Open access peer-reviewed chapter

Methods of Protein Detection in Cancer for Diagnosis, Prognosis and Therapy

Written By

Shenbagamoorthy Sundarraj, Gopalan Rajagopal, Balaji Sundaramahalingam, Madasamy Sundar and Ramar Thangam

Submitted: 10 July 2021 Reviewed: 04 October 2021 Published: 23 June 2022

DOI: 10.5772/intechopen.101050

From the Edited Volume

Protein Detection

Edited by Yusuf Tutar and Lütfi Tutar

Chapter metrics overview

321 Chapter Downloads

View Full Metrics

Abstract

Emerging proteomic technologies offer new insight in the study of malignant tumor to identify protein biomarkers for early detection, stratification, prediction and monitoring of treatment, as well as to detect target molecules for therapy. The tumor protein biomarker is responsible for the regulation of the cell cycle to promote cell proliferation and resistance to cell death. Important technologies include ELISA, immunohistochemistry, flow cytometry, western blot, mass spectrometry, protein microarray, and microfluidics for the study of screening, protein profiling, identification, qualitative and quantitative analysis of differential expressed oncoproteins relative to cancer tissues, counterparts at different stages of the disease from preneoplasia to neoplasia. It can also provide a detailed description of identifying tissue-specific protein biomarkers and to analysis the modification of protein activity in cancer conditions. In this chapter, we discuss current and emerging protein assays for improving cancer diagnosis, including trends toward advances in assay miniaturization, improve sensitivity and specificity, time and cost-effective, and accuracy in detection and measurement of protein activity. However, information from these protein diagnostic technologies should be integrated to obtain the optimal information required for the clinical management of a patient.

Keywords

  • cancer
  • protein biomarker
  • ELISA
  • protein microarray
  • mass spectrometry

1. Introduction

Cancer is the leading cause of death global population. As stated by the National Cancer Institute annual report revealed that there were 18.1 million new cases and 9.5 million cancer-related deaths globally in 2018. By 2040, the number of new cancer cases per year is anticipated to arise around 29.5 million and the number of cancer-related deaths to 16.4 million [1]. Cancer mortality can be reduced if cases are detected earlier and treated systematically and can result in a greater probability of survival rate and less morbidity [2]. Cancer diagnosis and prognosis have advanced dramatically during the last decades. Achieving this goal will necessitate not only improved therapies, but also enhanced methods for evaluating an individual’s risk of developing cancer, detecting cancers at an early stage when they can be treated more effectively, distinguishing aggressive from non-aggressive cancers, and monitoring recurrence and response to therapy.

Diagnostic imaging technologies can be used to detect people with cancer, these tests can be physically invasive, time-consuming and expensive to screen large groups of people who are asymptomatic and can cause unnecessary stress and worry. Furthermore, diagnostic imaging technologies frequently overlook minor lesions, resulting in the disease not being detected until it has progressed to the point when treatment intervention is less effective. However, insufficient diagnostics prohibit the detection of certain types of cancer until the advanced stage. For instance endoscopy with biopsy is the distinctive screening method for esophageal cancer and is generally performed after symptoms appear [3]. Other screens may be providing high levels of false positives or negatives. Hepatocellular carcinoma is generally detected by ultrasound, but this technique is subjected to operator mistake and often cannot distinguish between malignant and benign nodules [4]. Although mammography is the standard screening techniques for breast cancer, 20% of cases go undetected with this screening and specificity is 25%, leading to a large number of false positives [5].

Improving strategies for screening asymptomatic individuals for early-stage malignancies is a particularly difficult challenge. Overcome these challenges in the recent years, there has been a surge in interest in molecular markers as a cancer diagnosis, prognosis and therapeutic response [6]. The cancer antigen 15–3 (CA 15–3) act as a potential biomarker is used to screening and monitoring breast cancer [7]. The prostate-specific antigen (PSA) is a widely mentioned marker that is used to test male patients for prostate cancer [8]. The analysis of overexpression of human epidermal growth factor receptor type 2 (Her2) and estrogen receptor levels in breast cancer patients [9, 10].

Specifically, some modern molecule-oriented techniques used protein as a biomarker for monitoring of cancer progression and early tumor detection. Furthermore, tumor biomarker protein assays are suitable method for holding important clinical diagnostic tests in future because gene level studies may not correlation for the cancer alteration [11, 12]. Protein biomarkers played significant roles in accurate early diagnosis, therapy and prognosis in colorectal cancer [13].

Serum protein biomarkers are well developed tools for cancer diagnosis [14]. As proof, prostate-specific antigen (PSA), cancer antigen 125 (CA-125) and carcinoembryonic antigen (CEA) is extensively used for the diagnosis and management of various types of cancer, namely prostate, ovarian and gastrointestinal cancers [15]. The clinical sensitivity of a biomarker can simply be defined as the proportion of people with a confirmed disease who test positive for the biomarker assay whereas specificity refers to the proportion of healthy individuals who test negative for the biomarker assay [16]. Noninvasive assays, such as those using blood, stool, urine, or saliva are preferred because they cause less pain to the patient, have higher abidance rates and may be taken frequently for monitoring the treatment response [17]. Measurement of serum proteins ensures distinguishes between various types of malignancies from benign and thus leading imaging analysis, endoscopic examination and other diagnostic procedures and monitoring of the efficacy of the treatment.

The main objective of this chapter is to provide a new insight of emerging technologies based on protein detection for cancer diagnostic and prognostic. Proteins analytical techniques are especially suitable for the diagnosis of cancer are described here briefly along with their recent development. We also provide a brief description of techniques currently used to identify the protein activity in post-translational modification have been linked to cancer diagnosis and cancer progression.

Advertisement

2. Methods of proteins detection in cancer

The developments of proteomic patterns have emerged as effective method for diagnostics in the field of cancer proteomics in that it is represents new way for cancer detection and also clinically feasible. The enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC) and flow cytometry system represent the most reliable, sensitive and widely available protein-based testing platform in the clinic for the diagnosis, prognosis and treatment monitoring of cancer [18, 19]. Other protein analyses techniques such as mass spectrometry, Protein array and Microfluidics are currently at the laboratory level setup extensively used for cancer research purpose but these techniques are being developed for clinical applications (Figure 1 and Table 1).

Figure 1.

Schematic illustration of the different proteomic techniques for detection, identification, screening, protein profiling and modification of protein in various cancers.

2.1 Enzyme-linked immunosorbent assay

The enzyme-linked immunosorbent test (ELISA) has been widely utilised in regular clinical diagnostics and is still considered the gold standard for detecting cancer protein biomarkers in physiological samples [60, 61, 62]. Despite advances in developing cost-effective and label-free novel ELISA-based methods for future use in point-of-care cancer diagnostics, prognostics and therapy monitoring offers promise for improving the early detection of breast cancer (Figure 2). In conventional ELISA techniques, colorimetric or fluorescent readout signals are utilised to show FDA-approved protein biomarkers currently used in clinical practice (Table 2).

Figure 2.

Flowchart of nanocomposite used as a biosensing substrate and detection in breast cancer using nanoparticle coated arrays and ELISA based electrochemical immunosensor. First, a drop of blood from breast cancer (BC) patients is subjected to an nanoparticle fabricated array for a high-throughput screening of cancer biomarkers in breast cancer. Second, promising cancer biomarker candidates are selected from the array screening and validated in a large cohort of patients using ELISA, which can be used for early diagnosis, disease stratification, prediction of disease progression, or monitoring of drug responses. Finally, according to the function of nanoparticle coated array biomarker panel, biosensors could be designed and fabricated for clinical use in breast cancer.

Cancer Protein BiomarkerCancer typeSpecimenMethodologyReference
MidkineHepatocellular carcinomaSerumELISA[20]
Alpha-fetoprotein (AFP)Liver cancerSerumElectrochemical immunosensor, ELISA[21]
MidkineCancer cellsSerumElectromagnetically induced transparency (EIT)[22]
EGFR, CA125 and HE4Ovarian cancerSerumCIMA, ELISA[23]
Vim3, Mxi-2Renal carcinomaUrineELISA[24]
Cyclin D1Breast cancerTissueWB, IHC[25]
BAP31Ovarian cancerTissueIHC[26]
MDH2NSCLCUrineELISA[27]
Aminoacylase-1 (ACY-1)Rectal cancerTissueIHC[28]
MMP-1Oral cancerSaliva, UrineElectrochemical immunosensor[29]
Tumor-associated antigens (TAAbs)Lung cancerSerumProtein array, ELISA[30]
HPV E6 and E7 oncoproteinsCervical cancerSerumElectrospun PCL (ePCL) Fiber coated ELISA[31]
RAS Q61REpithelial-Myoepithelial CarcinomaTissueIHC[32]
Carcinoembryonic antigen, CA 15–3Breast cancerSerumElectrochemical aptasensor Redox probes labeled aptamers[33]
Integrin alpha VProstate cancerUrineELISA[34]
B-cell activating factor (BAFF)MelanomaSerumELISA, IHC[35]
QSOX1Colorectal CancerSerumELISA[36]
LRG1, TTR, CA 19–9Pancreatic cancerPlasmaELISA[37]
Carcinoembryonic antigenSerumMALDI-TOF MS[38]
Tumor-associated antigens (TAAbs)Gastric CancerSerumProteomic chips, ELISA[39]
PD-L1, HIF-1αBreast cancerSerumElectrochemical immunosensor[40]
FGL1SerumNanobody-based ELISA[41]
p16INK4aCervical cancercervical swabsELISA, IHC, WB[42]
FBLN1, ANT3Cervical cancerSerum, Plasma, tissueELISA, LC MS/MS[43]
ATX and LPAPancreatic cancerSerumELISA[44]
Tumor EndothelialColorectal cancerTissue, SerumIHC, ELISA[45]
Marker 8 (ANTXR1) SAS1BThyroid cancerSerumELISA[46]
Annexin A2Ovarian CancerPlasmaELISA[47]
DKK3Ovarian CancerSerumELISA[48]
Alpha-fetoproteinLiver cancerHuman cord serumELISA[29]
Desmoglein 3 (DSG3)Head and neck squamous cell carcinomaCell lysateMicrofluidic immunoarray[49]
FGFR3Bladder cancerRecombinant FGFR3 proteinElectrochemical impedance spectroscopy, ELISA[50]
BC2L-C lectinBreast cancerTissueIHC[51]
MUCIN-16/CA125Ovarian CancerPlasmaMS[52]
Matrix metalloproteinase-1Oral cancerSalivaMS[53]
HER2 and Ki67Breast cancerTissueWestern blot[9]
Perilipin-2Renal cancerUrineELISA, Bioplasmonic paper–based assay[54]
apo A-IV and LRG1Oral cancerPlasmaELISA, WB, LC MS/MS[55]
NANOGOvarian cancerTissueIHC, WB[56]
MUCIN-16/CA125Ovarian CancerPlasmaProximity extension assay[15]
ERBreast cancerTissueIHC[10]
rab31 and mucin-1 (CA15–3)Breast cancerTissueELISA, chemiluminescence immunoassay[57]
HER2Breast cancerTissueIHC-QD[58]
PSA and IL-6Prostate cancerSerumMicrofluidic electrochemical immunoassay[59]

Table 1.

List of cancer protein biomarkers and their detection methods in the current research practice.

BiomarkerClinical useCancer typeSpecimenMethodology
TP53Diagnosis and monitoring of diseaseLymphocytic Leukemia/lymphomaSerumImmunoassay
RETDiagnosisNSCLCSerum, plasmaImmunoassay
MS4A1(CD20 antigen)Diagnosis and monitoring of diseaseNon–Hodgkin’s LymphomaSerumImmunoassay
CCDC6-RET, KIF5B-RET, RETDiagnosisNSCLCSerum, plasmaImmunoassay
PPP2R2ADiscriminating cancer from benign DiseaseProstateSerumImmunoassay
IGHDiagnosis and monitoring of diseaseLymphocytic Leukemia/ lymphomaSerumImmunoassay
BRCAPrediction of malignancyOvarianSerumImmunoassay
MS4A1 (CD20 antigen)Prognosis, response to therapyLymphocytic LeukemiaSerumImmunoassay
NPM1Prognosis, response to therapyAcute Myeloid LeukemiaSerumImmunoassay
SSTRDiagnosis and monitoring of diseaseGastroenteropancreatic neuroendocrine tumorsSerum, plasmaImmunoassay
HLA-AAid in differential diagnosisMelanomaTissueIHC
FIP1L1-PDGFRADiagnosis and monitoring of diseaseEosinophilic LeukemiaSerumImmunoassay
MYD88Prediction of malignancyMacroglobulinemiaSerumImmunoassay
FLT3Diagnosis and monitoring of diseaseAcute Myeloid LeukemiaSerumImmunoassay
CD33Diagnosis and monitoring of diseaseAcute Myeloid LeukemiaSerumImmunoassay
NECTIN4Diagnosis and monitoring of diseaseMetastatic Urothelial CancerUrineLateral flow immunoassay
IDH1, IDH2Prognosis, response to therapyAcute Myeloid LeukemiaSerumImmunoassay
MYCNDetection of tumorsNeuroblastomaSerumImmunoassay
IL2RA (CD25 antigen)Prostate cancer diagnosisT-cell lymphomaSerumImmunoassay
ROS1Prognosis, response to therapyNSCLCSerumImmunoassay
RASProstate cancer diagnosisColorectal CancerFecesLateral flow immunoassay
METPrognosis, response to therapyNSCLCSerumImmunoassay
TNFRSF8 (CD30)progression of diseaseT-cell lymphomaSerumImmunoassay
CD274 (PD-L1)Aid in differential diagnosisMerkel Cell Carcinoma NSCLCTissueIHC
PDGFRADetection of tumorsGastrointestinal Stromal TumorFFPE tissueIHC
BRAFAid in differential diagnosisMelanomaTissueIHC
PML-RARAMorphologic diagnosis of APLAcute Promyelocytic leukemiaSerumImmunoassay
PIK3CAPrognosis, response to therapyBreastFFPE tissueIHC
Anaplastic lymphoma kinase (ALK)Prognosis, response to therapy for AL positive metastatic patientsNSCLCSerumImmunoassay
EGFRPrognosis, response to therapy for EGFR mutation-positive patientsNSCLCSerumImmunoassay
Pro2PSADiscriminating cancer from benign DiseaseProstateSerumImmunoassay
ROMA (HE4 + CA-125)Prediction of malignancyOvarianSerumImmunoassay
OVA1 (multiple proteins)Prediction of malignancyOvarianSerumImmunoassay
HE4Monitoring recurrence or progression of diseaseOvarianSerumImmunoassay
Fibrin/fibrinogen degradation product (DR-70)Monitoring progression of diseaseColorectalSerumImmunoassay
AFP-L3%Risk assessment for development of diseaseHepatocellularSerumHPLC, microfluidic electrophoresis
Circulating Tumor Cells (EpCAM, CD45, cytokeratins 8, 18+, 19+)Prediction of cancer progression and survivalBreastWhole bloodImmunomagnetic fluorescence
p63 proteinAid in differential diagnosisProstateFFPE tissueIHC
c-KitDetection of tumors, aid in selection of patientsGastrointestinal stromal tumorsFFPE tissueIHC
CA19–9Monitoring disease statusPancreaticSerum, plasmaImmunoassay
Estrogen receptor (ER)Prognosis, response to therapyBreastFFPE tissueIHC
Progesterone receptor (PR)Prognosis, response to therapyBreastFFPE tissueIHC
HER-2/neuAssessment for therapyBreastFFPE tissueIHC
CA-125Monitoring disease progression, response to therapyOvarianSerum, plasmaImmunoassay
CA15–3Monitoring disease response to therapyBreastSerum, plasmaImmunoassay
CA27.29Monitoring disease response to therapyBreastSerumImmunoassay
Free PSADiscriminating cancer from benign DiseaseProstateSerumImmunoassay
ThyroglobulinAid in monitoringThyroidSerum, plasmaImmunoassay
Nuclear Mitotic Apparatus proteinDiagnosis and monitoring of diseaseBladderUrineLateral flow immunoassay
(NuMA, NMP22)(professional and home use)
Alpha-fetoprotein (AFP)bManagement of cancerTesticularSerum, plasma amniotic fluidbImmunoassay
Total PSAProstate cancer diagnosis and MonitoringProstateSerumImmunoassay
Carcino-embryonic antigenAid in management and prognosisNot specifiedSerum, plasmaImmunoassay
Human hemoglobin (fecal occult blood)Detection of fecal occult blood (home use)ColorectalFecesLateral flow immunoassay

Table 2.

List of FDA-approved protein tumor markers currently used in clinical practice.

Stevens et al. generated a novel ELISA-based technique called plasmonic ELISA that uses gold nanoparticles as the probe to detect PSA in prostate cancer diagnosis [63, 64]. Shim et al. described a microfluidic droplet-based extremely flexible and sensitive diagnostic device for counting individual analyse molecules and identifying a biomarker for prostate cancer in buffer with a detection limit of 46 fM [65]. Zhang et al. described the estimation of PSA in a sandwich-type electrochemical ELISA with fabrication of PtNP-ferrocenedicarboxylic acid based infinite coordination polymer (ICP) in combination with polyamidoamine dendrimers modified sensor electrode [66]. Xu et al. described the simultaneous detection of triple cancer biomarkers, namely PSA, CEA and AFP using newly developed carbon and gold (CGN) nanocomposite-based immunoprobes [67].

We mainly focus on the recent advances made by a various group in improvement strategies for electrochemical ELISA-based immunosensors for improving access to diagnostics, increased detection sensitivity and specificity, magnification of the signal, ease of handling, potential for automation and combination with miniaturized analytical systems, low cost and comparative simplicity for mass production. The development of generation and characterization of double nanobody-based sandwich ELISA for the detection of FGL1 in cancer patient serum [41]. San Martin et al., developed the “gold standard” ELISA-based electrochemical immunosensor for the single determination of both proteins PD-L1 and HIF-1α in terms of assay time, compatibility making their use suitable for untrained users at the point of attention [40]. The fabrication of sandwich ELISA type electrochemical aptasensor is developed for the instantaneous determination of two important biomarkers arcinoembryonic antigen (CEA) and cancer antigen 15–3 (CA 15–3) in breast cancer. CEA and CA 15–3 aptamers linked to gold nanoparticles/redox probe/graphene nanocomposite were used as biosensing probes for signal amplification and to enhance the sensitivity of the immunoassay [33]. Poly(ε-caprolactone) electrospun scaffolds (ePCL) are used to arrange for a microstructured substrate with a high surface-to-volume ratio, capable of binding E7 oncoproteins when used for enzyme-linked immunosorbent assay (ELISA) tests [31]. Interestingly, the ultrasensitive detection of cancer biomarker matrix metalloproteinase-1 in urine, saliva, bovine serum, and cell culture media of oral and brain cancers using label-free electrochemical immunosensor based on gold nanoparticle/ polyethyleneimine/reduced graphene oxide nanocomposites [29]. Li et al., 2021 constructed a simple label-free electrochemical immunosensor based on worm-like platinum with a sandwich-like structure [21]. The fabricated electrochemical immunosensor showed a wide linear range, enhanced detection limit, good selectivity and stability for the determination of alpha-fetoprotein.

Applications of Enzyme-linked Immunosorbent Assay in cancer research

  • used to help diagnose certain diseases such as cancer

  • For determination and quantification of tumor markers

  • Detecting and measuring cell cycle checkpoint markers could be a promising pathway to better develop treatment strategies

2.2 Immunohistochemistry

Immunohistochemistry (IHC) is a fundamental method used for clinical decision making of diagnosis and prognosis of various cancers, such as breast [68], prostate [69], lung [70, 71]. It enables to find out the analysis of biomarker expression and tissue localization in cancer. Immunohistochemical techniques play critical roles a diagnosis and screening tools for distinguishing between malignant and benign with the help of biomarkers expression in lung cancer [72]. In recent years, the advancement of microfluidic-based immunohistochemistry represents clinically validated approaches to the standard chromogenic staining for rapid, accurate, and automated breast cancer diagnosis [73]. The automated chromogenic multiplexed immunohistochemistry assay approach provides an exclusive sample-sparing tool to characterize limited tissue samples in lung cancer and making it an emerging method in the clinical analysis for therapeutic decision making of advanced NSCLC, provided that validation in a larger population is performed. This implies limiting the number of tissue slides despite the existence of specific and sensitive biomarkers (ALK, ROS1, BRAF V600E, PD-L1) and the obligation to distinguish lung adenocarcinoma from squamous cell carcinoma [74].

Applications of Immunohistochemistry in cancer research

  • To predict the prognosis of tumors by identification of enzymes, tumor-specific antigens, oncogenes, tumor suppressor genes, and tumor cell proliferation markers.

  • To diagnosis of tumors of uncertain origin, primary as well as metastatic from unknown primary tumor.

  • To predict therapeutic response in two important tumors, i.e. carcinoma of breast and prostate.

  • Used to help tell the difference between different types of cancer

  • To determine the function of specific gene products in fundamental biological processes such as development and apoptosis.

2.3 Flow cytometry

Flow cytometry is a versatile technique with applications in a variety of fields, including immunology, virology, molecular biology, cancer biology, and infectious disease surveillance. Flow cytometry is a technique for swiftly analysing single cells that are suspended in a buffered salt solution and flow through one or more lasers. Each cell is subjected to a visible light scattering analysis as well as one or more fluorescence parameters. Visible light scatter is assessed in two directions: forward (Forward Scatter or FSC), which shows the cell’s relative size, and at 90° (Side Scatter or SSC), which reveals the cell’s internal complexity or granularity. Fluorescence measurements are performed on samples that have been transfected and expressed with fluorescent proteins (for example, Green Fluorescent Protein, GFP), stained with fluorescent dyes (for example, Propidium Iodide, DNA), or stained with fluorescently attached antibodies (e.g., CD3 FITC). It enables simultaneous characterization of mixed populations of cells from blood and bone marrow as well as dissociable solid tissues such as lymph nodes, spleen, mucosal tissues, and solid malignancies.

The availability of new reagents has resulted in an explosion in the number of parameters utilised in flow cytometry investigations during the last several years. The number of fluorochromes used to conjugate monoclonal antibodies has increased dramatically, including tandem dyes and polymer dyes. Additionally, the number of fluorescent proteins accessible for transfection beyond GFP has increased, including mCherry, mBanana, mOrange, and mNeptune. These advancements in fluorochromes and technology have enabled tests with over 30 parameters to be performed. Data analysis is the final step of a flow cytometry experiment. The two-parameter histogram (dot plot) gating and analysis method is still widely utilised. However, as the number of factors and complexity of experiments expand, newer cluster data analysis techniques such as PCA, SPADE, and tSNE are being used. These enhanced data mining techniques enable the extraction of relevant information from the high-dimensional data generated by flow cytometry.

Since Mack Fulwyler initially invented the present kind of flow cytometers in 1965 [75], flow cytometry has now been used in quite a broad range of clinical areas for assessing protein expression in cancer cells [76, 77]. It is commonly used to diagnose of acute lymphoblastic leukemia [78]. Flow cytometry is a rapid and sensitive diagnostic method that makes it possible to characterize more satisfactorily the heterogeneous group of acute lymphoblastic leukemias. Flow cytometry has been historically used to detect the expression of CD56 in the diagnosis of chronic myelomonocytic leukemia (CMML). CD56 is a cell surface marker that presents the surface of monocytes [79]. Following over a decade of extensive research, high-throughput image-based flow cytometry is now an accepted and widely used tool in scientific research, particularly in the field of cancer biology. Many researchers have replaced microscopy-based clinical tools with image-based flow cytometry. Erber’s team originally used image-based flow cytometry to identify the presence of promyelocytic leukemia (PML) bodies for the diagnosis and prognosis of acute myeloid leukemia (AML) [80].

Applications of Flow cytometry in cancer research

  • Emerging as a tool for diagnosis of cancer (abnormal DNA content)

  • Specific histopathological diagnosis (RNA for hematological cancers; surface markers for lymphoid and myeloid neoplasms)

  • Prognosis (adverse impact of aneuploidy and high S percentage)

  • Treatment (cytokinetically oriented, monoclonal antibodies, drug pharmacology)

2.4 Western blot

The Western blot (WB), also known as immunoblot, is an analytical and quantitative method for detecting particular proteins in various biological materials, including liquids and tissue/cellular homogenates [81]. Harry Towbin and colleagues developed the WB method in 1979. The WB approach provides clear and valuable information for assessing the phosphorylation state of a protein. We can evaluate the modified form of protein in the sample either qualitatively or quantitatively. Radenkovic et al., detected cyclin D1 expression in tumour and peritumoral tissue of breast cancer patients by Western blotting method to found that Cyclin D1 expression decreased significantly with each advanced clinical stage of disease and tumour size [25]. Kinase activity-tagged western blotting (KAT-WB) detected autophosphorylation of Tyr-kinase and site-specific phosphorylation by multiple kinases enables to interrogate multiple kinase signaling pathways without using radioactive substances [82]. Western blot analysis provides an opportunity to obtain more insight into cell cycle regulation factor in tumorigenesis, could spur the discovery of many more successful therapeutic targets [83].

Applications of Western blot in cancer research

  • To detect the presence of cancer proteins biomarkers

  • To determine the extent of post-translational modifications

  • To verify protein expression in cloning applications

  • To analyze protein and biomarker expression levels

  • In antibody epitope mapping

  • To test for markers of disease in clinical settings.

2.5 Mass spectrometry

The emphasis use of mass spectrometric analysis in clinical research has been on biomarker identification, which includes proteomics, lipidomics, and metabolomics [84, 85]. Metabolites, proteins and lipids have been shown to help distinguish between malignant and healthy tissue among the many compounds detectable with MS. To be identified as a qualifying biomarker, a molecule must be distinguishable from other molecules, ideally, the sample is simple, quick, and easy to collect, high sensitivity and specificity [86, 87], and is used to diagnose and prognosis for various cancer such as thyroid cancer [88], lung cancer [89, 90], bladder cancer [91], Pancreatic Cancer [92, 93], breast cancer [94], ovarian cancer [52], oral cancer [53], prostate cancer [95]. In general, various classes of molecules may function as biomarkers due to an imbalance of tumor-suppressing and promoting agents in cancer cells, regulating genetic changes, and changing the composition of lipids, metabolites, and proteins (Figure 3).

Figure 3.

Schematic representation showed that well established method immunohistochemistry was widely used in clinical diagnosis for cancer detection. In recent years, emerging proteomic technologies such as mass spectrometry and protein microarray has been developed for precise detection of cancer protein biomarkers.

The MALDI-TOF MS combined with magnetic bead used for detecting serum protein biomarkers and establishment of boosting decision tree model for diagnosis of colorectal cancer patients [96], breast cancer [97]. These differentially regulated proteins were considered as potential biomarkers for the patients with CRC in the serum. The emerging mass spectrometry methods of nanoLC–MS/MS, targeted LC–MS/MS, and stable isotope-labeled multiple reactions monitoring (MRM) MS coupled to test machine learning algorithms and logistic regression used to analyze plasma samples from colorectal cancer patients. The novel peptide biomarkers combination of PF4, ITIH4, and APOE achieved sensitivity 84.5%, specificity 97.5% and an AUC of 0.96 in CRC diagnosis [98]. Moran et al., developed an intact protein assay to analyze PSA by capillary electrophoresis-electrospray ionization-mass spectrometry after affinity purification from prostate cancer patients’ urine [99, 100].

The integrating mass spectrometry imaging and gold nanoparticle (AuNP)-based signal amplification was developed for quantitatively profiling protein biomarkers on the surface of exosomes in cancer diagnosis. Cancer protein biomarkers were modified with organic oligomers as mass tags and specific antibodies on AuNPs. Exosomes captured by the antibody-coated gold chip are recognized by the AuNPs probes, forming a sandwich immunoassay. Multiple protein biomarkers can be quantitatively detected from the exosomes with the mass tags by mass spectrometry imaging [101]. Park et al., 2019 developed a simple and robust cancer diagnostic method using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS)-based total serum protein fingerprinting to diagnose liver cancer [102]. The proteome profile of vimentin, tubulin beta 2C chain, tubulin alpha 1C chain, actin cytoplasmic 2, apolipoprotein A-I, and collagen alpha 2(VI) chain as a potential biomarker that exhibited differential expression in ovarian cancer using two-dimensional gel electrophoresis (2D-GE) and matrix-assisted laser desorption/ionization-time of flight mass spectrometric (MALDI-TOF MS) analysis [103]. The protein modifications in the N-glycosylation profile are usually associated with many cancers, like colorectal cancer. In turn, MALDI-TOF/MS and LC–MS methods are the most accurate technology in the quantification of N-glycans compositions in the serum and tissue of colorectal cancer patients [104]. The similar proteomic analysis was performed to identify potential lung cancer biomarkers such as CD5L, CLEC3B, ITIH4, SERFINF1, SAA4, SERFINC1, and C20ORF3 detected via a liquid biopsy-for the noninvasive diagnosis of lung cancer [105].

Applications of mass spectrometry in cancer research

  • Discovery-based proteomic experiments with increasing cohort size is imperative for this technology to transfer to the clinic

  • Application of tissue proteomics to cancer research is using it in a concerted effort to complement genomics.

  • Proteomics biomarker research in human body fluids, urine and alternative liquid biopsies

  • Mass spectrometry-based clinical proteomics in cancer biomarker research

  • Prospective versus retrospective profiling of clinical specimens

  • Human population heterogeneity & protein variability

2.6 Protein array

The analysis of protein biomarkers using these high throughput methods can provide robust and previously unachievable diagnostic and prognostic information for a variety of cancers [106, 107]. The analytical protein array is a useful developing technology that enables simultaneously analyzing >4000 protein samples in cell or tissues for biomarker discovery (Huang et al., 2017). The microarray is currently utilized to analyze biopsy samples in clinical research trials, essentially lead to the collection of information linked to posttranslational modifications of proteins reflecting the active status of signal pathways and networks [108]. This technique has the potential to enhance cancer detection, prognosis, and treatments. Protein microarray technology has been used effectively in fundamental and applied proteome research and affinity studies for protein identification, quantification, and functional analysis [109, 110]. A protein function array is made up of thousands of natural proteins that have been immobilised in a specific arrangement. When a functional protein array is used for serum protein profiling, autoantibodies are usually detected as biomarkers for diagnosis of cancer detection and for monitoring the cancer treatment due to their stability, specificity, and ease of detection, as compared with other serological components [111]. Protein microarrays have allowed researchers to examine functional protein dysregulation in various cancer namely colorectal cancers [112], pancreatic cancer [113]. Mirus et al., identified ERBB2, TNC and ESR1 in prediagnostic plasma from people that succumb to pancreatic ductal adenocarcinoma [114]. In addition to the understanding of the biological mechanisms, analytical protein arrays have also been applied to profile drug resistance [115].

In recent advancements in protein microarray is Reverse-phase protein array/microarray (RPPA/RPPM), which can precisely map functional proteomic profiling in individual cancer patient. The personalized therapy was prescribed by the identification of functional proteomics profilling. RPPA is an antibody-based highly quantitative proteomic technology, used for profiling the expression and modification of signaling proteins, mainly in low-abundance analytes cases. Clinical trials are using RPPA technology molecular-targeted therapeutics [116, 117]. Horton et al., 2021 found that the minimal effects on RPPA protein concentration distributions in peripheral blood and bone marrow, demonstrating that these preanalytical variables have been successfully managed in a multi-site clinical trial setting for leukemia [118]. A proteomic study was carried out for determining the levels of post-translational protein modifications and total protein expression in myeloproliferative neoplasms patients using RPPA [119]. These results highlight the robustness and the reproducibility of RPPA technology and its capacity to identify protein markers of cancer or response to therapy. Recent proteomics studies have focused on the expression of seven markers (CD5, CD10, BCL2, BCL6, MUM1, Ki-67, and C-MYC) is analyzed by RPPA using 37 diffuse large B-cell lymphomas (DLBCL) tissues [120]. These results suggest that RPPA could be applicable as a supportive tool for determining lymphoma prognosis. With all of these improvements, we believe that protein array technology will soon become a dominant tool for biomarker discovery in cancers.

Applications of protein array in cancer research

  • Analytical and functional protein array for cancer biomarker discovery

  • Personalized medicine in breast and ovarian cancers using protein microarray

  • Protein profiling of cancer cells using protein microarray

  • Drug discovery for target identification and validation

2.7 Microfluidics

Microfluidic technology, as new creativity has a great impact on automation and miniaturization via handling a small volume of materials and samples for cancer diagnosis [121]. This method has be considered as a primary screening tool for diagnosing breast cancer based on its robustness, high throughput, low energy requirements, excellent accuracy and accessibility to the general public [122]. A miniaturized instrument was developed for chemiluminescence detection and signal analysis with the advances in microfluidic technology. The system was validated by testing four biomarkers of colorectal cancer using plasma samples from patients [123]. Another design of the microfluidic device, magnetic nanoparticles (Fe3O4NPs) was successfully functionalized with an exosome-binding antibody (anti-CD9) to mediate the magnetic capture in a microdevice. The captured exosomes were then subjected to analysis of CA19–9, a protein often used to monitor pancreatic cancer patients [124]. In the line of discovery, the nuclear matrix protein 22 (NMP22) and bladder cancer antigen (BTA) from the urine samples was detected using the microfluidic paper-based analytical device (mu PAD) was developed by Jiang [125]. This method is feasible for home-based self-detection from urine samples within 10 min for the total process, which provides a new way for quick, economical, and convenient tumor diagnosis, prognosis evaluation, and drug response (Figure 4). The functions and recent development of microfluidic chip to provide great potential for advancing noninvasive cancer diagnosis [126, 127].

Figure 4.

Microfluidic technology, as new creativity has a great impact on automation and miniaturization via handling a small volume of materials and samples for cancer diagnosis. An effective management of cancer diagnosis screening by using body fluids and cancer protein biomarkers for diagnosis, prognosis, therapy and monitoring to treatment.

Microfluidic devices which used to mimic cancer metastasis process are usually applied to several cell types in order to culture two or more organoids. Different organoids are separated by some specific biomaterials, such as polydimethylsiloxane (PDMS), and connected with each other by channels and controllable fluids. Xu et al. designed and constructed a multi-organ microfluidic chip to mimic lung cancer metastasis to the brain, bone and liver. In this platform, organoids were divided into different chambers, including upstream lung organoid and three downstream organoids. Different types of cells were seeded in each chamber to culture different organoids and each organoid were linked by side channels. The culture medium flowed through microvascular channels to simulate blood circulation. At the same time, a circulating vacuum was applied to mimic the physiological breathing [128]. This system provided a physiologically relevant context to recapitulate the complex process of lung cancer metastasis and help us to effectively explore the underlying mechanism of lung cancer metastasis.

The integration between 3D bioprinting and microfluidic chip has given microfluidic chip greater potential to model cancers. Traditionally, in cancer modeling on chip, microfabrication such as micromachining, photolithography and injection molding, are used in the fabrication of microfluidic chips [129]. These methods have high resolution and accuracy, but their high cost, complex process and difficult reproducibility greatly limited the development of microfluidic chip [130]. The emerging of 3D printing technology greatly simplifies the fabrication process of microfluidic chips. The biggest characteristic of microfluidic chip is the customizability, which means microfluidic chip is a very flexible scientific tool that can accommodate with advanced technologies. To date, microfluidic chip shows tremendous promise in cancer diagnosis and treatment. Microfluidic chip can be applied in everything from anticancer drug development and screening to cancer modeling and diagnosis.

Applications of Microfluidics in cancer research

  • The development of cancer preclinical model; Animal models, 2D culture, 3D culture, as well as tumor organoid.

  • Detection of cancer biomarkers

  • Anti-cancer drug screening and nano-drug preparation

  • Exploring tumor heterogeneity on microfluidic chip

Advertisement

3. Conclusions

Over the past decade, protein profiling has emerged as a dynamic discipline, capable of generating a comprehensive perspective of protein patterns, modification of protein in various tissue-specific cancer types and mechanisms of cancer progression. Proteomics studies and analytical techniques are developed with modern materials for precise detection of tumor-specific alteration in proteins. Recent times, the proteomics technologies have fabricated with modern nanocomposites provide a new and more efficient methods for protein detection and identifying biomarkers for the early detection of cancer. The upgraded proteomics technology will modify the current pathological classification and grading methods of cancer during the next decade. Proteomic technologies will have an impact on the diagnosis and management of cancer.

References

  1. 1. Henley SJ, Ward EM, Scott S, Ma J, Anderson RN, Firth AU, et al. Annual report to the nation on the status of cancer, part I: National cancer statistics. Cancer. 2020;126(10):2225-2249.
  2. 2. de Martel C, Georges D, Bray F, Ferlay J, Clifford GM. Global burden of cancer attributable to infections in 2018: a worldwide incidence analysis. The Lancet Global Health. 2020;8(2):e180–e190.
  3. 3. Guo R, Pan C, Shen J, Liu C. New serum biomarkers for detection of esophageal carcinoma using Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Journal of cancer research and clinical oncology. 2011;137(3):513-519.
  4. 4. K Nowicki T, Markiet K, Szurowska E. Diagnostic imaging of hepatocellular carcinoma-A pictorial essay. Current Medical Imaging. 2017;13(2):140-153.
  5. 5. Løberg M, Lousdal ML, Bretthauer M, Kalager M. Benefits and harms of mammography screening. Breast Cancer Research. 2015;17(1):1-12.
  6. 6. Meric-Bernstam F, Mills GB. Overcoming implementation challenges of personalized cancer therapy. Nature reviews Clinical oncology. 2012;9(9):542-548.
  7. 7. Hing JX, Mok CW, Tan PT, Sudhakar SS, Seah CM, Lee WP, et al. Clinical utility of tumour marker velocity of cancer antigen 15-3 (CA 15-3) and carcinoembryonic antigen (CEA) in breast cancer surveillance. The Breast. 2020;52:95-101.
  8. 8. Kim DW, Chen M, Wu J, Huland H, Graefen M, Tilki D, et al. Prostate-specific antigen levels of≤ 4 and> 4 ng/mL and risk of prostate cancer–specific mortality in men with biopsy Gleason score 9 to 10 prostate cancer. Cancer. 2021;
  9. 9. Song B, Wang L, Zhang Y, Li N, Dai H, Xu H, et al. Combined detection of HER2, Ki67, and GSTP1 genes on the diagnosis and prognosis of breast cancer. Cancer biotherapy & radiopharmaceuticals. 2019;34(2):85-90.
  10. 10. Jayasekara H, MacInnis RJ, Chamberlain JA, Dite GS, Leoce NM, Dowty JG, et al. Mortality after breast cancer as a function of time since diagnosis by estrogen receptor status and age at diagnosis. International journal of cancer. 2019;145(12):3207-3217.
  11. 11. Tainsky MA. Genomic and proteomic biomarkers for cancer: a multitude of opportunities. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2009;1796(2):176-193.
  12. 12. Sokolenko AP, Imyanitov EN. Molecular diagnostics in clinical oncology. Frontiers in molecular biosciences. 2018;5:76.
  13. 13. Zhang X, Sun X-F, Shen B, Zhang H. Potential applications of DNA, RNA and protein biomarkers in diagnosis, therapy and prognosis for colorectal cancer: a study from databases to AI-assisted verification. Cancers. 2019;11(2):172.
  14. 14. Duffy MJ. Tumor markers in clinical practice: a review focusing on common solid cancers. Medical Principles and Practice. 2013;22(1):4-11.
  15. 15. Enroth S, Berggrund M, Lycke M, Broberg J, Lundberg M, Assarsson E, et al. High throughput proteomics identifies a high-accuracy 11 plasma protein biomarker signature for ovarian cancer. Communications biology. 2019;2(1):1-12.
  16. 16. Henderson MC, Hollingsworth AB, Gordon K, Silver M, Mulpuri R, Letsios E, et al. Integration of serum protein biomarker and tumor associated autoantibody expression data increases the ability of a blood-based proteomic assay to identify breast cancer. PLoS One. 2016;11(8):e0157692.
  17. 17. Lin D, Feng S, Pan J, Chen Y, Lin J, Chen G, et al. Colorectal cancer detection by gold nanoparticle based surface-enhanced Raman spectroscopy of blood serum and statistical analysis. Optics express. 2011;19(14):13565-13577.
  18. 18. Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426(6965):450-454.
  19. 19. Wong Y-L, Ramanathan A, Yuen KM, Mustafa WMW, Abraham MT, Tay KK, et al. Comparative sera proteomics analysis of differentially expressed proteins in oral squamous cell carcinoma. PeerJ. 2021;9:e11548.
  20. 20. El-Shayeb AF, El-Habachi NM, Mansour AR, Zaghloul MS. Serum midkine is a more sensitive predictor for hepatocellular carcinoma than Dickkopf-1 and alpha-L-fucosidase in cirrhotic HCV patients. Medicine. 2021;100(17).
  21. 21. Li S-S, Tan Y-Y, Zhang Y, Liu M, Liu A. A simple electrochemical immunosensor based on worm-like platinum for highly sensitive determination of alpha-fetoprotein. Bioelectrochemistry. 2021;140:107804.
  22. 22. Guo X, Zhang Z, Yang M, Bing P, Yan X, Yang Q, et al. Time-Frequency Double Domain Resolving by Electromagnetically Induced Transparency Metasensors for Rapid and Label-Free Detection of Cancer Biomarker Midkine. Optics and Lasers in Engineering. 2021;142:106566.
  23. 23. Chaudhary MK, Jaiswal R, Singh N, Pandey A, Ali W. Evaluation of Serum s EGFR in Ovarian Tumors, its Comparison with Serum CA125, HE4 and Correlation with Histopathological Types. Indian Journal of Gynecologic Oncology. 2021;19(2):1-6.
  24. 24. von Brandenstein M, Herden J, Köditz B, Huerta M, Nestler T, Heidenreich A, et al. Non-invasive urine markers for the differentiation between RCCs and oncocytoma. Journal of clinical laboratory analysis. 2021;35(5):e23762.
  25. 25. Radenkovic S, Konjevic G, Nikitovic M, Stojanovic-Rundic S, Plesinac-Karapandzic V, Milovic-Kovacevic M, et al. Evaluation of Cyclin D1 expression by western blotting methods and immunohistochemistry in breast cancer patients. Journal of BU ON: Official Journal of the Balkan Union of Oncology. 2021;26(2):475-482.
  26. 26. Liang H, Dong J, Cheng Z, Li Q, Feng D, Ling B. B-cell receptor-associated protein 31 promotes migration and invasion in ovarian cancer cells. Experimental and Therapeutic Medicine. 2021;22(2):1-12.
  27. 27. Ma Y, Tian P, Chen Z, Yue D, Liu C, Li C, et al. Urinary malate dehydrogenase 2 is a new biomarker for early detection of non-small-cell lung cancer. Cancer Science. 2021;112(6):2349.
  28. 28. Xu Z, Hu Y, Yu Z. Effect of the ACY-1 gene on HER2 and TRAIL expression in rectal carcinoma. Experimental and Therapeutic Medicine. 2021;22(2):1-7.
  29. 29. Liu X, Lin L-Y, Tseng F-Y, Tan Y-C, Li J, Feng L, et al. Label-free electrochemical immunosensor based on gold nanoparticle/polyethyleneimine/reduced graphene oxide nanocomposites for the ultrasensitive detection of cancer biomarker matrix metalloproteinase-1. Analyst. 2021;
  30. 30. Jiang D, Zhang X, Liu M, Wang Y, Wang T, Pei L, et al. Discovering panel of autoantibodies for early detection of lung cancer based on focused protein array. Frontiers in immunology. 2021;12:710.
  31. 31. Falcucci S, Paolini F, Mileo AM, Franconi R, Massa S, Rinaldi A, et al. ePCL Electrospun Microfibrous Layers for Immune Assays: Sensitive ELISA for the Detection of Serum Antibodies Against HPV16 E7 Oncoprotein. ACS omega. 2021;6(13):8778-8783.
  32. 32. Nakaguro M, Tanigawa M, Hirai H, Yamamoto Y, Urano M, Takahashi RH, et al. The Diagnostic Utility of RAS Q61R Mutation-specific Immunohistochemistry in Epithelial-Myoepithelial Carcinoma. The American journal of surgical pathology. 2021;45(7):885-894.
  33. 33. Shekari Z, Zare HR, Falahati A. Dual assaying of breast cancer biomarkers by using a sandwich–type electrochemical aptasensor based on a gold nanoparticles–3D graphene hydrogel nanocomposite and redox probes labeled aptamers. Sensors and Actuators B: Chemical. 2021;332:129515.
  34. 34. Zemskova MY, Marinets M V, Sivkov A V, Pavlova J V, Shibaev AN, Sorokin KS. Integrin Alpha V in Urine: A Novel Noninvasive Marker for Prostate Cancer Detection. Frontiers in oncology. 2020;10.
  35. 35. Lin Z, Süsskind D. Exploring the role of BAFF as biomarker in the detection of uveal melanoma metastases. Journal of Cancer Research and Clinical Oncology. 2021;147(5):1389-1405.
  36. 36. Ganig N, Baenke F, Thepkaysone M-L, Lin K, Rao VS, Wong FC, et al. Proteomic Analyses of Fibroblast-and Serum-Derived Exosomes Identify QSOX1 as a Marker for Non-invasive Detection of Colorectal Cancer. Cancers. 2021;13(6):1351.
  37. 37. Choi YJ, Yoon W, Lee A, Han Y, Byun Y, Kang JS, et al. Diagnostic model for pancreatic cancer using a multi-biomarker panel. Annals of surgical treatment and research. 2021;100(3):144.
  38. 38. Zhu Y-L, Lian Y-M, Wang J-K, Chen Z-P, Yu R-Q. Ultrasensitive detection of protein biomarkers by MALDI-TOF mass spectrometry based on ZnFe2O4 nanoparticles and mass tagging signal amplification. Talanta. 2021;224:121848.
  39. 39. Cui C, Duan Y, Qiu C, Wang P, Sun G, Ye H, et al. Identification of Novel Autoantibodies Based on the Human Proteomic Chips and Evaluation of Their Performance in the Detection of Gastric Cancer. Frontiers in oncology. 2021;11:342.
  40. 40. Muñoz-San Martín C, Gamella M, Pedrero M, Montero-Calle A, Pérez-Ginés V, Camps J, et al. Anticipating metastasis through electrochemical immunosensing of tumor hypoxia biomarkers. Analytical and bioanalytical chemistry. 2021;1-14.
  41. 41. Zhang W, Liu T, Wu M, Chen X, Han L, Shi Z, et al. Development of a nanobody-based immunoassay for the sensitive detection of fibrinogen-like protein 1. Acta Pharmacologica Sinica. 2021;1-9.
  42. 42. Leung SOA, Feldman S, Kalyanaraman R, Shanmugam V, Worley MJ, Berkowitz RS, et al. Triaging abnormal cervical cancer screening tests using p16INK4a detection by ELISA on fresh cervical samples. American Journal of Reproductive Immunology. 2021;e13394.
  43. 43. Hao Y, Ye M, Chen X, Zhao H, Hasim A, Guo X. Discovery and validation of FBLN1 and ANT3 as potential biomarkers for early detection of cervical cancer. Cancer cell international. 2021;21(1):1-13.
  44. 44. Chen J, Li H, Xu W, Guo X. Evaluation of serum ATX and LPA as potential diagnostic biomarkers in patients with pancreatic cancer. BMC gastroenterology. 2021;21(1):1-10.
  45. 45. Pietrzyk Ł, Korolczuk A, Matysek M, Arciszewski MB, Torres K. Clinical Value of Detecting Tumor Endothelial Marker 8 (ANTXR1) as a Biomarker in the Diagnosis and Prognosis of Colorectal Cancer. Cancer Management and Research. 2021;13:3113.
  46. 46. Yang HX, Yang Y, Li XD, Miao XM, Yang C, Zhi DF, et al. The detection of SAS1B in serum provides clues for early diagnosis of thyroid cancer. Eur Rev Med Pharmacol Sci. 2021;25(3):1410-1424.
  47. 47. Lokman NA, Ricciardelli C, Stephens AN, Jobling TW, Hoffmann P, Oehler MK. Diagnostic Value of Plasma Annexin A2 in Early-Stage High-Grade Serous Ovarian Cancer. Diagnostics 2021, 11, 69. s Note: MDPI stays neutral with regard to jurisdictional claims in …; 2021.
  48. 48. Nie X-C, He F, Lan C, Niu J-M, Xia P. Combined Serum DKK3 and Circulating CD133 Cells as Prognostic Biomarkers for Ovarian Cancer Patients. OncoTargets and therapy. 2021;14:427.
  49. 49. Sharafeldin M, Chen T, Ozkaya GU, Choudhary D, Molinolo AA, Gutkind JS, et al. Detecting cancer metastasis and accompanying protein biomarkers at single cell levels using a 3D-printed microfluidic immunoarray. Biosensors and Bioelectronics. 2021;171:112681.
  50. 50. Thangsunan P, Lal N, Tiede C, Moul S, Robinson JI, Knowles MA, et al. Affimer-based impedimetric biosensors for fibroblast growth factor receptor 3 (FGFR3): a novel tool for detection and surveillance of recurrent bladder cancer. Sensors and Actuators B: Chemical. 2021;326:128829.
  51. 51. Mawaribuchi S, Haramoto Y, Tateno H, Onuma Y, Aiki Y, Ito Y. rBC2LCN lectin as a potential probe of early-stage HER2-positive breast carcinoma. FEBS open bio. 2020;10(6):1056-1064.
  52. 52. Schuster-Little N, Madera S, Whelan R. Developing a mass spectrometry–based assay for the ovarian cancer biomarker CA125 (MUC16) using suspension trapping (STrap). Analytical and bioanalytical chemistry. 2020;412(24):6361-6370.
  53. 53. Hsiao Y-C, Lin S-Y, Chien K-Y, Chen S-F, Wu C-C, Chang Y-T, et al. An immuno-MALDI mass spectrometry assay for the oral cancer biomarker, matrix metalloproteinase-1, in dried saliva spot samples. Analytica chimica acta. 2020;1100:118-130.
  54. 54. Hu R, Gupta R, Wang Z, Wang C, Sun H, Singamaneni S, et al. Bioplasmonic paper–based assay for perilipin-2 non-invasively detects renal cancer. Kidney international. 2019;96(6):1417-1421.
  55. 55. Chang S-C, Lin W-L, Chang Y-F, Lee C-T, Wu J-S, Hsu P-H, et al. Glycoproteomic identification of novel plasma biomarkers for oral cancer. Journal of food and drug analysis. 2019;27(2):483-493.
  56. 56. Šuster NK, Meznaric M, Škorja N, Virant-Klun I, Verdenik I, Smrkolj Š. Cancer stem cell-related marker NANOG expression in ovarian serous tumors using Western blotting and immunohistochemistry: comparison of two techniques. European Journal of Gynaecological Oncology. 2019;40(6):948-952.
  57. 57. Kotzsch M, Kirchner T, Soelch S, Schäfer S, Friedrich K, Baretton G, et al. Inverse association of rab31 and mucin-1 (CA15-3) antigen levels in estrogen receptor-positive (ER+) breast cancer tissues with clinicopathological parameters and patients’ prognosis. American journal of cancer research. 2017;7(9):1959.
  58. 58. Miyashita M, Gonda K, Tada H, Watanabe M, Kitamura N, Kamei T, et al. Quantitative diagnosis of HER 2 protein expressing breast cancer by single-particle quantum dot imaging. Cancer medicine. 2016;5(10):2813-2824.
  59. 59. Chikkaveeraiah B V, Mani V, Patel V, Gutkind JS, Rusling JF. Microfluidic electrochemical immunoarray for ultrasensitive detection of two cancer biomarker proteins in serum. Biosensors and Bioelectronics. 2011;26(11):4477-4483.
  60. 60. Clark MF, Adams AN. Characteristics of the microplate method of enzyme-linked immunosorbent assay for the detection of plant viruses. Journal of general virology. 1977;34(3):475-483.
  61. 61. Engvall E, Perlmann P. Enzyme-linked immunosorbent assay (ELISA) quantitative assay of immunoglobulin G. Immunochemistry. 1971;8(9):871-874.
  62. 62. Reen DJ. Enzyme-linked immunosorbent assay (ELISA). Basic Protein and Peptide Protocols. 1994;461-466.
  63. 63. Oesterling JE. Prostate specific antigen: a critical assessment of the most useful tumor marker for adenocarcinoma of the prostate. The Journal of urology. 1991;145(5):907-923.
  64. 64. Freedland SJ, Humphreys EB, Mangold LA, Eisenberger M, Dorey FJ, Walsh PC, et al. Risk of prostate cancer–specific mortality following biochemical recurrence after radical prostatectomy. Jama. 2005;294(4):433-439.
  65. 65. Shim JU, Ranasinghe RT, Smith CA, Ibrahim SM, Hollfelder F, Huck WTS, et al. Ultrarapid generation of femtoliter microfluidic droplets for single-molecule-counting immunoassays. ACS Nano 7, 5955-5964. 2013.
  66. 66. Zhang B, Liu B, Chen G, Tang D. Redox and catalysis ‘all-in-one’infinite coordination polymer for electrochemical immunosensor of tumor markers. Biosensors and Bioelectronics. 2015;64:6-12.
  67. 67. Xu T, Liu N, Yuan J, Ma Z. Triple tumor markers assay based on carbon–gold nanocomposite. Biosensors and Bioelectronics. 2015;70:161-166.
  68. 68. Bonacho T, Rodrigues F, Liberal J. Immunohistochemistry for diagnosis and prognosis of breast cancer: a review. Biotechnic & Histochemistry. 2020; 95(2): 71-91.
  69. 69. Mandel P, Wenzel M, Hoeh B, Welte MN, Preisser F, Inam T, et al. Immunohistochemistry for Prostate Biopsy—Impact on Histological Prostate Cancer Diagnoses and Clinical Decision Making. Current Oncology. 2021;28(3):2123-2133.
  70. 70. Albero-González R, Munné-Collado J, Pijuan L, Simón M, Gimeno-Beltrán J, Mojal S, et al. Complementary value of electron microscopy and immunohistochemistry in the diagnosis of non-small cell lung cancer: A potential role for electron microscopy in the era of targeted therapy. Ultrastructural pathology. 2019;43(6):237-247.
  71. 71. Imai H, Wasamoto S, Yamaguchi O, Suzuki K, Sugiyama T, Uchino J, et al. Efficacy and safety of first-line pembrolizumab monotherapy in elderly patients (aged≥ 75 years) with non-small cell lung cancer. Journal of cancer research and clinical oncology. 2020;146(2):457-466.
  72. 72. Inamura K. Update on immunohistochemistry for the diagnosis of lung cancer. Cancers. 2018;10(3):72.
  73. 73. Aimi F, Procopio M-G, Flores MTA, Brouland J-P, Piazzon N, Brajkovic S, et al. Microfluidic-based immunohistochemistry for breast cancer diagnosis: a comparative clinical study. Virchows Archiv. 2019;475(3):313-323.
  74. 74. Ilie M, Hofman V, Ortholan C, Bonnetaud C, Coëlle C, Mouroux J, et al. Predictive clinical outcome of the intratumoral CD66b-positive neutrophil-to-CD8-positive T-cell ratio in patients with resectable nonsmall cell lung cancer. Cancer. 2012;118(6):1726-1737.
  75. 75. Fulwyler MJ. Electronic separation of biological cells by volume. Science. 1965;150(3698):910-911.
  76. 76. Orfao A, Ciudad J, Gonzalez M, Lopez A, Del Mar Abad M, Paz Bouza JI, et al. Flow cytometry in the diagnosis of cancer. Scandinavian Journal of Clinical and Laboratory Investigation. 1995;55(sup221):145-52.
  77. 77. Danova M, Torchio M, Comolli G, Sbrana A, Antonuzzo A, Mazzini G. The role of automated cytometry in the new era of cancer immunotherapy. Molecular and clinical oncology. 2018;9(4):355-361.
  78. 78. Starý J, Sokol L, Hausner P, Hrodek O, Sedmihorská J, Volejníková J, et al. Use of flow cytometry in the diagnosis of acute leukemias in childhood. Casopis lekaru ceskych. 1992;131(12):364-367.
  79. 79. Hudson CA, Burack WR, Bennett JM. Emerging utility of flow cytometry in the diagnosis of chronic myelomonocytic leukemia. Leukemia research. 2018;73:12-15.
  80. 80. Grimwade L, Gudgin E, Bloxham D, Scott MA, Erber WN. PML protein analysis using imaging flow cytometry. Journal of clinical pathology. 2011;64(5):447-450.
  81. 81. Ghosh R, Gilda JE, Gomes A V. The necessity of and strategies for improving confidence in the accuracy of western blots. Expert review of proteomics. 2014;11(5):549-560.
  82. 82. Eto M, Katsuki S, Tanaka Y, Takeya K. Kinase activity-tagged western blotting assay. BioTechniques. 2019;68(4):211-213.
  83. 83. Li L-H, Huang Q-M, Barbero M, Liu L, Nguyen T-T, Beretta-Piccoli M, et al. Quantitative proteomics analysis to identify biomarkers of chronic myofascial pain and therapeutic targets of dry needling in a rat model of myofascial trigger points. Journal of pain research. 2019;12:283.
  84. 84. Zhou W, Petricoin EF, Longo C. Mass spectrometry-based biomarker discovery. In: Molecular Profiling. Springer; 2017. p. 297-311.
  85. 85. Steffen P, Kwiatkowski M, Robertson WD, Zarrine-Afsar A, Deterra D, Richter V, et al. Protein species as diagnostic markers. Journal of proteomics. 2016;134:5-18.
  86. 86. Brower V. Biomarkers: Portents of malignancy. Nature. 2011;471(7339):S19–S20.
  87. 87. Rosenling T, Stoop MP, Smolinska A, Muilwijk B, Coulier L, Shi S, et al. The impact of delayed storage on the measured proteome and metabolome of human cerebrospinal fluid. Clinical chemistry. 2011;57(12):1703-1711.
  88. 88. Li F, Pan D, He Y, Wu Y, Peng J, Li J, et al. Using ultrasound features and radiomics analysis to predict lymph node metastasis in patients with thyroid cancer. BMC surgery. 2020;20(1):1-6.
  89. 89. Wu H, Pan Y, Kopylov AT, Zgoda V, Ma M, Wang C, et al. Assessment of Serological Early Biomarker Candidates for Lung Adenocarcinoma by using Multiple Reaction Monitoring-Mass Spectrometry. PROTEOMICS–Clinical Applications. 2020;14(4):1900095.
  90. 90. Zhou Y, Wu C, Lu G, Hu Z, Chen Q, Du X. FGF/FGFR signaling pathway involved resistance in various cancer types. Journal of Cancer. 2020;11(8):2000.
  91. 91. Nedjadi T, Benabdelkamal H, Albarakati N, Masood A, Al-Sayyad A, Alfadda AA, et al. Circulating proteomic signature for detection of biomarkers in bladder cancer patients. Scientific reports. 2020;10(1):1-10.
  92. 92. Jia K, Zhao X, Dang X. Mass spectrometry-based iTRAQ analysis of serum markers in patients with pancreatic cancer. Oncology letters. 2020;19(6):4106-4114.
  93. 93. Kim Y, Yeo I, Huh I, Kim J, Han D, Jang J-Y, et al. Development and Multiple Validation of the Protein Multi-marker Panel for Diagnosis of Pancreatic Cancer. Clinical Cancer Research. 2021;27(8):2236-2245.
  94. 94. Lee SB, Bose S, Ahn SH, Son BH, Ko BS, Kim HJ, et al. Breast cancer diagnosis by analysis of serum N-glycans using MALDI-TOF mass spectroscopy. Plos one. 2020;15(4):e0231004.
  95. 95. Geisler C, Gaisa NT, Pfister D, Fuessel S, Kristiansen G, Braunschweig T, et al. Identification and validation of potential new biomarkers for prostate cancer diagnosis and prognosis using 2D-DIGE and MS. BioMed research international. 2015;2015.
  96. 96. Liu C, Pan C, Shen J, Wang H, Yong L. MALDI-TOF MS combined with magnetic beads for detecting serum protein biomarkers and establishment of boosting decision tree model for diagnosis of colorectal cancer. International journal of medical sciences. 2011;8(1):39.
  97. 97. de Noo ME, Deelder A, van der Werff M, Özalp A, Mertens B, Tollenaar R. MALDI-TOF serum protein profiling for the detection of breast cancer. Oncology Research and Treatment. 2006;29(11):501-506.
  98. 98. Tsai I, Su EC-Y, Tsai I-L, Lin C-Y. Clinical assay for the early detection of colorectal cancer using mass spectrometric wheat germ agglutinin multiple reaction monitoring. Cancers. 2021;13(9):2190.
  99. 99. Otto JJ, Correll VL, Engstroem HA, Hitefield NL, Main BP, Albracht B, et al. Targeted Mass Spectrometry of a Clinically Relevant PSA Variant from Post-DRE Urines for Quantitation and Genotype Determination. PROTEOMICS–Clinical Applications. 2020;14(6):2000012.
  100. 100. Moran AB, Domínguez-Vega E, Nouta J, Pongracz T, de Reijke TM, Wuhrer M, et al. Profiling the proteoforms of urinary prostate-specific antigen by capillary electrophoresis–mass spectrometry. Journal of Proteomics. 2021;238:104148.
  101. 101. Wang Y, Zhang K, Huang X, Qiao L, Liu B. Mass spectrometry imaging of mass tag immunoassay enables the quantitative profiling of biomarkers from dozens of exosomes. Analytical Chemistry. 2020;93(2):709-714.
  102. 102. Park H-G, Jang K-S, Park H-M, Song W-S, Jeong Y-Y, Ahn D-H, et al. MALDI-TOF MS-based total serum protein fingerprinting for liver cancer diagnosis. Analyst. 2019;144(7):2231-2238.
  103. 103. Noreen S, Akhtar S, Batool T, Gardner QA, Akhtar MW. Tubulin Beta 2C Chain (TBB2C), a Potential Marker of Ovarian Cancer, an Insight from Ovarian Cancer Proteome Profile. ACS omega. 2021;6(16):10506-10514.
  104. 104. Coura M de, Barbosa EA, Brand GD, Bloch C, de Sousa JB. Identification of Differential N-Glycan Compositions in the Serum and Tissue of Colon Cancer Patients by Mass Spectrometry. Biology. 2021;10(4):343.
  105. 105. Choi E-S, Faruque H Al, Kim J-H, Kim KJ, Choi JE, Kim BA, et al. CD5L as an Extracellular Vesicle-Derived Biomarker for Liquid Biopsy of Lung Cancer. Diagnostics. 2021;11(4):620.
  106. 106. Grant GM, Fortney A, Gorreta F, Estep M, Del Giacco L, Van Meter A, et al. Microarrays in cancer research. Anticancer research. 2004;24(2A):441-448.
  107. 107. Russo G, Zegar C, Giordano A. Advantages and limitations of microarray technology in human cancer. Oncogene. 2003;22(42):6497-6507.
  108. 108. Liotta LA, Espina V, Mehta AI, Calvert V, Rosenblatt K, Geho D, et al. Protein microarrays: meeting analytical challenges for clinical applications. Cancer cell. 2003;3(4):317-325.
  109. 109. Poetz O, Schwenk JM, Kramer S, Stoll D, Templin MF, Joos TO. Protein microarrays: catching the proteome. Mechanisms of ageing and development. 2005;126(1):161-170.
  110. 110. Talapatra A, Rouse R, Hardiman G. Protein microarrays: challenges and promises. Pharmacogenomics. 2002;3(4):527-536.
  111. 111. Heo C-K, Bahk YY, Cho E-W. Tumor-associated autoantibodies as diagnostic and prognostic biomarkers. BMB reports. 2012;45(12):677.
  112. 112. Barderas R, Babel I, Diaz-Uriarte R, Moreno V, Suarez A, Bonilla F, et al. An optimized predictor panel for colorectal cancer diagnosis based on the combination of tumor-associated antigens obtained from protein and phage microarrays. Journal of proteomics. 2012;75(15):4647-4655.
  113. 113. Gerdtsson AS, Malats N, Säll A, Real FX, Porta M, Skoog P, et al. A multicenter trial defining a serum protein signature associated with pancreatic ductal adenocarcinoma. International journal of proteomics. 2015;2015.
  114. 114. Mirus JE, Zhang Y, Li CI, Lokshin AE, Prentice RL, Hingorani SR, et al. Cross-species antibody microarray interrogation identifies a 3-protein panel of plasma biomarkers for early diagnosis of pancreas cancer. Clinical Cancer Research. 2015;21(7):1764-1771.
  115. 115. Smith L, Watson MB, O’Kane SL, Drew PJ, Lind MJ, Cawkwell L. The analysis of doxorubicin resistance in human breast cancer cells using antibody microarrays. Molecular cancer therapeutics. 2006;5(8):2115-2120.
  116. 116. Masuda M, Yamada T. Signaling pathway profiling by reverse-phase protein array for personalized cancer medicine. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics. 2015;1854(6):651-7.
  117. 117. Elemike EE, Uzoh IM, Onwudiwe DC, Babalola OO. The role of nanotechnology in the fortification of plant nutrients and improvement of crop production. Applied Sciences. 2019;9(3):499.
  118. 118. Horton TM, Hoff FW, van Dijk A, Jenkins GN, Morrison D, Bhatla T, et al. The effects of sample handling on proteomics assessed by reverse phase protein arrays (RPPA): Functional proteomic profiling in leukemia. Journal of Proteomics. 2021;233:104046.
  119. 119. Tognon R, Almeida-e-Silva DC, Andraos-Rey R, Ristov M, Ambrósio L, de Almeida FC, et al. A proteomic study of myeloproliferative neoplasms using reverse-phase protein arrays. Leukemia & Lymphoma. 2020;61(13):3052-3065.
  120. 120. Suzuki M, Muroi A, Nojima M, Numata A, Takasaki H, Sakai R, et al. Utility of a Reverse Phase Protein Array to Evaluate Multiple Biomarkers in Diffuse Large B-Cell Lymphoma. PROTEOMICS–Clinical Applications. 2020;14(1):1900091.
  121. 121. Mahhengam N, fahem ghetran Khazaali A, Aravindhan S, Olegovna Zekiy A, Melnikova L, Siahmansouri H. Applications of Microfluidic Devices in the Diagnosis and Treatment of Cancer: A Review Study. Critical reviews in analytical chemistry. 2021;1-15.
  122. 122. Panesar S, Neethirajan S. Microfluidics: Rapid Diagnosis for Breast Cancer–A Review. Nano-micro letters. 2016;8(3):204-220.
  123. 123. Dai B, Yin C, Wu J, Li W, Zheng L, Lin F, et al. A flux-adaptable pump-free microfluidics-based self-contained platform for multiplex cancer biomarker detection. Lab on a Chip. 2021;21(1):143-153.
  124. 124. Sancho-Albero M, Sebastián V, Sesé J, Pazo-Cid R, Mendoza G, Arruebo M, et al. Isolation of exosomes from whole blood by a new microfluidic device: proof of concept application in the diagnosis and monitoring of pancreatic cancer. Journal of nanobiotechnology. 2020;18(1):1-15.
  125. 125. Jiang Q, Han T, Ren H, Aziz AUR, Li N, Zhang H, et al. Bladder cancer hunting: A microfluidic paper-based analytical device. Electrophoresis. 2020;41(16-17):1509-1516.
  126. 126. Garcia-Cordero JL, Maerkl SJ. Microfluidic systems for cancer diagnostics. Current opinion in biotechnology. 2020;65:37-44.
  127. 127. Guo Q, Zhang L, Liu J, Li Z, Li J, Zhou W, et al. Multifunctional microfluidic chip for cancer diagnosis and treatment. Nanotheranostics. 2021;5(1):73.
  128. 128. Xu Z, Li E, Guo Z, Yu R, Hao H, Xu Y, et al. Design and construction of a multi-organ microfluidic chip mimicking the in vivo microenvironment of lung cancer metastasis. ACS applied materials & interfaces. 2016;8(39):25840-25847.
  129. 129. Lee H, Cho D-W. One-step fabrication of an organ-on-a-chip with spatial heterogeneity using a 3D bioprinting technology. Lab on a Chip. 2016;16(14):2618-2625.
  130. 130. Yu F, Choudhury D. Microfluidic bioprinting for organ-on-a-chip models. Drug discovery today. 2019;24(6):1248-1257.

Written By

Shenbagamoorthy Sundarraj, Gopalan Rajagopal, Balaji Sundaramahalingam, Madasamy Sundar and Ramar Thangam

Submitted: 10 July 2021 Reviewed: 04 October 2021 Published: 23 June 2022