+ All Categories
Home > Documents > Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan...

Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan...

Date post: 17-Apr-2020
Category:
Upload: others
View: 5 times
Download: 0 times
Share this document with a friend
6
Immunohistochemistry Microarrays Huiyan Li, ,Gabrielle Brewer, ,Grant Ongo, ,Frederic Normandeau, ,Atilla Omeroglu, § and David Juncker* ,,,Biomedical Engineering Department, McGill University and Genome Quebec Innovation Centre, § Department of Pathology, McGill University Health Centre, and Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A0G1, Canada * S Supporting Information ABSTRACT: Immunohistochemistry (IHC) on tissue sections is widely used for quantifying the expression patterns of proteins and is part of the standard of care for cancer diagnosis and prognosis, but is limited to staining a single protein per tissue. Tissue microarray and microuidics staining methods have emerged as powerful high throughput techniques, but they either only permit the analysis of a single protein per slide or require complex instrumentation and expertise while only staining isolated areas. Here, we introduce IHC microarrays (IHCμA) for multiplexed staining of intact tissues with preserved histological and spatial information. Droplets of a dextran solution containing antibodies were prespotted on a slide and snapped onto a preprocessed formalin-xed, paran-embedded (FFPE) tissue section soaked in a polyethylene glycol solution. The antibodies are conned within the dextran droplets and locally stain the tissue below with a contrast similar to the one obtained by conventional IHC. The microarray of antibody droplets can be prespotted on a slide and stored, thus neither the preparation of the antibody solutions nor a sophisticated microarray spotter is needed. Sampling considerations with IHCμA were evaluated by taking three tissues with varying levels of cancer cells. A multiplex IHCμA with 180 spots targeting 8 cancer proteins was performed on a breast cancer tissue section to illustrate the potential of this method. This work opens the avenue of applying microarray technologies for conducting IHC on intact tissue slices and has great potential to be used in the discovery and validation of tissue biomarkers in human tumors. A nalysis of protein biomarkers in tumor tissues can help in cancer diagnosis, classication, prognosis, and treat- ment. 13 Compared to single protein analysis, a panel of multiple proteins can improve diagnostic accuracy. 4,5 To date, immunohistochemistry (IHC) is the most routinely used technology for the detection of proteins in tissue samples. 6 However, conventional IHC only detects one protein at a time, limiting its application for high-throughput discovery and validation of protein biomarkers in tumor tissues. Array-based technologies have been developed for the detection of proteins in tissues by arraying patient samples on slides. Tissue microarrays (TMA) comprise an array of tens to hundreds of paran embedded tissue disks, 7,8 with typical tissue core size ranging from 0.6 to 2 mm. 9 Likewise, reverse- phase protein arrays (RPPA) 10,11 are made of proteins extracted from either formalin- xed, paran-embedded (FFPE) or fresh frozen tissues. However, histological spatial information is lost not only for RPPA as only a homogenate is measured but also for TMA as the original sample is destroyed. Hence, eorts have been made to directly quantify multiple proteins in a single tissue slice which has the benet of providing multiplexed and contextual information. An early demonstration is the multiplex-immunostain chip formed in a silicone rubber comprising 50 wells, each 2 mm wide and 3 mm apart, 12 allowing 50 proteins to be analyzed simultaneously, but requiring tissue sections that are at least 30 × 15 mm 2 large, much larger than many human tumors that could be only 510 mm in diameter. 13 Recently, multiplexed IHC was realized using an aqueous two-phase system with microliter droplets of a dextran solution containing dierent antibodies applied to a slide soaked within a polyethylene glycol (PEG) solution. 14 However, the center-to-center spacing was even larger at 5 mm. Miniaturized, multiplexed IHC was rst proposed by Kim et al. using microuidic chips with 800 μm wide and 5 mm long channels for staining 4 proteins simultaneously. 15 105 human breast tumor tissues were studied, and the results correlated well with those from conventional IHC. Others stained 10 proteins in parallel, 16 peformed automated assays, 17 and used quantum dots to improve assay performance. 18 Lovchik et al. used a microuidic probe to locally stain tissues with 100 μm diameter tear drop-shaped patterns. 19 Here, contact was avoided, and sample consumption was much reduced; however, serial processing is required for staining with multiple Received: March 4, 2017 Accepted: August 1, 2017 Published: August 1, 2017 Technical Note pubs.acs.org/ac © XXXX American Chemical Society A DOI: 10.1021/acs.analchem.7b00807 Anal. Chem. XXXX, XXX, XXXXXX
Transcript
Page 1: Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan Li,†, ... methods have emerged as powerful high throughput techniques, but they either

Immunohistochemistry MicroarraysHuiyan Li,†,‡ Gabrielle Brewer,†,‡ Grant Ongo,†,‡ Frederic Normandeau,†,‡ Atilla Omeroglu,§

and David Juncker*,†,‡,∥

†Biomedical Engineering Department, ‡McGill University and Genome Quebec Innovation Centre, §Department of Pathology,McGill University Health Centre, and ∥Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A0G1,Canada

*S Supporting Information

ABSTRACT: Immunohistochemistry (IHC) on tissue sections is widelyused for quantifying the expression patterns of proteins and is part of thestandard of care for cancer diagnosis and prognosis, but is limited to staininga single protein per tissue. Tissue microarray and microfluidics stainingmethods have emerged as powerful high throughput techniques, but theyeither only permit the analysis of a single protein per slide or require complexinstrumentation and expertise while only staining isolated areas. Here, weintroduce IHC microarrays (IHCμA) for multiplexed staining of intacttissues with preserved histological and spatial information. Droplets of adextran solution containing antibodies were prespotted on a slide andsnapped onto a preprocessed formalin-fixed, paraffin-embedded (FFPE)tissue section soaked in a polyethylene glycol solution. The antibodies areconfined within the dextran droplets and locally stain the tissue below with acontrast similar to the one obtained by conventional IHC. The microarray ofantibody droplets can be prespotted on a slide and stored, thus neither the preparation of the antibody solutions nor asophisticated microarray spotter is needed. Sampling considerations with IHCμA were evaluated by taking three tissues withvarying levels of cancer cells. A multiplex IHCμA with 180 spots targeting 8 cancer proteins was performed on a breast cancertissue section to illustrate the potential of this method. This work opens the avenue of applying microarray technologies forconducting IHC on intact tissue slices and has great potential to be used in the discovery and validation of tissue biomarkers inhuman tumors.

Analysis of protein biomarkers in tumor tissues can help incancer diagnosis, classification, prognosis, and treat-

ment.1−3 Compared to single protein analysis, a panel ofmultiple proteins can improve diagnostic accuracy.4,5 To date,immunohistochemistry (IHC) is the most routinely usedtechnology for the detection of proteins in tissue samples.6

However, conventional IHC only detects one protein at a time,limiting its application for high-throughput discovery andvalidation of protein biomarkers in tumor tissues.Array-based technologies have been developed for the

detection of proteins in tissues by arraying patient sampleson slides. Tissue microarrays (TMA) comprise an array of tensto hundreds of paraffin embedded tissue disks,7,8 with typicaltissue core size ranging from 0.6 to 2 mm.9 Likewise, reverse-phase protein arrays (RPPA)10,11 are made of proteinsextracted from either formalin-fixed, paraffin-embedded(FFPE) or fresh frozen tissues. However, histological spatialinformation is lost not only for RPPA as only a homogenate ismeasured but also for TMA as the original sample is destroyed.Hence, efforts have been made to directly quantify multiple

proteins in a single tissue slice which has the benefit ofproviding multiplexed and contextual information. An earlydemonstration is the multiplex-immunostain chip formed in asilicone rubber comprising 50 wells, each 2 mm wide and 3 mm

apart,12 allowing 50 proteins to be analyzed simultaneously, butrequiring tissue sections that are at least 30 × 15 mm2 large,much larger than many human tumors that could be only 5−10mm in diameter.13 Recently, multiplexed IHC was realizedusing an aqueous two-phase system with microliter droplets ofa dextran solution containing different antibodies applied to aslide soaked within a polyethylene glycol (PEG) solution.14

However, the center-to-center spacing was even larger at ∼5mm.Miniaturized, multiplexed IHC was first proposed by Kim et

al. using microfluidic chips with 800 μm wide and 5 mm longchannels for staining 4 proteins simultaneously.15 105 humanbreast tumor tissues were studied, and the results correlatedwell with those from conventional IHC. Others stained 10proteins in parallel,16 peformed automated assays,17 and usedquantum dots to improve assay performance.18 Lovchik et al.used a microfluidic probe to locally stain tissues with ∼100 μmdiameter tear drop-shaped patterns.19 Here, contact wasavoided, and sample consumption was much reduced; however,serial processing is required for staining with multiple

Received: March 4, 2017Accepted: August 1, 2017Published: August 1, 2017

Technical Note

pubs.acs.org/ac

© XXXX American Chemical Society A DOI: 10.1021/acs.analchem.7b00807Anal. Chem. XXXX, XXX, XXX−XXX

Page 2: Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan Li,†, ... methods have emerged as powerful high throughput techniques, but they either

antibodies. Collectively, these microfluidic technologies dem-onstrate that multiplex, microscale IHC is possible on a singletissue slice. However, the microfluidic systems requiredmicrofabricated chips and specialized equipment such assyringe pumps and pressure controllers, which are not availableto typical biology and pathology laboratories.Here, we introduce immunohistochemistry microarrays

(IHCμA), a microarray-based multiplexed IHC that allowsfor (i) probing multiple proteins on a single tissue sectionwithout cross-reactivity; (ii) distributing multiple microspots(200 μm) at high density across the whole tissue for eachprotein, instead of macroscopic staining of one or a fewsubsections (e.g., TMA sampling method) of a tissue slice foreach protein, to ensure different locations of the tissue aresampled, thus better tackling the tissue heterogeneity issues andminimizing false-negative scenarios; (iii) saving reagents andreducing the cost of the assay; (iv) storing the prespottedantibody microarrays so that no sophisticated microarrayspotter is required by “end-users”. As a proof-of-concept, aFFPE breast tumor tissue slice was stained with an 8-plexIHCμA with 180 spots illustrating the potential of this method.

■ EXPERIMENTAL SECTIONThe assay procedure of the IHCμA is shown in Figure 1. Here,we apply the snap chip technology20,21 to transfer a microarray

of primary antibodies simultaneously onto a tissue slide,resulting in ∼200 μm antibody spots on the tissue slice. All theantibodies are prepared, spotted, and stored in advance todisassociate the reagent preparation and chip fabrication withthe assay procedure, so that neither antibody preparation nor amicroarray spotter is required on site.

The IHCμA is compatible with the most widely used FFPEtissue slides, and all the commonly used protocols for tissuepretreatment (deparaffinization, rehydration, antigen retrieval),blocking, and downstream processing after incubation ofprimary antibodies can be easily adapted in this method.Both fluorescence and chromogenic detection can beimplemented in the downstream processing steps.

■ RESULTS AND DISCUSSIONTo demonstrate the transfer of antibody droplets homoge-neously onto a tissue slice without cross-contaminationbetween adjacent spots, antibody microarrays were transferredonto 5 μm-thick FFPE tissue slides from a human breast tumorfor staining TIMP122 and estrogen receptor (ER) proteins.Although chromogenic detection is widely used in IHC, theresults are semiquantitative, and therefore, immunofluorescencewas used to quantify the signal intensities.23,24 We first spottedantibodies in PBS with 20% glycerol and transferred them ontothe tissue in a humidified environment, Figure S1a. Thefluorescence image of the stained spots showed an obvious ringeffect (Figure S1a,b) that might be due to the Marangonieffect23−25 or diffusion limitations when a large amount ofantibodies reached the tissue from the sides of each spot.26

To overcome the ring effect, we implemented the PEG-dextran two-phase system14,27 in the IHCμA, with antibodiesmixed in dextran solutions and the tissue slice covered with aPEG aqueous solution (see Figure S1c). To adapt the dextrandroplets to nanoliter-scale inkjet spotting, we decreased theconcentration of the dextran to 5% and also added 10% glycerolin the spotting solutions to avoid evaporation. The fluorescenceimage in Figure S1c and the quantitative analysis in Figure S1d,as well as the images in Figures 2 and 4, show that antibodies

Figure 1. Schematic illustrating the procedure of IHCμA. (a, b) TheFFPE tissue slide is pretreated (deparaffinization, rehydration, andantigen retrieval) and then blocked with BSA. (c) Primary antibodydroplets are spotted on a slide with an inkjet spotter, and the slide isstored. (d) To perform multiplexed IHCμA, the Ab slide is retrievedand mounted in a snap apparatus and snapped onto the tissue slide. (e,f) After incubation, the tissue slide is stained with either fluorescenceor chromogenic methods, followed by imaging and quantifying thestaining. (g) An example image of a ER positive breast tumor tissueslide stained with anti-ER antibody using the IHCμA method. Nucleiwere stained, consistent with the tumor sub-type determined byconventional IHC.

Figure 2. Images of stained cell monolayer with anti-ER antibodyusing (a) the IHCμA method and (b) conventional ICC (antibodieswere used at 10 μg/mL). Dashed circles in (a) delineate the boundaryof the stained spots. (c, d) Close-up of the spot within the dashed boxin (a) and (b), respectively.

Analytical Chemistry Technical Note

DOI: 10.1021/acs.analchem.7b00807Anal. Chem. XXXX, XXX, XXX−XXX

B

Page 3: Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan Li,†, ... methods have emerged as powerful high throughput techniques, but they either

Figure 3. Effect of microarray-based sampling on the percentage of stained cells on three different tissue slices with varying levels of cancerous cells.A 4.2 × 4.2 mm2 square tissue section that could accommodate 81 microspots was defined. Each virtual spot was 200 μm in diameter, and the center-to-center spacing between spots was 500 μm, the same as the IHCμA layout used in this work. (a−c) Layout of 81 microspots on three 4.2 × 4.2mm2 tissue sections, divided into 9 subsections. The surface coverage of the 81 spots represents 45% of the square. The percentage of stained cellsfor the whole tissue is 77% (score 3) for (a) targeting ER protein, 24% (score 2) for (b), and 6.6% (score 1) for (c) targeting PR protein. The close-up image of the spots on the row 2, column 9 of each tissue is shown in (d−f). The nuclei are revealed with diaminobenzidine staining and arebrown, and the counterstain conducted with hematoxylin is blue. Scale bars: 30 μm. (g) Box plots showing the percentage of stained cells for each ofthe 81 microspots on three tissues. (h−j) 3D column charts demonstrating the percentage of stained cells for each of the 81 spots on the 3 tissues.(k) Box plots showing the percentage of stained cells for 100 times sampling of 1−81 spots on each of the three tissues.

Analytical Chemistry Technical Note

DOI: 10.1021/acs.analchem.7b00807Anal. Chem. XXXX, XXX, XXX−XXX

C

Page 4: Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan Li,†, ... methods have emerged as powerful high throughput techniques, but they either

were homogeneously transferred onto the tissue or cellmonolayer slices, without the ring effect observed in mostimages, and individual spots were well separated. In Figure S1d,some spots showed higher intensity on the edge, whichreflected a lack of cells inside the spots rather than the ringeffect. We quantified the signal intensity of 49 replicated spotson a slide with a cell monolayer as shown in Figure S1c andnormalized the data to the number of cells in each spot. Thecoefficient of variation (CV) of the 49 replicated spots stainedusing IHCμA was 17%. A close-up image of a microspot stainedwith the anti-TIMP1 antibody is shown in Figure S2a, usingDAPI as the counterstain for the visualization of cells in theimage. Further, we optimized antibody concentration andfound that, for example, for TIMP1, 400 μg/mL of theantibodies gave the best S/N ratio of 9.2 (Figure S2b). Also, theresults of the storage of the antibody slides are shown in FigureS3.The histological architecture of a monolayer of MCF-7 breast

cancer cells, known to be positive for ER, was revealed bychromogenic staining using IHCμA or conventional IHC/ICCand found to be comparable, Figure 2. IHCμA required higherconcentrations of antibodies compared to conventionalstaining, probably because the aqueous two-phase systemwith glycerol decreased antibody−antigen binding efficiency,similar to the snap-chip immunoassays where much higherantibody concentrations (but less antibody overall) wereneeded compared to large-volume conventional assays.21 InFigure 2a, nuclei outside of the spots were sometimes stained aswell, probably due to the diffusion of the antibodies out of thedroplet. The center-to-center spacing of 500 μm used hereappears adequate to prevent cross contamination betweenspots. The slight variation in spot size was also attributed to thevariations in inkjet spotting due to the relatively viscousantibody solutions. These results indicate that the IHCμA canprovide comparable staining patterns to conventional IHC/ICC and can be used to reveal spatial protein expressionpatterns in tissues.Microscale staining of a single area may not be representative

of the whole tissue considering the heterogeneity of tissue andthe irregular shape of tumors. IHCμA can address this concernby using multiple replicate spots distributed across the wholetissue to stain for the same protein. In this work, we used thepercentage of stained cells to help evaluate the variationsamong microspots and the differences between microspot-based sampling and the whole-slice staining. As a proof-of-concept, three breast tumor tissue sections with varying degreesof ER and PR staining representing three positive but aquantitatively distinct percentage of stained cell scenarios in thewidely used H-score system were evaluated,28 Figure 3a−f.Conventional whole-slice staining was used to generate theimages, and a digital grid was used to define the 81 spots insilico resembling an IHCμA. The distribution of the percentageof stained cells (Figure 3g) shows that the number of positivecells within individual spots varies significantly, but whenincreasing the number of spots (up to 81) that are sampled, theaverage number of positive cell converges to the actual averageof the whole slice. Also, we observed that (Figure 3h−j), forexample, in Figure 3i, the percentage of stained cells from spotson the left side had higher counts than that of the spots in themiddle. These observations imply that the microspots shouldbe sampled across the whole area of interest in the tissue,instead of taking all from one subarea, to address the tissueheterogeneity issues. Therefore, to distribute the spots across

the tissue, we divided each tissue into 9 subsections, with 9spots in each subsection, as shown in Figure 3a−c. For clinicaldiagnosis of cancer, our first criterion would be to identifywhether there are cancer cells or not. Subsampling mightincrease the risk of false-negative diagnosis. Considering theheterogeneity of tissues, taking one or multiple spots from eachsubsection would help minimize false-negative results.Next, we investigated the effect of additional subsampling on

the percentage of stained cells when using between 1 and 81spots. For scenarios with (i) up to 9 spots, each spot wasselected from a different subsection, and (ii) for more than 9spots, the same amount of spots was first attributed to each ofthe 9 subsections; the remainders were distributed according to(i). In each subsection, the spot position was randomlyattributed. The percentage of stained cells for each of the 81scenarios was calculated from all spots. Each of the scenariosfrom 1 to 81 spots was run 100 times. The distribution of eachcoverage is shown in Figure 3k. For low number of spots, therewas a broad range in the number of stained cells whichconverges to the 81-spot average as more and more spots areconsidered. The graphs suggest that between 10 and 25 spotsmay be needed to obtain staining results that converge towardthe 81-spot average. With 10 spots, the CV of the percentage ofstained cells, for randomly sampling 100 times, are 12%, 15%,and 21% for the tissues with score 3, 2, and 1, respectively.With 25 spots, the CVs are 6.6%, 8.3%, and 10%, respectively.These results indicate that the accuracy of the IHCμA isdependent on many parameters, including the percentage ofstained cells of the whole tissue and the tissue heterogeneity.One should keep in mind that conventional IHC also suffersfrom sampling artifacts as a tissue section constitutes only asmall fraction of the tumor tissue. In IHCμA, the more spotsare used, the closer the results will converge to conventionalwhole tissue staining, but at the cost of multiplexing capability.Clinically, the H-score is widely used, and it will be importantto ensure that the H-score is conserved between methods,which could be achieved by deriving a confidence metric so thatcounts falling close to the threshold between two categories areidentified as ambiguous. In the case shown here, we found thatby using at least 8, 4, and 17 spots for each of the tissues,respectively, the same H-score was obtained as that byconventional IHC. The subsampling method could beenhanced using artificial intelligence and machine learning,29

allowing additional information to be extracted. If a result isinferred to be ambiguous with possible clinical consequences,then staining on additional slides could be performed. In anycase, rigorous clinical studies and statistical analysis will berequired before therapeutic choices can be made on the basis ofthe results of IHCμA.To demonstrate the multiplexing capability of the IHCμA,

we performed an 8-plex assay on a commercial FFPE breasttumor slide, with subtype of ER−, PR+, HER2−. The 8proteins include three markers for breast cancer subtyping (ER,PR, HER2) and 5 cancer-related proteins. The layout of themicroarray and the fluorescence image of IHCμA staining areshown in Figure 4a,b. As evaluated in Figure 3, we used 18spots for each protein, by randomly taking 2 spots from each ofthe 9 subsections as shown in Figure 3a−c. The distributions ofthe S/N ratios from 18 spots for each protein are shown inFigure 4c. PR was overexpressed, and the average signal levelsfrom HER2 and ER were similar to those from negativecontrols. These results are consistent with the tumor subtype asdetermined by conventional IHC. We found P53 and PTEN

Analytical Chemistry Technical Note

DOI: 10.1021/acs.analchem.7b00807Anal. Chem. XXXX, XXX, XXX−XXX

D

Page 5: Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan Li,†, ... methods have emerged as powerful high throughput techniques, but they either

were highly overexpressed in this tissue. This high expression ofP53 and PTEN in PR+ breast tumor is consistent with previousfindings in the same subtype.30,31 TIMP1 was also detected inthe tissue while CDK7 and CK7 were not highly expressed.The expression of some proteins, such as PR and TIMP1, wasrelatively heterogeneous in this tissue, showing a wide dynamicrange in S/N ratios, while other proteins such as ER and HER2were more homogeneous. To validate the multiplex IHCμA,these 8 proteins were also stained using whole-tissuefluorescence assays on adjacent tissue slices from the sametumor, and the S/N ratios against the staining with normal IgGwere calculated for each protein (Figure S4) and found to beconsistent across both methods. Our results suggested thatIHCμA may be a useful tool for quantitative, multiplexedprotein detection on FFPE tissue slides.

■ CONCLUSIONSIn this work, we developed a novel multiplexed immunohis-tochemistry using a snap chip. The microarray-based samplingconsiderations to tackle tissue heterogeneity issues and tominimize false-negative scenarios were discussed. An 8-plexassay with two negative controls using a total of 180 microspotswas performed on a breast tumor tissue slide, which is thelargest number of isolated stains on a single slice to date to thebest of our knowledge, indicating the multiplexing potential ofthis technology. Compared to previous array-based tissuestaining,12,14 our approach increased the array density by 2orders of magnitude, and thus, a small tumor tissue of 5 × 5mm2 could be stained with 100 microspots and used for mosttumors that reach this size at diagnosis. In this multiplex assay,108 nL of antibody solution was used per protein per tissueslice, equivalent to approximately $0.2 USD per target, thussaving reagent and reducing the cost of the assay.Similar to TMA, IHCμA is based on the concept of sampling,

which needs to be carefully optimized to ensure representativestaining results but which allows obtaining more informationfrom a given sample. The results obtained with IHCμA wereconsistent with whole-slice staining and, in the case ofambiguous results, could be resolved by staining additional

sections from the same tissue. A statistical analysis with asufficiently large number of tissue sections will be necessary todevelop theoretical and experimental models that will helpchoose the optimal number of spots to obtain staining resultswithin a predetermined confidence interval as compared toconventional IHC on a full slice. Further miniaturization mightopen the door to array-on-array staining by applying a highdensity antibody microarray on a lower density tissuemicroarray using a snap chip. For example, with 250 μmspot-to-spot spacing, 12 or 27 spots could be accommodatedon typical core sizes of TMA with 1 or 1.5 mm diameter. Webelieve that the IHCμA has the potential to be used in large-scale multiplexed protein profiling in archived FFPE tumortissue samples.

■ ASSOCIATED CONTENT*S Supporting InformationThe Supporting Information is available free of charge on theACS Publications website at DOI: 10.1021/acs.anal-chem.7b00807.

Reagents and materials; fabrication and storage of thesnap chip; pretreatment of the FFPE tissue slides;IHCμA using a snap chip; imaging of the slide and dataanalysis; primary antibodies used in IHCμA; optimiza-tion of the IHCμA; fluorescence image; comparison ofS/N ratios at different concentrations of antibodies;results of IHCμA targeting TIMP1 protein; results of theconventional staining of the 8 proteins on adjacent slicesof the human breast tumor tissue (PDF)

■ AUTHOR INFORMATIONCorresponding Author*E-mail: [email protected] Juncker: 0000-0002-7313-1162NotesThe authors declare no competing financial interest.

■ ACKNOWLEDGMENTSWe thank Rob Sladek for the use of the Nanoplotter inkjetspotter. We are thankful for the financial support from theCanadian Cancers Society Research Institute (CCSRI), theCanadian Institutes for Health Research (CIHR), the NaturalScience and Engineering Research Council of Canada(NSERC), and the Canada Foundation for Innovation (CFI).G.B. acknowledges a scholarship from the NSERC-CREATEIntegrated Sensor Systems program. D.J. acknowledges supportfrom a Canada Research Chair.

■ REFERENCES(1) Parise, C. A.; Caggiano, V. J. Cancer Epidemiol. 2014, 2014,469251.(2) Muller, P. A. J.; Vousden, K. H. Cancer Cell 2014, 25, 304−317.(3) Thomas, D. S.; Fourkala, E. O.; Apostolidou, S.; et al. Br. J. Cancer2015, 113, 268−274.(4) Xu, T.; Fang, Y.; Rong, A.; Wang, J. BMC Med. Res. Methodol.2015, 15, 94.(5) Kang, L.; Xiong, C.; Crane, P.; Tian, L. Stat. Med. 2013, 32, 631−643.(6) Hornick, J. L. Mod. Pathol. 2014, 27, S47−S63.(7) Kallioniemi, O.-P.; Wagner, U.; Kononen, J.; Sauter, G. Hum.Mol. Genet. 2001, 10, 657−662.

Figure 4. IHCμA detecting 8 proteins on a human breast tumor tissueslide with the tumor subtype of ER−, PR+, HER2−. (a) Layout of theantibodies targeting 8 cancer-related proteins. Normal mouse IgG (MIgG) and normal rabbit IgG (R IgG) were used as negative controls.(b) Fluorescence image of the staining results on the tissue slide. (c)Box plots of S/N ratios (the ratio of the net signal intensity to thenoise level of the antibody) for each of the 8 proteins and 2 IgGcontrols.

Analytical Chemistry Technical Note

DOI: 10.1021/acs.analchem.7b00807Anal. Chem. XXXX, XXX, XXX−XXX

E

Page 6: Immunohistochemistry Microarrays - McGill University · Immunohistochemistry Microarrays Huiyan Li,†, ... methods have emerged as powerful high throughput techniques, but they either

(8) Rimm, D. L.; Camp, R. L.; Charette, L. A.; et al. Cancer journal(Sudbury, Mass.). 2001, 7, 24−31.(9) Eckel-Passow, J. E.; Lohse, C. M.; Sheinin, Y.; Crispen, P. L.;Krco, C. J.; Kwon, E. D. Diagn. Pathol. 2010, 5, 48.(10) Wilson, B.; Liotta, L. A.; Petricoin, E., III Dis. Markers 2010, 28,225−232.(11) Assadi, M.; Lamerz, J.; Jarutat, T.; et al. Mol. Cell. Proteomics2013, 12, 2615−2622.(12) Furuya, T.; Ikemoto, K.; Kawauchi, S.; Oga, A.; Tsunoda, S. i.;Hirano, T.; Sasaki, K. J. Histochem. Cytochem. 2004, 52, 205−210.(13) Pitteri, S. J.; Kelly-Spratt, K. S.; Gurley, K. E.; et al. Cancer Res.2011, 71, 5090−5100.(14) Frampton, J. P.; Tsuei, M.; White, J. B.; Abraham, A. T.;Takayama, S. Biotechnol. J. 2015, 10, 121−125.(15) Kim, M. S.; Kim, T.; Kong, S.-Y.; Kwon, S.; et al. PLoS One2010, 5, e10441.(16) Kim, M. S.; Kwon, S.; Kim, T.; Lee, E. S.; Park, J.-K. Biomaterials2011, 32, 1396−1403.(17) Ciftlik, A. T.; Lehr, H.-A.; Gijs, M. A. M. Proc. Natl. Acad. Sci. U.S. A. 2013, 110, 5363−5368.(18) Kwon, S.; Cho, C. H.; Lee, E. S.; Park, J.-K. Anal. Chem. 2015,87, 4177−4183.(19) Lovchik, R. D.; Kaigala, G. V.; Georgiadis, M.; Delamarche, E.Lab Chip 2012, 12, 1040−1043.(20) Li, H.; Bergeron, S.; Juncker, D. Anal. Chem. 2012, 84, 4776−4783.(21) Li, H.; Munzar, J. D.; Ng, A.; Juncker, D. Sci. Rep. 2015, 5,11688.(22) Wurtz, S. Ø.; Wurtz, S. Ø.; Schrohl, A.-S.; Mouridsen, H.;Brunner, N. Acta Oncol. 2008, 47, 580−590.(23) Dupouy, D. G.; Ciftlik, A. T.; Fiche, M.; et al. Sci. Rep. 2016, 6,20277.(24) McCabe, A.; Dolled-Filhart, M.; Camp, R. L.; Rimm, D. L. J.Natl. Cancer Inst. 2005, 97, 1808−1815.(25) Rao, A. N.; Vandencasteele, N.; Gamble, L. J.; Grainger, D. W.Anal. Chem. 2012, 84, 10628−10636.(26) Pappaert, K.; Ottevaere, H.; Thienpont, H.; Van Hummelen, P.;Desmet, G. BioTechniques 2006, 41, 609−616.(27) Frampton, J. P.; White, J. B.; Simon, A. B.; Tsuei, M.; Paczesny,S.; Takayama, S. Sci. Rep. 2015, 4, 4878.(28) Rizzardi, A. E.; Johnson, A. T.; Vogel, R. I.; et al. Diagn. Pathol.2012, 7, 42.(29) Madabhushi, A.; Lee, G. Med. Image Anal. 2016, 33, 170−175.(30) Torhorst, J.; Bucher, C.; Kononen, J. Am. J. Pathol. 2001, 159,2249−2256.(31) Lebok, P.; Kopperschmidt, V.; Kluth, M.; et al. BMC Cancer2015, 15, 963.

Analytical Chemistry Technical Note

DOI: 10.1021/acs.analchem.7b00807Anal. Chem. XXXX, XXX, XXX−XXX

F


Recommended