Skip to content
Publicly Available Published by De Gruyter October 5, 2013

SERS-based detection of biomolecules

  • Dana Cialla , Sibyll Pollok , Carolin Steinbrücker , Karina Weber and Jürgen Popp EMAIL logo
From the journal Nanophotonics

Abstract

In order to detect biomolecules, different approaches using for instance biological, spectroscopic or imaging techniques are established. Due to the broad variety of these methods, this review is focused on surface enhanced Raman spectroscopy (SERS) as an analytical tool in biomolecule detection. Here, the molecular specificity of Raman spectroscopy is combined with metallic nanoparticles as sensor platform, which enhances the signal intensity by several orders of magnitude. Within this article, the characterization of diverse biomolecules by means of SERS is explained and moreover current application fields are presented. The SERS intensity and as a consequence thereof the reliable detection of the biomolecule of interest is effected by distance, orientation and affinity of the molecule towards the metal surface. Furthermore, the great capability of the SERS technique for cutting-edge applications like pathogen detection and cancer diagnosis is highlighted. We wish to motivate by this comprehensive and critical summary researchers from various scientific background to create their own ideas and schemes for a SERS-based detection and analysis of biomolecules.

Reviewed Publication:

Perelman Lev T.


Keywords: biomolecules; Raman; SERS

1 Introduction

The detection of biomolecules is essential in various analytical, medical, biochemical and pharmaceutical application fields. Since the term “biomolecule” is defined as a chemical substance generated by a living organism and includes amongst others nucleic acids, peptides/proteins and their building blocks, a variety of reliable detection schemes are established. In most cases, immunological or molecular biological methods such as enzyme-linked immunosorbent assay (ELISA) or polymerase chain reaction (PCR) are applied to detect peptides and proteins as well as to amplify nucleic acids for a reliable, sequence-specific detection, respectively [1, 2]. Moreover, optical methods like fluorescence microscopy [3–5], UV-VIS absorption [6] and vibrational spectroscopy (IR absorption, Raman spectroscopy) [7–9] are intensively used for bioanalytical purposes. Since every method needs a profound discussion about the capability of the detection strategy as well as presenting the current cutting-edge applications, this review focusses on surface enhanced Raman spectroscopy (SERS).

Nanostructured metal surfaces are the key element, which allow enhancing of the inherent weak Raman signals of different molecules. Thus, this technique combines the fingerprint specificity of the Raman method with an increased sensitivity by several orders of magnitude. A detailed description of the SERS method is found within the next chapter. When analyzing the SERS application fields based on the published items each year, it became evident, that SERS is preferentially applied in bioanalytics. This technique is used in recent years for studying not only the building blocks of life (nucleobases, amino acids) but also complex functional structures (nucleic acids and proteins) [10, 11]. Furthermore, SERS is an attractive tool for the investigation and characterization of whole prokaryotic cells, such as bacteria, bacterial spores, biofilm, viruses or even eukaryotic cells and tissues [12–14]. Also the use of SERS in biomedical and pharmaceutical research is a growing field in the last years [15, 16].

In order to investigate catalytic processes on metal surfaces like silver, gold or copper SERS is an excellent method due to the efficient enhancement of Raman modes involving in the rearrangement of atomic bonds directly at the surface [17]. Moreover, the detection of single molecules shows the great potential of this technique in sensitivity [18, 19]. To realize an increase of spatial resolution of SERS down to the nanometer scale, the technique is combined with scanning probe microscopy creating tip enhanced Raman spectroscopy [20, 21]. As an example, the TERS technique is applied to investigate inorganic samples like metalloid nanorods [22] or carbon nanotube composites [23], as well as biological samples such as insulin fibrils [24].

This review summarizes novel findings and trends in terms of biomolecules investigated by SERS, their characterization and detection introducing cutting-edge SERS-based application fields from the last 5 years.

2 Surface-enhanced Raman spectroscopy: a brief introduction

The optical properties of metallic nanoparticles were already used centuries ago, even before a detailed understanding of the nanoscale was developed. Using the example of the Late Roman Lycurgus Cup, gold nanoparticles were applied to color glass [25]. This phenomenon can be understood based on the interaction of light with metallic nanoparticles, whereby the electron cloud is oscillating with a frequency known as localized surface plasmon resonance [26]. The spectral position of the plasmon resonance is defined by the size, shape and material of the nanoparticle as well as the surrounding medium [27, 28] and is associated with the fulfillment of the plasmon resonance condition (Re(εi)≈-2εa; Im(εi)<<1; εi – dielectric constant of the metal; εa – dielectric constant of the surrounding medium) [29]. Finally a strong evanescent electromagnetic field is generated on the metallic surface of the nanoparticle [26]. Based on this strong field enhancement, various applications in ultrasensitive (bio)analytics are feasible.

To detect and investigate substances contactless with a molecular specificity combined with a simple sample preparation protocol, Raman spectroscopy is the method of choice in various application fields [8, 30–33]. Raman spectroscopy detects the vibrational modes of functional groups of molecules. This means that a Raman spectrum is dominated by group frequencies at distinct spectral positions and is therefore a molecular specific analytical tool. However, to detect analyt molecules in low concentrations, the application of Raman spectroscopy is often hindered by the inherent weak Raman intensity. Consequently, the effective field enhancement on the surface of metallic nanostructures is employed for the amplification of the Raman intensity of molecules located in close vicinity to the metallic surface (surface-enhanced Raman spectroscopy – SERS) [34–40]. Due to its high sensitivity based on the exploiting of the electromagnetic field enhancement on metal surfaces and fingerprint specificity from Raman spectroscopy, the SERS technique is proven as powerful method in chemical and biochemical applications [12, 41–44].

The SERS effect was first observed by investigating traces of pyridine adsorbed on roughened silver electrodes [45–47]. In order to explain the SERS enhancement two main mechanisms are discussed – the electromagnetic and the chemical enhancement, the electromagnetic being the main contribution with a maximum contribution to the enhancement factor in the order of 1011 [48]. When the excitation wavelength is resonant to the plasmon absorption profile of the nanoparticles, localized surface plasmon modes are excited and a strong evanescent electromagnetic field is induced on the metallic surface [29]. For a molecule, which is in close vicinity to the metallic surface, the Raman modes get efficiently enhanced because the Raman intensity is proportional to the squared local electromagnetic field intensity. Due to the characteristics of a Hertzian dipole the Raman-scattered light is – summarized for all orientations of the molecule – radiated in all spatial directions and finally collected by a microscope objective. Moreover, the Raman-scattered light undergoes a further enhancement when the Raman mode overlaps with the plasmonic profile. This effect is described in the literature as emission enhancement or second electromagnetic mechanism [49–51]. The chemical mechanism is understood as sum of different contributions: (1) a signal enhancement based on chemical interactions between molecule and nanoparticle in the ground state which are not based on an excitation within the system, (2) a SERS enhancement due to the resonant excitation of a charge transfer between nanoparticle and molecule, and (3) a resonance Raman enhancement due to the excitation of an electronic transition within the molecule [52]. One phenomenon in SERS spectroscopy called field gradient Raman effect is the detection of Raman modes under SERS conditions which are forbidden according to the spectroscopic selection rules, [53, 54]. Furthermore, variations in Raman intensity of different modes within the SERS spectrum are explainable by means of surface selection rules [55–57]. A molecule is characterized by its Raman tensor and orientation relative to the metallic surface. The enhancement of various Raman-active modes is dependent on the field components parallel and perpendicular to the surface. Only vibrations with a dynamic dipole normal to the surface are observed. In general, Raman modes with an orientation parallel to the surface are not or only with very weak signal intensity detectable. In summary, based on the discussed properties of the SERS mechanism several consequences for the reliable detection of molecules are expected. First, due to the range of the evanescent field on the surface of metallic nanostructures, the Raman signature is enhanced only in close vicinity up to roughly 10 nm by several orders of magnitude. Thus, the SERS response of large molecules like peptide and protein structures are dominated by the parts which are orientated very close to the metallic surface. Second, as already mentioned the orientation of a molecule relative to the metallic surface plays an important role in SERS, due to the surface selection rules. As a consequence, Raman modes perpendicular to the surface are preferably enhanced and thus, are dominating the SERS spectrum of the investigated biomolecules. Finally, since sulfur, nitrogen and oxygen containing chemical groups show a high affinity to silver and gold surface, these moieties bind preferably to the metal. Accordingly, vibrational modes of these surface-bound chemical groups are dominating the spectral response.

The development, preparation and characterization of SERS-active substrates are one of the most prominent research areas in SERS [25, 35, 36, 58]. Since thus is out of scope for this review article, this research field is discussed very briefly. Since the first observation of the SERS effect was done by investigating pyridine adsorbed on roughened silver electrodes [45–47, 59], historically the oldest SERS substrates are represented by roughened metal electrodes with nanostructured features generated by oxidation-reduction-cycles [60]. Today, metallic electrodes and their application as SERS substrate are of less importance. Most commonly, colloidal silver or gold nanoparticles are used for SERS-based investigations. Due to their simple and cost-efficient preparation protocol based on adding a reduction agent to an aqueous solution of, e.g., silver nitrate or chloroauric acid, colloidal metal nanoparticle suspensions are the most common used SERS substrate [58, 60, 61]. For the detection of single molecules by means of SERS, aggregated silver nanoparticles are used due to their excellent signal enhancement properties [36, 62, 63]. A third group of SERS substrates contains all plasmonic surfaces with randomly distributed or regular arranged metallic nanostructures. These are prepared by the deposition of metallic nanoparticles on a substrate, by the evaporation of pre-structured surfaces or by the application of lithographic methods [25, 35, 58, 60]. All here described SERS substrates consist mainly of silver, gold and copper structures, since for these materials the plasmon resonance condition is fulfilled within the visible and NIR spectral region.

3 SERS-based approaches to characterize and detect biomolecules

Biomolecules can be defined as any molecules that are produced by a living organism. From a chemical perspective the molecular precursors are carbon, hydrogen, oxygen, nitrogen, phosphor and sulfur. These atoms specifically associate to small biomolecules that are the building blocks of life: nucleotides, amino acids, fatty acids and monosaccharides. In general, the macromolecules itself tend to form larger biopolymers. These biopolymers belong to four major groups of essential components of prokaryotic as well as eukaryotic life: nucleic acids, proteins, lipids and carbohydrates. Besides, there are additional molecules with biological relevance that are synthesized within cells, like small or complex metabolites.

The characterization and detection schemes of nucleic acids and proteins and its building blocks using SERS as analytical tool are mainly addressed within this chapter. In literature, the band positions and the according assignment of Raman modes dominating the SERS spectra are in the focus of interest. As mentioned in the previous chapter, various effects influence the SERS intensity of vibrational modes and their intensity ratios and thus, play an important role in SERS analysis. (1) The orientation of a molecule relative to the metallic surface affects the SERS intensity, due to the surface selection rules. Raman modes perpendicular to the surface are preferably enhanced and are thus dominating the SERS spectrum. (2) Due to the evanescent field character of the electric field close to the metal surface, only Raman modes in close vicinity up to roughly 10 nm distance are enhanced by several orders of magnitude. (3) Vibrations of chemical groups which bind preferably to the metal – sulfur, nitrogen and oxygen showing a high affinity to silver and gold – are dominating the SERS spectrum. Within this chapter, an overview over the characterization and detection of biomolecules is given. Thus, the interested reader might be supported to evaluate the cutting-edge application fields of SERS introduced within the next chapter in relation to the own research area in terms of biomolecules.

3.1 Nucleotides and nucleic acids

Nucleotides are composed of a sugar, a phosphoryl group and a nucleobase, which can be a purine or a pyrimidine. There are two purine bases, adenine (A) and guanine (G) and three pyrimidine bases, cytosine (C), thymine (T) and uracil (U). Ribonucleic acid (RNA) and Deoxyribonucleic acid (DNA) are the polymers of nucleotides, which are designated by the type of the sugar in the backbone. In the case of RNA the sugar is D-ribose and for DNA it is 2-deoxy-D-ribose. The intrinsic genetic information of the DNA is translated via the messenger RNA (mRNA) into a defined peptide or protein sequence. Thus, mRNA molecules function as intermediate information storage. The expression of the DNA-coded information into a given amino acid chain can be regulated at various stages. One important regulation mechanism involves so-called non-coding RNAs that trigger gene silencing in eukaryotic cells.

Since the SERS spectra of DNA and RNA are dominated by adenine related modes [64, 65], the interpretation of adenine SERS spectra and the discussion of the high diversity of adenine compounds within the SERS fingerprints play an important role. Moreover, the attachment sites to the metal surface of adenine are often investigated and discussed in the literature. For clarification, the molecular structure of adenine is depicted in Figure 1. SERS spectra of adenine were recorded applying low concentrations of the nucleotide [66]. Adenine shows a strong Raman mode at 730 cm-1, which can be assigned to a breathing vibration and is dominating the SERS spectrum. In general, it is found that the SERS spectrum is dominated by in-plane vibrational modes. Thus, according to the surface selection rules, it is concluded, that the orientation of the adenine rings should be perpendicular to the silver surface. Due to the decreased intensity of the vibrational modes involving the nitrogen atoms N1, N3 and N9 as well as the preferentially enhanced SERS signals of Raman modes involving the external amino group and the nitrogen atom N7, it is suggested that the interaction between adenine and the silver surface takes place via the external amino group and N7. Furthermore, out-of-plane modes appear strongly weakened in the SERS spectrum but do not disappear completely. Thus, a slightly tilted orientation of the adenine molecule to the metal surface is probable. In further studies, the orientation of adenine and its derivatives polyadenine single stranded DNA (polyA) and adenosine monophosphate (AMP) were investigated at different pH values using SERS as well as surface enhanced IR absorption (SEIRA) on gold nanoshell particles [67]. Due to the protonation of adenine at various pH values, relative changes in the band intensity are found. The recorded SERS spectra of adenine and its investigated derivatives polyA and AMP are dominated by the adenine ring breathing mode at 735 cm-1 which is in consistency to other studies. Based on a detailed vibrational analysis of the recorded SERS as well as SEIRA spectra, the ‘end-on’ binding of adenine via the nitrogen atom N3 with the C-NH2 bond normal to the surface is suggested as the presumed orientation relative to the gold nanoshell surface. In the case of polyA and AMP, an adsorption via N3 is sterically hindered due to substituents bound at N9 and a binding via the external amino group is more likely. In order to increase the sensitivity for the detection of the nucleobases cytosine, adenine, thymine and guanine, the automated SERS detection at different pH values is developed [68] since the pH-sensitive SERS enhancement is known from various studies [65, 67, 69, 70]. As an example, the influence of the experimental conditions, like type of colloid, adenine concentration and pH value on the recorded SERS spectra is investigated [65]. It was found, that the SERS spectra are in agreement with the N1-protonated form of adenine at acidic pH values applying both silver and gold colloids. In contrast to previous studies, the recorded spectra at neutral and alkaline pH are assigned to the N9-deprotonated form rather than the neutral adenine molecule. Thus, the authors concluded that the ‘normal’ spectrum of adenine is attributed to the deprotonated form, similar to guanine and uracil. For low adenine concentrations applying silver colloids under neutral and alkaline conditions, the detected SERS spectra are attributed to different silver complexes of adenine. The high cross section of the silver complexes might be an explanation for the observed and difficult to interpret changes in intensity with varying the adenine concentrations. In addition, the SERS spectra of desoxyadenosine and 5′-desoxyadenosine monophosphate (5′-dAMP) are also dependent on the experimental conditions and the results indicate that both protonated and neutral forms of desoxyadenosine and 5′-dAMP are detectable as function of the pH value [70]. Since the ribose unit is substituted via the position N9, a deprotonated analogue to adenine is not detectable. Based on the weakened SERS intensity of the in-plane breathing modes, a much flatter orientation to the metal surface of desoxyadenosine is found for both the neutral and protonated forms compared to that of the corresponding forms of adenine. In the case of 5′-dAMP, a reorientation of the molecule on the metallic surface is observed as function of the concentration, pH and the used metal colloid. In summary, all introduced studies show that the SERS spectrum of adenine is dominated by the breathing mode of the entire molecule at around 730 cm-1, that the attachment of adenine is at least via the external amino group and moreover, hints are found for a N9-deprotonated form of the free adenine under normal conditions at neutral pH values.

Figure 1 Illustration of the molecular structure of adenine.
Figure 1

Illustration of the molecular structure of adenine.

In addition to the detailed investigation of nucleobases – especially adenine, also DNA and RNA are characterized by means of SERS. The here presented results are the starting point for a SERS-based application in label-free DNA and RNA analytics. In order to record reliable SERS spectra of nucleic acids, the interaction time between the molecules of interest and the metallic surface as well as the amount of molecules within the sample volume should be chosen with caution due to strong influences on the SERS signal [71]. A detailed analysis on the correlation of the molecular orientation and packing density of double-stranded DNA adsorbed on a metallic surface was performed [72]. In Figure 2, the detection strategy as well as the recorded SERS spectra is depicted. The SERS spectra were analyzed regarding their peak ratio of adenine and guanine vibrational modes as function of the DNA concentration. For high DNA concentrations which correspond to a high packing density, the adenine peak is more intense in comparison to guanine. In the case of low DNA concentrations and thus low packing densities, the guanine peak is increased with respect to the adenine vibrational mode. Based on these studies, higher adenine signal intensity indicates a less tilted orientation of the DNA molecule relative to the metal surface. A reorientation of double-stranded DNA towards an upright position was further done by means of insertion of polythymine single stranded DNA (polyT) which is detectable via an increased adenine peak intensity. Guanine-rich nucleic acid sequences are known to form a four-stranded DNA structure. The formation of these so-called G quadruplexes was characterized by means of SERS as a label-free analytical method [73]. The fluctuation in SERS intensity is quantitatively investigated before and after the quadruplex formation. Since the single DNA strands show a random orientation relative to the metal surface, the detailed analysis of the quadruplex SERS spectra suggests a perpendicular orientation of the G quadruplexes with respect to the metal surface. The characterization of the SERS signal as function of the numbers of G planes illustrates the improved stability of the quadruplex structure with an increasing number of G planes. In addition, the SERS technique is applied for the characterization of RNA. Adenine-containing microRNA chains adsorbed on a silver surface were investigated regarding their spectral features and moreover the structural arrangement relative to the metal surface [74]. The recorded SERS spectra, which are dominated by Raman modes of adenine, were analyzed in comparison with the spectral fingerprint of adenine, adenosine and the silver-adenine complex structure. Based on SERS and DFT results for adenine and adenosine, an interaction of microRNA via the N3 atom of adenine is proposed. Furthermore, the aromatic character of the pyrimidinic ring of adenine might favor a stronger metal-molecule interaction. Since the SERS signal is proportional to the amount of free residues interacting with the metallic surface, this technique was applied to monitor the catalysis of ribozyme cleavages – catalytic active hairpin structures found within RNA molecules [75]. In further studies, the interaction of small molecules containing three imidazole as well as three pyridine rings with single-stranded RNA polynucleotides, double-stranded DNA polynucleotides and calf thymus DNA was subject of research [76]. Due to the wavenumber downshift of imidazole and pyridine ring vibrations, an aromatic stacking structure of imidazole and pyridine aromatic moieties and DNA base-pairs is concluded. In summary, the results presented within this paragraph open the way to label-free DNA and RNA SERS-based detection schemes and moreover allow the investigation of interactions between small molecules and nucleic acids adsorbed on metallic surfaces. As a consequence of surface selection rules, Raman modes perpendicular to the surface are dominating the SERS spectrum. Due to the evanescent field on the metal surface, only Raman modes in close vicinity are observable under SERS conditions. Furthermore, vibrational modes of chemical groups which bind preferably to the metal are dominating the SERS spectrum. Thus, a change in the structural arrangement (reorientation) of nucleic acids with respect to the surface due to attachment of other molecule will cause a significant change in SERS intensity and band ratios of the Raman modes of the molecular compounds.

Figure 2 Characterization of the DNA orientation towards the metal surface. (A) Illustration of the SERS-based detection scheme on the basis of the double-stranded DNA tilt angle. (B) SERS response of double-stranded DNA for various concentrations (spectra 1–5 are related to the concentrations 40, 20, 10, 5 and 1.25 μm). Reprinted with permission from Barhoumi et al. [72]. Copyright 2008 American Chemical Society.
Figure 2

Characterization of the DNA orientation towards the metal surface. (A) Illustration of the SERS-based detection scheme on the basis of the double-stranded DNA tilt angle. (B) SERS response of double-stranded DNA for various concentrations (spectra 1–5 are related to the concentrations 40, 20, 10, 5 and 1.25 μm). Reprinted with permission from Barhoumi et al. [72]. Copyright 2008 American Chemical Society.

Miscellaneous proof-of-principle assays demonstrated the applicability of the SERS technique for the detection of adenine and its derivative as well as DNA or even RNA. As mentioned previously, adenine is one of the four RNA/DNA bases and therefore a main constituent of the nucleic acid metabolism in living organisms. Moreover adenine derivatives have pivotal roles in the cellular respiratory chain (e.g., adenosine triphosphate – ATP, nicotinamide adenine dinucleotide – NAD, flavin adenine dinucleotide – FAD) as well as second messengers in signal transduction processes (cyclic adenosine monophosphate – cAMP). Thus, the SERS-based detection of adenine or derivatives is addressed in recent articles. A planar chitosan-Ag SERS substrate was used for the quantification of adenine [77], whereas adenosine monophosphate (AMP) or deoxyadenosine monophosphate (dAMP) was detected by mean of SERS with noble metal particles [67, 78]. Also, DNA-aptamer biosensors were successfully used for analyzing adenosine/adenosine triphosphate [79–82]. Various papers introduced the applicability of the SERS-approach for the label-free detection of RNA or DNA molecules [83–85]. The successful use of SERS as a biosensor for DNA hybridization events with synthetic DNA or peptide nucleic acids (PNA) was described by several research groups [86–90]. Additionally, several sandwich hybridization assays concomitant with SERS detection were discussed in the literature [91–94]. The prominent spectral feature of adenine can also serve as an endogenous marker for a label-free SERS detection of DNA hybridization. As demonstrated in Figure 3 a DNA capture probe that harbors 2-aminopurine (2-AP) instead of adenine, which already preserved an adenine-like hybridization characteristic but lacked the 736 cm-1 breathing mode, was immobilized on a gold surface [64]. After the successful hybridization with the unlabeled complementary target DNA, that brought adenine within the sequence, the characteristic adenine peak was detectable. Another interesting tool for monitoring DNA hybridization was emphasized by dendrimer formation as a signal amplification strategy [95].

Figure 3 Label-free DNA detection applying SERS for read-out. Recorded SERS spectra of (A) adenine containing and (B) adenine non-containing DNA sequence are shown. Within the inset, the hybridization model between probe DNA having 2-aminopurine substituted for adenine and unlabeled target DNA is illustrated. Reprinted with permission from Barhoumi et al. [64]. Copyright 2010 American Chemical Society.
Figure 3

Label-free DNA detection applying SERS for read-out. Recorded SERS spectra of (A) adenine containing and (B) adenine non-containing DNA sequence are shown. Within the inset, the hybridization model between probe DNA having 2-aminopurine substituted for adenine and unlabeled target DNA is illustrated. Reprinted with permission from Barhoumi et al. [64]. Copyright 2010 American Chemical Society.

Damaged DNA, more precisely oxidized and UV-light exposed genomic DNA isolated from human cell lines, is detectable by label-free SERS [96]. The SERS community also implemented novel trends from the field of nucleic acid diagnostics, namely isothermal DNA amplification and microarray-based detection schemes. Rolling circle amplification, as one isothermal amplification strategy, allows the accumulation of target DNA and significantly improves the resulting SERS signal [97–99]. The creation of SERS substrates in array format on planar surfaces opens the possibility for multiplex detection by a plethora of dye-labeled oligonucleotides [100]. After the detailed discussion about the characterization as well detection of nucleotides and nucleic acids, innovative approaches related to the characterization of amino acids and their macromolecules peptides and proteins as well as promising detection schemes will be introduced within the next paragraph.

3.2 Amino acids and peptides/proteins

Amino acids are molecules containing an amine and a carboxylic group as well as a side-chain that specifies each amino acid. Amino acids itself are the most versatile group of biomolecules. They possess a variety of important biological function, which include precursors of hormones or neurotransmitters and, in particular, they serve as the building blocks of peptides and proteins. There are 22 proteinogenic amino acids which can be linked together to form a variety of smaller peptides or complex proteins. Within this paragraph, the characterization of amino acids, small peptides and proteins under SERS conditions is introduced. The following chapter includes the discussion about the preferred binding sites of amino acids relative to the metal surface which are important in spectral analysis, since the vibrational modes of these surface-binding moieties are dominating the SERS response. Moreover, the influences on the SERS signature of peptides and proteins are discussed and innovative detection schemes of these biomolecules are introduced.

In detailed studies by various research groups, the characteristic Raman modes of amino acids under SERS conditions and their dependencies as well as the adsorption behavior and orientation relative to the metallic surface was investigated. Applying SERS as analytical tool in combination with supporting DFT (density functional theory) calculations, the adsorption of the zwitterionic L-cysteine on the silver surface via the carboxylate, ammonium and sulphydryl groups is concluded [101]. Furthermore, it is found, that cysteine shows the tendency to form the corresponding dipeptide in solution. The characterization of L-tryptophan by means of SERS on silver nanoparticles shows the result that Raman modes of carboxylate and amino groups become strong under SERS conditions [102]. A time-dependent SERS study shows a unique spectral response, related with the most stable configuration and orientation to the metal surface, after a stabilization period [103]. Overall, it is concluded, that the attachment of this amino acid is preferably via the carboxylate and amino groups of L-tryptophan [102, 103]. A study on L-lysine attached to silver colloidal surfaces indicates no unique conformation and orientation relative to the metal surface [104]. Within the study, no identical SERS spectra were recorded under neutral pH conditions indicating various conformation of L-lysine with respect to the metal surface. In general, L-lysine interacts via the carboxylate and amino groups with the silver surface, which is also found for other amino acids. In the case of arginine, the preferably interaction between the amino acid and the silver surface is verified to be conducted through the guanidinium moiety and not via the carboxylate and amino groups [105]. Reorientation of amino acids adsorbed on silver surfaces is achieved by adding cations which becomes visible within the SERS response. It is found, that Ce3+ affects the structure of N-acetylalanine self-assembled at silver surfaces due to a binding reaction between Ce3+ and the carbonyl as well as the amino moieties [106]. The same is true for the effect of Pb2+ influencing the structure of L-glutathione due to the binding possibly occurring between the cation and the carboxyl and amino groups of the amino acids [107]. The adsorption behavior of enantiomeric and racemic forms of an amino acid was investigated by using methionine and collecting SERS spectra as function of pH value and electrode potential [108]. Non negligible spectral differences are found by comparing the SERS response of D-methionine and a racemic mixture of D- and L-methionine under acidic conditions for different electrode potentials. In comparison, in alkaline medium where both carboxylate and amino groups are responsible for the interaction with the electrode surface, no differences in spectral response are found in contrast to the observations under acidic conditions. Thus, indications are found for a chirality-dependent interaction based on intermolecular hydrogen bonds between adjacent molecules at pH 3. A further study shows the stereoselective interaction between phenylalanine and monolayers of cysteine adsorbed on rough silver surfaces [109]. Significant changes in SERS intensity were established for the interaction of L-phenylalanine with L-cysteine monolayer adsorbed on silver surfaces as well as for D-phenylalanine interacting with D-cysteine monolayers. This effect is attributed to variations in hydrogen bonding between the ammonium groups of the adsorbed cysteine molecule with the carboxylate groups of the phenylalanine. In summary, applying SERS for characterization of amino acids, a preferably binding via the carboxylate and amino moieties is found through the enhancement of the Raman intensity of vibrations which are influenced by the nature of the binding to the metal surface. A reorientation of amino acids is found for adding cations. Finally, indications are found that SERS is sensitive to the chirality of amino acids.

Within the following paragraph, the characterization of peptides, which are defined as small assemblies of amino acids, is introduced and the influences on the SERS response of peptides is discussed. The investigations are focused on studies of the orientation of peptides relative to the metallic surface and the therewith related marker modes within the SERS spectra. For L-carnosine an interaction primarily via the carboxylate group with the imidazole ring oriented perpendicular to the silver surface and the alanine moiety with parallel orientation having the amino group close to the silver surface is found [110]. A comprehensive study of alafosfalin and its analogs, which are phosphonodipeptides of alanine, was done by using colloidal silver particles as SERS platform supported by DFT calculations [111]. It is found, that these peptides adsorb as intact anionic species with the P-terminal acid group onto the silver surface. The detected SERS response indicate that both the constituent amino acids and the amide bond linking the N- and P-terminal residues interact with the metallic surface. Moreover, the adsorption mechanism of phosphonodipeptides containing N-terminal glycine on silver surface is subject of research [112]. Since a similar adsorption behavior is not found for all analogs, it is concluded, that the aliphatic spacer group play an important role within the interaction of these dipeptides with the silver surface. For dipeptides containing dehydroalanine and dehydrophenylalanine a strongly by dehydroresidues influenced SERS profile is detected [113]. Here, the most prominent SERS signal for the investigated dipeptides is attributed to the symmetric stretching vibration of the carboxylate group, which indicates the preferred adsorption via the deprotonated carboxyl group. Time-dependent and pH-dependent SERS studies were performed to characterize the binding characteristics of the homodipeptide diglycine to the silver surface [114]. A time-dependent increase in signal intensity is found for Raman modes representing the amino and amide moiety of the dipeptide. Furthermore, the pH value influences the way of adsorption due to the protonation as well as deprotonation of functional groups. Cysteine-containing aromatic peptides were characterized and the spectral response is compared with the SERS spectra of the cell-penetrating peptide oligomer penetratin [115]. It is concluded that, beside the protein backbone groups, aromatic amino acid residues provide the dominant features within the SERS spectra of peptides and proteins when present in the molecule of interest. In the case of phosphonate tripeptides, the adsorption on the metal surface is realized also via the phosphonate moiety which results in a reasonable SERS enhancement of the related vibrational modes [116]. Various studies are dealing with the characterization of bombesin, a 14-amino acid peptide, and its related fragments and analogues [117–121]. As a result of one of the comprehensive studies, the 8–14 bombesin fragment takes almost a parallel orientation with respect to the metal surface while the 1–5 peptide chain is orientated in a way, that it remains out of the range of the evanescent field caused by the metal surface [120]. Moreover, an effect on the orientation and adsorption mechanism is found when substituting amino acids within the chain. The same is true for fragments of human neurotensin and mutated analogues, where the SERS response indicates a slight influence on their adsorption behavior on silver surfaces due to the substitution of native amino acids in the investigated peptides [122]. When characterizing the bombesin subfamily peptides phyllolitorin and a peptide derived from Pseudophryne guntheri the moieties phenylalanine and tryptophan rings, the sulfur atom of methionine and the amide bond are in contact as well as in close vicinity to the metallic surface [123]. Comparative analysis of bombesin and five related peptides, among others a homolog in mammals known as neuromedin B, shows that the interaction between peptides and the metallic surface depends on the geometry of the tryptophan, amide bond, and S-C fragments of these molecules [121]. In a more detailed study, neuromedin B was investigated using different electrode materials and applied electrode potentials [124]. The authors demonstrated that the peptide adsorbs via the molecules backbone (including the phenylalanine and tryptophan rings) onto the silver, gold and copper electrode surfaces. Moreover, it is found, that the amide III mode, a typical Raman marker band of peptides and proteins, exhibit an electrochemical Stark effect which is reflected in potential depended frequencies. Furthermore, the carboxy terminal polypeptide of the β-subunit of human chorionic gonadotropin without carbohydrates moieties (P37) is characterized concerning the P37-metal interaction [125]. Here, the SERS analysis is performed based on the prior investigated oligopeptides with selected amino acid sequences MRKDV, ADEDRDA and LGRGISL [126, 127]. The SERS spectra of the investigated oligopeptides are dominated by the guanidinium moiety of the amino acid arginine which illustrates the orientation of the peptide relative to the metal surface that is driven by guanidinium [127]. In the case of ADEDRDA and LGRGISL, an additional interaction with the metal surface via other amino acids is found [126]. Based on these findings, the authors proposed the P37 metal interaction to be mediated via positively charged fragments of selected amino acids, mainly threonine, lysine and arginine [125]. Finally, evidences were found by means of SERS that the secondary structure of peptides is modified due to the adsorption on metallic surfaces [128]. To summarize, the SERS signature of peptides is not only depending on the orientation of the molecules to the metallic surface as well as on the chemical composition or more specifically the amino acid sequence of the peptide. Also the metal, pH value and electrode potential play a key role for the SERS spectrum. Finally, the paragraph illustrates how SERS spectra of peptides are influenced by various parameters. Thus the findings open a way to detect peptides in low concentrations and investigate interactions of peptides with other molecules.

The characterization of proteins, biological macromolecules consisting of one or more chains of amino acids, is in the focus of the next paragraph. Furthermore, approaches for an application of SERS as analytical tool for the detection of proteins are introduced. Fundamental studies on protein detection are introduced by investigation on the interaction of bovine serum albumin (BSA) with the surface of gold nanoparticles. The presence of the S-S stretching vibration bands of disulfide bridges indicates that the disulfide bonds are unbroken and the protein is not denatured at room temperature due to the attachment on the gold surface [129]. The adsorption of BSA to the metal surface is realized mainly via the tryptophan residue. Thus, BSA is specifically attached to the surface and undergoes only minor conformational changes when interacting with gold nanoparticles. In the case of lysozyme, the main Raman modes within the SERS spectrum are attributed to the amino acids tryptophan, tyrosine, phenylalanine and histidine, indicating the close vicinity of these moieties to the metal surface [130, 131]. In order to prevent spectral fluctuations due to reorientation of proteins during the analysis, an approach to bind proteins reproducible on silver surfaces is developed by fusing a silver-binding peptide to the C-terminus of maltose-binding protein [132]. Moreover, enzymes, which are selective catalysts and mostly proteins, were characterized by means of SERS. As a result of the orientation of coactivator-associated arginine methyltransferase 1 (CARM1) in relation to the nanostructured silver surface, strong Raman modes in the SERS spectrum occur which are attributed to amide vibrations and aromatic ring amino acids [133]. In the case of tyrosinase, the molecule is oriented in a way that the aromatic rings are parallel to the silver surface [134]. Furthermore, the adsorption behavior of the coenzyme Q10 radical intermediate as function of the potential is characterized [135]. For an applied potential lower than -0.30 V vs. SCE, a perpendicular orientation is found. The quinone ring of the reduced form adopts a face-on configuration for more positive electrode potentials than -0.30 V vs. SCE. The impact of the metal surface on the adsorption behavior of the protein bovine pancreatic trypsin inhibitor (BPTI) was investigated by using cetyltrimethylammonium bromide (CTAB)-protected gold nanoparticles in comparison to bare gold substrates [136]. While the disulfide stretching vibration of the protein deposited on the roughened gold substrate shows a significant heterogeneity, the S-S stretching signal remains unchanged for the application of CTAB-protected gold nanoparticle arrays. Thus, the authors concluded that the adsorption of BPTI on the protected nanoparticle array increases the stability of the protein. Cytochrome c, a small heme protein, was immobilized on 2-mercaptoethanesulfonate (MES) monolayers on silver nanostructures based on electrostatic attraction between the protein and the MES layer [137]. The linkage is characterized by SERS studies and finally the change of heme orientation relative to the metal surface deduced from the SERS response allowed the authors to propose protein dynamics. Furthermore, SERS demonstrated its benefit for the investigation of proteins on the basis of the characterization of myeloperoxidase (MPO), its corresponding antibody and their immunocomplex [138]. Within the SERS response variations in peak position and intensity can be assigned to conformational changes due to the immunocomplex formation. Another possible application of SERS is demonstrated by means of the characterization of amyloid beta peptide aggregates, which are related to the formation of neutric plaque, one of the primary pathological hallmarks of Alzheimer’s disease [139]. The spectral analysis of samples prepared by various aggregation procedures shows, that the Amide III band provides information about the secondary structure (α-helix and β-sheet) which allows understanding the properties of the protein aggregates at very low concentrations down to the femtomolar range. Using silver colloids as SERS substrate the cancer-promoting protein S100A4 was characterized [140]. Here, the SERS response of the dimer, tetramer and twelvemer aggregates demonstrates the possible discriminability of these species. Since dimeric forms are found in benign tumors, whereas oligomeric species are related to a late stage of cancer, this SERS study illustrates a possible application field of SERS in the field of biomolecule detection. Finally, the conformational changes of the photoactive yellow protein (PYP) are characterized on the single molecule level illustrating the high potential of SERS as analytical tool in detecting biomolecules [141]. In the ground state of the molecule, PYP has a para-Coumaric Acid (pCA) chromophore and its deprotonated phenolic oxygen is stabilized by means of hydrogen bridges. When absorbing a photon around 450 nm, the photocycle of PYP is initiated and yields a cascade of conformational changes. The detected spectral changes are attributed to temporal single molecule spectra and were related with photoisomerization, protonation of the chromophore and the carbonyl flip in the context of breaking hydrogen bridges.

Investigating protein detection with the SERS technique, the classical model thrombin is frequently chosen. Within this context, the measurement of free thrombin plasma levels represents a promising biomarker reflecting a patient’s individual hemostatic status to guide successful treatment decisions and to avoid bleeding or even thromboembolic complications [142]. Furthermore, thrombin is a useful marker for the diagnosis of pulmonary metastasis [143]. Aptamers, an alternative for antibodies, are artificial single-stranded oligonucleotides (ssRNA or ssDNA) that can bind to target molecules due to their specific three-dimensional structures [144]. Unsurprisingly, diverse aptamer-based immuno-SERS approaches for thrombin detection were published recently [145–151]. As an example, an aptamer-based detection scheme applying crystal violet as reporter molecule is depicted in Figure 4 [149]. The thrombin detection is performed according to the different amount of the reporter molecule crystal violet contributing to the overall SERS signal, based on electrostatic effects due to the changed structure of the thrombin-binding aptamer. A number of articles deal with the imaging of surface proteins by applying on-cell SERS. The purpose of one experimental setting was to elucidate the putative co-localization between the β2-adrenergic receptor and caveolin-3 on rat cardiomyoctes [152]. To that end, 4-(mercaptomethyl)-benzonitrile and d7-mercaptomethylbenzen are synthesized as Raman reporters for silver nanoparticle functionalization. Subsequently, 17% of the receptor co-localize with surface-exposed caveolin-3 protein. With the help of antibody-conjugated fluorescent SERS-dots the extracellular and respectively intracellular expression of CD34, Sca-1 and SP-C proteins are verified on bronchioveolar stem cell of the murine lung [153]. An immunolabeling process involving keratan-specific primary antibodies and Au-conjugated secondary antibodies that trigger silver particle localization is used for imaging of keratan sulfate within a corneal epithelium [154]. To understand mannoprotein membrane dynamics in a single living yeast cell, a SERS approach with silver nanoaggregates is employed [155].

Figure 4 Aptamer-based immuno-SERS detection scheme. (A) Using unmodified metallic nanostructured surfaces, a strong SERS signal of the positively charged reporter crystal violet is detected. (B) When binding a monolayer of aptamers, the amount of crystal violet molecules on the metal surface is decreased due to electrostatic effects. This results in a lower SERS signal. (C) In the case of thrombin binding to the aptamer structure, more crystal violet molecules are bound to the surface (B). Thus, the SERS signal increases after the thrombin binding, albeit to a lower level than in case (A). Reprinted with permission from Hu et al. [149]. Copyright 2009 American Chemical Society.
Figure 4

Aptamer-based immuno-SERS detection scheme. (A) Using unmodified metallic nanostructured surfaces, a strong SERS signal of the positively charged reporter crystal violet is detected. (B) When binding a monolayer of aptamers, the amount of crystal violet molecules on the metal surface is decreased due to electrostatic effects. This results in a lower SERS signal. (C) In the case of thrombin binding to the aptamer structure, more crystal violet molecules are bound to the surface (B). Thus, the SERS signal increases after the thrombin binding, albeit to a lower level than in case (A). Reprinted with permission from Hu et al. [149]. Copyright 2009 American Chemical Society.

Furthermore, the determination of enzymatic protein activity by means of SERS is addressed in recent studies. The monitoring of enzyme kinetics is an important issue of biochemistry, especially in the context of metabolism. With respect to this, SERS is a novel tool for determining the catalytic activity of an isolated succinate dehydrogenase by a color change of a redox dye. The artificial electron acceptor 2,6-dichlorphenolindophenol becomes SERS active when oxidized [156]. A study claims the applicability of SERS to quantitatively determine the proteolytic activity of a bovine protease on glass-immobilized substrates and rule out a detection limit of 0.43 mU/ml [157]. To detect the cAMP-protein kinase activity and further enzyme inhibition kinetics by diverse inhibitors, a SERS approach using anti-phosphoserine antibodies and gold nanoparticles is presented [158]. Moreover, the feasibility of SERS-based detection of two pterin entities is characterized by analyzing the serum from pterin-spiked rats [159]. A cascade of signal amplification reactions allows the SERS detection of lysozyme with a limit of detection of 10-15m [160]. These findings and results, which are summarized within the chapter, illustrate the potential of SERS for the detection of biomolecules. Since various detection schemes of biomolecules are often based on the interaction of a capture molecule with the biomolecule of interest, investigations regarding the interaction of various biomolecules are highlighted within the final section of this chapter.

3.3 Characterization of the interaction between biomolecules

The interaction of biomolecules due to the accumulation to affine groups is the basis of various detection schemes in bioanalytics. This section summarizes the recent findings regarding this important topic. Some immuno-SERS approaches, like the interaction of DNA-aptamers with target proteins and antigen-antibody recognition as specialized protein-protein interactions are highlighted in the relevant application chapters. The study of DNA-protein interaction in the background of transcription factor binding is characterized on the example of Wilms tumor 1 (WT1) [161]. Here, dye-labeled double-stranded DNA with a WT1 protein binding site is immobilized on metal nanoparticles and shows a significant SERS response dominated by the label molecule. The sequence-specific binding of the transcription factor to the DNA prevent the access of an exonuclease. Thus, the SERS signal of the dye label is still detectable. If no DNA-binding protein is present, the DNA gets accessible for the DNase, is exonucleolytic digested and the label molecule is released from the surface, which yields a lower SERS intensity. In a proof-of-principle approach, protein-protein and protein-small molecule interactions are detected on glass-surface immobilized human immunoglobulin G or avidin by the help of colloidal silver [162]. The combination of magnetic separation and SERS allows the detection of biotin-avidin interaction on Ag-coated magnetic particles [163]. Cell membranes are composed, amongst other biomolecules, of fatty acid chains that form a lipid bilayer. Since the incorporation of small molecules into cell membranes is of biological importance in terms of the characterization of drug effects, the insertion of ibuprofen into lipid layers is investigated [164]. Within these studies, a hybrid bilayer formed of dodecanethiol and phospholipids is prepared on the surface of gold nanoparticles. The intercalation of ibuprofen in the lipid structure is related to the enhancement of ibuprofen Raman marker modes showing a concentration dependency. On the basis of using silver nanoparticles as SERS substrate, the interaction of hypericin with low-density lipoproteins and phosphatidylcholine is characterized [165]. The authors observe that at high hypericin concentrations these molecules are placed in the outer shell of low-density lipoproteins forming aggregates. These results show the great potential of SERS for the investigation and detection of interactions between biomolecules. Within the next chapter which deals with cutting-edge application fields in bioanalytics of the SERS technique, the capability applying this method as analytical tool is demonstrated.

4 Cutting-edge application fields of SERS in bioanalytics

To reflect the publication activity in the field of SERS in conjunction with the term “biomolecule”, the detection of nucleic acids and proteins is mainly addressed within this chapter. In the following, state-of-the-art SERS applications in the fields of biomedicine and analytical chemistry are highlighted. Within this paragraph we report the use of the SERS technique for various biomedical applications, focusing, mainly on pathogen detection and cancer.

4.1 Pathogen sensing

The SERS technique is extensively studied since decades for the sensing of bacteria and viruses. This chapter gives an overview of the current SERS literature within this important biomedical field. A major challenge of modern medicine is a rapid and specific identification of bacterial pathogens in human body fluids. Efforts towards so-called point-of-care settings encompass the improvement of SERS with portable Raman microscopes, robust SERS substrates and even a simplification of sample preparation [166]. The current SERS publication activity points into the direction of an early diagnosis of bacteremia as well as urinary tract infections. Bacteremia, the presence of bacteria in the patient’s blood, resulted from severe infections at sites in the body, surgical wounds, or contaminated implanted devices and may lead to a sepsis. Sepsis is a cause of considerable mortality, morbidity, cost and health care utilization [167]. Urinary tract infections are common, with 50% of all women experiencing at least one in their lifetime. The rate of infection leads to high economic costs as well as patient morbidity [168]. The current gold standard for testing in hospitals is based on culturing and normally takes up to three days [169]. Due to our scope, that covers the SERS-based identification of biomolecules, the plethora of great publications that address whole bacteria capturing and detection is excluded from this review.

A whole bunch of articles reports a nucleic acid based identification of pathogens in conjunction with SERS. The advantages in this context are the capability of multiplexing and the improved sensitivity. Some proof-of-principle publications demonstrate the proper identification of various pathogens by using synthetic oligonucleotides. For example, the omp A gene of Clamydia trachomatis, a sexually transmitted bacteria, is used within an elegant exo-SERS assay [170]. Here, a sandwich-hybridization is performed with a magnetic bead immobilized capture probe, the target DNA and terminal phosphate/TAMRA-modified reporter probe. The 5′-phosphorylated reporter probe allow the partial degradation after the sequence-specific hybridization by a lambda-exonuclease [171]. Eventually, the shortened reporter probe is coated with silver nanoparticles and SERS spectra were recorded. Another sandwich hybridization assay highlight the usage of oligonucleotide-gold and oligonucleotide-silver nanoparticles for the probing of Staphylococcus aureus [172]. Two studies claim multiplexed detection of several E. coli strains by using either different dye-labeled reporter probes or even Ag/Au-colloids and multivariate data analysis [173, 174]. The diagnosis of pathogens is also presented in conjunction with the amplification of organism’s genomic DNA and SERS measurements. A multiplexed gold particle-on-wire sensor allowed the identification of Staphylococcus aureus and Vibrio vulnificus, which are causatives for severe human diseases like sepsis or gastroenteritis [175]. The detection limit of this sensor is depicted as 10 pM. The usability of SERS for the multiplexed detection of hospital-acquired methicillin-resistant Staphylococcus aureus is addressed [176]. To that end target genes are amplified by PCR. In case of hybridization with the complementary fluorescence-probe, the formed dsDNA did not adsorb on the surface of silver nanoparticles which result in a SERS signal reduction. Another assay based on SERS-primers and silver nanoparticles for the identification of Staphylococcus epidermis is depicted in Figure 5 [177]. In case of the absence of target DNA the SERS primer is closed, thus the DNA is predominantly double-stranded and has a lower affinity to adsorb to the nanoparticles, resulting in a low SERS response. The improved SERS assay using PCR and enzyme digestion to generate dye-labeled single-stranded DNA to detect pathogens like Staphylococcus aureus is highlighted in a recent article [178]. A specific detection of Staphylococcus aureus DNA through the combination of TaqMan assay with SERS is also realizable [179]. A successful multiplexed SERS detection of Mycoplasma mycoides, the causative agent of contagious bovine pleuropneumonia, is realized via combining magnetic nanoparticles and DNA hybridization [180]. SERS is able to provide a vibrational spectrum of the diverse cell wall molecules of a single bacterium within a few seconds. A fingerprint bacterial identification via their O-antigens, a part of the lipopolysaccharides that are exposed on the outer bacterial wall, is described [181]. In the proof-of-principle paper the reliable differentiation between Salmonella typhimurium and E. coli O16 is enabled by characteristic vibrational modes in the spectral regions 1250–1130 cm-1, 1030–730 cm-1 and 650–430 cm-1 that represented carbohydrates, most probably O-antigens. Using a liquid core photonic crystal fiber a detection limit of 106Shewanella oneides bacteria is achieved [182]. For an immuno-SERS approach metallic nanoparticles were functionalized with ligands like antibodies or aptamers that recognize target biomolecules on the surface of the pathogen [183]. The simultaneous detection of three different bacteria is accomplished by using gold, silver and Ag-Au core shell nanoparticles co-immobilized with anti-Salmonella typhimurium aptamers, anti-Staphylococcus aureus and anti-Escherichia coli O157:H7 antibodies respectively and unique Raman reporter molecules [184]. Gold nanoparticles with a coat of anti-protein A and DTNB as Raman reporter could be used for identification of Staphylococcus aureus via the bacterial surface antigen. A detection limit of 1 pg/ml of protein A is achieved [185]. Gold nanoparticles are coated with DSNB and antibodies, which are generated against a surface antigen of Mycobacterium avium subsp. paratuberculosis [186]. This bacillus is the causative of a cattle disease, which is responsible for devastating losses in the worldwide dairy production. The sandwich immuno-SERS assay allows the quantification of 500 mycobacteria/ml milk.

Figure 5 Illustration of the detection scheme of DNA based on the application of silver nanoparticles (labeled as Ag NP in the figure). (A) When no target DNA is present, the SERS primer is closed, which results in a low SERS signal. (B) In the case of the presence of complementary DNA, the loop structure is opened. Thus, the dye label is free to adsorb on the silver nanoparticle surface producing a high SERS signal. Reprinted with permission from van Lierop et al. [177]. Copyright 2011 American Chemical Society.
Figure 5

Illustration of the detection scheme of DNA based on the application of silver nanoparticles (labeled as Ag NP in the figure). (A) When no target DNA is present, the SERS primer is closed, which results in a low SERS signal. (B) In the case of the presence of complementary DNA, the loop structure is opened. Thus, the dye label is free to adsorb on the silver nanoparticle surface producing a high SERS signal. Reprinted with permission from van Lierop et al. [177]. Copyright 2011 American Chemical Society.

Similar to bacterial sensing, the fast diagnosis and therapy of viral infections is of great importance. The West Nile virus (WNV), a pathogen with a RNA genome, is the causative of West Nile fever and encephalitis. WNV is transmitted to its vertebrate hosts by an infected mosquito during blood feeding. The replication of the virus was documented in human monocytes in vitro, which could lead to transmission via blood transfusion [187]. Thus, the screening of donor blood is desirable and SERS techniques were implemented to identify WNV. Various sandwich hybridization assays, that utilize synthetic target DNA sequence instead of the WNV-RNA, are applied on either gold or paramagnetic iron nanoparticles [188, 189]. These approaches enable a limit of detection for WNV target nucleic acid of 10 pM and the authors claim a future adaption to a clinical detection or on-site platform using low-cost, handheld Raman instrumentation. A multiplexed immuno-SERS detection addressing envelope and capsid antigens of WNV is also realized [190]. The identification of new infections with human immunodeficiency virus (HIV) is crucial to prevent transmission as well as slowing disease progression by early medical intervention. Thus, HIV RNA-based tests may allow a very early diagnosis even before a common antibody test may become positive. In clinical laboratories the assessment of HI virus load is done by so-called branched DNA technologies [191]. A SERS-approach use HIV-1 DNA sequence for sandwich hybridization that induced the formation of a molecular junction based biosensor [192]. With regard to this platform, a concentration as low as 10-19 M HIV-1 DNA is detectable. An interesting experimental setup was invented to detect dengue virus sequences by on-chip SERS [193]. To date, the diagnosis of dengue, the most common mosquito-borne global disease, remains challenging. Infection by the RNA virus could lead to dengue fever or severe dengue illness [194]. Applying a microfluidic SERS assay, synthetic DNA sequences that mimic viral dengue serotypes are used as TAMRA-labeled targets. The complementary capture probes are immobilized on gold nanoparticles and the SERS measurement is performed within the optofluidic SERS-chip. Characteristic peaks are found at 1653 cm-1, 1360 cm-1 and 1219 cm-1 [193]. Of further clinical importance is the fast and reliable detection of influenza viruses. Nowadays an extensive menu of diagnostic tools is available [195]. Within this diagnostic topic a SERS immunosensor experiment is conducted [196]. Gold binding polypetides with hemagglutinin H1 antigen are immobilized on a nanoforest-structured substrate, followed by an immersion with anti-H1 detection antibodies. The relevant SERS peaks are found at 993 cm-1 and 1525 cm-1, which correspond to phenylalanine and tryptophan peaks, respectively. Moreover, within a short communication an aptamer-based detection of influenza nucleoproteins is presented on the basis of silver nanorods [197]. A label-free detection of DNA hybridization limited to short oligonucleotides is introduced for the analysis of the respiratory syncytial virus (RSV) [198]. This pathogen is the major causative of lower respiratory tract infections prevalent among infants and elderly [199]. The Epstein-Barr virus (EBV)-associated expression of latent membrane protein 1 (LMP1), a tumor marker for nasopharyngeal carcinoma, is addressed by immuno-SERS [200]. Au/Ag core-shell nanoparticles coated with LMP1-specific antibody and 4-MBA as a Raman reporter are used for the detection of LMP1 in paraffin-embedded nasopharyngeal carcinoma tissues of patients, that were EBV-positive and healthy volunteers. The authors claim that with the LMP1-SERS, 97.1% of the cancer patients are tested positive for the virus-encoded protein, whereas only in 64.7% the protein was detectable when conventional immunohistochemical staining technique was applied.

To summarize, the SERS technique is successfully applied to detect pathogen contamination on the basis of their building blocks such as DNA or proteins. Various detection schemes are introduced to open the way towards point-of-care applications of the SERS technique.

4.2 Cancer diagnosis and prognosis

Nowadays, cancer belongs like every other disease to the risks of life of every person. Everyone has to deal with a cancer diagnosis or treatment either in the personal life, via family members or within the circle of friends. Often the person affected ask “why me” and moreover a late diagnosis has the consequence of a high mortality. Thus, a reliable method for cancer diagnosis and prognosis is of great importance. Within this chapter we address important SERS activities in the field of cancer diagnosis/prognosis in conjunction with the terms non-invasive cancer detection, splice variants and single nucleotide polymorphisms, epigenetics, chemotherapeutics as well as theranostics.

The chances of curing cancer often correlate with the time of diagnosis. The earlier cancer is detected the better is the patient’s chance to be cured by an early treatment. In addition the risk for metastasis is reduced by early detection. Much effort has been made to develop rapid and non-invasive cancer screening tests. The preferable material for such a non-invasive diagnosis is blood plasma or serum and the sample preparation is simple during initial screening as well as following the patient’s treatment. This non-invasive cancer detection in conjunction with SERS includes the analysis of circulating nucleic acids, circulating tumor cells, immunophenotyping of cancer cells as well as the detection of various tumor markers and paves the way towards personalized medicine. Recently, a combination of membrane electrophoresis and SERS is applied for the optical detection of gastric and nasopharyngeal cancer [201, 202]. To that end, blood plasma from cancer patients and healthy volunteers is collected; the albumin and globulin protein fractions are separated by membrane electrophoresis, eluted and mixed with silver nanoparticles. A PCA reveals that the cancer and healthy group form distinct non-overlapping clusters. Again, silver nanoparticles are used as SERS substrate and directly mixed with blood plasma of gastric cancer patients or healthy subjects [203]. A subsequent PCA-LDA multivariate analysis point out that distinctive spectral SERS features and intensity differences between the two groups could reflect cellular changes associated with malignant transformation. The authors found for instance that the SERS peak at 725 cm-1 corresponding to the C-H in-plane bending mode of adenine exhibits higher signal in the cancer panel, indicating that there is an increase in the relative amount of nucleic acids in blood of cancer patient. This finding is matching reports that so-called circulating nucleic acids are found in higher concentrations when subjects are affected by a neoplastic disease [204]. Further cancer entities, namely nasopharyngeal, colorectal, esophageal and cervical cancer are analyzed using silver or gold colloids and blood plasma samples [205–208]. An increase of distinct SERS signals are observed for patients with pathologically confirmed carcinoma that indicate an abnormal nucleic acid metabolism. The reasons for an increase in the relative amount of cell-free nucleic acids in cancer patients’ blood have been proposed in the literature by cell death-related cell disintegration or release of intact cells in the blood stream followed by their lysis [209]. On the other hand a decrease of signal intensities for SERS-bands, which originate from tyrosine, serine, galactosamine and mannose reflects a higher tumor metabolism [206–208]. An overall decrease of the 1400 cm-1 peak intensity in the cancer patient’s plasma, reflecting the CH2 bending mode of collagen might account for elevated concentrations of matrix metalloproteinases that cleave the structure protein in cancer dysplasia [208]. A label-free serum RNA analysis enabled the detection of colorectal cancer by using 3D silver nanofilm as a SERS-active substrate [210]. The authors describe that RNA-SERS combined with PCA-LDA multivariate analysis was applicable for the differentiation of colorectal cancer patients from healthy subjects with a diagnostic sensitivity of 89.1% and a specificity of 95.6%. These results employing the SERS technique put forward the possibility of detecting circulating nucleic acids from blood plasma samples as biomarkers for monitoring cancer occurrence. Malignant cells can disseminate from the solid tumor and float as so called circulating tumor cells (CTCs) in the bloodstream of patients, potentially manifesting a metastasis [211]. The capturing from patient’s blood and the analysis of these rare cancer cells is of great importance for treatment decisions and monitoring. SERS is successfully applied for the detection of CTCs, even in complex matrices like blood [212, 213]. Magnetic beads functionalized with anti-epithelial cell adhesion molecule (EpCAM) antibodies are used for the capturing of SKBR3 cells, which mimic CTCs, spiked in human blood. Subsequent, the captured EpCAM-overexpessing cancer cells are detected with Nanoplex SERS tags [213]. Another experimental setting uses gold nanoparticles coated with antibodies against epidermal growth factor (EGF) to specifically capture CTCs in the blood of patients with head-and-neck cancer [212]. Here, 1–720 CTCs per ml of white blood cells get enriched. The diagnostic immunophenotyping of cancer cells is an important issue concerning the identification and of subpopulations and has a great value in predicting the invasiveness and metastatic potential of a tumor. In general, the amount of surface marker proteins, the cluster of differentiation (CD), is evaluated by flow cytometry. Antibodies against CD24 and CD44 antigens immobilized on gold nanoparticles enable the SERS-subtyping of three breast cancer cell populations [214]. The non-invasive but selective targeting of hematologic malignancies, e.g., chronic lymphocytic leukemia (CLL) using SERS gold nanoparticles is recently reported [215, 216]. To that end, e.g. anti-CD19 antibodies are conjugated covalently to gold nanoparticles. The antibodies recognize the CD19 surface antigen on CLL cells prepared from leukemia patients. The growing amount of publications which deals with cancer biomarker detection in patient’s blood/serum or on target cells by means of SERS reflects the efforts that are made within this field. Well characterized tumor markers are two ErbB family members: epidermal growth factor receptor (EGFR, ErbB-1) and human epidermal growth factor receptor 2 (HER2, ErbB-2). The EGFR is significantly overexpressed in aberrant crypt foci, which represent putative colon cancer precursors [217]. Immuno-SERS techniques using antibody- or affibody-conjugated SERS nanoparticles are applied to monitor the presence of EGFR in vivo [218, 219]. The authors conclude that the approach is capable of targeting EGFR biomarker in human cancer cells as well as in xenograft mouse tumor models. The Olivo group uses photonic crystal fibers filled with lysate of cancer cell lines to immobilize the target protein EGFR [220]. Then anti-EGFR-antibody conjugated SERS nanotags are flown through the hollow fiber and specifically bind to their target protein. An antibody-based SERS platform allows the multiplex detection of EGFR and HER2 on cancer cell lines [221]. Various publications report the identification of HER2-positive breast cancer cell lines by means of immuno-SERS [222–224]. The sequence-specific DNA analysis using sentinel nanoprobes is exemplarily demonstrated for the breast cancer marker HER2 and the proliferation marker Ki-67 [225]. The researchers use two different DNA-aptamer structures conjugated to silver nanoparticles for sensing the presence of synthetic HER2/Ki-67 target DNA. A well-known marker for tumor-associated angiogenesis is the vascular endothelial growth factor (VEGF) [226]. The functionality of a sandwich immunoassay is documented by the combination of the target VEGF immobilization on the gold array via antibodies and the detection by VEGF-aptamer-nanoparticles [227]. VEGF is also detectable by an immunosensor setting in blood plasma from breast cancer patients [228]. Hybrid Ag-Au-aptamer structures labeled with Rhodamine 6G are constructed that are capable of recognizing mucin 1 (MUC1) [229]. The transmembrane protein MUC1 is overexpressed in primary and metastatic breast cancers [230]. In their experimental section the authors demonstrate that MUC1-positive MCF-7 cells show a characteristic SERS signal, whereas MUC1-negative HepG2 or MCF-10A cells did not show any signal of the reporter. The screening for the cancer biomarker mucin 4 (MUC4) in the blood serum of patients can help to early diagnose pancreatic cancer [231]. Within this project, an immuno-SERS principle is performed to monitor the presence of MUC4 in various serum samples. The results indicate that sera from patients with prostate cancer produce significantly higher SERS response for MUC4 compared to sera from healthy subjects. Also, an antibody-based recognition of two important markers in routine cancer diagnosis, namely carcinoembryonic antigen (CEA) and α-fetoprotein (AFP), respectively, is demonstrated recently by means of SERS [232]. Here, a duplex SERS nanoprobe is created by coupling two antibodies against the cancer biomarkers on magnetic beads that allow the simultaneous detection of both markers in blood serum with a single exitation wavelength. A gold patterned microarray is used as a matrix for the immuno-based detection of AFP and angiogenin, the latter one seems to have an important role in tumor angiogenesis [233–235]. The multiplexed detection of the prostate specific antigen (PSA) in paraffin prostate cancer sections is investigated with antibody-conjugated nanoparticles or the antibody-fluorochrome-conjugate [236]. Both detection reagents possess similar performance in the tissue, supporting the development of Raman probes for PSA tumor marker screening in situ. Soluble PSA as a target protein is also detectable by antibody-conjugated hybrid Fe3O4/Au nanoparticles, emphasizing the advantages of magnetic particles in the bioassays [237]. Two further serum biomarkers, matrix metalloproteinase 7 (MMP-7) and carbohydrate antigen 19-9 (CA 19-9) are suitable for the diagnosis of early stage pancreatic cancer [238]. To that end, human serum was spiked with the recombinant proteins and the subsequent immuno-SERS allowed the measurement of these biomarkers. A biorecognition between the bacterial redox protein azurin and human p53 is exploited to realize a SERS-based detection of the tumor suppressor at concentrations of 500 fM in human serum [239, 240]. It is well documented in the literature that wild-type as well as mutant p53 protein is found in elevated levels in tumor cells concomitant with altered levels in the blood serum [241–243]. The SERS approaches for ultrasensitive detection of p53 involve a coupling of the protein via the interaction with 4-amithiophenol to gold nanoparticles and the subsequent interaction with immobilized azurin molecules on a glass surface [239, 240]. Azurin exhibit strong association with both, the wild-type as well as the mutated p53 protein and represents due its combination with SERS a promising approach for future tumor marker screenings. Another member of the p53 tumor suppressor family, p65, is addressed by an immuno-SERS approach together with the cyclin-dependent kinase inhibitor p21cip [244]. Here, the two proteins are spiked into human blood serum and are detected in a multiplexed fashion by antibody-functionalized Au-Ag nanorods. Immuno-SERS microscopy is applied for the fast detection of p63 in paraffin-embedded prostate biopsies [245, 246].

Single nucleotide polymorphisms (SNPs) are single base pair differences in the DNA among individuals that account for more than 90% of all sequence variations [247, 248]. Roughly one of every 100–300 bases in the human genome is a SNP [248, 249]. Numerous SNPs are detected as reliable diagnostic biomarkers for selection of appropriate drug therapies in the upcoming field of personalized medicine. For instance, a single nucleotide substitution in different codon positions of the proto-oncogenic Kirsten rat sarcoma (KRAS) is responsible for an activating mutation. Thus patients with this type of mutations will not respond to certain therapies [250]. An expansion of the SNP screening method repertoire is facilitated by SERS. A midsequence SNP discrimination by using synthetic oligonucleotides and gold nanoparticles is realized as dye-label-free SERS [251]. Another interesting approach utilizing sequence-specific hybridization and the ligase detection reaction (LDR) is introduced for discriminating KRAS SNPs [252, 253]. When the SNP sequence matches the discriminating primer, two adjacent oligonucleotide sequences get ligated and a SERS active product result. With the help of this LDR-SERS method a multiplexed SNP genotyping of the KRAS oncogene is possible down to a limit of detection of 10 pM [252]. A further SNP-related study depicts the application of gold nanowire/nanofilm-hybrids and a S1 nuclease digestion for assaying Wilson disease [254]. The SNP at codon 504 of the breast cancer 1 (BRCA1) gene is chosen to be detected by SERS [255]. The authors demonstrate the usability of a SERS molecular sentinel that comprises a silver nanoparticle plus the associated DNA hairpin structure that gets opened when the target DNA hybridizes to it. Thus the Raman label is physically separated from the particle and quenches the SERS signal.

In addition to SNP analysis, the detection of BRCA1 splice variants is of great importance. Changes in the alternative mRNA splicing profile of the BRCA1 gene is associated with malignant transformation in breast cancer [256]. The identification of cell and tissue specific splicing patterns helps to unravel the role of such genes in tumor development. In two proof-of-concept papers a SERS approach for a multiplexed detection of BRCA1 splice variants through nanoparticle-DNA-Raman-tag setup is presented with synthetic DNA [257, 258]. An advanced gene expression assay by SERS, involving the isolation of RNA from breast cancer cell lines followed by an translation into complementary DNA, sensitively quantify the expression levels of splice junction, skipping exon 9 and 10, of the BRCA1 gene [259, 260]. Moreover, SERS experiments are conducted that allowed the screening for wild-type and mutated BRCA1 peptides involving gold nanocuboids [261].

The SERS technique is also introduced by numerous groups to detect epigenetic modifications in the context of cancer diagnosis and prognosis. Epigenetic variations within the human genome are realized by means of non-coding RNAs or covalent DNA modifications. Micro RNAs (miRNAs) are small non-coding RNA molecules that regulate eukaryotic gene expression by mRNA degradation or inhibition of translation [262, 263]. It has been suggested that up to 30% of the human genome may be regulated by miRNAs [264]. Of further importance is the fact, that miRNA expression profiling can serve as diagnostic/prognostic biomarker of human cancers. Recent studies highlight the frequent downregulation of miR-15/16 in chronic lymphocytic leukemia [265, 266]. In addition, a reduced expression of multiple let-7 miRNA members has been found to be associated with human cancers [267]. Emerging techniques for the detection of miRNA alterations are bead-based flow cytometry and RNA-primed array-based Klenow extention (RAKE) that significantly reduced assay time [268, 269]. Nevertheless, the requirement of a hybridization step remained. To further accelerate analysis time, SERS is introduced as a label-free method for the detection and classification of miRNA. The applicability of SERS for miRNA analysis is demonstrated using silver nanorods and synthetic human miRNAs, like the pathogenic relevant miR-16 or let-7f [270–273]. The findings indicate that miRNA sequences could be accurately discriminated and quantified even in multicomponent mixtures. The fact, that let-7 miRNA family members produced SERS spectra very similar to one other in number and location of peaks is remarkable. But the spectra differed significantly in the relative intensities of some bands [270]. The SERS-based approaches enable rapid as well as quantitative miRNA detection using minimal sample volumes concomitant with an elimination of probe labeling and hybridization steps. Thus, the SERS technique is successfully adopted for the growing field of miRNA expression profiling. In eukaryotic cells methylated cytosine (mC) in so called CpG islands is the most common epigenetic mark [274]. More recently, hydroxymethylcytosine (hmC) was discovered as a novel DNA methylation tag [275, 276]. Methylated DNA represents an important epigenetic modification that provides information for altering the gene expression. Moreover, aberrant DNA methylation has been found to strongly correlate with human cancer. In that context the relationship of hypermethylation of CpG islands in the promotor regions of tumor-suppressor genes like p16INK4A and the promotion of tumor progression is reported [277, 278]. New scientific findings point in the direction of using methylation profiles as diagnostic or predictive biomarkers [278–280]. So far, a whole bunch of techniques have been developed for DNA methylation screening [281]. Recently, an article claims the single base extension reaction-based SERS for the detection of methylated p16INK4A-DNA [282]. Additionally, a SERS-based analysis enables the differentiation of the post-replicative DNA modifications mC and hmC [283]. The spectral variations between normal and hydroxymethylated DNA are significant, so an increase in the 665 cm-1 peak is an obvious marker of the presence of hmC. The acquired data point into the direction that SERS is a reliable method for discrimination between mC and hmC.

Besides the usefullness of the SERS technique for cancer diagnosis and prognosis, it can also be successfully exerted for monitoring the effect of anticancer drugs on target cells. With the help of a gold nanoflower-array the immobilization of living HepG2 liver cells is realized and this cell-based chip is used for studying the various effects of anticancer drugs on the cell behavior [284]. The chosen drugs are 5-fluorouracil and hydroxyurea which both interfere with DNA synthesis and cyclophosphamide, a DNA damaging agent. The recorded Raman spectra reflect a significant change in the cellular composition of nucleic acids and even proteins after the chemotherapeutical treatment. The measurement of drug concentrations in patient saliva instead of blood is a further non-invasive option to monitor drug metabolism and regulate drug dosage administration. In a proof-of-principle experiment disposable SERS-active capillaries filled with silver layers are tested for evaluation of 5-fluorouracil and its metabolite concentrations in saliva [285]. Furthermore, a DNA approach is performed to illuminate the interaction with the chemotherapy agent cisplatin [286]. After incubation of dsDNA with cisplatin the resulting intrastrand and interstrand adducts evoke spectral changes. These characteristic time-dependent spectra variations are caused by conformational distortions due to the ligand binding. The intracellular release of the chemotherapeutic agent 6-mercaptopurine, which is coupled to gold nanoparticles, can be triggered by glutathione. The release monitoring is performed by using SERS [287].

Within the emerging field of personalized medicine the development of so called theranostics is of great interest. Theranostics is referred to as a treatment combining therapeutics and diagnostics [288]. The aim is to integrate diagnostic approach and therapeutic intervention, allowing a simultaneous diagnosis, treatment as well as efficacy monitoring [289]. The earlier mentioned report about the binding of SERS-active nanostructures to MUC1-expressing cancer cell lines has also outlined the capability of absorbing near-infrared irradiation. They are therefore eligible for the photothermal therapy to selectively induce cell death of the targeted cancer cells without destroying surrounding tissues [230]. Further reports on photodynamic tumor therapy implementing SERS are presented in the literature [290, 291]. In general the photodynamic therapy (PDT) involves the selective uptake and accumulation of a photosensitizing agent in the target cells and the subsequent irradiation with light. The induced generation of reactive oxygen species (ROS) causes damaging of intracellular biomolecules, leading to cell death by apoptotic processes [292]. The intracellular accumulation of the photosensitizer-coated nanoparticles in cancer cells is realizable by either the use of cell-penetrating peptides [291] or enhanced permeability and retention (EPR) [290]. The latter EPR effect accounts for the uptake of gold nanorods by tumor cells and a large area of dead cells could be clearly seen in PDT-treated tumors. A non-invasive in situ release monitoring possibility is introduced very recently by the help of SERS [293]. The authors demonstrate the controlled release of the cargo doxorubicin from gold nanocages by applying heating, pointing into the direction of controlled drug delivery. The development of plasmonic vesicles assembled from SERS-active gold nanoparticles for targeted drug delivery, which can be tracked by Raman spectroscopy is reported [294]. The gold particles contain the Raman reporter BGLA, mixed brushes of hydrophilic poly ethylene glycol (PEG) and pH-sensitive hydrophobic copolymers of methyl methacrylate and 4-vinylpyridine (PMMAVP) to allow drug release, HER2 antibodies for cancer cell targeting and encapsulated doxorubicin as an anticancer drug. To verify the targeting properties concomitant with pH-triggered payload release, studies with HER2-overexpressing breast cancer cells were performed. The vesicle-coated cancer cells display a strong fingerprint Raman signal of the BGLA probe that gradually decrease with time caused by acidic driven disruption of the vesicles and a drug release. The entire strategy of this approach is depicted in Figure 6.

Figure 6 Theranostics strategy based on plasmonic vesicles built of gold nanoparticles. (A) Scheme of the applied gold nanoparticles coated with mixed polymer brushes and a Raman reporter as well as the drug-loaded plasmonic vesicle coated with antibodies for cancer cell targeting. (B) Illustration of the plasmonic vesicle’s attachment to the outer cell membrane, their uptake and finally the disruption of the SERS-active, pH-sensitive vesicle structure. Reprinted with permission from Song et al. [294]. Copyright 2012 American Chemical Society.
Figure 6

Theranostics strategy based on plasmonic vesicles built of gold nanoparticles. (A) Scheme of the applied gold nanoparticles coated with mixed polymer brushes and a Raman reporter as well as the drug-loaded plasmonic vesicle coated with antibodies for cancer cell targeting. (B) Illustration of the plasmonic vesicle’s attachment to the outer cell membrane, their uptake and finally the disruption of the SERS-active, pH-sensitive vesicle structure. Reprinted with permission from Song et al. [294]. Copyright 2012 American Chemical Society.

This chapter summarizes innovative strategies based on the SERS application in cancer diagnostics and prognosis. It is shown, that the SERS technique is used by various detection schemes based on silver and gold nanoparticles to contribute to the research field of non-invasive cancer detection, which includes the detection of circulating nucleic acids and tumor markers. Moreover, the sequence specific DNA detection to investigate splice variants and single nucleotide polymorphisms as well as the contribution to the research on epigenetics is highlighted by using SERS as analytical tool. The detection of chemotherapeutics is of great importance in terms of personalized medicine and drug treatment. Finally, the findings within the research field theranostics illustrate the great capability of SERS to detect and to release drugs within cancer cells.

4.3 Further SERS-based application strategies in biomedicine and biomolecule detection

As described in the previous paragraphs, SERS is an alternative method to detect pathogens and cancer alterations. In addition, SERS can be applied in other important biomedical fields. Thus, the following overview summarizes SERS applications concerning disease-related mutation detection, cell death and differentiation verification, forensics, drug and neurotransmitter screening, metabolism monitoring, prion protein detection in neurons, oxygen-binding protein sensing as well as targeting of immunoglobulins in blood. By employing SERS, the discrimination between single nucleotide and triplet deletion mutations as well as wild type of cystic fibrosis transmembrane regulator (CFTR) gene is accomplished [295, 296]. CFTR mutations cause cystic fibrosis, which is one of the most common life-threatening inherited diseases in Caucasians [297]. The use of an electroactive label for so-called SERS-E-melting experiments that enable the CTFR gene discrimination at the attomole level is reported [295]. A further SERS-E-melting approach is introduced for the discrimination of short tandem repeats [298]. These polymorphic short sequences are commonly used to determine genetic profiles for forensic individual identifications. Programmed cell death, alias apoptosis, is a highly important cellular process in vivo and the selective induction of apoptosis in cancer tissue is an established chemotherapeutic strategy. A label-free detection of apoptosis at the single-cell level is realized by silver nanoparticles [299]. After apoptosis induction by detergents only a weak SERS signal of DNA is observed due to specific cleavage of DNA strands as a late apoptotic event. The verification of cell differentiation is also accomplished by SERS [300, 301]. The in vitro differentiation of neuronal progenitor cells into terminal differentiated neurons is realized by staurosporine. To record SERS spectra corresponding to nucleic acids, the accumulation of gold nanoparticles within the nucleus of the different cell types is done by using the nuclear localization signal of the SV40-large T antigen [301]. The tracking of differentiation of mouse embryonic stem cells by intracellular nanoparticles show the following results: SERS peaks derived from undifferentiated cells are mostly attributed to nucleic acids (high proliferation rate) whereas the SERS peak at 1604 cm-1 originated from mitochondria of the terminally differentiated cardiomyocytes (high metabolic activity) [300]. As mentioned earlier, aptamers are a versatile tool for bioanalytical purposes. In conjunction with SERS the conformational changes in the aptamers induced by target molecule interaction are monitored on a label-free level. To that end the drug cocaine is chosen as a model substance [302, 303]. An antibody-based immuno-SERS approach use silver-coated carbon nanotubes for the detection of the major cocaine metabolite benzoylecgonine [304]. Serotonin, a ubiquitous neurotransmitter is quantified in a background of other indoles using SERS [305]. Lastly, SERS assisted ultra-fast peptide screening is described as a novel promising tool for drug discovery [306]. The authors claim the usability of microbeads with Au-Ag nanoparticle coat as a solid support for the synthesis of combinatorial peptide libraries. The subsequent SERS analysis enables the identification of several tripeptide combinations as a consequence of the unique vibrational signature of each amino acid. A transcutaneous in vivo glucose measurement in a rat model is realized by SERS [307]. This work represents a significant step towards an implantable, real time glucose SERS-sensor. Another example of metabolism monitoring is the detection of lactic acid at physiological concentrations in blood samples [308]. The study of prion proteins is performed by several SERS approaches [309–312]. The focus lay on the native cellular prion protein (PrPc), which is mainly expressed on the cell surface of neurons. The physiological function of PrPc remains elusive, although it has been related to copper metabolism [313]. SERS allow the tracking of PrPc-Cu2+ interaction on the cell membrane of different neuronal cells [309, 312]. The detection of PrPc either in bovine serum or in blood samples with silver nanorods is displayed [310, 311]. A proof-of-concept paper focuses on the combination of isotachophoretic flow electrophoresis on a chip-device and SERS for the detection of myoglobin, an abundant oxygen-binding protein in muscle cells [314]. To gain relevant information about putative drug effects on different hemoglobin subpopulations in intact erythrocytes, SERS based on silver colloids is performed [315]. The group shows that submembrane and cytosolic hemoglobin molecules display significant conformation differences. Beyond that, an immuno-SERS approach is applied to detect immunoglobulin E (IgE) in blood serum of rabbits [316]. Gold nanoparticles are coated with protein A/G and malachite green as a Raman tag. The formed immuncomplex allow the monitoring of rabbit serum IgEs by SERS and represents an alternative to classical ELISA formats. Mouse or human IgG is chosen as a model protein to demonstrate the feasibility of SERS as a novel spectroscopic technique in clinical diagnostics [317, 318].

Finally, novel strategies regarding SERS for toxin monitoring in water and food, identification of genetically modified organisms, extraterrestrial life sensing and even art evaluation are presented. As an example, ricin, a potential bioterrorism agent, is successfully quantified by either an aptamer-based SERS approach or a pre-immunomagnetic separation, respectively [319, 320]. At least 100 ng/ml ricin B is detectable in complex matrices like milk. This highlighted the promising potential of the SERS technique for bioweapon screening and the feasibility of using portable Raman instruments for on-site detection [319]. The assembly of gold nanorods is used for the principle detection of microcystin, an algae toxin pollution in drinking water reservoirs [321]. The presence of the toxin interfered with the antibody-antigen triggered assembly of nanorod chains, thus significantly reducing the SERS signal for 4-ATP. In a study, the screening for genetically modified organisms is performed by combining magnetic pre-concentration of the target DNA, which was the 35S gene promoter, with further SERS measurements. A real sample analysis with Bt-176 maize DNA demonstrates the feasibility of the assay concept [322]. Surprisingly, SERS was utilized for analyzing nucleobases that mimic microbial life on pyroxene rocks [323]. The authors depict that the experimental procedure could be easily adapted for in situ recognition of life traces in extraterrestrial environments, mainly detecting the marker band of adenine at 735 cm-1. Moreover, SERS is introduced in artworks. Here, the precise identification and localization of proteins, such as ovalbumin, collagen or casein is demonstrated by indirect immuno-SERS in multilayered paintings [324].

In addition to the application fields of SERS pathogen sensing and cancer diagnostics, this section shows the potential of SERS in further research areas. The innovative, here described approaches might be the starting point for the establishment of new application schemes employing the SERS technique.

5 Conclusion

This review article summarizes the recent results and developments of using SERS for the detection of biomolecules. First, the characterization of biomolecules and their detection schemes were discussed. In general, the SERS intensity depends on various parameters like the distance of the molecule from, its orientation relative to and its affinity to the molecule towards the metal surface. The great capability of the SERS technique as an analytical tool to detect biomolecules is illustrated by cutting-edge applications in the fields of pathogen sensing and cancer diagnosis. In the case of pathogen sensing, the bacterial or viral contamination is detected via their building blocks such as DNA and proteins. Moreover, innovative strategies in cancer diagnosis are introduced. As an example, the SERS technique is applied for non-invasive cancer detection (which includes the detection of circulating nucleic acids and tumor markers), epigenetics, chemotherapeutic drug monitoring and theranostics. This highlights the contribution of the SERS technique among others to personalized medicine, which means an inclusion of individual circumstances to the treatment of patients. Finally, novel strategies in cell investigations, forensics, biomarker screening as well as environment monitoring and even art are introduced. This comprehensive summary in the field of SERS-based biomolecule detection might be a starting point for the interested reader from different research areas to create own approaches for a SERS-based detection in biochemical, biological or biomedical applications.


Corresponding author: Jürgen Popp, Institute of Photonic Technology (IPHT) Jena, Albert-Einstein-Straße 9, 07745 Jena, Germany; and Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743 Jena, Germany, e-mail:
aThe authors contributed equally to this manuscript.

Acknowledgement

Funding of the research projects “QuantiSERS” and “Jenaer Biochip Initiative 2.0” within the framework “Unternehmen Region – InnoProfile Transfer” the Federal Ministry of Education and Research, Germany (BMBF) is gratefully acknowledged.

References

[1] Ma L-n, Zhang J, Chen H-t, Zhou J-h, Ding YZ, Liu YS. An overview on ELISA techniques for FMD. Virology J 2011;8: Article no. 419, 9 pages.10.1186/1743-422X-8-419Search in Google Scholar PubMed PubMed Central

[2] Malou N, Raoult D. Immuno-PCR: a promising ultrasensitive diagnostic method to detect antigens and antibodies. Trends in Microbiol 2011;19:295–302.10.1016/j.tim.2011.03.004Search in Google Scholar PubMed

[3] Seibel J, Koenig S, Goehler A, Doose S, Memmel E, Bertleff N, Sauer M. Investigating infection processes with a workflow from organic chemistry to biophysics: the combination of metabolic glycoengineering, super-resolution fluorescence imaging and proteomics. Expert Rev Proteomics 2013;10:25–31.10.1586/epr.12.72Search in Google Scholar PubMed

[4] Petryayeva E, Algar WR, Medintz IL. Quantum dots in bioanalysis: a review of applications across various platforms for fluorescence spectroscopy and imaging. Appl Spectrosc 2013;67:215–52.10.1366/12-06948Search in Google Scholar PubMed

[5] Pampaloni F, Ansari N, Stelzer EHK. High-resolution deep imaging of live cellular spheroids with light-sheet-based fluorescence microscopy. Cell Tissue Res 2013;352:161–77.10.1007/s00441-013-1589-7Search in Google Scholar PubMed

[6] Nicklas JA, Buel E. Quantification of DNA in forensic samples. Anal Bioanal Chem 2003;376:1160–7.10.1007/s00216-003-1924-zSearch in Google Scholar PubMed

[7] Ataka K, Kottke T, Heberle J. Thinner, smaller, faster: IR techniques to probe the functionality of biological and biomimetic systems. Angew Chem-Int Edit 2010;49:5416–24.10.1002/anie.200907114Search in Google Scholar PubMed

[8] Brauchle E, Schenke-Layland K. Raman spectroscopy in biomedicine - non-invasive in vitro analysis of cells and extracellular matrix components in tissues. Biotechnol J 2013;8:288–97.10.1002/biot.201200163Search in Google Scholar PubMed PubMed Central

[9] Downes A, Elfick A. Raman spectroscopy and related techniques in biomedicine. Sensors 2010;10:1871–89.10.3390/s100301871Search in Google Scholar PubMed PubMed Central

[10] Culha M. Surface-enhanced raman scattering: an emerging label-free detection and identification technique for proteins. Appl Spectrosc 2013;67:355–64.10.1366/12-06895Search in Google Scholar PubMed

[11] Bantz KC, Meyer AF, Wittenberg NJ, Im H, Kurtulus O, Lee SH, Lindquist NC, Oh SH, Haynes CL. Recent progress in SERS biosensing. Phys Chem Chem Phys 2011;13:11551–67.10.1039/c0cp01841dSearch in Google Scholar PubMed PubMed Central

[12] Pahlow S, Maerz A, Seise B, Hartmann K, Freitag I, Kämmer E, Böhme R, Deckert V, Weber K, Cialla D, Popp J. Bioanalytical application of surface- and tip-enhanced Raman spectroscopy. Eng Life Sci 2012;12:131–43.10.1002/elsc.201100056Search in Google Scholar

[13] Negri P, Dluhy RA. Ag nanorod based surface-enhanced Raman spectroscopy applied to bioanalytical sensing. J Biophotonics 2013;6:20–35.10.1002/jbio.201200133Search in Google Scholar PubMed PubMed Central

[14] Vitol EA, Orynbayeva Z, Friedman G, Gogotsi Y. Nanoprobes for intracellular and single cell surface-enhanced Raman spectroscopy (SERS). J Raman Spectrosc 2012;43:817–27.10.1002/jrs.3100Search in Google Scholar

[15] Vendrell M, Maiti KK, Dhaliwal K, Chang Y-T. Surface-enhanced Raman scattering in cancer detection and imaging. Trends Biotechnolo 2013;31:249–57.10.1016/j.tibtech.2013.01.013Search in Google Scholar PubMed

[16] Kho KW, Fu CY, Dinish US, Olivo M. Clinical SERS: are we there yet?. J Biophotonics 2011;4:667–84.10.1002/jbio.201100047Search in Google Scholar PubMed

[17] Joseph V, Engelbrekt C, Zhang J, Gernert U, Ulstrup J, Kneipp J. Characterizing the kinetics of nanoparticle-catalyzed reactions by surface-enhanced Raman scattering. Angew Chem-Int Edit 2012;51:7592–6.10.1002/anie.201203526Search in Google Scholar PubMed

[18] Le Ru, EC, Etchegoin PG. Single-molecule surface-enhanced Raman spectroscopy. In: Johnson MA, Martinez TJ, eds. Annu Rev Phys Chem 2012;63:65–87.Search in Google Scholar

[19] Kneipp J, Kneipp H, Kneipp K. SERS - a single-molecule and nanoscale tool for bioanalytics. Chem Soc Rev 2008;37: 1052–60.10.1039/b708459pSearch in Google Scholar PubMed

[20] Stadler J, Schmid T, Zenobi R. Developments in and practical guidelines for tip-enhanced Raman spectroscopy. Nanoscale 2012;4:1856–70.10.1039/C1NR11143DSearch in Google Scholar PubMed

[21] Treffer R, Boehme R, Deckert-Gaudig T, Lau K, Tiede S, Lin X, Deckert V. Advances in TERS (tip-enhanced Raman scattering) for biochemical applications. Biochem Soc T 2012;40:609–14.10.1042/BST20120033Search in Google Scholar PubMed

[22] Reparaz JS, Peica N, Kirste R, Goni AR, Wagner MR, Callsen G, Alonso MI, Garriga M, Marcus IC, Ronda A, Berbezier I, Maultzsch J, Thomsen C, Hoffmann A. Probing local strain and composition in Ge nanowires by means of tip-enhanced Raman scattering. Nanotechnology 2013;24:185704.10.1088/0957-4484/24/18/185704Search in Google Scholar PubMed

[23] Suzuki T, Yan X, Kitahama Y, Sato H, Itoh T, Miura T, Ozaki Y. Tip-enhanced Raman spectroscopy study of local interactions at the interface of styrene-butadiene rubber/multiwalled carbon nanotube nanocomposites. J Phys Chem C 2013;117:1436–40.10.1021/jp309217ySearch in Google Scholar

[24] Kurouski D, Deckert-Gaudig T, Deckert V, Lednev IK. Structural characterization of insulin fibril surfaces using Tip Enhanced Raman Spectroscopy (TERS). Biophys J 2013;104:49A–49A.10.1016/j.bpj.2012.11.311Search in Google Scholar

[25] Love SA, Marquis BJ, Haynes CL. Recent advances in nanomaterial plasmonics: fundamental studies and applications. Appl Spectros 2008;62:346A–362A.10.1366/000370208786822331Search in Google Scholar PubMed

[26] Willets KA, Van Duyne RP. Localized surface plasmon resonance spectroscopy and sensing. Annu Rev Phys Chem 2007;58:267–97.10.1146/annurev.physchem.58.032806.104607Search in Google Scholar PubMed

[27] Xia Y, Halas NJ. Shape-controlled synthesis and surface plasmonic properties of metallic nanostructures. MRS Bull 2005;30:338–48.10.1557/mrs2005.96Search in Google Scholar

[28] Haes AJ, Haynes CL, McFarland AD, Schatz GC, Van Duyne RP, Zou S. Plasmonic materials for surface-enhanced sensing and spectroscopy. MRS Bull 2005;30:368–75.10.1557/mrs2005.100Search in Google Scholar

[29] Schatz GC, Van Duyne RP. Electromagnetic mechanism of surface-enhanced spectroscopy. Handbook of Vibrational Spectroscopy (Wiley & Sons, Chichester) 2002;1:759–74.Search in Google Scholar

[30] Schmitt M, Popp J. Raman spectroscopy at the beginning of the twenty-first century. J Raman Spectros 2006;37:20–8.10.1002/jrs.1486Search in Google Scholar

[31] Kudelski A. Analytical applications of Raman spectroscopy. Talanta 2008;76:1–8.10.1016/j.talanta.2008.02.042Search in Google Scholar PubMed

[32] Petry R, Schmitt M, Popp J. Raman spectroscopy-a prospective tool in the life sciences. ChemPhysChem 2003;4:14–30.10.1002/cphc.200390004Search in Google Scholar PubMed

[33] Ferrari AC, Basko DM. Raman spectroscopy as a versatile tool for studying the properties of graphene. Nat Nanotechnol 2013;8:235–46.10.1038/nnano.2013.46Search in Google Scholar PubMed

[34] Hering K, Cialla D, Ackermann K, Dörfer T, Möller R, Schneidewind H, Mattheis R, Fritzsche W, Rösch P, Popp J. SERS: a versatile tool in chemical and biochemical diagnostics. Anal Bioanal Chem 2008;390:113–24.10.1007/s00216-007-1667-3Search in Google Scholar PubMed

[35] Cialla D, Maerz A, Boehme R, Theil F, Weber K, Schmitt M, Popp J. Surface-enhanced Raman spectroscopy (SERS): progress and trends. Anal Bioanal Chem 2012;403:27–54.10.1007/s00216-011-5631-xSearch in Google Scholar PubMed

[36] Kleinman SL, Frontiera RR, Henry A-I, Dieringer JA, Van Duyne RP. Creating, characterizing, and controlling chemistry with SERS hot spots. Phys Chem Chem Phys 2013;15:21–36.10.1039/C2CP42598JSearch in Google Scholar

[37] Wang Y, Schluecker S. Rational design and synthesis of SERS labels. Analyst 2013;138:2224–38.10.1039/c3an36866aSearch in Google Scholar PubMed

[38] Bell SEJ, Sirimuthu NMS. Quantitative surface-enhanced Raman spectroscopy. Chem Soc Rev 2008;37:1012–24.10.1039/b705965pSearch in Google Scholar PubMed

[39] Smith WE. Practical understanding and use of surface enhanced Raman scattering/ surface enhanced resonance Raman scattering in chemical and biological analysis. Chem Soc Rev 2008;37:955–64.10.1039/b708841hSearch in Google Scholar PubMed

[40] Stiles PL, Dieringer JA, Shah NC, Van Duyne RP. Surface-enhanced Raman spectroscopy. Annu Rev Anal Chem 2008;1:601–26.10.1146/annurev.anchem.1.031207.112814Search in Google Scholar PubMed

[41] Xie W, Schluecker S. Medical applications of surface-enhanced Raman scattering. Phys Chem Chem Phys 2013;15:5329–44.10.1039/c3cp43858aSearch in Google Scholar PubMed

[42] Anker JN, Hall WP, Lyandres O, Shah NC, Zhao J, Van Duyne RP. Biosensing with plasmonic nanosensors. Nature Mater 2008;7:442–53.10.1038/nmat2162Search in Google Scholar PubMed

[43] Graham D, Goodacre R. Chemical and bioanalytical applications of surface enhanced Raman scattering spectroscopy. Chem Soc Rev 2008;37:883–4.10.1039/b804297gSearch in Google Scholar

[44] Huh YS, Chung AJ, Erickson D. Surface enhanced Raman spectroscopy and its application to molecular and cellular analysis. Microfluidics and Nanofluidics 2009;6:285–97.10.1007/s10404-008-0392-3Search in Google Scholar

[45] Fleischmann M, Hendra PJ, McQuillan AJ. Raman spectra of pyridine adsorbed at a silver electrode. Chem Phys Lett 1974;26:163–6.10.1016/0009-2614(74)85388-1Search in Google Scholar

[46] Albrecht MG, Creighton JA. Anomalously intense Raman spectra of pyridine at a silver electrode. J Am Chem Soc 1977;99:5215–7.10.1021/ja00457a071Search in Google Scholar

[47] Jeanmaire DL, Van Duyne RP. Surface Raman spectroelectrochemistry. Part I. Heterocyclic, aromatic, and aliphatic amines adsorbed on the anodized silver electrode. J Electroanal Chem Interf Electr 1977;84:1–20.Search in Google Scholar

[48] Xu H, Aizpurua J, Kaell M, Apell P. Electromagnetic contributions to single-molecule sensitivity in surface-enhanced Raman scattering. Phys Rev E: Stat, Nonlin Soft Matter Phys 2000;62:4318–24.10.1103/PhysRevE.62.4318Search in Google Scholar

[49] Cialla D, Petschulat J, Huebner U, Schneidewind H, Zeisberger M, Mattheis R, Pertsch T, Schmitt M, Möller R, Popp J. Investigation on the second part of the electromagnetic SERS enhancement and resulting fabrication strategies of anisotropic plasmonic arrays. ChemPhysChem 2010;11:1918–24.10.1002/cphc.200901009Search in Google Scholar PubMed

[50] Itoh T, Yoshida K, Biju V, Kikkawa Y, Ishikawa M, Ozaki Y. Second enhancement in surface-enhanced resonance Raman scattering revealed by an analysis of anti-Stokes and Stokes Raman spectra. Phys Rev B 2007;76:085405/085401–085405/085405.10.1103/PhysRevB.76.129901Search in Google Scholar

[51] Zhang WH, Fischer H, Schmid T, Zenobi R, Martin OJF. Mode-selective surface-enhanced Raman spectroscopy Using nanofabricated plasmonic dipole antennas. J Phys Chem C 2009;113:14672–5.10.1021/jp9042304Search in Google Scholar

[52] Jensen L, Aikens CM, Schatz GC. Electronic structure methods for studying surface-enhanced Raman scattering. Chem Soc Rev 2008;37:1061–73.10.1039/b706023hSearch in Google Scholar PubMed

[53] Knoll P, Marchl M, Kiefer W. Raman-spectroscopy of microparticles in laser-light traps. Indian J Pure Ap Phy 1988;26:268–77.Search in Google Scholar

[54] Ayars EJ, Hallen HD. Electric field gradient effects in Raman spectroscopy. Phys Rev Lett 2000;85:4180–3.10.1103/PhysRevLett.85.4180Search in Google Scholar PubMed

[55] Otto A, Mrozek I, Grabhorn H, Akemann W. Surface-enhanced Raman scattering. J Phys: Condens Matter 1992;4:1143–212.10.1088/0953-8984/4/5/001Search in Google Scholar

[56] Gao X, Davies JP, Weaver MJ. Test of surface selection rules for surface-enhanced Raman scattering: the orientation of adsorbed benzene and monosubstituted benzenes on gold. J Phys Chem 1990;94:6858–64.10.1021/j100380a059Search in Google Scholar

[57] Moskovits M, Suh JS. Surface selection rules for surface-enhanced Raman spectroscopy: calculations and application to the surface-enhanced Raman spectrum of phthalazine on silver. J Phys Chem 1984;88:5526–30.10.1021/j150667a013Search in Google Scholar

[58] Brown RJC, Milton MJT. Nanostructures and nanostructured substrates for surface-enhanced Raman scattering (SERS). J Raman Spectros 2008;39:1313–26.10.1002/jrs.2030Search in Google Scholar

[59] Wu D-Y, Li J-F, Ren B, Tian Z-Q. Electrochemical surface-enhanced Raman spectroscopy of nanostructures. Chem Soc Rev 2008;37:1025–41.10.1039/b707872mSearch in Google Scholar

[60] Vo-Dinh T. Surface-enhanced Raman spectroscopy using metallic nanostructures. TrAC, T Anal Chem 1998;17:557–82.10.1016/S0165-9936(98)00069-7Search in Google Scholar

[61] Vo Dinh T, Stokes DL. SERS-based Raman Probes. Handbook of Vibrational Spectroscopy (Wiley & Sons, Chichester) 2002;2:1302–17.Search in Google Scholar

[62] Kneipp K, Wang Y, Kneipp H, Perelman LT, Itzkan I, Dasari RR, Feld MS. Single molecule detection using surface-enhanced Raman scattering (SERS). Phys Rev Lett 1997;78:1667–70.10.1103/PhysRevLett.78.1667Search in Google Scholar

[63] Kneipp K, Kneipp H, Itzkan I, Dasari RR, Feld MS. Single molecule detection using near infrared surface-enhanced Raman scattering. Springer Ser Chem Phys 2001;67:144–60.10.1007/978-3-642-56544-1_8Search in Google Scholar

[64] Barhoumi A, Halas NJ. Label-free detection of dna hybridization using surface enhanced Raman spectroscopy. J Am Chem Soc 2010;132:12792–3.10.1021/ja105678zSearch in Google Scholar PubMed

[65] Papadopoulou E, Bell SEJ. Structure of adenine on metal nanoparticles: pH equilibria and formation of Ag+ complexes detected by surface-enhanced Raman spectroscopy. J Phys Chem C 2010;114:22644–51.10.1021/jp1092256Search in Google Scholar

[66] Feng F, Zhi G, Jia HS, Cheng L, Tian YT, Li XJ. SERS detection of low-concentration adenine by a patterned silver structure immersion plated on a silicon nanoporous pillar array. Nanotechnology 2009;20: Article no. 295501, 6 pages.10.1088/0957-4484/20/29/295501Search in Google Scholar PubMed

[67] Kundu J, Neumann O, Janesko BG, Zhang D, Lal S, Barhoumi A, Scuseria G, Halas NJ. Adenine- and Adenosine Monophosphate (AMP)-gold binding interactions studied by surface-enhanced Raman and infrared spectroscopies. J Phys Chem C 2009;113:14390–7.10.1021/jp903126fSearch in Google Scholar

[68] Carrillo-Carrion C, Armenta S, Simonet BM, Valcarcel M, Lendl B. Determination of pyrimidine and purine bases by reversed-phase capillary liquid chromatography with at-line surface-enhanced raman spectroscopic detection employing a novel sers substrate based on zns/cdse silver-quantum dots. Anal Chem 2011;83:9391–8.10.1021/ac201821qSearch in Google Scholar PubMed

[69] Primera-Pedrozo OM, Rodriguez GDM, Castellanos J, Felix-Rivera H, Resto O, Hernández-Rivera SP. Increasing surface enhanced Raman spectroscopy effect of RNA and DNA components by changing the pH of silver colloidal suspensions. SpectrochimActa A 2012;87:77–85.10.1016/j.saa.2011.11.012Search in Google Scholar PubMed

[70] Papadopoulou E, Bell SEJ. Surface-enhanced Raman evidence of protonation, reorientation, and Ag+ complexation of Deoxyadenosine and Deoxyadenosine-5 ′-Monophosphate (dAMP) on Ag and Au surfaces. J Phys Chem C 2011;115:14228–35.10.1021/jp204369fSearch in Google Scholar

[71] Muntean CM, Leopold N, Halmagyi A, Valimareanu S. Surface-enhanced Raman spectroscopy of DNA from leaves of in vitro grown apple plants. J Raman Spectros 2011;42:844–50.10.1002/jrs.2780Search in Google Scholar

[72] Barhoumi A, Zhang D, Halas NJ. Correlation of molecular orientation and packing density in a dsDNA self-assembled monolayer observable with surface-enhanced-Raman spectroscopy. J Am Chem Soc 2008;130:14040–1.10.1021/ja804367cSearch in Google Scholar PubMed

[73] Rusciano G, De Luca AC, Pesce G, Sasso A, Oliviero G, Amato J, Borbone N, D′Errico S, Piccialli V, Piccialli G, Mayol L. Label-free probing of G-quadruplex formation by surface-enhanced Raman scattering. Anal Chem 2011;83:6849–55.10.1021/ac201783hSearch in Google Scholar PubMed

[74] Muniz-Miranda M, Gellini C, Pagliai M, Innocenti M, Salvi PR, Schettino V. SERS and computational studies on MicroRNA chains adsorbed on silver surfaces. J Phys Chem C 2010;114:13730–5.10.1021/jp103304rSearch in Google Scholar

[75] Percot A, Lecomte S, Vergne J, Maurel M-C. Hairpin ribozyme catalysis: a surface-enhanced Raman spectroscopy study. Biopolymers 2009;91:384–90.10.1002/bip.21143Search in Google Scholar PubMed

[76] Miljanic S, Dijanosic A, Piantanida I, Meic Z, Albelda MT, Sornosa-Ten A, García-Espana E. Surface-enhanced Raman study of the interactions between tripodal cationic polyamines and polynucleotides. Analyst 2011;136:3185–93.10.1039/c0an01035aSearch in Google Scholar PubMed

[77] Luo XL, Buckhout-White S, Bentley WE, Rubloff GW. Biofabrication of chitosan-silver composite SERS substrates enabling quantification of adenine by a spectroscopic shift. Biofabrication 2011;3: Article no. 034108, 9 pages.10.1088/1758-5082/3/3/034108Search in Google Scholar PubMed

[78] Yin P-G, Jiang L, Lang X-F, Guo L, Yang S. Quantitative analysis of mononucleotides by isotopic labeling surface-enhanced Raman scattering spectroscopy. Biosens Bioelectron 2011;26:4828–31.10.1016/j.bios.2011.05.042Search in Google Scholar PubMed

[79] Chen J-W, Liu X-P, Feng K-J, Liang Y, Jiang JH, Shen GL, Yu RQ. Detection of adenosine using surface-enhanced Raman scattering based on structure-switching signaling aptamer. Biosens Bioelectron 2008;24:66–71.10.1016/j.bios.2008.03.013Search in Google Scholar PubMed

[80] Kim NH, Lee SJ, Moskovits M. Aptamer-mediated surface-enhanced Raman spectroscopy intensity amplification. Nano Letters 2010;10:4181–5.10.1021/nl102495jSearch in Google Scholar PubMed

[81] Li M, Zhang J, Suri S, Sooter LJ, Ma D, Wu N. Detection of adenosine triphosphate with an aptamer biosensor based on surface-enhanced Raman scattering. Anal Chem 2012;84:2837–42.10.1021/ac203325zSearch in Google Scholar PubMed

[82] Ye S, Xiao J, Guo Y, Zhang S. Aptamer-based SERS assay of ATP and lysozyme by using primer self-generation. Chemistry (Weinheim an der Bergstrasse, Germany) 2013;19:8111–6.10.1002/chem.201300126Search in Google Scholar PubMed

[83] Li M, Zhang J, Suri S, Sooter LJ, Ma D, Wu N. Quantitative label-free RNA detection using surface-enhanced Raman spectroscopy. Chem Commun 2011;47:7425–7.10.1039/c1cc11925gSearch in Google Scholar PubMed

[84] Rao S, Raj S, Balint S, Bardina Fons C, Campoy S, Llagostera M, Petrov D. Single DNA molecule detection in an optical trap using surface-enhanced Raman scattering. Appl Phys Lett 2010;96:213701.10.1063/1.3431628Search in Google Scholar

[85] Liu R, Zhu S, Si M, Liu Z, Zhang D. Surface-enhanced Raman scattering-based approach for DNA detection at low concentrations via polyvinyl alcohol-protected silver grasslike patterns. J Raman Spectros 2012;43:370–9.10.1002/jrs.3040Search in Google Scholar

[86] Yuan W, Ho HP, Lee RKY, Kong SK. Surface-enhanced Raman scattering biosensor for DNA detection on nanoparticle island substrates. Appl Optics 2009;48:4329–37.10.1364/AO.48.004329Search in Google Scholar PubMed

[87] Fang C, Agarwal A, Buddharaju KD. Khalid NM, Salim SM, Widjaja E, Garland MV, Balasubramanian N, Kwong DL. DNA detection using nanostructured SERS substrates with Rhodamine B as Raman label. Biosens Bioelectron 2008;24:216–21.10.1016/j.bios.2008.03.032Search in Google Scholar PubMed

[88] Strelau KK, Kretschmer R, Moller R, Fritzsche W, Popp J. SERS as tool for the analysis of DNA-chips in a microfluidic platform. Anal Bioanal Chem 2010;396:1381–4.10.1007/s00216-009-3374-8Search in Google Scholar PubMed

[89] Papadopoulou E, Bell SEJ. Label-free detection of single-base mismatches in DNA by surface-enhanced Raman spectroscopy. Angew Chem-Int Edit 2011;50:9058–61.10.1002/anie.201102776Search in Google Scholar PubMed PubMed Central

[90] Li J-M, Ma W-F, You L-J, Guo J, Hu J, Wang CC. Highly sensitive detection of target ssDNA based on SERS liquid chip using suspended magnetic nanospheres as capturing substrates. Langmuir 2013;29:6147–55.10.1021/la4006828Search in Google Scholar PubMed

[91] Zhang Z, Wen Y, Ma Y, Luo J, Jiang L, Song Y. Mixed DNA-functionalized nanoparticle probes for surface-enhanced Raman scattering-based multiplex DNA detection. Chem Commun 2011;47:7407–9.10.1039/c1cc11062dSearch in Google Scholar PubMed

[92] Thompson DG, Faulds K, Smith WE, Graham D. Precise control of the assembly of dye-coded oligonucleotide silver nanoparticle conjugates with single base mismatch discrimination using surface enhanced resonance Raman scattering. J Phys Chem C 2010;114:7384–9.10.1021/jp909639bSearch in Google Scholar

[93] Thuy NTB, Yokogawa R, Yoshimura Y, Fujimoto K, Koyano M, Maenosono S. Surface-enhanced Raman spectroscopy for facile DNA detection using gold nanoparticle aggregates formed via photoligation. Analyst 2010;135:595–602.10.1039/b919969aSearch in Google Scholar PubMed

[94] He Y, Su S, Xu T, Zhong Y, Zapien JA, Li J, Fan C, Lee S.-T. Silicon nanowires-based highly-efficient SERS-active platform for ultrasensitive DNA detection. Nano Today 2011;6:122–30.10.1016/j.nantod.2011.02.004Search in Google Scholar

[95] Sun Y-H, Kong R-M, Lu D-Q, Zhang X-B, Meng HM, Tan M, Shen GL, Yu RQ. A nanoscale DNA-Au dendrimer as a signal amplifier for the universal design of functional DNA-based SERS biosensors. Chem Commun 2011;47:3840–2.10.1039/c0cc05133kSearch in Google Scholar PubMed

[96] Panikkanvalappil SR, Mackey MA, El-Sayed MA. Probing the unique dehydration-induced structural modifications in cancer cell dna using surface enhanced raman spectroscopy. J Am Chem Soc 2013;135:4815–21.10.1021/ja400187bSearch in Google Scholar PubMed

[97] Hu J, Zhang C-y. Sensitive detection of nucleic acids with rolling circle amplification and surface-enhanced raman scattering spectroscopy. Anal Chem 2010;82:8991–7.10.1021/ac1019599Search in Google Scholar PubMed

[98] Ye S, Yang Y, Xiao J, Zhang S. Surface-enhanced Raman scattering assay combined with autonomous DNA machine for detection of specific DNA and cancer cells. Chem Commun 2012;48:8535–7.10.1039/c2cc32740fSearch in Google Scholar PubMed

[99] Yan J, Su S, He S, He Y, Zhao B, Wang D, Zhang H, Huang Q, Song S, Fan C. Nano rolling-circle amplification for enhanced SERS hot spots in protein microarray analysis. Anal Chem 2012;84:9139–45.10.1021/ac301809eSearch in Google Scholar PubMed

[100] Hering KK, Moller R, Fritzsche W, Popp J. Microarray-based detection of dye-labeled DNA by SERRS using particles formed by enzymatic silver deposition. ChemPhysChem 2008;9:867–72.10.1002/cphc.200700591Search in Google Scholar PubMed

[101] Diaz Fleming G, Finnerty JJ, Campos-Vallette M, Celis F, Aliaga AE, Fredes C, Koch R. Experimental and theoretical Raman and surface-enhanced Raman scattering study of cysteine. J Raman Spectros 2009;40:632–8.10.1002/jrs.2175Search in Google Scholar

[102] Chuang C-H, Chen Y-T. Raman scattering of L-tryptophan enhanced by surface plasmon of silver nanoparticles: vibrational assignment and structural determination. J Raman Spectros 2009;40:150–6.10.1002/jrs.2097Search in Google Scholar

[103] Aliaga AE, Osorio-Roman I, Leyton P, Garrido C, Carcamo J, Caniulef C, Celis F, Diaz F, Clavijo E, Gomez-Jeria JS, Campos-Vallete MM. Surface-enhanced Raman scattering study of L-tryptophan. J Raman Spectros 2009;40:164–9.10.1002/jrs.2099Search in Google Scholar

[104] Aliaga AE, Osorio-Roman I, Garrido C, Leyton P, Cárcamo J, Clavijo E, Gómez-Jeria JS, Díaz F, Campos-Vallette MM. Surface enhanced Raman scattering study of L-lysine. Vib Spectros 2009;50:131–5.10.1016/j.vibspec.2008.09.018Search in Google Scholar

[105] Aliaga AE, Garrido C, Leyton P, Diaz FG, Gomez-Jeria JS, Aguayo T, Clavijo E, Campos-Vallette MM, Sanchez-Cortes S. SERS and theoretical studies of arginine. Spectrochim Acta A 2010;76:458–63.10.1016/j.saa.2010.01.007Search in Google Scholar PubMed

[106] Yang H, Zhu X, Song W, Sun Y, Duan G, Zhao X, Zhang Z. N-acetylalanine monolayers at the silver surface investigated by surface enhanced Raman scattering spectroscopy and X-ray photoelectron spectroscopy: effect of metallic ions. J Phys Chem C 2008;112:15022–7.10.1021/jp8042544Search in Google Scholar

[107] Sheng C, Zhao H, Gu F, Yang H. Effect of Pb2+ on L-glutathione monolayers on a silver surface investigated by surface-enhanced Raman scattering spectroscopy. J Raman Spectros 2009;40:1274–8.10.1002/jrs.2277Search in Google Scholar

[108] Graff M, Bukowska J. Surface-enhanced Raman scattering (SERS) spectroscopy of enantiomeric and racemic methionine on a silver electrode-evidence for chiral discrimination in interactions between adsorbed molecules. Chem Phys Lett 2011;509:58–61.10.1016/j.cplett.2011.04.089Search in Google Scholar

[109] Graff M, Bukowska J. Enantiomeric recognition of phenylalanine by self-assembled monolayers of cysteine: Surface enhanced Raman scattering evidence. Vib Spectros 2010;52:103–7.10.1016/j.vibspec.2009.11.003Search in Google Scholar

[110] Thomas S, Biswas N, Malkar VV, Mukherjee T, Kapoor S. Studies on adsorption of carnosine on silver nanoparticles by SERS. Chem Phys Lett 2010;491:59–64.10.1016/j.cplett.2010.03.059Search in Google Scholar

[111] Podstawka E, Andrzejak M, Kafarski P, Proniewicz LM. Comparison of adsorption mechanism on colloidal silver surface of alafosfalin and its analogs. J Raman Spectros 2008;39:1238–49.10.1002/jrs.1977Search in Google Scholar

[112] Podstawka E, Kafarski P, Proniewicz LM. Effect of an aliphatic spacer group on the adsorption mechanism of phosphonodipeptides containing N-terminal glycine on the colloidal silver surface. J Raman Spectros 2008;39:1396–407.10.1002/jrs.2010Search in Google Scholar

[113] Malek K, Makowski M, Krolikowska A, Bukowska J. Comparative studies on IR, Raman, and surface enhanced Raman scattering spectroscopy of dipeptides containing Delta Ala and Delta Phe. J Phys Chem B 2012;116:1414–25.10.1021/jp208586jSearch in Google Scholar PubMed

[114] Yuan X, Gu H, Wu J. Surface-enhanced Raman spectrum of Gly-Gly adsorbed on the silver colloidal surface. J Mol Struct 2010;977:56–61.10.1016/j.molstruc.2010.05.009Search in Google Scholar

[115] Wei F, Zhang D, Halas NJ, Hartgerink JD. Aromatic amino acids providing characteristic motifs in the Raman and SERS spectroscopy of peptides. J Phys Chem B 2008;112:9158–64.10.1021/jp8025732Search in Google Scholar PubMed

[116] Podstawka E, Kafarski P, Proniewicz LM. Structural properties of L-X-L-Met-L-Ala phosphonate tripeptides: a combined FT-IR, FT-RS, and SERS spectroscopy studies and DFT calculations. J Phys Chem A 2008;112:11744–55.10.1021/jp803674qSearch in Google Scholar PubMed

[117] Podstawka E. Effect of amino acid modifications on the molecular structure of adsorbed and nonadsorbed bombesin 6-14 fragments on an electrochemically roughened silver surface. J Raman Spectros 2008;39:1290–1305.10.1002/jrs.1996Search in Google Scholar

[118] Podstawka E, Niaura G, Proniewicz LM. Potential-dependent studies on the interaction between phenylalanine-substituted bombesin fragments and roughened Ag, Au, and Cu electrode surfaces. J Phys Chem B 2010;114:1010–29.10.1021/jp909268cSearch in Google Scholar PubMed

[119] Podstawka E, Ozaki Y, Proniewicz LM. Structures and bonding on a colloidal silver surface of the various length carboxyl terminal fragments of bombesin. Langmuir 2008;24:10807–16.10.1021/la8012415Search in Google Scholar PubMed

[120] Podstawka-Proniewicz E, Ozaki Y, Kim Y, Xu Y, Proniewicz LM. Surface-enhanced Raman scattering studies on bombesin, its selected fragments and related peptides adsorbed at the silver colloidal surface. Appl Surf Sci 2011;257:8246–52.10.1016/j.apsusc.2011.02.012Search in Google Scholar

[121] Podstawka E, Proniewicz LM. The orientation of BN-related peptides adsorbed on SERS-active silver nanoparticles: comparison with a silver electrode surface. J Phys Chem B 2009;113:4978–85.10.1021/jp8110716Search in Google Scholar PubMed

[122] Podstawka-Proniewicz E, Kudelski A, Kim Y, Proniewicz LM. Structure and binding of specifically mutated neurotensin fragments on a silver substrate: vibrational studies. J Phys Chem B 2011;115:7097–108.10.1021/jp201316nSearch in Google Scholar PubMed

[123] Podstawka-Proniewicz E, Ignatjev I, Niaura G, Proniewicz LM. Phe-MetNH(2) terminal bombesin subfamily peptides: potential induced changes in adsorption on Ag, Au, and Cu electrodes monitored by SERS. J Phys Chem C 2012;116:4189–200.10.1021/jp2126027Search in Google Scholar

[124] Ignatjev I, Podstawka-Proniewicz E, Niaura G, Lombardi JR, Proniewicz LM. Potential induced changes in neuromedin B adsorption on Ag, Au, and Cu electrodes monitored by surface-enhanced Raman scattering. J Phys Chem B 2011;115:10525–36.10.1021/jp2026863Search in Google Scholar PubMed

[125] Aliaga AE, Aguayo T, Garrido C, Clavijo E, Hevia E, Gómez-Jeria JS, Leyton P, Campos-Vallette MM, Sanchez-Cortes S. Surface-enhanced Raman scattering and theoretical studies of the C-terminal peptide of the beta-subunit human chorionic gonadotropin without linked carbohydrates. Biopolymers 2011;95:135–43.10.1002/bip.21542Search in Google Scholar PubMed

[126] Garrido C, Aliaga AE, Gomez-Jeria JS, Clavijo RE, Campos-Vallette MM, Sanchez-Cortes S. Adsorption of oligopeptides on silver nanoparticles: surface-enhanced Raman scattering and theoretical studies. J Raman Spectros 2010;41:1149–55.10.1002/jrs.2583Search in Google Scholar

[127] Aliaga AE, Ahumada H, Sepulveda K, Gomez-Jeria JS, Garrido C, Weiss-Lopez BE, Campos-Vallette MM. SERS, molecular dynamics and molecular orbital studies of the MRKDV peptide on silver and membrane surfaces. J Phys Chem C 2011;115:3982–9.10.1021/jp1107153Search in Google Scholar

[128] Podstawka-Proniewicz E, Kosior M, Kim Y, Rolka K, Proniewicz LM. Nociceptin and Its natural and specifically-modified fragments: structural studies. Biopolymers 2010;93:1039–54.10.1002/bip.21516Search in Google Scholar PubMed

[129] Iosin M, Toderas F, Baldeck PL, Astilean S. Study of protein-gold nanoparticle conjugates by fluorescence and surface-enhanced Raman scattering. J Mole Struct 2009;924–26:196–200.10.1016/j.molstruc.2009.02.004Search in Google Scholar

[130] Das R, Jagannathan R, Sharan C, Kumar U, Poddar P. Mechanistic study of surface functionalization of enzyme lysozyme synthesized Ag and Au nanoparticles using surface enhanced Raman spectroscopy. J Phys Chem C 2009;113:21493–500.10.1021/jp905806tSearch in Google Scholar

[131] Chandra G, Ghosh KS, Dasgupta S, Roy A. Evidence of conformational changes in adsorbed lysozyme molecule on silver colloids. Int J Biol Macromol 2010;47:361–5.10.1016/j.ijbiomac.2010.05.020Search in Google Scholar PubMed

[132] Sengupta A, Thai CK, Sastry MSR, Matthaei JF, Schwartz DT, Davis EJ, Baneyx F. A genetic approach for controlling the binding and orientation of proteins on nanoparticles. Langmuir 2008;24:2000–8.10.1021/la702079eSearch in Google Scholar PubMed

[133] Kumar GVP, Selvi R, Kishore AH, KunduTK, Narayana C. Surface-enhanced Raman, spectroscopic studies of coactivator-associated arginine methyltransferase 1. J Phys Chem B 2008;112:6703–7.10.1021/jp711594zSearch in Google Scholar PubMed

[134] Kudelski A. In situ SERS studies on the adsorption of tyrosinase on bare and alkanethiol-modified silver substrates. Vib Spectros 2008;46:34–8.10.1016/j.vibspec.2007.08.005Search in Google Scholar

[135] Li D, Li D-W, Fossey JS, Long Y-T. In situ surface-enhanced Raman scattering and X-ray photoelectron spectroscopic investigation of coenzyme Q(10) on silver electrode. Phys Chem Chem Phys 2011;13:2259–65.10.1039/C0CP01449DSearch in Google Scholar

[136] Kaminska A, Forster RJ, Keyes TE. The impact of adsorption of bovine pancreatic trypsin inhibitor on CTAB-protected gold nanoparticle arrays: a Raman spectroscopic comparison with solution denaturation. J Raman Spectros 2010;41:130–5.Search in Google Scholar

[137] Krolikowska A, Bukowska J. Surface-enhanced resonance Raman spectroscopic characterization of cytochrome c immobilized on 2-mercaptoethanesulfonate monolayers on silver. J Raman Spectros 2010;41:1621–31.10.1002/jrs.2618Search in Google Scholar

[138] Papazoglou ES, Babu S, Hansberry DR, Mohapatra S, Patel C. SERS study on myeloperoxidase and its immunocomplex: Identification of binding interactions. Spectros Int J 2010;24:183–90.10.1155/2010/169292Search in Google Scholar

[139] Choi I, Huh YS, Erickson D. Ultra-sensitive, label-free probing of the conformational characteristics of amyloid beta aggregates with a SERS active nanofluidic device. Microfluidics and Nanofluidics 2012;12:663–9.10.1007/s10404-011-0879-1Search in Google Scholar

[140] Abdali S, De Laere B, Poulsen M, Grigorian M, Lukanidin E, Klingelhöfer J. Toward methodology for detection of cancer-promoting S100A4 protein conformations in subnanomolar concentrations using Raman and SERS. J Phys Chem C 2010;114:7274–9.10.1021/jp908335zSearch in Google Scholar

[141] Singhal K, Kalkan AK. Surface-enhanced raman scattering captures conformational changes of single photoactive yellow protein molecules under photoexcitation. J Am Chem Soc 2010;132:429–31.10.1021/ja9028704Search in Google Scholar PubMed

[142] Mueller J, Becher T, Braunstein J, Berdel P, Gravius S, Rohrbach F, Oldenburg J, Mayer G, Pötzsch B. Profiling of active thrombin in human blood by supramolecular complexes. Angew Chem-Int Edit 2011;50:6075–8.10.1002/anie.201007032Search in Google Scholar PubMed

[143] Nierodzik ML, Karpatkin S. Thrombin induces tumor growth, metastasis, and angiogenesis: Evidence for a thrombin-regulated dormant tumor phenotype. Cancer Cell 2006;10:355–62.10.1016/j.ccr.2006.10.002Search in Google Scholar PubMed

[144] Song K-M, Lee S, Ban C. Aptamers and their biological applications. Sensors 2012;12:612–31.10.3390/s120100612Search in Google Scholar PubMed PubMed Central

[145] Wu Z, Liu Y, Zhou X, Shen A, Hu J. A "turn-off′ SERS-based detection platform for ultrasensitive detection of thrombin based on, enzymatic assays. Biosens Bioelectron 2013;44:10–15.10.1016/j.bios.2013.01.006Search in Google Scholar PubMed

[146] Ochsenkuehn MA, Campbell CJ. Probing biomolecular interactions using surface enhanced Raman spectroscopy: label-free protein detection using a G-quadruplex DNA aptamer. Chem Commun 2010;46:2799–801.10.1039/b920941gSearch in Google Scholar PubMed

[147] Pagba CV, Lane SM, Cho H, Wachsmann-Hogiu S. Direct detection of aptamer-thrombin binding via surface-enhanced Raman spectroscopy. J Biomed Optics 2010;15.10.1117/1.3465594Search in Google Scholar PubMed

[148] Fabris L, Dante M, Nguyen TQ, Tok JBH, Bazan GC. SERS aptatags: New responsive metallic nanostructures for heterogeneous protein detection by surface enhanced Raman spectroscopy. Adv Funct Mater 2008;18:2518–25.10.1002/adfm.200800301Search in Google Scholar

[149] Hu J, Zheng P-C, Jiang J-H, Shen G-L, Yu RQ, Liu GK. Electrostatic interaction based approach to thrombin detection by surface-enhanced Raman spectroscopy. Anal Chem 2009;81:87–93.10.1021/ac801431mSearch in Google Scholar PubMed

[150] Wang YL, Lee K, Irudayaraj J. SERS aptasensor from nanorod-nanoparticle junction for protein detection. Chem Commun 2010;46:613–5.10.1039/B919607BSearch in Google Scholar PubMed

[151] Yoon J, Choi N, Ko J, Kim K, Lee S, Choo J. Highly sensitive detection of thrombin using SERS-based magnetic aptasensors. Biosens Bioelectron 2013;47:62–7.10.1016/j.bios.2013.03.003Search in Google Scholar PubMed

[152] Kennedy DC, Hoop KA, Tay L-L, Pezacki JP. Development of nanoparticle probes for multiplex SERS imaging of cell surface proteins. Nanoscale 2010;2:1413–6.10.1039/c0nr00122hSearch in Google Scholar PubMed

[153] Woo MA, Lee S-M, Kim G, Baek J, Noh MS, Kim JE, Park SJ, Minai-Tehrani A, Park SC, Seo YT, Kim YK, Lee YS, Jeong DH, Cho MH. Multiplex immunoassay using fluorescent-surface enhanced Raman spectroscopic dots for the detection of bronchioalveolar stem Cells in murine lung. Anal Chem 2009;81:1008–15.10.1021/ac802037xSearch in Google Scholar PubMed

[154] Hodges MD, Kelly JG, Bentley AJ, Fogarty S, Patel II, Martin FL, Fullwood NJ. Combining immunolabeling and surface-enhanced Raman spectroscopy on cell membranes. ACS Nano 2011;5:9535–41.10.1021/nn202652hSearch in Google Scholar PubMed

[155] Sujith A, Itoh T, Abe H, Yoshida K-i, Kiran MS, Biju V, Ishikawa M. Imaging the cell wall of living single yeast cells using surface-enhanced Raman spectroscopy. Anal Bioanal Chem 2009;394:1803–9.10.1007/s00216-009-2883-9Search in Google Scholar PubMed

[156] Hollywood KA, Shadi IT, Goodacre R. Monitoring the succinate dehydrogenase activity isolated from mitochondria by surface enhanced Raman scattering. J Phys Chem C 2010;114: 7308–13.10.1021/jp908950xSearch in Google Scholar

[157] Yazgan NN, Boyaci IH, Temur E, Tamer U, Topcu A. A high sensitive assay platform based on surface-enhanced Raman scattering for quantification of protease activity. Talanta 2010;82:631–9.10.1016/j.talanta.2010.05.023Search in Google Scholar PubMed

[158] Li T, Liu D, Wang Z. Microarray-based Raman spectroscopic assay for kinase inhibition by gold nanoparticle probes. Biosens Bioelectron 2009;24:3335–9.10.1016/j.bios.2009.04.033Search in Google Scholar PubMed

[159] Stevenson R, Stokes RJ, MacMillan D, Armstrong D, Faulds K, Wadsworth R, Kunuthur S, Suckling CJ, Graham D. In situ detection of pterins by SERS. Analyst 2009;134:1561–4.10.1039/b905562bSearch in Google Scholar PubMed

[160] He P, Zhang Y, Liu L, Qiao W, Zhang S. Ultrasensitive SERS detection of lysozyme by a target-triggering multiple cycle amplification strategy based on a gold substrate. Chemistry (Weinheim an der Bergstrasse, Germany) 2013;19:7452–60.10.1002/chem.201203224Search in Google Scholar PubMed

[161] Joshi B, Chakrabarty A, Bruot C, Ainsworth H, Fraizer G, Wei QH. DNA-WT1 protein interaction studied by surface-enhanced Raman spectroscopy. Anal Bioanal Chem 2010;396:1415–21.10.1007/s00216-009-3364-xSearch in Google Scholar PubMed

[162] Han XX, Kitahama Y, Tanaka Y, Guo J, Xu WQ, Zhao B, Ozaki Y. Simplified protocol for detection of protein-ligand interactions via surface-enhanced resonance Raman scattering and surface-enhanced fluorescence. Anal Chem 2008;80:6567–72.10.1021/ac800642gSearch in Google Scholar PubMed

[163] Chen L, Hong W, Guo Z, Sa Y, Wang X, Jung YM, Zhao B. Magnetic assistance highly sensitive protein assay based on surface-enhanced resonance Raman scattering. J Colloid Interf Sci 2012;368:282–6.10.1016/j.jcis.2011.10.069Search in Google Scholar PubMed

[164] Levin CS, Kundu J, Janesko BG, Scuseria GE, Raphael RM, Halas NJ. Interactions of Ibuprofen with hybrid lipid bilayers probed by complementary surface-enhanced vibrational spectroscopies. J Phys Chem B 2008;112:14168–75.10.1021/jp804374eSearch in Google Scholar PubMed PubMed Central

[165] Lajos G, Jancura D, Miskovsky P, Garcia-Ramos JV, Sanchez-Cortes S. Interaction of the photosensitizer hypericin with low-density lipoproteins and phosphatidylcholine: a surface-enhanced raman scattering and surface-enhanced fluorescence study. J Phys Chem C 2009;113:7147–54.10.1021/jp8112528Search in Google Scholar

[166] Premasiri WR, Sauer-Budge AF, Lee JC, Klapperich CM, Ziegler LD. Rapid bacterial diagnostics via surface-enhanced Raman microscopy. Spectroscopy - Special Issues 2012;27:s8–s21.Search in Google Scholar

[167] O’Brien JM, Jr, Ali NA, Aberegg SK, Abraham E. Sepsis. Am J Med 2007;120:1012–22.10.1016/j.amjmed.2007.01.035Search in Google Scholar PubMed

[168] Brumbaugh AR, Mobley HLT. Preventing urinary tract infection: progress toward an effective Escherichia coli vaccine. Expert Rev Vaccines 2012;11:663–76.10.1586/erv.12.36Search in Google Scholar PubMed PubMed Central

[169] Wertheim H, Verbrugh HA, van Pelt C, de Man P, van Belkum A, Vos MC. Improved detection of methicillin-resistant Staphylococcus aureus using phenyl mannitol broth containing aztreonam and ceftizoxime. J Clin Microbiol 2001;39:2660–2.10.1128/JCM.39.7.2660-2662.2001Search in Google Scholar PubMed PubMed Central

[170] Dougan JA, MacRae D, Graham D, Faulds K. DNA detection using enzymatic signal production and SERS. Chem Commun 2011;47:4649–51.10.1039/c0cc05216gSearch in Google Scholar PubMed

[171] Kujau MJ, Wolfl S. Efficient preparation of single-stranded DNA for in vitro selection. Mole Biotechnol 1997;7:333–5.10.1007/BF02740823Search in Google Scholar PubMed

[172] Graham D, Stevenson R, Thompson DG, Barrett L, Dalton C, Faulds K. Combining functionalised nanoparticles and SERS for the detection of DNA relating to disease. Faraday Discuss 2011;149:291–9.10.1039/C005397JSearch in Google Scholar

[173] Faulds K, Jarvis R, Smith WE, Graham D, Goodacre R. Multiplexed detection of six labelled oligonucleotides using surface enhanced resonance Raman scattering (SERRS). Analyst 2008;133:1505–12.10.1039/b800506kSearch in Google Scholar PubMed

[174] Papadopoulou E, Bell SEJ. Label-Free detection of nanomolar unmodified single- and double-stranded DNA by using surface-enhanced Raman spectroscopy on Ag and Au colloids. Chem Eur J 2012;18:5394–400.10.1002/chem.201103520Search in Google Scholar PubMed

[175] Kang T, Yoo SM, Yoon I, Lee SY, Kim B. Patterned multiplex pathogen DNA detection by Au particle-on-wire SERS sensor. Nano Letters 2010;10:1189–93.10.1021/nl1000086Search in Google Scholar PubMed

[176] MacAskill A, Crawford D, Graham D, Faulds K. DNA sequence detection using surface-enhanced resonance Raman spectroscopy in a homogeneous multiplexed assay. Anal Chem 2009;81:8134–40.10.1021/ac901361bSearch in Google Scholar PubMed

[177] van Lierop D, Faulds K, Graham D. Separation free DNA detection using surface enhanced Raman scattering. Anal Chem 2011;83:5817–21.10.1021/ac200514eSearch in Google Scholar PubMed

[178] van Lierop D, Larmour IA, Faulds K, Graham D. SERS primers and their mode of action for pathogen DNA detection. Anal Chem 2013;85:1408–14.10.1021/ac302254hSearch in Google Scholar PubMed

[179] Harper MM, Robertson B, Ricketts A, Faulds K. Specific detection of DNA through coupling of a TaqMan assay with surface enhanced Raman scattering (SERS). Chem Commun 2012;48:9412–14.10.1039/c2cc34859dSearch in Google Scholar PubMed

[180] Strelau KK, Brinker A, Schnee C, Weber K, Möller R, Popp J. Detection of PCR products amplified from DNA of epizootic pathogens using magnetic nanoparticles and SERS. J Raman Spectros 2011;42:243–50.10.1002/jrs.2730Search in Google Scholar

[181] Osorio-Roman IO, Aroca RF, Astudillo J, Matsuhiro B, Vásquez C, Pérez JM. Characterization of bacteria using its O-antigen with surface-enhanced Raman scattering. Analyst 2010;135:1997–2001.10.1039/c0an00061bSearch in Google Scholar PubMed

[182] Yang X, Gu C, Qian F, Li Y, Zhang JZ. Highly sensitive detection of proteins and bacteria in aqueous solution using surface-enhanced Raman scattering and optical fibers. Anal Chem 2011;83:5888–94.10.1021/ac200707tSearch in Google Scholar PubMed

[183] Arruebo M, Valladares M, Gonzalez-Fernandez A. Antibody-conjugated nanoparticles for biomedical applications. J Nanomaterials 2009;2009: Article ID 439389, 24 pages (doi: 10.1155/2009/439389).Search in Google Scholar

[184] Ravindranath SP, Wang Y, Irudayaraj J. SERS driven cross-platform based multiplex pathogen detection. Sensor Actuat B-Chem 2011;152:183–90.10.1016/j.snb.2010.12.005Search in Google Scholar

[185] Lin C-C, Yang Y-M, Chen Y-F, Yang T-S, Chang H-C. A new protein A assay based on Raman reporter labeled immunogold nanoparticles. Biosens Bioelectron 2008;24:178–83.10.1016/j.bios.2008.03.035Search in Google Scholar PubMed

[186] Yakes BJ, Lipert RJ, Bannantine JP, Porter MD. Detection of Mycobacterium avium subsp paratuberculosis by a sonicate immunoassay based on surface-enhanced Raman scattering. Clin Vaccine Immunol 2008;15:227–34.10.1128/CVI.00334-07Search in Google Scholar PubMed PubMed Central

[187] Colpitts TM, Conway MJ, Montgomery RR, Fikrig E. West nile virus: biology, transmission, and human infection. Clin microbiol rev 2012;25:635–48.10.1128/CMR.00045-12Search in Google Scholar PubMed PubMed Central

[188] Zhang H, Harpster MH, Park HJ, Johnson PA. Surface-enhanced Raman scattering detection of DNA derived from the west nile virus genome using magnetic capture of Raman-active gold nanoparticles. Anal Chem 2011;83:254–60.10.1021/ac1023843Search in Google Scholar PubMed

[189] Zhang H, Harpster MH, Wilson WC, Johnson PA. Surface-enhanced Raman scattering detection of DNAs derived from virus genomes using Au-coated paramagnetic nanoparticles. Langmuir 2012;28:4030–7.10.1021/la204890tSearch in Google Scholar PubMed

[190] Neng J, Harpster MH, Wilson WC, Johnson PA. Surface-enhanced Raman scattering (SERS) detection of multiple viral antigens using magnetic capture of SERS-active nanoparticles. Biosens Bioelectron 2013;41:316–21.10.1016/j.bios.2012.08.048Search in Google Scholar PubMed

[191] Tsongalis GJ. Branched DNA technology in molecular diagnostics. Am J Clin Pathol 2006;126:448–53.10.1309/90BU6KDXANFLN4RJSearch in Google Scholar PubMed

[192] Hu J, Zheng P-C, Jiang J-H, Shen G-L, Yu RQ, Liu GK. Sub-attomolar HIV-1 DNA detection using surface-enhanced Raman spectroscopy. Analyst 2010;135:1084–9.10.1039/b920358cSearch in Google Scholar PubMed

[193] Huh YS, Chung AJ, Cordovez B, Erickson D. Enhanced on-chip SERS based biomolecular detection using electrokinetically active microwells. Lab on a Chip 2009;9:433–9.10.1039/B809702JSearch in Google Scholar

[194] Tang KF, Ooi EE. Diagnosis of dengue: an update. Expert Rev Anti-Infective Therapy 2012;10:895–907.10.1586/eri.12.76Search in Google Scholar PubMed

[195] Kumar S, Henrickson KJ. Update on influenza diagnostics: lessons from the novel H1N1 Influenza a pandemic. Clin microbiol rev 2012;25:344–61.10.1128/CMR.05016-11Search in Google Scholar PubMed PubMed Central

[196] Seol M-L, Choi S-J, Baek DJ, Park TJ, Ahn J-H, Lee SY, Choi YK. A nanoforest structure for practical surface-enhanced Raman scattering substrates. Nanotechnology 2012;23: Article no. 095301, 7 pages.10.1088/0957-4484/23/9/095301Search in Google Scholar PubMed

[197] Negri P, Kage A, Nitsche A, Naumann D, Dluhy RA. Detection of viral nucleoprotein binding to anti-influenza aptamers via SERS. Chem Commun 2011;47:8635–7.10.1039/c0cc05433jSearch in Google Scholar PubMed

[198] Marotta NE, Beavers KR, Bottomley LA. Limitations of surface enhanced Raman scattering in sensing DNA hybridization demonstrated by label-free DNA oligos as molecular rulers of distance-dependent enhancement. Anal Chem 2013;85:1440–6.10.1021/ac302454jSearch in Google Scholar PubMed

[199] Chang J. Current progress on development of respiratory syncytial virus vaccine. Bmb Reports 2011;44:232–7.10.5483/BMBRep.2011.44.4.232Search in Google Scholar PubMed

[200] Chen Y, Zheng X, Chen G, He C, Zhu W, Feng S, Xi G, Chen R, Lan F, Zeng H. Immunoassay for LMP1 in nasopharyngeal tissue based on surface-enhanced Raman scattering. Int J Nanomed 2012;7:73–82.Search in Google Scholar

[201] Lin J, Chen R, Feng S, Pan J, Li B, Chen G, Lin S, Li C, Sun L, Huang Z, Zeng H. Surface-enhanced Raman scattering spectroscopy for potential noninvasive nasopharyngeal cancer detection. J Raman Spectros 2012;43:497–502.10.1002/jrs.3072Search in Google Scholar

[202] 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;7:655–63.10.1016/j.nano.2011.01.012Search in Google Scholar PubMed

[203] Feng SY, Chen R, Lin JQ, Pan JJ, 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;26:3167–74.10.1016/j.bios.2010.12.020Search in Google Scholar PubMed

[204] Pinzani P, Salvianti F, Pazzagli M, Orlando C. Circulating nucleic acids in cancer and pregnancy. Methods 2010;50:302–7.10.1016/j.ymeth.2010.02.004Search in Google Scholar PubMed

[205] Li S-X, Zeng Q-Y, Li L-F, Zhang Y-J, Wan MM, Liu ZM, Xiong HL, Guo ZY, Liu SH. Study of support vector machine and serum surface-enhanced Raman spectroscopy for noninvasive esophageal cancer detection. J Biomed Opt 2013;18:027008.10.1117/1.JBO.18.2.027008Search in Google Scholar PubMed

[206] Lin D, Feng S, Pan J, Chen Y, Lin J, Chen G, Xie S, Zeng H, Chen R. Colorectal cancer detection by gold nanoparticle based surface-enhanced Raman spectroscopy of blood serum and statistical analysis. Opt Express 2011;19:13565–77.10.1364/OE.19.013565Search in Google Scholar PubMed

[207] Feng S, Chen R, Lin J, Pan J, Chen G, Li Y, Cheng M, Huang Z, Chen J, Zeng H. Nasopharyngeal cancer detection based on blood plasma surface-enhanced Raman spectroscopy and multivariate analysis. Biosens Bioelectron 2010;25:2414–9.10.1016/j.bios.2010.03.033Search in Google Scholar PubMed

[208] Feng S, Lin D, Lin J, Li B, Huang Z, Chen G, Zhang W, Wang L, Pan J, Chen R, Zeng H. Blood plasma surface-enhanced Raman spectroscopy for non-invasive optical detection of cervical cancer. Analyst 2013;138:3967–74.10.1039/c3an36890dSearch in Google Scholar PubMed

[209] Gormally E, Caboux E, Vineis P, Hainaut P. Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res-Rev Mutat 2007;635:105–117.10.1016/j.mrrev.2006.11.002Search in Google Scholar PubMed

[210] Chen Y, Chen G, Feng S, Pan J, Zheng X, Su Y, Chen Y, Huang Z, Lin X, Lan F, Chen R, Zeng H. Label-free serum ribonucleic acid analysis for colorectal cancer detection by surface-enhanced Raman spectroscopy and multivariate analysis. J Biomed Opt 2012;17:067003.10.1117/1.JBO.17.6.067003Search in Google Scholar PubMed

[211] Arya SK, Lim B, Rahman ARA. Enrichment, detection and clinical significance of circulating tumor cells. Lab on a Chip 2013;13:1995–2027.10.1039/c3lc00009eSearch in Google Scholar PubMed

[212] Wang X, Qian X, Beitler JJ, Chen ZG, Khuri FR, Lewis MM, Shin HJ, Nie S, Shin DM. Detection of circulating tumor cells in human peripheral blood using surface-enhanced Raman scattering nanoparticles. Cancer Res 2011;71: 1526–32.10.1158/0008-5472.CAN-10-3069Search in Google Scholar PubMed PubMed Central

[213] Sha MY, Xu H, Natan MJ, Cromer R. Surface-enhanced Raman scattering tags for rapid and homogeneous detection of circulating tumor cells in the presence of human whole blood. J Am Chem Soc 2008;130:17214–5.10.1021/ja804494mSearch in Google Scholar PubMed PubMed Central

[214] Lee K, Drachev VP, Irudayaraj J. DNA-Gold nanoparticle reversible networks grown on cell surface marker sites: application in diagnostics. Acs Nano 2011;5:2109–17.10.1021/nn1030862Search in Google Scholar PubMed

[215] MacLaughlin CM, Mullaithilaga N, Yang G, Ip SY, Wang C, Walker GC. Surface-enhanced Raman scattering dye-labeled Au nanoparticles for triplexed detection of leukemia and lymphoma cells and SERS flow cytometry. Langmuir 2013;29:1908–19.10.1021/la303931cSearch in Google Scholar PubMed

[216] Nguyen CT, Nguyen JT, Rutledge S, Zhang J, Wang C, Walker GC. Detection of chronic lymphocytic leukemia cell surface markers using surface enhanced Raman scattering gold nanoparticles. Cancer Lett 2010;292:91–7.10.1016/j.canlet.2009.11.011Search in Google Scholar PubMed

[217] Dougherty U, Sehdev A, Cerda S, Mustafi R, Little N, Yuan W, Jagadeeswaran S, Chumsangsri A, Delgado J, Tretiakova M, Joseph L, Hart J, Cohen EE, Aluri L, Fichera A, Bissonnette M. Epidermal growth factor receptor controls flat dysplastic aberrant crypt foci development and colon cancer progression in the rat azoxymethane model. Clin Cancer Res 2008;14: 2253–62.10.1158/1078-0432.CCR-07-4926Search in Google Scholar PubMed

[218] Qian X, Peng X-H, Ansari DO, Yin-Goen Q, Chen GZ, Shin DM, Yang L, Young AN, Wang MD, Nie S. In vivo tumor targeting and spectroscopic detection with surface-enhanced Raman nanoparticle tags. Nat Biotechnol 2008;26, 83–90.10.1038/nbt1377Search in Google Scholar PubMed

[219] Jokerst JV, Miao Z, Zavaleta C, Cheng Z, Gambhir SS. Affibody-functionalized gold-silica nanoparticles for Raman molecular imaging of the epidermal growth factor receptor. Small 2011;7:625–33.10.1002/smll.201002291Search in Google Scholar PubMed PubMed Central

[220] Dinish US, Fu CY, Soh KS, Bhuvaneswari R, Kumar A, Olivo M. Highly sensitive SERS detection of cancer proteins in low sample volume using hollow core photonic crystal fiber. Biosens Bioelectron 2012;33:293–8.10.1016/j.bios.2011.12.056Search in Google Scholar PubMed

[221] Maiti KK, Samanta A, Vendrell M, Soh K-S, Olivo M, Chang YT. Multiplex cancer cell detection by SERS nanotags with cyanine and triphenylmethine Raman reporters. Chem Commun 2011;47:3514–6.10.1039/c0cc05265eSearch in Google Scholar PubMed

[222] 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;402:1093–100.10.1007/s00216-011-5577-zSearch in Google Scholar PubMed

[223] Lee S, Chon H, Lee M, Choo J, Shin SY, Lee YH, Rhyu IJ, Son SW, Oh CH. Surface-enhanced Raman scattering imaging of HER2 cancer markers overexpressed in single MCF7 cells using antibody conjugated hollow gold nanospheres. Biosens Bioelectron 2009;24:2260–3.10.1016/j.bios.2008.10.018Search in Google Scholar PubMed

[224] Park H, Lee S, Chen L, Lee EK, Shin SY, Lee YH, Son SW, Oh CH, Song JM, Kang SH, Choo J. SERS imaging of HER2-overexpressed MCF7 cells using antibody-conjugated gold nanorods. Phys Chem Chem Phys 2009;11:7444–9.10.1039/b904592aSearch in Google Scholar PubMed

[225] Wang HN, Vo-Dinh T. Multiplex detection of breast cancer biomarkers using plasmonic molecular sentinel nanoprobes. Nanotechnology 2009;20:065101.10.1088/0957-4484/20/6/065101Search in Google Scholar PubMed PubMed Central

[226] Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev 2004;25:581–611.10.1210/er.2003-0027Search in Google Scholar PubMed

[227] Ko J, Lee S, Lee EK, Chang S-I, Chen L, Yoon SY, Choo J. SERS-based immunoassay of tumor marker VEGF using DNA aptamers and silica-encapsulated hollow gold nanospheres. Phys Chem Chem Phys 2013;15:5379–85.10.1039/C2CP43155FSearch in Google Scholar PubMed

[228] Li M, Cushing SK, Zhang J, Suri S, Evans R, Petros WP, Gibson LF, Ma D, Liu Y, Wu N. Three-dimensional hierarchical plasmonic nano-architecture enhanced surface-enhanced Raman scattering immunosensor for cancer biomarker detection in blood plasma. ACS Nano 2013;7:4967–76.10.1021/nn4018284Search in Google Scholar PubMed PubMed Central

[229] Wu P, Gao Y, Zhang H, Cai C. Aptamer-guided silver-gold bimetallic nanostructures with highly active surface-enhanced Raman scattering for specific detection and near-infrared photothermal therapy of human breast cancer cells. Anal Chem 2012;84:7692–9.10.1021/ac3015164Search in Google Scholar PubMed

[230] Yu C, Hu Y, Duan J, Yuan W, Wang C, Xu H, Yang X.-D. Novel aptamer-nanoparticle bioconjugates enhances delivery of anticancer drug to MUC1-positive cancer cells in vitro. Plos One 2011;6:e24077.10.1371/journal.pone.0024077Search in Google Scholar PubMed PubMed Central

[231] Wang G, Lipert RJ, Jain M, Kaur S, Chakraboty S, Torres MP, Batra SK, Brand RE, Porter MD. Detection of the potential pancreatic cancer marker MUC4 in serum using surface-enhanced Raman scattering. Anal Chem 2011;83:2554–61.10.1021/ac102829bSearch in Google Scholar PubMed PubMed Central

[232] Chon H, Lee S, Yoon S-Y, Chang S-I, Lim DW, Choo J. Simultaneous immunoassay for the detection of two lung cancer markers using functionalized SERS nanoprobes. Chem Commun 2011;47:12515–17.10.1039/c1cc15707hSearch in Google Scholar PubMed

[233] Lee M, Lee S, Lee JH, Lim HW, Seong GH, Lee EK, Chang SI, Oh CH, Choo J. Highly reproducible immunoassay of cancer markers on a gold-patterned microarray chip using surface-enhanced Raman scattering imaging. Biosens Bioelectron 2011;26:2135–41.10.1016/j.bios.2010.09.021Search in Google Scholar PubMed

[234] Lee M, Lee K, Kim KH, Oh KW, Choo J. SERS-based immunoassay using a gold array-embedded gradient microfluidic chip. Lab on a Chip 2012;12:3720–7.10.1039/c2lc40353fSearch in Google Scholar PubMed

[235] Srisa-Art M, Kang D-K, Hong J, Park H, Leatherbarrow RJ, Edel JB, Chang SI, deMello AJ. Analysis of protein-protein interactions by using droplet-based microfluidics. Chembiochem 2009;10:1605–11.10.1002/cbic.200800841Search in Google Scholar PubMed

[236] Lutz B, Dentinger C, Sun L, Nguyen L, Zhang J, Chmura A, Allen A, Chan S, Knudsen B. Raman nanoparticle probes for antibody-based protein detection in tissues. J Histochem Cytochem 2008;56:371–9.10.1369/jhc.7A7313.2007Search in Google Scholar PubMed PubMed Central

[237] Zhou X, Xu W, Wang Y, Kuang Q, Shi Y, Zhong L, Zhang Q. Fabrication of cluster/shell Fe3O4/Au nanoparticles and application in protein detection via a SERS method. J Phys Chem C 2010;114:19607–13.10.1021/jp106949vSearch in Google Scholar

[238] Granger JH, Granger MC, Firpo MA, Mulvihill SJ, Porter MD. Toward development of a surface-enhanced Raman scattering (SERS)-based cancer diagnostic immunoassay panel. Analyst 2013;138:410–6.10.1039/C2AN36128KSearch in Google Scholar

[239] Domenici F, Bizzarri AR, Cannistraro S. SERS-based nanobiosensing for ultrasensitive detection of the p53 tumor suppressor. Int J Nanomed 2011;6:2033–42.Search in Google Scholar

[240] Domenici F, Bizzarri AR, Cannistraro S. Surface-enhanced Raman scattering detection of wild-type and mutant p53 proteins at very low concentration in human serum. Anal Biochem 2012;421:9–15.10.1016/j.ab.2011.10.010Search in Google Scholar PubMed

[241] Toledo F, Wahl GM. Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat Rev Cancer 2006;6:909–23.10.1038/nrc2012Search in Google Scholar PubMed

[242] Wu M, Mao C, Chen Q, Cu X-W, Zhang W-S. Serum p53 protein and anti-p53 antibodies are associated with increased cancer risk: a case-control study of 569 patients and 879 healthy controls. Mol Biol Rep 2010;37:339–43.10.1007/s11033-009-9744-7Search in Google Scholar PubMed

[243] Balogh GA, Mailo DA, Corte MM, Roncoroni P, Nardi H, Vincent E, Martinez D, Cafasso ME, Frizza A, Ponce G, Vincent E, Barutta E, Lizarraga P, Lizarraga G, Monti C, Paolillo E, Vincent R, Quatroquio R, Grimi C, Maturi H, Aimale M, Spinsanti C, Montero H, Santiago J, Shulman L, Rivadulla M, Machiavelli M, Salum G, Cuevas MA, Picolini J, Gentili A, Gentili R, Mordoh J. Mutant p53 protein in serum could be used as a molecular marker in human breast cancer. Int J Oncol 2006; 28:995–1002.10.3892/ijo.28.4.995Search in Google Scholar PubMed

[244] Wu L, Wang Z, Zong S, Chen H, Wang C, Xu S, Cui Y. Simultaneous evaluation of p53 and p21 expression level for early cancer diagnosis using SERS technique. The Analyst 2013;138:3450–6.10.1039/c3an00181dSearch in Google Scholar PubMed

[245] Schutz M, Steinigeweg D, Salehi M, Kompe K, Schlucker S. Hydrophilically stabilized gold nanostars as SERS labels for tissue imaging of the tumor suppressor p63 by immuno-SERS microscopy. Chem Commun 2011;47:4216–8.10.1039/c0cc05229aSearch in Google Scholar PubMed

[246] Salehi M, Steinigeweg D, Ströbel P, Marx A, Packeisen J, Schlücker S. Rapid immuno-SERS microscopy for tissue imaging with single-nanoparticle sensitivity. J Biophotonics 2013, DOI: 10.1002/jbio.201200148.10.1002/jbio.201200148Search in Google Scholar PubMed

[247] Wang DG, Fan JB, Siao CJ, Berno A, Young P, Sapolsky R, Ghandour G, Perkins N, Winchester E, Spencer J, Kruglyak L, Stein L, Hsie L, Topaloglou T, Hubbell E, Robinson E, Mittmann M, Morris MS, Shen N, Kilburn D, Rioux J, Nusbaum C, Rozen S, Hudson TJ, Lipshutz R, Chee M, Lander ES. Large-scale identification, mapping, and genotyping of single-nucleotide polymorphisms in the human genome. Science 1998;280:1077–82.10.1126/science.280.5366.1077Search in Google Scholar PubMed

[248] Sachidanandam R, Weissman D, Schmidt SC, Kakol JM, Stein LD, Marth G, Sherry S, Mullikin JC, Mortimore BJ, Willey DL, Hunt SE, Cole CG, Coggill PC, Rice CM, Ning Z, Rogers J, Bentley DR, Kwok PY, Mardis ER, Yeh RT, Schultz B, Cook L, Davenport R, Dante M, Fulton L, Hillier L, Waterston RH, McPherson JD, Gilman B, Schaffner S, Van Etten WJ, Reich D, Higgins J, Daly MJ, Blumenstiel B, Baldwin J, Stange-Thomann N, Zody MC, Linton L, Lander ES, Altshuler D; International SNP Map Working Group. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 2001;409:928–33.10.1038/35057149Search in Google Scholar PubMed

[249] Aouacheria A, Navratil V, Wen WY, Jiang M, Mouchiroud D, Gautier C, Gouy M, Zhang M. In silico whole-genome scanning of cancer-associated nonsynonymous SNPs and molecular characterization of a dynein light chain tumour variant. Oncogene 2005;24:6133–42.10.1038/sj.onc.1208745Search in Google Scholar PubMed

[250] Martini M, Vecchione L, Siena S, Tejpar S, Bardelli A. Targeted therapies: how personal should we go? Nature Rev Clin Oncology 2012;9:87–97.10.1038/nrclinonc.2011.164Search in Google Scholar PubMed

[251] Moody B, McCarty G. Statistically significant Raman detection of midsequence single nucleotide polymorphisms. Anal Chem 2009;81:2013–16.10.1021/ac802247ySearch in Google Scholar PubMed

[252] Lowe AJ, Huh YS, Strickland AD, Erickson D, Batt CA. Multiplex single nucleotide polymorphism genotyping utilizing ligase detection reaction coupled surface enhanced Raman spectroscopy. Anal Chem 2010;82:5810–4.10.1021/ac100921bSearch in Google Scholar PubMed

[253] Huh YS, Lowe AJ, Strickland AD, Batt CA, Erickson D. Surface-enhanced Raman scattering based ligase detection reaction. J Am Chem Soc 2009;131:2208–13.10.1021/ja807526vSearch in Google Scholar PubMed PubMed Central

[254] Yoo SM, Kang T, Kim B, Lee SY. Detection of single nucleotide polymorphisms by a gold nanowire-on-film SERS sensor coupled with S1 nuclease treatment. Chem Eur J 2011;17: 8657–62.10.1002/chem.201003372Search in Google Scholar PubMed

[255] Wabuyele MB, Yan F, Vo-Dinh T. Plasmonics nanoprobes: detection of single-nucleotide polymorphisms in the breast cancer BRCA1 gene. Anal Bioanal Chem 2010;398:729–36.10.1007/s00216-010-3992-1Search in Google Scholar PubMed

[256] Okumura N, Yoshida H, Kitagishi Y, Nishimura Y, Matsuda S. Alternative splicings on p53, BRCA1 and PTEN genes involved in breast cancer. Biochem Biophys Res Commun 2011;413:395–9.10.1016/j.bbrc.2011.08.098Search in Google Scholar PubMed

[257] Choi N, Lee K, Lim DW, Lee EK, Chang SI, Oh KW, Choo J. Simultaneous detection of duplex DNA oligonucleotides using a SERS-based micro-network gradient chip. Lab on a Chip 2012;12:5160–7.10.1039/c2lc40890bSearch in Google Scholar PubMed

[258] Sun L, Yu CX, Irudayaraj J. Raman multiplexers for alternative gene splicing. Anal Chem 2008;80:3342–9.10.1021/ac702542nSearch in Google Scholar PubMed

[259] Sun L, Irudayaraj J. PCR-free quantification of multiple splice variants in a cancer gene by surface-enhanced Raman spectroscopy. J Phys Chem B 2009;113:14021–5.10.1021/jp908225fSearch in Google Scholar PubMed PubMed Central

[260] Sun L, Irudayaraj J. Quantitative surface-Enhanced raman for gene expression estimation. Biophys J 2009;96:4709–16.10.1016/j.bpj.2009.03.021Search in Google Scholar PubMed PubMed Central

[261] Das G, Chirumamilla M, Toma A, Gopalakrishnan A, Zaccaria RP, Alabastri A, Leoncini M, Di Fabrizio E. Plasmon based biosensor for distinguishing different peptides mutation states. Scientific reports 2013;3:1792.10.1038/srep01792Search in Google Scholar

[262] Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004;116:281–97.10.1016/S0092-8674(04)00045-5Search in Google Scholar

[263] Zhang B, Wang Q, Pan X. MicroRNAs and their regulatory roles in animals and plants. J Cell Physiol 2007;210:279–89.10.1002/jcp.20869Search in Google Scholar PubMed

[264] Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005;120:15–20.10.1016/j.cell.2004.12.035Search in Google Scholar PubMed

[265] Calin GA, Croce CM. Genomics of chronic lymphocytic leukemia microRNAs as new players with clinical significance. Semin Oncol 2006;33:167–73.10.1053/j.seminoncol.2006.01.010Search in Google Scholar PubMed

[266] Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, Wojcik SE, Aqeilan RI, Zupo S, Dono M, Rassenti L, Alder H, Volinia S, Liu CG, Kipps TJ, Negrini M, Croce CM. miR-15 and miR-16 induce apoptosis by targeting BCL2. P Natl Acad Sci USA 2005;102:13944–9.10.1073/pnas.0506654102Search in Google Scholar PubMed PubMed Central

[267] Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer 2006;6:259–69.10.1038/nrc1840Search in Google Scholar PubMed

[268] Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR. MicroRNA expression profiles classify human cancers. Nature 2005;435:834–8.10.1038/nature03702Search in Google Scholar PubMed

[269] Nelson PT, BaldwinDA, Scearce LM, Oberholtzer JC, Tobias JW, Mourelatos Z. Microarray-based, high-throughput gene expression profiling of microRNAs. Nat Method 2004;1:155–61.10.1038/nmeth717Search in Google Scholar PubMed

[270] Driskell JD, Seto AG, Jones LP, Jokela S, Dluhy RA, Zhao YP, Tripp RA. Rapid microRNA (miRNA) detection and classification via surface-enhanced Raman spectroscopy (SERS). Biosens Bioelectron 2008;24:917–22.10.1016/j.bios.2008.07.060Search in Google Scholar PubMed

[271] Driskell JD, Primera-Pedrozo OM, Dluhy RA, Zhao Y, Tripp RA. Quantitative surface-enhanced Raman spectroscopy based analysis of MicroRNA mixtures. Appl Spectros 2009;63: 1107–14.10.1366/000370209789553183Search in Google Scholar PubMed

[272] Driskell JD, Tripp RA. Label-free SERS detection of microRNA based on affinity for an unmodified silver nanorod array substrate. Chem Commun 2010;46:3298–300.10.1039/c002059aSearch in Google Scholar PubMed

[273] Abell JL, Garren JM, Driskell JD, Tripp RA, Zhao Y. Label-free detection of Micro-RNA hybridization using surface-enhanced Raman spectroscopy and least-squares analysis. J Am Chem Soc 2012;134:12889–92.10.1021/ja3043432Search in Google Scholar PubMed

[274] Esteller M. Molecular origins of cancer: epigenetics in cancer. N Engl J Med 2008;358:1148–59.10.1056/NEJMra072067Search in Google Scholar PubMed

[275] Kriaucionis S, Heintz N. The nuclear DNA base 5-hydroxymethylcytosine is present in purkinje neurons and the brain. Science 2009;324:929–30.10.1126/science.1169786Search in Google Scholar PubMed PubMed Central

[276] Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, Agarwal S, Iyer LM, Liu DR, Aravind L, Rao A. Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1. Science 2009;324:930–5.10.1126/science.1170116Search in Google Scholar PubMed PubMed Central

[277] Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene 2002;21:5427–40.10.1038/sj.onc.1205600Search in Google Scholar PubMed

[278] Laird PW. The power and the promise of DNA methylation markers. Nat Rev Cancer 2003;3:253–66.10.1038/nrc1045Search in Google Scholar PubMed

[279] Muller HM, Widschwendter A, Fiegl H, Ivarsson L, Goebel G, Perkmann E, Marth C, Widschwendter M. DNA methylation in serum of breast cancer patients: an independent prognostic marker. Cancer Res 2003;63:7641–5.Search in Google Scholar

[280] Yu J, Cheng YY, Tao Q, Cheung KF, Lam CN, Geng H, Tian LW, Wong YP, Tong JH, Ying JM, Jin H, To KF, Chan FK, Sung JJ. Methylation of protocadherin 10, a novel tumor suppressor, is associated with poor prognosis in patients with gastric cancer. Gastroenterology 2009;136:640–51.10.1053/j.gastro.2008.10.050Search in Google Scholar PubMed

[281] Shames DS, Minna JD, Gazdar AF. Methods for detecting DNA methylation in tumors: From bench to bedside. Cancer Lett 2007;251:187–98.10.1016/j.canlet.2006.10.014Search in Google Scholar PubMed

[282] Hu J, Zhang C-y. Single base extension reaction-based surface enhanced Raman spectroscopy for DNA methylation assay. Biosens Bioelectron 2012;31:451–7.10.1016/j.bios.2011.11.014Search in Google Scholar PubMed

[283] Barhoumi A, Halas NJ. Detecting chemically modified DNA bases using surface-enhanced Raman spectroscopy. J Phys Chem Lett 2011;2:3118–23.10.1021/jz201423bSearch in Google Scholar PubMed PubMed Central

[284] El-Said WA, Kim TH, Kim H, Choi JW. Detection of effect of chemotherapeutic agents to cancer cells on gold nanoflower patterned substrate using surface-enhanced Raman scattering and cyclic voltammetry. Biosens Bioelectron 2010;26:1486–92.10.1016/j.bios.2010.07.089Search in Google Scholar PubMed

[285] Farquharson S, Gift A, Shende C, Inscore F, Ordway B, Farquharson C, Murren J. Surface-enhanced Raman spectral measurements of 5-fluorouracil in saliva. Molecules 2008;13:2608–27.10.3390/molecules13102608Search in Google Scholar PubMed PubMed Central

[286] Barhoumi A, Zhang D, Tam F, Halas NJ. Surface-enhanced Raman spectroscopy of DNA. J Am Chem Soc 2008;130:5523–9.10.1021/ja800023jSearch in Google Scholar PubMed

[287] Ock K, Jeon WI, Ganbold EO, Kim M, Park J, Seo JH, Cho K, Joo SW, Lee SY. Real-time monitoring of glutathione-triggered thiopurine anticancer drug release in live cells investigated by surface-enhanced Raman scattering. Anal Chem 2012;84:2172–8.10.1021/ac2024188Search in Google Scholar PubMed

[288] Wang L-S, Chuang M-C, Ho J-a A. Nanotheranostics - a review of recent publications. Int J Nanomed 2012;7:4679–95.Search in Google Scholar

[289] Lee JE, Lee N, Kim T, Kim J, Hyeon T. Multifunctional mesoporous silica nanocomposite nanoparticles for theranostic applications. Accounts Chem Res 2011;44:893–902.10.1021/ar2000259Search in Google Scholar PubMed

[290] Zhang Y, Qian J, Wang D, Wang Y, He S. Multifunctional gold nanorods with ultrahigh stability and tunability for in vivo fluorescence imaging, SERS detection, and photodynamic therapy. Angew Chem-Int Edit 2013;52:1148–51.10.1002/anie.201207909Search in Google Scholar PubMed

[291] Fales AM, Yuan H, Vo-Dinh T. Cell-penetrating Peptide enhanced intracellular Raman imaging and photodynamic therapy. Mole pharmaceutics 2013;10:2291–8.10.1021/mp300634bSearch in Google Scholar PubMed PubMed Central

[292] Plaetzer K, Kiesslich T, Oberdanner CB, Krammer B. Apoptosis following photodynamic tumor therapy: Induction, mechanisms and detection. Curr Pharm Design 2005;11:1151–65.10.2174/1381612053507648Search in Google Scholar PubMed

[293] Tian L, Gandra N, Singamaneni S. Monitoring controlled release of payload from gold nanocages using surface enhanced Raman scattering. ACS Nano 2013;7:4252–60.10.1021/nn400728tSearch in Google Scholar PubMed

[294] Song J, Zhou J, Duan H. Self-assembled plasmonic vesicles of SERS-encoded amphiphilic gold nanoparticles for cancer cell targeting and traceable intracellular drug delivery. J Am Chem Soc 2012;134:13458–69.10.1021/ja305154aSearch in Google Scholar PubMed

[295] Mahajan S, Richardson J, Brown T, Bartlett PN. SERS-melting: a new method for discriminating mutations in DNA sequences. J Am Chem Soc 2008;130:15589–601.10.1021/ja805517qSearch in Google Scholar PubMed

[296] Mahajan S, Richardson J, Ben Gaied N, Zhao ZY, Brown T, Bartlett PN. The use of an electroactive marker as a SERS label in an e-melting mutation discrimination assay. Electroanalysis 2009;21:2190–7.10.1002/elan.200904681Search in Google Scholar

[297] Lubamba B, Dhooghe B, Noel S, Leal T. Cystic fibrosis: Insight into CFTR pathophysiology and pharmacotherapy. Clin Biochem 2012;45:1132–44.10.1016/j.clinbiochem.2012.05.034Search in Google Scholar PubMed

[298] Corrigan DK, Gale N, Brown T, Bartlett PN. Analysis of short tandem repeats by using SERS monitoring and electrochemical melting. Angew Chem-Int Edit 2010;49:5917–20.10.1002/anie.201001389Search in Google Scholar PubMed

[299] Jiang X, Jiang Z, Xu T, Su S, Zhong Y, Peng F, Su Y, He Y. Surface-enhanced Raman scattering-based sensing in vitro: facile and label-free detection of apoptotic cells at the single-cell level. Anal Chem 2013;85:2809–16.10.1021/ac303337bSearch in Google Scholar PubMed

[300] Sathuluri RR, Yoshikawa H, Shimizu E, Saito M, Tamiya E. Gold nanoparticle-based surface-enhanced Raman scattering for noninvasive molecular probing of embryonic stem cell differentiation. Plos One 2011;6:e22802.10.1371/journal.pone.0022802Search in Google Scholar PubMed PubMed Central

[301] Huefner A, Kuan W-L, Barker RA, Mahajan S. Intracellular SERS nanoprobes for distinction of different neuronal cell types. Nano Lett 2013;13:2463–70.10.1021/nl400448nSearch in Google Scholar PubMed PubMed Central

[302] Neumann O, Zhang DM, Tam F, Lal S, Wittung-Stafshede P, Halas NJ. Direct optical detection of aptamer conformational changes induced by target molecules. Anal Chem 2009;81:10002–6.10.1021/ac901849kSearch in Google Scholar PubMed

[303] Chen JW, Jiang JH, Gao X, Liu GK, Shen G, Yu R. A new aptameric biosensor for cocaine based on surface-enhanced Raman scattering spectroscopy. Chem Eur J 2008;14:8374–82.10.1002/chem.200701307Search in Google Scholar PubMed

[304] Sanles-Sobrido M, Rodriguez-Lorenzo L, Lorenzo-Abalde S, Gonzalez-Fernandez A, Correa-Duarte MA, Alvarez-Puebla RA, Liz-Marzán LM. Label-free SERS detection of relevant bioanalytes on silver-coated carbon nanotubes: the case of cocaine. Nanoscale 2009;1:153–8.10.1039/b9nr00059cSearch in Google Scholar PubMed

[305] Tu Q, Eisen J, Chang C. Surface-enhanced Raman spectroscopy study of indolic molecules adsorbed on gold colloids. J Biomed Opt 2010;15:020512.10.1117/1.3400660Search in Google Scholar PubMed

[306] Perez-Pineiro R, Correa-Duarte MA, Salgueirino V, Alvarez-Puebla RA. SERS assisted ultra-fast peptidic screening: a new tool for drug discovery. Nanoscale 2012;4:113–6.10.1039/C1NR11293GSearch in Google Scholar PubMed

[307] Yuen JM, Shah NC, Walsh JT, Jr, Glucksberg MR, Van Duyne RP. Transcutaneous glucose sensing by surface-enhanced spatially offset Raman spectroscopy in a rat model. Anal Chem 2010;82:8382–5.10.1021/ac101951jSearch in Google Scholar PubMed PubMed Central

[308] Hsu P-H, Chiang HK. Surface-enhanced Raman spectroscopy for quantitative measurement of lactic acid at physiological concentration in human serum. J Raman Spectros 2010;41:1610–4.10.1002/jrs.2609Search in Google Scholar

[309] Manno D, Filippo E, Fiore R, Serra A, Urso E, Rizzello A, Maffia M. Monitoring prion protein expression in complex biological samples by SERS for diagnostic applications. Nanotechnology 2010;21:165502.10.1088/0957-4484/21/16/165502Search in Google Scholar PubMed

[310] Alvarez-Puebla RA, Zubarev ER, Kotov NA, Liz-Marzan LM. Self-assembled nanorod supercrystals for ultrasensitive SERS diagnostics. Nano Today 2012;7:6–9.10.1016/j.nantod.2011.11.001Search in Google Scholar

[311] Alvarez-Puebla RA, Agarwal A, Manna P, Khanal BP, Aldeanueva-Potel P, Carbó-Argibay E, Pazos-Pérez N, Vigderman L, Zubarev ER, Kotov NA, Liz-Marzán LM. Gold nanorods 3D-supercrystals as surface enhanced Raman scattering spectroscopy substrates for the rapid detection of scrambled prions. P Natl Acad Sci USA 2011;108:8157–61.10.1073/pnas.1016530108Search in Google Scholar PubMed PubMed Central

[312] Serra A, Manno D, Filippo E, Buccolieri A, Urso E, Rizzello A, Maffia M. SERS based optical sensor to detect prion protein in neurodegenerate living cells. Sensor Actuat B-Chem 2011;156:479–85.10.1016/j.snb.2011.04.019Search in Google Scholar

[313] Lorca RA, Varela-Nallar L, Inestrosa NC, Huidobro-Toro JP. The cellular prion protein prevents copper-induced inhibition of P2X(4) receptors. Int J Alzheimer’s Disease 2011;2011: Article ID 706576, 6 pages (doi: 10.4061/2011/706576).Search in Google Scholar

[314] Becker M, Budich C, Deckert V, Janasek D. Isotachophoretic free-flow electrophoretic focusing and SERS detection of myoglobin inside a miniaturized device. Analyst 2009;134:38–40.10.1039/B816717FSearch in Google Scholar PubMed

[315] Brazhe NA, Abdali S, Brazhe AR, Luneva OG, Bryzgalova NY, Parshina EY, Sosnovtseva OV, Maksimov GV. New insight into erythrocyte through in vivo surface-enhanced raman spectroscopy. Biophys J 2009;97:3206–14.10.1016/j.bpj.2009.09.029Search in Google Scholar PubMed PubMed Central

[316] Neng J, Harpster MH, Zhang H, Mecham JO, Wilson WC, Johnson PA. A versatile SERS-based immunoassay for immunoglobulin detection using antigen-coated gold nanoparticles and malachite green-conjugated protein A/G. Biosens Bioelectron 2010;26:1009–1015.10.1016/j.bios.2010.08.015Search in Google Scholar PubMed

[317] Chen Y, Cheng H, Tram K, Zhang S, Zhao Y, Han L, Chen Z, Huan S. A paper-based surface-enhanced resonance Raman spectroscopic (SERRS) immunoassay using magnetic separation and enzyme-catalyzed reaction. Analyst 2013;138:2624–31.10.1039/c3an36647bSearch in Google Scholar PubMed

[318] Chen J-W, Lei Y, Liu X-J, Jiang J-H, Shen GL, Yu RQ. Immunoassay using surface-enhanced Raman scattering based on aggregation of reporter-labeled immunogold nanoparticles. Anal Bioanal Chem 2008;392:187–93.10.1007/s00216-008-2237-zSearch in Google Scholar PubMed

[319] He L, Deen B, Rodda T, Ronningen I, Blasius T, Haynes C, Diez-Gonzalez F, Labuza TP. Rapid detection of ricin in milk using immunomagnetic separation combined with surface-enhanced Raman spectroscopy. J Food Science 2011;76:N49–53.10.1111/j.1750-3841.2011.02196.xSearch in Google Scholar PubMed

[320] He L, Lamont E, Veeregowda B, Sreevatsan S, Haynes CL, Diez-Gonzalez F, Labuza TP. Aptamer-based surface-enhanced Raman scattering detection of ricin in liquid foods. Chem Science 2011;2:1579–82.10.1039/c1sc00201eSearch in Google Scholar

[321] Zhu Y, Kuang H, Xu L, Ma W, Peng C, Hua Y, Wang L, Xu C. Gold nanorod assembly based approach to toxin detection by SERS. J Mater Chem 2012;22:2387–91.10.1039/C2JM15238JSearch in Google Scholar

[322] Guven B, Boyaci IH, Tamer U, Calik P. A rapid method for detection of genetically modified organisms based on magnetic separation and surface-enhanced Raman scattering. Analyst 2012;137:202–8.10.1039/C1AN15629BSearch in Google Scholar PubMed

[323] Muniz-Miranda M, Gellini C, Salvi PR, Pagliai M. Surface-enhanced Raman micro-spectroscopy of DNA/RNA bases adsorbed on pyroxene rocks as a test of in situ search for life traces on Mars. J Raman Spectros 2010;41:12–15.10.1002/jrs.2398Search in Google Scholar

[324] Arslanoglu J, Zaleski S, Loike J. An improved method of protein localization in artworks through SERS nanotag-complexed antibodies. Anal Bioanal Chem 2011;399:2997–3010.10.1007/s00216-010-4378-0Search in Google Scholar PubMed

Received: 2013-6-19
Accepted: 2013-9-5
Published Online: 2013-10-5
Published in Print: 2014-12-1

©2014 Science Wise Publishing & De Gruyter

Downloaded on 22.5.2024 from https://www.degruyter.com/document/doi/10.1515/nanoph-2013-0024/html
Scroll to top button