Skip to main content
Top
Published in: Annals of Surgical Oncology 3/2024

05-12-2023 | Cholangiocarcinoma | Translational Research

Impact of Neutrophil Extracellular Traps Identified by Citrullinated Histone H3 Immunohistochemistry for Postoperative Prognosis in Patients with Extrahepatic Cholangiocarcinomas

Authors: Hiroyuki Yamamoto, MD, Yoshitsugu Nakanishi, MD, PhD, Tomoko Mitsuhashi, MD, PhD, Yutaka Hatanaka, PhD, Kanako Hatanaka, PhD, Ayae Nange, MS, Yusuke Yoshida, MD, Norito Ino, MD, Masaru Go, MD, Keisuke Okamura, MD, PhD, Takahiro Tsuchikawa, MD, PhD, Toru Nakamura, MD, PhD, Takehiro Noji, MD, PhD, Toshimichi Asano, MD, PhD, Aya Matsui, MD, PhD, Kimitaka Tanaka, MD, PhD, Soichi Murakami, MD, PhD, Yuma Ebihara, MD, PhD, Yo Kurashima, MD, PhD, Toshiaki Shichinohe, MD, PhD, Satoshi Hirano, MD, PhD

Published in: Annals of Surgical Oncology | Issue 3/2024

Login to get access

Abstract

Background

Neutrophil extracellular traps (NETs) are extracellular chromatin structures composed of cytoplasmic, granular, and nuclear components of neutrophils. Recently, NETs have received much attention for their role in tumor biology; however, their impact on the postoperative prognosis of patients with extrahepatic cholangiocarcinomas (EHCCs) remains unclear. The purpose of this study was to clarify the impact of NETs identified by immunohistochemical citrullinated histone H3 (Cit-H3) staining on postoperative overall survival (OS) in patients with perihilar cholangiocarcinoma (PHCC) and distal cholangiocarcinoma (DCC).

Methods

This study included 318 patients with EHCC (PHCC, n = 192; DCC, n = 126) who underwent surgical resection with curative intent. Neutrophils and NETs were identified by immunohistochemistry using antibodies against CD15 and Cit-H3, respectively. Based on the distribution of CD15 and Cit-H3 expression in the tumor bed, the patients were classified into four groups: one negative group and three subgroups of the positive group (diffuse, intermediate, and focal subgroups).

Results

No significant difference was found in the postoperative OS rate depending on the distribution of CD15 expression in patients with PHCC or DCC. However, the three subgroups with positive Cit-H3 expression had significantly poorer OS than the negative group for both PHCC and DCC. Moreover, positive Cit-H3 was an independent OS factor in the multivariable analyses of PHCC (hazard ratio [HR] 1.69, 95% confidence interval [CI] 1.11–2.59, P = 0.0115) and DCC (HR 2.03; 95% CI 1.21–3.42, P = 0.0057).

Conclusions

The presence of NETs in the tumor microenvironment may have adverse prognostic effects in patients with EHCCs.
Appendix
Available only for authorised users
Literature
1.
go back to reference Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15:e493-503.PubMedCrossRef Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15:e493-503.PubMedCrossRef
2.
go back to reference Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30:1073–81.PubMedCrossRef Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30:1073–81.PubMedCrossRef
3.
go back to reference Smith HA, Kang Y. The metastasis-promoting roles of tumor-associated immune cells. J Mol Med (Berl). 2013;91:411–29.PubMedCrossRef Smith HA, Kang Y. The metastasis-promoting roles of tumor-associated immune cells. J Mol Med (Berl). 2013;91:411–29.PubMedCrossRef
5.
go back to reference Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–5.ADSPubMedCrossRef Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–5.ADSPubMedCrossRef
6.
go back to reference Saffarzadeh M, Juenemann C, Queisser MA, et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS ONE. 2012;7:e32366.ADSPubMedPubMedCentralCrossRef Saffarzadeh M, Juenemann C, Queisser MA, et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS ONE. 2012;7:e32366.ADSPubMedPubMedCentralCrossRef
8.
go back to reference Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. 2018;18:134–47.PubMedCrossRef Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. 2018;18:134–47.PubMedCrossRef
9.
go back to reference Jorch SK, Kubes P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med. 2017;23:279–87.PubMedCrossRef Jorch SK, Kubes P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med. 2017;23:279–87.PubMedCrossRef
10.
go back to reference Anderson CD, Pinson CW, Berlin J, Chari RS. Diagnosis and treatment of cholangiocarcinoma. Oncologist. 2004;9:43–57.PubMedCrossRef Anderson CD, Pinson CW, Berlin J, Chari RS. Diagnosis and treatment of cholangiocarcinoma. Oncologist. 2004;9:43–57.PubMedCrossRef
11.
go back to reference Amin MB, Edge SB, Greene FL. AJCC cancer staging manual. 8th edn. New York: Springer; 2017. Amin MB, Edge SB, Greene FL. AJCC cancer staging manual. 8th edn. New York: Springer; 2017.
12.
go back to reference Banales JM, Marin JJG, Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol. 2020;17:557–88.PubMedPubMedCentralCrossRef Banales JM, Marin JJG, Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol. 2020;17:557–88.PubMedPubMedCentralCrossRef
13.
go back to reference van der Gaag NA, Kloek JJ, de Bakker JK, et al. Survival analysis and prognostic nomogram for patients undergoing resection of extrahepatic cholangiocarcinoma. Ann Oncol. 2012;23:2642–9.PubMedCrossRef van der Gaag NA, Kloek JJ, de Bakker JK, et al. Survival analysis and prognostic nomogram for patients undergoing resection of extrahepatic cholangiocarcinoma. Ann Oncol. 2012;23:2642–9.PubMedCrossRef
15.
go back to reference Akita M, Ajiki T, Matsumoto T, et al. Preoperative cholangitis affects survival outcome in patients with extrahepatic bile duct cancer. J Gastrointest Surg. 2017;21:983–9.PubMedCrossRef Akita M, Ajiki T, Matsumoto T, et al. Preoperative cholangitis affects survival outcome in patients with extrahepatic bile duct cancer. J Gastrointest Surg. 2017;21:983–9.PubMedCrossRef
16.
go back to reference Wang Y, Fu W, Tang Z, Meng W, Zhou W, Li X. Effect of preoperative cholangitis on prognosis of patients with hilar cholangiocarcinoma: a systematic review and meta-analysis. Medicine (Baltimore). 2018;97:e12025.PubMedCrossRef Wang Y, Fu W, Tang Z, Meng W, Zhou W, Li X. Effect of preoperative cholangitis on prognosis of patients with hilar cholangiocarcinoma: a systematic review and meta-analysis. Medicine (Baltimore). 2018;97:e12025.PubMedCrossRef
17.
go back to reference Cho JY, Han HS, Yoon YS, et al. Preoperative cholangitis and metastatic lymph node have a negative impact on survival after resection of extrahepatic bile duct cancer. World J Surg. 2012;36:1842–7.PubMedCrossRef Cho JY, Han HS, Yoon YS, et al. Preoperative cholangitis and metastatic lymph node have a negative impact on survival after resection of extrahepatic bile duct cancer. World J Surg. 2012;36:1842–7.PubMedCrossRef
20.
go back to reference Kiriyama S, Kozaka K, Takada T, et al. Tokyo Guidelines 2018: diagnostic criteria and severity grading of acute cholangitis (with videos). J Hepatobiliary Pancreat Sci. 2018;25:17–30.PubMedCrossRef Kiriyama S, Kozaka K, Takada T, et al. Tokyo Guidelines 2018: diagnostic criteria and severity grading of acute cholangitis (with videos). J Hepatobiliary Pancreat Sci. 2018;25:17–30.PubMedCrossRef
21.
go back to reference Nakazawa D, Tomaru U, Suzuki A, et al. Abnormal conformation and impaired degradation of propylthiouracil-induced neutrophil extracellular traps: implications of disordered neutrophil extracellular traps in a rat model of myeloperoxidase antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. 2012;64:3779–87.PubMedCrossRef Nakazawa D, Tomaru U, Suzuki A, et al. Abnormal conformation and impaired degradation of propylthiouracil-induced neutrophil extracellular traps: implications of disordered neutrophil extracellular traps in a rat model of myeloperoxidase antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. 2012;64:3779–87.PubMedCrossRef
22.
go back to reference Yoshida M, Sasaki M, Sugisaki K, Yamaguchi Y, Yamada M. Neutrophil extracellular trap components in fibrinoid necrosis of the kidney with myeloperoxidase-ANCA-associated vasculitis. Clin Kidney J. 2013;6:308–12.PubMedPubMedCentralCrossRef Yoshida M, Sasaki M, Sugisaki K, Yamaguchi Y, Yamada M. Neutrophil extracellular trap components in fibrinoid necrosis of the kidney with myeloperoxidase-ANCA-associated vasculitis. Clin Kidney J. 2013;6:308–12.PubMedPubMedCentralCrossRef
23.
go back to reference Nakazawa D, Shida H, Tomaru U, et al. Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA–associated microscopic polyangiitis. J Am Soc Nephrol. 2014;25:990–7.PubMedPubMedCentralCrossRef Nakazawa D, Shida H, Tomaru U, et al. Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA–associated microscopic polyangiitis. J Am Soc Nephrol. 2014;25:990–7.PubMedPubMedCentralCrossRef
24.
go back to reference Masuda S, Nakazawa D, Shida H, et al. NETosis markers: quest for specific, objective, and quantitative markers. Clin Chim Acta. 2016;459:89–93.PubMedCrossRef Masuda S, Nakazawa D, Shida H, et al. NETosis markers: quest for specific, objective, and quantitative markers. Clin Chim Acta. 2016;459:89–93.PubMedCrossRef
25.
go back to reference Mousset A, Lecorgne E, Bourget I, et al. Neutrophil extracellular traps formed during chemotherapy confer treatment resistance via TGF-β activation. Cancer Cell. 2023;41:757-75.e10.PubMedCrossRef Mousset A, Lecorgne E, Bourget I, et al. Neutrophil extracellular traps formed during chemotherapy confer treatment resistance via TGF-β activation. Cancer Cell. 2023;41:757-75.e10.PubMedCrossRef
26.
go back to reference Zhang H, Lv H, Weng M, et al. Preoperative leukocytosis is associated with increased tumor-infiltrating neutrophil extracellular traps and worse outcomes in esophageal cancer. Ann Transl Med. 2020;8:441.PubMedPubMedCentralCrossRef Zhang H, Lv H, Weng M, et al. Preoperative leukocytosis is associated with increased tumor-infiltrating neutrophil extracellular traps and worse outcomes in esophageal cancer. Ann Transl Med. 2020;8:441.PubMedPubMedCentralCrossRef
28.
go back to reference Tohme S, Yazdani HO, Al-Khafaji AB, et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 2016;76:1367–80.PubMedPubMedCentralCrossRef Tohme S, Yazdani HO, Al-Khafaji AB, et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 2016;76:1367–80.PubMedPubMedCentralCrossRef
29.
go back to reference Zhang Y, Hu Y, Ma C, et al. Diagnostic, therapeutic predictive, and prognostic value of neutrophil extracellular traps in patients with gastric adenocarcinoma. Front Oncol. 2020;10:1036.PubMedPubMedCentralCrossRef Zhang Y, Hu Y, Ma C, et al. Diagnostic, therapeutic predictive, and prognostic value of neutrophil extracellular traps in patients with gastric adenocarcinoma. Front Oncol. 2020;10:1036.PubMedPubMedCentralCrossRef
30.
go back to reference Park J, Wysocki RW, Amoozgar Z, et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med. 2016;8:361–138.CrossRef Park J, Wysocki RW, Amoozgar Z, et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med. 2016;8:361–138.CrossRef
31.
go back to reference Arelaki S, Arampatzioglou A, Kambas K, et al. Gradient infiltration of neutrophil extracellular traps in colon cancer and evidence for their involvement in tumour growth. PLoS ONE. 2016;11:e0154484.PubMedPubMedCentralCrossRef Arelaki S, Arampatzioglou A, Kambas K, et al. Gradient infiltration of neutrophil extracellular traps in colon cancer and evidence for their involvement in tumour growth. PLoS ONE. 2016;11:e0154484.PubMedPubMedCentralCrossRef
32.
go back to reference Li M, Lin C, Leso A, Nefedova Y. Quantification of citrullinated histone H3 bound DNA for detection of neutrophil extracellular traps. Cancers. 2020;12:3424.PubMedPubMedCentralCrossRef Li M, Lin C, Leso A, Nefedova Y. Quantification of citrullinated histone H3 bound DNA for detection of neutrophil extracellular traps. Cancers. 2020;12:3424.PubMedPubMedCentralCrossRef
33.
go back to reference Yoshikawa-Kimura A, Taira K, Maruyama H, et al. Influence of a biliary stent in patients with advanced pancreatic cancer treated with modified FOLFIRINOX. Medicine (Baltimore). 2022;101:e32150.PubMedCrossRef Yoshikawa-Kimura A, Taira K, Maruyama H, et al. Influence of a biliary stent in patients with advanced pancreatic cancer treated with modified FOLFIRINOX. Medicine (Baltimore). 2022;101:e32150.PubMedCrossRef
34.
go back to reference Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis. 2012;33:949–55.PubMedCrossRef Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis. 2012;33:949–55.PubMedCrossRef
35.
go back to reference Granot Z, Jablonska J. Distinct functions of neutrophil in cancer and its regulation. Mediators Inflamm. 2015;701067. Granot Z, Jablonska J. Distinct functions of neutrophil in cancer and its regulation. Mediators Inflamm. 2015;701067.
37.
go back to reference Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med. 2010;207:1853–62.PubMedPubMedCentralCrossRef Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med. 2010;207:1853–62.PubMedPubMedCentralCrossRef
38.
go back to reference Oto J, Navarro S, Larsen AC, et al. MicroRNAs and neutrophil activation markers predict venous thrombosis in pancreatic ductal adenocarcinoma and distal extrahepatic cholangiocarcinoma. Int J Mol Sci. 2020;21:840.PubMedPubMedCentralCrossRef Oto J, Navarro S, Larsen AC, et al. MicroRNAs and neutrophil activation markers predict venous thrombosis in pancreatic ductal adenocarcinoma and distal extrahepatic cholangiocarcinoma. Int J Mol Sci. 2020;21:840.PubMedPubMedCentralCrossRef
40.
go back to reference Thålin C, Hisada Y, Lundström S, Mackman N, Wallén H. Neutrophil extracellular traps: villains and targets in arterial, venous, and cancer-associated thrombosis. Arterioscler Thromb Vasc Biol. 2019;39:1724–38.PubMedPubMedCentralCrossRef Thålin C, Hisada Y, Lundström S, Mackman N, Wallén H. Neutrophil extracellular traps: villains and targets in arterial, venous, and cancer-associated thrombosis. Arterioscler Thromb Vasc Biol. 2019;39:1724–38.PubMedPubMedCentralCrossRef
41.
42.
go back to reference Yang L, Liu Q, Zhang X, et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature. 2020;583:133–8.ADSPubMedCrossRef Yang L, Liu Q, Zhang X, et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature. 2020;583:133–8.ADSPubMedCrossRef
43.
go back to reference Cai Z, Zhang M, Boafo Kwantwi L, et al. Breast cancer cells promote self-migration by secreting interleukin 8 to induce NET formation. Gene. 2020;754:144902.PubMedCrossRef Cai Z, Zhang M, Boafo Kwantwi L, et al. Breast cancer cells promote self-migration by secreting interleukin 8 to induce NET formation. Gene. 2020;754:144902.PubMedCrossRef
44.
go back to reference Lerman I, Hammes SR. Neutrophil elastase in the tumor microenvironment. Steroids. 2018;133:96–101.PubMedCrossRef Lerman I, Hammes SR. Neutrophil elastase in the tumor microenvironment. Steroids. 2018;133:96–101.PubMedCrossRef
45.
go back to reference Okeke EB, Louttit C, Fry C, et al. Inhibition of neutrophil elastase prevents neutrophil extracellular trap formation and rescues mice from endotoxic shock. Biomaterials. 2020;238:119836.PubMedPubMedCentralCrossRef Okeke EB, Louttit C, Fry C, et al. Inhibition of neutrophil elastase prevents neutrophil extracellular trap formation and rescues mice from endotoxic shock. Biomaterials. 2020;238:119836.PubMedPubMedCentralCrossRef
46.
go back to reference Cools-Lartigue J, Spicer J, McDonald B, et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Investig. 2013;123:3446–58.PubMedPubMedCentralCrossRef Cools-Lartigue J, Spicer J, McDonald B, et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Investig. 2013;123:3446–58.PubMedPubMedCentralCrossRef
47.
go back to reference Leal AC, Mizurini DM, Gomes T, et al. Tumor-derived exosomes induce the formation of neutrophil extracellular traps: implications for the establishment of cancer-associated thrombosis. Sci Rep. 2017;7:6438.ADSPubMedPubMedCentralCrossRef Leal AC, Mizurini DM, Gomes T, et al. Tumor-derived exosomes induce the formation of neutrophil extracellular traps: implications for the establishment of cancer-associated thrombosis. Sci Rep. 2017;7:6438.ADSPubMedPubMedCentralCrossRef
Metadata
Title
Impact of Neutrophil Extracellular Traps Identified by Citrullinated Histone H3 Immunohistochemistry for Postoperative Prognosis in Patients with Extrahepatic Cholangiocarcinomas
Authors
Hiroyuki Yamamoto, MD
Yoshitsugu Nakanishi, MD, PhD
Tomoko Mitsuhashi, MD, PhD
Yutaka Hatanaka, PhD
Kanako Hatanaka, PhD
Ayae Nange, MS
Yusuke Yoshida, MD
Norito Ino, MD
Masaru Go, MD
Keisuke Okamura, MD, PhD
Takahiro Tsuchikawa, MD, PhD
Toru Nakamura, MD, PhD
Takehiro Noji, MD, PhD
Toshimichi Asano, MD, PhD
Aya Matsui, MD, PhD
Kimitaka Tanaka, MD, PhD
Soichi Murakami, MD, PhD
Yuma Ebihara, MD, PhD
Yo Kurashima, MD, PhD
Toshiaki Shichinohe, MD, PhD
Satoshi Hirano, MD, PhD
Publication date
05-12-2023
Publisher
Springer International Publishing
Published in
Annals of Surgical Oncology / Issue 3/2024
Print ISSN: 1068-9265
Electronic ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-023-14638-2

Other articles of this Issue 3/2024

Annals of Surgical Oncology 3/2024 Go to the issue