Next Article in Journal
Odorant Binding Causes Cytoskeletal Rearrangement, Leading to Detectable Changes in Endothelial and Epithelial Barrier Function and Micromotion
Next Article in Special Issue
Plasmonic Sensors: A New Frontier in Nanotechnology
Previous Article in Journal
A Minireview for Recent Development of Nanomaterial-Based Detection of Antibiotics
Previous Article in Special Issue
Screening and Evaluation of Thiamethoxam Aptamer Based on Pressurized GO-SELEX and Its Sensor Application
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Trends in SERS-Based Plasmonic Sensors for Disease Diagnostics, Biomolecules Detection, and Machine Learning Techniques

Advanced Centre of Research in High Energy Materials (ACRHEM), DRDO Industry Academia—Centre of Excellence (DIA-COE), University of Hyderabad, Hyderabad 500046, Telangana, India
*
Author to whom correspondence should be addressed.
Biosensors 2023, 13(3), 328; https://doi.org/10.3390/bios13030328
Submission received: 31 January 2023 / Revised: 20 February 2023 / Accepted: 24 February 2023 / Published: 27 February 2023
(This article belongs to the Special Issue Plasmonic Sensors: A New Frontier in Nanotechnology)

Abstract

:
Surface-enhanced Raman spectroscopy/scattering (SERS) has evolved into a popular tool for applications in biology and medicine owing to its ease-of-use, non-destructive, and label-free approach. Advances in plasmonics and instrumentation have enabled the realization of SERS’s full potential for the trace detection of biomolecules, disease diagnostics, and monitoring. We provide a brief review on the recent developments in the SERS technique for biosensing applications, with a particular focus on machine learning techniques used for the same. Initially, the article discusses the need for plasmonic sensors in biology and the advantage of SERS over existing techniques. In the later sections, the applications are organized as SERS-based biosensing for disease diagnosis focusing on cancer identification and respiratory diseases, including the recent SARS-CoV-2 detection. We then discuss progress in sensing microorganisms, such as bacteria, with a particular focus on plasmonic sensors for detecting biohazardous materials in view of homeland security. At the end of the article, we focus on machine learning techniques for the (a) identification, (b) classification, and (c) quantification in SERS for biology applications. The review covers the work from 2010 onwards, and the language is simplified to suit the needs of the interdisciplinary audience.

Graphical Abstract

1. Introduction

Plasmonics is the study of electron oscillations in metal nanostructures and their interaction with electromagnetic radiation. Since its conception in the 1950s, researchers have been interested in studying the fundamentals of the effects of shape, surrounding medium, material, and their interaction with light of different wavelengths [1]. With this well-established knowledge, plasmonics is witnessing an enormous potential for applications in different fields, including forensics [2]; environmental safety [3]; biosensing [4,5,6,7,8,9,10,11], e.g., SARS-CoV-2 detection [12]; and homeland security [13]. The applications of plasmonics majorly rely on surface plasmon resonance (SPR) or localized surface plasmon resonance (LSPR) effects [14]. Some of the significant techniques that were developed using these include higher-order harmonic generation, microscopy, drug delivery, photovoltaics, surface-enhanced Raman spectroscopy (SERS) and fluorescence, and surface-enhanced infrared absorption spectroscopy (SEIAS) and waveguides. The use of plasmonics in these techniques has significantly improved their efficiency over existing conventional techniques, offering flexibility, signal enhancement, and ease of use [15]. Advents in plasmonics have led to the emergence of SERS with impressive signal enhancements over traditional Raman spectroscopy [16]. SERS-based sensing is being widely used for the trace detection of different molecules, such as explosives [17], pesticides [18,19], food adulterants [20,21], drugs [22], biomolecules [23,24,25,26,27], medicine [28,29,30], and microorganisms [31].
SERS typically utilizes localized surface plasmon resonances in metal nanostructures to enhance the weak Raman signal significantly. The phenomenon was first observed by Fleischmann in 1974 while studying pyridine adsorbed on a roughened silver electrode [32]. However, the enhancement was attributed to increased surface area for adsorption. It took further experiments in 1977 by two independent groups, Jeanmaire and van Duyne [33] and Albrecht and Creighton [34], to understand the origin of the enhancement. Now it is established that the enhancement predominantly comes from two mechanisms: electromagnetic enhancement (EE) and chemical enhancement (CE) [35]. The electromagnetic enhancement in SERS is a two-step process, and the total enhancement is multiplicative. When a molecule of interest is in the vicinity of a plasmonic nanostructure, it experiences an enhanced field called local field enhancement (LFE). The molecule then radiates with increased efficiency, referred to as radiation enhancement [36,37]. In addition, there is chemical enhancement which occurs because of charge-transfer mechanisms between nanoparticles and the analyte. Figure 1 summarizes the two enhancement mechanisms in SERS. The type of the plasmonic material, choice of wavelength, surface coverage of the molecules, and concentration of the analyte are the factors that influence SERS’s efficiency [38]. This technique is label-free, rapid, non-destructive, and water compatible and offers the fingerprint of the molecule, making it suitable for numerous applications. Nobel metals such as Au, Ag, and Cu and their alloys are the widely used materials for SERS for their tunability in the visible and IR region, inertness, sensitivity, and compatibility [39,40]. Despite the superior performance of Ag owing to its high-quality resonance in the visible region, Au is the preferred material, as it is known to be biocompatible and non-reactive in an oxygen atmosphere. The near-field enhancement in SERS is dependent on the shape and size of the nanostructures, in addition to the distance between the nanoparticles and distribution of probe molecules around the nanoparticles [41]. The different morphologies of nanoparticles, such as core–shell, rods, spherical, triangular, stars, and nanopyramids, are synthesized by widely reported chemical routes in bottom-up or top-down approaches [42]. Anisotropic nanostructures such as dendrites, rods, stars, and triangle are considered highly desirable for SERS since they enable lower detection limits owing to the lightening-rod effect [43,44]. The performance of SERS is also dependent on the choice of wavelength, and most biological tissues are transparent in the IR region, making it a preferred choice [45]. Recently, there is also growing interest in the UV and deep UV SERS for applications concerning biomolecules such as amino acids and DNA bases because they have electronic transitions in the UV region [39].
With a growing population and, consequently, the diseases worldwide, there is a need to develop point-of-care (POC) devices that are easy to use, reliable, rapid, and low cost. Over the years, SERS has been proven to possess all of these advantages, including trace detection with sub-picomolar sensitivity. Particularly, there are many reasons for the surge of using SERS for biosensing. Firstly, given the low scattering cross-section of water, SERS is extremely compatible with liquid samples, paving the way for use in biology applications, including liquid biopsy [46,47]. SERS has been widely used for disease diagnosis using urine, blood, serum, plasma, saliva, breath, and tear samples, establishing its compatibility. Measurements in SERS can be performed using liquids, gases, solids, and powders, unlike traditional tests. Secondly, SERS gives specific molecular information, which is often a vibrational fingerprint of the molecule or cell under study. Biomarkers that are Raman active are extensively used for the identification of different diseases, using SERS [48]. Frequently, when the variations are unrecognizable to the human eye, machine learning techniques are used to extract the patterns and discriminate the samples [49]. This was successfully used to classify normal and cancer cells [50], identify microorganism species [51], and monitor disease progression [52]. Thirdly, SERS is a rapid technique, can accomplish trace detection, and has a test time of three to five minutes [53]. Combined with recent developments in flexible SERS sensors, it also offers easy sample-collection methods, such as swabbing from an uneven surface [54]. Lastly, advances in portable instrumentation and low-cost lasers leveraged the usage of SERS for real-world applications [55]. The easy availability of IR lasers that have a low damage threshold with biology samples, as well as quench fluorescence, has favored the development of SERS for biosensing. All of these advantages have made SERS a popular choice for biosensing recently.
There have been many review articles concerning the applications of plasmonics for biosensing and biosensors over the years. Salazar et al. and Han et al. reviewed different techniques, including LSPR, Chiral Plasmonic Biosensors, Magnetoplasmonic Biosensors, and Quantum Plasmonics Biosensors [56,57]. Anand et al. published a comprehensive review on plasmonic biosensors for the detection of viruses, with a special focus on COVID-19. They have focused on LSPR, SPR, SERS, SEF, and SEIAS techniques [58]. There are reviews and book chapters elaborating specifically on various SPR [59,60,61] and LSPR [62] techniques that are currently being used for biosensing. Similarly, Sarah et al. focused exclusively on LSPR techniques and associated challenges in the detection [63]. Alexandre reviewed the future of plasmonic biosensing with a goal of single-molecule and single-particle sensing [64]. Juanjuan et al. discussed on the challenges and future of using plasmonic materials for point-of-need applications [65]. However, although significant work has been performed using SERS for biology applications, no reviews of the literature can be found in this area. Here, we present a review of the work conducted in SERS for biosensing and the recent developments, with a special focus on machine learning techniques that are being used for the same. The article covers work from 2010 onwards and is organized into different sections, as shown in the index. Figure 2a,b illustrate the statistics of publications in different areas discussed in this review article. The data presented in the figure indicate that there is a growing interest in the usage of SERS for cancer-related applications and the usage of machine learning techniques for biosensing using SERS. There is also relative growth in using SERS for respiratory disease diagnosis recently owing to the COVID-19 situation.

2. SERS for Disease Diagnosis

With growing zoonotic diseases, cancers, diabetes, and other ailments, there is a pressing need to develop low-cost and POC identification techniques. Early and rapid diagnosis is the key to saving a life and prevent the rapid transmission of diseases. A trace detection technique such as SERS will aid in tracking the minute changes in cells or biomarkers, thus enabling early diagnosis. SERS is being extensively used for the same in both labeled and label-free approaches, often targeting specific biomarkers of the disease expression [30]. In the label-free approach, the sample is directly studied in contact with the plasmonic material, whereas in the labeled approach, a Raman reporter, such as fluorophores, antibodies, or ligands, is attached to the sample for detection and imaging [66,67]. Different biomarkers, such as proteins, antibodies, miRNAs, exosomes, and DNA, are used as indicators for the presence of the disease. In our observation, where full cells, tissues, or body fluids are studied, a machine learning algorithm is used hand in hand for accurate identification. SERS has been used for the detection of conditions such as Alzheimer’s [68,69,70,71], PCOS [72], diabetes [73,74], inflammation [74], Crohn’s disease [75], and single Hb molecule [76], to name a few. Here we review the progress on SERS for the diagnosis of (a) cancer, paying special attention to lung and breast cancer, as they are the leading causes of deaths due to cancer; and (b) respiratory viruses, including COVID-19.

2.1. Cancer Diagnosis and Theranostics

Cancer is the new pandemic and a leading cause of deaths in the modern world [77]. There is an increase in the incidence of various types of cancers, including mouth, gastric, lungs, ovaries, skin, and blood cancer. Numerous factors, such as environment, diet, lifestyle, and smoking, can trigger cancer. The early diagnosis of cancer is extremely important, as it is lifesaving with existing treatment protocols. Conventional cancer diagnosis is often performed using imaging techniques such as X-ray, computerized tomography scan (CT), positron emission tomography (PET), ultrasound, and magnetic resonance imaging (MRI). These techniques are often destructive, posing the risk of radiation ionization, and are often not compatible with patients with pre-existing conditions and medical devices such as pacemakers [78]. These are also expensive, involve sophisticated instruments, are time-consuming, and are often performed with multiple tests to avoid ambiguity [79]. Recently, there has been an increase in using plasmonic biosensing for cancer diagnosis and therapy, with review articles summarizing the progress in the same [80,81,82,83,84,85,86]. They are established to be minimally invasive, rapid, low cost, and offer point-of-care testing [87,88]. Of all plasmonic-based detection techniques, SERS is being extensively used for cancer identification, monitoring, and other theranostics, including imaging and chemo/photothermal therapy [89,90,91,92,93,94,95,96]. Figure 2 also indicates the growing interest in the last decade for the use of SERS-based plasmonic techniques for cancer diagnosis. SERS facilitates liquid biopsy [96] by using urine, saliva, and serum, thus making it low cost and enabling easier frequent sampling compared to the existing tissue-biopsy techniques, which are often destructive [97]. Different cancer biomarkers, such as miRNA [98,99], proteins, exosomes [100,101], circulating tumor DNA (ctDNA), genes [102], peptides [103], and blood plasma [104], are studied using SERS for disease identification. SERS tags that specifically bind to the targets under study are widely used for analyzing cancer samples [105,106,107,108,109]. Machine learning algorithms are used to analyze complex patterns and recognize buried signals overcoming noise from undesirable constituents of cells and other bio-fluids. Here, we focus only on SERS-based plasmonic biosensing for cancer-related applications in recent times, focusing on lung and breast cancers.

2.1.1. Lung Cancer

Lung cancer is known as the most fatal and frequently diagnosed cancer of all [110]. The cited report projected 2.89 million cases of lung cancer by 2030. Smoking, the presence of carcinogenic substances in the environment, and lifestyle are considered to be the main causes of lung cancer [88]. There are two kinds of lung cancers, non-small cell (NSCLC) and small cell lung cancer (SCLC). NSCLC is the most common kind of lung cancer, accounting for 80% of the cases. SCLC is the most fatal and fast-spreading cancer, and it is often diagnosed only at the later stages. SERS has been successfully used for the diagnosis of both kinds of lung cancers, with a prospect of developing point-of-care and rapid testing.
Using DNA-based complexes as SERS tags and miRNA as a biomarker, Mao et al. have developed a lateral-flow-assay-based SERS substrate for the rapid detection and quantification of lung cancer biomarkers in less than 30 min [111]. Two types of biomarkers, miR-21 and miR-196a-5p, were detected simultaneously and with comparable accuracy with the existing qRT-PCR techniques. Similar studies were carried out with a flexible filter paper substrate [112] and with different biomarkers [113,114]. They extended their studies with ctDNA as the biomarker, thus implying the versatility of SERS [115]. Similarly, miRNA has been used for the detection of lung cancer by using circular exponential amplification reaction (EXPAR)-based SERS [116]. With a combination of asymmetric PCR and SERS, in regard to mutation genes in ctDNA, Guo et al. achieved a highly specific (100%) and sensitive (75%) lung-cancer-detection method in blood samples. Asymmetric PCA was performed to obtain single-stranded DNA, followed by the SERS-based detection using specifically labeled Au substrates [117]. With exosomes as biomarkers, lung cancers at different stages were identified accurately (~90%) by using SERS and a deep learning algorithm to classify healthy and malignant samples, exhibiting potential for early diagnosis, as shown in Figure 3 [118]. Serum samples of normal and lung-cancer patients were analyzed using PCA and PLS analysis to discriminate and identify the cancer samples with SERS spectra and achieved an accuracy of 92% [119]. Similarly, using a core-satellite type of plasmonic materials and SERS, serum samples of healthy, benign, and malignant cases of lung cancers were classified with a combination of principal component analysis (PCA) and support vector machines (SVMs) [120]. CtDNA-based identification of lung cancer using a DNA-rN1-DNA-mediated SERS frequency shift method was developed to achieve sub-femtomolar sensitivity [121]. Similarly, exosomes derived from bronchoalveolar fluid were used for the detection [122]. Whole-exosome SERS spectra have been analyzed using PCA to classify lung cancer and normal samples with 95.3% sensitivity and 97.3% specificity [123]. Lung-cancer biomarkers (aldehydes) and cells were identified rapidly using renewable porous CuFeSe2/Au nanostructures achieving an LOD of 1 ppb [124]. Challenging gaseous biomarkers called volatile organic compounds, which serve as indicators for lung cancer, were detected using ZIF-8-coated gold superparticles for the sensitive identification of lung cancer [125]. Pleural effusions of lung cancer and normal samples were studied using SERS and machine learning techniques to achieve a classification accuracy of 85% [126]. A combination of PCA and LDA has been used to classify lung cancer and normal samples with SERS analysis of serum samples, achieving sensitivity and specificity of 100% and 90%, respectively [127]. The same technique was used for SERS-based classification of lung-cancer-tissue slices [128]. Using common protein carcinoembryonic antigen (CEA) and a-fetoprotein (AFP) as biomarkers, SERS-based detection was performed for the diagnosis of lung cancer [129]. A non-destructive photothermal therapy targeting lung cancer cells (A549 cells) was developed using NIR radiation and Ag-Au shell–core structures were used for the SERS-based detection of the A549 cells. These nanostructures are highly specific and have different affinities for cancerous and non-cancerous cells, thus helping in tagging the cells. Based on the SERS activity of R6G molecules, the detection and phototherapy can be monitored [130]. Similar studies were carried out using reduced graphene oxide plasmonic substrates [131]. A multivariate analysis (SVM and PCA) of SERS data was used to identify and classify different types of lung cancers with an accuracy of 95% [132]. Choosing aldehydes in exhaled breath as biomarkers, highly sensitive, portable detection was performed, achieving LOD of 1.35 nM [133]. Chemometric techniques coupled with slippery liquid-infused porous surface-enhanced Raman spectroscopy were used for concentrating blood samples in a small area and thus enhancing the SERS signal for trace detection [134]. A gap mode plasmonic SERS substrate with a combination of Ag nanocubes and Au nanorods was used for the identification of lung-cancer-related exosomes [135]. A SERS analysis of saliva samples was performed to classify healthy and cancerous samples, using SVM and random forest, with a sensitivity of 95% and 97%, respectively [136]. With adenosine as a biomarker, urine samples were analyzed using SERS with Fe3O4/Au/Ag-based substrates, achieving good reproducibility, stability, and sensitivity of 10−10 M [137].

2.1.2. Breast Cancer

Breast cancer is considered to be the second leading cause of death among women after lung cancer [138]. Breast cancer is often diagnosed by a mammogram, ultrasound, MRI, or biopsy. Furthermore, it is often concluded by a histopathological test, which is unfortunately time-consuming and is highly prone to human interpretation error. In addition to identification using urine, serum [139], and tear samples [140], SERS has also been used to understand the drug carrier mechanism [141] and classification of different stages of breast cancer [142], as shown in Figure 4.
Using the epidermal growth factor receptor as a biomarker, a gold-nanorods-based SERS tool that can identify and image the spatial and temporal distribution of breast cancer cells was developed by Xiao et al. [145]. Sialic acid, with its specificity towards a phenyboronic-acid-based nanoprobe, was used as biomarker for identification and imaging of breast cancer in human cells and saliva [146,147]. The miRNA of breast cancer was detected with a high sensitivity of 10−10 M, using a hybrid SERS substrate of GaN nanostructures with Au/Ag [148]. Functionalized SERS substrates with specific tags were used for the simultaneous isolation and detection of breast cancer cell lines [149]. Zheng et al. developed a SERS-based microfluidic channel for detection and quantification of prominent breast cancer biomarkers in real samples [150]. A combination of SERS and electrochemical biosensor was developed to monitor the drug response of DNA associated with breast cancer cells [52]. Hameed et al. have worked on fabricating anisotropic gold nano-stars that showed specific affinity to breast cancer cells compared to normal cells, aiding in the detection of the same [151]. Multiple SERS tags were used for understanding the drug-carrier mechanisms in breast cancer cells for the antineoplastic drug tamoxifen [141]. Similar studies were carried out with estrogen receptor alpha (ER-α) as the biomarker [152]. ER-α-based SERS has also been used for understanding cellular uptake mechanisms in breast cancer [153]. Labeled hollow silica-encapsulated gold nano-spheres were used for identifying and quantifying breast cancer biomarkers [154]. Choi et al. developed SERS nanotags by using Ag-Au hollow nanospheres that are durable, reproducible, and sensitive for the detection of various biomarkers for SERS [155]. A SERS-based 3D holograph was developed to detect and quantify nine miRNAs corresponding to breast cancer. Hairpin-like DNA was used as SERS tags along with Raman reporters for each miRNA that are spatially separated on the SERS substrate [156]. Similar studies were carried out by Weng et al. [157], Li et al. [158], and Lee et al. [159]. A ratio-type method was developed for the discrimination of breast cancer and non-cancer cells, using the SERS technique. A plasmonic material with Rh6G as a tag for breast cancer biomarker (MMP-2), along with a standard (2-NT), was used for analyzing the live cells based on the ratio of SERS signals in standard and R6G [160]. Li et al. also performed similar studies [161] and was also used for quantitative molecular phenotyping in a different study [162]. Recently, ratiometric SERS has been used for the identification of breast cancer using Au@Ag and GO nanostructures [163]. Cell suspensions of normal and breast cancer cells were analyzed using SERS coupled with Random Forest classifier to understand the differences. It was found that breast cancer cells have high cholesterol, lipids, proteins, and nucleic acids relative to the normal cells, and the classification accuracy was nearly 78% [164]. A comparison was made between SERS and Raman performance for the classification of different stages of breast cancers, using PCA and PLS-LDA, and found that SERS leads to better accuracy (94%) relative to the Raman (83%) method [142]. PLS-LDA was also used by Zheng et al. for the identification of breast-cancer biomarkers, using HAp [165]. PLS-SVM, PLS-LDA, and PCA-LDA were used for the classification of breast cancer and the normal group [166,167,168,169,170,171,172,173,174,175,176]. An exosome-based CNN model was developed for the classification of breast cancer and normal samples, with an accuracy of 95% [177]. A systematic analysis of SERS spectra obtained from urine and serum samples was performed, and it was found that the urine samples demonstrated better accuracy in the classification [170]. Biomarkers tracking the epithelial–mesenchymal transition in the plasma samples of breast cancer cells were identified using SERS immunoassay [178]. SERS-based cancer cell imaging was performed using gold nanoparticles based on the specific affinity of phenylalanine [179]. Different methods for the preparation of plasmonic Ag nanoparticles and their effects in SERS signal were discussed by Beata et al. [180]. Photothermal therapy and SERS-based identification of breast cancer were performed using gold nanorods [181] and gold nanobipyramids [182]. NIR and SERS-based phototherapy and detection were also performed [183]. A three-in-one tool consisting of photoacoustic imaging, thermosurgery, and SERS was developed to address the concern of residual microtumors in breast cancer [144]. By combining artificial intelligence and SERS, researchers developed a label-free detection method of breast cancer exosomes with 100% accuracy. This was also used to assess the outcomes of the surgeries [143]. Au/HCP-PS nanospheres were used for the SERS-based detection of breast cancer, using tears from asymptomatic patients, along with chemometric analysis [140]. A Pt-based SERS template was developed using cost-effective methods for the detection of breast cancer exosomes that achieved a sensitivity of 83.3% and a specificity of 95.8% [184]. A combination of 2D graphene and plasmonic gold nanostars was used for trace identification of exosomes [185]. There are many reports of researchers using exosomes as biomarkers for the identification of cancer, using SERS [186]. A highly sensitive (EF ~105) and reproducible (2.7%) method was developed using Au@Ag nanospheres for the detection of breast-cancer-based extracellular metabolites [187]. Systematic experiments were performed to understand the effects of laser power and acquisition time on the reproducibility in immune-SERS microscopy and found that a longer acquisition time and higher laser power lead to poor reproducibility [188].

2.1.3. Miscellaneous

With the mechanism for detection being the same, SERS has been extensively used for the detection of several other cancers, including gastric [189], oral, liver, ovarian [190,191], and prostate cancers [192,193,194,195,196,197]. Gastric cancer diagnoses have been performed using different plasmonic materials by analyzing SERS spectra of serum samples [198,199,200], blood plasma [201], exosomes [202], extracellular vesicles [203], telomerase [204], saliva [205], and ctDNA [206]. In a breakthrough study, a breath analysis based on SERS was performed to identify different stages of gastric cancers by analyzing the Raman bands [207,208]. Different chemometric techniques such as PCA [209,210], PCA-LDA [201,211,212,213], SVM [214], ANN [215], and PCA-QDA [216] were also used for classification and identification of gastric cancers. Li et al. used a combination of classification algorithms, such as PCA-LDA, PCA-SVM, and PCA-CART, for identifying gastric diseases in serum samples [217]. Blood samples from healthy and normal patients were analyzed for different cancers, such as liver cancer, colonic cancer, esophageal cancer, nasopharyngeal cancer, gastric cancer along with PCA-SVM and achieved an accuracy of 96% [218]. Based on the SERS profiling of urine samples, bladder cancer was studied using machine learning algorithms with miRNA as a biomarker [219]. PCA, random forest, KNN, and naive Bayes algorithms were used for the identification of renal cancer, with the SERS profiling of serum samples achieving accuracy greater than 75% [139]. Taking advantage of the coffee-ring effect, the serum samples of lung and prostate cancer patients were identified with 100% accuracy, using PLS-SVM algorithms on SERS data [220]. Gaussian-based CNNs were used for the same application elsewhere [221]. Recently, there was a review article specifically focusing on SERS-based biosensing for liver cancer detection applications [222]. Zhang et al. elaborated on the existing literature for oral cancer diagnosis and therapy with gold nanoparticles, highlighting the current progress and challenges [223]. A similar review article was also published for the case of ovarian cancer [224]. Oral cancer was studied using saliva samples and the miRNA of normal and cancer patients with the SERS technique [225,226]. SVM in combination with SERS has been used for the early detection of oral cancer among patients using serum and saliva samples and achieved an accuracy of 80% [227]. Prostate cancer has been extensively studied and successfully identified using different techniques, such as serum analysis combined with PCA-SVM [228]; detection of prostate specific antigens [229,230,231,232]; EVs combined with CNN [230]; miRNAs [233]; different multivariate techniques, e.g., PCA-LDA and PCA-SVM [234]; and urine profiling [235].

2.2. SARS-CoV-2 and Other Respiratory Diseases

With the onset of the pandemic and the fast-spreading variants, there was a need to rapidly identify, detect, and quarantine the infected population. Surveying the presence of antibodies in large populations, often called a serological survey, was important to access the percentage of population infected and to monitor community transmission [236]. The dominant existing technique for the identification of SARS-CoV was PCR, which relies on analyzing the genetic material of the virus [237]. However, the test is expensive, thus preventing wide usage and also is time consuming. The Raman spectrum of a whole organism, including viruses, is contributed to by the proteins, carbohydrates, and nucleic acids that make up the organism [238]. The expression of these building blocks is controlled by the genetic material of the organism, hence helping in the unique identification [239]. SERS has enabled trace, point-of-care (POC), sample-collection-friendly, rapid, flexible, and cost-effective covid detection alternatives with the use of diverse nanomaterials [10,240,241,242]. In addition, both portable and handheld systems have indeed enabled point-of-care testing based on Raman spectroscopy [56,243]. SERS has also been widely used for the detection of other respiratory zoonotic diseases, such as H1N1, H7N9, H3N2, and H5N1; and other coronaviruses, such as MERS-CoV [244,245]. Often, machine learning algorithms are used in combination to enable the identification of patterns that are not apparent to the human eye [246,247]. The availability of large data and the ease of collection have accelerated the potential of machine learning algorithms in identifying viruses and their variants with reliable accuracies for POC devices [248,249]. In addition to trace identification, SERS has also enabled quantification of viral load to access the severity of the infection [250,251].
SERS in combination with LDA has been used for the rapid (2 min) identification of respiratory viruses, including SARS-CoV-2, human adenovirus type 7, and H1N1, using label-free silver nanoparticles [252]. Fe3O4@Ag nanoparticles tagged with specific antibodies were used for the detection of adenovirus and influenza virus [253]. Eleven different respiratory pathogens were identified using SERS, with nanoparticles tagged with nucleic acids achieving remarkable LODs in the sub-picomolar range [254]. Gold nanoparticles functionalized with a specific enzyme were used for the detection of S protein expressed by the COVID-19 viruses with SERS-based sensing in water [255]. Trace S protein detection has also been performed with SERS substrates enabling both chemical and electromagnetic enhancement [256] and using DNA-aptamer-based substrates, achieving a 0.7 fg mL−1 LOD [257]. Influenza-infected cells were identified based on proteins, using SERS and PCA [258]. Influenza and covid viruses were detected in human nasal fluid and saliva, using SERS [259], and also in untreated saliva [260]. A portable breath analyzer for covid detection based on the presence of organic volatile compounds was developed, achieving a sensitivity greater than 95% with less than 5 min of detection time [53]. A lateral-flow-immunoassay-based SERS was proposed for the quantitative detection of SARS-CoV-2 [261]. Similar work was performed for the trace detection of SARS-CoV-2 antibodies and spike proteins [262,263,264]. Li et al. optimized the silver nanostructures to increase the LOD for SARS-CoV-2 detection [265]. In a unique study, Kim et al. studied the efficacy of the Oxford–AstraZeneca vaccine by using SERS studies on tear samples and achieved excellent reproducibility and LOD in the femtomolar regime [266]. Machine learning algorithms such as PCA and SVM were used for the classification of normal and SARS-CoV-2 saliva samples with SERS data, with an accuracy of 95% [267]. Different respiratory viruses and their variants were identified using a silver-nanorods-based SERS sensor [268]. Different respiratory syncytial viruses have been identified and classified using SERS and classification algorithms such as PCA and HCA [269]. A deep-learning-based on-site SERS detection was developed to detect the SARS-CoV-2 virus based on the spike protein with 87% accuracy. This work also studied Raman modes of the spike protein theoretically and established a database [270]. Different variants of the SARS-CoV-2 virus, including wild-type, Alpha, Delta, and Omicron, were successfully identified using specific antibody-tagged 3D porous Ag-based SERS substrates [271]. SERS has also shown the potential of simultaneous detection of influenza virus (H1N1), SARS-CoV-2, and respiratory syncytial virus by using magnetic-tags-based SERS substrates with extended studies in throat swabs [272]. Label-free SERS was performed on serum samples of patients after 4 to 16 days of testing positive for COVID-19, and chemometric techniques were used to find significant difference in the SERS spectral features [273]. Figure 5 summarizes different techniques that are used for the SERS-based detection of SARS-CoV-2.

3. SERS-Based Detection of Microorganisms

3.1. Bacteria Sensing

A bacterium is a living cell and falls under the class of prokaryotic microorganisms. Bacteria come in different shapes, including spheres, rods, spiral, and comma, and have a typical size of few micrometers [274]. Bacterial cells are omnipresent, as they are found in water, food, soil, air, and the human body, and, interestingly, the human body contains 10 times more bacterial cells than human cells. However, only 3% of the bacteria are pathogenic, while the other 97% are essential for the survival of different life forms on the earth [275]. The identification of bacteria is important to assess the quality and contamination of food, soil, and water as a measure of public health. In some cases, the presence of bacteria is also desirable to ensure the decomposition of undesirable contaminants through a process called bioremediation [276,277,278]. Conventionally, PCR, plate culture, and flow cytometry are used for the detection of bacteria. However, all of them are time-consuming and need 2 to 3 days to arrive at conclusions [279]. SERS-based sensing for bacteria is extensively used for its proven advantages of being specific, sensitive [280,281,282], rapid [283], and water compatible to perform in situ measurements [284], as well as having the ability to quantify [285,286,287] and potential for trace detection [288,289,290,291,292,293]. Point-of-care devices for detection of bacteria can also be realized through SERS [294,295]. The sensitivity of SERS even enabled the detection of the single bacterium [295]. It is even possible to distinguish between live and dead bacteria cells by using SERS [296]. With the use of appropriate machine learning techniques, researchers achieved strain-level distinction using SERS spectra [297].
A SERS biosensor using aptamer (aptamer–Fe3O4@Au) and antibiotic (Vancomycin–Au@MBA) molecules has been used for the detection and quantification of pathogenic bacteria achieving a LOD of 3 cells/mL [298]. Vancomycin tagged NPs were also used in fabricating a sandwich such as SERS substrate for identification and photothermal elimination of bacteria in blood samples [299]. Different bacteria species such as S. typhi, E. coli, and L. mono were identified using SERS with Fe3O4@Au magnetic nanoparticles and demonstrated good accuracy in real world samples such as beef, saliva, and urine [300]. Wang et al. have also used magnetic nanoparticles for the detection of S. aureus [301,302]. Inspired by polyphenolic chemistry, SERS substrates with metal phenolic networks were designed for the detection of E. coli and S. aureus [303]. In addition to E. coli detection, antibiotic susceptibility was studied using core–shell Au@Ag nanorods. This study was also extended to mice blood, implying practical usage [304]. Bacteria present in serum and human blood sampl was identified using SERS based sensing [305,306]. Polymer mats prepared by force spinning were used for the detection of S. aureus, P. aeruginosa, and S. Typhimurium in blood plasma [307]. Using external magnetic field and plasmonic magnetic nanoparticles, the sensitive detection of Gram-negative bacteria was performed by concentrating the sample to a small area [308]. Similar work was accomplished using a microfluidic device to analyze drinking water for bacterial contamination [309]. The quantification of Salmonella typhimurium was performed using 3D DNA-based SERS substrates [310]. SERS-based immunoassay was used for the ultrasensitive and quantitative detection of different bacteria species simultaneously [311]. Multiplexing was also demonstrated by Hayleigh et al. [312] and Gracie et al., who then went on to conduct quantification in multiplexing [313]. A ceramic-filter-based SERS substrate, along with metal nanoparticles, was used for the detection of E. coli and Shewanella putrefaciens [314]. Nine different species of E. coli were studied using a SERS microfluidic device and discriminated with 92% accuracy, using support vector machine analysis [315]. The label-free and portable detection of various foodborne bacteria was studied using SERS and different chemometric techniques, e.g., PCA and PLS-DA [316]. Silver nanoparticles synthesized using leaf extract were used for the detection of two bacteria species [317]. SERS, in combination with deep learning techniques, was used for the accurate identification of Staphylococcus aureus to achieve an accuracy of ~98% [318].

3.2. Sensing of Biohazardous Molecules for Homeland Security

Bioterrorism is the new threat facing the world and is equally potent to cause large-scale destruction of civil, animal, and plant life. Often, biological agents are easy to prepare and scale up; can be contaminated in food, water, and soil; and are easy to carry, making them the future weapons. Many countries keep them in their military stockpiles despite the regulations [319]. According to the Centre for Disease Control and Prevention (CDC), a biohazardous material is defined as any infectious agent or biological material that poses a threat to human health, the environment, and animals. A review by Lister et al. summarized different biological agents that concern homeland security [320]. Different pathogens and biological agents, such as toxins, venom, and allergens, are some examples of biohazardous materials. Nerve agents are a big concern owing to their high solubility, high toxicity, and durability, with the Tokyo event in 1995 being an example [321,322]. Nerve agents can be classified into G-series, representing agents developed by Germans; V-series for venomous agents; GV series for the combination of G- and V-series; and Novichock series [323]. It is imperative to have a detection system that is sensitive, rapid, portable, and functional for different background media, such as liquids and gases, for the detection of these nerve agents. Plasmonic sensors are widely used for the detection of chemical and biological war threats [324,325]. Of all, SERS has its own advantages for the reasons discussed in the Introduction section and hence is widely used for the detection of biological threats, with a potential for field applications using portable devices [326] and chemometrics [327]. Here we focus on nerve agents, risk-grade-two and -three bacteria species, or their biomarkers’ sensing, using SERS, with an interest in homeland security.
A sensitive and selective identification of the nerve agents Tabun, Cyclosarin, and VX was performed using gold- and silver-coated Si nanostructures both without [328] and with a tag (antidote) [329] in two different studies. VX and its hydrolysis products were studied elsewhere, too [330,331]. Sarin, an organophosphorus nerve agent, was detected using plasmonic Si nanocone structures [332]. Three nerve agents, i.e., isopropyl methylphosphonofluoridate (GB), pinacolyl methylphosphonofluoridate (GD), and cyclohexyl methylphosphonofluoridate (GF), were identified, and their hydrolysis degradation was distinguished using SERS [322]. A mustard simulant, pathogenic bacteria, and cyanide were detected using SERS [333]. A reproducible (7%), rapid (30 s), and sensitive (1 ppb) was used for the detection of a nerve simulant, pinacolyl methyl phosphonic acid (PMPA) [334]. Gaseous warfare agents such as dimethyl methylphosphonate were identified using SERS on LiCl microlenses [335]. Various G-series and VX nerve agents were identified using novel pinhole shell-isolated Au nanoparticles substrates achieving sensitivity of 10 ng/L and 20 ng/L, respectively [336]. Using plasmonic 3D fractal structures, a G-series nerve agent called dimethyl methylphosphonate (DMMP) was detected in the gaseous state, with a sensitivity of 12 ppmV [337]. Bacillus anthracis is a highly infectious bacteria that causes the fatal disease anthrax in humans. It is a cause for concern because of its recent usage as a biowarfare agent by many countries [338]. Farrell et al. summarized different anthrax biomarkers and existing detection techniques [339]. Plasmonic metal decorated anisotropic Ni nanostructures were used for detection of dipicolinic acid (DPA), a biomarker for anthrax [340]. Specifically, tagged SERS substrates were used for the detection of anthrax protective antigens, achieving a remarkable LOD of 1 pg/mL [341]. A magnetic microfluidic SERS sensor using specifically tagged Au nanoparticles was used for the detection of the anthrax biomarker poly-γ-D-glutamic acid, with an LOD of 100 pg/mL [342]. Reusable and sensitive laser-ablated Au nanostructures were used for the detection of dipicolinic acid (DPA) with a LOD of 0.83 pg/L and signal enhancement of ~1012 [343]. A selective SERS substrate that can discriminate between different strains of bacteria by specifically binding to Bacillus anthracis was designed with DPA as a biomarker [344]. Gold nanorods were also employed for the sensitive detection of DPA and anthrax-protective antigen [345,346]. The trace detection of DPA, equivalent to nearly 18 spores, was achieved using super-hydrophobic SERS sensors [347]. The effects of aggregation of NPs and pH on the SERS performance for the detection of components of cell wall and endospores of Bacillus thuringiensis were studied extensively [348]. Different chemical and biological warfare agents were classified using techniques such as PCA, PLS-DA, as well as hierarchical classification techniques based on the SERS spectra [328,349].

4. Machine Learning in SERS-Based Biosensing

4.1. Introduction to Machine Learning

In recent times, machine learning is widely being used for many applications including spectroscopy for both data pre- and postprocessing. Machine learning (ML), as the name suggests, is a technique in which the algorithm learns patterns from the existing data and will attempt to make accurate predictions on the unknown based on the trained data. The potential for its ability to find complex patterns from big data sets has given an opportunity to extract and model data purposefully. There are different existing algorithms, both supervised and unsupervised, depending on the problem at hand. Deep learning is a subdomain of machine learning inspired by the human brain that uses multilayered neural networks for modeling data. Throughout this article, machine learning also implies deep learning techniques. Advances in computation facilities and with increasing availability and complexity of big data, deep learning, which is a kind of machine learning, has found its place. Some popular and relevant examples of ML being classification of emails as span and not span, identifying cancer in early stage using medical images, face recognition and weather prediction. ML algorithms can be broadly classified into three types, namely supervised for labeled observations, unsupervised for unlabeled observations, and reinforcement learning for models that learn from the errors to improve accuracy [350], as summarized in the Figure 6 below.
With the ease of data collection and availability of open source Raman spectroscopy data, SERS has also seen a surge in machine learning models [49,351,352]. The trend is welcoming and desirable as the nature of existing challenges in SERS involving trace detection, signal fluctuations, quantification and identification are complex with many variables calling for an analytical tool that has the ability to capture the patterns devoid of experts [353]. Trace detection implies identifying signal from a noisy background where ML could be aided. SERS is also known to have inherent signal fluctuations owing to localization of hotspots. Especially in the case of bio samples, they have background contribution from different undesirable components thus interfering with the signal and need ML algorithms to extract the useful information [2,354,355,356,357]. The process of data collection, identification of chemical composition and quantification is non-linear and is highly dependent on human intelligence making it a barrier to carry the benefits of SERS to onsite [358]. Some of the widely used techniques include Principal Component Analysis (PCA), Support Vector Machine (SVM), Partial Least Squares (PLS), Decision Trees (DTs) and Convolutional Neural Networks (CNNs). PCA is a dimensionality reduction technique where components representative of the data with large variance are preserved. This is extensively used a preprocessing step in order to reduce complexity of the models or also as a classification technique [359,360,361]. SVM is a nonlinear ML technique that can be used for both regression and classification [360]. It works by finding a hyperplane that distinguishes two or more classes using a kernel function [362]. If the data set is small and the number of variables is large, PLS is useful for its ability to still extract useful information and is often used for quantitative studies [363,364]. DTs are widely used for classification of the data using a method bootstrapping [365]. CNNs are a kind of neural networks which employ filters and pooled layers in the architecture and often used if the size of the data set is large enough and if images are involved in the modeling [366]. Specifically, in the field of biophotonics, machine learning models using SERS can be efficiently classified into three domains: identification, classification, and quantification, with interests such as disease and molecular diagnosis [367,368]; microorganism classification, identification, etc. [369,370,371,372]; and cancer diagnosis [373], as shown in Figure 7. In addition, machine learning was also used to improve data collection to overcome signal fluctuations and enhance the usability on site [374], to estimate the effect of scattering [375] and for the SERS signal enhancement itself [376]. In further sections, we discuss different ML techniques that were used in SERS for biology applications.

4.2. Identification

SERS provides the vibrational fingerprint of many biomolecules, including amino acids, peptides, carbohydrates, pathogens, and nuclei acids [377]. It is also label free and non-destructive, making it desirable for in situ and rapid identification. Often in real-world situations of biology sample analysis, there are undesirable effects from background cell signals or with the similarity of spectra from two subspecies. Machine learning models can be successfully trained to capture these complex differences and distinguish two similar spectra devoid of the background helping in identification of the sample. Figure 8 summarizes the work so far in using ML for identification applications in biosensing with SERS. CNNs were used for identification of cancer using SERS with gold multi-branched nanoparticles (AuMs), functionalized with different chemical groups, and achieved 100% accuracy in identifying the structural changes [378]. Drug-sensitive and drug-resistant bacterial strains were identified using SERS with a combination of CNNs and achieved 100% accuracy [379]. Different classification algorithms such as LDA, SVM, and KNN were used for the classification of bacterial extracellular vesicles for E. coli by strain and culture time using label-free approach of SERS [380]. SVM was successfully used for the identification of different drugs in human urine at trace levels with an accuracy greater than 92% [381]. A SERS chip was designed to identify a cancer marker, TIMP-1, and combined it with ML to identify lung and colon cancer in patients [382]. A label-free SERS, in combination with different machine learning algorithms, such as random forest, PCA-LDA, and decision trees, was used for the identification of colon cancer using serum samples. It was found that the random forest model outperformed the other two models in terms of accuracy and specificity [383]. SERS combined with ANN was used for the identification of different pollen samples despite many spectral contributions using Au NPs [384]. A microfluidic-chip-based SERS substrate with Au nanoparticles was used for the identification of l Jurkat, THP-1, and MONO-MAC-6 leukemia cell lysates, using SVM, and achieved 99% accuracy [385]. A lab-on-chip SERS device was fabricated and used for the successful identification of different species of mycobacteria [386]. The machine learning models PLS-DA and CNN were used to identify different stages of kidney malfunction in dialysis patients by using serum analysis by SERS. The CNN model achieved an accuracy of 96%, which is better than that of PLS-DA, with 84% [387]. The SVM outperformed other techniques in the identification of cyanobacteria, using SERS spectra of mutant and wild-type strains [388]. Using a dimensionality reduction technique, followed by a probabilistic ML model, SARS-CoV-2 identification was performed with an accuracy of ~85% [389]. SERS coupled with SVM was also used for the identification of lung cancers [96].

4.3. Quantification

One of the interests of using SERS for sensing also lies in its ability to detect trace and ultra-trace molecules. The intensity and concentration relation for a peak of choice in the SERS spectrum is often non-linear due to many factors, such as the inhomogeneous distribution of hotspots, non-uniform adsorption of molecules, and localization of the hotspots [361,390]. This calls for machine learning models that have the ability to capture non-linear patterns of intensity and concentration relation and further predict the unknown concentration. As the problem demands, regression ML models such as PCR, PLSR, SVR, and XGBR are used for the quantification of trace biomolecules.
A quantitative analysis of antibiotics and a mixture of antibiotics was performed using PLSR with an accuracy of 96% [391]. An SERS-based lateral flow assay was used for the quantification of E. coli in milk and beef, using the Bayesian ridge regression (BRR), support vector regression (SVR), elastic net regression (ENR), and extreme gradient boosting regression (XGBR) algorithm, as shown in Figure 9 [392]. A SERS substrate with plasmonic nanogaps was fabricated and used for the trace sensing of pyocyanin, a secondary metabolite of Pseudomonas aeruginosa, from a complex background. Furthermore, using machine learning algorithms, the quantification of pyocyanin was performed with an accuracy until five significant digits, using PLS [393]. The quantification of very low concentrations of fumonicins in maize was performed using different chemometric techniques such as PCR and PLSR and achieved an accuracy above 90% [394]. Thiols found in the whole blood of umbilical cords were quantified using a PLSR model on SERS spectra collected using silver nanoparticles as plasmonic substrates [395]. PCA, followed by SVR, was used for the quantification of histamine, an allergen, in seafood, using spectral data from a combination of TLS and SERS [396].

4.4. Classification

The goal of the classification algorithms employed for data analysis in SERS for biosensing is often differentiating different classes, species, and spectra corresponding to different stages of the disease or different diseases themselves. So far, classification algorithms such as SVM, KNN, and PCA; and different neural networks, such as CNN, were used for the problems stated.
Different bacteria species were classified and identified using SVM, with an accuracy of 87% by using SERS with bacterial cellulose nanocrystals (BCNCs) decorated with Au nanoparticles [397]. K-nearest neighbor and decision trees were used for the classification of SERS-based liquid biopsy assay to identify five protein biomarkers (CA19-9, HE4, MUC4, MMP7, and mesothelin) in pancreatic cancer patients, ovarian cancer patients, pancreatitis patients, and healthy individuals [398]. The direct serum analysis of liver cancer samples is performed using Au-Ag nano complex-decorated ZnO nanopillars on paper for the classification of different stages of cancer using CNNs. This method achieved an accuracy of 97.78% [399]. SERS combined with machine learning was also used for the screening of PCOS, using classification algorithms on SERS data. Samples of follicular fluids and plasma from healthy and PCOS patients were successfully classified, with an accuracy of 89%, using stacked models for both [400]. Protein species with similar spectral profiles were classified using principal component analysis (PCA) applied to SERS spectra [401]. CNNs without any preprocessing steps were used for the classification of different grades of bladder cancer tissue, using Raman spectra, and different species of E. coli, using SERS spectra. Different classification algorithms, such as KNN, PCA, SVM, and ANN, were used, but CNN was found to outperform the others in terms of accuracy [402]. Using Non-Structural Protein 1 (NS1) as a biomarker for dengue, extreme learning machine and PCA models were used for the classification of dengue patients with 100% accuracy towards a goal of early diagnosis [403]. Bacterial endotoxins of twelve different species were identified and classified using SERS spectra and machine learning algorithms such as KNN, RF, SVM, and RamanNet. While the other algorithms achieved accuracy greater than 90%, RamanNet outperformed them, with 100% accuracy [404]. With a goal to identify cancer at an early stage, a point-of-care diagnosis system using a novel hydrophobic SERS substrate combined with machine learning techniques was used, as shown in Figure 10 [50]. The SERS spectra of serum samples collected from nearly 690 patients, including normal and different cancers (breast cancer, leukemia, and hepatitis B virus), were collected and analyzed using deep learning techniques to achieve 100% accuracy in successfully classifying the data. They performed external testing with an accuracy of 98%, indicating potential usage in the real world.

5. Conclusions and Scope

The vast existing literature and continued interest in the SERS technique for biosensing is a promising sign to realize point-of-care devices based on SERS. This would revolutionize disease diagnosis due to its ability to identify traces, enabling early detection, cost effectiveness, and rapid diagnosis. Under optimized conditions, a single bacterial cell was also detected using SERS, thus demonstrating its sensitivity [295]. Using SERS, it is possible to identify disease biomarkers in a variety of bio-fluids, such as urine, saliva, plasma, and blood, as well as in volatile compounds and gases. Unlike many commercial techniques, SERS is reagent free and does not need sequential procedures for the identification of disease biomarkers. Machine learning techniques are extensively being used in SERS for their ability to recognize complex and intricate patterns devoid of background noise. Different models, such as PCA, SVM, ANN, CNN, KNN, and PLS, were used for identification, quantification, and classification of microorganisms and different diseases, including cancers. In regard to cancer diagnosis using SERS, the distinguishment between normal and cancerous samples, including cells and liquids, and the discrimination of different stages of cancer have also been performed. These methods are cost efficient, rapid, and sensitive, as opposed to the existing cancer-screening techniques. In response to the pandemic situation, SERS has been extensively used for the detection of novel COVID-19 virus and also for tracking the efficiency of the vaccines [266]. Furthermore, SERS has been widely used for the detection of various nerve agents and other bio-warfare agents, thus expanding its application in homeland security. Commercialization of SERS is already underway, with many lateral flow and point-of-care devices that have been developed in response to the pandemic [261] and diagnosis of other diseases with equal and par performance as existing commercial techniques [150]. For example, it is established that SERS performs better than the commercial enzyme-linked immunosorbent assay (ELISA) test kits in cancer detection, allowing multiplexing with very less sample volume [405]. It was also shown to achieve a lower LOD than radioimmunoassay (RIA) and ELISA in a different study [406]. In a recent study, SERS was compared with a clinically available method for quantification of glucose in blood sugar and shown to perform equally good [407]. It was found that SERS is 16-to-32-times more sensitive than the commercial lateral flow assay and >400-times more sensitive than the ELISA with the same reagents for the detection of covid [264]. The major components of a Raman system consist of a laser source, a probe for excitation and signal collection, and a detection system with a spectrometer [408]. Recent advances on all of these fronts for miniaturizing and reducing the cost are enabling the widespread usage of SERS-based detection with portable systems.
Despite its merits, there are few challenges that stand in the way of scaling up SERS for biosensing in the real world. Firstly, there are reasons inherent to the SERS enhancement mechanisms that turn out to be undesirable, often causing signal fluctuations and poor reproducibility. Due to the localization of dense field enhancement areas (“hotspots”) and metal-sample adsorption artefacts, SERS signals are known to fluctuate. Upon laser illumination, these hotspots are also known to diffuse or transform, thus adding further to the poor reproducibility. A substrate with homogeneous field enhancement promises good reproducibility but comes at the cost of enhancement and eventually limiting trace detection [374]. Secondly, SERS substrates majorly comprise Au or Ag nanoparticles/nanostructures. These nanostructures are generally not stable for long durations, with a risk of rapid oxidation upon exposure to ambient atmosphere. Owing to their large surface charge, they also tend to aggregate to form clusters. Often, aggregation and oxidation are prevented by the addition of capping agents or ligands which could affect the SERS signal and compatibility with the bio-samples. In view of commercialization, there is also a question of the reusability of the SERS substrates. Thirdly, SERS substrates that are used in the lab are optimized under specific instrument conditions, such as laser wavelength, acquisition time, power, and focusing conditions. In regard to point-of-care applications, it is a challenge to have the same experimental conditions, thus limiting the substrate efficiency. Field applications also call for cost-effective and miniature devices that are easy to operate by a non-expert [39]. Specifically in regard to biosensing, SERS-based detection in the field is a challenge because of the lack of disease specificity. In most of the scenarios, the biomarkers for disease detection, such as proteins and antigens, are not disease specific and need further evaluation in order for researchers to arrive at conclusions [409]. In the case of whole-organism or cell/tissue studies, it is difficult to ascertain the peaks because of contribution from various components, such as proteins, DNA bases, lipids, and other cell components. Figure 11 summarizes some best practices for quantification and qualitative analysis in SERS at various stages of experiments. The current research in SERS for biosensing is moving in the right direction to overcome these challenges with exploration in the direction of instrumentation, standoff detection, and the usage of ML techniques to improve data collection and identification without expertise. We believe that, in the coming years, these challenges will be successfully met and SERS will realize its full potential in real-world low-cost biosensing.

Author Contributions

Conceptualization, R.B. and V.R.S.; methodology, R.B. and V.R.S.; resources, V.R.S. and K.R.V.; writing—original draft preparation, R.B.; writing—review and editing, V.R.S. and K.R.V.; visualization, R.B. and V.R.S.; supervision, V.R.S. and K.R.V.; project administration, V.R.S.; funding acquisition, V.R.S. All authors have read and agreed to the published version of the manuscript.

Funding

The authors thank DRDO for the funding [Project #ERIP/ER/1501138/M/01/319/D(R&D)]. V.R. Soma also thanks the University of Hyderabad (UoH) for financial support through the Institute of Eminence (IoE) project [No. UOH/IOE/RC1/RC1-20-016]. The IoE scheme was granted to the UoH by the Ministry of Education, Government of India, vide MHRD notification F11/9/2019-U3(A).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stewart, M.E.; Anderton, C.R.; Thompson, L.B.; Maria, J.; Gray, S.K.; Rogers, J.A.; Nuzzo, R.G. Nanostructured Plasmonic Sensors. Chem. Rev. 2008, 108, 494–521. [Google Scholar] [CrossRef]
  2. Costanzo, H.; Gooch, J.; Frascione, N. Nanomaterials for Optical Biosensors in Forensic Analysis. Talanta 2023, 253, 123945. [Google Scholar] [CrossRef]
  3. Chen, G.; Chen, Y.; Huang, W.; Shi, Y. Plasmonic Nanobiosensors for Detection of Different Targets. In Proceedings of the Second International Conference on Medical Imaging and Additive Manufacturing (ICMIAM 2022), Xiamen, China, 25–27 February 2022. [Google Scholar] [CrossRef]
  4. Sadani, K.; Nag, P.; Thian, X.Y.; Mukherji, S. Enzymatic Optical Biosensors for Healthcare Applications. Biosens. Bioelectron. X 2022, 12, 100278. [Google Scholar] [CrossRef]
  5. Erkmen, C.; Selcuk, O.; Unal, D.N.; Kurbanoglu, S.; Uslu, B. Layer-by-Layer Modification Strategies for Electrochemical Detection of Biomarkers. Biosens. Bioelectron. X 2022, 12, 100270. [Google Scholar] [CrossRef]
  6. Spillman, W.B. Fiber Optic Biosensors; Elsevier: Amsterdam, The Netherlands, 2011; Volume 3, ISBN 9780470126844. [Google Scholar]
  7. Kazanskiy, N.L.; Khonina, S.N.; Butt, M.A.; Kaźmierczak, A.; Piramidowicz, R. State-of-the-Art Optical Devices for Biomedical Sensing Applications—A Review. Electronics 2021, 10, 973. [Google Scholar] [CrossRef]
  8. Ramirez, J.C.; Grajales García, D.; Maldonado, J.; Fernández-Gavela, A. Current Trends in Photonic Biosensors: Advances towards Multiplexed Integration. Chemosensors 2022, 10, 398. [Google Scholar] [CrossRef]
  9. Chadha, U.; Bhardwaj, P.; Agarwal, R.; Rawat, P.; Agarwal, R.; Gupta, I.; Panjwani, M.; Singh, S.; Ahuja, C.; Selvaraj, S.K.; et al. Recent Progress and Growth in Biosensors Technology: A Critical Review. J. Ind. Eng. Chem. 2022, 109, 21–51. [Google Scholar] [CrossRef]
  10. Dutta, G. Next-Generation Nanobiosensor Devices for Point-of-Care Diagnostics; Springer: Singapore, 2023; ISBN 9789811971303. [Google Scholar] [CrossRef]
  11. Ahangari, A.; Mahmoodi, P.; Mohammadzadeh, A. Advanced Nano Biosensors for Rapid Detection of Zoonotic Bacteria. Biotechnol. Bioeng. 2022, 120, 41–56. [Google Scholar] [CrossRef]
  12. Taha, B.A.; Al Mashhadany, Y.; Bachok, N.N.; Ashrif, A.; Bakar, A.; Hafiz Mokhtar, M.H.; Dzulkefly Bin Zan, M.S.; Arsad, N. Detection of COVID-19 Virus on Surfaces Using Photonics: Challenges and Perspectives. Diagnostics 2021, 11, 1119. [Google Scholar] [CrossRef]
  13. Soma, V.R.; Podagatlapalli, G.K.; Hamad, S.; Mechanics, F. Ultrafast Laser Ablation in Liquids for Nanomaterials and Applications. J. Nanosci. Nanotechnol. 2014, 14, 1364–1388. [Google Scholar] [CrossRef]
  14. Soler, M.; Lechuga, L.M. Principles, Technologies, and Applications of Plasmonic Biosensors. J. Appl. Phys. 2021, 129, 111102. [Google Scholar] [CrossRef]
  15. Barbillon, G. Plasmonics and Its Applications. Materials 2019, 12, 1502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Michaels, A.M.; Jiang, J.; Brus, L. Ag Nanocrystal Junctions as the Site for Surface-Enhanced Raman Scattering of Single Rhodamine 6G Molecules. J. Phys. Chem. B 2000, 104, 11965–11971. [Google Scholar] [CrossRef] [Green Version]
  17. Golightly, R.S.; Doering, W.E.; Natan, M.J. Surface-Enhanced Raman Spectroscopy and Homeland Security: A Perfect Match? ACS Nano 2009, 3, 2859–2869. [Google Scholar] [CrossRef] [PubMed]
  18. Vendamani, V.S.; Beeram, R.; Nageswara Rao, S.V.S.; Pathak, A.P.; Soma, V.R. Trace Level Detection of Explosives and Pesticides Using Robust, Low-Cost, Free-Standing Silver Nanoparticles Decorated Porous Silicon. Opt. Express 2021, 29, 30045. [Google Scholar] [CrossRef]
  19. Liu, C.; Xu, D.; Dong, X.; Huang, Q. A Review: Research Progress of SERS-Based Sensors for Agricultural Applications. Trends Food Sci. Technol. 2022, 128, 90–101. [Google Scholar] [CrossRef]
  20. Zhang, D.; Pu, H.; Huang, L.; Sun, D.W. Advances in Flexible Surface-Enhanced Raman Scattering (SERS) Substrates for Nondestructive Food Detection: Fundamentals and Recent Applications. Trends Food Sci. Technol. 2021, 109, 690–701. [Google Scholar] [CrossRef]
  21. Li, Y.; Liu, X.; Lin, Z. Recent Developments and Applications of Surface Plasmon Resonance Biosensors for the Detection of Mycotoxins in Foodstuffs. Food Chem. 2012, 132, 1549–1554. [Google Scholar] [CrossRef]
  22. Dies, H.; Raveendran, J.; Escobedo, C.; Docoslis, A. Rapid Identification and Quantification of Illicit Drugs on Nanodendritic Surface-Enhanced Raman Scattering Substrates. Sens. Actuators B Chem. 2018, 257, 382–388. [Google Scholar] [CrossRef]
  23. Vendamani, V.S.; Beeram, R.; Rao, S.V.S.N.; Rao, S.V. Protocol for Designing AuNP-Capped Ag Dendrites as Surface-Enhanced Raman Scattering Sensors for Trace Molecular Detection Protocol for Designing AuNP-Capped Ag Dendrites as Surface-Enhanced Raman Scattering Sensors for Trace Molecular Detection. STAR Protoc. 2023, 4, 102068. [Google Scholar] [CrossRef]
  24. He, L.; Rodda, T.; Haynes, C.L.; Deschaines, T.; Strother, T.; Diez-Gonzalez, F.; Labuza, T.P. Detection of a Foreign Protein in Milk Using Surface-Enhanced Raman Spectroscopy Coupled with Antibody-Modified Silver Dendrites. Anal. Chem. 2011, 83, 1510–1513. [Google Scholar] [CrossRef]
  25. Jebakumari, K.A.E.; Murugasenapathi, N.K. Engineered Two-Dimensional Nanostructures as SERS Substrates for Biomolecule Sensing: A Review. Biosensors 2023, 13, 102. [Google Scholar] [CrossRef]
  26. Bantz, K.C.; Meyer, A.F.; Wittenberg, N.J.; Im, H.; Kurtuluş, Ö.; Lee, S.H.; Lindquist, N.C.; Oh, S.H.; Haynes, C.L. Recent Progress in SERS Biosensing. Phys. Chem. Chem. Phys. 2011, 13, 11551–11567. [Google Scholar] [CrossRef] [PubMed]
  27. Vendamani, V.S.; Nageswara Rao, S.V.S.; Venugopal Rao, S.; Kanjilal, D.; Pathak, A.P. Three-Dimensional Hybrid Silicon Nanostructures for Surface Enhanced Raman Spectroscopy Based Molecular Detection. J. Appl. Phys. 2018, 123, 014301. [Google Scholar] [CrossRef] [Green Version]
  28. Huang, Z.; Zhang, A.; Zhang, Q.; Cui, D. Nanomaterial-Based SERS Sensing Technology for Biomedical Application. J. Mater. Chem. B 2019, 7, 3755–3774. [Google Scholar] [CrossRef]
  29. Szaniawska, A.; Kudelski, A. Applications of Surface-Enhanced Raman Scattering in Biochemical and Medical Analysis. Front. Chem. 2021, 9, 664134. [Google Scholar] [CrossRef]
  30. Chen, Y.; An, Q.; Teng, K.; Liu, C.; Sun, F.; Li, G. Application of SERS in In-Vitro Biomedical Detection. Chem. Asian J. 2022, 18, e202201194. [Google Scholar] [CrossRef]
  31. Hegde, M.; Pai, P.; Gangadhar Shetty, M.; Sundara Babitha, K. Gold Nanoparticle Based Biosensors for Rapid Pathogen Detection: A Review. Environ. Nanotechnol. Monit. Manag. 2022, 18, 100756. [Google Scholar] [CrossRef]
  32. Fleischmann, M.; Hendra, P.J.; McQuillan, A.J. Raman Spectra of Pyridine Adsorbed at a Silver Electrode. Chem. Phys. Lett. 1974, 26, 163–166. [Google Scholar] [CrossRef]
  33. Jeanmaire, D.L.; Duyne, R.P.V.A.N. Surface Raman spectroelectrochemistry: Part I. Heterocyclic, aromatic, and aliphatic amines adsorbed on the anodized silver electrode. J. Electroanal. Chem. Interfacial Electrochem. 1977, 84, 1–20. [Google Scholar] [CrossRef]
  34. Albrecht, M.G.; Creighton, J.A. Anomalously Intense Raman Spectra of Pyridine at a Silver Electrode. J. Am. Chem. Soc. 1977, 99, 5215–5217. [Google Scholar] [CrossRef]
  35. Pilot, R.; Signorini, R.; Durante, C.; Orian, L.; Bhamidipati, M.; Fabris, L. A Review on Surface-Enhanced Raman Scattering. Biosensors 2019, 9, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Le Ru, E.; Etchegoin, P. Principles of Surface Enhanced Raman Spectroscopy and Related Plasmonic Effects; Elseveir: Amsterdam, The Netherlands, 2008. [Google Scholar]
  37. Etchegoin, P.G.; Ru, E.C.L. Basic Electromagnetic Theory of SERS. In Surface Enhanced Raman Spectroscopy: Analytical, Biophysical and Life Science Applications; John Wiley & Sons: Hoboken, NJ, USA, 2011; pp. 1–37. ISBN 9783527325672. [Google Scholar]
  38. Le Ru, E.C.; Blackie, E.; Meyer, M.; Etchegoint, P.G. Surface Enhanced Raman Scattering Enhancement Factors: A Comprehensive Study. J. Phys. Chem. C 2007, 111, 13794–13803. [Google Scholar] [CrossRef]
  39. Sharma, B.; Frontiera, R.R.; Henry, A.I.; Ringe, E.; Van Duyne, R.P. SERS: Materials, Applications, and the Future. Mater. Today 2012, 15, 16–25. [Google Scholar] [CrossRef]
  40. Li, Q.; Huo, H.; Wu, Y.; Chen, L.; Su, L.; Zhang, X.; Song, J.; Yang, H. Design and Synthesis of SERS Materials for In Vivo Molecular Imaging and Biosensing. Adv. Sci. 2023, 2023, 2202051. [Google Scholar] [CrossRef]
  41. Israelsen, N.D.; Hanson, C.; Vargis, E. Nanoparticle Properties and Synthesis Effects on Surface-Enhanced Raman Scattering Enhancement Factor: An Introduction. Sci. World J. 2015, 2015, 124582. [Google Scholar] [CrossRef] [Green Version]
  42. Wang, A.X.; Kong, X. Review of Recent Progress of Plasmonic Materials and Nano-Structures for Surface-Enhanced Raman Scattering. Materials 2015, 8, 3024–3052. [Google Scholar] [CrossRef]
  43. Moram, S.S.B.; Byram, C.; Soma, V.R. Gold-Nanoparticle- and Nanostar-Loaded Paper-Based SERS Substrates for Sensing Nanogram-Level Picric Acid with a Portable Raman Spectrometer. Bull. Mater. Sci. 2020, 43, 53. [Google Scholar] [CrossRef]
  44. Zhang, Z.; Guan, R.; Li, J. Engineering Rational SERS Nanotags for Parallel Detection of Multiple Cancer Circulating Biomarkers. Chemosensors. 2023, 11, 110. [Google Scholar] [CrossRef]
  45. Pilot, R.; Massari, M. Silver Nanoparticle Aggregates: Wavelength Dependence of Their SERS Properties in the First Transparency Window of Biological Tissues. Chem. Phys. Impact 2021, 2, 100014. [Google Scholar] [CrossRef]
  46. Zhang, Y.; Mi, X.; Tan, X.; Xiang, R. Recent Progress on Liquid Biopsy Analysis Using Surface-Enhanced Raman Spectroscopy. Theranostics 2019, 9, 491–525. [Google Scholar] [CrossRef] [PubMed]
  47. Aitekenov, S.; Sultangaziyev, A.; Ilyas, A.; Dyussupova, A.; Boranova, A.; Gaipov, A.; Bukasov, R. Surface-Enhanced Raman Spectroscopy (SERS) for Protein Determination in Human Urine. Sens. Bio-Sens. Res. 2022, 38, 100535. [Google Scholar] [CrossRef]
  48. Akgönüllü, S.; Denizli, A. Recent Advances in Optical Biosensing Approaches for Biomarkers Detection. Biosens. Bioelectron. X 2022, 12, 100269. [Google Scholar] [CrossRef]
  49. Lussier, F.; Thibault, V.; Charron, B.; Wallace, G.Q.; Masson, J.F. Deep Learning and Artificial Intelligence Methods for Raman and Surface-Enhanced Raman Scattering. TrAC Trends Anal. Chem. 2020, 124, 115796. [Google Scholar] [CrossRef]
  50. Lin, X.; Lin, D.; Chen, Y.; Lin, J.; Weng, S.; Song, J.; Feng, S. High Throughput Blood Analysis Based on Deep Learning Algorithm and Self-Positioning Super-Hydrophobic SERS Platform for Non-Invasive Multi-Disease Screening. Adv. Funct. Mater. 2021, 31, 2103382. [Google Scholar] [CrossRef]
  51. Breuch, R.; Klein, D.; Siefke, E.; Hebel, M.; Herbert, U.; Wickleder, C.; Kaul, P. Differentiation of Meat-Related Microorganisms Using Paper-Based Surface-Enhanced Raman Spectroscopy Combined with Multivariate Statistical Analysis. Talanta 2020, 219, 121315. [Google Scholar] [CrossRef]
  52. Ilkhani, H.; Hughes, T.; Li, J.; Zhong, C.J.; Hepel, M. Nanostructured SERS-Electrochemical Biosensors for Testing of Anticancer Drug Interactions with DNA. Biosens. Bioelectron. 2016, 80, 257–264. [Google Scholar] [CrossRef] [PubMed]
  53. Leong, S.X.; Leong, Y.X.; Tan, E.X.; Sim, H.Y.F.; Koh, C.S.L.; Lee, Y.H.; Chong, C.; Ng, L.S.; Chen, J.R.T.; Pang, D.W.C.; et al. Noninvasive and Point-of-Care Surface-Enhanced Raman Scattering (SERS)-Based Breathalyzer for Mass Screening of Coronavirus Disease 2019 (COVID-19) under 5 Min. ACS Nano 2022, 16, 2629–2639. [Google Scholar] [CrossRef]
  54. Bharati, M.S.S.; Soma, V.R. Flexible SERS Substrates for Hazardous Materials Detection: Recent Advances. Opto-Electron. Adv. 2021, 4, 210048. [Google Scholar] [CrossRef]
  55. Ali, A.; Nettey-Oppong, E.E.; Effah, E.; Yu, C.Y.; Muhammad, R.; Soomro, T.A.; Byun, K.M.; Choi, S.H. Miniaturized Raman Instruments for SERS-Based Point-of-Care Testing on Respiratory Viruses. Biosensors 2022, 12, 590. [Google Scholar] [CrossRef]
  56. Mejía-Salazar, J.R.; Oliveira, O.N. Plasmonic Biosensing. Chem. Rev. 2018, 118, 10617–10625. [Google Scholar] [CrossRef]
  57. Han, X.; Liu, K.; Sun, C. Plasmonics for Biosensing. Materials 2019, 12, 1411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Shrivastav, A.M.; Cvelbar, U.; Abdulhalim, I. A Comprehensive Review on Plasmonic-Based Biosensors Used in Viral Diagnostics. Commun. Biol. 2021, 4, 70. [Google Scholar] [CrossRef] [PubMed]
  59. Wang, Q.; Ren, Z.H.; Zhao, W.M.; Wang, L.; Yan, X.; Zhu, A.S.; Qiu, F.M.; Zhang, K.K. Research Advances on Surface Plasmon Resonance Biosensors. Nanoscale 2022, 14, 564–591. [Google Scholar] [CrossRef] [PubMed]
  60. Homola, J. Present and Future of Surface Plasmon Resonance Biosensors. Anal. Bioanal. Chem. 2003, 377, 528–539. [Google Scholar] [CrossRef]
  61. Piliarik, M.; Vaisocherová, H.; Homola, J. Surface Plasmon Resonance Biosensing. In Biosensors and Biodetection; Humana Press: Totowa, NJ, USA, 2009; pp. 65–88. [Google Scholar] [CrossRef]
  62. Hong, Y.; Huh, Y.M.; Yoon, D.S.; Yang, J. Nanobiosensors Based on Localized Surface Plasmon Resonance for Biomarker Detection. J. Nanomater. 2012, 2012, 759830. [Google Scholar] [CrossRef] [Green Version]
  63. Unser, S.; Bruzas, I.; He, J.; Sagle, L. Localized Surface Plasmon Resonance Biosensing: Current Challenges and Approaches. Sensors 2015, 15, 15684–15716. [Google Scholar] [CrossRef]
  64. Brolo, A.G. Plasmonics for Future Biosensors. Nat. Photonics 2012, 6, 709–713. [Google Scholar] [CrossRef]
  65. Liu, J.; Jalali, M.; Mahshid, S.; Wachsmann-Hogiu, S. Are Plasmonic Optical Biosensors Ready for Use in Point-of-Need Applications? Analyst 2020, 145, 364–384. [Google Scholar] [CrossRef] [Green Version]
  66. Moore, T.J.; Moody, A.S.; Payne, T.D.; Sarabia, G.M.; Daniel, A.R.; Sharma, B. In Vitro and in Vivo Sers Biosensing for Disease Diagnosis. Biosensors 2018, 8, 46. [Google Scholar] [CrossRef] [Green Version]
  67. Alvarez-Puebla, R.A.; Liz-Marzán, L.M. SERS-Based Diagnosis and Biodetection. Small 2010, 6, 604–610. [Google Scholar] [CrossRef] [PubMed]
  68. Zhang, X.; Liu, S.; Song, X.; Wang, H.; Wang, J.; Wang, Y.; Huang, J.; Yu, J. Robust and Universal SERS Sensing Platform for Multiplexed Detection of Alzheimer’s Disease Core Biomarkers Using PAapt-AuNPs Conjugates. ACS Sens. 2019, 4, 2140–2149. [Google Scholar] [CrossRef] [PubMed]
  69. Park, H.J.; Cho, S.; Kim, M.; Jung, Y.S. Carboxylic Acid-Functionalized, Graphitic Layer-Coated Three-Dimensional SERS Substrate for Label-Free Analysis of Alzheimer’s Disease Biomarkers. Nano Lett. 2020, 20, 2576–2584. [Google Scholar] [CrossRef]
  70. Dang, H.; Joung, Y.; Jeong, C.; Jeon, C.S.; Pyun, S.H.; Park, S.-G.; Choo, J. Nanoplasmonic Assay Platforms for Reproducible SERS Detection of Alzheimer’s Disease Biomarker. Bull. Korean Chem. Soc. 2023, 2023, 1. [Google Scholar] [CrossRef]
  71. Momenpour, A.; Lima, P.D.A.; Chen, Y.-A.; Tzeng, C.-R.; Tsang, B.K.; Anis, H. Surface-Enhanced Raman Scattering for the Detection of Polycystic Ovary Syndrome. Biomed. Opt. Express 2018, 9, 801. [Google Scholar] [CrossRef] [PubMed]
  72. Lyandres, O.; Shah, N.C.; Yonzon, C.R.; Walsh, J.T.; Glucksberg, M.R.; Van Duyne, R.P. Real-Time Glucose Sensing by Surface-Enhanced Raman Spectroscopy in Bovine Plasma Facilitated by a Mixed Decanethiol/Mercaptohexanol Partition Layer. Anal. Chem. 2005, 77, 6134–6139. [Google Scholar] [CrossRef]
  73. Qi, G.; Jia, K.; Fu, C.; Xu, S.; Xu, W. A Highly Sensitive SERS Sensor for Quantitative Analysis of Glucose Based on the Chemical Etching of Silver Nanoparticles. J. Opt. 2015, 17, 114020. [Google Scholar] [CrossRef]
  74. Rong, Z.; Xiao, R.; Xing, S.; Xiong, G.; Yu, Z.; Wang, L.; Jia, X.; Wang, K.; Cong, Y.; Wang, S. SERS-Based Lateral Flow Assay for Quantitative Detection of C-Reactive Protein as an Early Bio-Indicator of a Radiation-Induced Inflammatory Response in Nonhuman Primates. Analyst 2018, 143, 2115–2121. [Google Scholar] [CrossRef]
  75. Li, B.; Wu, Y.; Wang, Z.; Xing, M.; Xu, W.; Zhu, Y.; Du, P.; Wang, X.; Yang, H. Non-Invasive Diagnosis of Crohn’s Disease Based on SERS Combined with PCA-SVM. Anal. Methods 2021, 13, 5264–5273. [Google Scholar] [CrossRef]
  76. Xu, H.; Bjerneld, E.J.; Käll, M.; Börjesson, L. Spectroscopy of Single Hemoglobin Molecules by Surface Enhanced Raman Scattering. Phys. Rev. Lett. 1999, 83, 4357–4360. [Google Scholar] [CrossRef] [Green Version]
  77. World Health Organization Report. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 30 January 2023).
  78. Blanco-Formoso, M.; Alvarez-Puebla, R.A. Cancer Diagnosis through Sers and Other Related Techniques. Int. J. Mol. Sci. 2020, 21, 2253. [Google Scholar] [CrossRef] [Green Version]
  79. Guerrini, L.; Pazos-Perez, N.; Garcia-Rico, E.; Alvarez-Puebla, R. Cancer Characterization and Diagnosis with SERS-Encoded Particles. Cancer Nanotechnol. 2017, 8, 5. [Google Scholar] [CrossRef]
  80. Kaur, B.; Kumar, S.; Kaushik, B.K. Recent Advancements in Optical Biosensors for Cancer Detection. Biosens. Bioelectron. 2022, 197, 113805. [Google Scholar] [CrossRef]
  81. Thenrajan, T.; Wilson, J. Biosensors for Cancer Theranostics. Biosens. Bioelectron. X 2022, 12, 100232. [Google Scholar] [CrossRef]
  82. Falkowski, P.; Lukaszewski, Z.; Gorodkiewicz, E. Potential of Surface Plasmon Resonance Biosensors in Cancer Detection. J. Pharm. Biomed. Anal. 2021, 194, 113802. [Google Scholar] [CrossRef]
  83. Fu, Q.; Zhang, X.; Song, J.; Yang, H. Plasmonic Gold Nanoagents for Cancer Imaging and Therapy. View 2021, 2, 20200149. [Google Scholar] [CrossRef]
  84. Azzouz, A.; Hejji, L.; Kim, K.H.; Kukkar, D.; Souhail, B.; Bhardwaj, N.; Brown, R.J.C.; Zhang, W. Advances in Surface Plasmon Resonance–Based Biosensor Technologies for Cancer Biomarker Detection. Biosens. Bioelectron. 2022, 197, 113767. [Google Scholar] [CrossRef] [PubMed]
  85. Sugumaran, S.; Jamlos, M.F.; Ahmad, M.N.; Bellan, C.S.; Schreurs, D. Nanostructured Materials with Plasmonic Nanobiosensors for Early Cancer Detection: A Past and Future Prospect. Biosens. Bioelectron. 2018, 100, 361–373. [Google Scholar] [CrossRef] [PubMed]
  86. Fattahi, Z.; Khosroushahi, A.Y.; Hasanzadeh, M. Recent Progress on Developing of Plasmon Biosensing of Tumor Biomarkers: Efficient Method towards Early Stage Recognition of Cancer. Biomed. Pharmacother. 2020, 132, 110850. [Google Scholar] [CrossRef] [PubMed]
  87. Bellassai, N.; D’Agata, R.; Jungbluth, V.; Spoto, G. Surface Plasmon Resonance for Biomarker Detection: Advances in Non-Invasive Cancer Diagnosis. Front. Chem. 2019, 7, 570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Usman, F.; Dennis, J.O.; Aljameel, A.I.; Ali, M.K.M.; Aldaghri, O.; Ibnaouf, K.H.; Zango, Z.U.; Beygisangchin, M.; Alsadig, A.; Meriaudeau, F. Plasmonic Biosensors for the Detection of Lung Cancer Biomarkers: A Review. Chemosensors 2021, 9, 326. [Google Scholar] [CrossRef]
  89. Yin, B.; Ho, W.K.H.; Xia, X.; Chan, C.K.W.; Zhang, Q.; Ng, Y.M.; Lam, C.Y.K.; Cheung, J.C.W.; Wang, J.; Yang, M.; et al. A Multilayered Mesoporous Gold Nanoarchitecture for Ultraeffective Near-Infrared Light-Controlled Chemo/Photothermal Therapy for Cancer Guided by SERS Imaging. Small 2023, 2023, 2206762. [Google Scholar] [CrossRef] [PubMed]
  90. Constantinou, M.; Hadjigeorgiou, K.; Abalde-cela, S.; Andreou, C. Label-Free Sensing with Metal Nanostructure-Based Surface- Enhanced Raman Spectroscopy for Cancer Diagnosis. ACS Appl. Nano Mater. 2022, 5, 12276–12299. [Google Scholar] [CrossRef] [PubMed]
  91. Rajput, S.; Pink, D.; Findlay, S.; Woolner, E.; Lewis, J.D.; McDermott, M.T. Application of Surface-Enhanced Raman Spectroscopy to Guide Therapy for Advanced Prostate Cancer Patients. ACS Sens. 2022, 7, 827–838. [Google Scholar] [CrossRef]
  92. Avci, E.; Yilmaz, H.; Sahiner, N.; Tuna, B.G.; Cicekdal, M.B.; Eser, M.; Basak, K.; Altıntoprak, F.; Zengin, I.; Dogan, S.; et al. Label-Free Surface Enhanced Raman Spectroscopy for Cancer Detection. Cancers 2022, 14, 5021. [Google Scholar] [CrossRef]
  93. Guerrini, L.; Alvarez-puebla, R.A. Surface-Enhanced Raman Spectroscopy in Cancer Diagnosis, Prognosis and Monitoring. Cancers 2019, 11, 748. [Google Scholar] [CrossRef] [Green Version]
  94. Pollap, A.; Paweł, S. Recent Advances in Sandwich SERS Immunosensors for Cancer Detection. Int. J. Mol. Sci. 2022, 23, 4740. [Google Scholar] [CrossRef]
  95. Vendrell, M.; Maiti, K.K.; Dhaliwal, K.; Chang, Y.T. Surface-Enhanced Raman Scattering in Cancer Detection and Imaging. Trends Biotechnol. 2013, 31, 249–257. [Google Scholar] [CrossRef]
  96. Moisoiu, V.; Iancu, S.D.; Stefancu, A.; Moisoiu, T.; Pardini, B.; Dragomir, M.P.; Crisan, N.; Avram, L.; Crisan, D.; Andras, I.; et al. SERS Liquid Biopsy: An Emerging Tool for Medical Diagnosis. Colloids Surfaces B Biointerfaces 2021, 208, 112064. [Google Scholar] [CrossRef]
  97. Shanmugasundaram, K.B.; Li, J.; Sina, A.I.; Wuethrich, A.; Trau, M. Toward Precision Oncology: SERS Microfluidic Systems for Multiplex Biomarker Analysis in Liquid Biopsy. Mater. Adv. 2022, 3, 1459–1471. [Google Scholar] [CrossRef]
  98. Song, C.Y.; Yang, Y.J.; Yang, B.Y.; Sun, Y.Z.; Zhao, Y.P.; Wang, L.H. An Ultrasensitive SERS Sensor for Simultaneous Detection of Multiple Cancer-Related MiRNAs. Nanoscale 2016, 8, 17365–17373. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, H.N.; Crawford, B.M.; Norton, S.J.; Vo-Dinh, T. Direct and Label-Free Detection of MicroRNA Cancer Biomarkers Using SERS-Based Plasmonic Coupling Interference (PCI) Nanoprobes. J. Phys. Chem. B 2019, 123, 10245–10251. [Google Scholar] [CrossRef]
  100. Guerrini, L.; Garcia-Rico, E.; O’loghlen, A.; Giannini, V.; Alvarez-Puebla, R.A. Surface-Enhanced Raman Scattering (Sers) Spectroscopy for Sensing and Characterization of Exosomes in Cancer Diagnosis. Cancers. 2021, 13, 2179. [Google Scholar] [CrossRef] [PubMed]
  101. Lee, J.U.; Kim, S.; Sim, S.J. SERS-Based Nanoplasmonic Exosome Analysis: Enabling Liquid Biopsy for Cancer Diagnosis and Monitoring Progression. BioChip J. 2020, 14, 231–241. [Google Scholar] [CrossRef]
  102. Vo-Dinh, T.; Allain, L.R.; Stokes, D.L. Cancer Gene Detection Using Surface-Enhanced Raman Scattering (SERS). J. Raman Spectrosc. 2002, 33, 511–516. [Google Scholar] [CrossRef]
  103. Zhu, D.; Li, A.; Di, Y.; Wang, Z.; Shi, J.; Ni, X.; Wang, Y. Interference-Free SERS Nanoprobes for Labeling and Imaging of MT1-MMP in Breast Cancer Cells. Nanotechnology 2022, 33, 115702. [Google Scholar] [CrossRef]
  104. Lin, J.; Chen, R.; Feng, S.; Pan, J.; Li, Y.; Chen, G.; Cheng, M.; Huang, Z.; Yu, Y.; Zeng, H. A Novel Blood Plasma Analysis Technique Combining Membrane Electrophoresis with Silver Nanoparticle-Based SERS Spectroscopy for Potential Applications in Noninvasive Cancer Detection. Nanomed. Nanotechnol. Biol. Med. 2011, 7, 655–663. [Google Scholar] [CrossRef]
  105. Fabris, L. SERS Tags: The Next Promising Tool for Personalized Cancer Detection? ChemNanoMat 2016, 2, 249–258. [Google Scholar] [CrossRef]
  106. Davis, R.M.; Campbell, J.L.; Burkitt, S.; Qiu, Z.; Kang, S.; Mehraein, M.; Miyasato, D.; Salinas, H.; Liu, J.T.C.; Zavaleta, C. A Raman Imaging Approach Using CD47 Antibody-Labeled SERS Nanoparticles for Identifying Breast Cancer and Its Potential to Guide Surgical Resection. Nanomaterials 2018, 8, 953. [Google Scholar] [CrossRef] [Green Version]
  107. Yang, J.; Wang, Z.; Zong, S.; Song, C.; Zhang, R.; Cui, Y. Distinguishing Breast Cancer Cells Using Surface-Enhanced Raman Scattering. Anal. Bioanal. Chem. 2012, 402, 1093–1100. [Google Scholar] [CrossRef]
  108. Dinish, U.S.; Balasundaram, G.; Chang, Y.T.; Olivo, M. Actively Targeted in Vivo Multiplex Detection of Intrinsic Cancer Biomarkers Using Biocompatible SERS Nanotags. Sci. Rep. 2014, 4, 4075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Liu, H.; Gao, X.; Xu, C.; Liu, D. SERS Tags for Biomedical Detection and Bioimaging. Theranostics 2022, 12, 1870–1903. [Google Scholar] [CrossRef] [PubMed]
  110. Report on Lung Cancer. Available online: https://www.chestnet.org/newsroom/chest-news/2020/07/world-lung-cancer-day-2020-fact-sheet (accessed on 30 January 2023).
  111. Mao, Y.; Sun, Y.; Xue, J.; Lu, W.; Cao, X. Ultra-Sensitive and High Efficiency Detection of Multiple Non-Small Cell Lung Cancer-Related MiRNAs on a Single Test Line in Catalytic Hairpin Assembly-Based SERS-LFA Strip. Anal. Chim. Acta 2021, 1178, 338800. [Google Scholar] [CrossRef] [PubMed]
  112. Xia, J.; Liu, Y.; Ran, M.; Lu, D.; Cao, X.; Wang, Y. SERS Platform Based on Bimetallic Au-Ag Nanowires-Decorated Filter Paper for Rapid Detection of MiR-196ain Lung Cancer Patients Serum. J. Chem. 2020, 2020, 5073451. [Google Scholar] [CrossRef]
  113. Cao, X.; Sun, Y.; Mao, Y.; Ran, M.; Liu, Y.; Lu, D.; Bi, C. Rapid and Sensitive Detection of Dual Lung Cancer-Associated MiRNA Biomarkers by a Novel SERS-LFA Strip Coupling with Catalytic Hairpin Assembly Signal Amplification. J. Mater. Chem. C 2021, 9, 3661–3671. [Google Scholar] [CrossRef]
  114. Cao, X.; Mao, Y.; Gu, Y.; Ge, S.; Lu, W.; Gu, Y.; Li, Z. Highly Sensitive and Simultaneous Detection of CtDNAs Related to Non-Small Cell Lung Cancer in Serum Using a Catalytic Hairpin Assembly Strategy in a SERS Microfluidic Chip. J. Mater. Chem. B 2022, 10, 6194–6206. [Google Scholar] [CrossRef]
  115. Cao, X.; Ge, S.; Zhou, X.; Mao, Y.; Sun, Y.; Lu, W.; Ran, M. A Dual-Signal Amplification Strategy Based on Pump-Free SERS Microfluidic Chip for Rapid and Ultrasensitive Detection of Non-Small Cell Lung Cancer-Related Circulating Tumour DNA in Mice Serum. Biosens. Bioelectron. 2022, 205, 114110. [Google Scholar] [CrossRef]
  116. Ye, L.P.; Hu, J.; Liang, L.; Zhang, C.Y. Surface-Enhanced Raman Spectroscopy for Simultaneous Sensitive Detection of Multiple MicroRNAs in Lung Cancer Cells. Chem. Commun. 2014, 50, 11883–11886. [Google Scholar] [CrossRef]
  117. Guo, T.; Li, W.; Qian, L.; Yan, X.; Cui, D.; Zhao, J.; Ni, H.; Zhao, X.; Zhang, Z.; Li, X.; et al. Highly-Selective Detection of EGFR Mutation Gene in Lung Cancer Based on Surface Enhanced Raman Spectroscopy and Asymmetric PCR. J. Pharm. Biomed. Anal. 2020, 190, 113522. [Google Scholar] [CrossRef]
  118. Shin, H.; Oh, S.; Hong, S.; Kang, M.; Kang, D.; Ji, Y.G.; Choi, B.H.; Kang, K.W.; Jeong, H.; Park, Y.; et al. Early-Stage Lung Cancer Diagnosis by Deep Learning-Based Spectroscopic Analysis of Circulating Exosomes. ACS Nano 2020, 14, 5435–5444. [Google Scholar] [CrossRef]
  119. Lei, J.; Yang, D.; Li, R.; Dai, Z.X.; Zhang, C.; Yu, Z.; Wu, S.; Pang, L.; Liang, S.; Zhang, Y. Label-Free Surface-Enhanced Raman Spectroscopy for Diagnosis and Analysis of Serum Samples with Different Types Lung Cancer. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2021, 261, 120021. [Google Scholar] [CrossRef]
  120. Wang, Z.; Hong, Y.; Yan, H.; Luo, H.; Zhang, Y.; Li, L.; Lu, S.; Chen, Y.; Wang, D.; Su, Y.; et al. Fabrication of Optoplasmonic Particles through Electroless Deposition and the Application in SERS-Based Screening of Nodule-Involved Lung Cancer. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 279, 121483. [Google Scholar] [CrossRef] [PubMed]
  121. Zhang, J.; Dong, Y.; Zhu, W.; Xie, D.; Zhao, Y.; Yang, D.; Li, M. Ultrasensitive Detection of Circulating Tumor DNA of Lung Cancer via an Enzymatically Amplified SERS-Based Frequency Shift Assay. ACS Appl. Mater. Interfaces 2019, 11, 18145–18152. [Google Scholar] [CrossRef] [PubMed]
  122. Jonak, S.T.; Liu, Z.; Liu, J.; Li, T.; D’Souza, B.V.; Schiaffino, A.; Oh, S.; Xie, Y.-H. Analyzing Bronchoalveolar Fluid Derived Small Extracellular Vesicles Using Single-Vesicle SERS for Non-Small Cell Lung Cancer Detection. Sens. Diagn. 2023, 2, 90–99. [Google Scholar] [CrossRef]
  123. Park, J.; Hwang, M.; Choi, B.; Jeong, H.; Jung, J.H.; Kim, H.K.; Hong, S.; Park, J.H.; Choi, Y. Exosome Classification by Pattern Analysis of Surface-Enhanced Raman Spectroscopy Data for Lung Cancer Diagnosis. Anal. Chem. 2017, 89, 6695–6701. [Google Scholar] [CrossRef]
  124. Wen, H.; Wang, H.; Hai, J.; He, S.; Chen, F.; Wang, B. Photochemical Synthesis of Porous CuFeSe 2 /Au Heterostructured Nanospheres as SERS Sensor for Ultrasensitive Detection of Lung Cancer Cells and Their Biomarkers. ACS Sustain. Chem. Eng. 2019, 7, 5200–5208. [Google Scholar] [CrossRef]
  125. Qiao, X.; Su, B.; Liu, C.; Song, Q.; Luo, D.; Mo, G.; Wang, T. Selective Surface Enhanced Raman Scattering for Quantitative Detection of Lung Cancer Biomarkers in Superparticle@MOF Structure. Adv. Mater. 2018, 30, 1702275. [Google Scholar] [CrossRef]
  126. Perumal, J.; Lee, P.; Dev, K.; Lim, H.Q.; Dinish, U.S.; Olivo, M. Machine Learning Assisted Real-Time Label-Free SERS Diagnoses of Malignant Pleural Effusion Due to Lung Cancer. Biosensors 2022, 12, 940. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, K.; Liu, X.; Man, B.; Yang, C.; Zhang, C.; Liu, M.; Zhang, Y.; Liu, L.; Chen, C. Label-Free and Stable Serum Analysis Based on Ag-NPs/PSi Surface-Enhanced Raman Scattering for Noninvasive Lung Cancer Detection. Biomed. Opt. Express 2018, 9, 4345. [Google Scholar] [CrossRef]
  128. Zhang, K.; Hao, C.; Huo, Y.; Man, B.; Zhang, C.; Yang, C.; Liu, M.; Chen, C. Label-Free Diagnosis of Lung Cancer with Tissue-Slice Surface-Enhanced Raman Spectroscopy and Statistical Analysis. Lasers Med. Sci. 2019, 34, 1849–1855. [Google Scholar] [CrossRef]
  129. Chon, H.; Lee, S.; Yoon, S.Y.; Chang, S.I.; Lim, D.W.; Choo, J. Simultaneous Immunoassay for the Detection of Two Lung Cancer Markers Using Functionalized SERS Nanoprobes. Chem. Commun. 2011, 47, 12515–12517. [Google Scholar] [CrossRef] [PubMed]
  130. Wu, P.; Gao, Y.; Lu, Y.; Zhang, H.; Cai, C. High Specific Detection and Near-Infrared Photothermal Therapy of Lung Cancer Cells with High SERS Active Aptamer-Silver-Gold Shell-Core Nanostructures. Analyst 2013, 138, 6501–6510. [Google Scholar] [CrossRef]
  131. Chen, Y.W.; Liu, T.Y.; Chen, P.J.; Chang, P.H.; Chen, S.Y. A High-Sensitivity and Low-Power Theranostic Nanosystem for Cell SERS Imaging and Selectively Photothermal Therapy Using Anti-EGFR-Conjugated Reduced Graphene Oxide/Mesoporous Silica/AuNPs Nanosheets. Small 2016, 12, 1458–1468. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, Y.; Ye, X.; Xu, G.; Jin, X.; Luan, M.; Lou, J.; Wang, L.; Huang, C.; Ye, J. Identification and Distinction of Non-Small-Cell Lung Cancer Cells by Intracellular SERS Nanoprobes. RSC Adv. 2016, 6, 5401–5407. [Google Scholar] [CrossRef]
  133. Huang, Y.; Xie, T.; Zou, K.; Gu, Y.; Yang, G.; Zhang, F.; Qu, L.L.; Yang, S. Ultrasensitive SERS Detection of Exhaled Biomarkers of Lung Cancer Using a Multifunctional Solid Phase Extraction Membrane. Nanoscale 2021, 13, 13344–13352. [Google Scholar] [CrossRef]
  134. Cai, C.; Liu, Y.; Li, J.; Wang, L.; Zhang, K. Serum Fingerprinting by Slippery Liquid-Infused Porous SERS for Non-Invasive Lung Cancer Detection. Analyst 2022, 147, 4426–4432. [Google Scholar] [CrossRef]
  135. Sivashanmugan, K.; Huang, W.L.; Lin, C.H.; Liao, J.D.; Lin, C.C.; Su, W.C.; Wen, T.C. Bimetallic Nanoplasmonic Gap-Mode SERS Substrate for Lung Normal and Cancer-Derived Exosomes Detection. J. Taiwan Inst. Chem. Eng. 2017, 80, 149–155. [Google Scholar] [CrossRef]
  136. Qian, K.; Wang, Y.; Hua, L.; Chen, A.; Zhang, Y. New Method of Lung Cancer Detection by Saliva Test Using Surface-Enhanced Raman Spectroscopy. Thorac. Cancer 2018, 9, 1556–1561. [Google Scholar] [CrossRef]
  137. Yang, T.; Guo, X.; Wu, Y.; Wang, H.; Fu, S.; Wen, Y.; Yang, H. Facile and Label-Free Detection of Lung Cancer Biomarker in Urine by Magnetically Assisted Surface-Enhanced Raman Scattering. ACS Appl. Mater. Interfaces 2014, 6, 20985–20993. [Google Scholar] [CrossRef]
  138. Breast Cancer Report. Available online: https://www.cancer.org/cancer/breast-cancer/about/how-common-is-breast-cancer.html (accessed on 30 January 2023).
  139. Moisoiu, T.; Iancu, S.D.; Burghelea, D.; Dragomir, M.P.; Iacob, G.; Stefancu, A.; Cozan, R.G.; Antal, O.; Bálint, Z.; Muntean, V.; et al. SERS Liquid Biopsy Profiling of Serum for the Diagnosis of Kidney Cancer. Biomedicines 2022, 10, 233. [Google Scholar] [CrossRef]
  140. Kim, S.; Kim, T.G.; Lee, S.H.; Kim, W.; Bang, A.; Moon, S.W.; Song, J.; Shin, J.H.; Yu, J.S.; Choi, S. Label-Free Surface-Enhanced Raman Spectroscopy Biosensor for On-Site Breast Cancer Detection Using Human Tears. ACS Appl. Mater. Interfaces 2020, 12, 7897–7904. [Google Scholar] [CrossRef] [PubMed]
  141. Teixeira, R.A.R.; Lataliza, A.A.B.; Raposo, N.R.B.; Costa, L.A.S.; Sant’Ana, A.C. Insights on the Transport of Tamoxifen by Gold Nanoparticles for MCF-7 Breast Cancer Cells Based on SERS Spectroscopy. Colloids Surf. B Biointerfaces 2018, 170, 712–717. [Google Scholar] [CrossRef] [PubMed]
  142. Nargis, H.F.; Nawaz, H.; Bhatti, H.N.; Jilani, K.; Saleem, M. Comparison of Surface Enhanced Raman Spectroscopy and Raman Spectroscopy for the Detection of Breast Cancer Based on Serum Samples. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2021, 246, 119034. [Google Scholar] [CrossRef] [PubMed]
  143. Xie, Y.; Su, X.; Wen, Y.; Zheng, C.; Li, M. Artificial Intelligent Label-Free SERS Profiling of Serum Exosomes for Breast Cancer Diagnosis and Postoperative Assessment. Nano Lett. 2022, 22, 7910–7918. [Google Scholar] [CrossRef] [PubMed]
  144. Wen, Y.; Truong, V.X.; Li, M. Real-Time Intraoperative Surface-Enhanced Raman Spectroscopy-Guided Thermosurgical Eradication of Residual Microtumors in Orthotopic Breast Cancer. Nano Lett. 2021, 21, 3066–3074. [Google Scholar] [CrossRef]
  145. Xiao, L.; Harihar, S.; Welch, D.R.; Zhou, A. Imaging of Epidermal Growth Factor Receptor on Single Breast Cancer Cells Using Surface-Enhanced Raman Spectroscopy. Anal. Chim. Acta 2014, 843, 73–82. [Google Scholar] [CrossRef] [Green Version]
  146. Liang, L.; Shen, Y.; Zhang, J.; Xu, S.; Xu, W.; Liang, C.; Han, B. Identification of Breast Cancer through Spectroscopic Analysis of Cell-Membrane Sialic Acid Expression. Anal. Chim. Acta 2018, 1033, 148–155. [Google Scholar] [CrossRef]
  147. Hernández-Arteaga, A.; de Jesús Zermeño Nava, J.; Kolosovas-Machuca, E.S.; Velázquez-Salazar, J.J.; Vinogradova, E.; José-Yacamán, M.; Navarro-Contreras, H.R. Diagnosis of Breast Cancer by Analysis of Sialic Acid Concentrations in Human Saliva by Surface-Enhanced Raman Spectroscopy of Silver Nanoparticles. Nano Res. 2017, 10, 3662–3670. [Google Scholar] [CrossRef]
  148. Han, Y.; Qiang, L.; Gao, Y.; Gao, J.; He, Q.; Liu, H.; Han, L.; Zhang, Y. Large-Area Surface-Enhanced Raman Spectroscopy Substrate by Hybrid Porous GaN with Au/Ag for Breast Cancer MiRNA Detection. Appl. Surf. Sci. 2021, 541, 148456. [Google Scholar] [CrossRef]
  149. Yarbakht, M.; Nikkhah, M.; Moshaii, A.; Weber, K.; Matthäus, C.; Cialla-May, D.; Popp, J. Simultaneous Isolation and Detection of Single Breast Cancer Cells Using Surface-Enhanced Raman Spectroscopy. Talanta 2018, 186, 44–52. [Google Scholar] [CrossRef]
  150. Zheng, Z.; Wu, L.; Li, L.; Zong, S.; Wang, Z.; Cui, Y. Simultaneous and Highly Sensitive Detection of Multiple Breast Cancer Biomarkers in Real Samples Using a SERS Microfluidic Chip. Talanta 2018, 188, 507–515. [Google Scholar] [CrossRef] [PubMed]
  151. Hameed, M.K.; Parambath, J.B.M.; Gul, M.T.; Khan, A.A.; Park, Y.; Han, C.; Mohamed, A.A. Arylated Gold Nanostars Aided SERS Study of Breast Cancer Cells. Appl. Surf. Sci. 2022, 583, 152504. [Google Scholar] [CrossRef]
  152. Kapara, A.; Brunton, V.G.; Graham, D.; Faulds, K. Characterisation of Estrogen Receptor Alpha (ERα) Expression in Breast Cancer Cells and Effect of Drug Treatment Using Targeted Nanoparticles and SERS. Analyst 2020, 145, 7225–7233. [Google Scholar] [CrossRef] [PubMed]
  153. Kapara, A.; Brunton, V.; Graham, D.; Faulds, K. Investigation of Cellular Uptake Mechanism of Functionalised Gold Nanoparticles into Breast Cancer Using SERS. Chem. Sci. 2020, 11, 5819–5829. [Google Scholar] [CrossRef]
  154. Lee, S.; Chon, H.; Lee, J.; Ko, J.; Chung, B.H.; Lim, D.W.; Choo, J. Rapid and Sensitive Phenotypic Marker Detection on Breast Cancer Cells Using Surface-Enhanced Raman Scattering (SERS) Imaging. Biosens. Bioelectron. 2014, 51, 238–243. [Google Scholar] [CrossRef]
  155. Choi, N.; Dang, H.; Das, A.; Sim, M.S.; Chung, I.Y.; Choo, J. SERS Biosensors for Ultrasensitive Detection of Multiple Biomarkers Expressed in Cancer Cells. Biosens. Bioelectron. 2020, 164, 112326. [Google Scholar] [CrossRef]
  156. Meng, S.; Chen, R.; Xie, J.; Li, J.; Cheng, J.; Xu, Y.; Cao, H.; Wu, X.; Zhang, Q.; Wang, H. Surface-Enhanced Raman Scattering Holography Chip for Rapid, Sensitive and Multiplexed Detection of Human Breast Cancer-Associated MicroRNAs in Clinical Samples. Biosens. Bioelectron. 2021, 190, 113470. [Google Scholar] [CrossRef]
  157. Weng, S.; Lin, D.; Lai, S.; Tao, H.; Chen, T.; Peng, M.; Qiu, S.; Feng, S. Highly Sensitive and Reliable Detection of MicroRNA for Clinically Disease Surveillance Using SERS Biosensor Integrated with Catalytic Hairpin Assembly Amplification Technology. Biosens. Bioelectron. 2022, 208, 114236. [Google Scholar] [CrossRef]
  158. Li, Y.; Qi, X.; Lei, C.; Qifeng, Q.; Zhang, S. Simultaneous SERS Detection and Imaging of Two Biomarkers on the Cancer Cell Surface by Self-Assembly of Branched DNA-Gold Nanoaggregates. Chem. Commun. 2014, 50, 9907–9909. [Google Scholar] [CrossRef] [Green Version]
  159. Lee, J.U.; Kim, W.H.; Lee, H.S.; Park, K.H.; Sim, S.J. Quantitative and Specific Detection of Exosomal MiRNAs for Accurate Diagnosis of Breast Cancer Using a Surface-Enhanced Raman Scattering Sensor Based on Plasmonic Head-Flocked Gold Nanopillars. Small 2019, 15, 1804968. [Google Scholar] [CrossRef]
  160. Zhong, Q.; Zhang, K.; Huang, X.; Lu, Y.; Zhao, J.; He, Y.; Liu, B. In Situ Ratiometric SERS Imaging of Intracellular Protease Activity for Subtype Discrimination of Human Breast Cancer. Biosens. Bioelectron. 2022, 207, 114194. [Google Scholar] [CrossRef] [PubMed]
  161. Li, L.; Liao, M.; Chen, Y.; Shan, B.; Li, M. Surface-Enhanced Raman Spectroscopy (SERS) Nanoprobes for Ratiometric Detection of Cancer Cells. J. Mater. Chem. B 2019, 7, 815–822. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, Y.; Kang, S.; Khan, A.; Ruttner, G.; Leigh, S.Y.; Murray, M.; Abeytunge, S.; Peterson, G.; Rajadhyaksha, M.; Dintzis, S.; et al. Quantitative Molecular Phenotyping with Topically Applied SERS Nanoparticles for Intraoperative Guidance of Breast Cancer Lumpectomy. Sci. Rep. 2016, 6, 21242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Zhang, Q.; Ma, R.; Zhang, Y.; Zhao, J.; Wang, Y.; Xu, Z. Dual-Aptamer-Assisted Ratiometric SERS Biosensor for Ultrasensitive and Precise Identification of Breast Cancer Exosomes. ACS Sens. 2023. [Google Scholar] [CrossRef]
  164. Shen, L.S.N.; Du, Y.; Wei, N.; Li, Q.; Li, S.M.; Sun, T.M.; Xu, S.; Wang, H.; Man, X.X.; Han, B. SERS Studies on Normal Epithelial and Cancer Cells Derived from Clinical Breast Cancer Specimens. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 237, 118364. [Google Scholar] [CrossRef]
  165. Lin, Y.; Gao, S.; Zheng, M.; Tang, S.; Lin, K.; Xie, S.; Yu, Y.; Lin, J. A Microsphere Nanoparticle Based-Serum Albumin Targeted Adsorption Coupled with Surface-Enhanced Raman Scattering for Breast Cancer Detection. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2021, 261, 120039. [Google Scholar] [CrossRef]
  166. Lin, Y.; Gao, J.; Tang, S.; Zhao, X.; Zheng, M.; Gong, W.; Xie, S.; Gao, S.; Yu, Y.; Lin, J. Label-Free Diagnosis of Breast Cancer Based on Serum Protein Purification Assisted Surface-Enhanced Raman Spectroscopy. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2021, 263, 120234. [Google Scholar] [CrossRef]
  167. Moisoiu, V.; Socaciu, A.; Stefancu, A.; Iancu, S.D.; Boros, I.; Alecsa, C.D.; Rachieriu, C.; Chiorean, A.R.; Eniu, D.; Leopold, N.; et al. Breast Cancer Diagnosis by Surface-Enhanced Raman Scattering (SERS) of Urine. Appl. Sci. 2019, 9, 806. [Google Scholar] [CrossRef] [Green Version]
  168. Akbar, S.; Majeed, M.I.; Nawaz, H.; Rashid, N.; Tariq, A.; Hameed, W.; Shakeel, S.; Dastgir, G.; Bari, R.Z.A.; Iqbal, M.; et al. Surface-Enhanced Raman Spectroscopic (SERS) Characterization of Low Molecular Weight Fraction of the Serum of Breast Cancer Patients with Principal Component Analysis (PCA) and Partial Least Square-Discriminant Analysis (PLS-DA). Anal. Lett. 2022, 55, 1588–1604. [Google Scholar] [CrossRef]
  169. Feng, S.; Huang, S.; Lin, D.; Chen, G.; Xu, Y.; Li, Y.; Huang, Z.; Pan, J.; Chen, R.; Zeng, H. Surface-Enhanced Raman Spectroscopy of Saliva Proteins for the Noninvasive Differentiation of Benign and Malignant Breast Tumors. Int. J. Nanomed. 2015, 10, 537–547. [Google Scholar] [CrossRef] [Green Version]
  170. Iancu, S.D.; Cozan, R.G.; Stefancu, A.; David, M.; Moisoiu, T.; Moroz-Dubenco, C.; Bajcsi, A.; Chira, C.; Andreica, A.; Leopold, L.F.; et al. SERS Liquid Biopsy in Breast Cancer. What Can We Learn from SERS on Serum and Urine? Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 273, 120992. [Google Scholar] [CrossRef]
  171. Lin, X.; Jia, X.; Lin, J.Y.; Wu, P.H.; Weng, Y.; Feng, S. A Comparative Study Based on Serum SERS Spectra in and on the Coffee Ring for High Precision Breast Cancer Detection. J. Raman Spectrosc. 2022, 53, 1371–1379. [Google Scholar] [CrossRef]
  172. Știufiuc, G.F.; Toma, V.; Buse, M.; Mărginean, R.; Morar-Bolba, G.; Culic, B.; Tetean, R.; Leopold, N.; Pavel, I.; Lucaciu, C.M.; et al. Solid Plasmonic Substrates for Breast Cancer Detection by Means of SERS Analysis of Blood Plasma. Nanomaterials 2020, 10, 1212. [Google Scholar] [CrossRef] [PubMed]
  173. Cervo, S.; Mansutti, E.; Del Mistro, G.; Spizzo, R.; Colombatti, A.; Steffan, A.; Sergo, V.; Bonifacio, A. SERS Analysis of Serum for Detection of Early and Locally Advanced Breast Cancer. Anal. Bioanal. Chem. 2015, 407, 7503–7509. [Google Scholar] [CrossRef] [PubMed]
  174. Lin, D.; Wang, Y.; Wang, T.; Zhu, Y.; Lin, X.; Lin, Y.; Feng, S. Metabolite Profiling of Human Blood by Surface-Enhanced Raman Spectroscopy for Surgery Assessment and Tumor Screening in Breast Cancer. Anal. Bioanal. Chem. 2020, 412, 1611–1618. [Google Scholar] [CrossRef] [PubMed]
  175. Gao, N.; Wang, Q.; Tang, J.; Yao, S.; Li, H.; Yue, X.; Fu, J.; Zhong, F.; Wang, T.; Wang, J. Non-Invasive SERS Serum Detection Technology Combined with Multivariate Statistical Algorithm for Simultaneous Screening of Cervical Cancer and Breast Cancer. Anal. Bioanal. Chem. 2021, 413, 4775–4784. [Google Scholar] [CrossRef]
  176. Vargas-Obieta, E.; Martínez-Espinosa, J.C.; Martínez-Zerega, B.E.; Jave-Suárez, L.F.; Aguilar-Lemarroy, A.; González-Solís, J.L. Breast Cancer Detection Based on Serum Sample Surface Enhanced Raman Spectroscopy. Lasers Med. Sci. 2016, 31, 1317–1324. [Google Scholar] [CrossRef]
  177. Ma, X.; Xiong, H.; Guo, J.; Liu, Z.; Han, Y.; Liu, M. Label-Free Breast Cancer Detection and Classification by Convolutional Neural Network-Based on Exosomes Surface-Enhanced Raman Scattering. J. Innov. Opt. Health Sci. 2022, 2022, 2244001. [Google Scholar] [CrossRef]
  178. Zhang, Z.; Wang, J.; Shanmugasundaram, K.B.; Yeo, B.; Möller, A.; Wuethrich, A.; Lin, L.L.; Trau, M. Tracking Drug-Induced Epithelial–Mesenchymal Transition in Breast Cancer by a Microfluidic Surface-Enhanced Raman Spectroscopy Immunoassay. Small 2020, 16, 1905614. [Google Scholar] [CrossRef]
  179. Zhu, J.; Zhou, J.; Guo, J.; Cai, W.; Liu, B.; Wang, Z.; Sun, Z. Surface-Enhanced Raman Spectroscopy Investigation on Human Breast Cancer Cells. Chem. Cent. J. 2013, 7, 37. [Google Scholar] [CrossRef] [Green Version]
  180. Brozek-Pluska, B.; Kopec, M.; Surmacki, J. Surface-Enhanced Raman Spectroscopy Analysis of Human Breast Cancer via Silver Nanoparticles: An Examination of Fabrication Methods. J. Spectrosc. 2018, 2018, 4893274. [Google Scholar] [CrossRef]
  181. Narayanan, N.; Kim, J.H.; Santhakumar, H.; Joseph, M.M.; Karunakaran, V.; Shamjith, S.; Saranya, G.; Sujai, P.T.; Jayasree, R.S.; Barman, I.; et al. Nanotheranostic Probe Built on Methylene Blue Loaded Cucurbituril [8] and Gold Nanorod: Targeted Phototherapy in Combination with SERS Imaging on Breast Cancer Cells. J. Phys. Chem. B 2021, 125, 13415–13424. [Google Scholar] [CrossRef] [PubMed]
  182. Feng, J.; Chen, L.; Xia, Y.; Xing, J.; Li, Z.; Qian, Q.; Wang, Y.; Wu, A.; Zeng, L.; Zhou, Y. Bioconjugation of Gold Nanobipyramids for SERS Detection and Targeted Photothermal Therapy in Breast Cancer. ACS Biomater. Sci. Eng. 2017, 3, 608–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Zeng, L.; Pan, Y.; Wang, S.; Wang, X.; Zhao, X.; Ren, W.; Lu, G.; Wu, A. Raman Reporter-Coupled Agcore@Aushell Nanostars for in Vivo Improved Surface Enhanced Raman Scattering Imaging and Near-Infrared-Triggered Photothermal Therapy in Breast Cancers. ACS Appl. Mater. Interfaces 2015, 7, 16781–16791. [Google Scholar] [CrossRef] [PubMed]
  184. Xinyue, L.; Keshavarz, M.; Panagiotis, K.; Roddan, A.; Yeatman, E.; Thompson, A. SERS Detection of Breast Cancer-Derived Exosomes Using a Nanostructured Pt-Black Template. Adv. Sens. Res. 2023, 2023, 2200039. [Google Scholar] [CrossRef]
  185. Pramanik, A.; Mayer, J.; Patibandla, S.; Gates, K.; Gao, Y.; Davis, D.; Seshadri, R.; Ray, P.C. Mixed-Dimensional Heterostructure Material-Based SERS for Trace Level Identification of Breast Cancer-Derived Exosomes. ACS Omega 2020, 5, 16602–16611. [Google Scholar] [CrossRef]
  186. Li, G.; Zhu, N.; Zhou, J.; Kang, K.; Zhou, X.; Ying, B.; Yi, Q.; Wu, Y. A Magnetic Surface-Enhanced Raman Scattering Platform for Performing Successive Breast Cancer Exosome Isolation and Analysis. J. Mater. Chem. B 2021, 9, 2709–2716. [Google Scholar] [CrossRef]
  187. Yang, Z.; Su, H.S.; You, E.M.; Liu, S.; Li, Z.; Zhang, Y. High Uniformity and Enhancement Au@AgNS 3D Substrates for the Diagnosis of Breast Cancer. ACS Omega 2022, 7, 15223–15230. [Google Scholar] [CrossRef]
  188. Wang, X.P.; Walkenfort, B.; König, M.; König, L.; Kasimir-Bauer, S.; Schlücker, S. Fast and Reproducible ISERS Microscopy of Single HER2-Positive Breast Cancer Cells Using Gold Nanostars as SERS Nanotags. Faraday Discuss. 2017, 205, 377–386. [Google Scholar] [CrossRef]
  189. Chen, Z.; Shen, X.; Chen, S.; Dai, K. Gastric Cancer Prewarning and Early Diagnosis System; Springer: Berlin/Heidelberg, Germany, 2017; ISBN 9789402409499. [Google Scholar]
  190. Hunter, R.A.; Asare-Werehene, M.; Mandour, A.; Tsang, B.K.; Anis, H. Determination of Chemoresistance in Ovarian Cancer by Simultaneous Quantification of Exosomes and Exosomal Cisplatin with Surface Enhanced Raman Scattering. Sens. Actuators B Chem. 2022, 354, 131237. [Google Scholar] [CrossRef]
  191. Moothanchery, M.; Perumal, J.; Mahyuddin, A.P.; Singh, G.; Choolani, M.; Olivo, M. Rapid and Sensitive Detection of Ovarian Cancer Biomarker Using a Portable Single Peak Raman Detection Method. Sci. Rep. 2022, 12, 12459. [Google Scholar] [CrossRef]
  192. Sarkar, S.; Gogoi, M.; Mahato, M.; Joshi, A.B.; Baruah, A.J.; Kodgire, P.; Boruah, P. Biosensors for Detection of Prostate Cancer: A Review. Biomed. Microdevices 2022, 24, 32. [Google Scholar] [CrossRef]
  193. Turan, E.; Zengin, A.; Suludere, Z.; Kalkan, N.Ö.; Tamer, U. Construction of a Sensitive and Selective Plasmonic Biosensor for Prostate Specific Antigen by Combining Magnetic Molecularly-Imprinted Polymer and Surface-Enhanced Raman Spectroscopy. Talanta 2022, 237, 122926. [Google Scholar] [CrossRef]
  194. Haroon, M.; Tahir, M.; Nawaz, H.; Majeed, M.I.; Al-Saadi, A.A. Surface-Enhanced Raman Scattering (SERS) Spectroscopy for Prostate Cancer Diagnosis: A Review. Photodiagn. Photodyn. Ther. 2022, 37, 102690. [Google Scholar] [CrossRef] [PubMed]
  195. Ashrafizadeh, M.; Aghamiri, S.; Tan, S.C.; Zarrabi, A.; Sharifi, E.; Rabiee, N.; Kadumudi, F.B.; Pirouz, A.D.; Delfi, M.; Byrappa, K.; et al. Nanotechnological Approaches in Prostate Cancer Therapy: Integration of Engineering and Biology. Nano Today 2022, 45, 101532. [Google Scholar] [CrossRef]
  196. Gaba, F.; Tipping, W.J.; Salji, M.; Faulds, K.; Graham, D.; Leung, H.Y. Raman Spectroscopy in Prostate Cancer: Techniques, Applications and Advancements. Cancers. 2022, 14, 1535. [Google Scholar] [CrossRef]
  197. Pandey, A.; Sarkar, S.; Pandey, S.K.; Srivastava, A. Silica Nanospheres Coated Silver Islands as an Effective Opto-Plasmonic SERS Active Platform for Rapid and Sensitive Detection of Prostate Cancer Biomarkers. Molecules 2022, 27, 7821. [Google Scholar] [CrossRef] [PubMed]
  198. Wei, Y.; Zhu, Y.Y.; Wang, M.L. Surface-Enhanced Raman Spectroscopy of Gastric Cancer Serum with Gold Nanoparticles/Silicon Nanowire Arrays. Optik 2016, 127, 7902–7907. [Google Scholar] [CrossRef]
  199. Ito, H.; Inoue, H.; Hasegawa, K.; Hasegawa, Y.; Shimizu, T.; Kimura, S.; Onimaru, M.; Ikeda, H.; Kudo, S. ei Use of Surface-Enhanced Raman Scattering for Detection of Cancer-Related Serum-Constituents in Gastrointestinal Cancer Patients. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 599–608. [Google Scholar] [CrossRef]
  200. Ge, S.; Ran, M.; Mao, Y.; Sun, Y.; Zhou, X.; Li, L.; Cao, X. A Novel DNA Biosensor for the Ultrasensitive Detection of DNA Methyltransferase Activity Based on a High-Density ‘Hot Spot’ SERS Substrate and Rolling Circle Amplification Strategy. Analyst 2021, 146, 5326–5336. [Google Scholar] [CrossRef]
  201. Feng, S.; Chen, R.; Lin, J.; Pan, J.; Wu, Y.; Li, Y.; Chen, J.; Zeng, H. Gastric Cancer Detection Based on Blood Plasma Surface-Enhanced Raman Spectroscopy Excited by Polarized Laser Light. Biosens. Bioelectron. 2011, 26, 3167–3174. [Google Scholar] [CrossRef]
  202. Pan, H.; Dong, Y.; Gong, L.; Zhai, J.; Song, C.; Ge, Z.; Su, Y.; Zhu, D.; Chao, J.; Su, S.; et al. Sensing Gastric Cancer Exosomes with MoS2-Based SERS Aptasensor. Biosens. Bioelectron. 2022, 215, 114553. [Google Scholar] [CrossRef]
  203. Liu, Z.; Li, T.; Wang, Z.; Liu, J.; Huang, S.; Min, B.H.; An, J.Y.; Kim, K.M.; Kim, S.; Chen, Y.; et al. Gold Nanopyramid Arrays for Non-Invasive Surface-Enhanced Raman Spectroscopy-Based Gastric Cancer Detection via SEVs. ACS Appl. Nano Mater. 2022, 5, 12506–12517. [Google Scholar] [CrossRef]
  204. Gayoung, E.; Hongki, K.; Ahreum, H.; Hye-Young, S.; Yuna, C.; Jeong, M.; Donghyeong, K.; Miyeon, L.; Eun-Kyung, L.; Jinyoung, J.; et al. Nanogap-Rich Au Nanowire SERS Sensor for Ultrasensitive Telomerase Activity Detection. Adv. Funct. Mater. 2017, 27, 1701832. [Google Scholar] [CrossRef]
  205. Chen, Y.; Cheng, S.; Zhang, A.; Song, J.; Chang, J.; Wang, K.; Gao, G.; Zhang, Y.; Li, S.; Liu, H.; et al. Salivary Analysis Based on Surface Enhanced Raman Scattering Sensors Distinguishes Early and Advanced Gastric Cancer Patients from Healthy Persons. J. Biomed. Nanotechnol. 2018, 14, 1773–1784. [Google Scholar] [CrossRef]
  206. Cao, X.; Ge, S.; Hua, W.; Zhou, X.; Lu, W.; Gu, Y.; Li, Z.; Qian, Y. A Pump-Free and High-Throughput Microfluidic Chip for Highly Sensitive SERS Assay of Gastric Cancer-Related Circulating Tumor DNA via a Cascade Signal Amplification Strategy. J. Nanobiotechnol. 2022, 20, 271. [Google Scholar] [CrossRef]
  207. Chen, Y.; Zhang, Y.; Pan, F.; Liu, J.; Wang, K.; Zhang, C.; Cheng, S.; Lu, L.; Zhang, W.; Zhang, Z.; et al. Breath Analysis Based on Surface-Enhanced Raman Scattering Sensors Distinguishes Early and Advanced Gastric Cancer Patients from Healthy Persons. ACS Nano 2016, 10, 8169–8179. [Google Scholar] [CrossRef]
  208. Huang, L.; Zhu, Y.; Xu, C.; Cai, Y.; Yi, Y.; Li, K.; Ren, X.; Jiang, D.; Ge, Y.; Liu, X.; et al. Noninvasive Diagnosis of Gastric Cancer Based on Breath Analysis with a Tubular Surface-Enhanced Raman Scattering Sensor. ACS Sens. 2022, 7, 1439–1450. [Google Scholar] [CrossRef]
  209. Cao, D.; Lin, H.; Liu, Z.; Qiu, J.; Ge, S.; Hua, W.; Cao, X.; Qian, Y.; Xu, H.; Zhu, X. PCA-TLNN-Based SERS Analysis Platform for Label-Free Detection and Identification of Cisplatin-Treated Gastric Cancer. Sens. Actuators B Chem. 2023, 375, 132903. [Google Scholar] [CrossRef]
  210. Guo, L.; Li, Y.; Huang, F.; Dong, J.; Li, F.; Yang, X.; Zhu, S.; Yang, M. Identification and Analysis of Serum Samples by Surface-Enhanced Raman Spectroscopy Combined with Characteristic Ratio Method and PCA for Gastric Cancer Detection. J. Innov. Opt. Health Sci. 2019, 12, 1950003. [Google Scholar] [CrossRef] [Green Version]
  211. Ma, J.; Zhou, H.; Gong, L.; Liu, S.; Zhou, Z.; Mao, W.; Zheng, R. Distinction of Gastric Cancer Tissue Based on Surface-Enhanced Raman Spectroscopy. Opt. Health Care Biomed. Opt. V 2012, 8553, 855328. [Google Scholar] [CrossRef]
  212. Chen, Y.; Chen, Y.; Chen, G.; Zheng, X.; He, C.; Feng, S.; Lin, X.; Chen, R.; Zeng, H. Discrimination of Gastric Cancer from Normal by Serum RNA Based on Surface-Enhanced Raman Spectroscopy (SERS) and Multivariate Analysis. Med. Phys. 2012, 39, 5664–5668. [Google Scholar] [CrossRef] [Green Version]
  213. Feng, S.Y.; Pan, J.J.; Wu, Y.A.; Lin, D.; Chen, Y.P.; Xi, G.Q.; Lin, J.Q.; Chen, R. Study on Gastric Cancer Blood Plasma Based on Surface-Enhanced Raman Spectroscopy Combined with Multivariate Analysis. Sci. China Life Sci. 2011, 54, 828–834. [Google Scholar] [CrossRef] [Green Version]
  214. Aslam, M.A.; Xue, C.; Wang, K.; Chen, Y.; Zhang, A.; Cai, W.; Ma, L.; Yang, Y.; Sun, X.; Liu, M.; et al. SVM Based Classification and Prediction System for Gastric Cancer Using Dominant Features of Saliva. Nano Biomed. Eng. 2020, 12, 1–13. [Google Scholar] [CrossRef]
  215. Aslam, M.A.; Xue, C.; Liu, M.; Wang, K.; Cui, D. Classification and Prediction of Gastric Cancer from Saliva Diagnosis Using Artificial Neural Network. Eng. Lett. 2020, 29, 2. [Google Scholar]
  216. Avram, L.; Iancu, S.D.; Stefancu, A.; Moisoiu, V.; Colnita, A.; Marconi, D.; Donca, V.; Buzdugan, E.; Craciun, R.; Leopold, N.; et al. SERS-Based Liquid Biopsy of Gastrointestinal Tumors Using a Portable Raman Device Operating in a Clinical Environment. J. Clin. Med. 2020, 9, 212. [Google Scholar] [CrossRef] [Green Version]
  217. Li, X.; Yang, T.; Li, S.; Wang, D.; Song, Y.; Yu, K. Different Classification Algorithms and Serum Surface Enhanced Raman Spectroscopy for Noninvasive Discrimination of Gastric Diseases. J. Raman Spectrosc. 2016, 47, 917–925. [Google Scholar] [CrossRef]
  218. Li, S.X.; Zhang, Y.J.; Zeng, Q.Y.; Li, L.F.; Guo, Z.Y.; Liu, Z.M.; Xiong, H.L.; Liu, S.H. Potential of Cancer Screening with Serum Surface-Enhanced Raman Spectroscopy and a Support Vector Machine. Laser Phys. Lett. 2014, 11, 065603. [Google Scholar] [CrossRef]
  219. Moisoiu, T.; Dragomir, M.P.; Iancu, S.D.; Schallenberg, S.; Birolo, G.; Ferrero, G.; Burghelea, D.; Stefancu, A.; Cozan, R.G.; Licarete, E.; et al. Combined MiRNA and SERS Urine Liquid Biopsy for the Point-of-Care Diagnosis and Molecular Stratification of Bladder Cancer. Mol. Med. 2022, 28, 39. [Google Scholar] [CrossRef]
  220. Gao, S.; Lin, Y.; Zhao, X.; Gao, J.; Xie, S.; Gong, W.; Yu, Y.; Lin, J. Label-Free Surface Enhanced Raman Spectroscopy Analysis of Blood Serum via Coffee Ring Effect for Accurate Diagnosis of Cancers. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 267, 120605. [Google Scholar] [CrossRef]
  221. Meng, C.; Li, H.; Chen, C.; Wu, W.; Gao, J.; Lai, Y.; Ka, M.; Zhu, M.; Lv, X.; Chen, F.; et al. Serum Raman Spectroscopy Combined with Gaussian—Convolutional Neural Network Models to Quickly Detect Liver Cancer Patients. Spectrosc. Lett. 2022, 55, 79–90. [Google Scholar] [CrossRef]
  222. Ni, J.-T.; Huang, M.-Y.; Ji, W.; Wang, L.; Sun, T.-D. Recent Advances in Surface-Enhanced Raman Scattering for Liver Cancer Detection. Chin. J. Anal. Chem. 2022, 50, 100180. [Google Scholar] [CrossRef]
  223. Zhang, Q.; Hou, D.; Wen, X.; Xin, M.; Li, Z.; Wu, L.; Pathak, J.L. Gold Nanomaterials for Oral Cancer Diagnosis and Therapy: Advances, Challenges, and Prospects. Mater. Today Bio 2022, 15, 100333. [Google Scholar] [CrossRef] [PubMed]
  224. Zadeh, F.A.; Shahhosseini, E.; Rasoolzadegan, S.; Özbolat, G.; Farahbod, F. Au Nanoparticles in the Diagnosis and Treatment of Ovarian Cancer: A New Horizon in the Personalized Medicine. Nanomed. Res. J. 2022, 7, 1–18. [Google Scholar] [CrossRef]
  225. Wang, Y.; Zhang, Y.; Du, Q.; Cao, D.; Lu, X.; Meng, Z. Sensitive SERS Detection of Oral Squamous Cell Carcinoma-Related MiRNAs in Saliva via a Gold Nanohexagon Array Coupled with Hybridization Chain Reaction Amplification. Anal. Methods 2022, 14, 4563–4575. [Google Scholar] [CrossRef]
  226. Fălămaș, A.; Rotaru, H.; Hedeșiu, M. Surface-Enhanced Raman Spectroscopy (SERS) Investigations of Saliva for Oral Cancer Diagnosis. Lasers Med. Sci. 2020, 35, 1393–1401. [Google Scholar] [CrossRef]
  227. Wang, K.; Qiu, Y.; Wu, C.; Wen, Z.N.; Li, Y. Surface-Enhanced Raman Spectroscopy and Multivariate Analysis for the Diagnosis of Oral Squamous Cell Carcinoma. J. Raman Spectrosc. 2023. [Google Scholar] [CrossRef]
  228. Lin, Y.; Lin, J.; Zheng, M.; Gong, W.; Li, H.; Shu, Z.; Du, W.; Gao, S.; Yu, Y. Quantitative and Direct Serum Albumin Detection by Label-Free SERS Using Tunable Hydroxyapatite Nanostructure for Prostate Cancer Detection. Anal. Chim. Acta 2022, 1221, 340101. [Google Scholar] [CrossRef]
  229. Zhao, J.; Wang, J.; Liu, Y.; Han, X.X.; Xu, B.; Ozaki, Y.; Zhao, B. Detection of Prostate Cancer Biomarkers via a SERS-Based Aptasensor. Biosens. Bioelectron. 2022, 216, 114660. [Google Scholar] [CrossRef]
  230. Lu, Y.; Zhan, C.; Yu, L.; Yu, Y.; Jia, H.; Chen, X.; Zhang, D.; Gao, R. Multifunctional Nanocone Array as Solid Immunoassay Plate and SERS Substrate for the Early Diagnosis of Prostate Cancer on Microfluidic Chip. Sens. Actuators B Chem. 2023, 376, 133046. [Google Scholar] [CrossRef]
  231. Munteanu, V.C.; Munteanu, R.A.; Gulei, D.; Mărginean, R.; Schițcu, V.H.; Onaciu, A.; Toma, V.; Știufiuc, G.F.; Coman, I.; Știufiuc, R.I. New Insights into the Multivariate Analysis of SER Spectra Collected on Blood Samples for Prostate Cancer Detection: Towards a Better Understanding of the Role Played by Different Biomolecules on Cancer Screening: A Preliminary Study. Cancers 2022, 14, 3227. [Google Scholar] [CrossRef] [PubMed]
  232. Stefancu, A.; Moisoiu, V.; Couti, R.; Andras, I.; Rahota, R.; Crisan, D.; Pavel, I.E.; Socaciu, C.; Leopold, N.; Crisan, N. Combining SERS Analysis of Serum with PSA Levels for Improving the Detection of Prostate Cancer. Nanomedicine 2018, 13, 2455–2467. [Google Scholar] [CrossRef] [Green Version]
  233. Liyanage, T.; Alharbi, B.; Quan, L.; Esquela-Kerscher, A.; Slaughter, G. Plasmonic-Based Biosensor for the Early Diagnosis of Prostate Cancer. ACS Omega 2022, 7, 2411–2418. [Google Scholar] [CrossRef] [PubMed]
  234. Zhao, X.; Xu, Q.; Lin, Y.; Du, W.; Bai, X.; Gao, J.; Li, T.; Huang, Y.; Yu, Y.; Wu, X.; et al. Label-free surface-enhanced Raman spectroscopy detectionof prostate cancer combined with multivariate statistical algorithm. J. Raman Spectrosc. 2022, 53, 1861–1870. [Google Scholar] [CrossRef]
  235. Sayyadi, N.; Justiniano, I.; Wang, Y.; Zheng, X.; Zhang, W.; Jiang, L.; Polikarpov, D.M.; Willows, R.D.; Gillatt, D.; Campbell, D.; et al. Detection of Rare Prostate Cancer Cells in Human Urine Offers Prospect of Non-Invasive Diagnosis. Sci. Rep. 2022, 12, 18452. [Google Scholar] [CrossRef] [PubMed]
  236. Lu, S.; Lin, S.; Zhang, H.; Liang, L.; Shen, S. Methods of Respiratory Virus Detection: Advances towards Point-of-Care for Early Intervention. Micromachines 2021, 12, 697. [Google Scholar] [CrossRef] [PubMed]
  237. Omidifar, N.; Lankarani, K.B.; Moghadami, M.; Shokripour, M.; Chashmpoosh, M.; Mousavi, S.M.; Hashemi, S.A.; Gholami, A. Different Laboratory Diagnosis Methods of COVID-19: A Systematic Review. Arch. Clin. Infect. Dis. 2021, 16, e110667. [Google Scholar] [CrossRef]
  238. Mousavi, S.M.; Hashemi, S.A.; Rahmanian, V.; Kalashgrani, M.Y. Highly Sensitive Flexible SERS-Based Sensing Platform for Detection of Biosensors Highly Sensitive Flexible SERS-Based Sensing Platform for Detection of COVID-19. Biosensors 2022, 12, 466. [Google Scholar] [CrossRef]
  239. Stöckel, S.; Kirchhoff, J.; Neugebauer, U.; Rösch, P.; Popp, J. The Application of Raman Spectroscopy for the Detection and Identification of Microorganisms. J. Raman Spectrosc. 2016, 47, 89–109. [Google Scholar] [CrossRef]
  240. Soler, M.; Estevez, M.C.; Cardenosa-Rubio, M.; Astua, A.; Lechuga, L.M. How Nanophotonic Label-Free Biosensors Can Contribute to Rapid and Massive Diagnostics of Respiratory Virus Infections: COVID-19 Case. ACS Sens. 2020, 5, 2663–2678. [Google Scholar] [CrossRef]
  241. Iravani, S. Nano- And Biosensors for the Detection of SARS-CoV-2: Challenges and Opportunities. Mater. Adv. 2020, 1, 3092–3103. [Google Scholar] [CrossRef]
  242. Zhang, D.; Zhang, X.; Ma, R.; Deng, S.; Wang, X.; Wang, X.; Zhang, X.; Huang, X.; Liu, Y.; Li, G.; et al. Ultra-Fast and Onsite Interrogation of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) in Waters via Surface Enhanced Raman Scattering (SERS). Water Res. 2021, 200, 117243. [Google Scholar] [CrossRef] [PubMed]
  243. Joung, Y.; Kim, K.; Lee, S.; Chun, B.-S.; Lee, S.; Hwang, J.; Choi, S.; Kang, T.; Lee, M.-K.; Chen, L.; et al. Rapid and Accurate On-Site Immunodiagnostics of Highly Contagious Severe Acute Respiratory Syndrome Coronavirus 2 Using Portable Surface-Enhanced Raman Scattering-Lateral Flow Assay Reader. ACS Sens. 2022, 7, 3470–3480. [Google Scholar] [CrossRef] [PubMed]
  244. Saviñon-Flores, F.; Méndez, E.; López-Castaños, M.; Carabarin-Lima, A.; López-Castaños, K.A.; González-Fuentes, M.A.; Méndez-Albores, A. A Review on Sers-Based Detection of Human Virus Infections: Influenza and Coronavirus. Biosensors 2021, 11, 66. [Google Scholar] [CrossRef] [PubMed]
  245. Lim, J.Y.; Nam, J.S.; Yang, S.E.; Shin, H.; Jang, Y.H.; Bae, G.U.; Kang, T.; Lim, K.I.; Choi, Y. Identification of Newly Emerging Influenza Viruses by Surface-Enhanced Raman Spectroscopy. Anal. Chem. 2015, 87, 11652–11659. [Google Scholar] [CrossRef] [PubMed]
  246. Zhang, K.; Wang, Z.; Liu, H.; Perea-López, N.; Ranasinghe, J.C.; Bepete, G.; Minns, A.M.; Rossi, R.M.; Lindner, S.E.; Huang, S.X.; et al. Understanding the Excitation Wavelength Dependence and Thermal Stability of the SARS-CoV-2 Receptor-Binding Domain Using Surface-Enhanced Raman Scattering and Machine Learning. ACS Photonics 2022, 9, 2963–2972. [Google Scholar] [CrossRef]
  247. Yang, Y.; Xu, B.; Murray, J.; Haverstick, J.; Chen, X.; Tripp, R.A.; Zhao, Y. Rapid and Quantitative Detection of Respiratory Viruses Using Surface-Enhanced Raman Spectroscopy and Machine Learning. Biosens. Bioelectron. 2022, 217, 114721. [Google Scholar] [CrossRef]
  248. Ye, J.; Yeh, Y.; Xue, Y.; Wang, Z.; Zhang, N.; Liu, H.; Zhang, K.; Ricker, R.; Yu, Z.; Roder, A. Accurate Virus Identi Fi Cation with Interpretable Raman Signatures by Machine Learning. Proc. Natl. Acad. Sci. USA 2022, 199, e2118836119. [Google Scholar] [CrossRef] [PubMed]
  249. Carlomagno, C.; Bertazioli, D.; Gualerzi, A.; Picciolini, S.; Banfi, P.I.; Lax, A.; Messina, E.; Navarro, J.; Bianchi, L.; Caronni, A.; et al. COVID-19 Salivary Raman Fingerprint: Innovative Approach for the Detection of Current and Past SARS-CoV-2 Infections. Sci. Rep. 2021, 11, 4943. [Google Scholar] [CrossRef]
  250. Zavyalova, E.; Ambartsumyan, O.; Zhdanov, G.; Gribanyov, D.; Gushchin, V.; Tkachuk, A.; Rudakova, E.; Nikiforova, M.; Kuznetsova, N.; Popova, L.; et al. Sers-Based Aptasensor for Rapid Quantitative Detection of Sars-Cov-2. Nanomaterials 2021, 11, 1394. [Google Scholar] [CrossRef]
  251. Hwang, C.S.H.; Lee, S.; Lee, S.; Kim, H.; Kang, T.; Lee, D.; Jeong, K.H. Highly Adsorptive Au-TiO2 Nanocomposites for the SERS Face Mask Allow the Machine-Learning-Based Quantitative Assay of SARS-CoV-2 in Artificial Breath Aerosols. ACS Appl. Mater. Interfaces 2022, 14, 54550–54557. [Google Scholar] [CrossRef] [PubMed]
  252. Zhang, Z.; Jiang, S.; Wang, X.; Dong, T.; Wang, Y.; Li, D.; Gao, X.; Qu, Z.; Li, Y. A Novel Enhanced Substrate for Label-Free Detection of SARS-CoV-2 Based on Surface-Enhanced Raman Scattering. Sens. Actuators B Chem. 2022, 359, 131568. [Google Scholar] [CrossRef] [PubMed]
  253. Wang, C.; Wang, C.; Wang, X.; Wang, K.; Zhu, Y.; Rong, Z.; Wang, W.; Xiao, R.; Wang, S. Magnetic SERS Strip for Sensitive and Simultaneous Detection of Respiratory Viruses. ACS Appl. Mater. Interfaces 2019, 11, 19495–19505. [Google Scholar] [CrossRef]
  254. Zhang, D.; Huang, L.; Liu, B.; Ge, Q.; Dong, J.; Zhao, X. Rapid and Ultrasensitive Quantification of Multiplex Respiratory Tract Infection Pathogen via Lateral Flow Microarray Based on SERS Nanotags. Theranostics 2019, 9, 4849–4859. [Google Scholar] [CrossRef] [PubMed]
  255. Yang, Y.; Peng, Y.; Lin, C.; Long, L.; Hu, J.; He, J.; Zeng, H.; Huang, Z.; Li, Z.Y.; Tanemura, M.; et al. Human ACE2-Functionalized Gold “Virus-Trap” Nanostructures for Accurate Capture of SARS-CoV-2 and Single-Virus SERS Detection. Nano-Micro Lett. 2021, 13, 109. [Google Scholar] [CrossRef]
  256. Peng, Y.; Lin, C.; Long, L.; Masaki, T.; Tang, M.; Yang, L.; Liu, J.; Huang, Z.; Li, Z.; Luo, X.; et al. Charge-Transfer Resonance and Electromagnetic Enhancement Synergistically Enabling MXenes with Excellent SERS Sensitivity for SARS-CoV-2 S Protein Detection. Nano-Micro Lett. 2021, 13, 52. [Google Scholar] [CrossRef]
  257. Gu, M.M.; Guan, P.C.; Xu, S.S.; Li, H.M.; Kou, Y.C.; Lin, X.D.; Kathiresan, M.; Song, Y.; Zhang, Y.J.; Jin, S.Z.; et al. Ultrasensitive Detection of SARS-CoV-2 S Protein with Aptamers Biosensor Based on Surface-Enhanced Raman Scattering. J. Chem. Phys. 2023, 158, 024203. [Google Scholar] [CrossRef]
  258. Lim, J.Y.; Nam, J.S.; Shin, H.; Park, J.; Song, H.I.; Kang, M.; Lim, K.I.; Choi, Y. Identification of Newly Emerging Influenza Viruses by Detecting the Virally Infected Cells Based on Surface Enhanced Raman Spectroscopy and Principal Component Analysis. Anal. Chem. 2019, 91, 5677–5684. [Google Scholar] [CrossRef]
  259. Eom, G.; Hwang, A.; Kim, H.; Yang, S.; Lee, D.K.; Song, S.; Ha, K.; Jeong, J.; Jung, J.; Lim, E.K.; et al. Diagnosis of Tamiflu-Resistant Influenza Virus in Human Nasal Fluid and Saliva Using Surface-Enhanced Raman Scattering. ACS Sens. 2019, 4, 2282–2287. [Google Scholar] [CrossRef]
  260. Zhang, M.; Li, X.; Pan, J.; Zhang, Y.; Zhang, L.; Wang, C.; Yan, X.; Liu, X.; Lu, G. Ultrasensitive Detection of SARS-CoV-2 Spike Protein in Untreated Saliva Using SERS-Based Biosensor. Biosens. Bioelectron. 2021, 190, 113421. [Google Scholar] [CrossRef]
  261. Yadav, S.; Sadique, M.A.; Ranjan, P.; Kumar, N.; Singhal, A.; Srivastava, A.K.; Khan, R. Sers Based Lateral Flow Immunoassay for Point-of-Care Detection of Sars-Cov-2 in Clinical Samples. ACS Appl. Bio Mater. 2021, 4, 2974–2995. [Google Scholar] [CrossRef]
  262. Chen, S.; Meng, L.; Wang, L.; Huang, X.; Ali, S.; Chen, X.; Yu, M.; Yi, M.; Li, L.; Chen, X.; et al. SERS-Based Lateral Flow Immunoassay for Sensitive and Simultaneous Detection of Anti-SARS-CoV-2 IgM and IgG Antibodies by Using Gap-Enhanced Raman Nanotags. Sens. Actuators B Chem. 2021, 348, 130706. [Google Scholar] [CrossRef]
  263. Liu, H.; Dai, E.; Xiao, R.; Zhou, Z.; Zhang, M.; Bai, Z.; Shao, Y.; Qi, K.; Tu, J.; Wang, C.; et al. Development of a SERS-Based Lateral Flow Immunoassay for Rapid and Ultra-Sensitive Detection of Anti-SARS-CoV-2 IgM/IgG in Clinical Samples. Sens. Actuators B Chem. 2021, 329, 129196. [Google Scholar] [CrossRef] [PubMed]
  264. Antoine, D.; Mohammadi, M.; Vitt, M.; Dickie, J.M.; Jyoti, S.S.; Tilbury, M.A.; Johnson, P.A.; Wawrousek, K.E.; Wall, J.G. Rapid, Point-of-Care ScFv-SERS Assay for Femtogram Level Detection of SARS-CoV-2. ACS Sens. 2022, 7, 866–873. [Google Scholar] [CrossRef] [PubMed]
  265. Li, Z.; Luo, Y.; Song, Y.; Zhu, Q.; Xu, T.; Zhang, X. One-Click Investigation of Shape Influence of Silver Nanostructures on SERS Performance for Sensitive Detection of COVID-19. Anal. Chim. Acta 2022, 1234, 340523. [Google Scholar] [CrossRef]
  266. Kim, W.; Kim, S.; Han, J.; Kim, T.G.; Bang, A.; Choi, H.W.; Min, G.E.; Shin, J.H.; Moon, S.W.; Choi, S. An Excitation Wavelength-Optimized, Stable SERS Biosensing Nanoplatform for Analyzing Adenoviral and AstraZeneca COVID-19 Vaccination Efficacy Status Using Tear Samples of Vaccinated Individuals. Biosens. Bioelectron. 2022, 204, 114079. [Google Scholar] [CrossRef] [PubMed]
  267. Karunakaran, V.; Joseph, M.M.; Yadev, I.; Sharma, H.; Shamna, K.; Saurav, S.; Sreejith, R.P.; Anand, V.; Beegum, R.; Regi David, S.; et al. A Non-Invasive Ultrasensitive Diagnostic Approach for COVID-19 Infection Using Salivary Label-Free SERS Fingerprinting and Artificial Intelligence. J. Photochem. Photobiol. B Biol. 2022, 234, 112545. [Google Scholar] [CrossRef]
  268. Shanmukh, S.; Jones, L.; Driskell, J.; Zhao, Y.; Dluhy, R.; Tripp, R.A. Rapid and Sensitive Detection of Respiratory Virus Molecular Signatures Using a Silver Nanorod Array SERS Substrate. Nano Lett. 2006, 6, 2630–2636. [Google Scholar] [CrossRef] [PubMed]
  269. Dluhy, R.A.; Shanmukh, S.; Jones, L.; Zhao, Y.P.; Driskell, J.D.; Tripp, R.A. Identification and Classification of Respiratory Syncytial Virus (RSV) Strains by Surface-Enhanced Raman Spectroscopy and Multivariate Statistical Techniques. Anal. Bioanal. Chem. 2008, 390, 1551–1555. [Google Scholar] [CrossRef]
  270. Huang, J.; Wen, J.; Zhou, M.; Ni, S.; Le, W.; Chen, G.; Wei, L.; Zeng, Y.; Qi, D.; Pan, M.; et al. On-Site Detection of SARS-CoV-2 Antigen by Deep Learning-Based Surface-Enhanced Raman Spectroscopy and Its Biochemical Foundations. Anal. Chem. 2021, 93, 9174–9182. [Google Scholar] [CrossRef] [PubMed]
  271. Yeh, Y.J.; Le, T.N.; Hsiao, W.W.W.; Tung, K.L.; Ostrikov, K.; Chiang, W.H. Plasmonic Nanostructure-Enhanced Raman Scattering for Detection of SARS-CoV-2 Nucleocapsid Protein and Spike Protein Variants. Anal. Chim. Acta 2023, 1239, 340651. [Google Scholar] [CrossRef] [PubMed]
  272. Liu, Z.; Wang, C.; Zheng, S.; Yang, X.; Han, H.; Dai, Y.; Xiao, R. Simultaneously Ultrasensitive and Quantitative Detection of Influenza A Virus, SARS-CoV-2, and Respiratory Syncytial Virus via Multichannel Magnetic SERS-Based Lateral Flow Immunoassay. Nanomed. Nanotechnol. Biol. Med. 2023, 47, 102624. [Google Scholar] [CrossRef] [PubMed]
  273. Chisanga, M.; Williams, H.; Boudreau, D.; Pelletier, J.N.; Trottier, S. Label-Free SERS for Rapid Differentiation of SARS-CoV-2-Induced Serum Metabolic Profiles in Non-Hospitalized Adults. Anal. Chem. 2023, 95, 3638–3646. [Google Scholar] [CrossRef] [PubMed]
  274. Bacteria. Available online: https://microbiologysociety.org/why-microbiology-matters/what-is-microbiology/bacteria.html#:~:text=Bacteria%20are%20classified%20into%20five,)%20or%20corkscrew%20(spirochaetes) (accessed on 30 January 2023).
  275. Bacteria as Pathogens. Available online: https://sphweb.bumc.bu.edu/otlt/mph-modules/ph/ph709_infectiousagents/PH709_InfectiousAgents4.html#:~:text=While%20only%20about%205%25%20of,of%20human%20disease%20and%20death (accessed on 30 January 2023).
  276. Nanda, M.; Kumar, V.; Sharma, D.K. Multimetal Tolerance Mechanisms in Bacteria: The Resistance Strategies Acquired by Bacteria That Can Be Exploited to ‘Clean-up’ Heavy Metal Contaminants from Water. Aquat. Toxicol. 2019, 212, 1–10. [Google Scholar] [CrossRef] [PubMed]
  277. Glick, B.R. Phytoremediation: Synergistic Use of Plants and Bacteria to Clean up the Environment. Biotechnol. Adv. 2003, 21, 383–393. [Google Scholar] [CrossRef]
  278. Kulshreshtha, A.; Agrawal, R.; Barar, M.; Saxena, S. A Review on Bioremediation of Heavy Metals in Contaminated Water. IOSR J. Environ. Sci. Toxicol. Food Technol. 2014, 8, 44–50. [Google Scholar] [CrossRef]
  279. Zhou, X.; Hu, Z.; Yang, D.; Xie, S.; Jiang, Z.; Niessner, R.; Haisch, C.; Zhou, H.; Sun, P. Bacteria Detection: From Powerful SERS to Its Advanced Compatible Techniques. Adv. Sci. 2020, 7, 2001739. [Google Scholar] [CrossRef] [PubMed]
  280. Wang, R.; Kim, K.; Choi, N.; Wang, X.; Lee, J.; Jeon, J.H.; Rhie, G.E.; Choo, J. Highly Sensitive Detection of High-Risk Bacterial Pathogens Using SERS-Based Lateral Flow Assay Strips. Sens. Actuators B Chem. 2018, 270, 72–79. [Google Scholar] [CrossRef]
  281. Zhu, T.; Hu, Y.; Yang, K.; Dong, N.; Yu, M.; Jiang, N. A Novel SERS Nanoprobe Based on the Use of Core-Shell Nanoparticles with Embedded Reporter Molecule to Detect E. coli O157:H7 with High Sensitivity. Microchim. Acta 2018, 185, 30. [Google Scholar] [CrossRef]
  282. Chisanga, M.; Muhamadali, H.; Ellis, D.I.; Goodacre, R. Surface-Enhanced Raman Scattering (SERS) in Microbiology: Illumination and Enhancement of the Microbial World. Appl. Spectrosc. 2018, 72, 987–1000. [Google Scholar] [CrossRef]
  283. Kim, J.A.; Wales, D.J.; Thompson, A.J.; Yang, G.Z. Fiber-optic SERS probes fabricated using two-photon polymerization for rapid detection of bacteria. Adv. Opt. Mater. 2020, 8, 1901934. [Google Scholar] [CrossRef] [Green Version]
  284. Zhou, H.; Yang, D.; Ivleva, N.P.; Mircescu, N.E.; Niessner, R.; Haisch, C. SERS Detection of Bacteria in Water by in Situ Coating with Ag Nanoparticles. Anal. Chem. 2014, 86, 1525–1533. [Google Scholar] [CrossRef]
  285. Yang, Y.; Zeng, C.; Huang, J.; Wang, M.; Qi, W.; Wang, H.; He, Z. Specific and Quantitative Detection of Bacteria Based on Surface Cell Imprinted SERS Mapping Platform. Biosens. Bioelectron. 2022, 215, 114524. [Google Scholar] [CrossRef]
  286. Pearson, B.; Wang, P.; Mills, A.; Pang, S.; McLandsborough, L.; He, L. Innovative Sandwich Assay with Dual Optical and SERS Sensing Mechanisms for Bacterial Detection. Anal. Methods 2017, 9, 4732–4739. [Google Scholar] [CrossRef]
  287. Hudson, S.D.; Chumanov, G. Bioanalytical Applications of SERS (Surface-Enhanced Raman Spectroscopy). Anal. Bioanal. Chem. 2009, 394, 679–686. [Google Scholar] [CrossRef] [PubMed]
  288. Mosier-Boss, P.A. Review on SERS of Bacteria. Biosensors 2017, 7, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  289. Jarvis, R.M.; Goodacre, R. Characterisation and Identification of Bacteria Using SERS. Chem. Soc. Rev. 2008, 37, 931–936. [Google Scholar] [CrossRef]
  290. Xia, J.; Li, W.; Sun, M.; Wang, H. Application of SERS in the Detection of Fungi, Bacteria and Viruses. Nanomaterials 2022, 12, 3572. [Google Scholar] [CrossRef]
  291. Wang, C.; Meloni, M.M.; Wu, X.; Zhuo, M.; He, T.; Wang, J.; Wang, C.; Dong, P. Magnetic Plasmonic Particles for SERS-Based Bacteria Sensing: A Review. AIP Adv. 2019, 9, 010701. [Google Scholar] [CrossRef] [Green Version]
  292. Efrima, S.; Zeiri, L. Understanding SERS of Bacteria. J. Raman Spectrosc. 2009, 40, 277–288. [Google Scholar] [CrossRef]
  293. Liu, H.B.; Du, X.J.; Zang, Y.X.; Li, P.; Wang, S. SERS-Based Lateral Flow Strip Biosensor for Simultaneous Detection of Listeria Monocytogenes and Salmonella Enterica Serotype Enteritidis. J. Agric. Food Chem. 2017, 65, 10290–10299. [Google Scholar] [CrossRef]
  294. Mungroo, N.A.; Oliveira, G.; Neethirajan, S. SERS Based Point-of-Care Detection of Food-Borne Pathogens. Microchim. Acta 2016, 183, 697–707. [Google Scholar] [CrossRef]
  295. Lin, H.Y.; Huang, C.H.; Hsieh, W.H.; Liu, L.H.; Lin, Y.C.; Chu, C.C.; Wang, S.T.; Kuo, I.T.; Chau, L.K.; Yang, C.Y. On-Line SERS Detection of Single Bacterium Using Novel SERS Nanoprobes and a Microfl Uidic Dielectrophoresis Device. Small 2014, 10, 4700–4710. [Google Scholar] [CrossRef]
  296. Wang, Y.; Lee, K.; Irudayaraj, J. Silver Nanosphere SERS Probes for Sensitive Identification of Pathogens. J. Phys. Chem. C 2010, 114, 16122–16128. [Google Scholar] [CrossRef]
  297. Witkowska, E.; Korsak, D.; Kowalska, A.; Janeczek, A.; Kamińska, A. Strain-Level Typing and Identification of Bacteria—A Novel Approach for SERS Active Plasmonic Nanostructures. Anal. Bioanal. Chem. 2018, 410, 5019–5031. [Google Scholar] [CrossRef] [Green Version]
  298. Pang, Y.; Wan, N.; Shi, L.; Wang, C.; Sun, Z.; Xiao, R.; Wang, S. Dual-Recognition Surface-Enhanced Raman Scattering (SERS)Biosensor for Pathogenic Bacteria Detection by Using Vancomycin-SERS Tags and Aptamer-Fe3O4@Au. Anal. Chim. Acta 2019, 1077, 288–296. [Google Scholar] [CrossRef] [PubMed]
  299. Gao, X.; Yin, Y.; Wu, H.; Hao, Z.; Li, J.; Wang, S.; Liu, Y. Integrated SERS Platform for Reliable Detection and Photothermal Elimination of Bacteria in Whole Blood Samples. Anal. Chem. 2021, 93, 1569–1577. [Google Scholar] [CrossRef]
  300. Zhou, Z.; Xiao, R.; Cheng, S.; Wang, S.; Shi, L.; Wang, C.; Qi, K.; Wang, S. A Universal SERS-Label Immunoassay for Pathogen Bacteria Detection Based on Fe3O4@Au-Aptamer Separation and Antibody-Protein A Orientation Recognition. Anal. Chim. Acta 2021, 1160, 338421. [Google Scholar] [CrossRef]
  301. Wang, J.; Wu, X.; Wang, C.; Rong, Z.; Ding, H.; Li, H.; Li, S.; Shao, N.; Dong, P.; Xiao, R.; et al. Facile Synthesis of Au-Coated Magnetic Nanoparticles and Their Application in Bacteria Detection via a SERS Method. ACS Appl. Mater. Interfaces 2016, 8, 19958–19967. [Google Scholar] [CrossRef]
  302. Wang, C.; Wang, J.; Li, M.; Qu, X.; Zhang, K.; Rong, Z.; Xiao, R.; Wang, S. A Rapid SERS Method for Label-Free Bacteria Detection Using Polyethylenimine-Modified Au-Coated Magnetic Microspheres and Au@Ag Nanoparticles. Analyst 2016, 141, 6226–6238. [Google Scholar] [CrossRef]
  303. Huang, L.; Sun, D.W.; Wu, Z.; Pu, H.; Wei, Q. Reproducible, Shelf-Stable, and Bioaffinity SERS Nanotags Inspired by Multivariate Polyphenolic Chemistry for Bacterial Identification. Anal. Chim. Acta 2021, 1167, 338570. [Google Scholar] [CrossRef] [PubMed]
  304. Bi, L.; Wang, X.; Cao, X.; Liu, L.; Bai, C.; Zheng, Q.; Choo, J.; Chen, L. SERS-Active Au@Ag Core-Shell Nanorod (Au@AgNR) Tags for Ultrasensitive Bacteria Detection and Antibiotic-Susceptibility Testing. Talanta 2020, 220, 121397. [Google Scholar] [CrossRef] [PubMed]
  305. Hunter, R.; Sohi, A.N.; Khatoon, Z.; Berthiaume, V.R.; Alarcon, E.I.; Godin, M.; Anis, H. Optofluidic Label-Free SERS Platform for Rapid Bacteria Detection in Serum. Sens. Actuators B Chem. 2019, 300, 126907. [Google Scholar] [CrossRef]
  306. Sivanesan, A.; Witkowska, E.; Adamkiewicz, W.; Dziewit, Ł.; Kamińska, A.; Waluk, J. Nanostructured Silver-Gold Bimetallic SERS Substrates for Selective Identification of Bacteria in Human Blood. Analyst 2014, 139, 1037–1043. [Google Scholar] [CrossRef]
  307. Witkowska, E.; Szymborski, T.; Kamińska, A.; Waluk, J. Polymer Mat Prepared via ForcespinningTM as a SERS Platform for Immobilization and Detection of Bacteria from Blood Plasma. Mater. Sci. Eng. C 2017, 71, 345–350. [Google Scholar] [CrossRef]
  308. Zhang, L.; Xu, J.; Mi, L.; Gong, H.; Jiang, S.; Yu, Q. Multifunctional Magnetic-Plasmonic Nanoparticles for Fast Concentration and Sensitive Detection of Bacteria Using SERS. Biosens. Bioelectron. 2012, 31, 130–136. [Google Scholar] [CrossRef]
  309. Krafft, B.; Tycova, A.; Urban, R.D.; Dusny, C.; Belder, D. Microfluidic device for concentration and SERS-based detection of bacteria in drinking water. Electrophoresis 2021, 42, 86–94. [Google Scholar] [CrossRef]
  310. Yang, E.; Li, D.; Yin, P.; Xie, Q.; Li, Y.; Lin, Q.; Duan, Y. A Novel Surface-Enhanced Raman Scattering (SERS) Strategy for Ultrasensitive Detection of Bacteria Based on Three-Dimensional (3D) DNA Walker. Biosens. Bioelectron. 2021, 172, 112758. [Google Scholar] [CrossRef]
  311. Wang, C.; Wang, C.; Li, J.; Tu, Z.; Gu, B.; Wang, S. Ultrasensitive and Multiplex Detection of Four Pathogenic Bacteria on a Bi-Channel Lateral Flow Immunoassay Strip with Three-Dimensional Membrane-like SERS Nanostickers. Biosens. Bioelectron. 2022, 214, 114525. [Google Scholar] [CrossRef]
  312. Kearns, H.; Goodacre, R.; Jamieson, L.E.; Graham, D.; Faulds, K. SERS Detection of Multiple Antimicrobial-Resistant Pathogens Using Nanosensors. Anal. Chem. 2017, 89, 12666–12673. [Google Scholar] [CrossRef] [Green Version]
  313. Gracie, K.; Correa, E.; Mabbott, S.; Dougan, J.A.; Graham, D.; Goodacre, R.; Faulds, K. Simultaneous Detection and Quantification of Three Bacterial Meningitis Pathogens by SERS. Chem. Sci. 2014, 5, 1030–1040. [Google Scholar] [CrossRef] [Green Version]
  314. Mosier-Boss, P.A.; Sorensen, K.C.; George, R.D.; Obraztsova, A. SERS Substrates Fabricated Using Ceramic Filters for the Detection of Bacteria. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2016, 153, 591–598. [Google Scholar] [CrossRef] [PubMed]
  315. Walter, A.; März, A.; Schumacher, W.; Rösch, P.; Popp, J. Towards a Fast, High Specific and Reliable Discrimination of Bacteria on Strain Level by Means of SERS in a Microfluidic Device. Lab Chip 2011, 11, 1013–1021. [Google Scholar] [CrossRef]
  316. Wu, X.; Xu, C.; Tripp, R.A.; Huang, Y.W.; Zhao, Y. Detection and Differentiation of Foodborne Pathogenic Bacteria in Mung Bean Sprouts Using Field Deployable Label-Free SERS Devices. Analyst 2013, 138, 3005–3012. [Google Scholar] [CrossRef] [PubMed]
  317. Ankamwar, B.; Sur, U.K.; Das, P. SERS Study of Bacteria Using Biosynthesized Silver Nanoparticles as the SERS Substrate. Anal. Methods 2016, 8, 2335–2340. [Google Scholar] [CrossRef]
  318. Ciloglu, F.U.; Caliskan, A.; Saridag, A.M.; Kilic, I.H.; Tokmakci, M.; Kahraman, M.; Aydin, O. Drug-Resistant Staphylococcus Aureus Bacteria Detection by Combining Surface-Enhanced Raman Spectroscopy (SERS) and Deep Learning Techniques. Sci. Rep. 2021, 11, 18444. [Google Scholar] [CrossRef]
  319. Nerve Agents, BBC Report. Available online: https://www.bbc.com/news/uk-43431537 (accessed on 30 January 2023).
  320. Lister, A.P.; Sellors, W.J.; Howle, C.R.; Mahajan, S. Raman Scattering Techniques for Defense and Security Applications. Anal. Chem. 2021, 93, 417–429. [Google Scholar] [CrossRef]
  321. Sadayoshi, O.H.B.U.; Yamashina, A.; Takasu, N.; Yamaguchi, T.; Murai, T.; Nakano, K.; Hinohara, S. Sarin poisoning on Tokyo subway. South. Med. J. 1997, 90, 587–593. [Google Scholar]
  322. Inscore, F.E.; Gift, A.D.; Maksymiuk, P.; Farquharson, S. Characterization of Chemical Warfare G-Agent Hydrolysis Products by Surface-Enhanced Raman Spectroscopy. Chem. Biol. Point Sens. Homel. Def. II 2004, 5585, 46. [Google Scholar] [CrossRef]
  323. Mukherjee, S.; Gupta, R.D. Organophosphorus Nerve Agents: Types, Toxicity, and Treatments. J. Toxicol. 2020, 2020, 3007984. [Google Scholar] [CrossRef]
  324. Saylan, Y.; Akgönüllü, S.; Denizli, A. Plasmonic Sensors for Monitoring Biological and Chemical Threat Agents. Biosensors 2020, 10, 142. [Google Scholar] [CrossRef] [PubMed]
  325. Primera-Pedrozo, O.M.; Jerez-Rozo, J.I.; De La Cruz-Montoya, E.; Luna-Pineda, T.; Pacheco-Londoño, L.C.; Hernández-Rivera, S.P. Nanotechnology-Based Detection of Explosives and Biological Agents Simulants. IEEE Sens. J. 2008, 8, 963–973. [Google Scholar] [CrossRef]
  326. Yan, F.; Vo-Dinh, T. Surface-Enhanced Raman Scattering Detection of Chemical and Biological Agents Using a Portable Raman Integrated Tunable Sensor. Sens. Actuators B Chem. 2007, 121, 61–66. [Google Scholar] [CrossRef]
  327. Pearman, W.F.; Fountain, A.W. Classification of Chemical and Biological Warfare Agent Simulants by Surface-Enhanced Raman Spectroscopy and Multivariate Statistical Techniques. Appl. Spectrosc. 2006, 60, 356–365. [Google Scholar] [CrossRef] [PubMed]
  328. Hakonen, A.; Rindzevicius, T.; Schmidt, M.S.; Andersson, P.O.; Juhlin, L.; Svedendahl, M.; Boisen, A.; Käll, M. Detection of Nerve Gases Using Surface-Enhanced Raman Scattering Substrates with High Droplet Adhesion. Nanoscale 2016, 8, 1305–1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Juhlin, L.; Mikaelsson, T.; Hakonen, A.; Stenbæk, M. Selective Surface-Enhanced Raman Scattering Detection of Tabun, VX and Cyclosarin Nerve Agents Using 4-Pyridine Amide Oxime Functionalized Gold Nanopillars Talanta Selective Surface-Enhanced Raman Scattering Detection of Tabun, VX and Cyclosarin Nerve A. Talanta 2020, 211, 120721. [Google Scholar] [CrossRef]
  330. Farquharson, S.; Gift, A.; Maksymiuk, P.; Inscore, F. Surface-Enhanced Raman Spectra of VX and Its Hydrolysis Products. Appl. Spectrosc. 2005, 59, 654–660. [Google Scholar] [CrossRef]
  331. Heleg-Shabtai, V.; Sharabi, H.; Zaltsman, A.; Ron, I.; Pevzner, A. Surface-Enhanced Raman Spectroscopy (SERS) for Detection of VX and HD in the Gas Phase Using a Hand-Held Raman Spectrometer. Analyst 2020, 145, 6334–6341. [Google Scholar] [CrossRef]
  332. Zhao, Q.; Liu, G.; Zhang, H.; Zhou, F.; Li, Y.; Cai, W. SERS-Based Ultrasensitive Detection of Organophosphorus Nerve Agents via Substrate’s Surface Modification. J. Hazard. Mater. 2017, 324, 194–202. [Google Scholar] [CrossRef]
  333. Spencer, K.M.; Sylvia, J.M.; Clauson, S.L.; Janni, J.A. Surface-Enhanced Raman as a Water Monitor for Warfare Agents. Vib. Spectrosc. Sens. Syst. 2002, 4577, 158. [Google Scholar] [CrossRef]
  334. Bertone, J.F.; Cordeiro, K.L.; Sylvia, J.M.; Spencer, K.M. A Nanoengineered Sensor to Detect Vibrational Modes of Warfare Agents/Explosives Using Surface-Enhanced Raman Scattering. Sens. Command. Control. Commun. Intell. Technol. Homel. Secur. Homel. Def. III 2004, 5403, 387. [Google Scholar] [CrossRef]
  335. Kim, Y.T.; Kim, D.; Park, S.; Zhexembekova, A.; Byeon, M.; Hong, T.E.; Lee, J.; Lee, C.Y. Aqueous Microlenses for Localized Collection and Enhanced Raman Spectroscopy of Gaseous Molecules. Adv. Opt. Mater. 2021, 9, 2101209. [Google Scholar] [CrossRef]
  336. Wu, J.; Zhu, Y.; Gao, J.; Chen, J.; Feng, J.; Guo, L.; Xie, J. A Simple and Sensitive Surface-Enhanced Raman Spectroscopic Discriminative Detection of Organophosphorous Nerve Agents. Anal. Bioanal. Chem. 2017, 409, 5091–5099. [Google Scholar] [CrossRef] [PubMed]
  337. Lafuente, M.; Berenschot, E.J.W.; Tiggelaar, R.M.; Mallada, R.; Tas, N.R.; Pina, M.P. 3D Fractals as SERS Active Platforms: Preparation and Evaluation for Gas Phase Detection of G-Nerve Agents. Micromachines 2018, 9, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  338. Goel, A.K. Anthrax: A Disease of Biowarfare and Public Health Importance. World J. Clin. Cases 2015, 3, 20. [Google Scholar] [CrossRef] [PubMed]
  339. Farrell, M.E.; Pellegrino, P.M. Army Relevant Biological Hazards Detection with Commercial SERS Substrates. Biosen. Nanomed. V 2012, 8460, 84600J. [Google Scholar] [CrossRef]
  340. Sajanlal, P.R.; Pradeep, T. Functional Hybrid Nickel Nanostructures as Recyclable SERS Substrates: Detection of Explosives and Biowarfare Agents. Nanoscale 2012, 4, 3427–3437. [Google Scholar] [CrossRef]
  341. Wang, T.; Dong, P.; Zhu, C.; Sha, P.; Gao, W.; Wu, Y.; Wu, X. Trace Detection of Anthrax Protective Antigens via a Competitive Method Based on Surface-Enhanced Raman Scattering. Sens. Actuators B Chem. 2021, 346, 130467. [Google Scholar] [CrossRef]
  342. Gao, R.; Ko, J.; Cha, K.; Ho Jeon, J.; Rhie, G.E.; Choi, J.; de Mello, A.J.; Choo, J. Fast and Sensitive Detection of an Anthrax Biomarker Using SERS-Based Solenoid Microfluidic Sensor. Biosens. Bioelectron. 2015, 72, 230–236. [Google Scholar] [CrossRef]
  343. Naqvi, T.K.; Bajpai, A.; Bharati, M.S.S.; Kulkarni, M.M.; Siddiqui, A.M.; Soma, V.R.; Dwivedi, P.K. Ultra-Sensitive Reusable SERS Sensor for Multiple Hazardous Materials Detection on Single Platform. J. Hazard. Mater. 2021, 407, 124353. [Google Scholar] [CrossRef]
  344. Sengupta, A.; Shende, C.; Farquharson, S.; Inscore, F. Detection of Bacillus Anthracis Spores Using Peptide Functionalized SERS-Active Substrates. Int. J. Spectrosc. 2012, 2012, 176851. [Google Scholar] [CrossRef] [Green Version]
  345. Yilmaz, M.; Senlik, E.; Biskin, E.; Yavuz, M.S.; Tamer, U.; Demirel, G. Combining 3-D Plasmonic Gold Nanorod Arrays with Colloidal Nanoparticles as a Versatile Concept for Reliable, Sensitive, and Selective Molecular Detection by SERS. Phys. Chem. Chem. Phys. 2014, 16, 5563–5570. [Google Scholar] [CrossRef]
  346. Li, B.; Wang, T.; Bai, W.; Su, Q.; Wu, X.; Dong, P. Label-Free and Rapid Detection of Anthrax Protective Antigen by Surface-Enhanced Raman Scattering on Au Nanorods. IEEE Sens. J. 2021, 21, 18425–18434. [Google Scholar] [CrossRef]
  347. Cheung, M.; Lee, W.W.Y.; Cowcher, D.P.; Goodacre, R.; Bell, S.E.J. SERS of Meso-Droplets Supported on Superhydrophobic Wires Allows Exquisitely Sensitive Detection of Dipicolinic Acid, an Anthrax Biomarker, Considerably below the Infective Dose. Chem. Commun. 2016, 52, 9925–9928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  348. Félix-Rivera, H.; González, R.; Rodríguez, G.D.M.; Primera-Pedrozo, O.M.; Ríos-Velázquez, C.; Hernández-Rivera, S.P. Improving SERS Detection of Bacillus Thuringiensis Using Silver Nanoparticles Reduced with Hydroxylamine and with Citrate Capped Borohydride. Int. J. Spectrosc. 2011, 2011, 989504. [Google Scholar] [CrossRef] [Green Version]
  349. FountainIII, A.W.; Pearman, W.F. Multivariate Statistical Classification of Surface Enhanced Raman Spectra of Chemical and Biological Warfare Agent Simulants. Chem. Biol. Sens. Ind. Environ. Secur. 2005, 5994, 180–193. [Google Scholar] [CrossRef]
  350. Arano-Martinez, J.A.; Martínez-González, C.L.; Salazar, M.I.; Torres-Torres, C. A Framework for Biosensors Assisted by Multiphoton Effects and Machine Learning. Biosensors 2022, 12, 710. [Google Scholar] [CrossRef]
  351. Luo, R.; Popp, J.; Bocklitz, T. Deep Learning for Raman Spectroscopy: A Review. Analytica 2022, 3, 287–301. [Google Scholar] [CrossRef]
  352. Ralbovsky, N.M.; Lednev, I.K. Towards Development of a Novel Universal Medical Diagnostic Method: Raman Spectroscopy and Machine Learning. Chem. Soc. Rev. 2020, 49, 7428–7453. [Google Scholar] [CrossRef]
  353. Ding, Y.; Sun, Y.; Liu, C.; Jiang, Q.Y.; Chen, F.; Cao, Y. SERS-Based Biosensors Combined with Machine Learning for Medical Application. ChemistryOpen 2023, 12, e202200192. [Google Scholar] [CrossRef]
  354. Cui, F.; Yue, Y.; Zhang, Y.; Zhang, Z.; Zhou, H.S. Advancing Biosensors with Machine Learning. ACS Sens. 2020, 5, 3346–3364. [Google Scholar] [CrossRef]
  355. Lv, R.; Wang, Z.; Ma, Y.; Li, W.; Tian, J. Machine Learning Enhanced Optical Spectroscopy for Disease Detection. J. Phys. Chem. Lett. 2022, 13, 9238–9249. [Google Scholar] [CrossRef]
  356. Schackart, K.E.; Yoon, J.Y. Machine Learning Enhances the Performance of Bioreceptor-Free Biosensors. Sensors 2021, 21, 5519. [Google Scholar] [CrossRef]
  357. Lussier, F.; Missirlis, D.; Spatz, J.P.; Masson, J.F. Machine-Learning-Driven Surface-Enhanced Raman Scattering Optophysiology Reveals Multiplexed Metabolite Gradients Near Cells. ACS Nano 2019, 13, 1403–1411. [Google Scholar] [CrossRef] [PubMed]
  358. Nguyen, P.H.L.; Hong, B.; Rubin, S.; Fainman, Y. Machine Learning for Composition Analysis of SsDNA Using Chemical Enhancement in SERS. Biomed. Opt. Express 2020, 11, 5092. [Google Scholar] [CrossRef] [PubMed]
  359. Narla, L.M.; Rao, S.V. Identification of Metals and Alloys Using Color CCD Images of Laser-Induced Breakdown Emissions Coupled with Machine Learning. Appl. Phys. B Lasers Opt. 2020, 126, 113. [Google Scholar] [CrossRef]
  360. Beeram, R.; Banerjee, D.; Narlagiri, L.M.; Soma, V.R. Machine Learning for Rapid Quantification of Trace Analyte Molecules Using SERS and Flexible Plasmonic Paper Substrates. Anal. Methods 2022, 14, 1788–1796. [Google Scholar] [CrossRef]
  361. Murthy, N.L.; Abdul Salam, S.; Rao, S.V. Stand-off Femtosecond Laser Induced Breakdown Spectroscopy of Metals, Soil, Plastics and Classification Studies. In Proceedings of the 2019 Workshop on Recent Advances in Photonics (WRAP), Guwahati, India, 13–14 December 2019; pp. 1–3. [Google Scholar] [CrossRef]
  362. Boehmke, B.; Greenwell, B. Hands-On Machine Learning with R; Chapman and Hall/CRC: Boca Raton, FL, USA, 2019; ISBN 9781492032649. [Google Scholar]
  363. Li, D.; Zhang, Q.; Deng, B.; Chen, Y.; Ye, L. Rapid, Sensitive Detection of Ganciclovir, Penciclovir and Valacyclovir-Hydrochloride by Artificial Neural Network and Partial Least Squares Combined with Surface Enhanced Raman Spectroscopy. Appl. Surf. Sci. 2021, 539, 148224. [Google Scholar] [CrossRef]
  364. Boulesteix, A.L.; Strimmer, K. Partial Least Squares: A Versatile Tool for the Analysis of High-Dimensional Genomic Data. Brief. Bioinform. 2007, 8, 32–44. [Google Scholar] [CrossRef] [Green Version]
  365. Deng, W.; Huang, Z.; Zhang, J.; Xu, J. A Data Mining Based System for Transaction Fraud Detection. In Proceedings of the 2021 IEEE International Conference on Consumer Electronics and Computer Engineering (ICCECE), Guangzhou, China, 15–17 January 2021; pp. 542–545. [Google Scholar] [CrossRef]
  366. Fan, X.; Ming, W.; Zeng, H.; Zhang, Z.; Lu, H. Deep Learning-Based Component Identification for the Raman Spectra of Mixtures. Analyst 2019, 144, 1789–1798. [Google Scholar] [CrossRef]
  367. Zhou, H.; Xu, L.; Ren, Z.; Zhu, J.; Lee, C. Machine Learning-Augmented Surface-Enhanced Spectroscopy toward next-Generation Molecular Diagnostics. Nanoscale Adv. 2023, 5, 538–570. [Google Scholar] [CrossRef]
  368. Malinick, A.S.; Stuart, D.D.; Lambert, A.S.; Cheng, Q. Surface Plasmon Resonance Imaging (SPRi) in Combination with Machine Learning for Microarray Analysis of Multiple Sclerosis Biomarkers in Whole Serum. Biosens. Bioelectron. X 2022, 10, 100127. [Google Scholar] [CrossRef]
  369. Pradhan, P.; Guo, S.; Ryabchykov, O.; Popp, J.; Bocklitz, T.W. Deep Learning a Boon for Biophotonics? J. Biophotonics 2020, 13, e201960186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  370. Moon, G.; Lee, J.; Lee, H.; Yoo, H.; Ko, K.; Im, S.; Kim, D. Machine Learning and Its Applications for Plasmonics in Biology. Cell Rep. Phys. Sci. 2022, 3, 101042. [Google Scholar] [CrossRef]
  371. Sun, Y.; Shi, L.; Mi, L.; Guo, R.; Li, T. Recent Progress of SERS Optical Nanosensors for MiRNA Analysis. J. Mater. Chem. B 2020, 8, 5178–5183. [Google Scholar] [CrossRef] [PubMed]
  372. Raji, H.; Tayyab, M.; Sui, J.; Mahmoodi, S.R.; Javanmard, M. Biosensors and Machine Learning for Enhanced Detection, Stratification, and Classification of Cells: A Review. Biomed. Microdevices 2022, 24, 26. [Google Scholar] [CrossRef] [PubMed]
  373. Banerjee, A.; Maity, S.; Mastrangelo, C.H. Nanostructures for biosensing, with a brief overview on Cancer Detection, IoT, and the Role of Machine Learning In Smart Biosensors. Sensors 2021, 21, 1253. [Google Scholar] [CrossRef]
  374. Beeram, R.; Vendamani, V.S.; Soma, V.R. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy Deep Learning Approach to Overcome Signal Fluctuations in SERS for Efficient On-Site Trace Explosives Detection. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2023, 289, 122218. [Google Scholar] [CrossRef] [PubMed]
  375. Moon, G.; Son, T.; Lee, H.; Kim, D. Deep Learning Approach for Enhanced Detection of Surface Plasmon Scattering. Anal. Chem. 2019, 91, 9538–9545. [Google Scholar] [CrossRef]
  376. Gupta, A.K.; Hsu, C.H.; Lai, C.S. Enhancement of the Au/ZnO-NA Plasmonic SERS Signal Using Principal Component Analysis as a Machine Learning Approach. IEEE Photonics J. 2020, 12, 1–11. [Google Scholar] [CrossRef]
  377. Vendamani, V.S.; Beeram, R.; Neethish, M.M.; Rao, S.V.S.N.; Rao, S.V. Wafer-Scale Silver Nanodendrites with Homogeneous Distribution of Gold Nanoparticles for Biomolecules Detection. iScience 2022, 25, 104849. [Google Scholar] [CrossRef]
  378. Erzina, M.; Trelin, A.; Guselnikova, O.; Dvorankova, B.; Strnadova, K.; Perminova, A.; Ulbrich, P.; Mares, D.; Jerabek, V.; Elashnikov, R.; et al. Precise Cancer Detection via the Combination of Functionalized SERS Surfaces and Convolutional Neural Network with Independent Inputs. Sens. Actuators B Chem. 2020, 308, 127660. [Google Scholar] [CrossRef]
  379. Wang, S.; Dong, H.; Shen, W.; Yang, Y.; Li, Z.; Liu, Y.; Wang, C.; Gu, B.; Zhang, L. Rapid SERS Identification of Methicillin-Susceptible and Methicillin-Resistant: Staphylococcus Aureus via Aptamer Recognition and Deep Learning. RSC Adv. 2021, 11, 34425–34431. [Google Scholar] [CrossRef]
  380. Kazemzadeh, M.; Hisey, C.L.; Dauros-Singorenko, P.; Swift, S.; Zargar-Shoshtari, K.; Xu, W.; Broderick, N.G.R. Label-Free Classification of Bacterial Extracellular Vesicles by Combining Nanoplasmonic Sensors with Machine Learning. IEEE Sens. J. 2022, 22, 1128–1137. [Google Scholar] [CrossRef]
  381. Dong, R.; Weng, S.; Yang, L.; Liu, J. Detection and Direct Readout of Drugs in Human Urine Using Dynamic Surface-Enhanced Raman Spectroscopy and Support Vector Machines. Anal. Chem. 2015, 87, 2937–2944. [Google Scholar] [CrossRef] [PubMed]
  382. Lin, C.; Liang, S.; Li, Y.; Peng, Y.; Huang, Z.; Li, Z.; Yang, Y.; Luo, X. Localized Plasmonic Sensor for Direct Identifying Lung and Colon Cancer from the Blood. Biosens. Bioelectron. 2022, 211, 114372. [Google Scholar] [CrossRef] [PubMed]
  383. Peng, S.; Lu, D.; Zhang, B.; You, R.; Chen, J.; Xu, H.; Lu, Y. Machine Learning—Assisted Internal Standard Calibration Label—Free SERS Strategy for Colon Cancer Detection. Anal. Bioanal. Chem. 2023. [Google Scholar] [CrossRef] [PubMed]
  384. Seifert, S.; Merk, V.; Kneipp, J. Identification of Aqueous Pollen Extracts Using Surface Enhanced Raman Scattering (SERS) and Pattern Recognition Methods. J. Biophotonics 2016, 9, 181–189. [Google Scholar] [CrossRef] [PubMed]
  385. Hassoun, M.; Rüger, J.; Kirchberger-Tolstik, T.; Schie, I.W.; Henkel, T.; Weber, K.; Cialla-May, D.; Krafft, C.; Popp, J. A Droplet-Based Microfluidic Chip as a Platform for Leukemia Cell Lysate Identification Using Surface-Enhanced Raman Scattering. Anal. Bioanal. Chem. 2018, 410, 999–1006. [Google Scholar] [CrossRef] [PubMed]
  386. Mühlig, A.; Bocklitz, T.; Labugger, I.; Dees, S.; Henk, S.; Richter, E.; Andres, S.; Merker, M.; Stöckel, S.; Weber, K.; et al. LOC-SERS: A Promising Closed System for the Identification of Mycobacteria. Anal. Chem. 2016, 88, 7998–8004. [Google Scholar] [CrossRef] [PubMed]
  387. Bratchenko, L.A.; Al-Sammarraie, S.Z.; Tupikova, E.N.; Konovalova, D.Y.; Lebedev, P.A.; Zakharov, V.P.; Bratchenko, I.A. Analyzing the Serum of Hemodialysis Patients with End-Stage Chronic Kidney Disease by Means of the Combination of SERS and Machine Learning. Biomed. Opt. Express 2022, 13, 4926. [Google Scholar] [CrossRef] [PubMed]
  388. Gao, K.; Zhu, H.; Charron, B.; Mochizuki, T.; Dong, C.; Ding, H.; Cui, Y.; Lu, M.; Peng, W.; Zhu, S.; et al. Combining Dense Au Nanoparticle Layers and 2D Surface-Enhanced Raman Scattering Arrays for the Identification of Mutant Cyanobacteria Using Machine Learning. J. Phys. Chem. C 2022, 126, 9446–9455. [Google Scholar] [CrossRef]
  389. Ikponmwoba, E.; Ukorigho, O.; Moitra, P.; Pan, D.; Gartia, M.R.; Owoyele, O. A Machine Learning Framework for Detecting COVID-19 Infection Using Surface-Enhanced Raman Scattering. Biosensors 2022, 12, 589. [Google Scholar] [CrossRef] [PubMed]
  390. Pérez-Jiménez, A.I.; Lyu, D.; Lu, Z.; Liu, G.; Ren, B. Surface-Enhanced Raman Spectroscopy: Benefits, Trade-Offs and Future Developments. Chem. Sci. 2020, 11, 4563–4577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  391. Hou, M.; Huang, Y.; Ma, L.; Zhang, Z. Quantitative Analysis of Single and Mix Food Antiseptics Basing on SERS Spectra with PLSR Method. Nanoscale Res. Lett. 2016, 11, 296. [Google Scholar] [CrossRef] [Green Version]
  392. Yan, S.; Liu, C.; Fang, S.; Ma, J.; Qiu, J.; Xu, D.; Li, L.; Yu, J.; Li, D.; Liu, Q. SERS-Based Lateral Flow Assay Combined with Machine Learning for Highly Sensitive Quantitative Analysis of Escherichia coli O157:H7. Anal. Bioanal. Chem. 2020, 412, 7881–7890. [Google Scholar] [CrossRef] [PubMed]
  393. Nguyen, C.Q.; Thrift, W.J.; Bhattacharjee, A.; Ranjbar, S.; Gallagher, T.; Darvishzadeh-Varcheie, M.; Sanderson, R.N.; Capolino, F.; Whiteson, K.; Baldi, P.; et al. Longitudinal Monitoring of Biofilm Formation via Robust Surface-Enhanced Raman Scattering Quantification of Pseudomonas Aeruginosa -Produced Metabolites. ACS Appl. Mater. Interfaces 2018, 10, 12364–12373. [Google Scholar] [CrossRef] [PubMed]
  394. Lee, K.M.; Herrman, T.J. Determination and Prediction of Fumonisin Contamination in Maize by Surface–Enhanced Raman Spectroscopy (SERS). Food Bioprocess Technol. 2016, 9, 588–603. [Google Scholar] [CrossRef]
  395. Kuligowski, J.; El-Zahry, M.R.; Sánchez-Illana, Á.; Quintás, G.; Vento, M.; Lendl, B. Surface Enhanced Raman Spectroscopic Direct Determination of Low Molecular Weight Biothiols in Umbilical Cord Whole Blood. Analyst 2016, 141, 2165–2174. [Google Scholar] [CrossRef] [Green Version]
  396. Tan, A.; Zhao, Y.; Sivashanmugan, K.; Squire, K.; Wang, A.X. Quantitative TLC-SERS Detection of Histamine in Seafood with Support Vector Machine Analysis. Food Control 2019, 103, 111–118. [Google Scholar] [CrossRef] [Green Version]
  397. Rahman, A.; Kang, S.; Wang, W.; Huang, Q.; Kim, I.; Vikesland, P.J. Lectin-Modified Bacterial Cellulose Nanocrystals Decorated with Au Nanoparticles for Selective Detection of Bacteria Using Surface-Enhanced Raman Scattering Coupled with Machine Learning. ACS Appl. Nano Mater. 2022, 5, 259–268. [Google Scholar] [CrossRef]
  398. Banaei, N.; Moshfegh, J.; Mohseni-Kabir, A.; Houghton, J.M.; Sun, Y.; Kim, B. Machine Learning Algorithms Enhance the Specificity of Cancer Biomarker Detection Using SERS-Based Immunoassays in Microfluidic Chips. RSC Adv. 2019, 9, 1859–1868. [Google Scholar] [CrossRef] [Green Version]
  399. Cheng, N.; Chen, D.; Lou, B.; Fu, J.; Wang, H. A Biosensing Method for the Direct Serological Detection of Liver Diseases by Integrating a SERS-Based Sensor and a CNN Classifier. Biosens. Bioelectron. 2021, 186, 113246. [Google Scholar] [CrossRef] [PubMed]
  400. Zhang, X.; Liang, B.; Zhang, J.; Hao, X.; Xu, X.; Chang, H.M.; Leung, P.C.K.; Tan, J. Raman Spectroscopy of Follicular Fluid and Plasma with Machine-Learning Algorithms for Polycystic Ovary Syndrome Screening. Mol. Cell. Endocrinol. 2021, 523, 111139. [Google Scholar] [CrossRef] [PubMed]
  401. Barucci, A.; D’Andrea, C.; Farnesi, E.; Banchelli, M.; Amicucci, C.; De Angelis, M.; Hwang, B.; Matteini, P. Label-Free SERS Detection of Proteins Based on Machine Learning Classification of Chemo-Structural Determinants. Analyst 2021, 146, 674–682. [Google Scholar] [CrossRef]
  402. Kazemzadeh, M.; Hisey, C.L.; Zargar-Shoshtari, K.; Xu, W.; Broderick, N.G.R. Deep Convolutional Neural Networks as a Unified Solution for Raman Spectroscopy-Based Classification in Biomedical Applications. Opt. Commun. 2022, 510, 127977. [Google Scholar] [CrossRef]
  403. Othman, N.H.; Yoot Lee, K.; Mohd Radzol, A.R.; Mansor, W.; Amanina Yusoff, N. PCA-Polynomial-ELM Model Optimal for Detection of NS1 Adulterated Salivary SERS Spectra. J. Phys. Conf. Ser. 2019, 1372, 012064. [Google Scholar] [CrossRef]
  404. Yang, Y.; Xu, B.; Haverstick, J.; Ibtehaz, N.; Muszyński, A.; Chen, X.; Chowdhury, M.E.H.; Zughaier, S.; Zhao, Y. Differentiation and Classification of Bacterial Endotoxins Based on Surface Enhanced Raman Scattering and Advanced Machine Learning. Nanoscale 2022, 14, 8806–8817. [Google Scholar] [CrossRef]
  405. Lin, D.; Hsieh, C.L.; Hsu, K.C.; Liao, P.H.; Qiu, S.; Gong, T.; Yong, K.T.; Feng, S.; Kong, K.V. Geometrically Encoded SERS Nanobarcodes for the Logical Detection of Nasopharyngeal Carcinoma-Related Progression Biomarkers. Nat. Commun. 2021, 12, 3430. [Google Scholar] [CrossRef]
  406. Wang, G.; Lipert, R.J.; Jain, M.; Kaur, S.; Chakraboty, S.; Torres, M.P.; Batra, S.K.; Brand, R.E.; Porter, M.D. Detection of the Potential Pancreatic Cancer Marker MUC4 in Serum Using Surface-Enhanced Raman Scattering. Anal. Chem. 2011, 83, 2554–2561. [Google Scholar] [CrossRef] [Green Version]
  407. Lu, L.; Guan, S.; Guan, Y.; Hong, M. Dual-Modal Fluorescence-SERS Detection of Blood Glucose Engineered by Hierarchical Laser-Induced Micro/Nano Structures for Diabetes Screening. Adv. Mater. Interfaces 2022, 9, 2102532. [Google Scholar] [CrossRef]
  408. Sun, J.; Gong, L.; Wang, W.; Gong, Z.; Wang, D.; Fan, M. Surface-Enhanced Raman Spectroscopy for on-Site Analysis: A Review of Recent Developments. Luminescence 2020, 35, 808–820. [Google Scholar] [CrossRef] [PubMed]
  409. Kho, K.W.; Fu, C.Y.; Dinish, U.S.; Olivo, M. Clinical SERS: Are We There Yet? J. Biophotonics 2011, 4, 667–684. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic of total enhancement in SERS via electromagnetic and chemical enhancement mechanisms.
Figure 1. Schematic of total enhancement in SERS via electromagnetic and chemical enhancement mechanisms.
Biosensors 13 00328 g001
Figure 2. (a) Trends in research on SERS based plasmonic applications in the detection of microorganisms, cancers, respiratory diseases, other diseases such as heart ailments and diabetes and the use of different machine learning techniques for SERS based biosensing. (b) Bar chart with percentage contribution from each area shown on the label for the past 12 years. Source: Scopus search with the keywords mentioned in both the panels as on 5 January 2023.
Figure 2. (a) Trends in research on SERS based plasmonic applications in the detection of microorganisms, cancers, respiratory diseases, other diseases such as heart ailments and diabetes and the use of different machine learning techniques for SERS based biosensing. (b) Bar chart with percentage contribution from each area shown on the label for the past 12 years. Source: Scopus search with the keywords mentioned in both the panels as on 5 January 2023.
Biosensors 13 00328 g002
Figure 3. Schematic of work created by Hyunku et al. in lung cancer identification via a combination of SERS and deep learning. (a) Exosomes that were used as biomarkers for sensing. (b) Sample preparation and data collection. (c) Deep learning model used for the classification of normal and cancer cells exosomes using SERS spectra. Reproduced with permission from [118]. Copyright (2020), American Chemical Society.
Figure 3. Schematic of work created by Hyunku et al. in lung cancer identification via a combination of SERS and deep learning. (a) Exosomes that were used as biomarkers for sensing. (b) Sample preparation and data collection. (c) Deep learning model used for the classification of normal and cancer cells exosomes using SERS spectra. Reproduced with permission from [118]. Copyright (2020), American Chemical Society.
Biosensors 13 00328 g003
Figure 4. Breast cancer detection using SERS. (a) Schematic of workflow for (A) sample collection and (B) deep learning model-based breast cancer detection with exosomes-based SERS sensor. Reproduced with permission from [143]. Copyright (2022), American Chemical Society. (b) SERS and Photoacoustic (PA) imaging of breast cancer cells (A): (i) brightfield microscopic images and SERS mapping area, and (ii) corresponding SERS spectra in different regions labeled in (i). (B) (i) Photo of mouse with tumor, (ii) corresponding representative PA images for different post injection times, and (iii) PA intensity at 750 nm. (C) (i) Optical image of tumor, (ii) SERS image of tumor, and (iii) corresponding spectra for different regions in the image. Reproduced with permission from [144]. Copyright (2021), American Chemical Society.
Figure 4. Breast cancer detection using SERS. (a) Schematic of workflow for (A) sample collection and (B) deep learning model-based breast cancer detection with exosomes-based SERS sensor. Reproduced with permission from [143]. Copyright (2022), American Chemical Society. (b) SERS and Photoacoustic (PA) imaging of breast cancer cells (A): (i) brightfield microscopic images and SERS mapping area, and (ii) corresponding SERS spectra in different regions labeled in (i). (B) (i) Photo of mouse with tumor, (ii) corresponding representative PA images for different post injection times, and (iii) PA intensity at 750 nm. (C) (i) Optical image of tumor, (ii) SERS image of tumor, and (iii) corresponding spectra for different regions in the image. Reproduced with permission from [144]. Copyright (2021), American Chemical Society.
Biosensors 13 00328 g004
Figure 5. (a) Optical images of the workflow used for SERS-based detection of COVID-19 disease using breath analysis with a detection time of 5 min, achieving sensitivity >95% in nearly 500 participants, establishing the rapidness and specificity of SERS. Reproduced with permission from [53]. Copyright (2022), American Chemical Society. (b) Schematic of nasopharyngeal-swab-based Covid detection using SERS with flexible substrates enabling sensitive detection. Reproduced with permission from [238]. Copyright (2022), MDPI.
Figure 5. (a) Optical images of the workflow used for SERS-based detection of COVID-19 disease using breath analysis with a detection time of 5 min, achieving sensitivity >95% in nearly 500 participants, establishing the rapidness and specificity of SERS. Reproduced with permission from [53]. Copyright (2022), American Chemical Society. (b) Schematic of nasopharyngeal-swab-based Covid detection using SERS with flexible substrates enabling sensitive detection. Reproduced with permission from [238]. Copyright (2022), MDPI.
Biosensors 13 00328 g005
Figure 6. Flow chart illustrating the classification of different machine learning algorithms as supervised, unsupervised, and reinforcement models.
Figure 6. Flow chart illustrating the classification of different machine learning algorithms as supervised, unsupervised, and reinforcement models.
Biosensors 13 00328 g006
Figure 7. Schematic of applications of machine learning for biosensing using SERS based plasmonic sensors.
Figure 7. Schematic of applications of machine learning for biosensing using SERS based plasmonic sensors.
Biosensors 13 00328 g007
Figure 8. Applications of machine learning techniques used in the identification of biological samples, using the SERS technique for the (A) identification of lung and colon cancers from exosomes HCT-116 (colon cancer biomarker), A549 (lung cancer biomarker), and non-cancerous samples performed with (a) PCA, (b) PLSDA, and (c) SVM, with 60% of training set and 40% of test set. (d) Predicted labels for the test set using the SVM model. Value 1 is a prediction for normal plasma, Value 2 is a prediction for A549 (lung cancer) exosomes, and Value 3 is a prediction for HCT-116 (colon cancer) exosomes. The highlighted portion shows labels that are wrongly identified. Reproduced with permission from [382]. Copyright (2022), Elsevier. (B) Identification of 14 commercially available pollen species using SERS spectra combined with an artificial neural network, using a winner-takes-all (WTA) method. Reproduced with permission from [384]. Copyright (2015), Wiley. (C) Schematic for SERS-based ML model used in the identification of methicillin-susceptible Staphylococcus aureus (MSSA) and methicillin-resistant Staphylococcus aureus (MRSA) bacteria, using a CNN model. Reproduced with permission from [379]. Copyright (2021), RSC.
Figure 8. Applications of machine learning techniques used in the identification of biological samples, using the SERS technique for the (A) identification of lung and colon cancers from exosomes HCT-116 (colon cancer biomarker), A549 (lung cancer biomarker), and non-cancerous samples performed with (a) PCA, (b) PLSDA, and (c) SVM, with 60% of training set and 40% of test set. (d) Predicted labels for the test set using the SVM model. Value 1 is a prediction for normal plasma, Value 2 is a prediction for A549 (lung cancer) exosomes, and Value 3 is a prediction for HCT-116 (colon cancer) exosomes. The highlighted portion shows labels that are wrongly identified. Reproduced with permission from [382]. Copyright (2022), Elsevier. (B) Identification of 14 commercially available pollen species using SERS spectra combined with an artificial neural network, using a winner-takes-all (WTA) method. Reproduced with permission from [384]. Copyright (2015), Wiley. (C) Schematic for SERS-based ML model used in the identification of methicillin-susceptible Staphylococcus aureus (MSSA) and methicillin-resistant Staphylococcus aureus (MRSA) bacteria, using a CNN model. Reproduced with permission from [379]. Copyright (2021), RSC.
Biosensors 13 00328 g008
Figure 9. (A) Schematic of ML-based quantification of E. coli O157:H7, using (a) SERS nanotags and (b) lateral flow assay along with different regression models, including SVR, BNR, and XGBR. Reproduced with permission from [392]. Copyright (2020), Springer. (B) (a) (i) Optical images of the lateral flow strips and (ii) SERS mapping region of the prominent peak in the SERS intensity profile. (iii) Corresponding SERS spectra of the test lines. (iv) Intensity and concentration fit. (b) Machine-learning-based regression fits for (i) Bayesian ridge regression (BRR), (ii) support vector regression (SVR), and the (iii) elastic net regression (ENR) and (iv) extreme gradient boosting regression (XGBR). (c) PCR image for the E. coli detection. Reproduced with permission from [392]. Copyright (2020), Springer.
Figure 9. (A) Schematic of ML-based quantification of E. coli O157:H7, using (a) SERS nanotags and (b) lateral flow assay along with different regression models, including SVR, BNR, and XGBR. Reproduced with permission from [392]. Copyright (2020), Springer. (B) (a) (i) Optical images of the lateral flow strips and (ii) SERS mapping region of the prominent peak in the SERS intensity profile. (iii) Corresponding SERS spectra of the test lines. (iv) Intensity and concentration fit. (b) Machine-learning-based regression fits for (i) Bayesian ridge regression (BRR), (ii) support vector regression (SVR), and the (iii) elastic net regression (ENR) and (iv) extreme gradient boosting regression (XGBR). (c) PCR image for the E. coli detection. Reproduced with permission from [392]. Copyright (2020), Springer.
Biosensors 13 00328 g009
Figure 10. (A) Schematic of the architecture used for classification of different cancers and normal serum samples using SERS spectra collected from nearly 695 patients. (B) Learning curves for the model implemented with loss and (C) accuracy as metrics. (D) Confusion matrix for the training (E) test data sets communicating good accuracy. Reproduced with permission from [50]. Copyright (2021), Wiley.
Figure 10. (A) Schematic of the architecture used for classification of different cancers and normal serum samples using SERS spectra collected from nearly 695 patients. (B) Learning curves for the model implemented with loss and (C) accuracy as metrics. (D) Confusion matrix for the training (E) test data sets communicating good accuracy. Reproduced with permission from [50]. Copyright (2021), Wiley.
Biosensors 13 00328 g010
Figure 11. Steps for best practices for quantitative and qualitative detection using SERS to overcome the challenges at each stage with a goal of real-world applications. Reproduced with permission from [390]. Copyright (2020), RSC.
Figure 11. Steps for best practices for quantitative and qualitative detection using SERS to overcome the challenges at each stage with a goal of real-world applications. Reproduced with permission from [390]. Copyright (2020), RSC.
Biosensors 13 00328 g011
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Beeram, R.; Vepa, K.R.; Soma, V.R. Recent Trends in SERS-Based Plasmonic Sensors for Disease Diagnostics, Biomolecules Detection, and Machine Learning Techniques. Biosensors 2023, 13, 328. https://doi.org/10.3390/bios13030328

AMA Style

Beeram R, Vepa KR, Soma VR. Recent Trends in SERS-Based Plasmonic Sensors for Disease Diagnostics, Biomolecules Detection, and Machine Learning Techniques. Biosensors. 2023; 13(3):328. https://doi.org/10.3390/bios13030328

Chicago/Turabian Style

Beeram, Reshma, Kameswara Rao Vepa, and Venugopal Rao Soma. 2023. "Recent Trends in SERS-Based Plasmonic Sensors for Disease Diagnostics, Biomolecules Detection, and Machine Learning Techniques" Biosensors 13, no. 3: 328. https://doi.org/10.3390/bios13030328

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop