - Xiao-Yu Chen
- Department of Pathology, Affiliated Tumor Hospital of Guang Xi Medical University, Nanning, People's Republic of China
| |
Collapse
|
18
|
Alrawashdeh W, Jones R, Dumartin L, Radon TP, Cutillas PR, Feakins RM, Dmitrovic B, Demir IE, Ceyhan GO, Crnogorac‐Jurcevic T. Perineural invasion in pancreatic cancer: proteomic analysis and in vitro modelling. Mol Oncol 2019; 13:1075-1091. [PMID: 30690892 PMCID: PMC6487729 DOI: 10.1002/1878-0261.12463] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/27/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022] Open
Abstract
Perineural invasion (PNI) is a common and characteristic feature of pancreatic ductal adenocarcinoma (PDAC) that is associated with poor prognosis, tumor recurrence, and generation of pain. However, the molecular alterations in cancer cells and nerves within PNI have not previously been comprehensively analyzed. Here, we describe our proteomic analysis of the molecular changes underlying neuro-epithelial interactions in PNI using liquid chromatography-mass spectrometry (LC-MS/MS) in microdissected PNI and non-PNI cancer, as well as in invaded and noninvaded nerves from formalin-fixed, paraffin-embedded PDAC tissues. In addition, an in vitro model of PNI was developed using a co-culture system comprising PDAC cell lines and PC12 cells as the neuronal element. The overall proteomic profiles of PNI and non-PNI cancer appeared largely similar. In contrast, upon invasion by cancer cells, nerves demonstrated widespread plasticity with a pattern consistent with neuronal injury. The up-regulation of SCG2 (secretogranin II) and neurosecretory protein VGF (nonacronymic) in invaded nerves in PDAC tissues was further validated using immunohistochemistry. The tested PDAC cell lines were found to be able to induce neuronal plasticity in PC12 cells in our in vitro established co-culture model. Changes in expression levels of VGF, as well as of two additional proteins previously reported to be overexpressed in PNI, Nestin and Neuromodulin (GAP43), closely recapitulated our proteomic findings in PDAC tissues. Furthermore, induction of VGF, while not necessary for PC12 survival, mediated neurite extension induced by PDAC cell lines. In summary, here we report the proteomic alterations underlying PNI in PDAC and confirm that PDAC cells are able to induce neuronal plasticity. In addition, we describe a novel, simple, and easily adaptable co-culture model for in vitro study of neuro-epithelial interactions.
Collapse
Affiliation(s)
- Wasfi Alrawashdeh
- Centre for Molecular OncologyBarts Cancer InstituteQueen Mary University of LondonUK
| | | | - Laurent Dumartin
- Centre for Molecular OncologyBarts Cancer InstituteQueen Mary University of LondonUK
| | - Tomasz P. Radon
- Centre for Molecular OncologyBarts Cancer InstituteQueen Mary University of LondonUK
| | - Pedro R. Cutillas
- Centre for Haemato‐OncologyBart Cancer InstituteQueen Mary University of LondonUK
| | | | - Branko Dmitrovic
- Department of Pathology and Forensic MedicineFaculty of MedicineUniversity of OsijekCroatia
| | - Ihsan Ekin Demir
- Department of SurgeryKlinikum rechts der Isar Technische UniversitätMunichGermany
| | - Guralp O. Ceyhan
- Department of SurgeryKlinikum rechts der Isar Technische UniversitätMunichGermany
| | | |
Collapse
|
19
|
Huang C, Li Y, Guo Y, Zhang Z, Lian G, Chen Y, Li J, Su Y, Li J, Yang K, Chen S, Su H, Huang K, Zeng L. MMP1/PAR1/SP/NK1R paracrine loop modulates early perineural invasion of pancreatic cancer cells. Theranostics 2018; 8:3074-3086. [PMID: 29896303 PMCID: PMC5996366 DOI: 10.7150/thno.24281] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 03/27/2018] [Indexed: 01/09/2023] Open
Abstract
The molecular mechanism of perineural invasion (PNI) is unclear, and insufficient detection during early-stage PNI in vivo hampers its investigation. We aimed to identify a cytokine paracrine loop between pancreatic ductal adenocarcinoma (PDAC) cells and nerves and established a noninvasive method to monitor PNI in vivo. Methods: A Matrigel/ dorsal root ganglia (DRG) system was used to observe PNI in vitro, and a murine sciatic nerve invasion model was established to examine PNI in vivo. PNI was assessed by MRI with iron oxide nanoparticle labeling. We searched publicly available datasets as well as obtained PDAC tissues from 30 patients to examine MMP1 expression in human tumor and non-tumor tissues. Results: Our results showed that matrix metalloproteinase-1 (MMP1) activated AKT and induced protease-activated receptor-1 (PAR1)-expressing DRG to release substance P (SP), which, in turn, activated neurokinin 1 receptor (NK1R)-expressing PDAC cells and enhanced cellular migration, invasion, and PNI via SP/NK1R/ERK. In animals, hind limb paralysis and a decreased hind paw width were observed approximately 20 days after inoculation of cancer cells in the perineurium. MMP1 silencing with shRNA or treatment with either a PAR1 or an NK1R antagonist inhibited PNI. MRI detected PNI as early as 10 days after implantation of PDAC cells. PNI also induced PDAC liver metastasis. Bioinformatic analyses and pathological studies on patient tissues corroborated the clinical relevance of these findings. Conclusion: In this study, we provided evidence that the MMP1/PAR1/SP/NK1R paracrine loop contributes to PNI during the early stage of primary tumor formation. Furthermore, we established a sensitive and non-invasive method to detect nerve invasion using iron oxide nanoparticles and MRI.
Collapse
|
20
|
Tummers WS, Miller SE, Teraphongphom NT, Gomez A, Steinberg I, Huland DM, Hong S, Kothapalli SR, Hasan A, Ertsey R, Bonsing BA, Vahrmeijer AL, Swijnenburg RJ, Longacre TA, Fisher GA, Gambhir SS, Poultsides GA, Rosenthal EL. Intraoperative Pancreatic Cancer Detection using Tumor-Specific Multimodality Molecular Imaging. Ann Surg Oncol 2018; 25:1880-1888. [PMID: 29667116 DOI: 10.1245/s10434-018-6453-2] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Operative management of pancreatic ductal adenocarcinoma (PDAC) is complicated by several key decisions during the procedure. Identification of metastatic disease at the outset and, when none is found, complete (R0) resection of primary tumor are key to optimizing clinical outcomes. The use of tumor-targeted molecular imaging, based on photoacoustic and fluorescence optical imaging, can provide crucial information to the surgeon. The first-in-human use of multimodality molecular imaging for intraoperative detection of pancreatic cancer is reported using cetuximab-IRDye800, a near-infrared fluorescent agent that binds to epidermal growth factor receptor. METHODS A dose-escalation study was performed to assess safety and feasibility of targeting and identifying PDAC in a tumor-specific manner using cetuximab-IRDye800 in patients undergoing surgical resection for pancreatic cancer. Patients received a loading dose of 100 mg of unlabeled cetuximab before infusion of cetuximab-IRDye800 (50 mg or 100 mg). Multi-instrument fluorescence imaging was performed throughout the surgery in addition to fluorescence and photoacoustic imaging ex vivo. RESULTS Seven patients with resectable pancreatic masses suspected to be PDAC were enrolled in this study. Fluorescence imaging successfully identified tumor with a significantly higher mean fluorescence intensity in the tumor (0.09 ± 0.06) versus surrounding normal pancreatic tissue (0.02 ± 0.01), and pancreatitis (0.04 ± 0.01; p < 0.001), with a sensitivity of 96.1% and specificity of 67.0%. The mean photoacoustic signal in the tumor site was 3.7-fold higher than surrounding tissue. CONCLUSIONS The safety and feasibilty of intraoperative, tumor-specific detection of PDAC using cetuximab-IRDye800 with multimodal molecular imaging of the primary tumor and metastases was demonstrated.
Collapse
Affiliation(s)
- Willemieke S Tummers
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA.,Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Sarah E Miller
- Department of Otolaryngology, Stanford University, Stanford, CA, USA
| | | | - Adam Gomez
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Idan Steinberg
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA
| | - David M Huland
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA
| | - Steve Hong
- Department of Otolaryngology, Stanford University, Stanford, CA, USA
| | - Sri-Rajasekhar Kothapalli
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA
| | - Alifia Hasan
- Department of Otolaryngology, Stanford University, Stanford, CA, USA
| | - Robert Ertsey
- Department of Otolaryngology, Stanford University, Stanford, CA, USA
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Teri A Longacre
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - George A Fisher
- Department of Medical Oncology, Stanford University, Stanford, CA, USA
| | - Sanjiv S Gambhir
- Departments of Radiology, Bioengineering, and Materials Science & Engineering, Molecular Imaging Program at Stanford; Canary Center at Stanford for Early Cancer Detection, Stanford University, Stanford, CA, USA
| | | | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University, Stanford, CA, USA. .,Stanford Cancer Center, Stanford University, Stanford, CA, USA.
| |
Collapse
|
21
|
Zhang M, Liang XH, Tang YL. [Advances in the molecular mechanisms of perineural invasion in salivary adenoid cystic carcinoma]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2018; 36:204-211. [PMID: 29779285 PMCID: PMC7030335 DOI: 10.7518/hxkq.2018.02.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 12/02/2017] [Indexed: 02/05/2023]
Abstract
Salivary adenoid cystic carcinoma is a very common malignant neoplasm in the salivary glands. One of the most important features of salivary cystic carcinoma is perineural invasion, which renders its complete removal is very difficult through clinical surgery. Consequently, tumor recurrence occurs easily after clinical surgery. Therefore, the molecular mechanisms of perineural invasion in salivary adenoid cystic carcinoma are necessary to explore. These mechanisms are reviewed in this paper.
Collapse
Affiliation(s)
- Mei Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
22
|
Tummers WS, Kimura RH, Abou-Elkacem L, Beinat C, Vahrmeijer AL, Swijnenburg RJ, Willmann JK, Gambhir SS. Development and Preclinical Validation of a Cysteine Knottin Peptide Targeting Integrin αvβ6 for Near-infrared Fluorescent-guided Surgery in Pancreatic Cancer. Clin Cancer Res 2018; 24:1667-1676. [DOI: 10.1158/1078-0432.ccr-17-2491] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/20/2017] [Accepted: 12/28/2017] [Indexed: 11/16/2022]
|
23
|
Duconseil P, Périnel J, Autret A, Adham M, Sauvanet A, Chiche L, Mabrut JY, Tuech JJ, Mariette C, Régenet N, Fabre JM, Bachellier P, Delpéro JR, Paye F, Turrini O. Resectable invasive IPMN versus sporadic pancreatic adenocarcinoma of the head of the pancreas: Should these two different diseases receive the same treatment? A matched comparison study of the French Surgical Association (AFC). Eur J Surg Oncol 2017; 43:1704-1710. [PMID: 28687431 DOI: 10.1016/j.ejso.2017.06.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/25/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To compare survival and impact of adjuvant chemotherapy in patients who underwent pancreaticoduodenectomy (PD) for invasive intraductal papillary mucinous neoplasm (IIPMN) and sporadic pancreatic ductal adenocarcinoma (PDAC). METHODS From 2005 to 2012, 240 patients underwent pancreatectomy for IIPMN and 1327 for PDAC. Exclusion criteria included neoadjuvant treatment, pancreatic resection other than PD, vascular resection, carcinoma in situ, or <11 examined lymph nodes. Thus, 82 IIPMN and 506 PDAC were eligible for the present study. Finally, The IIPMN group was matched 1:2 to compose the PDAC group according to TNM disease stage, perineural invasion, lymph node ratio, and margin status. RESULTS There was no difference in patient's characteristics, intraoperative parameters, postoperative outcomes, and histologic parameters. Overall survival and disease-free survival times were comparable between the 2 groups. In each group, overall survival time was significantly poorer in patients who did not achieve adjuvant chemotherapy (p = 0.03 for the IIPMN group; p = 0.03 for the PDAC group). In lymph-node negative patients of the IIPMN group, adjuvant chemotherapy did not have any significant impact on overall survival time (OR = 0.57; 95% CI [0.24-1.33]). Considering the whole population (i.e. patients with IIPMN and PDAC; n = 246), patients who did not achieve adjuvant chemotherapy had poorer survival (p < 0.01). CONCLUSIONS The courses of IIPMN and PDAC were similar after an optimized stage-to-stage comparison. Adjuvant chemotherapy was efficient in both groups. However, in lymph node negative patients, adjuvant chemotherapy seemed not to have a significant impact.
Collapse
Affiliation(s)
- P Duconseil
- Department of Surgery, Hôpital Nord, Marseille, France.
| | - J Périnel
- Department of Surgery, Hôpital Edouard-Herriot, HCL, UCBL1, Lyon, France
| | - A Autret
- Department of Biostatistics, Institut Paoli-Calmettes, Marseille, France
| | - M Adham
- Department of Surgery, Hôpital Edouard-Herriot, HCL, UCBL1, Lyon, France
| | - A Sauvanet
- Department of Surgery, Hôpital Beaujon, Paris, France
| | - L Chiche
- Department of Surgery, Maison du Haut-Lévêque, Bordeaux, France
| | - J-Y Mabrut
- Department of Surgery, Hôpital de la Croix Rousse, Lyon, France
| | - J-J Tuech
- Department of Surgery, Hôpital Charles Nicolle, Rouen, France
| | - C Mariette
- Department of Surgery, Hôpital Claude-Huriez, Lille, France
| | - N Régenet
- Department of Surgery, Hôpital Hôtel Dieu, Nantes, France
| | - J-M Fabre
- Department of Surgery, Hôpital Saint Eloi, Montpellier, France
| | - P Bachellier
- Department of Surgery, Hôpital de Hautepierre, Strasbourg, France
| | - J-R Delpéro
- Department of Surgery, Institut Paoli-Calmettes, Marseille, France
| | - F Paye
- Department of Surgery, Hôpital Saint Antoine, Paris, France
| | - O Turrini
- Department of Surgery, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
24
|
Tummers WS, Farina-Sarasqueta A, Boonstra MC, Prevoo HA, Sier CF, Mieog JS, Morreau J, van Eijck CH, Kuppen PJ, van de Velde CJ, Bonsing BA, Vahrmeijer AL, Swijnenburg RJ. Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:56816-56828. [PMID: 28915633 PMCID: PMC5593604 DOI: 10.18632/oncotarget.18232] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/01/2017] [Indexed: 12/21/2022] Open
Abstract
Discrimination of pancreatic ductal adenocarcinoma (PDAC) from chronic pancreatitis (CP) or peritumoral inflammation is challenging, both at preoperative imaging and during surgery, but it is crucial for proper therapy selection. Tumor-specific molecular imaging aims to enhance this discrimination and to help select and stratify patients for resection. We evaluated various biomarkers for the specific identification of PDAC and associated lymph node metastases. Using immunohistochemistry (IHC), expression levels and patterns were investigated of integrin αvβ6, carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), Cathepsin E (Cath E), epidermal growth factor receptor (EGFR), hepatocyte growth factor receptor (c-MET), thymocyte differentiation antigen 1 (Thy1), and urokinase-type plasminogen activator receptor (uPAR). In a first cohort, multiple types of pancreatic tissue were evaluated (n=62); normal pancreatic tissue (n=8), CP (n=7), PDAC (n=9), tumor associated lymph nodes (n=32), and PDAC after neoadjuvant radiochemotherapy (n=6). In a second cohort, tissues were investigated (n=55) with IHC and immunofluorescence (IF) for concordance of biomarker expression in all tissue types, obtained from an individual patient. Integrin αvβ6 and CEACAM5 showed significantly higher expression levels in PDAC versus normal pancreatic tissue (P=0.001 and P<0.001, respectively) and CP (P=0.003 and P<0.001, respectively). Avβ6 and CEACAM5 expression identified tumor-positive lymph nodes correctly in 84% and 68%, respectively, and in 100% of tumor-negative nodes for both biomarkers. In conclusion, αvβ6 and CEACAM5 are excellent biomarkers to differentiate PDAC from surrounding tissue and to identify lymph node metastases. Individually or combined, these biomarkers are promising targets for tumor-specific molecular imaging of PDAC.
Collapse
Affiliation(s)
- Willemieke S Tummers
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Martin C Boonstra
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Hendrica A Prevoo
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis F Sier
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan S Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Casper H van Eijck
- Department of Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter J Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
25
|
Laeseke PF, Chen R, Jeffrey RB, Brentnall TA, Willmann JK. Combining in Vitro Diagnostics with in Vivo Imaging for Earlier Detection of Pancreatic Ductal Adenocarcinoma: Challenges and Solutions. Radiology 2016; 277:644-61. [PMID: 26599925 DOI: 10.1148/radiol.2015141020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth-leading cause of cancer-related death in the United States and is associated with a dismal prognosis, particularly when diagnosed at an advanced stage. Overall survival is significantly improved if PDAC is detected at an early stage prior to the onset of symptoms. At present, there is no suitable screening strategy for the general population. Available diagnostic serum markers are not sensitive or specific enough, and clinically available imaging modalities are inadequate for visualizing early-stage lesions. In this article, the role of currently available blood biomarkers and imaging tests for the early detection of PDAC will be reviewed. Also, the emerging biomarkers and molecularly targeted imaging agents being developed to improve the specificity of current imaging modalities for PDAC will be discussed. A strategy incorporating blood biomarkers and molecularly targeted imaging agents could lead to improved screening and earlier detection of PDAC in the future. (©) RSNA, 2015.
Collapse
Affiliation(s)
- Paul F Laeseke
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - Ru Chen
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - R Brooke Jeffrey
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - Teresa A Brentnall
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - Jürgen K Willmann
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| |
Collapse
|
26
|
Prognostic Value of Perineural Invasion in Esophageal and Esophagogastric Junction Carcinoma: A Meta-Analysis. DISEASE MARKERS 2016; 2016:7340180. [PMID: 27051075 PMCID: PMC4802032 DOI: 10.1155/2016/7340180] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/16/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Here we aimed to clarify the prognostic significance of perineural invasion (PNI) in esophageal and esophagogastric junction (EGJ) carcinoma. METHODS A comprehensive literature search for relevant reports published up to July 2015 was performed using Pubmed and Embase databases. The pooled HR and 95% CI for overall survival (OS) and disease-free survival (DFS) were used to assess the prognostic value. The association of PNI with pathological characteristics was evaluated by OR and 95% CI. RESULTS A total of 13 cohorts were retrieved, covering 2770 patients treated by surgery. The cumulative analysis revealed a statistical correlation between PNI and poor OS (HR = 1.76, 95% CI: 1.54-2.20, and P < 0.00001), as well as poor DFS (HR = 1.96, 95% CI: 1.42-2.71, and P < 0.001). Moreover, analysis of 1475 patients showed improved PNI in T3 + T4 (OR = 0.39, 95% CI: 0.21-0.70, and P = 0.002), N+ (OR = 0.52, 95% CI: 0.40-0.69, and P < 0.00001), and G3 + G4 (OR = 0.66, 95% CI: 0.48-0.90, and P = 0.008) patients compared with T1 + T2, N-, and G1 + G2 ones, respectively. No significant heterogeneity was found between the studies. CONCLUSIONS PNI is an adverse prognostic biomarker in esophageal and EGJ carcinoma. Moreover, PNI implies advanced T, N stage and poor cell differentiation.
Collapse
|
27
|
Eskander MF, Bliss LA, Tseng JF. Pancreatic adenocarcinoma. Curr Probl Surg 2016; 53:107-54. [DOI: 10.1067/j.cpsurg.2016.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022]
|
28
|
Abstract
Pancreatic nerves undergo prominent alterations during the evolution and progression of human chronic pancreatitis and pancreatic cancer. Intrapancreatic nerves increase in size (neural hypertrophy) and number (increased neural density). The proportion of autonomic and sensory fibres (neural remodelling) is switched, and are infiltrated by perineural inflammatory cells (pancreatic neuritis) or invaded by pancreatic cancer cells (neural invasion). These neuropathic alterations also correlate with neuropathic pain. Instead of being mere histopathological manifestations of disease progression, pancreatic neural plasticity synergizes with the enhanced excitability of sensory neurons, with Schwann cell recruitment toward cancer and with central nervous system alterations. These alterations maintain a bidirectional interaction between nerves and non-neural pancreatic cells, as demonstrated by tissue and neural damage inducing neuropathic pain, and activated neurons releasing mediators that modulate inflammation and cancer growth. Owing to the prognostic effects of pain and neural invasion in pancreatic cancer, dissecting the mechanism of pancreatic neuroplasticity holds major translational relevance. However, current in vivo models of pancreatic cancer and chronic pancreatitis contain many discrepancies from human disease that overshadow their translational value. The present Review discusses novel possibilities for mechanistically uncovering the role of the nervous system in pancreatic disease progression.
Collapse
Affiliation(s)
- Ihsan Ekin Demir
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675 Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675 Munich, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675 Munich, Germany
| |
Collapse
|
29
|
Afsar CU, Gunaldi M, Kum P, Sahin B, Erkisi M, Kara IO, Paydas S, Duman BB, Ercolak V, Karaca F, Uyeturk U, Guner SI. Pancreatic carcinoma, thrombosis and mean platelet volume: single center experience from the southeast region of Turkey. Asian Pac J Cancer Prev 2015; 15:9143-6. [PMID: 25422192 DOI: 10.7314/apjcp.2014.15.21.9143] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The aim of this study was to investigate the general characteristics of patients with deep vein thrombosis (DVT) and pancreatic cancer as well as evaluate the relationship between mean platelet volume (MPV), DVT and survival. MATERIALS AND METHODS Seventy-seven patients with pancreatic cancer, who were admitted to Cukurova University Medical Faculty, Department of Medical Oncology, were enrolled in the study RESULTS The mean age was 59±20. Forty-nine (63.6%) were men and 28 women (36.4%) . Sixty-eight (88.3%) patients had adenocarcinoma and 9 (11.7%) had a malignant epithelial tumor. Thirty-six (46.7%) had liver metastasis at diagnosis. Twenty-six (33.8%) patients were alive, 20 (26%) were dead and in 31 (40.2%) the status was unknown. Only 14 (18.1%) patients had DVT. In 42 (54.5%) patients MPV values were normal, in 28 (36.4%) patients they were above normal, and in 7 (9.1%) patients they were below normal. There was no statistically significant difference between gender, tumour localization, chemotherapy and survival rates (p:0.56, p:0.11, p:0.21). There was no significant difference between DVT, gender, localisation, histological subtype, the presence of metastasis, stage and if the patient had been treated with chemotherapy (p:0.5, p:0.6, p:0.2, p:0.32, p:0.1, p:0.84). There was also no significant difference between MPV and DVT (p:0.57) but there was a significant difference between liver metastasis and DVT (p:0.02). Age, stage, the presence of metastasis and DVT were prognostic in pancreatic cancer patients. CONCLUSIONS Cases of pancreatic cancer with liver metastasis should be studied more carefully as thrombosis is more common in these patients.
Collapse
Affiliation(s)
- Cigdem Usul Afsar
- Department of Medical Oncology, Bahcelievler Medical Park Hospital, Istanbul, Turkey E-mail :
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yang Y, Huang X, Sun J, Gao P, Song Y, Chen X, Zhao J, Wang Z. Prognostic value of perineural invasion in colorectal cancer: a meta-analysis. J Gastrointest Surg 2015; 19:1113-22. [PMID: 25663635 DOI: 10.1007/s11605-015-2761-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 01/21/2015] [Indexed: 01/31/2023]
Abstract
BACKGROUND The prognostic value of perineural invasion (PNI) in colorectal cancer (CRC) does not reach a consensus. METHODS A comprehensive literature search for relevant reports published up to October 2014 was performed using the electronic databases: PubMed, Embase, and Web of Science. The pooled hazard ratio (HR) with 95 % confidence intervals (CI) was used to estimate the prognostic effects. RESULT Thirty-eight studies comprising 12,661 CRC patients were analyzed. Our results showed that PNI is significantly associated with poor prognosis in OS (overall survival) (HR = 2.07, 95 % CI = 1.87-2.29, P < 0.01) and DFS (disease-free survival) (HR = 2.23, 95 % CI = 1.79-2.78, P < 0.01). There was no significant prognostic difference in DFS between stage II CRC patients with PNI(+) and stage III patients (HR = 1.67, 95 % CI = 0.53-5.25, P = 0.38). Further subgroup analysis revealed that the significance of the association between PNI and worse prognosis in CRC patients is not affected by many factors, including geographic setting, PNI positive rate, treatment, TNM stage, tumor site, and quality of the study. CONCLUSIONS The meta-analysis indicates that PNI is a poor prognostic factor in CRC patients. The postoperative survival of stage II CRC patients with PNI(+) is probably more similar to that of stage III patients. Currently available adjuvant therapy should be considered in stage II CRC patients with PNI(+).
Collapse
Affiliation(s)
- Yuchong Yang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Bliss LA, Witkowski ER, Yang CJ, Tseng JF. Outcomes in operative management of pancreatic cancer. J Surg Oncol 2014; 110:592-8. [PMID: 25111970 DOI: 10.1002/jso.23744] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/18/2014] [Indexed: 12/18/2022]
Abstract
Pancreatic cancer is a highly lethal malignancy that often presents at an advanced stage. Surgical resection can prolong survival and offers the only potential for cure. However, pancreatectomy is associated with significant morbidity and mortality. This article reviews perioperative outcomes, post-resection long-term survival, and innovations in the surgical management of pancreatic cancer.
Collapse
Affiliation(s)
- Lindsay A Bliss
- Department of Surgery and Surgical Outcomes Analysis & Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | | | | |
Collapse
|
32
|
Zhou L, Li J, Zhao YP, Cui QC, Zhou WX, Guo JC, You L, Wu WM, Zhang TP. The prognostic value of Cyclin B1 in pancreatic cancer. Med Oncol 2014; 31:107. [DOI: 10.1007/s12032-014-0107-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/25/2014] [Indexed: 02/08/2023]
|
33
|
Jiang YM, Li G, Sun BC, Zhao XL, Zhou ZK. Study on the Relationship Between CXCR4 Expression and Perineural Invasion in Pancreatic Cancer. Asian Pac J Cancer Prev 2014; 15:4893-6. [DOI: 10.7314/apjcp.2014.15.12.4893] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
34
|
Zhang S, Wen DQ, Kong YL, Li YL, Zhang HY. Effects of secondary left-sided portal hypertension on the radical operation rate and prognosis in patients with pancreatic cancer. Asian Pac J Cancer Prev 2014; 15:2239-44. [PMID: 24716963 DOI: 10.7314/apjcp.2014.15.5.2239] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To investigate the effects of secondary left-sided portal hypertension (LSPH) on the radical operation rate of patients with pancreatic cancer and systemically evaluate the prognosis of patients with LSPH secondary to pancreatic cancer after radical surgery. MATERIALS AND METHODS The data of patients with pancreatic cancer who underwent laparotomy over a 15-year period in Department of Hepatobiliary Surgery of Chinese PLA Air Force General Hospital from Jan. 1, 1997, to Jun. 30, 2012 was retrospectively reviewed. RESULTS A total of 362 patients with pancreatic cancer after laparotomy were selected, including 73 with LSPH and 289 without LSPH. Thirty-five patients with LSPH (47.9%) and 147 without non-LSPH (50.9%) respectively underwent radical operations. No significant difference was found between these two groups regarding the total resection rate and stratified radical resection rate according to different pathological types and cancer locations. The mean and median survival time of patients after radical operation in LSPH group were 13.9 ± 1.3 months and 14.8 months, respectively, while those in non-LSPH group were 22.6 ± 1.4 months and 18.4 months, respectively(P<0.05). CONCLUSIONS Radical operations for pancreatic cancer and secondary LSPH are safe and effective. Because high-grade malignancy and poor prognosis are closely associated, the decision for radical surgery should be made more meticulously for the patients with pancreatic cancer.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Obstetrics and Gynecology, Chinese PLA Air Force General Hospital, Beijing, China E-mail : ,
| | | | | | | | | |
Collapse
|