• No results found

Immunoassays or LC-MS/MS?

N/A
N/A
Protected

Academic year: 2021

Share "Immunoassays or LC-MS/MS?"

Copied!
58
0
0

Loading.... (view fulltext now)

Full text

(1)

19-X5

Immunoassays

or

LC-MS/MS?

A

Comparison

Revealing

the

Properties

of

Modern

Methods

for

Insulin,

Pro-insulin,

C-peptide

and

Glucagon

Quantification

Anna

Ivert

Nordén,

Temis

Martinez,

Casper

Tiger,

Viktor

Umenius,

Ruta

Upite,

Susanna

Wärmegård

Beställare:

Mercodia

AB

Beställarrepresentant:

Johan

Ledin

Handledare:

Lena

Henriksson

1MB332,

Självständigt

arbete

i

molekylär

bioteknik,

15

hp,

vt

2019

Civilingenjörsprogrammet

i

molekylär

bioteknik

(2)

Abstract 

 

The purpose of this report is to compare seven different methods for biomarker detection  and quantification based on previously published papers. The methods investigated are  ELISA, LC-MS/MS, UPLC-MS/MS, LC-IM/MS, IA-LC-MS/MS, MSIA-HR/AM, HTRF and  AlphaLISA​® .

 

 

The focus lies on biomarkers relevant for diabetes, obesity and cardiovascular diseases.  Namely insulin, proinsulin, glucagon and C-peptide. Particular significance is assigned to  the comparison of the currently widest used method, ELISA, with various types of 

LC-MS/MS. 

 

 

The report concludes ELISA being superior to LC-MS/MS methods in terms of recovery  and precision, while LC-MS/MS is superior in accuracy, multiplexing, specificity, 

throughput and sample cost. This suggests that different types of LC-MS/MS has the  potential to gain momentum in the field of biomarker quantification if they become more  available.   

   

(3)

Table of Contents   

1. Vocabulary and Abbreviations

2. Background

2.1 Rationale for this project 6 

2.2 Detection today 6 

3. Workflow

4. Technology Descriptions

4.1 ELISA 9 

4.2 Liquid Chromatography and Mass Spectroscopy Methods 10 

4.2.1 LC-MS/MS 10 

4.2.2 UPLC-MS/MS 12 

4.2.3 LC-IM-MS 12 

4.2.4 IA-LC-MS/MS 13 

4.2.5 MSIA-HR-AM 15 

4.3 Other Methods for Detection and Quantification 16 

4.3.1 HTRF 16 

4.3.2 AlphaLISA® 17 

5. Explanation of parameters upon comparison 19 

6. Summarised comparison of the methods 21 

6.1 Insulin 21 

6.2 Proinsulin 22 

6.3 C-peptide 23 

6.4 Glucagon 24 

7. Remarks regarding comparison of the methods 25 

7.1 Comparison Between ELISA and LC-MS Methods 25 

7.1.1 Accuracy for LC-MS/MS 25 

7.1.2 Automatisation and multiplexing 26  7.1.3 Sample preparation with respect to complex matrices 26 

7.1.4 UPLC-MS/MS 27 

7.1.5 LC-IM-MS 27 

7.1.6 Availability of data about the biomarkers among the methods 27  7.2 Comparison Between ELISA and other immunochemical methods 28 

7.2.1 AlphaLISA® 28 

(4)

7.2.3 Hook effect in immunochemical methods 29 

7.3 Economics 29 

8. Conclusion 30 

9. Ethical Discussion 31 

9.1 Production of Antibodies 31 

9.2 Diagnostics or Research Only 32 

9.3 New Standard Technology for Detection and Quantification 32 

9.4 Production and Transport 33 

10. Acknowledgements 34  11. Statement of Contribution 34  12. References 36  12.1 Articles 36  12.2 Online sources 39  Appendix 1 41 

Not Selected Technologies 41 

(5)

1. Vocabulary and Abbreviations 

2D-method: ​A method that uses two different liquid chromatography columns to separate  the sample before being analyzed by two mass spectrometers in series. 

Biomarker:​ A measurable biological indicator. In this report, biomarker is referred to as the  molecules insulin, proinsulin, C-peptide and glucagon. 

Cross-reactions:​ In this report - cross-reactivity of an antibody in the context of 

immunology. Reliant on an antibody’s antigen-binding site several proteins/peptides might  get bound if the target molecule shares similarity meaning that the same antibody will  detect several molecules. Depending on the purpose of immuno-recognition this might  lead to false positives upon detecting a specific target molecule. 

CV​: Coefficient of variation upon cross validation. Performed with analytes in different  concentrations. Smaller variation between the used data sets indicate greater precision.  Endogenous peptide: ​Peptide that is produced in the sampled organism. 

Exogenous peptide: ​Peptide added to a sample/sampled organism. 

FRET: ​Fluorescence Resonance Energy Transfer, upon close contact (<10nm) two molecules  can transfer energy to one another without using radiation. 

Hyphenated techniques​: Hyphenation refers to coupling different techniques. Hyphenated  methods in this report are combinations of different liquid chromatography and mass  spectroscopic techniques as well as immunoaffinity based separation techniques. 

Immunochemical methods:​ Methods using antibodies for the purpose of detecting specific  proteins/peptides. ELISA and RIA are examples of immunochemical methods. 

Immunosorbent:​ An insoluble support for an antigen or antibodies used to adsorb 

antibodies or antigen from a mixture. The adsorbed material can then be rinsed out in pure  form for analysis. Many different substances are used, including sepharose, glutaraldehyde,  copolymers of anhydrides, polyacrylamides and others. 

(6)

Matrix-effects:​ Matrix depending signals. Non-target molecules can affect quantification  upon measurements. Interferences may enhance or suppress signals.  

Multiplexing:​ Detecting multiple peptides simultaneously. In biomarker detection it is of  interest to detect isoforms arising from, for example, posttranslational modifications as  well as relevant exogenous analogs of the same peptide. 

PTM: ​Posttranslational modifications 

Recovery​: A measure of accuracy, quantifying agreement between a known reference value  (standard) and the found value. 

RIA:​ Radioimmunoassay, quantifies analyte by detecting radioactivity of radiolabeled  antigens that are released from a complex as the interesting antigen outcompetes the  radioactive ones from an antibody-antigen complex. 

Signal-to-noise ratio: ​Describes the ratio between the wanted signal and the unwanted  background noise from the sample. 

Targeted Mass Spectrometry (MRM/SRM):​ Multiple reaction monitoring (MRM, also  known as Selective Reaction Monitoring – SRM) is a highly specific and sensitive mass  spectrometry technique. It uses triple quadrupole MS where the molecule is fragmented in  several steps and the target ions as well as their daughter ions are tracked. The technique  is especially suitable for detection of bigger peptides with mass range outside of the  measurement range of simpler MS instruments. 

Top down and bottom up analysis: ​ Top down analysis uses the whole protein in the  MS/MS analysis. Bottom up analysis breaks down the compound into smaller components  prior to the MS/MS analysis. Bottom up is more complex but more suitable for bigger  molecules. 

(7)

2. Background

 

In this report we aimed to explore available methods for biomarker quantification and  detection aside from the currently most widely used method, Enzyme-linked 

ImmunoSorbent Assay (ELISA) . In particular, various liquid chromatography tandem mass  spectrometry (LC-MS/MS) methods were of interest. Other antibody utilising methods  such as Homogeneous Time Resolved Fluorescence (HTRF) and AlphaLISA​® are reviewed as 

well. LC-MS/MS methods are of interest as they have seen an upswing in popularity during  the last decade as they offer improvements of quantification that traditional methods like  ELISA do not support. 

2.1 Rationale for this project 

This study was requested by Mercodia AB in order to gain a summarising view of what  types of methods for biodetection that currently exist. The company develops ELISA kits  for complex matrices such as blood serum and plasma. Therefore, the articles analysed in  this study concerns primarily these matrices. Following biomarkers were researched:  insulin, proinsulin, C-peptide and glucagon, as these are the most important biomarkers  that Merodia AB develops kits for and are therefore of greatest interest. 

 

Identifying and quantifying biomarkers is important for tracing and detecting a wide  variety of diseases. Thus biomarker detection is a foundation in many areas of research in  order to understand the biological processes causing disorders in living organisms. 

Diabetes, obesity and cardiovascular diseases can for example be linked to the biomarkers  we aim to look at in this study. Societal costs for these diseases, due to their effect on the  quality of life, contributes to demand for reliable biomarker detection. The interest to  detect biomarkers is also of great importance in other fields of research, such as common  animal studies. Biomarkers in this study have multifaceted functions in organisms, 

understanding of their function may lead to increased knowledge in other areas beyond the  mentioned diseases. 

2.2 Detection today 

(8)

of more than 20 SEK (Sakamoto ​et al.​ 2018). Moreover, there is a risk for interference from  other molecules, such as cross reactivity with similar compounds to the target antigen,  which results in false positives. Also, ELISA has a relatively long total runtime, around 2-4  hours, with many steps involved during the procedure (Farino ​et al.​ 2016). Lastly, ELISA  requires tailored and specific antibodies for every biomarker of interest.  

 

These are the reasons for exploring potential new methods for biodetection. Furthermore,  it is shown that consensus for concentrations among different ways of measuring 

biomarker levels is not reached among methods used today. MS-based methods have been  suggested to offer more coinciding results due to streamlined methodology that minimizes  manual labour.​ However, many of the methods studied in this report solve some or most of  these issues, but as we will see, they do come with some disadvantages of their own.  

(9)

3. Workflow 

A literary study was conducted to gain background knowledge about different ways of  quantifying the biomarkers of interest from serum and plasma. Information about several  technologies was gathered and stored. The different biomarkers had almost an equal  number of articles, but insulin is overall the most studied. Comparative articles between  different technologies and ELISA were collected as well.  

 

Later, a list was made with all the twelve technologies found we deemed interesting:  LC-MS/MS, IA-LC-MS/MS, LC-IM/MS, UPLC-MS/MS, HTRF, HRMS, MSIA-HR/AM,  AlphaLISA​® , ECLIA, CLIA, IRMA and IECMA. These were assigned among the group 

members and each person investigated how two to three of these technologies functioned.  Shortly thereafter, another round of method selection was carried out based on the 

individual research.   

After the initial investigation, we chose to research further about the following methods, all  which will be described in the next section of the report: ELISA, LC-MS/MS, 

UPLC-MS/MS, IA-LC-MS/MS, LC-IM/MS, MSIA-HR/AM, HTRF and AlphaLISA​® . The 

choice was based on the amount of information that was found and our judgement of the  potential of the technologies in comparison to ELISA. Each member then searched for  information regarding two of these technologies, in articles where they were applied on  the four biomarkers of interest for this report. The methods that were not chosen from the  initial investigation, namely ECLIA, CLIA, IRMA and IECMA, are shortly described in 

Appendix 1 to show why these technologies were not selected.    

Finally, summarising tables 1.1, 1.2, 2.1, 2.2, 3.1, 4.1, 4.2, 4.3, 4.4, 5.1, 6.1, 6.2, 6.3, 7.1, 7.2 and 7.3  were compiled to the result tables 1, 2, 3 and 4 for an easier overview.  

(10)

4. Technology Descriptions 

Below follows a description of how the seven chosen methods work. It also includes an  explanation of ELISA. Their estimated prices are included. To simplify the comparison  between technologies all the prices, as cost per sample and cost of instrumentation, were  converted to SEK. The publication date of the article was taken in consideration for the  conversion ratios that year. 

4.1 ELISA 

Enzyme linked immunosorbent assay (ELISA) is an immunochemical method, and the  currently most widespread method for biomarker detection. There are many variations of  ELISA, namely indirect ELISA, direct ELISA, sandwich ELISA and competitive ELISA. They  all differ slightly from one another but share the core mechanics. Looking closer on one  method, Sandwich ELISA (​Figure 1​), which works by connecting an overabundance of  antibodies specific to the biomarker of interest to a chemically inert plate. The sample is  then washed over the plate, resulting in only the biomarkers of interest binding to the  antibodies and the rest being washed away. Then a second antibody with a connected  enzyme is washed over the plate, binding only where antigen has already been bound with  the first sets of antibodies. This ensures specificity. By adding a substrate catalysed by the  enzyme, a detectable product that is directly proportional to the amount of bound antigen  will be produced. Measuring these will give an estimation of the concentration of the target  biomarker within the sample. (Sakamoto ​et al.​ 2018) 

 

In contrast to sandwich ELISA, direct and indirect ELISA let everything in the sample first  bind to the plate and then wash with antibodies for detection. Competitive ELISA uses  different types of antibodies that competes for the antigen and then the concentration is  calculated through the ratio of catalysed substrate compared to the number of antibodies  added of each type. The typical cost per sample for ELISA is 20-50 SEK and no expensive  machines are required other than for detection of the coloured product from the enzyme  reaction. This equipment is standard lab equipment however and will not be a problem for  most research teams. A typical ELISA kit might cost around 5000 SEK for an assay covering  around 100 wells (50 SEK per sample, Thermo Fisher Scientific​TM​, 2019). (Sakamoto ​et al. 

(11)

Figure 1. Description of Sandwich ELISA. 1) Sample is washed over plate with antibodies 2) Antigen of interest  binds 3) A second wave of antibodies with a linked enzyme is washed over the plate 4) The second antibody binds to  the antigen 5) Substrate is added which are catalysed by the enzyme into detectable product. Figure created with  BioRender.com. 

4.2 Liquid Chromatography and Mass Spectroscopy Methods  

4.2.1 LC-MS/MS 

LC-MS/MS is a liquid chromatography separation combined with tandem mass 

spectrometry. The sample is first separated through a liquid chromatography as shown in 

(12)

mass analyser, in a similar way as in the first mass analyser, and finally detected (see ​Figure 

3). (Pitt 2009)   

When analysing a complex matrix, one has to take into account the matrix effects. These  are caused by all components in the sample that are not quantified and can affect the  reproducibility of the experiment. Therefore complex samples often need a pre-treatment  before they are injected into the LC-MS/MS machine. (Smeraglia ​et al.​ 2002) 

 

Depending on the type of LC machine used, the price can range from 10 000- 400 000 SEK  (Liquid Chromatographs (HPLC), Conquer Scientific. 2019). The same applies to the MS/MS  machines, where second hand machines can range in price from 500 000-2 500 000 SEK  (​LabX. 2019​). 

 

Figure 2. LC workflow. 1) The mobile phase is pumped into the column. 2) The sample is added. 3) The sample is  separated based on the affinity of the column. 4) The sample is detected. 5) Sample is analysed in a computer. 6)  Output from the separated sample for further investigation. Figure created with BioRender.com. 

(13)

Figure 3. MS/MS workflow 1) The inserted sample is ionised. 2) The ionised sample is separated in the first mass  analyser. 3) The separated sample is fragmented 4) The fragmented ions are separated in the second mass analyser  5) Detection of the wanted compounds. Figure created with BioRender.com. 

4.2.2 UPLC-MS/MS 

UPLC-MS/MS stands for Ultra Performance/Pressure Liquid Chromatography – tandem  mass spectrometry and is also known under the name UHPLC-MS/MS where the H stands  for “high”. It is a method for separation of components that combines qualitative liquid  chromatography with mass spectrometry. This liquid chromatography is better than the  ordinary because of the special columns that are used. These columns have a particle size  diameter of less than two micrometers which makes the system more efficient. In addition,  the eluent is driven through the column at a higher pressure than with a HPLC. Altogether  the properties of UPLC contributes to a time reduction. (Guillarme & Veuthey 2017) 

  

The system needs a mobile phase, a pumping system, an autosampler, a special column,  detectors in series and a data system (Chromatography & Mass Spectrometry Solutions).  Almost everything in the system can be controlled by the data system. This type of 

instrumentation could cost 2 000 000 SEK (Cross & Hornshaw 2016). However, according  to the article by Howard ​et al.​ (2017) the price will be ameliorated after a couple of years  due to the systems multiplexing ability. 

4.2.3 LC-IM-MS 

Liquid chromatography ion mobility mass spectrometry (LC-IM-MS or LC-IMS-MS) utilises  three coupled separations to create one powerful method which separates molecules with  regard to both mobility and mass/charge. The liquid chromatography and mass 

(14)

 

All the instruments needed in this method have high prices, including the IMS which cost  around 350 000 SEK​ (CBRNE Tech Index 2018​). The most time-consuming step in this  method is LC while IMS only takes a few seconds. In recent years, IMS has seen an  increased use in the detection of biomolecules, such as peptides. As can be seen in an  article by Lapthorn ​et al.​ (2013) the peak capacity for LC-IM-MS is 100 times higher than  LC-MS and greater by a factor of 25 000 compared to UHPLC. Peak capacity is a 

measurement of how good the separation is by calculating a number of maximum number  of peaks that fit in a 2D method (Lapthorn ​et al.​ 2013). 

4.2.4 IA-LC-MS/MS 

Immunoaffinity liquid chromatography tandem mass spectrometry (IA-LC-MS/MS) uses  antibodies to isolate and capture analytes of interest in a complex matrix and then runs a  LC-MS/MS for analysis. The immunoaffinity can be done in two main ways, either by using  an offline approach or by using an online approach. The online-column based method  usually requires less sample preparations, since most steps are combined in a single online  method (Dufield & Radabaugh 2012). For this, one uses affinity columns where the 

chromatography media is covalently coupled with antibodies, as shown in ​Figure 4​. The  online immunoaffinity is directly coupled to an analytical technique for analysis. With a  relatively high concentration of antibodies, this approach gives a rapid enrichment of the  sample and can be regenerated and used for several runs. Disadvantages with using this  method is that there is a risk for sample transfer and it has difficulties running large sample  volumes.  

 

The offline based approach of enrichment uses either magnetic particles coupled with  antibodies (see ​Figure 5​), beads coupled with antibodies or immobilised antibodies in pipet  tips. The advantages with the offline method is that samples can be processed in parallel by  using larger sample volumes. (Whiteaker & Paulovich 2011) 

 

The price for this method, excluding the price for LC and MS equipment, depends on the  immunoaffinity approach. A bead-based approach using Dynabeads™​ ​(Thermo Fisher 

Scientific​TM​, 2019) costs between 24 000 to 33 000 SEK for 10 mL. Using the columns 

(15)

Figure 4. Online column based immunoaffinity which is directly coupled with an analytical technique. 1) Sample is  added to an antibody coated column. 2) After rinsing, everything but the antigen bound to the antibodies will leave  the column. 3) Only antigens remain from the sample, they are then eluated out and introduced into an 

LC-MS/MS system. Figure created with BioRender.com.  

 

Figure 5. Offline bead based immunoaffinity. 1) Fill tube with magnetic beads. 2) Added antibodies will bind to the  beads. 3) After addition of sample, antigen will bind to the antibodies. 4) Using a magnet to hold the beads,  everything else is removed. 5) Eluate out beads with antigen and wash off antigen, then transfer into an 

(16)

4.2.5 MSIA-HR-AM 

Mass spectrometric immunoassay - high resolution - accurate mass (MSIA-HR-AM) applies  immunoaffinity based enrichment of biomarkers with high resolution LC-MS/MS for  quantification of the peptides of interest. Use of MSIA microcolumns allows streamlined,  multiplexed screening for biomarkers as the workflow is partially automatised. The  monolithic microcolumns used for MSIA are activated with an antibody and fixed in a  pipette tip. Immunoaffinity retrieval is obtained with 100-250 cycles of aspiration and  dispensation on a pipettor (see ​Figure 6) (Oran ​et al. 2011, Peterman ​et al. 2014, Nedelkov ​et 

al.​ 2018).   

Enrichment with help of a polyclonal antibody serves as a precondition for further analysis  in MS. Antibodies are polyclonal with respect to isoforms and PTMs as several forms of  analysed peptides are recognized. The elute from the MSIA step is loaded in a LC-MS/MS  system. To gain high-resolution data that fulfills HR-AM standards, a spectrometer with  sufficient resolution is required. Data analysis is then performed using software such as  Pinpoint™ Software (Thermo Fisher Scientific​TM​) in order to quantify biomarkers as well as 

to perform a qualitative analysis of the obtained peaks (Peterman ​et al.​ 2014).    

A MSIA step combined with LC-MS/MS offers a top down analysis. MSIA can be 

hyphenated with selected reaction monitoring (SRM) - a bottom up approach that obtains  an even higher resolution. 

 

Throughput of the method is highly dependent on the pipettor used. Thermo Fisher  Scientific​TM​ offers the MSIA tips and several products applicable to MSIA protocols - 

multichannel pipette, a liquid handler system and a fully automated system. Studies  available and used in our comparison uses a multichannel pipette or a 96-channel liquid  handling system. This system (Versette​TM​ system for MSIA application, Thermo Fisher 

Scientific​TM​, 2019) costs 400 000 SEK. The custom MSIA D.A.R.T.’S​TM​ (Disposable 

(17)

Figure 6. Immunoenrichment steps in MSIA tips. 1) Pipette tips are connected to the pipettor. 2) Closer look at the  pipette tip. 3) The sample is filtered by pipetting the sample up and down multiple times through the antibody filled  column within the pipette tip, antigens bind to the antibodies. 4) The antigens are eluted. Figure created with  BioRender.com.  

4.3 Other Methods for Detection and Quantification 

4.3.1 HTRF

 

 

Homogeneous Time Resolved Fluorescence (HTRF) (​Figure 7​) is a method that utilises  Fluorescence Resonance Energy Transfer (FRET) between two molecules attached to two  separate antibodies. The first molecule is called the “donor” and this excites the second  molecule, the “acceptor”, via FRET. When the antibodies are not connected to the antigen,  the molecules attached to the antibodies are too far apart (>10nm) for FRET to occur. When  both antibodies bind to the antigen on two different sites the distance is shortened so  FRET can occur. When administering energy, the wavelength of both the excited donor and  acceptor will be detected if FRET occurred, and only the donors wavelength if FRET did not  occur. Since antibodies are added in excess, the amount of fluorescence from the acceptor  is directly linked to the number of antigens in the sample. 

 

(18)

 

Figure 7. 1) No FRET may occur because of distance 2) Distance is shortened after antibodies bind to antigen 3)  FRET occurs, donor gives energy to acceptor, acceptor gives out light of a specific wavelength that is measured. 

Figure created with BioRender.com.  

4.3.2 AlphaLISA​

®  

AlphaLISA​® is a commercial method branded by PerkinElmer, that is claimed to be an 

improved version of ELISA when working with difficult matrices or running several assays  of the same sample (​PerkinElmer. 2019. Alpha Reagents​). As shown in ​Figure 8​, it is a  sandwich assay with two antibodies coupled to two separate beads. A Biotinylated  Anti-analyte Antibody (antibody with a biotin bound to it) first binds to the analyte of  interest and then an Anti-Analyte Antibody with an AlphaLISA​® Acceptor Bead binds to the 

analyte. A Streptavidin-coated Alpha​ ​Donor Bead binds to the Biotinylated Anti-analyte 

Antibody and form a complex. This complex is transported to a laser reader, which excite  the donor bead, causing it to release an oxygen molecule to the acceptor bead. As a result,  the acceptor bead emits a sharp light peak at 615 nm which can be detected. 

(PerkinElmer. 2011. TDS – Human Insulin.)   

To implement an AlphaLISA​® assay the kit itself as well as a plate reader with laser 

excitation are needed. One plate reader branded by Perkin Elmer is EnVision Multimode  Plate Reader. A refurbished instrument costs up to 170 000 SEK and new ones have  significantly higher prices, giving this method a high starting cost. Apart from that, it is  useful to have dark plates, something to cover the plates with and a dimmed room since  the Donor beads are light-sensitive (​PerkinElmer. 2011. TDS – Human Insulin.​).​ ​AlphaLISA​®  

(19)

 

Figure 8. Schematic figure over the AlphaLISA​® reaction. An oxygen molecule is released from the Streptavidin on 

the donor bead when it is excited by the 680 nm wave emissions. This excites the molecule connected to the  acceptor bead and this emits a 615 nm wave which is measured. This may only occur if they are in close proximity,  prompted by the antibodies binding to the antigen. Figure created with BioRender.com.  

(20)

5. Explanation of parameters upon comparison 

To compare the different methods, fourteen parameters were considered. The parameters  sample volume, sensitivity, measure range, throughput, sample cost, multiplexing, 

specificity, required equipment were desired by Mercodia AB. Equipment requirements are  described above in the technology description. Number of samples, accuracy, precision,  LOD, LOQ and recovery were added since these were often given in the articles and thus  used to obtain a better comparison. In Appendix 2, the parameter ​number of samples  describes the number of individuals used in the study. These are included to ensure the  reliability of the article. The ​sample volume​ is referred to as the volume of plasma or  serum, in μL, that is required to conduct the experiment, or how much that is inserted in a  well. In the latter case, it is explained in the table. Note: the volumes are generally the  sample volumes from the patient e.g. plasma or serum, that is needed prior to adding  buffers or similar for the analytical sample preparation. ​Accuracy ​is how close the results  are to the true value. Values are presented in percent and ppm or with phrases as “good”  according to the descriptions in the original article. ​Precision​ is how close the results are  relative to each other, here the values are given in intra- and interassay percentage.    

Sensitivity​ describes the smallest amount of the sample that can be detected by the  system. The parameter is displayed as “good”, “high” or in g/mL or in mol/L depending on  the article it is taken from. This is closely connected to ​LOD​, which stands for Limit Of  Detection and is given in mol/L or g/mL and ​LOQ​, which represent the Limit Of 

Quantification in mol/L or g/mL. Table 2.2, 4.4, 7.2 and 7.3 Appendix 2 show the value for  Lowest Limit Of Quantification (LLOQ). ​Measure range​ is an interval between the lowest  and highest amount of the biomarker that can be detected with reasonable accuracy, given  in concentration per volume or mass per volume. Note: different units are compiled in the  tables as they are taken directly from the corresponding articles. The ​throughput​ refers to  the time (minutes or hours) it takes to do the analysis, excluding sample preparations.  Sample cost​ is the cost in SEK for each sample without taking the equipment in  consideration. 

 

Multiplexing​ is a yes or no parameter that describes whether or not the method can  effectively analyse several biomarkers in one run. ​Recovery​ describes the concentration of  the target molecule that is reported recovered/detected by the method in contrast to a  known concentration within a sample. The result agrees with a standard in a percentage  that could be over 100. The closer to 100% the better. 

 

(21)

correspondence to identify true m/z ratios. This ranking is biomarker specific, regards  each table separately and must not be compared across different tables. The scale goes  from A to D, where A is the highest score in each separate table. 

 

Values for the parameters did not have the same units and we have avoided to convert the  units in order not to tamper with interpretations, thus some parameters are given with  several units.  

 

All values presented in the tables are for biomarkers in complex matrices - serum or  plasma. 

 

(22)

6. Summarised comparison of the methods  

Below, all the seven different methods and ELISA are compared to one another in respect  to the four different biomarkers mentioned in the background of this report. The values are  merged from the gathered data presented in Appendix 2. Values for the parameters did not  have the same units and we have avoided to convert the units. This in order not to tamper  with interpretations, thus some parameters are given in several units. 

 

6.1 Insulin 

Table 1. Values in the respective fields are assembled from the corresponding insulin tables for each method in  Appendix 2. Upon different values in the same field there were no clear consensus among the same methods in  the tables found in Appendix 2. 

    ELISA  ELISA  Ultra-  sensitive  insulin  LC-MS/MS  UPLC-MS/ MS 

LC-IM/MS  IA-LC-MS/MS  MSIA-HR-A

HTRF  AlphaLISA​®   Sample  volume  25 μL per  well  25 μL per  well  150 - 250  μL (<5 µL)  250 - 500  µL (<5 µL)  200 μL  100 - 500 μL  500 μL  20 μL  per well  1 - 10 µL per  well  Accuracy   -  -  92.8-112 %  Inter-  assay:   87-96 %  Intra-  assay:   80-92 %  -   92 - 99 %  85 -127 %  0.96 - 22 ppm  Very  good  -  Precision   Inter-  assay:  4.0-5.0 %  Intra-  assay:  2.8-4.0 %  Inter-  assay:  5.4-6.0 %  Intra-  assay:  4.2-5.3 %  Inter-  Assay:  3.0-13.0 %  Intra-  assay:   1.27-14.0%  -  12 - 19 %  6-25%  Inter-  assay:   6.8-22.5%  Intra-  assay:   2.2-10.7%  Inter-  assay: <8%  Inter-  assay:  7.6% -  9.8%   Intra-  assay :  3.4-4.3%   Inter-  assay:   7.5-12.3 %  Intra-  assay:   2.9-8.5 %   

Sensitivity   -  -  ≤50pg/mL  -  Good  Good  High  High  - 

Specificity   D  D  A  A  B  A  A  D  D 

Measure  range  3 - 200  mU/L  0.15 - 20  mU/L    38.5 -  38500 pg/ mL   -  0 - 4.0  ng/mL  See table 4.1  1.5 - 1920  pmol/L  -  0.8 - 3000  µlU/mL  Through-  put  Incubation  60+15 min  Incubation  60+30 min    LC: <8 min  per sample  25  samples/  day.   35 min/  high  sensitivity  assay  12 min/  sample   IA: 1 - 2 h  LC-MS/MS:   4 - 33 min  High  Medium  through -put  3 h 

Sample cost  -  -  -  -  Low  -  -  ≈ 2 SEK/ 

sample 

(23)

(0.43  μg/L)  μ mU/L  (0.0065  μg/L)  (10.8  pmol/L)  ng/mL  LOQ  -  -  ≥ 38.46  pg/mL  200 pg/mol  0.3 ng/mL  0.5 ng/mL  Alt.   0.78  pmol/L  1.5 - 15  pmol/L  0.345  ng/mL  -  Recovery  Upon  addition  94-113 %  Upon  addition  97-107 %  68-113 %  -   64 %  33-44 %  alt.   76-108 %   -  104-126  %  94-112 % 

6.2 Proinsulin 

Table 2. Values in the respective fields are assembled from the corresponding proinsulin tables for each method  in Appendix 2. Upon different values in the same field there were no clear consensus among the same methods  in the tables found in Appendix 2. 

 

  ELISA  LC-MS/MS  UPLC-MS/MS  LC-IM/MS  IA-LC-MS/MS  MSIA-HR-A

HTRF  AlphaLISA​®   Sample  volume  50 μL per well  -  -  -  100 μL  -  -  -  Accuracy  -  -  -  -  85 - 127 %  -  -  -  Precision  Intraassay:   2.5 - 3.2 %  Interassay:   5.0 - 6.1 %  -  -  -  6 - 25 %  -  -  -  Sensitivity  -  -  -  -  ~0.05 ng/mL  -  -  -  Specificity  B  -  -  -  A  -  -  -  Measure  range  3.3 - 130  pmol/L  -  -  -  -  Through-  put  Incubation  60+60+15 min  -  -  -  IA: 1 - 2 h  LC-MS  /MS:   ~33 min  -  -  -  Sample cost  -  -  -  -  Multi-  plexing  No  -  -  -  Yes  -  -  -  LOD  1.7 pmol/L  -  -  -  ~0.05 ng/mL  -  -  -  LOQ  -  -  -  - 

Recovery  Upon dilution: 

(24)

6.3 C-peptide 

Table 3. Values in the respective fields are assembled from the corresponding C-peptide tables for each method  in Appendix 2. Upon different values in the same field there were no clear consensus among the same methods  in the tables found in Appendix 2. 

    ELISA  ELISA  Ultrasensitiv e   C-peptide  LC-MS/MS  UPLC-MS/ MS  LC-IM/MS  IA-LC-MS/ MS  MSIA-HR-A HTRF  AlphaLISA​®   Sample  volume  25 μL per  well  50 μL per well  -  -  -  100-150 µL  -  -  1-10 µL 

Accuracy  -  -  -  85 - 127 %  -  Very good  - 

Precision  Intra-  assay:  < 6.8 %  Inter-  assay:  < 4.8 %  Intra-  assay:  3.9-6.2 %  Inter-  assay:   2.2-5.3 %  -  -  -  4-25 %  -  Intra-  assay:   3.5-5.0 %  Inter-  assay:  4.9-8.8 %   Intra-  assay:  3.9-4.8 %  Inter-  assay:  7.7-11.8 % 

Sensitivity  -  -  -  Good  -  High  Good 

Specificity  C  C  -  -  -  A  -  C+  B  Measure  range  100-4000  pmol/L   (0.3-12.0  ng/mL)  5-280 pmol/L   (0.015-0.846  ng/mL)  -  -  -  0.11 - 27.7  ng/ml  -  -  23.5 -   30 000  pg/mL  Through-  put  Incu-  bation  60+60+15  min  Incu-  bation  60+60+30 min  -  -  -  IA: 1 - 2 h  LC-MS  /MS:  4 - 33 min   -  -  3 h  Sample  cost  -  -  -  ~ 20 - 30  SEK/  sample  Multi-  plexing 

No  -  -  -  -  Yes  -  No  No 

(25)

6.4 Glucagon 

Table 4. Values in the respective fields are assembled from the corresponding glucagon tables for each method  in Appendix 2. Upon different values in the same field there were no clear consensus among the same methods  in the tables found in Appendix 2. 

 

  ELISA  LC-MS/MS  UPLC-MS/MS  LC-IM/MS  IA-LC-MS/MS  MSIA-HR-A

HTRF  AlphaLISA​®   Sample  volume  25 μL per  well  200-250 μL  (<5 µL)  400 µL  (<5 µL)  -  500 µL    -  -  1 - 10 µL  Accuracy  -  Interassay:  <4.0 %  Intraassay::  <12 %  68.6 - 87.4 %    -  -  -  -  -  Precision  Intraassay:  3.3 - 5.1 %  Interassay:  7.5 - 9.5 %  Intraassay:  <13 %   Interassay:  <14 %  Intra- and  interassay  >20 %  -  Intraassay:   2.2 - 10.7 %  Interassay:   6.7 - 22.5 %  -  -  Intraassay: 12%  Interassay: 13% 

Sensitivity  1 pmol/L  Good  High  -  Good   -  -  Good 

Specificity  C  B  A  -  A  -  -  D 

Measure  range  1.5 - 120  pmol/L   0.5 - 100  pmol/L  alt.  10 – 10 000  pg/mL  15 - 1000  pg/mL  -  Detection  range:   2 - 750 mg/dL   -  -  20 - 30 000  pg/mL  Throughput  18-22 h  (overnight) +  15 min  substrate  incubation  -  3.6 - 5 min/  sample 

-  IA: 1h & 15 min  

LC-MS/MS:  ~13 min  

-  -  3 h 

Sample cost  -  -  Low/very low  -  -  -  -  ≈ 20 - 30 SEK/ 

sample  Multi- 

plexing 

No  -  Yes  -  Yes  -  -  No 

(26)

7. Remarks regarding comparison of the methods 

Here we discuss the results between the seven methods and ELISA in respect to the  different parameters within Table 1-4. There is also a short economic discussion regarding  the prices of the different methods.  

 

The measuring ranges in our results were not consistently documented. Quantification  ranges are more common than detection ranges and thus used for comparing measuring  range. Also, LLOD (lowest limit of detection) is documented more frequently than ULOD  (upper limit of detection) and serves as a guideline for sensitivity comparisons which are  more relevant to the studied biomarkers as they occur in low concentrations. 

 

The number of samples used in the different experiments is shown in tables in Appendix 2,  where the information was available. This information is not compiled in the result tables  since the number of samples are different between the articles and therefore difficult to  compile into an interval.

 

7.1 Comparison Between ELISA and LC-MS Methods  

From the various articles read across this project, some clear strengths and weaknesses for  both ELISA and LC-MS/MS methods have been noted. As described in many of the articles,  and elegantly summarised by Cross & Hornshaw (2016)​,​

ELISAs main strength lies in the  high amount of sensitivity that can be achieved. This without the need of any expensive  equipment, while at the same time being fairly easy to use. Most of the articles use ELISA to  verify the results of other methods. 

7.1.1 Accuracy for LC-MS/MS 

As described in the result, accuracy is a parameter of great importance to the comparison  since it defines reliability. The accuracy depends on both sensitivity and specificity as it  includes aspects of detecting the right compound as well as the right amount. While  LC-MS/MS methods can achieve a high specificity, one of the greater problems is the  method sensitivity. This is due to poor fragmentation, formation of multiple charged ions,  and matrix effects such as adsorption of analytes to the containers and instability of its  compounds (Lapko ​et al.​ 2013). However, a high sensitivity can be achieved with 

(27)

LC-MS/MS offers a more accurate quantification with less false positives than other  immunochemical methods such as ELISA (Cross & Hornshaw 2016). This technology is  quickly developing to be a powerful technique in the field of quantifying peptides,  previously only analysed by immunochemical assays (Lapko ​et al.​ 2013). 

 

 

However, in Table 1, 2, 3 and 4 it is shown that ELISA overall has better recovery rates  indicating higher accuracy than LC-MS/MS based methods. The conflicting results can be  partially explained by the fact that accuracy is not that well documented in most studies,  and for some biomarkers not studied at all. In general, the recovery rates were either very  much lower than for ELISA, or relatively close but broader and less precise.

 

 

For immunochemistry-based methods, specificity is depending on antibody-antigen  interactions and thus different interaction conditions will affect specificity. MS-based  methods may use immunoreactions but only for the purpose of enriching the sample,  allowing a more concentrated sample to reach the MS-step. The immunoaffinity step is not  specificity-determinant. LC-MS/MS methods have high specificity due to having a 

detection mechanism where the mass/charge ratio is a very specific characteristic, it does  require a spectral library in order to map the right peak to the right molecule. 

7.1.2 Automatisation and multiplexing 

Modern instruments allow LC-MS/MS workflow to be more automatised, which reduces  the influence of the human error. Automatisation makes it possible to run many samples  unsupervised which increases the throughput, lets the system perform with minor sample  volumes and comes with high intra- and interassay reproducibility (Cross & Hornshaw  2016). In addition, the successful development of LC-MS/MS has given the method the  quality of multiplexing, an advantage that is lacking in antibody-based methods.

 

 

Features discussed above are offered by LC-MS/MS methods due to the multiple 

instrumentation that forms the system. The workflow might require supervision and the  results gained are often complicated graphs that need training to be understood. In other  words, these methods require qualified personnel with experience in the subject, 

something that might be hard to find. 

 

7.1.3 Sample preparation with respect to complex matrices 

When it comes to complexity of a sample, it can affect the analytical correctness and  reproducibility of an experiment when using a LC-MS/MS method. Without sufficient  sample preparation, matrix effects will occur. Therefore, it is important with a thorough,  sample preparation to get a good signal-to-noise ratio. This is of utter importance when  working with complex matrices such as plasma and serum (Smeraglia ​et al.​ 2002). 

(28)

LC-MS/MS. However, the reference material used for calibrating the detection curve with  ELISA needs to be chosen with care to avoid misdetection among analogs or isoforms. The  used antibody need to have corresponding specificity towards both the analyte and the  standards used as reference (Lapko ​et al.​ 2013). The secondary structure might vary among  different references due to minor changes in primary structure, which affects 

immunochemistry-based methods but not MS-based ones, as quantification relies solely on  the integrity of the analytes primary structure. 

 

 

In immunoaffinity LC-MS/MS, antibodies are used in the purification step of the analysis.  This combines the specificity of the LC-MS/MS method with the sensitivity of the 

immunoaffinity, making it possible to match the sensitivity of current immunoassays  (Taylor ​et al.​ 2016). A drawback with this method is the cost of antibodies and the  incubation time of the sample preparation, which affects the throughput of the method  negatively.

 

7.1.4 UPLC-MS/MS 

Results referring to UPLC-MS/MS are shown in Table 1 and 4. It has the disadvantage of  not covering C-peptide nor proinsulin. However, with the existing data for glucagon and  insulin, it has the highest specificity score and a quite low cost per sample. In Table 4 we  see that the method presents a high sensitivity and a LOQ of 15 pg/mL and a wide 

measuring range of 15 - 1000 pg/mL. It is debatable whether an accuracy from 68.6 - 87.4  % is good because of the lack of information from ELISA. Nonetheless, it is an advantage to  avoid using antibodies due to cross-reactivity and the false positives that they might 

contribute to. ELISA in general had a poorer detection range than the LC-MS/MS methods.  UPLC-MS/MS was shown to have the best overall detection range with very low LOQ  values.

 

7.1.5 LC-IM-MS 

During this project we were pleasantly surprised about the great potential of LC-IM-MS.  Unfortunately, very little information was found about this method, specially for the 

biomarkers of interest. The method showcases better precision than several other methods  with the lowest LOQ in our results, see Table 1. LC-IM-MS has potential to give great 

results in the future, tentatively in combination with bottom up approach, and therefore  this method should be more investigated for biomarker quantification.

 

7.1.6 Availability of data about the biomarkers among the methods 

(29)

for its absence in the studies. (Thomas ​et al.​ 2017)

 

 

Another difficult biomarker to quantify is C-peptide which showed to be difficult to analyse  by the LC-MS/MS methods. Only IA-LC-MS/MS was able to detect it among the LC-MS  based methods. This is most likely because IA-LC-MS/MS utilises an immunochemical  separation step which the other LC methods do not. We do not otherwise know what  factor makes it hard to find via LC-MS methods. 

 

7.2 Comparison Between ELISA and other immunochemical 

methods 

ELISA has better recovery rate in comparison to the other methods, although AlphaLISA​®  

and ELISA performed almost identically when screening for insulin and had comparable  results for C-peptide. AlphaLISA​® had a slightly worse performance than ELISA when 

analysing glucagon. HTRF was also similar but in general had larger intervals in recovery  percentages than ELISA. 

7.2.1 AlphaLISA​

®

 

When comparing AlphaLISA​® and ELISA, the former has a lower sample volume and is 

faster due to the washing-steps not being required. In addition, AlphaLISA​® has lower 

cross-reactivity to proinsulin than some commercially available ELISA kits when analysing  C-peptide. This makes AlphaLISA​® a method that potentially can outshine ELISA. Yet, there 

does not exist any commercially available AlphaLISA​® kits for proinsulin, while it does exist 

for ELISA. ELISA is a broader method with the advantage that it can be used on biomarkers  that are not covered by AlphaLISA​® . AlphaLISA​® also requires a special laser instrument for 

detection, making the method more expensive to begin with.   

When compared, Table 3 presents the need of 25 to 50 µL of the sample for ELISA while  AlphaLISA​® only needs 10 µL to quantify C-peptide. The latter method also has a better 

specificity and faster throughput for the same biomarker. Table 4 shows that ELISA needs  25 µL in sample volume while AlphaLISA​® still only needs 10 µL to quantify glucagon. In 

addition, AlphaLISA​® has faster throughput while ELISA has a better specificity.  

7.2.2 HTRF 

The major differences between ELISA and HTRF are mainly price, time of execution and  sample preparation. HRTF is a faster analytic method. The whole process takes half the  time compared to ELISA, it only has about 5 steps while ELISA has approximately 13. 

(30)

 

We believe that every immunochemical method mentioned in this report should in theory  work for all the same biomarkers as ELISA does, since they all operate based on antibodies.  Although, sufficient data could not always be found, such as for HTRF on proinsulin and  glucagon.  

7.2.3 Hook effect in immunochemical methods 

Articles studied during this project showed an inaccurate specificity for all the 

immunoassay methods. This could be a consequence of cross-reactivity, which causes false  positives or of the “Hook effect” which causes false negatives. Hook effects occurs when  the concentration of antibodies is much greater than the concentration of analytes. All the  antigens will bind to one pair of antibodies until there is no free antigens left. This high  concentration of unbound antibodies will bind to the antigens in solution and will be  washed away in the final washing step. This leads to underestimation of the true antigen  concentration as some are removed from the sample. The Hook effect only occurs when  the sandwich principle is used, and it is named after the point where the solution starts to  be saturated, the so-called Hook point. (Einhorn & Krapfenbauer 2015) 

7.3 Economics 

When it comes to expenses, ELISA is the method least dependent of equipment and thus  require no large investment in order to be used. This comes at the price of having the  highest sample cost out of all the methods that were researched. In contrast, most of the  LC-MS/MS methods have a very low sample cost but all require expensive equipment to be  used. HTRF and AlphaLISA​® can be considered the middle ground, where they require 

some equipment but offer a lower sample cost than ELISA, in the case of HTRF even a  significantly lower sample cost. An interesting note is that the articles we found stated a  price range of 2 - 8.4 SEK. These ranges were for the material costs only, if one were to  perform the method independently (Einhorn & Krapfenbauer 2015). In contrast, some ready  to use HTRF kit products that were found online could have a cost per sample as high as 28  SEK and we found none cheaper than 11.5 SEK. To reach that price you had to buy a kit of  10 000 wells for 115 000 SEK, which is much too expensive for most studies. 

 

 

(31)

8. Conclusion  

To conclude, our results indicate that advantages of immunoassays include great recovery  and precision, in particular when it comes to ELISA. The availability of these diverse 

commercial kits strengthens the expanse of the methods which is manifested in our  results. However, parameters as specificity and throughput are proved to be superior for  LC-MS/MS methods.

 

 

In addition, accuracy, LOD and LOQ are also better for LC-MS/MS methods when  compared to immunoassays, which makes the methods an alternative to ELISA. 

Multiplexing is a great advantage of LC-MS/MS. The cost per sample is another asset, but  this advantage comes at the expense of higher equipment prices. Furthermore, it requires  more expertise to interpret the results from these methods.  

 

In summary, LC-MS/MS methods are extensively supported by scientific articles reporting  on biomarker detection and quantification. The methods demand streamlining and 

standardisation for each analyte and ELISA will be used to evaluate reliability of LC-MS/MS  for years to come.

 

 

So, the question remains: Immunoassay or LC-MS/MS? Our answer is that it depends on  the purpose of the quantification and the requirements for each parameter.  

(32)

9. Ethical Discussion  

This is meant as an additional section to raise the readers awareness about potential  ethical dilemmas that could occur within the fields of biomarker detection. It also touches  upon some problems that Mercodia AB might have to deal with in the future in case there  is a big shift into LC-MS/MS technologies within the research field.  

9.1 Production of Antibodies  

Antibodies are truly valuable tools in research such as detection and quantification of  biomarkers in complex samples and treatment of, for example, immune deficiencies. There  are many advantages of using antibodies for these applications due to their high sensitivity  and specificity. But there are some disadvantages, especially during the production of the  antibodies. To produce polyclonal antibodies, one need to inject a chosen substance into  laboratory or farm animals such as mice, rabbits or horses. The immune systems of the  animal then starts to produce antibodies directed against the injected substance. The issue  of using animals to obtain antibodies is that the antibodies become species-specific, 

leading to the need of being humanised before the antibodies can be used for human  samples. The binding parts of the antibodies will keep its animal origin but the non-binding  parts are exchanged for human types. (Fredholm. 2007)  

 

To collect the antibodies the animal is first heavily anaesthetised, then a needle is inserted  directly into the heart. When no more blood can be extracted the animal is put down with a  lethal injection of pentobarbitone (​Animalresearchinfo. 2014).​ Some may argue that this is a  harsh treatment of animals and some may say that it is pain-free and necessary. One thing  is certain: ​we are finding ourselves crossing into an animal rights discussion for research  purposes. According to ​the Swedish Animal Protection Act a strict requirement for animal  use is that the research can not be accomplished in any other satisfactory manner without  the use of animals (Riksdagsförvaltningen. 2018).  

  

From the production method described above we receive polyclonal antibodies. These  antibodies come from different B-cells that can recognise multiple epitopes on the same  antigen. Monoclonal antibodies represent, in contrast to polyclonal antibodies, antibodies  that only bind to a specific epitope on an antigen which make them favorable for 

(33)

survive the fusion process (​Denovo Biotechnology. 2017​). There is also some benefits with  polyclonal antibodies because they are more tolerant of minor changes in the antigen. 

9.2 Diagnostics or Research Only  

The kits produced by companies such as Mercodia AB has the main purpose of being used  for research and not for clinical diagnostic purposes. Nevertheless, the products are being  used for diagnostic purposes in some places around the world and therefore reliability is  essential to avoid misdiagnosed patients. In such cases, companies like Mercodia AB could  be argued to have a responsibility to produce reliable products when it comes to clinical  diagnostics as well, ​even though that is not their initial purpose. However, it is debatable  whether the producer carries an ethical responsibility rather than judicial for how their  products are applied by the clients. Since there is an interest for diagnostic methods, by  expanding the area of use, the company would probably gain customers and also probably  profit a lot from it.  

 

In this report we have compared many different technologies and methods for detection  and quantification of biomarkers. Some of them seem to be better than ELISA and could  therefore be better suited for diagnostic purposes. However, some of them require 

expensive equipment and a lot of experience which probably would not make it suitable for  diagnostic purposes. The cost for reliable tests may not be a price everyone is able to pay  which further divides the rich from the poor and increases the social gaps. So, what is a  company, such as Mercodia AB, supposed to do? Stay with the same technology and try to  make it more available, or go for the already more advanced technologies and join the  future?  

9.3 New Standard Technology for Detection and 

Quantification  

Let us say for the sake of discussion that we found out through our research that the best  way to detect and quantify biomarkers in complex matrices is through a form of 

LC-MS/MS method. These are methods that, once again, require expensive equipment that  not all research facilities have access to nor can afford. An UHPLC (second hand) can cost  up to 400 000 SEK (Liquid Chromatographs (HPLC), ​Conquer Scientific. 2019​) and a mass  spectrometer-LCMS system (second hand) can cost over 1 000 000 SEK (Mass 

(34)

equipment regularly. Many small research facilities may not have the funds to do this and  therefore will not be able to perform the same high-quality experiments.   

 

If this became the new standard for all kits that companies like Mercodia AB would adapt  their methods for, then it could potentially cripple small research facilities that cannot  afford the required equipment. They would perhaps be forced to send their samples for  analysis to a contract research organisation such as Sci Life in Sweden, and that may  increase the cost and could affect the reliability of the result due to the changes of  environment. This may also mean a loss of customers for companies that would start  developing kits customised for LC-MS/MS methods instead of for example ELISA.  

9.4 Production and Transport 

(35)

10. Acknowledgements 

The authors would like to thank:   

Our supervisor, Lena Henriksson, for guidance throughout the project.   

Lecturers Elisabeth Sköld and Lennart Köhler for instructions in project organization and  report writing. 

 

Representative of Mercodia AB, Johan Ledin for answering our questions and providing  useful feedback. 

 

Mercodia AB for the project and an interesting site visit.   

Last but not least, a big thanks to our fellow students on the course for valuable responsive  comments on our report.  

11. Statement of Contribution 

All authors have conducted the literature study together in which the data was gathered.  All authors have been taking active parts in discussing all aspects of the report and given  feedback to one another. All authors have had a hand in the formation of each section in  this report.  

 

C. Tiger was the main contributor to the background section of the report and wrote the  technology sections on ELISA and HTRF. He created fig 1-3 and 7 and wrote figure texts for  figures beside 2 and 3.  

 

A. Ivert has contributed to the report by writing the IA-LC-MS/MS method description and  the ethical discussion. She also designed the tables. 

 

T. Martinez has written the description of the method UPLC-LC/MS and data collection of  HTRF. She also contributed with the workflow, the explanation of parameters and the  conclusion. 

 

R. Upite has contributed with the MSIA-HR/AM method description, figure 6 and 

collecting data for this method used in the comparisons. She also compiled the “Vocabulary  and Abbreviations”. 

(36)

V. Umenius has contributed to this report with the method description for LC-MS/MS and  the figure texts for figure 2 and 3. He also contributed with the data collection of 

LC-MS/MS.   

S. Wärmegård contributed mostly to the method description and data collection for 

LC-IM-MS and AlphaLISA​® . She also created figure 8, contributed to the explanation of the 

parameters and designed the tables. 

(37)

12. References 

12.1 Articles 

 

Blackburn M. 2013. Advances in the quantitation of therapeutic insulin analogues by  LC–MS/MS. Bioanalysis 5: 2933–2946. 

Carslake HB, Pinchbeck GL, McGowan CM. 2017. Evaluation of a Chemiluminescent  Immunoassay for Measurement of Equine Insulin. Journal of Veterinary Internal  Medicine 31: 568–574. 

Chambers EE, Fountain KJ, Smith N, Ashraf L, Karalliedde J, Cowan D, Legido-Quigley C.  2014. Multidimensional LC-MS/MS Enables Simultaneous Quantification of Intact  Human Insulin and Five Recombinant Analogs in Human Plasma. Analytical 

Chemistry 86: 694–702. 

Chen Z, Caulfield MP, McPhaul MJ, Reitz RE, Taylor SW, Clarke NJ. 2013. Quantitative  Insulin Analysis Using Liquid Chromatography-Tandem Mass Spectrometry in a  High-Throughput Clinical Laboratory. Clinical Chemistry 59: 1349–1356. 

Cross TG, Hornshaw MP. 2016. Can LC and LC-MS ever replace immunoassays? Journal of  Applied Bioanalysis 2: 108–116. 

Dong S, Gu Y, Wei G, Si D, Liu C. 2018. Direct comparison of LC–MS/MS and RIA methods  for the pharmacokinetics assessment of human insulin in preclinical development.  Biomedical Chromatography 32: e4323. 

Dufield DR, Radabaugh MR. 2012. Online immunoaffinity LC/MS/MS. A general method to  increase sensitivity and specificity: How do you do it and what do you need? 

Methods 56: 236–245. 

Einhorn L, Krapfenbauer K. 2015. HTRF: a technology tailored for biomarker 

determination—novel analytical detection system suitable for detection of specific  autoimmune antibodies as biomarkers in nanogram level in different body fluids.  The EPMA Journal, doi 10.1186/s13167-015-0046-y. 

Farino ZJ, Morgenstern TJ, Vallaghe J, Gregor N, Donthamsetti P, Harris PE, Pierre N,  Freyberg R, Charrier-Savournin F, Javitch JA, Freyberg Z. 2016. Development of a  Rapid Insulin Assay by Homogenous Time-Resolved Fluorescence. PLOS ONE 11:  e0148684. 

Fredholm L. 2007. Antikroppar som läkemedel. WWW-dokument 2007-12-01: 

(38)

Guillarme D, Veuthey J-L. 2017. Chapter 1 - Theory and Practice of UHPLC and UHPLC–MS.  I: Holčapek M, Byrdwell WmC (red.). Handbook of Advanced Chromatography/Mass  Spectrometry Techniques, s. 1–38. AOCS Press,  

Henry H, Lannoy D, Simon N, Seguy D, D’Herbomez M, Barthélémy C, Décaudin B, Dine T,  Odou P. 2017. Immunoassay quantification of human insulin added to ternary  parenteral nutrition containers: comparison of two methods. Analytical and  Bioanalytical Chemistry 409: 3677–3684. 

Heyduk E, Moxley MM, Salvatori A, Corbett JA, Heyduk T. 2010. Homogeneous Insulin and  C-Peptide Sensors for Rapid Assessment of Insulin and C-Peptide Secretion by the  Islets. Diabetes 59: 2360–2365. 

Howard JW, Kay RG, Jones B, Cegla J, Tan T, Bloom S, Creaser CS. 2017. Development of a  UHPLC–MS/MS (SRM) method for the quantitation of endogenous glucagon and  dosed GLP-1 from human plasma. Bioanalysis 9: 733–751. 

Howard JW, Kay RG, Tan T, Minnion J, Ghatei M, Bloom S, Creaser CS. 2014. Development  of a high-throughput UHPLC–MS/MS (SRM) method for the quantitation of  endogenous glucagon from human plasma. Bioanalysis 6: 3295–3309. 

Kanu AB, Dwivedi P, Tam M, Matz L, Hill HH. 2008. Ion mobility–mass spectrometry.  Journal of Mass Spectrometry 43: 1–22. 

Kinumi T, Mizuno R, Takatsu A. 2014. Quantification of serum C-peptide by isotope-dilution  liquid chromatography–tandem mass spectrometry: Enhanced detection using  chemical modification and immunoaffinity purification. Journal of Chromatography  B 953–954: 138–142. 

Lapko VN, Miller PS, Brown GP, Islam R, Peters SK, Sukovaty RL, Ruhn PF, Kafonek CJ. 2013.  Sensitive glucagon quantification by immunochemical and LC–MS/MS methods.  Bioanalysis 5: 2957–2972. 

Lapthorn C, Pullen F, Chowdhry BZ. 2013. Ion mobility spectrometry-mass spectrometry  (IMS-MS) of small molecules: Separating and assigning structures to ions. Mass  Spectrometry Reviews 32: 43–71. 

Lee AYH, Chappell DL, Bak MJ, Judo M, Liang L, Churakova T, Ayanoglu G, Castro-Perez J,  Zhou H, Previs S, Souza SC, Lassman ME, Laterza OF. 2016. Multiplexed 

Quantification of Proglucagon-Derived Peptides by Immunoaffinity Enrichment and  Tandem Mass Spectrometry after a Meal Tolerance Test. Clinical Chemistry 62:  227–235. 

Legg KM, Labay LM, Aiken SS, Logan BK. 2019. Validation of a Fully Automated 

(39)

double-antibody sandwich ELISA for rapid detection to C-peptide in human urine.  Journal of Pharmaceutical and Biomedical Analysis 162: 179–184. 

Miyachi A, Kobayashi M, Mieno E, Goto M, Furusawa K, Inagaki T, Kitamura T. 2017.  Accurate analytical method for human plasma glucagon levels using liquid  chromatography-high resolution mass spectrometry: comparison with 

commercially available immunoassays. Analytical and Bioanalytical Chemistry 409:  5911–5918. 

Moser AC, Hage DS. 2010. Immunoaffinity chromatography: an introduction to applications  and recent developments. Bioanalysis 2: 769–790. 

Nedelkov D, Niederkofler EE, Oran PE, Peterman S, Nelson RW. 2018. Top-down mass  spectrometric immunoassay for human insulin and its therapeutic analogs. Journal  of Proteomics 175: 27–33. 

Oran PE, Jarvis JW, Borges CR, Sherma ND, Nelson RW. 2011. Mass spectrometric  immunoassay of intact insulin and related variants for population proteomics  studies. PROTEOMICS – Clinical Applications 5: 454–459. 

Peterman S, Niederkofler EE, Phillips DA, Krastins B, Kiernan UA, Tubbs KA, Nedelkov D,  Prakash A, Vogelsang MS, Schoeder T, Couchman L, Taylor DR, Moniz CF, Vadali G,  Byram G, Lopez MF. 2014. An automated, high-throughput method for targeted  quantification of intact insulin and its therapeutic analogs in human serum or  plasma coupling mass spectrometric immunoassay with high resolution and  accurate mass detection (MSIA-HR/AM). PROTEOMICS 14: 1445–1456. 

Pitt JJ. 2009. Principles and Applications of Liquid Chromatography-Mass Spectrometry in  Clinical Biochemistry. The Clinical Biochemist Reviews 30: 19–34. 

Riksdagsförvaltningen. 2018. Djurskyddslag (2018:1192) Svensk författningssamling  2018:2018:1192 - Riksdagen. WWW document: 

https://www.riksdagen.se/sv/dokument-lagar/dokument/svensk-forfattningssa mling/djurskyddslag-20181192_sfs-2018-1192. Accessed 4 June 2019. 

Sakamoto S, Putalun W, Vimolmangkang S, Phoolcharoen W, Shoyama Y, Tanaka H,  Morimoto S. 2018. Enzyme-linked immunosorbent assay for the 

quantitative/qualitative analysis of plant secondary metabolites. Journal of Natural  Medicines 72: 32–42. 

Simmons E, Johnson D, Delhaye R, Colwell J, Clor E, Santa P, Bowsher RR. 2019. 

(40)

Smeraglia J, Baldrey SF, Watson D. 2002. Matrix effects and selectivity issues in LC-MS-MS.  Chromatographia 55: S95–S99. 

Taylor SW, Clarke NJ, Chen Z, McPhaul MJ. 2016. A high-throughput mass spectrometry  assay to simultaneously measure intact insulin and C-peptide. Clinica Chimica Acta  455: 202–208. 

Thomas A, Brinkkötter PT, Schänzer W, Thevis M. 2017. Simultaneous determination of  insulin, DesB30 insulin, proinsulin, and C-peptide in human plasma samples by  liquid chromatography coupled to high resolution mass spectrometry. Forensic  Toxicology 35: 106–113. 

Thomas A, Schänzer W, Thevis M. 2014. Determination of human insulin and its analogues  in human blood using liquid chromatography coupled to ion mobility mass 

spectrometry (LC-IM-MS). Drug Testing and Analysis 6: 1125–1132. 

Warnken T, Huber K, Feige K. 2016. Comparison of three different methods for the  quantification of equine insulin. BMC Veterinary Research, doi 

10.1186/s12917-016-0828-z. 

Whiteaker JR, Paulovich AG. 2011. Peptide immunoaffinity enrichment coupled with mass  spectrometry for peptide and protein quantification. Clinics in laboratory medicine  31: 385–396. 

12.2 Online sources 

Animalresearchinfo. 2014. Antibody production. WWW-document 11-11-2014: 

http://www.animalresearch.info/en/designing-research/experimental-design-an alysis/antibody-production/​. ​Accessed ​28 May 2019. 

 

CBRNE Tech Index. 2018. Ion Mobility Spectrometry (IMS). WWW-document: 

http://www.cbrnetechindex.com/Chemical-Detection/Technology-CD/Spectrom etry-CD-T/Ion-Mobility-Spectrometry-CD-S​. ​Accessed ​28 May 2019. 

 

Conquer Scientific. 2019. Liquid Chromatographs (HPLC). WWW-document: 

https://conquerscientific.com/lab-equipment/liquid-chromatographs-hplc/​.  Accessed ​4 June 2019. 

 

Conquer Scientific. 2019. Mass Spectrometers-LCMS Systems. WWW-document: 

https://conquerscientific.com/lab-equipment/mass-spectrometers-lcms-systems /​. ​Accessed ​4 June 2019. 

 

(41)

Denovo Biotechnology 2017. What are the advantages and disadvantages of using  Monoclonal antibodies Vs Plyclonal antibosies? WWW-document: 

https://www.denovobiotech.com/support-center/view-article/2-what-are-the-a dvantages-and-disadvantages-of-using-monoclonal-antibodies-vs-polyclonal-antib odies​. ​Accessed ​28 May 2019. 

 

Eurofins (1). 2019. HTRF kit 10 000 wells. WWW-document: 

https://www.eurofinsdiscovery.com/33-036-pi-3-kinase-htrf-assay-10-000-wells Accessed 5 June 2019. 

 

Eurofins (2). 2019. HTRF kits 384 wells. WWW-document: 

https://www.eurofinsdiscovery.com/33-016-33-016-pi-3-kinase-htrf-assay-384-wells​ Accessed 5 June 2019. 

 

LabX. 2019. Mass spectrometer Listings. WWW-document: 

https://www.labx.com/mass-spectrometer​. ​Accessed ​28 May 2019.   

Pacific Immunology. 2019. WWW-documents: 

https://www.pacificimmunology.com/resources/antibody-introduction/polyclon al-vs-monoclonal-antibodies/​. Accessed 7 June 2019. 

  

PerkinElmer. 2011. TDS – Human Insulin. WWW-document: 

https://www.perkinelmer.com/lab-solutions/resources/docs/TDS_AlphaLISA_A L204.pdf​. ​Accessed ​28 May 2019 

 

PerkinElmer. 2012. TDS – Human Connecting Peptide (C-peptide). WWW-document:  https://www.perkinelmer.com/lab-solutions/resources/docs/TDS_AlphaLISA_AL299.p

df​. ​Accessed ​28 May 2019.   

PerkinElmer. 2019. Alpha Reagents. WWW-document: 

https://www.perkinelmer.com/se/category/alpha-reagents​. Accessed ​28 May  2019.  

 

PerkinElmer. 2012. TDS – Glucagon AlphaLISA Immunoassay Kit. WWW-document: 

http://www.perkinelmer.com/lab-solutions/resources/docs/TDS_AlphaLISA_AL 312.pdf​ ​Accessed ​28 May 2019. 

 

Thermo Fisher Scientific​TM​. 2019. MSIA Microcolumns. WWW-document:  

References

Related documents

In Paper VI, the modified COW algorithm was used as a pre-processing tool for PARAFAC modelling of the LC-MS data obtained for five runs (#a-#e) of a peptide standard.. 18 shows

Therefore, in order to improve our understanding on MeHg chemical speciation, we developed a novel analytical method for direct determination of MeHg complexes

139 Essentially, what has been described above as a social struggle over the idea of a human right to food, situated in the sphere of global civil society and the force field

To create a smooth hair strand curve based on the control points of the guide strands, the control points are used to evaluate the guide curves at a discrete number of positions

The study sequence (in bold) appears together with tick species of the I. A) Phylogenetic comparison of partial Ixodes 16S rRNA gene sequences inferred by using the Maximum

Detta skulle kunna leda till stora kostnader eller att drönarna inte kommer tillbaka i tid restaurangerna..

tolkningar av olika personers berättelser från att mötas i vården. Dock möjliggjorde den valda metoden att inkludera ett mer heterogent urval med fler representanter från varje

Data analysis and metabolite annotation is one of the most time consuming and challenging steps in any metabolomics study and the reduced cluster and adduct formation, achieved