“Faster diagnosis, better outcomes: Biosensors pave the way for a brighter future for cancer patients”. As one of the top causes of death worldwide, cancer must be addressed with the help of innovative treatments and state-of-the-art diagnostic techniques. Due to stress, poor lifestyle choices, and environmental factors, cancer incidence is worryingly on the rise in India, especially among the younger generation. In India, 1 in 5 persons may receive a cancer diagnosis by 2025, potentially impacting 1.57 million people, even though 30–50% of cancers are preventable. Even though standard screening techniques are frequently too costly and impracticable for everyday use, early detection is vital. Alternatives that show promise include emerging biosensor technologies, which give quick, accurate, and customized diagnostic results. Due to its capacity to quickly and automatically identify biological changes, ultra-sensitive biosensing systems utilizing single chips have revolutionized cancer detection. Since they are more effective than conventional techniques, point-of-care (PoC) biosensors—such as innovative nano-sensing devices for exosomal micro-RNA analysis—are becoming increasingly popular. Developing sophisticated diagnostic instruments like bio-computers and resonant mirrors is made easier by these biosensors, which combine analytes, receptors, and electrical sensors to detect cancer biomarkers in biological samples. The accuracy and usability of detection are further improved by advancements in wearable technologies, microfluidics, and electrochemical and graphene-based sensors. BrCyS-Q and NanoLiposomes provide improved photodynamic treatment and targeted medication delivery, respectively. Improved patient outcomes and early intervention are anticipated using the i-Genbox, a colorimetric sensor based on LAMP technology, and DNA-SWCNT-based sensors that further improve biomarker identification for gynecologic tumors.
Citation: Dinesh Bhatia, Tania Acharjee, Monika Bhatia. Advancements in nanosensors for cancer detection[J]. AIMS Biophysics, 2024, 11(4): 527-583. doi: 10.3934/biophy.2024028
“Faster diagnosis, better outcomes: Biosensors pave the way for a brighter future for cancer patients”. As one of the top causes of death worldwide, cancer must be addressed with the help of innovative treatments and state-of-the-art diagnostic techniques. Due to stress, poor lifestyle choices, and environmental factors, cancer incidence is worryingly on the rise in India, especially among the younger generation. In India, 1 in 5 persons may receive a cancer diagnosis by 2025, potentially impacting 1.57 million people, even though 30–50% of cancers are preventable. Even though standard screening techniques are frequently too costly and impracticable for everyday use, early detection is vital. Alternatives that show promise include emerging biosensor technologies, which give quick, accurate, and customized diagnostic results. Due to its capacity to quickly and automatically identify biological changes, ultra-sensitive biosensing systems utilizing single chips have revolutionized cancer detection. Since they are more effective than conventional techniques, point-of-care (PoC) biosensors—such as innovative nano-sensing devices for exosomal micro-RNA analysis—are becoming increasingly popular. Developing sophisticated diagnostic instruments like bio-computers and resonant mirrors is made easier by these biosensors, which combine analytes, receptors, and electrical sensors to detect cancer biomarkers in biological samples. The accuracy and usability of detection are further improved by advancements in wearable technologies, microfluidics, and electrochemical and graphene-based sensors. BrCyS-Q and NanoLiposomes provide improved photodynamic treatment and targeted medication delivery, respectively. Improved patient outcomes and early intervention are anticipated using the i-Genbox, a colorimetric sensor based on LAMP technology, and DNA-SWCNT-based sensors that further improve biomarker identification for gynecologic tumors.
Artificial Bee Colony-Based Backpropagation Neural Network
Artificial Intelligence
Albumin
Acute Lymphoblastic Leukemia Image Database
Artificial Neural Network
American Society of Hematology
Gold Nanoparticles
Beta Human Chorionic Gonadotropin
Before the Common Era
Cancer Antigen 125
Carbohydrate Antigen 19-9
Cluster of Differentiation 63 (a protein commonly found on exosomes)
Carcinoembryonic Antigen
Cell-Free DNA
Convolutional Neural Network
Circulating Tumor Cells
Circulating Tumor Cells
Colorectal Cancer
Deoxyribonucleic Acid
DNA-Single-Walled Carbon Nanotube
Digital Breast Tomosynthesis
Digital Mammography
Discrete Orthogonal Stockwell Transform
Enzyme-Linked Immunosorbent Assay
Enhanced Permeability and Retention
Food and Drug Administration
Fluorescence-Activated Cell Sorting
Fluorescence Immunophenotyping and Interphase Cytogenetics as a Tool for Investigation of Neoplasms
Fluorescence in Situ Hybridization
Histological Image Analysis
Human Epididymis Protein 4
Human Papillomavirus
High-Performance Liquid Chromatography
Internet of Medical Things
Infrared
K-Nearest Neighbors
Lung Cancer
Linear Discriminant Analysis
Lung-Metastasis Initiating Stem Cells
Loop-Mediated Isothermal Amplification
Magnetic-Activated Cell Sorting
Metal Insulator Metal
Metastasis-Initiating Stem Cells
MicroRNA
MicroRNAs
Multiple Myeloma
Multidrug Resistance
Non-Communicable Diseases
Next-Generation Sequencing
Natural Killer Cells
Near-Infrared
Nanoparticles
Positron Emission Tomography
Principal Component Analysis
Prostate-Specific Antigen
Phosphoserine
Photodynamic Therapy
Ribonucleic Acid
Reduced Graphene Oxide/Gold Nanoparticles
Residual Convolutional Neural Network with 34 layers
Random Forest
Reactive Oxygen Species
Surface-Enhanced Raman Spectroscopy
Support Vector Machine
Single-Walled Carbon Nanotube
Tumor-Derived Exosomes
Tumor-Derived Exosomes
Units per Milliliter
Volatile Organic Compounds
White Blood Cells
X-radiation (a form of electromagnetic radiation)
Chitinase-3-like Protein 1
[1] | Printz C (2019) American Cancer Society study: the percentage of cancers associated with excess body weight varies by state. Cancer 125: 1956-1957. https://doi.org/10.1002/cncr.32194 |
[2] | Harris E (2024) Prostate cancer cases might rise to 3 million globally by 2040. JAMA 331: 1698. https://doi.org/10.1001/jama.2024.6729 |
[3] | Ozkan-Ariksoysal D (2022) Current perspectives in graphene oxide-based electrochemical biosensors for cancer diagnostics. Biosensors 12: 607. https://doi.org/10.3390/bios12080607 |
[4] | Atikukke G, Alkhateeb A, Porter L, et al. (2020) P-370 Comprehensive targeted genomic profiling and comparative genomic analysis to identify molecular mechanisms driving cancer progression in young-onset sporadic colorectal cancer. Ann Oncol 31: S209-S210. https://doi.org/10.1016/j.annonc.2020.04.452 |
[5] | Cheng WT, Yao YH, Li DY, et al. (2023b) Asymmetrically split DNAzyme-based colorimetric and electrochemical dual-modal biosensor for detection of breast cancer exosomal surface proteins. Biosens Bioelectron 238: 115552. https://doi.org/10.1016/j.bios.2023.115552 |
[6] | Doval D (2017) Commentary: eight year survival analysis of patients with T-triple negative breast cancer in India. J Cancer Treat Diagn 1: 4-5. https://doi.org/10.29245/2578-2967/2018/1.110 |
[7] | Mathey-Andrews C (2018) Small but mighty: microRNAs as novel signalling molecules in cancer. Doctoral dissertation, Harvard University . https://doi.org/10.14800/rd.627 |
[8] | Bosch X (2000) Earliest stages of tumour-induced angiogenesis dissected. Lancet 355: 382. https://doi.org/10.1016/s0140-6736(05)74005-8 |
[9] | Wang L (2018) Novel exosome proteins as potential biomarkers for early detection of lung cancer. International Conference on Cancer Research & Diagnostics 16th Asia Pacific Biotechnology Congress. J Cancer Diagn 3: 11-16. https://doi.org/10.4172/2476-2253-c1-001 |
[10] | Somatostatin analogue scintigraphy in patients with small cell lung cancer (SCLC) and non small cell lung cancer (NSCLC). Lung Cancer (1994) 11: 65. https://doi.org/10.1016/0169-5002(94)94020-7 |
[11] | Gao J, Lv F, Li J, et al. (2014) Serum cytokeratin 19 fragment, CK19-2G2, as a newly identified biomarker for lung cancer. Plos One 9: e101979. https://doi.org/10.1371/journal.pone.0101979 |
[12] | Tutanov OS, Glass SE, Coffey RJ, et al. (2023) Emerging connections between GPI-anchored proteins and their extracellular carriers in colorectal cancer. Extracell Vesicles Circ Nucl Acids 4: 195-217. https://doi.org/10.20517/evcna.2023.17 |
[13] | Borah N, Gogoi D, Ghosh NN, et al. (2023) SP-AuNP@Tollens' complex as a highly sensitive plasmonic nanosensor for detection of formaldehyde and benzaldehyde in preserved food products. Food Chem 399: 133975. https://doi.org/10.1016/j.foodchem.2022.133975 |
[14] | Tobita A, Takao J, Endo T, et al. (2023) Single cycle selection of CD63-targeting aptamers using a microscale electrophoretic filtration device. BUNSEKI KAGAKU 72: 111-116. https://doi.org/10.2116/bunsekikagaku.72.111 |
[15] | Seddon AB (2013) Mid-infrared (IR)—A hot topic: The potential for using mid-IR light for non-invasive early detection of skin cancer in vivo. Phys Status Solidi (B) 250: 1020-1027. https://doi.org/10.1002/pssb.201248524 |
[16] | Sandberg AA (1987) 21 Cytogenetic definition of cancer subtypes. Cancer Genet Cytogen 28: 34. https://doi.org/10.1016/0165-4608(87)90300-1 |
[17] | Rastogi N, Mishra DP, et al. (2012) Therapeutic targeting of cancer cell cycle using proteasome inhibitors. Cell Div 7: 26. https://doi.org/10.1186/1747-1028-7-26 |
[18] | Goutzanis L (2022) Differential retrospective analysis in oral cancerous, pre-cancerous, and benign tissue biopsies. Cureus 14: e24956. https://doi.org/10.7759/cureus.24956 |
[19] | Ghosh SK (2017) Giovanni Battista Morgagni (1682–1771): father of pathologic anatomy and pioneer of modern medicine. Anat Sci Int 92: 305-312. https://doi.org/10.1007/s12565-016-0373-7 |
[20] | Melicow MM (1975) Percivall Pott (1713–1788) 200th anniversary of first report of occupation-induced cancer of scrotum in chimney sweepers (1775). Urology 6: 745-749. https://doi.org/10.1016/0090-4295(75)90812-2 |
[21] | Thomsen LT, Kjær SK (2021) Human papillomavirus (HPV) testing for cervical cancer screening in a middle-income country: comment on a large real-world implementation study in China. BMC Med 19: 165. https://doi.org/10.1186/s12916-021-02051-z |
[22] | Hoppenrath T (2006) Silent waves: theory and practice of lymph drainage therapy, ed 2. Phys Ther 86: 146-147. https://doi.org/10.1093/ptj/86.1.146 |
[23] | Feng Z, Zhang L, Liu Y, et al. (2022) NCAPG2 contributes to the progression of malignant melanoma through regulating proliferation and metastasis. Biochem Cell Biol 100: 473-484. https://doi.org/10.1139/bcb-2022-0048 |
[24] | JAMA Revisited.Wilhelm Konrad Roentgen—The centennial of his birth—semicentennial of the X-Rays. JAMA (2020) 323: 1512. https://doi.org/10.1001/jama.2019.13400 |
[25] | Munaron L, Antoniotti S, Lovisolo D (2004) Intracellular calcium signals and control of cell proliferation: how many mechanisms?. J Cell Mol Med 8: 161-168. https://doi.org/10.1111/j.1582-4934.2004.tb00271.x |
[26] | Sakai K, Shiina M, Ishihara N, et al. (1984) Thorotrast-induced multiple primary malignant tumors of the liver--cholangiocarcinoma and malignant hemangioendothelioma. Jpn J Clin Oncol 14: 411-416. https://doi.org/10.1093/oxfordjournals.jjco.a038994 |
[27] | Kumar S, Carter LF (2011) Giant cell tumor of soft tissue of hand: simple but rare diagnosis, which is often missed. Clinics Pract 1: e54. https://doi.org/10.4081/cp.2011.e54 |
[28] | Wahrenbrock MG, Varki A (2006) Multiple hepatic receptors cooperate to eliminate secretory mucins aberrantly entering the bloodstream: are circulating cancer mucins the “tip of the iceberg”?. Cancer Res 66: 2433-2441. https://doi.org/10.1158/0008-5472.can-05-3851 |
[29] | Imai K, Ichinose Y, Kubota Y, et al. (1994) Clinical significance of prostate specific antigen for early stage prostate cancer detection. Jpn J Clin Oncol 24: 160-165. https://doi.org/10.1093/oxfordjournals.jjco.a039697 |
[30] | Liu J, Xing Y, Sun LQ, et al. (2022) Commentary: the tumor markers and blood inflammation markers are more likely to be the indicators for differentiating benign and malignant pancreatic mucinous cystic neoplasms. Front Oncol 12: 831355. https://doi.org/10.3389/fonc.2022.901010 |
[31] | Anumula KR, Schulz R, Back N (1989) Quantitative determination of kinins released by trypsin using enzyme-linked immunosorbent assay (ELISA) and identification by high-performance liquid chromatography (HPLC). Biochem Pharmacol 38: 2421-2427. https://doi.org/10.1016/0006-2952(89)90085-3 |
[32] | Zhang T, Mubeen S, Myung NV, et al. (2008) Recent progress in carbon nanotube-based gas sensors. Nanotechnology 19: 332001. https://doi.org/10.1088/0957-4484/19/33/332001 |
[33] | Kretschmer C, Sterner-Kock A, Siedentopf F, et al. (2011) Identification of early molecular markers for breast cancer. Mol Cancer 10: 15. https://doi.org/10.1186/1476-4598-10-15 |
[34] | ACOG committee opinion No. 727 summary: cascade testing: testing women for known hereditary genetic mutations associated with cancer. Obstet Gynecol (2018) 131: 194-195. https://doi.org/10.1097/aog.0000000000002451 |
[35] | Bamodu OA, Chung CC, Pisanic II TR, et al. (2023) Harnessing liquid biopsies: exosomes and ctDNA as minimally invasive biomarkers for precision cancer medicine. J Liq Biopsy 2: 100126. https://doi.org/10.1016/j.jlb.2023.100126 |
[36] | Rengasamy G, Priya VV (2024) NGS revolutionizing oral cancer: from genetic profiling to personalized therapy. Oral Oncol Rep 10: 100456. https://doi.org/10.1016/j.oor.2024.100456 |
[37] | Bernardi D, Gentilini MA, De Nisi M, et al. (2020) Effect of implementing digital breast tomosynthesis (DBT) instead of mammography on population screening outcomes including interval cancer rates: results of the Trento DBT pilot evaluation. Breast 50: 135-140. https://doi.org/10.1016/j.breast.2019.09.012 |
[38] | Huang T, Li G, Guo Y, et al. (2023) Recent advances in PAMAM dendrimer-based CT contrast agents for molecular imaging and theranostics of cancer. Sens Diagn 2: 1145-1157. https://doi.org/10.1039/d3sd00101f |
[39] | Hu X, Tang Y, Hu Y, et al. (2019) Gadolinium-chelated conjugated polymer-based nanotheranostics for photoacoustic/magnetic resonance/NIR-II fluorescence imaging-guided cancer photothermal therapy. Theranostics 9: 4168-4181. https://doi.org/10.7150/thno.34390 |
[40] | Zhang M, Kim HS, Jin TF, et al. (2016) Ultrasound-guided photoacoustic imaging for the selective detection of EGFR-expressing breast cancer and lymph node metastases. Biomed Opt Express 7: 1920. https://doi.org/10.1364/boe.7.001920 |
[41] | Bernardi D, Gentilini MA, De Nisi M, et al. (2020) Effect of implementing digital breast tomosynthesis (DBT) instead of mammography on population screening outcomes including interval cancer rates: results of the Trento DBT pilot evaluation. Breast 50: 135-140. https://doi.org/10.1016/j.breast.2019.09.012 |
[42] | Spangler ML, Zuley ML, Sumkin JH, et al. (2011) Detection and classification of calcifications on digital breast tomosynthesis and 2D digital mammography: a comparison. Am J Roentgenol 196: 320-324. https://doi.org/10.2214/ajr.10.4656 |
[43] | Tagliafico A, Mariscotti G, Durando M, et al. (2015) Characterisation of microcalcification clusters on 2D digital mammography (FFDM) and digital breast tomosynthesis (DBT): does DBT underestimate microcalcification clusters? Results of a multicentre study. Eur Radiol 25: 9-14. https://doi.org/10.1007/s00330-014-3402-8 |
[44] | Seo M, Chang JM, Kim SA, et al. (2016) Addition of digital breast tomosynthesis to full-field digital mammography in the diagnostic setting: additional value and cancer detectability. J Breast Cancer 19: 438. https://doi.org/10.4048/jbc.2016.19.4.438 |
[45] | Houssami N, Hofvind S, Soerensen AL, et al. (2021) Interval breast cancer rates for digital breast tomosynthesis versus digital mammography population screening: an individual participant data meta-analysis. EClinicalMedicine 34: 100804. https://doi.org/10.1016/j.eclinm.2021.100804 |
[46] | Brenner H, Stock C, Hoffmeister M, et al. (2014) Effect of screening sigmoidoscopy and screening colonoscopy on colorectal cancer incidence and mortality: systematic review and meta-analysis of randomised controlled trials and observational studies. BMJ 348: g2467. https://doi.org/10.1136/bmj.g2467 |
[47] | Atkin WS, Northover JM, Cuzick J, et al. (1993) Prevention of colorectal cancer by once-only sigmoidoscopy. Lancet 341: 736-740. https://doi.org/10.1016/0140-6736(93)90499-7 |
[48] | Lin OS, Kozarek RA, Cha JM (2014) Impact of sigmoidoscopy and colonoscopy on colorectal cancer incidence and mortality: an evidence-based review of published prospective and retrospective studies. Intest Res 12: 268. https://doi.org/10.5217/ir.2014.12.4.268 |
[49] | Diaz LA, Bardelli A (2014) Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol 32: 579-586. https://doi.org/10.1200/jco.2012.45.2011 |
[50] | Kapeleris J, Kulasinghe A, Warkiani ME, et al. (2018) The prognostic role of circulating tumor cells (CTCs) in lung cancer. Front Oncol 8: 311. https://doi.org/10.3389/fonc.2018.00311 |
[51] | Avram L, Iancu SD, Stefancu A, et al. (2020) SERS-based liquid biopsy of gastrointestinal tumors using a portable Raman device operating in a clinical environment. J Clin Med 9: 212. https://doi.org/10.3390/jcm9010212 |
[52] | De La Escosura-Muñiz A, Parolo C, Merkoçi A, et al. (2010) Immunosensing using nanoparticles. Mater Today 13: 24-34. https://doi.org/10.1016/s1369-7021(10)70125-5 |
[53] | Bick U, Trimboli RM, Athanasiou A, et al. (2020) Image-guided breast biopsy and localisation: recommendations for information to women and referring physicians by the European society of breast imaging. Insights Imaging 11: 12. https://doi.org/10.1186/s13244-019-0803-x |
[54] | Tam AL, Lim HJ, Wistuba II, et al. (2015) Image-guided biopsy in the era of personalized cancer care: proceedings from the society of interventional radiology research consensus panel. J Vasc Interv Radiol 27: 8-19. https://doi.org/10.1016/j.jvir.2015.10.019 |
[55] | Ciliberti V, Maffei E, D'Ardia A, et al. (2023) Combined fine needle aspiration cytology and core needle biopsy in the same setting: a two-years' experience. Cytopathology 35: 78-91. https://doi.org/10.1111/cyt.13318 |
[56] | Serra-García L, Eliana-Radonich J, Marti-Marti I, et al. (2022) Diagnostic accuracy of image-guided biopsies for diagnosis of metastatic melanoma in a real-life setting. Acta Dermato Venereol 102: adv00833. https://doi.org/10.2340/actadv.v102.3981 |
[57] | Mosele F, Remon J, Mateo J, et al. (2020) Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Anna Oncol 35: 588-606. https://doi.org/10.1016/j.annonc.2024.04.005 |
[58] | Kage H, Shinozaki-Ushiku A, Ishigaki K, et al. (2023) Clinical utility of todai oncopanel in the setting of approved comprehensive cancer genomic profiling tests in Japan. Cancer Sci 114: 1710-1717. https://doi.org/10.1111/cas.15717 |
[59] | Naito Y, Aburatani H, Toraji Amano T, et al. (2020) Clinical practice guidance for next-generation sequencing in cancer diagnosis and treatment (edition 2.1). Int J Clin Oncol 26: 233-283. https://doi.org/10.1007/s10147-020-01831-6 |
[60] | Glenn TC (2011) Field guide to next-generation DNA sequencers. Mol Ecol Resour 11: 759-769. https://doi.org/10.1111/j.1755-0998.2011.03024.x |
[61] | Moter A, Göbel UB (2000) Fluorescence in situ hybridization (FISH) for direct visualization of microorganisms. J Microbiol Methods 41: 85-112. https://doi.org/10.1016/s0167-7012(00)00152-4 |
[62] | Palumbo A, Avet-Loiseau H, Oliva S, et al. (2015) Revised international staging system for multiple myeloma: a report from international myeloma working group. J Clin Oncol 33: 2863-2869. https://doi.org/10.1200/jco.2015.61.2267 |
[63] | Grønborg M, Kristiansen TZ, Iwahori A, et al. (2006) Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach. Mol Cell Proteomics 5: 157-171. https://doi.org/10.1074/mcp.m500178-mcp200 |
[64] | Parkhurst MR, Yang YC, Langan RC, et al. (2011) T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther 19: 620-626. https://doi.org/10.1038/mt.2010.272 |
[65] | Morrissey JJ, Mobley J, Figenshau RS, et al. (2015) Urine aquaporin 1 and perilipin 2 differentiate renal carcinomas from other imaged renal masses and bladder and prostate cancer. Mayo Clin Proc 90: 35-42. https://doi.org/10.1016/j.mayocp.2014.10.005 |
[66] | Rissin DM, Kan CW, Campbell TG, et al. (2010) Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat Biotechnol 28: 595-599. https://doi.org/10.1038/nbt.1641 |
[67] | Wu C, Garden PM, Walt DR, et al. (2020) Ultrasensitive detection of attomolar protein concentrations by dropcast single molecule assays. J Am Chem Soc 142: 12314-12323. https://doi.org/10.1021/jacs.0c04331 |
[68] | Li J, Zhang Z, Trau M, et al. (2023) Digital platforms enabling single-molecule analysis for cancer detection. TrAC-Trend Anal Chem 171: 117502. https://doi.org/10.1016/j.trac.2023.117502 |
[69] | Parkhurst MR, Yang JC, Langan RC, et al. (2011) T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther 19: 620-626. https://doi.org/10.1038/mt.2010.272 |
[70] | Piagnerelli R, Marrelli D, Roviello G, et al. (2015) Clinical value and impact on prognosis of peri-operative CA 19-9 serum levels in stage I and II adenocarcinoma of the pancreas. Tumor Biol 37: 1959-1966. https://doi.org/10.1007/s13277-015-3986-x |
[71] | Scambia G, Gadducci A, Panici PB, et al. (1994) Combined use of CA 125 and CA 15-3 in patients with endometrial carcinoma. Gynecol Oncol 54: 292-297. https://doi.org/10.1006/gyno.1994.1213 |
[72] | Chambers MC, Maclean B, Burke R, et al. (2012) A cross-platform toolkit for mass spectrometry and proteomics. Nat Biotechnol 30: 918-920. https://doi.org/10.1038/nbt.2377 |
[73] | Petricoin EF, Ardekani AM, Hitt BA, et al. (2002) Use of pteomic ptterns in srum to ientify oarian cancer. Obstet Gynecol Surv 57: 352-353. https://doi.org/10.1097/00006254-200206000-00015 |
[74] | Lilley KS, Razzaq A, Dupree P, et al. (2002) Two-dimensional gel electrophoresis: recent advances in sample preparation, detection and quantitation. Curr Opin Chem Biol 6: 46-50. https://doi.org/10.1016/s1367-5931(01)00275-7 |
[75] | Chen G, Gharib TG, Huang CC, et al. (2002) Proteomic analysis of lung adenocarcinoma: identification of a highly expressed set of proteins in tumors. Clin Cancer Res 8: 2298-2305. |
[76] | Liang C, Shi S, Qin Y, et al. (2019) Localisation of PGK1 determines metabolic phenotype to balance metastasis and proliferation in patients with SMAD4-negative pancreatic cancer. Gut 69: 888-900. https://doi.org/10.1136/gutjnl-2018-317163 |
[77] | Edelsberg J, Weycker D, Atwood M, et al. (2018) Cost-effectiveness of an autoantibody test (earlyCDT-lung) as an aid to early diagnosis of lung cancer in patients with incidentally detected pulmonary nodules. Plos One 13: e0197826. https://doi.org/10.1371/journal.pone.0197826 |
[78] | Singh P, Pandey SK, Singh J, et al. (2015) Biomedical perspective of electrochemical nanobiosensor. Nano-Micro Lett 8: 193-203. https://doi.org/10.1007/s40820-015-0077-x |
[79] | Wu X, Zhao B, Wu P, et al. (2009) Effects of ionic liquids on enzymatic catalysis of the glucose oxidase toward the oxidation of glucose. J Phys Chem B 113: 13365-13373. https://doi.org/10.1021/jp905632k |
[80] | Wu C, Pan TM, Wu CS, et al. (2012) Label-free detection of prostate specific antigen using a silicon nanobelt field-effect transistor. Int Electrochem Sci 7: 4432-4442. https://doi.org/10.1016/s1452-3981(23)19551-4 |
[81] | Wu D, Yu Y, Jin D, et al. (2020) Dual-aptamer modified graphene field-effect transistor nanosensor for label-free and specific detection of hepatocellular carcinoma-derived microvesicles. Anal Chem 92: 4006-4015. https://doi.org/10.1021/acs.analchem.9b05531 |
[82] | Machado RF, Laskowski D, Deffenderfer O, et al. (2005) Detection of lung cancer by sensor array analyses of exhaled breath. Am J Respir Crit Care Med 171: 1286-1291. https://doi.org/10.1164/rccm.200409-1184oc |
[83] | Yonzon C, Stuart DA, Xiaoyu Zhang XY, et al. (2005) Towards advanced chemical and biological nanosensors—an overview. Talanta 67: 438-448. https://doi.org/10.1016/j.talanta.2005.06.039 |
[84] | Khan I, Saeed K, Khan I, et al. (2017) Nanoparticles: properties, applications and toxicities. Arab J Chem 12: 908-931. https://doi.org/10.1016/j.arabjc.2017.05.011 |
[85] | Patra JK, Das G, Fraceto LF, et al. (2018) Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol 16: 71. https://doi.org/10.1186/s12951-018-0392-8 |
[86] | Ta HT, Arndt N, Wu Y, et al. (2018) Activatable magnetic resonance nanosensor as a potential imaging agent for detecting and discriminating thrombosis. Nanoscale 10: 15103-15115. https://doi.org/10.1039/c8nr05095c |
[87] | Sztandera K, Gorzkiewicz M, Klajnert-Maculewicz B, et al. (2018) Gold nanoparticles in cancer Treatment. Mol Pharmaceutics 16: 1-23. https://doi.org/10.1021/acs.molpharmaceut.8b00810 |
[88] | Kobeleva ES, Uvarov MN, Kravetset NV, et al. (2023) Fluorinated carbon nanotubes as nonvolatile additive to the active layer of polymer/fullerene solar cells. Fuller Nanotub Car N 31: 464-473. https://doi.org/10.1080/1536383x.2023.2179618 |
[89] | Raverot V, Richet C, Morel Y, et al. (2016) Establishment of revised diagnostic cut-offs for adrenal laboratory investigation using the new roche diagnostics elecsys® cortisol II assay. Ann Endocrinol 77: 620-622. https://doi.org/10.1016/j.ando.2016.05.002 |
[90] | Brennan JD, Brown RS, McClintock CP, et al. (1990) Fluorescence transduction of an enzyme-substrate reaction by modulation of lipid membrane structure. Anal Chim Acta 237: 253-263. https://doi.org/10.1016/s0003-2670(00)83927-6 |
[91] | Lu M, Zhu H, Hong L, et al. (2020) Wavelength-tunable optical fiber localized surface plasmon resonance biosensor via a diblock copolymer-templated nanorod monolayer. ACS Appl Mater Interfaces 12: 50929-50940. https://doi.org/10.1021/acsami.0c09711 |
[92] | Liu C, Zeng X, An ZJ, et al. (2018) Sensitive detection of exosomal proteins via a compact surface plasmon resonance biosensor for cancer diagnosis. ACS Sens 3: 1471-1479. https://doi.org/10.1021/acssensors.8b00230 |
[93] | Xiao Y, Bi MY, Guo HY, et al. (2022) Multi-omics approaches for biomarker discovery in early ovarian cancer diagnosis. EBioMedicine 79: 104001. https://doi.org/10.1016/j.ebiom.2022.104001 |
[94] | Hirsch M, Duffy J, Davis CJ, et al. (2016) Diagnostic accuracy of cancer antigen 125 for endometriosis: a systematic review and meta-analysis. BJOG: Int J Obstet Gynaecol 123: 1761-1768. https://doi.org/10.1111/1471-0528.14055 |
[95] | Büyüktiryaki S, Say R, Denizli A, et al. (2017) Phosphoserine imprinted nanosensor for detection of cancer Antigen 125. Talanta 167: 172-180. https://doi.org/10.1016/j.talanta.2017.01.093 |
[96] | Van Gorp T, Cadron I, Despierre E, et al. (2011) HE4 and CA125 as a diagnostic test in ovarian cancer: prospective validation of the risk of ovarian malignancy algorithm. Brit J Cancer 104: 863-870. https://doi.org/10.1038/sj.bjc.6606092 |
[97] | Kabawat SE, Bast RC, Bhan AK, et al. (1983) Tissue distribution of a coelomic- epithelium-related antigen recognized by the monoclonal antibody OC125. Int J Gynecol Pathol 2: 275-285. https://doi.org/10.1097/00004347-198303000-00005 |
[98] | Luk'yanchuk B, Zheludev NI, Maier SA, et al. (2010) The Fano resonance in plasmonic nanostructures and metamaterials. Nat Mater 9: 707-715. https://doi.org/10.1038/nmat2810 |
[99] | Eckschlager T, Plch J, Stiborova M, et al. (2017) Histone deacetylase inhibitors as anticancer drugs. Int Journal Mol Sci 18: 1414. https://doi.org/10.3390/ijms18071414 |
[100] | Park JH, Byun JY, Mun HY, et al. (2014) A regeneratable, label-free, localized surface plasmon resonance (LSPR) aptasensor for the detection of ochratoxin A. Biosens Bioelectron 59: 321-327. https://doi.org/10.1016/j.bios.2014.03.059 |
[101] | Kaur A, Pandey K, Kaur R, et al. (2022) Nanocomposites of carbon quantum dots and graphene quantum dots: environmental applications as sensors. Chemosensors 10: 367. https://doi.org/10.3390/chemosensors10090367 |
[102] | Ng E, Yao CY, Shultz TO, et al. (2018) Magneto-nanosensor smartphone platform for the detection of HIV and leukocytosis at point-of-care. Nanomed Nanotechnol Biol Med 16: 10-19. https://doi.org/10.1016/j.nano.2018.11.007 |
[103] | Fox J, Velaiutham S, Yang N, et al. (2024) Abstract PS05-03: magneto-nanosensor® HER2, a molecularly targeted magnetic resonance imaging agent for the detection of axillary nodal metastasis in subjects with human epidermal growth factor receptor 2 positive (HER2+) breast cancer. Cancer Res 84: PS05-03. https://doi.org/10.1158/1538-7445.sabcs23-ps05-03 |
[104] | Amiri M, Salavati-Niasari M, Akbari A, et al. (2019) Magnetic nanocarriers: evolution of spinel ferrites for medical applications. Adv Colloid Interfac 265: 29-44. https://doi.org/10.1016/j.cis.2019.01.003 |
[105] | 01. ExoSense: a microprobe-based method for single-step isolation and genetic of exosomes, Poster Presentation (2021). Available from: https://digitalcommons.latech.edu/undergraduate-research-symposium/2021/poster-presentations/9/. |
[106] | Kumar A, Kim S, Nam JM, et al. (2016) Plasmonically engineered nanoprobes for biomedical applications. J Am Chem Soci 138: 14509-14525. https://doi.org/10.1021/jacs.6b09451 |
[107] | Yan LX, Huang XF, Shao Q, et al. (2008) MicroRNA miR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA 14: 2348-2360. https://doi.org/10.1261/rna.1034808 |
[108] | Xu H, Liao C, Zuo P, et al. (2018) Magnetic-based microfluidic device for on-chip isolation and detection of tumor-derived exosomes. Anal Chem 90: 13451-13458. https://doi.org/10.1021/acs.analchem.8b03272 |
[109] | Liu L, Li N, Huang ZM, et al. (2020) Gold nanoflares with computing function as smart diagnostic automata for multi-miRNA patterns in living cells. Anal Chem 92: 10925-10929. https://doi.org/10.1021/acs.analchem.0c02325 |
[110] | Chen MX, Duan RL, Xu SJ, et al. (2021) Photoactivated DNA walker based on DNA nanoflares for signal-amplified microRNA imaging in single living cells. Anal Chem 93: 16264-16272. https://doi.org/10.1021/acs.analchem.1c04505 |
[111] | Premachandran S, Dhinakaran AK, Das S, et al. (2024) Detection of lung cancer metastasis from blood using L-MISC nanosensor: targeting circulating metastatic cues for improved diagnosis. Biosens Bioelectron 243: 115782. https://doi.org/10.1016/j.bios.2023.115782 |
[112] | Makarov SV, Tsypkin AN, Voytova TA, et al. (2016) Self-adjusted all-dielectric metasurfaces for deep ultraviolet femtosecond pulse generation. Nanoscale 8: 17809-17814. https://doi.org/10.1039/c6nr04860a |
[113] | Vijayakumar SC, Venkatakrishnan K, Tan B, et al. (2017) SERS active nanobiosensor functionalized by self-assembled 3D nickel nanonetworks for glutathione detection. ACS Appl Mater Interfaces 9: 5077-5091. https://doi.org/10.1021/acsami.6b13576 |
[114] | Failli M, Demir S, Río-Álvarez ÁD, et al. (2023) Computational drug prediction in hepatoblastoma by integrating pan-cancer transcriptomics with pharmacological response. Hepatology 80: 55-68. https://doi.org/10.1097/hep.0000000000000601 |
[115] | Rotari A, Failli M, Cairo S, et al. (2023) Understanding nfe2l2/keap1-mediated drug resistance in hepatoblastoma. Klinische Pädiatrie . https://doi.org/10.1055/s-0043-1768538 |
[116] | Felfoul O, Mohammadi M, Taherkhani S, et al. (2016) Magneto-aerotactic bacteria deliver drug-containing nanoliposomes to tumour hypoxic regions. Nat Nanotechnol 11: 941-947. https://doi.org/10.1038/nnano.2016.137 |
[117] | Chen X, Zou LQ, Niu J, et al. (2015) The stability, sustained release and cellular antioxidant activity of curcumin nanoliposomes. Molecules 20: 14293-14311. https://doi.org/10.3390/molecules200814293 |
[118] | Nolan KA, Doncaster JR, Dunstan MS, et al. (2009) Synthesis and biological evaluation of coumarin-based inhibitors of NAD(P)H: quinone oxidoreductase-1 (NQO1). J Med Chem 52: 7142-7156. https://doi.org/10.1021/jm9011609 |
[119] | Li ZP, Feng QC, Hou JT, et al. (2024) NQO-1 activatable NIR photosensitizer for visualization and selective killing of breast cancer cells. Bioorg Chem 143: 107021. https://doi.org/10.1016/j.bioorg.2023.107021 |
[120] | Miller KD, Siegel RL, Lin CC, et al. (2016) Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin 66: 271-289. https://doi.org/10.3322/caac.21349 |
[121] | Ford D, Easton DF, Stratton M, et al. (1998) Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. Am J Human Genet 62: 676-689. https://doi.org/10.1086/301749 |
[122] | Khalid A, Mehmood A, Alabrah A, et al. (2023) Breast cancer detection and prevention using machine learning. Diagnostics 13: 3113. https://doi.org/10.3390/diagnostics13193113 |
[123] | Kharel N, Alsadoon A, Prasad PWC, et al. (2017) Early diagnosis of breast cancer using contrast limited adaptive histogram equalization (CLAHE) and morphology methods. Proceedings of the 2017 8th International Conference on information and communication systems : 120. https://doi.org/10.1109/IACS.2017.7921957 |
[124] | Nguyen HQ, Nguyen VD, Nguyen HV, et al. (2020) Quantification of colorimetric isothermal amplification on the smartphone and its open-source app for point-of-care pathogen detection. Sci Rep 10: 15123. https://doi.org/10.1038/s41598-020-72095-3 |
[125] | Bukhari M, Yasmin S, Sammad S, et al. (2022) A deep learning framework for leukemia cancer detection in microscopic blood samples using squeeze and excitation learning. Math Probl Eng 2022: 1-18. https://doi.org/10.1155/2022/2801227 |
[126] | Dong Y, Shi O, Zeng QX, et al. (2020) Leukemia incidence trends at the global, regional, and national level between 1990 and 2017. Exp Hematol Oncol 9: 14. https://doi.org/10.1186/s40164-020-00170-6 |
[127] | Nasir MU, Khan MF, Khan MA, et al. (2023) Hematologic cancer detection using white blood cancerous cells empowered with transfer learning and image processing. J Healthc Eng 2023: 1-20. https://doi.org/10.1155/2023/1406545 |
[128] | Sridhar B, Sridhar S, Nanchariah V, et al. (2021) Cluster medical image segmentation using morphological adaptive bilateral filter based BSA algorithm. 2021 5th International Conference on Trends in Electronics and Informatics : 726-731. https://doi.org/10.1109/icoei51242.2021.9452816 |
[129] | Bibi N, Sikandar M, Din IU, et al. (2020b) IoMT-based automated detection and classification of leukemia using deep learning. J Healthc Eng 2020: 1-12. https://doi.org/10.1155/2020/6648574 |
[130] | Das SK, Islam KS, Neha TA, et al. (2021) Towards the segmentation and classification of white blood cell cancer using hybrid mask-recurrent neural network and transfer learning. Contrast Media Mol Imag 2021: 1-12. https://doi.org/10.1155/2021/4954854 |
[131] | Bukhari M, Yasmin S, Sammad S, et al. (2022) A deep learning framework for leukemia cancer detection in microscopic blood samples using squeeze and excitation learning. Math Probl Eng 2022: 1-18. https://doi.org/10.1155/2022/2801227 |
[132] | Karimi-Maleh H, Alizadeh M, Orooji Y, et al. (2021) Guanine-based DNA biosensor amplified with Pt/SWCNTs nanocomposite as analytical tool for nanomolar determination of daunorubicin as an anticancer drug: a docking/experimental investigation. Ind Eng Chem Res 60: 816-823. https://doi.org/10.1021/acs.iecr.0c04698 |
[133] | Shibata A, Goto Y, Saito H, et al. (2006) Comparison of SYBR green I and SYBR gold stains for enumerating bacteria and viruses by epifluorescence microscopy. Aquat Microb Ecol 43: 223-231. https://doi.org/10.3354/ame043223 |
[134] | Chen XX, Zeng KX, Xu M, et al. (2018) SP1-induced lncRNA-ZFAS1 contributes to colorectal cancer progression via the miR-150-5p/VEGFA axis. Cell Death Dis 9: 982. https://doi.org/10.1038/s41419-018-0962-6 |
[135] | Toiyama Y, Takahashi M, Hur K, et al. (2013) Serum miR-21 as a diagnostic and prognostic biomarker in colorectal cancer. J Natl Cancer I 105: 849-859. https://doi.org/10.1093/jnci/djt101 |
[136] | Chen KZ, Zhao H, Shi YB, et al. (2019) Perioperative dynamic changes in circulating tumor DNA in patients with lung cancer (DYNAMIC). Clin Cancer Res 25: 7058-7067. https://doi.org/10.1158/1078-0432.ccr-19-1213 |
[137] | Ratajczak K, Grel H, Olejnik P, et al. (2023) Current progress, strategy, and prospects of PD-1/PDL-1 immune checkpoint biosensing platforms for cancer diagnostics, therapy monitoring, and drug screening. Biosens Bioelectron 240: 115644. https://doi.org/10.1016/j.bios.2023.115644 |