Open Access
13 October 2017 Noninvasive detection of nasopharyngeal carcinoma based on saliva proteins using surface-enhanced Raman spectroscopy
Author Affiliations +
Abstract
The present study evaluated the capability of saliva analysis combining membrane protein purification with surface-enhanced Raman spectroscopy (SERS) for noninvasive detection of nasopharyngeal carcinoma (NPC). A rapid and convenient protein purification method based on cellulose acetate membrane was developed. A total of 659 high-quality SERS spectra were acquired from purified proteins extracted from the saliva samples of 170 patients with pathologically confirmed NPC and 71 healthy volunteers. Spectral analysis of those saliva protein SERS spectra revealed specific changes in some biochemical compositions, which were possibly associated with NPC transformation. Furthermore, principal component analysis combined with linear discriminant analysis (PCA-LDA) was utilized to analyze and classify the saliva protein SERS spectra from NPC and healthy subjects. Diagnostic sensitivity of 70.7%, specificity of 70.3%, and diagnostic accuracy of 70.5% could be achieved by PCA-LDA for NPC identification. These results show that this assay based on saliva protein SERS analysis holds promising potential for developing a rapid, noninvasive, and convenient clinical tool for NPC screening.

1.

Introduction

According to Global Cancer Statistics, cancer is one of the most common causes of death, with about 14.1 million people being diagnosed and 8.2 million people dying of cancer in 2012 worldwide.1 Nasopharyngeal carcinoma (NPC), a head and neck malignancy, is an endemic disease with high mortality rates in certain regions of southeastern Asia, including Hong Kong, Guangdong province of China, Fujian province of China, and some other South Asian countries.25 The five-year overall survival rate is about 90% for stage I NPC patients; however, the stage IV group has a poor five-year overall survival rate of 30.3%.6,7 Hence, early screening and effective treatment are essential for preventing the advancement of NPC and improving the five-year overall survival rate. Unfortunately, early detection remains a great challenge due to the insidious nature of NPC and the relative anatomical inaccessibility of the nasopharynx. At present, the common detection and screening methods for NPC mainly include electronic nasopharyngoscope examination, nasopharyngeal and neck magnetic resonance imaging, body positron emission tomography, bone emission CT, and biopsy, which suffer the disadvantages of being time-consuming, high-cost, complex procedures, and based on subjective judgment of clinicians with varying levels of experience.810 Therefore, development of an alternative diagnostic method is of significant clinical value for NPC screening.

Raman spectroscopy (RS) is a powerful optical analytical tool with many advantages over conventional optical detection techniques. For example, RS has low water interference, high signal-to-noise ratio, and high sensitivity compared with Fourier infrared spectroscopy.11 Additionally, RS has a narrow peak width and multiplexing detection ability, leading to better analytical efficiency than fluorescence spectroscopy.12,13 RS has attracted significant attentions for its material analysis and biomedical applications, especially for cancer diagnosis, due to its ability to provide structural- and biochemical-specific information of macromolecules, such as proteins, nucleic acids, lipids, and so on.14,15 However, the conventional RS technique has some deficiencies, including its inherent weak Raman scattering efficiency and strong fluorescence background. For instance, the Raman signal of a biological molecule is 1/10,000 of its fluorescence signal. To overcome these problems, surface-enhanced Raman spectroscopy (SERS) technology exploits the interaction between the biomolecules and metal nanoparticles (NPs) surface to dramatically increase the Raman signal (enhancement factors of up to 1013 to 1015), providing a more powerful and sensitive detection approach for cancer detection.2,16,17 Currently, SERS technology has been widely used to detect various cancers in human samples, such as DNA, RNA, cell, blood, and tissue.18 Compared with traditional polymerase chain reactions or immunoassays, biomarker detection based on SERS avoids use of expensive reagents and complex sample preparation steps. In addition, SERS has many advantages over traditional fluorescent method, such as better multiplex capability and less photobleaching.19,20

Human saliva, as a body fluid, contains a large number of serum components, abundant protein, and metabolites, which may change when associated with neoplastic transformation.21 Comparing the above-mentioned human samples, saliva is regarded as an ideal medium for cancer diagnostics due to its rapid, noninvasive, and convenient collection procedure.2224 Recently, application of SERS in saliva analysis was investigated extensively to detect several types of cancer.19,2527 These preliminary results demonstrated the potential of the saliva SERS method for cancer detection.

To further verify the efficiency of saliva SERS in NPC detection, in this work, a large number of NPC saliva samples were collected, and a saliva sample analysis based on membrane protein purification was developed for saliva SERS detection. Moreover, principal component analysis combined with a linear discriminant analysis (PCA-LDA) diagnostic algorithm was employed to classify the saliva protein SERS spectra from the NPC group and the healthy group.

2.

Materials and Methods

2.1.

Preparation of Human Saliva Samples

In this study, the saliva samples were collected from 170 NPC patients with confirmed clinical and histopathological diagnosis of NPC lesions and 71 healthy volunteers. Detailed information on these samples is presented in Table 1. This study obtained ethical approval from Fujian Provincial Cancer Hospital (Fuzhou, Fujian, China). The collection process of the saliva samples is as follows: (1) after 12 h of overnight fasting, 1.5 ml of saliva was collected from the study subjects who washed their mouth with water three times between 7 and 8 am; (2) the collected saliva was centrifuged at 13,900 rpm for 10 min to remove oral epithelial cells and any residual food debris; and (3) the pure saliva samples were frozen at 80°C until used.

Table 1

Clinical diagnosis of NPC patients and healthy subjects.

CharacteristicNPC patientsHealthy volunteers
Total number17071
Gender
Male10641
Female6430
Age (years)
509236
>507835
Clinical stage of NPC
I and II39NA
IV and III131NA
Note: NPC, nasopharyngeal carcinoma and NA, not applicable.

2.2.

Preparation of Silver Colloids

Silver (Ag) colloids were synthesized using hydroxylamine hydrochloride and Ag nitrate according to the deoxidizing method reported by Li et al.28 At first, 4.5 ml sodium hydroxide solution was mixed with 5 ml of 0.06 M hydroxylamine hydrochloride solution. After that, the mixture was added rapidly to 90 ml of 0.0011 M silver nitrate aqueous solution. The resulting mixture was stirred to obtain uniformly gray Ag colloids. Meanwhile, the maximum absorption peak of Ag colloids was 418 nm, and the Ag colloids NPs sizes were represented with a mean diameter of 35 nm by transmission electron microscopy (TEM) [Fig. 1(a)]. Finally, the final Ag colloids were obtained by centrifuging the solution at 10,000 rpm for 10 min and removing the supernatant as the SERS substrate.

Fig. 1

(a) UV–visible–NIR absorption spectra of Ag colloids and TEM micrograph of the Ag colloidal surface and (b) the schematic diagram of saliva protein purification and SERS detection.

JBO_22_10_105004_f001.png

2.3.

Saliva Protein Purification and Surface-Enhanced Raman Spectroscopy Detection

Figure 1(b) shows the schematic diagram of saliva protein purification and SERS detection. In brief, 10  μl of the purified saliva sample was blotted onto the cellulose acetate (CA) membrane (2×8  cm) by a trace pipette gun two times. In total, 20  μL of each purified saliva sample was used. After the saliva sample was completely absorbed for about 5 min, the membrane was washed for 5 min in 400 ml of a special solution made up of 180 ml of 95% ethanol, 200 ml of distilled water, and 20 ml of glacial acetic acid. This step aimed to remove any other components contained in the saliva sample and leave only the saliva proteins in the CA membrane. Then, the membrane was patted dry with a filter paper for about 10 min. After that, the position of the CA membrane that only contained saliva proteins was cut into pieces and collected in a tube. Next, 150  μl of acetic acid was added into the tube to dissolve the membrane fragments into a transparent gel. Then, 150  μl of a prepared Ag colloids solution was added into the tube and the tube was placed into 37°C water for 20 min. Finally, 2  μl of liquid supernatant (protein–Ag NP mixture) was dripped onto a clean aluminum plate (Guantai Metal, Hebei, China) for SERS measurements. All SERS spectra of the saliva protein were collected with a confocal Raman microspectrometer (Renishaw plc, Gloucestershire, UK) using a Peltier cooled charge-coupled device camera under a 785-nm diode laser (a maximum power output of 5 mW) in the range of 600 to 1750  cm1. All spectra were recorded with 50× objective, 2cm1 spectral resolution, and 10 s acquisition time. The software package WIRE 2.0 was used for raw spectral acquisition.

2.4.

Data Processing and Multivariate Statistical Analysis

All the raw SERE spectra of saliva proteins contain Raman scattering, fluorescence background, and noise signals. To obtain better Raman spectra, a Vancouver Raman algorithm based on a fifth-order polynomial fitting method was used to remove fluorescence background and noise signals.29 Then, all background-subtracted SERS spectra were normalized to the integrated area under the curve in the range of 600 to 1750  cm1 for a better comparison of SERS spectral shape in the analysis. This sophisticated and robust diagnostic model based on PCA-LDA was used for Raman spectral analysis in saliva detection. In this study, PCA was used to reduce complex data sets and select the principal components (PCs) that account for the maximal variances in the multidimensional data sets.30 To further analyze saliva protein SERS data, three diagnostically significant PCs (p<0.05) were selected for one independent sample t-test. LDA was used to generate effective diagnostics using three diagnostically significant PCs with leave-one-out cross-validation methods. Receiver operating characteristic (ROC) curves were generated by successively changing the discrimination threshold levels to additionally evaluate the classifications of the multivariate statistical method for NPC diagnosis.30

3.

Results and Discussions

Figures 2(a) and 2(b) show the SERS spectra of untreated saliva samples of 10 patients with NPC and saliva protein samples after membrane protein purification from the same samples, respectively. Great variations in spectral features, including spectral intensity, position, and width, could be found in the untreated saliva SERS within the cancer group, making it difficult to achieve efficient diagnostics using SERS spectral analysis. This is explainable. A saliva sample contains a variety of native constituents (proteins, peptides, polynucleotides, and electrolytes) and exogenous substances (nonadherent oral bacteria, food remainders, traces of medications, or chemical products).31,32 Although preprocessing such as washing the mouth for saliva collection was implemented, there remained some exogenous substances in the saliva samples that generated prominent SERS signals. Thus, great SERS spectral variation from untreated saliva samples was probably attributed to the different exogenous substances among subjects. Interestingly, the reproducibility of SERS spectra [Fig. 2(b)] was dramatically improved by the saliva protein purification method developed. This method not only circumvented the limitations of raw saliva SERS detection but also provided a unique opportunity to use SERS to explore the changes in saliva proteins associated with cancer transformation.

Fig. 2

(a) Untreated saliva SERS spectra from 10 NPC saliva samples and (b) saliva protein SERS spectra using membrane protein purification method from the same samples.

JBO_22_10_105004_f002.png

Figure 3(a) shows the mean normalized SERS spectrum of purified whole proteins obtained from normal (blue line, n=71) and NPC (red line, n=170) saliva samples. The prominent SERS peaks located at around 621, 642, 760, 854, 878, 935, 959, 1004, 1031, 1049, 1123, 1175, 1208, 1265, 1337, 1445, 1552, and 1684  cm1 were clearly observed in both normal and NPC saliva protein samples. These prominent SERS peaks could be tentatively assigned to explain the changes in biological constituents in saliva protein samples as shown in Table 2.3336 The strongest peaks at 760, 1004, 1265, 1445, and 1684  cm1 existed in the measured saliva protein SERS spectra. Compared with the SERS spectra of NPC saliva proteins, the SERS spectra of normal saliva proteins had higher intensities at 621, 935, and 1049  cm1 and lower intensities at 1004, 1031, 1208, 1123, and 1684  cm1. The comparisons of saliva proteins SERS spectral intensities between the NPC and normal groups could be viewed more clearly in Figs. 3(b) and 3(c). These differences in spectral intensities demonstrated that saliva proteins SERS has a potential role for NPC detection. For instance, the SERS bands of phenylalanine (1004 and 1031  cm1) were all related to the molecular stretching and bending mode of proteins, and they showed a lower SERS signal in the normal saliva protein samples than in the NPC saliva protein samples, indicating that an increase in the percentage of these proteins content in the total SERS-active in the NPC patients. Our group also observed that the content of phenylalanine was increased when associated with malignant transformation in cervical and NPC blood plasma by the SERS technique.2,18 However, the intensity of the saliva protein SERS spectrum exhibited decreased peaks at 935  cm1 (praline and valine) and 1049  cm1 (proteins) in NPC subjects, indicating that cancer patient saliva may be associated with a decreased concentration of these proteins. These results revealed that specific-protein changes between the NPC and normal groups could be detected by SERS, suggesting promising potential of saliva protein SERS for NPC screening.

Fig. 3

(a) Comparison of the mean normalized SERS spectrum of purified proteins obtained from normal saliva (blue line, n=71) and cancer saliva (red line, n=170), (b) difference spectrum calculated from the mean SERS spectra between the NPC and normal groups, and (c) comparison of the mean intensities of the selected peaks between cancer (red pillar) and normal (black pillar) samples.

JBO_22_10_105004_f003.png

Table 2

SERS peak positions and tentative vibrational mode assignments.

Peak position (cm−1)Vibrational modeMajor assignments
621CC twisting modePhenylalanine
642ν (CS)Tyrosine
760Ring breathing modeTyptophan
854Ring breathing modeTyrosine
878ν (CC)Hydro-oxyproline
935ν (CC)Proline
959ν (CC)α-Helix, proline, valine
1004νs (CC)Phenylalanine
1031δ (CH)Phenylalanine
1049ν (CO), ν (CN)Proteins
1123ν (CN)Proteins
1175δ (CH)Tyrosine
1208ν (CC6H5)Typtophan
1265ν (CN), δ (NH)Amide III, collagen
1337CH3CH2 waggingCollagen
1445δ (CH)Collagen, lipids
1552ν (CC)Typtophan
1684ν (CC)Amide I
Note: ν, stretching vibration; δ, bending vibration; and νs, symmetric stretch.

It should be noted that the simplistic peak intensities analysis above only uses limited information of SERS peaks, and more valuable diagnostic information contained in the SERS spectra has not been employed for spectral classification. Therefore, the PCA-LDA diagnostic algorithm was used to improve the diagnostic efficiency of the SERS technique by analyzing and classifying the saliva protein SERS spectra from NPC and normal subjects.

This sophisticated and robust diagnostic model based on PCA-LDA has been widely used for Raman spectral analysis in tissue, cell, saliva, and blood detection.27,37,38 In this study, PCA was used first to reduce intensity variables within the raw Raman spectrum of saliva protein into a few PCs. Then, three PCs (PC5, PC6, and PC7) were selected to be the most diagnostically significant (p<0.0001) as defined for discriminating normal and NPC groups by an independent sample t-test.

To further analyze saliva protein SERS data, all three diagnostically significant PCs were loaded into the LDA with the leave-one-out cross-validation method for generating an effective diagnostic model for saliva protein sample classification. Figure 4(a) shows the posterior probability plot of the healthy group (circles) and NPC group (triangles) based on the results of the PCA-LDA diagnostic model. As we can see, there are some plots overlapping in this figure. A discrimination threshold of 0.54 was employed for accurate discriminate between the healthy volunteers and NPC patients, yielding the diagnostic sensitivity, specificity, and accuracy of 70.7% (304/430), 70.3% (161/229), and 70.5% (465/659), respectively. To further evaluate the performance of saliva protein SERS for NPC diagnosis, an ROC curve was generated from the PCA-LDA data as shown in Fig. 4(b). The integration area under the ROC curve was 0.795. These results further indicated that saliva protein SERS technology combining the PCA-LDA diagnostic algorithm has the potential as a rapid and noninvasive diagnosis method for NPC detection.

Fig. 4

(a) Scatter plots of the posterior probability of NPC (red triangles) and normal (blue circles) saliva samples using the PCA-LDA algorithm. The separation threshold was 0.54. (b) The ROC curve of discrimination results for saliva protein SERS spectra. The integration area under the ROC curve was 0.795.

JBO_22_10_105004_f004.png

4.

Conclusions

A rapid and convenient saliva SERS analysis method was developed for NPC detection. Using the membrane protein purification method, high-quality and reproducibility SERS spectra of saliva proteins were obtained, making it possible to reveal specific changes in proteins associated with cancer transformation. Furthermore, a diagnostic sensitivity of 70.7% and specificity of 70.3% could be achieved by the PCA-LDA diagnostic algorithm for NPC identification, demonstrating promising potential of this saliva SERS analysis method to be a noninvasive body fluid test for clinical NPC screening.

Disclosures

The authors declare that there are no conflicts of interest related to this article.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (Nos. U1605253, 61210016, 61405036, and 61575043), the Innovation Team Development Plan by the Ministry of Education of China (No. IRT15R10), the National Natural Science Foundation of Fujian, China (Nos. 2017J01499 and 2015J01436), the Key Clinical Specialty Discipline Construction Program of Fujian (China), and the National Clinical Key Specialty Construction Program.

References

1. 

L. A. Torre et al., “Global cancer statistics, 2012,” CA-Cancer J. Clin., 65 (2), 87 –108 (2015). http://dx.doi.org/10.3322/caac.21262 Google Scholar

2. 

S. Feng et al., “Nasopharyngeal cancer detection based on blood plasma surface-enhanced Raman spectroscopy and multivariate analysis,” Biosens. Bioelectron., 25 (11), 2414 –2419 (2010). http://dx.doi.org/10.1016/j.bios.2010.03.033 BBIOE4 0956-5663 Google Scholar

3. 

J. Ferlay et al., “Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012,” Int. J. Cancer, 136 (5), E359 –E386 (2015). http://dx.doi.org/10.1002/ijc.29210 IJCNAW 1097-0215 Google Scholar

4. 

M. C. Yu and J.-M. Yuan, “Epidemiology of nasopharyngeal carcinoma,” Semin. Cancer Biol., 12 421 –429 (2002). SECBE7 1044-579X Google Scholar

5. 

L.-L. Tang et al., “Global trends in incidence and mortality of nasopharyngeal carcinoma,” Cancer Lett., 374 (1), 22 –30 (2016). http://dx.doi.org/10.1016/j.canlet.2016.01.040 CALEDQ 0304-3835 Google Scholar

6. 

X. Sun et al., “Long-term outcomes of intensity-modulated radiotherapy for 868 patients with nasopharyngeal carcinoma: an analysis of survival and treatment toxicities,” Radiother. Oncol., 110 (3), 398 –403 (2014). http://dx.doi.org/10.1016/j.radonc.2013.10.020 RAONDT 0167-8140 Google Scholar

7. 

Y. Chen et al., “Progress report of a randomized trial comparing long-term survival and late toxicity of concurrent chemoradiotherapy with adjuvant chemotherapy versus radiotherapy alone in patients with stage III to IVB nasopharyngeal carcinoma from endemic regions of China,” Cancer, 119 (12), 2230 –2238 (2013). http://dx.doi.org/10.1002/cncr.28049 CANCAR 0008-543X Google Scholar

8. 

A. King et al., “The impact of 18F-FDG PET/CT on assessment of nasopharyngeal carcinoma at diagnosis,” Br. J. Radiol., 81 (964), 291 –298 (2014). http://dx.doi.org/10.1259/bjr/73751469 Google Scholar

9. 

S.-X. Zhang et al., “Intravoxel incoherent motion MRI: emerging applications for nasopharyngeal carcinoma at the primary site,” Eur. Radiol., 24 (8), 1998 –2004 (2014). http://dx.doi.org/10.1007/s00330-014-3203-0 Google Scholar

10. 

H. Kwok et al., “Genomic diversity of Epstein-Barr virus genomes isolated from primary nasopharyngeal carcinoma biopsy samples,” J. Virol., 88 (18), 10662 –10672 (2014). http://dx.doi.org/10.1128/JVI.01665-14 JOVIAM 0022-538X Google Scholar

11. 

H. Kimura-Suda and T. Ito, “Bone quality characteristics obtained by Fourier transform infrared and Raman spectroscopic imaging,” J. Oral Biosci., 591 142 –145 (2017). http://dx.doi.org/10.1016/j.job.2017.04.002 Google Scholar

12. 

S. Nie and S. R. Emory, “Probing single molecules and single nanoparticles by surface-enhanced Raman scattering,” Science, 275 (5303), 1102 –1106 (1997). http://dx.doi.org/10.1126/science.275.5303.1102 SCIEAS 0036-8075 Google Scholar

13. 

S. Schlücker, “Surface-enhanced Raman spectroscopy: concepts and chemical applications,” Angew. Chem. Int. Ed., 53 (19), 4756 –4795 (2014). http://dx.doi.org/10.1002/anie.201205748 Google Scholar

14. 

S. Hong et al., “Live-cell stimulated Raman scattering imaging of alkyne-tagged biomolecules,” Angew. Chem. Int. Ed., 53 (23), 5827 –5831 (2014). http://dx.doi.org/10.1002/anie.201400328 Google Scholar

15. 

K. Kong et al., “Raman spectroscopy for medical diagnostics—from in-vitro biofluid assays to in-vivo cancer detection,” Adv. Drug Delivery Rev., 89 121 –134 (2015). http://dx.doi.org/10.1016/j.addr.2015.03.009 ADDREP 0169-409X Google Scholar

16. 

D. Lin et al., “Colorectal cancer detection by gold nanoparticle based surface-enhanced Raman spectroscopy of blood serum and statistical analysis,” Opt. Express, 19 (14), 13565 –13577 (2011). http://dx.doi.org/10.1364/OE.19.013565 OPEXFF 1094-4087 Google Scholar

17. 

J. Kneipp, H. Kneipp and K. Kneipp, “SERS—a single-molecule and nanoscale tool for bioanalytics,” Chem. Soc. Rev., 37 (5), 1052 –1060 (2008). http://dx.doi.org/10.1039/b708459p CSRVBR 0306-0012 Google Scholar

18. 

S. Feng et al., “Blood plasma surface-enhanced Raman spectroscopy for non-invasive optical detection of cervical cancer,” Analyst, 138 (14), 3967 –3974 (2013). http://dx.doi.org/10.1039/c3an36890d ANLYAG 0365-4885 Google Scholar

19. 

M. Vendrell et al., “Surface-enhanced Raman scattering in cancer detection and imaging,” Trends Biotechnol., 31 (4), 249 –257 (2013). http://dx.doi.org/10.1016/j.tibtech.2013.01.013 TRBIDM 0167-7799 Google Scholar

20. 

K. C. Bantz et al., “Recent progress in SERS biosensing,” Phys. Chem. Chem. Phys., 13 (24), 11551 –11567 (2011). http://dx.doi.org/10.1039/c0cp01841d PPCPFQ 1463-9076 Google Scholar

21. 

P. Dowling et al., “Analysis of the saliva proteome from patients with head and neck squamous cell carcinoma reveals differences in abundance levels of proteins associated with tumour progression and metastasis,” J. Proteomics, 71 (2), 168 –175 (2008). http://dx.doi.org/10.1016/j.jprot.2008.04.004 Google Scholar

22. 

M. Castagnola et al., “Potential applications of human saliva as diagnostic fluid,” Acta Otorhinolaryngol. Ital., 31 (6), 347 –357 (2011). AOITDU 0392-100X Google Scholar

23. 

K. Thomadaki et al., “Whole-saliva proteolysis and its impact on salivary diagnostics,” J. Dent. Res., 90 (11), 1325 –1330 (2011). http://dx.doi.org/10.1177/0022034511420721 JDREAF 0022-0345 Google Scholar

24. 

L. R. Bigler et al., “Salivary biomarkers for the detection of malignant tumors that are remote from the oral cavity,” Clin. Lab. Med., 29 (1), 71 –85 (2009). http://dx.doi.org/10.1016/j.cll.2009.01.004 CLMED6 0272-2712 Google Scholar

25. 

K. W. Kho et al., “Surface enhanced Raman spectroscopic (SERS) study of saliva in the early detection of oral cancer,” Proc. SPIE, 5702 84 –91 (2005). http://dx.doi.org/10.1117/12.590142 Google Scholar

26. 

X. Li, T. Yang and J. Lin, “Spectral analysis of human saliva for detection of lung cancer using surface-enhanced Raman spectroscopy,” J. Biomed. Opt., 17 (3), 037003 (2012). http://dx.doi.org/10.1117/1.JBO.17.3.037003 JBOPFO 1083-3668 Google Scholar

27. 

S. Qiu et al., “Non-invasive detection of nasopharyngeal carcinoma using saliva surface-enhanced Raman spectroscopy,” Oncol. Lett., 11 (1), 884 –890 (2016). http://dx.doi.org/10.3892/ol.2015.3969 Google Scholar

28. 

S.-M. Li et al., “Rapid microwave-assisted preparation and characterization of cellulose-silver nanocomposites,” Carbohydr. Polym., 83 (2), 422 –429 (2011). http://dx.doi.org/10.1016/j.carbpol.2010.08.003 CAPOD8 0144-8617 Google Scholar

29. 

J. Zhao et al., “Automated autofluorescence background subtraction algorithm for biomedical Raman spectroscopy,” Appl. Spectrosc., 61 (11), 1225 –1232 (2007). http://dx.doi.org/10.1366/000370207782597003 APSPA4 0003-7028 Google Scholar

30. 

U. Demšar et al., “Principal component analysis on spatial data: an overview,” Ann. Assoc. Am. Geogr., 103 (1), 106 –128 (2013). http://dx.doi.org/10.1080/00045608.2012.689236 AAAGAK Google Scholar

31. 

P. D. V. de Almeida et al., “Saliva composition and functions: a comprehensive review,” J. Contemp. Dent., 9 (3), 72 –80 (2008). Google Scholar

32. 

S. Singh et al., “Saliva as a prediction tool for dental caries: an in vivo study,” J. Oral Biol. Craniofac. Res., 5 (2), 59 –64 (2015). http://dx.doi.org/10.1016/j.jobcr.2015.05.001 Google Scholar

33. 

L.-J. Xu et al., “Label-free detection of native proteins by surface-enhanced Raman spectroscopy using iodide-modified nanoparticles,” Anal. Chem., 86 (4), 2238 –2245 (2014). http://dx.doi.org/10.1021/ac403974n ANCHAM 0003-2700 Google Scholar

34. 

D. Kurouski et al., “Amide I vibrational mode suppression in surface (SERS) and tip (TERS) enhanced Raman spectra of protein specimens,” Analyst, 138 (6), 1665 –1673 (2013). http://dx.doi.org/10.1039/c2an36478f ANLYAG 0365-4885 Google Scholar

35. 

Z. Huang et al., “Near-infrared Raman spectroscopy for optical diagnosis of lung cancer,” Int. J. Cancer, 107 (6), 1047 –1052 (2003). http://dx.doi.org/10.1002/(ISSN)1097-0215 IJCNAW 1097-0215 Google Scholar

36. 

S. Teh et al., “Diagnostic potential of near-infrared Raman spectroscopy in the stomach: differentiating dysplasia from normal tissue,” Br. J. Cancer, 98 (2), 457 –465 (2008). http://dx.doi.org/10.1038/sj.bjc.6604176 BJCAAI 0007-0920 Google Scholar

37. 

D. Lin et al., “Label-free blood plasma test based on surface-enhanced Raman scattering for tumor stages detection in nasopharyngeal cancer,” Sci. Rep., 4 4751 (2014). http://dx.doi.org/10.1038/srep04751 SRCEC3 2045-2322 Google Scholar

38. 

Z. Li et al., “Raman microspectroscopy as a diagnostic tool to study single living nasopharyngeal carcinoma cell lines,” Biochem. Cell Biol., 91 (3), 182 –186 (2013). http://dx.doi.org/10.1139/bcb-2012-0024 BCBIEQ 0829-8211 Google Scholar

Biography

Xueliang Lin received his BS degree in physics from Longyan University in 2015. He is currently working at the Institute of Laser and Optoelectronics Technology and working toward his master’s degree in optics at Fujian Normal University, China. His research focuses on the application of surfaced-enhanced Raman spectroscopy (SERS) in the diagnosis of nasopharyngeal carcinoma (NPC).

Duo Lin received his master’s degree in physical electronics from Fujian Normal University in 2012. He is currently working at the Institute of Laser and Optoelectronics Technology and working toward his PhD in optics at Fujian Normal University, China. His research focuses on the application of SERS in biomedical diagnosis.

Xiaosong Ge received his BS degree communication engineering from Changzhou University in 2015. He is currently working at the Institute of Laser and Optoelectronics Technology and working toward his master’s degree in physical electronics at Fujian Normal University, China. His research focuses on the application of SERS in biomedical diagnosis.

Sufang Qiu received her PhD from Fujian Medical University in 2016. Currently, she is a chief physician of Fujian Tumor Hospital, China. She is an oncologist and a master tutor. Her research interest focuses on radiation therapy and diagnosis of NPC.

Shangyuan Feng received his PhD from Fujian Normal University in 2011. He started a postdoctoral position at BC Cancer Agency, Canada, in 2013. Currently, he is an associate professor at the School of Optoelectronics and Information Engineering, Fujian Normal University, China. His research interest focuses on the application of SERS in biomedical diagnosis.

Rong Chen was the director of Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education. Currently, he is a doctoral tutor at the School of Optoelectronics and Information Engineering, Fujian Normal University, China. His research interest is the application of Raman spectral to detect NPC.

© 2017 Society of Photo-Optical Instrumentation Engineers (SPIE) 1083-3668/2017/$25.00 © 2017 SPIE
Xueliang Lin, Duo Lin, Xiaosong Ge, Sufang Qiu, Shangyuan Feng, and Rong Chen "Noninvasive detection of nasopharyngeal carcinoma based on saliva proteins using surface-enhanced Raman spectroscopy," Journal of Biomedical Optics 22(10), 105004 (13 October 2017). https://doi.org/10.1117/1.JBO.22.10.105004
Received: 27 June 2017; Accepted: 21 September 2017; Published: 13 October 2017
Lens.org Logo
CITATIONS
Cited by 23 scholarly publications.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Proteins

Surface enhanced Raman spectroscopy

Diagnostics

Cancer

Silver

Raman spectroscopy

Statistical analysis

Back to Top