Skip to main content
Top
Published in: Cancer Cell International 1/2022

Open Access 01-12-2022 | Artificial Intelligence | Review

Advances in biomarkers and techniques for pancreatic cancer diagnosis

Authors: Haotian Wu, Suwen Ou, Hongli Zhang, Rui Huang, Shan Yu, Ming Zhao, Sheng Tai

Published in: Cancer Cell International | Issue 1/2022

Login to get access

Abstract

Pancreatic cancer is the most lethal type of malignancy and is characterized by high invasiveness without severe symptoms. It is difficult to detect PC at an early stage because of the low diagnostic accuracy of existing routine methods, such as abdominal ultrasound, CT, MRI, and endoscopic ultrasound (EUS). Therefore, it is of value to develop new diagnostic techniques for early detection with high accuracy. In this review, we aim to highlight research progress on novel biomarkers, artificial intelligence, and nanomaterial applications on the diagnostic accuracy of pancreatic cancer.
Literature
2.
go back to reference Egawa S, et al. Japan Pancreatic Cancer Registry; 30th year anniversary: Japan Pancreas Society. Pancreas. 2012;41(7):985–92.PubMedCrossRef Egawa S, et al. Japan Pancreatic Cancer Registry; 30th year anniversary: Japan Pancreas Society. Pancreas. 2012;41(7):985–92.PubMedCrossRef
3.
go back to reference Buscail E, et al. Liquid biopsy approach for pancreatic ductal adenocarcinoma. Cancers (Basel). 2019;11(6):852.CrossRef Buscail E, et al. Liquid biopsy approach for pancreatic ductal adenocarcinoma. Cancers (Basel). 2019;11(6):852.CrossRef
4.
go back to reference Gandolfi L, et al. The role of ultrasound in biliary and pancreatic diseases. Eur J Ultrasound. 2003;16(3):141–59.PubMedCrossRef Gandolfi L, et al. The role of ultrasound in biliary and pancreatic diseases. Eur J Ultrasound. 2003;16(3):141–59.PubMedCrossRef
5.
go back to reference Francis IR. Pancreatic adenocarcinoma: diagnosis and staging using multidetector-row computed tomography (MDCT) and magnetic resonance imaging (MRI). Cancer Imaging. 2007;7 Spec No A(Special issue A):S160–5.PubMedCrossRef Francis IR. Pancreatic adenocarcinoma: diagnosis and staging using multidetector-row computed tomography (MDCT) and magnetic resonance imaging (MRI). Cancer Imaging. 2007;7 Spec No A(Special issue A):S160–5.PubMedCrossRef
6.
go back to reference Fattahi R, et al. Pancreatic diffusion-weighted imaging (DWI): comparison between mass-forming focal pancreatitis (FP), pancreatic cancer (PC), and normal pancreas. J Magn Reson Imaging. 2009;29(2):350–6.PubMedCrossRef Fattahi R, et al. Pancreatic diffusion-weighted imaging (DWI): comparison between mass-forming focal pancreatitis (FP), pancreatic cancer (PC), and normal pancreas. J Magn Reson Imaging. 2009;29(2):350–6.PubMedCrossRef
7.
go back to reference Treadwell JR, et al. Imaging tests for the diagnosis and staging of pancreatic adenocarcinoma: a meta-analysis. Pancreas. 2016;45(6):789–95.PubMedCrossRef Treadwell JR, et al. Imaging tests for the diagnosis and staging of pancreatic adenocarcinoma: a meta-analysis. Pancreas. 2016;45(6):789–95.PubMedCrossRef
8.
go back to reference Kurihara K, Hanada K, Shimizu A. Endoscopic ultrasonography diagnosis of early pancreatic cancer. Diagnostics (Basel). 2020;10(12):1086.CrossRef Kurihara K, Hanada K, Shimizu A. Endoscopic ultrasonography diagnosis of early pancreatic cancer. Diagnostics (Basel). 2020;10(12):1086.CrossRef
9.
go back to reference Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol. 2007;33(3):266–70.PubMedCrossRef Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol. 2007;33(3):266–70.PubMedCrossRef
10.
go back to reference Scara S, Bottoni P, Scatena R. CA 19-9: biochemical and clinical aspects. Adv Exp Med Biol. 2015;867:247–60.PubMedCrossRef Scara S, Bottoni P, Scatena R. CA 19-9: biochemical and clinical aspects. Adv Exp Med Biol. 2015;867:247–60.PubMedCrossRef
11.
go back to reference Ferrone CR, et al. Perioperative CA19-9 levels can predict stage and survival in patients with resectable pancreatic adenocarcinoma. J Clin Oncol. 2006;24(18):2897–902.PubMedCrossRef Ferrone CR, et al. Perioperative CA19-9 levels can predict stage and survival in patients with resectable pancreatic adenocarcinoma. J Clin Oncol. 2006;24(18):2897–902.PubMedCrossRef
12.
go back to reference Wu Z, Kuntz AI, Wadleigh RG. CA 19-9 tumor marker: is it reliable? A case report in a patient with pancreatic cancer. Clin Adv Hematol Oncol. 2013;11(1):50–2.PubMed Wu Z, Kuntz AI, Wadleigh RG. CA 19-9 tumor marker: is it reliable? A case report in a patient with pancreatic cancer. Clin Adv Hematol Oncol. 2013;11(1):50–2.PubMed
13.
go back to reference Gu YL, et al. Applicative value of serum CA19-9, CEA, CA125 and CA242 in diagnosis and prognosis for patients with pancreatic cancer treated by concurrent chemoradiotherapy. Asian Pac J Cancer Prev. 2015;16(15):6569–73.PubMedCrossRef Gu YL, et al. Applicative value of serum CA19-9, CEA, CA125 and CA242 in diagnosis and prognosis for patients with pancreatic cancer treated by concurrent chemoradiotherapy. Asian Pac J Cancer Prev. 2015;16(15):6569–73.PubMedCrossRef
15.
go back to reference Henriksen SD, et al. Cell-free DNA promoter hypermethylation in plasma as a predictive marker for survival of patients with pancreatic adenocarcinoma. Oncotarget. 2017;8(55):93942–56.PubMedPubMedCentralCrossRef Henriksen SD, et al. Cell-free DNA promoter hypermethylation in plasma as a predictive marker for survival of patients with pancreatic adenocarcinoma. Oncotarget. 2017;8(55):93942–56.PubMedPubMedCentralCrossRef
16.
go back to reference BACALBASA N, et al. Does sentinel node biopsy play a role in pancreatic adenocarcinoma? J Arch Balkan Med Union. 2019;54(2):334–8.CrossRef BACALBASA N, et al. Does sentinel node biopsy play a role in pancreatic adenocarcinoma? J Arch Balkan Med Union. 2019;54(2):334–8.CrossRef
18.
go back to reference Eissa MAL, et al. Promoter methylation of ADAMTS1 and BNC1 as potential biomarkers for early detection of pancreatic cancer in blood. Clin Epigenet. 2019;11(1):59.CrossRef Eissa MAL, et al. Promoter methylation of ADAMTS1 and BNC1 as potential biomarkers for early detection of pancreatic cancer in blood. Clin Epigenet. 2019;11(1):59.CrossRef
19.
go back to reference Nishizawa N, et al. Diagnostic potential of hypermethylation of the cysteine dioxygenase 1 gene (CDO1) promoter DNA in pancreatic cancer. Cancer Sci. 2019;110(9):2846–55.PubMedPubMedCentralCrossRef Nishizawa N, et al. Diagnostic potential of hypermethylation of the cysteine dioxygenase 1 gene (CDO1) promoter DNA in pancreatic cancer. Cancer Sci. 2019;110(9):2846–55.PubMedPubMedCentralCrossRef
20.
go back to reference Kisiel JB, et al. New DNA methylation markers for pancreatic cancer: discovery, tissue validation, and pilot testing in pancreatic juice. Clin Cancer Res. 2015;21(19):4473–81.PubMedPubMedCentralCrossRef Kisiel JB, et al. New DNA methylation markers for pancreatic cancer: discovery, tissue validation, and pilot testing in pancreatic juice. Clin Cancer Res. 2015;21(19):4473–81.PubMedPubMedCentralCrossRef
21.
go back to reference Schultz NA, et al. MicroRNA biomarkers in whole blood for detection of pancreatic cancer. JAMA. 2014;311(4):392–404.PubMedCrossRef Schultz NA, et al. MicroRNA biomarkers in whole blood for detection of pancreatic cancer. JAMA. 2014;311(4):392–404.PubMedCrossRef
23.
go back to reference Yu Y, et al. Identification of Serum microRNA-25 as a novel biomarker for pancreatic cancer. Medicine (Baltimore). 2020;99(52): e23863.CrossRef Yu Y, et al. Identification of Serum microRNA-25 as a novel biomarker for pancreatic cancer. Medicine (Baltimore). 2020;99(52): e23863.CrossRef
25.
26.
go back to reference Debernardi S, et al. Noninvasive urinary miRNA biomarkers for early detection of pancreatic adenocarcinoma. Am J Cancer Res. 2015;5(11):3455–66.PubMedPubMedCentral Debernardi S, et al. Noninvasive urinary miRNA biomarkers for early detection of pancreatic adenocarcinoma. Am J Cancer Res. 2015;5(11):3455–66.PubMedPubMedCentral
27.
go back to reference Yang JY, et al. MicroRNAs in stool samples as potential screening biomarkers for pancreatic ductal adenocarcinoma cancer. Am J Cancer Res. 2014;4(6):663–73.PubMedPubMedCentral Yang JY, et al. MicroRNAs in stool samples as potential screening biomarkers for pancreatic ductal adenocarcinoma cancer. Am J Cancer Res. 2014;4(6):663–73.PubMedPubMedCentral
28.
go back to reference Machida T, et al. miR-1246 and miR-4644 in salivary exosome as potential biomarkers for pancreatobiliary tract cancer. Oncol Rep. 2016;36(4):2375–81.PubMedCrossRef Machida T, et al. miR-1246 and miR-4644 in salivary exosome as potential biomarkers for pancreatobiliary tract cancer. Oncol Rep. 2016;36(4):2375–81.PubMedCrossRef
29.
go back to reference Ma Z, et al. Long non-coding RNA SNHG15 inhibits P15 and KLF2 expression to promote pancreatic cancer proliferation through EZH2-mediated H3K27me3. Oncotarget. 2017;8(48):84153–67.PubMedPubMedCentralCrossRef Ma Z, et al. Long non-coding RNA SNHG15 inhibits P15 and KLF2 expression to promote pancreatic cancer proliferation through EZH2-mediated H3K27me3. Oncotarget. 2017;8(48):84153–67.PubMedPubMedCentralCrossRef
30.
go back to reference Guo XB, Yin HS, Wang JY. Evaluating the diagnostic and prognostic value of long non-coding RNA SNHG15 in pancreatic ductal adenocarcinoma. Eur Rev Med Pharmacol Sci. 2018;22(18):5892–8.PubMed Guo XB, Yin HS, Wang JY. Evaluating the diagnostic and prognostic value of long non-coding RNA SNHG15 in pancreatic ductal adenocarcinoma. Eur Rev Med Pharmacol Sci. 2018;22(18):5892–8.PubMed
31.
34.
go back to reference Li J, et al. Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res. 2018;37(1):177.PubMedPubMedCentralCrossRef Li J, et al. Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res. 2018;37(1):177.PubMedPubMedCentralCrossRef
35.
go back to reference Li Z, et al. Tumor-released exosomal circular RNA PDE8A promotes invasive growth via the miR-338/MACC1/MET pathway in pancreatic cancer. Cancer Lett. 2018;432:237–50.PubMedCrossRef Li Z, et al. Tumor-released exosomal circular RNA PDE8A promotes invasive growth via the miR-338/MACC1/MET pathway in pancreatic cancer. Cancer Lett. 2018;432:237–50.PubMedCrossRef
36.
go back to reference Shen X, et al. Identification of Circ_001569 as a potential biomarker in the diagnosis and prognosis of pancreatic cancer. Technol Cancer Res Treat. 2021;20:1533033820983302.PubMedPubMedCentralCrossRef Shen X, et al. Identification of Circ_001569 as a potential biomarker in the diagnosis and prognosis of pancreatic cancer. Technol Cancer Res Treat. 2021;20:1533033820983302.PubMedPubMedCentralCrossRef
39.
go back to reference Sun L, et al. CPA4 is a promising diagnostic serum biomarker for pancreatic cancer. Am J Cancer Res. 2016;6(1):91–6.PubMed Sun L, et al. CPA4 is a promising diagnostic serum biomarker for pancreatic cancer. Am J Cancer Res. 2016;6(1):91–6.PubMed
40.
go back to reference Sogawa K, et al. Identification of a novel serum biomarker for pancreatic cancer, C4b-binding protein α-chain (C4BPA) by quantitative proteomic analysis using tandem mass tags. Br J Cancer. 2016;115(8):949–56.PubMedPubMedCentralCrossRef Sogawa K, et al. Identification of a novel serum biomarker for pancreatic cancer, C4b-binding protein α-chain (C4BPA) by quantitative proteomic analysis using tandem mass tags. Br J Cancer. 2016;115(8):949–56.PubMedPubMedCentralCrossRef
41.
go back to reference Fukutake N, et al. A novel multivariate index for pancreatic cancer detection based on the plasma free amino acid profile. PLoS ONE. 2015;10(7):e0132223.PubMedPubMedCentralCrossRef Fukutake N, et al. A novel multivariate index for pancreatic cancer detection based on the plasma free amino acid profile. PLoS ONE. 2015;10(7):e0132223.PubMedPubMedCentralCrossRef
42.
go back to reference Kaur S, et al. A combination of MUC5AC and CA19-9 improves the diagnosis of pancreatic cancer: a multicenter study. Am J Gastroenterol. 2017;112(1):172–83.PubMedCrossRef Kaur S, et al. A combination of MUC5AC and CA19-9 improves the diagnosis of pancreatic cancer: a multicenter study. Am J Gastroenterol. 2017;112(1):172–83.PubMedCrossRef
43.
go back to reference Poruk KE, et al. Serum osteopontin and tissue inhibitor of metalloproteinase 1 as diagnostic and prognostic biomarkers for pancreatic adenocarcinoma. Pancreas. 2013;42(2):193–7.PubMedPubMedCentralCrossRef Poruk KE, et al. Serum osteopontin and tissue inhibitor of metalloproteinase 1 as diagnostic and prognostic biomarkers for pancreatic adenocarcinoma. Pancreas. 2013;42(2):193–7.PubMedPubMedCentralCrossRef
44.
go back to reference Radon TP, et al. Identification of a three-biomarker panel in urine for early detection of pancreatic adenocarcinoma. Clin Cancer Res. 2015;21(15):3512–21.PubMedPubMedCentralCrossRef Radon TP, et al. Identification of a three-biomarker panel in urine for early detection of pancreatic adenocarcinoma. Clin Cancer Res. 2015;21(15):3512–21.PubMedPubMedCentralCrossRef
45.
go back to reference Hogendorf P, et al. Neutrophil Gelatinase-Associated Lipocalin (NGAL) concentration in urine is superior to CA19-9 and Ca 125 in differentiation of pancreatic mass: preliminary report. Cancer Biomark. 2016;16(4):537–43.PubMedCrossRef Hogendorf P, et al. Neutrophil Gelatinase-Associated Lipocalin (NGAL) concentration in urine is superior to CA19-9 and Ca 125 in differentiation of pancreatic mass: preliminary report. Cancer Biomark. 2016;16(4):537–43.PubMedCrossRef
47.
go back to reference Zhang WH, et al. Advances on diagnostic biomarkers of pancreatic ductal adenocarcinoma: a systems biology perspective. Comput Struct Biotechnol J. 2020;18:3606–14.PubMedPubMedCentralCrossRef Zhang WH, et al. Advances on diagnostic biomarkers of pancreatic ductal adenocarcinoma: a systems biology perspective. Comput Struct Biotechnol J. 2020;18:3606–14.PubMedPubMedCentralCrossRef
48.
go back to reference Terai K, et al. Levels of soluble LR11/SorLA are highly increased in the bile of patients with biliary tract and pancreatic cancers. Clin Chim Acta. 2016;457:130–6.PubMedCrossRef Terai K, et al. Levels of soluble LR11/SorLA are highly increased in the bile of patients with biliary tract and pancreatic cancers. Clin Chim Acta. 2016;457:130–6.PubMedCrossRef
49.
go back to reference Du T, et al. The diagnosis and staging of pancreatic cancer: a comparison of endoscopic ultrasound and computed tomography with pancreas protocol. Am J Surg. 2018;215(3):472–5.PubMedCrossRef Du T, et al. The diagnosis and staging of pancreatic cancer: a comparison of endoscopic ultrasound and computed tomography with pancreas protocol. Am J Surg. 2018;215(3):472–5.PubMedCrossRef
50.
go back to reference Walczak S, Velanovich V. An evaluation of artificial neural networks in predicting pancreatic cancer survival. J Gastrointest Surg. 2017;21(10):1606–12.PubMedCrossRef Walczak S, Velanovich V. An evaluation of artificial neural networks in predicting pancreatic cancer survival. J Gastrointest Surg. 2017;21(10):1606–12.PubMedCrossRef
51.
go back to reference Le Berre C, et al. Application of artificial intelligence to gastroenterology and hepatology. Gastroenterology. 2020;158(1):76-94.e2.PubMedCrossRef Le Berre C, et al. Application of artificial intelligence to gastroenterology and hepatology. Gastroenterology. 2020;158(1):76-94.e2.PubMedCrossRef
52.
go back to reference Liang M, et al. Low-dose CT screening for lung cancer: computer-aided detection of missed lung cancers. Radiology. 2016;281(1):279–88.PubMedCrossRef Liang M, et al. Low-dose CT screening for lung cancer: computer-aided detection of missed lung cancers. Radiology. 2016;281(1):279–88.PubMedCrossRef
53.
go back to reference Le MH, et al. Automated diagnosis of prostate cancer in multi-parametric MRI based on multimodal convolutional neural networks. Phys Med Biol. 2017;62(16):6497–514.PubMedCrossRef Le MH, et al. Automated diagnosis of prostate cancer in multi-parametric MRI based on multimodal convolutional neural networks. Phys Med Biol. 2017;62(16):6497–514.PubMedCrossRef
54.
go back to reference Lu Y, et al. Identification of metastatic lymph nodes in MR imaging with faster region-based convolutional neural networks. Cancer Res. 2018;78(17):5135–43.PubMedCrossRef Lu Y, et al. Identification of metastatic lymph nodes in MR imaging with faster region-based convolutional neural networks. Cancer Res. 2018;78(17):5135–43.PubMedCrossRef
55.
go back to reference Li X, et al. Diagnosis of thyroid cancer using deep convolutional neural network models applied to sonographic images: a retrospective, multicohort, diagnostic study. Lancet Oncol. 2019;20(2):193–201.PubMedCrossRef Li X, et al. Diagnosis of thyroid cancer using deep convolutional neural network models applied to sonographic images: a retrospective, multicohort, diagnostic study. Lancet Oncol. 2019;20(2):193–201.PubMedCrossRef
56.
go back to reference Goyal H, et al. Application of artificial intelligence in pancreaticobiliary diseases. Ther Adv Gastrointest Endosc. 2021;14:2631774521993059.PubMedPubMedCentral Goyal H, et al. Application of artificial intelligence in pancreaticobiliary diseases. Ther Adv Gastrointest Endosc. 2021;14:2631774521993059.PubMedPubMedCentral
57.
go back to reference Prasoppokakorn T, et al. Application of artificial intelligence for diagnosis of pancreatic ductal adenocarcinoma by EUS: a systematic review and meta-analysis. Endosc Ultrasound. 2022;11(1):17–26.PubMed Prasoppokakorn T, et al. Application of artificial intelligence for diagnosis of pancreatic ductal adenocarcinoma by EUS: a systematic review and meta-analysis. Endosc Ultrasound. 2022;11(1):17–26.PubMed
58.
go back to reference Chassagnon G, et al. Artificial intelligence applications for thoracic imaging. Eur J Radiol. 2020;123:108774.PubMedCrossRef Chassagnon G, et al. Artificial intelligence applications for thoracic imaging. Eur J Radiol. 2020;123:108774.PubMedCrossRef
59.
go back to reference Sheth D, Giger ML. Artificial intelligence in the interpretation of breast cancer on MRI. J Magn Reson Imaging. 2020;51(5):1310–24.PubMedCrossRef Sheth D, Giger ML. Artificial intelligence in the interpretation of breast cancer on MRI. J Magn Reson Imaging. 2020;51(5):1310–24.PubMedCrossRef
60.
go back to reference Liu KL, et al. Deep learning to distinguish pancreatic cancer tissue from non-cancerous pancreatic tissue: a retrospective study with cross-racial external validation. Lancet Digit Health. 2020;2(6):e303–13.PubMedCrossRef Liu KL, et al. Deep learning to distinguish pancreatic cancer tissue from non-cancerous pancreatic tissue: a retrospective study with cross-racial external validation. Lancet Digit Health. 2020;2(6):e303–13.PubMedCrossRef
61.
go back to reference Liu SL, et al. Establishment and application of an artificial intelligence diagnosis system for pancreatic cancer with a faster region-based convolutional neural network. Chin Med J (Engl). 2019;132(23):2795–803.CrossRef Liu SL, et al. Establishment and application of an artificial intelligence diagnosis system for pancreatic cancer with a faster region-based convolutional neural network. Chin Med J (Engl). 2019;132(23):2795–803.CrossRef
62.
go back to reference Gao X, Wang X. Performance of deep learning for differentiating pancreatic diseases on contrast-enhanced magnetic resonance imaging: a preliminary study. Diagn Interv Imaging. 2020;101(2):91–100.PubMedCrossRef Gao X, Wang X. Performance of deep learning for differentiating pancreatic diseases on contrast-enhanced magnetic resonance imaging: a preliminary study. Diagn Interv Imaging. 2020;101(2):91–100.PubMedCrossRef
63.
go back to reference Li S, et al. An effective computer aided diagnosis model for pancreas cancer on PET/CT images. Comput Methods Progr Biomed. 2018;165:205–14.CrossRef Li S, et al. An effective computer aided diagnosis model for pancreas cancer on PET/CT images. Comput Methods Progr Biomed. 2018;165:205–14.CrossRef
64.
go back to reference Das A, et al. Digital image analysis of EUS images accurately differentiates pancreatic cancer from chronic pancreatitis and normal tissue. Gastrointest Endosc. 2008;67(6):861–7.PubMedCrossRef Das A, et al. Digital image analysis of EUS images accurately differentiates pancreatic cancer from chronic pancreatitis and normal tissue. Gastrointest Endosc. 2008;67(6):861–7.PubMedCrossRef
65.
go back to reference Zhu M, et al. Differentiation of pancreatic cancer and chronic pancreatitis using computer-aided diagnosis of endoscopic ultrasound (EUS) images: a diagnostic test. PLoS ONE. 2013;8(5):e63820.PubMedPubMedCentralCrossRef Zhu M, et al. Differentiation of pancreatic cancer and chronic pancreatitis using computer-aided diagnosis of endoscopic ultrasound (EUS) images: a diagnostic test. PLoS ONE. 2013;8(5):e63820.PubMedPubMedCentralCrossRef
66.
go back to reference Săftoiu A, et al. Quantitative contrast-enhanced harmonic EUS in differential diagnosis of focal pancreatic masses (with videos). Gastrointest Endosc. 2015;82(1):59–69.PubMedCrossRef Săftoiu A, et al. Quantitative contrast-enhanced harmonic EUS in differential diagnosis of focal pancreatic masses (with videos). Gastrointest Endosc. 2015;82(1):59–69.PubMedCrossRef
67.
go back to reference Momeni-Boroujeni A, Yousefi E, Somma J. Computer-assisted cytologic diagnosis in pancreatic FNA: an application of neural networks to image analysis. Cancer Cytopathol. 2017;125(12):926–33.PubMedCrossRef Momeni-Boroujeni A, Yousefi E, Somma J. Computer-assisted cytologic diagnosis in pancreatic FNA: an application of neural networks to image analysis. Cancer Cytopathol. 2017;125(12):926–33.PubMedCrossRef
68.
go back to reference Zhang K, et al. Clinically applicable AI system for accurate diagnosis, quantitative measurements, and prognosis of COVID-19 pneumonia using computed tomography. Cell. 2020;181(6):1423-1433.e11.PubMedPubMedCentralCrossRef Zhang K, et al. Clinically applicable AI system for accurate diagnosis, quantitative measurements, and prognosis of COVID-19 pneumonia using computed tomography. Cell. 2020;181(6):1423-1433.e11.PubMedPubMedCentralCrossRef
69.
go back to reference Yang Y, et al. Diagnosis of pancreatic carcinoma based on combined measurement of multiple serum tumor markers using artificial neural network analysis. Chin Med J (Engl). 2014;127(10):1891–6. Yang Y, et al. Diagnosis of pancreatic carcinoma based on combined measurement of multiple serum tumor markers using artificial neural network analysis. Chin Med J (Engl). 2014;127(10):1891–6.
70.
go back to reference Lee J, et al. Identification of circulating serum miRNAs as novel biomarkers in pancreatic cancer using a penalized algorithm. Int J Mol Sci. 2021;22(3):1007.PubMedPubMedCentralCrossRef Lee J, et al. Identification of circulating serum miRNAs as novel biomarkers in pancreatic cancer using a penalized algorithm. Int J Mol Sci. 2021;22(3):1007.PubMedPubMedCentralCrossRef
71.
go back to reference Burke BP, Cawthorne C, Archibald SJ. Multimodal nanoparticle imaging agents: design and applications. Philos Trans A Math Phys Eng Sci. 2017;375(2107):20170261.PubMed Burke BP, Cawthorne C, Archibald SJ. Multimodal nanoparticle imaging agents: design and applications. Philos Trans A Math Phys Eng Sci. 2017;375(2107):20170261.PubMed
72.
go back to reference Asem H, et al. Biodistribution of biodegradable polymeric nano-carriers loaded with busulphan and designed for multimodal imaging. J Nanobiotechnol. 2016;14(1):82.CrossRef Asem H, et al. Biodistribution of biodegradable polymeric nano-carriers loaded with busulphan and designed for multimodal imaging. J Nanobiotechnol. 2016;14(1):82.CrossRef
73.
go back to reference Barrefelt Å, et al. Fluorescence labeled microbubbles for multimodal imaging. Biochem Biophys Res Commun. 2015;464(3):737–42.PubMedCrossRef Barrefelt Å, et al. Fluorescence labeled microbubbles for multimodal imaging. Biochem Biophys Res Commun. 2015;464(3):737–42.PubMedCrossRef
74.
go back to reference Ye F, et al. Advances in nanotechnology for cancer biomarkers. J Nano Today. 2018;18:103–23.CrossRef Ye F, et al. Advances in nanotechnology for cancer biomarkers. J Nano Today. 2018;18:103–23.CrossRef
75.
go back to reference Hahn MA, et al. Nanoparticles as contrast agents for in-vivo bioimaging: current status and future perspectives. Anal Bioanal Chem. 2011;399(1):3–27.PubMedCrossRef Hahn MA, et al. Nanoparticles as contrast agents for in-vivo bioimaging: current status and future perspectives. Anal Bioanal Chem. 2011;399(1):3–27.PubMedCrossRef
76.
go back to reference Rosenberger I, et al. Targeted diagnostic magnetic nanoparticles for medical imaging of pancreatic cancer. J Control Release. 2015;214:76–84.PubMedCrossRef Rosenberger I, et al. Targeted diagnostic magnetic nanoparticles for medical imaging of pancreatic cancer. J Control Release. 2015;214:76–84.PubMedCrossRef
77.
go back to reference Luo Y, et al. Hyaluronic acid-mediated multifunctional iron oxide-based MRI nanoprobes for dynamic monitoring of pancreatic cancer. RSC Adv. 2019;9:10486–93.PubMedPubMedCentralCrossRef Luo Y, et al. Hyaluronic acid-mediated multifunctional iron oxide-based MRI nanoprobes for dynamic monitoring of pancreatic cancer. RSC Adv. 2019;9:10486–93.PubMedPubMedCentralCrossRef
78.
go back to reference Boyer C, et al. The design and utility of polymer-stabilized iron-oxide nanoparticles for nanomedicine applications. NPG Asia Mater. 2009;2:23–30.CrossRef Boyer C, et al. The design and utility of polymer-stabilized iron-oxide nanoparticles for nanomedicine applications. NPG Asia Mater. 2009;2:23–30.CrossRef
81.
go back to reference Sun X, et al. Tracking stem cells and macrophages with gold and iron oxide nanoparticles-The choice of the best suited particles. Appl Mater Today. 2019;15:267–79.CrossRef Sun X, et al. Tracking stem cells and macrophages with gold and iron oxide nanoparticles-The choice of the best suited particles. Appl Mater Today. 2019;15:267–79.CrossRef
82.
go back to reference Zhuo Y, et al. Functionalized SiO2 labeled CA19-9 antibodies: a new strategy for signal amplification of antigen-antibody sensing processes. Analyst. 2010;135(8):2036–42.PubMedCrossRef Zhuo Y, et al. Functionalized SiO2 labeled CA19-9 antibodies: a new strategy for signal amplification of antigen-antibody sensing processes. Analyst. 2010;135(8):2036–42.PubMedCrossRef
83.
go back to reference Jin W, et al. A simple MWCNTs@paper biosensor for CA19-9 detection and its long-term preservation by vacuum freeze drying. Int J Biol Macromol. 2020;144:995–1003.PubMedCrossRef Jin W, et al. A simple MWCNTs@paper biosensor for CA19-9 detection and its long-term preservation by vacuum freeze drying. Int J Biol Macromol. 2020;144:995–1003.PubMedCrossRef
84.
go back to reference Morgan NY, et al. Real time in vivo non-invasive optical imaging using near-infrared fluorescent quantum dots. Acad Radiol. 2005;12(3):313–23.PubMedCrossRef Morgan NY, et al. Real time in vivo non-invasive optical imaging using near-infrared fluorescent quantum dots. Acad Radiol. 2005;12(3):313–23.PubMedCrossRef
85.
go back to reference Raffa V, et al. Progress in nanotechnology for healthcare. Minim Invasive Ther Allied Technol. 2010;19(3):127–35.PubMedCrossRef Raffa V, et al. Progress in nanotechnology for healthcare. Minim Invasive Ther Allied Technol. 2010;19(3):127–35.PubMedCrossRef
86.
go back to reference Gu B, et al. ZnO quantum dot labeled immunosensor for carbohydrate antigen 19–9. Biosens Bioelectron. 2011;26(5):2720–3.PubMedCrossRef Gu B, et al. ZnO quantum dot labeled immunosensor for carbohydrate antigen 19–9. Biosens Bioelectron. 2011;26(5):2720–3.PubMedCrossRef
87.
go back to reference Kumar R, et al. Covalently dye-linked, surface-controlled, and bioconjugated organically modified silica nanoparticles as targeted probes for optical imaging. ACS Nano. 2008;2(3):449–56.PubMedCrossRef Kumar R, et al. Covalently dye-linked, surface-controlled, and bioconjugated organically modified silica nanoparticles as targeted probes for optical imaging. ACS Nano. 2008;2(3):449–56.PubMedCrossRef
Metadata
Title
Advances in biomarkers and techniques for pancreatic cancer diagnosis
Authors
Haotian Wu
Suwen Ou
Hongli Zhang
Rui Huang
Shan Yu
Ming Zhao
Sheng Tai
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2022
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-022-02640-9

Other articles of this Issue 1/2022

Cancer Cell International 1/2022 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine