ebook img

optimization of whole genome amplification products for sequencing applicati PDF

12 Pages·2017·1.52 MB·English
by  
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview optimization of whole genome amplification products for sequencing applicati

Shoetal.BMCCancer (2017) 17:457 DOI10.1186/s12885-017-3447-6 RESEARCH ARTICLE Open Access Precision oncology using a limited number of cells: optimization of whole genome amplification products for sequencing applications Shonan Sho1,2* , Colin M. Court1,2, Paul Winograd1,2, Sangjun Lee3, Shuang Hou3, Thomas G. Graeber3, Hsian-Rong Tseng3 and James S. Tomlinson1,2,4 Abstract Background: Sequencing analysis of circulating tumor cells(CTCs) enables “liquid biopsy” to guide precision oncology strategies. However, this requires low-template wholegenomeamplification(WGA) thatis prone to errors and biases from uneven amplifications.Currently, quality control (QC) methods for WGAproducts, as well as the number ofCTCs needed for reliable downstream sequencing, remain poorly defined. We sought to define strategies for selecting and generating optimal WGA products from low-template input as it relates to their potential applications inprecision oncology strategies. Methods: Singlepancreatic cancer cells (HPAF-II) were isolated using laser microdissection. WGA was performed using multiple displacement amplification (MDA), multiple annealing and looping based amplification (MALBAC) and PicoPLEX. Qualityof amplified DNA products were assessed using a multiplex/RT-qPCR based method that evaluates for 8-cancer related genesand QC-scores were assigned. We utilized this scoring system to assess the impact of de novo modifications to the WGA protocol. WGA products were subjected to Sanger sequencing,array comparative genomic hybridization (aCGH)and next generation sequencing (NGS) to evaluate their performances inrespective downstream analysesproviding validation of theQC-score. Results: Single-cell WGA products exhibited a significant sample-to-sample variability in amplified DNA qualityas assessed by our 8-gene QCassay.Single-cellWGA products that passed thepre-analysis QC had lower amplification biasand improved aCGH/NGS performance metrics when compared to single-cell WGA products that failedthe QC. Increasing thenumberof cellular input resulted in improved QC-scores overall, buta resultant WGA product that consistentlypassedthe QC step required a starting cellularinput of atleast 20-cells. Our modified-WGA protocol effectively reduced this number, achieving reproducible high-quality WGAproducts from ≥5-cells as a starting template. Astartingcellular input of 5to 10-cells amplified using the modified-WGA achieved aCGH and NGSresults that closelymatched that ofunamplified, batch genomic DNA. Conclusion: The modified-WGA protocol coupled with the8-gene QC serve as an effective strategyto enhance the quality of low-template WGA reactions. Furthermore, a threshold numberof 5–10 cellsare likely needed for a reliable WGA reactionand product withhighfidelity to theoriginal starting template. Keywords: Precision oncology, Whole genome amplification, Single-cellsequencing, Next generation sequencing, Multiple displacement amplification *Correspondence:[email protected] 1DepartmentofSurgery,UniversityofCaliforniaLosAngeles,10833LeConte Ave,California,LosAngeles90095,USA 2DepartmentofSurgery,GreaterLosAngelesVeteran’sAffairsAdministration, 11301WilshireBlvd,California,LosAngeles90073,USA Fulllistofauthorinformationisavailableattheendofthearticle ©TheAuthor(s).2017OpenAccessThisarticleisdistributedunderthetermsoftheCreativeCommonsAttribution4.0 InternationalLicense(http://creativecommons.org/licenses/by/4.0/),whichpermitsunrestricteduse,distribution,and reproductioninanymedium,providedyougiveappropriatecredittotheoriginalauthor(s)andthesource,providealinkto theCreativeCommonslicense,andindicateifchangesweremade.TheCreativeCommonsPublicDomainDedicationwaiver (http://creativecommons.org/publicdomain/zero/1.0/)appliestothedatamadeavailableinthisarticle,unlessotherwisestated. Shoetal.BMCCancer (2017) 17:457 Page2of12 Background originalgenomictemplate.Therefore,onemustbeableto “Liquidbiopsy”ofcirculatingtumorcells(CTCs)hasbeen accurately differentiate between high- and low- quality suggested in many recent studies as an ideal biopsy tech- WGA products in order to ensure accurate sequencing niqueforprecisiononcologyapplications[1–5].CTCsare resultsforguidingcancertherapy. thoughttoarisefrombothprimaryandmetastaticlesions, Currently, however, quality control metrics and selection allowing for a more comprehensive representation of the criteria for high-quality WGA products from single cells tumor genomic make-up [6]. Furthermore, the need for have not been sufficiently defined. Much of the existing only a simple peripheral blood draw in “liquid biopsy” literature utilizing whole genome amplified DNA lack ana- makes it amenable to repeated samplings without incur- lysis of the quality of WGA samples being used. Given the ring significant costs or risks to patients. Although known sample-to-sample variability in minimal template successful CTC enrichment, capture and downstream WGA products, WGA quality must be defined in order to molecular analysis has been described, major obstacles accuratelyinterpretandcompareWGA-DNAderiveddata. still remain prior to its clinical translation [6, 7] (Fig. 1). Furthermore, although prior studies have shown improved One major challenge is the limited number of CTCs WGAqualitywithincreasingamountofgenomictemplate available for molecular analysis. The number of CTCs input [19], the number of CTCs needed to generate a obtainable from a single peripheral blood remains highly WGA product suitable for sequence analysis to be utilized limited, with most studies showing <5 CTC/mL from a inprecisiononcologystrategiesremainunknown. single peripheral blood [3, 8]. For many GIcancers, espe- In the current study, we sought to define strategies for cially pancreas ductal adenocarcinoma, CTCs are even selecting and generating optimal WGA products from more limited [9, 10]. Thus, for molecular analysis to be samples ranging from one to twenty cells as it relates to performed using CTCs, the limited amount of genomic their potential applications in precision oncology strat- materials available from CTCs must undergo whole gen- egies. To this end, we developed a quality control (QC) omeamplification(WGA)togenerateadequatequantities assay to help facilitate the selection of high quality ofDNAfordownstreamsequencinganalysis. WGA product suitable for use in downstream sequen- Despite recent advances in WGA techniques, amplifi- cing applications, including point mutation detection, cation processes remain prone to uneven amplifications, array comparative genomic hybridization (aCGH) and resulting in amplification bias [11–14]. For heterozygous next generation sequencing (NGS). In order to better sites, this can result in a total loss of one allele, a understand the key determinants of WGA quality, we phenomenon called allele drop out (ADO) [15, 16]. This compared various WGA methods in addition to the is especially problematic when working with a small number of input cells to determine their influence upon number of cells, as in CTC analysis, as the stochastic amplification reactions. We then used our findings to variation in the WGA process is exacerbated when start- develop a modified multiple displacement amplification ing with low copies of genomic input [17, 18]. This (MDA) protocol with a notable improvement in ampli- results in considerable variability in sample-to-sample fied DNA quality over the conventional MDA proto- quality when working with CTCs [15]. Low-quality col. Ultimately, we utilized these findings to determine WGA products with significant degrees of amplification the “threshold” number of cells needed for reliable bias and ADO are inappropriate for precision oncology molecular analysis that could be utilized in precision applications as they fail to accurately represent the oncology strategies. Fig.1WorkflowofCTCsequencingandassociatedchallenges.SeveralchallengesexistinCTCsequencinganalysis.Qualitycontrol(QC)forthe amplifiedDNAproductisparamount,aslowqualityWGAproductsleadtofailed/inaccuratedownstreamsequencingapplications.AQCstep priortocostlydownstreamsequencingapplicationshelpsreduceavoidablecostsassociatedwithfailedsequencing Shoetal.BMCCancer (2017) 17:457 Page3of12 Methods reduced individual reaction volumes (3ul instead of Celllines 50ul), all taking place in parallel. Following the amplifi- Pancreatic cancer cell line HPAF-II was obtained from cation step, contents within the 16-wells were collected American Type Culture Collection (ATCC, Virginia, into one tube, followed by the purification and quantifi- USA), and grown using EMEM medium (ATCC) supple- cation stepasdescribed above. mented with 10% fetal bovine serum (ATCC) and 100 U/ mL penicillin-streptomycin (ATCC). All cell lines were Developmentofqualitycontrol(QC)-scoreforWGA-DNA grownat37°Cwith5%CO andwereroutinelypassaged Prior literatures have described the role of multiplex 2 at 80% confluence using an iso-osmotic sodium citrate PCR in evaluating DNA quality isolated from formalin- solutionforcellrelease(Thermo,Massachusetts,USA). fixed paraffin embedded (FFPE) tissues prior to down- stream analysis using aCGH [20]. Based on this concept, Lasermicrodissection we developed an 8-gene multiplex PCR/quantitative In preparation for laser microdissection, cells were PCR (qPCR) based QC assay for evaluation of WGA- released from the culture plates using the iso-osmotic DNA quality. We selectively chose for 8 cancer-related sodium citrate solution (Thermo).Followingawashwith genes that are considered molecular targets for targeted the culture medium, each cellline wasdiluted to adens- cancer therapy and therefore highly implicated in guid- ity of 1000 cells per 100 μL. Approximately 1000 cells ing therapeutic decisions. This way, the QC assay identi- (100 μL) were smeared on PEN membrane slides (Leica, fies WGA products suitable for precision oncology Wetzlar, Germany), air-dried for 10 min, and fixed with applications by evaluating for the coverage and “acces- 100 μL of 100% ethanol. Cells were then isolated using sibility” of these important genomic locations within the PALM MicroBeam laser microdissection system the WGA-DNA. Genes evaluated by the QC assay in- (Zeiss, Oberkochen, Germany). 1, 5, 10 or 20 cells were cluded BRAF, EGFR, KIT, KRAS, NRAS, PIK3CA, laser microdissected and collected into 200 μL opaque PTEN and P53. tube caps (Zeiss) using the laser pressure catapult Score of 0–8 was assigned based on the number of function. Cell transfer to the tube cap was confirmed by genes successfully amplified and detected using multi- imagingthe cap priortocapclosure usingthe cap-check plex PCR and qPCR. Failure to detect one or more of function. the 8-gene from the WGA product signifies lack of coverage or potential ADO during the WGA process, Wholegenomeamplification both of which indicate a poor quality WGA product and Isolated cells were subjected to genomic DNA isolation jeopardizes accurate representation of the original start- and WGA using one of the three commercially available ing genome. Thus, we only gave WGA-DNA products single-cell WGA kits according to the manufacturer’s with 8 out of 8 score a “pass” and deemed them fitting protocol: REPLI-g Single-Cell Kit (Qiagen, California, forfurtherdownstreamanalysis. USA), Multiple Annealing and Looping Based Amplifi- A secondary QC-score was generated to confirm the cation Cycles (MALBAC) Single-cell WGA Kit (Yikon internalvalidity of the original QC-score based onthe 8- Genomics, Beijing, China) and PicoPlex Single-Cell cancer related genes. The secondary QC-score was WGA Kit (Rubicon, Michigan). Reactions were per- generatedfrom adistinct setof8primerpairsrepresent- formed a minimum of 3 times for all conditions tested. ing 8 housekeeping genes: NDUFA, UQCRC, ACTG, WGA products were purified using the QIAquick PCR CYB5A,GABA-RAPL,MIF,MYC,PRPH. Purification Kit (Qiagen) and quantified with NanoDrop 2000 (Thermo), in keeping with a previously described MultiplexPCRandquantitativePCR(qPCR) CTCmolecular analysismethodology[15]. WGA products were subjected to multiplex PCR pre- Modified MDA protocol was performed using the amplification followed by qPCR assay for detection of same reagents available from the REPLI-g Single Cell Kit amplified targeted genes. Multiplex PCR was performed (Qiagen), with modifications made in the cell lysis step using primer sets representing genes mentioned above. and the final amplification step. Cell lysis was performed Primer sets used for the primary QC assay included: over a course of 30 min to ensure complete lysis of gen- BRAF (forward 5′ – TAC TGC TCT TTC TTC TCC omic material from isolated cells (as opposed to 10 min AAC AC – 3′; reverse 5′ – CCT GAT TGT ATT TGA recommended by the manufacturer). Prior to the final GAT CTA GTA GGG – 3′) EGFR (forward 5′-CAG amplification step, the 50-ul MDA reaction mix was CCT TCT CCG TAA TTA GCA T – 3′; reverse 5′ – mixed for 30 s by pipetting up and down and then TGA CAC AGA TAA TTG TCC CAC AG – 3′), KIT partitioned into 16 individual reactions (approximately (forward 5′ – GGC ATT GAG GAG GGA TAG TAA 3ul each) and the amplification occurred at 30 °C for AT – 3′; reverse 5′ – CTG AAC AAT TTG CTT GAA 8 h. This resulted in 16 individual MDA reactions with TGT TGG – 3′), KRAS (forward 5′ – GTG TTA CTT Shoetal.BMCCancer (2017) 17:457 Page4of12 ACC TGT CTT GTC TTTG – 3′; reverse 5′ – GCC and 95 °C at 0.5 °C intervals. Real-time PCR data were TTC TAG AAC AGT AGA CAC AA – 3′), NRAS reviewed and analyzed using the CFX manager (BioRad). (forward 5′ – AAT GGA ATC CCG TAA CTC TTG G Specificity of the PCR amplification product was deter- –3′;reverse 5′–GATGATGTACCTATG GTG CTA mined using melting curve analysis. PCR products with GTG – 3′), PIK3CA (forward 5′ – AGG GCA AAT melting-temperature (Tm) matching the expected value AATAGT GGT GAT CT – 3′; reverse 5′ – CAG CAA based on primer sequence, and threshold cycle (Ct) <30 TTA CTT GTT CTG GTA CAC – 3′), PTEN (forward were counted asreliableamplificationanddetection. 5′ – CTT TCT CTA GGT GAA GCT GTA CT – 3′; reverse 5′ – GGT TCATTG TCA CTA ACATCT GG Array-CGH – 3′) and P53 (forward 5′ – AAG AGA AGC AAG Sample WGA-DNA and reference DNA were differen- AGG CAG TAA G – 3′; reverse 5′ – CTT AGG CTC tially labeled with cyanine-3 (CY3) and cyanine-5 (Cy5) CAG AAA GGA CAA G – 3′). Primer sets used for the dyes using the GenetiSure Amplification and Labeling secondary QC assay included: NDUFA7 (forward 5′ – Kit (Agilent) according to the manufacturer’s protocol. TGC TCT GGA TGT GAA GAT GCC A – 3′; reverse Briefly, a 15.5 ul reaction mixture containing sample or – 5′ – TTC CAG GTA AAT CCA GCC CAG G – 3′), reference DNA, Random Primer Mix and water was UQCRC1 (forward 5′ – CAG CCA GTC AGC ATC placed in 98 °C for 3 min for DNA denaturation and ATC CAA C – 3′; reverse 5′ – GAA AGC CGG ATT transferred to 4 °C for 3 min. Next, 9.5 ul of Labeling GCG GTA ACA T - 3′), ACTG1 (forward 5′ - GCT Master Mix containing 5ul of 5× Reaction Buffer, 2.5ul CAA TGG GGT ACT TCA GGG T – 3′; reverse 5′ – of 10× dNTP Mix, 1.5 ul of Cy5-dUTP/Cy3-dUTP and GTG GAC GTTACG TAA AAG GCC C – 3′), CYB5A 0.5 Exo (−) Klenow were added to each tube containing (forward 5′ – GGC AAC GCT TAG ACT CTG TGT G the sample/reference. DNA labeling reaction was per- – 3′; reverse 5′ – CTG CCC TTG GCC TAA CTA formed in 37 °C for 45 min followed by inactivation step ACC T – 3′), GABARAPL2 (forward 5′ – CCA GCC in 65C for 10 min. Labeled DNA was purified using the AAT TCA TGA GTC GGT G – 3′; reverse 5′ – CCT Post Labeling Purification Columns (Agilent) according GAC AAC TCG CAA GTA GCAC – 3′), MIF (forward to the manufacturer’s protocol. Purified labeled DNA 5′ – AGA AGT CAG GCA CGT AGC TCA G – 3′; samples were prepared for hybridization, which took reverse 5′ – GGC ACGTTG GTG TTTACGATG A – place on Agilent 8x60K CGH microarray slides at 67 °C 3′), MYC (forward 5′ – GGA TAG CTC TGC AAG for 6 h. Following the hybridization, the slides were GGG AGA G – 3′; reverse 5′ –TCG TCG CAG TAG washed per manufacturer protocol, and prepared for AAA TAC GGC T – 3′), PRPH (forward 5′ – GTT scanningusingtheAgilentSureScanMicroarrayScanner CCT CAA GAA GCT GCA CGA G – 3′; reverse 5′ – (Agilent). Microarray image was prepared and analyzed CGT TAG ACT CTG GAT CTG GCG T – 3′). Details usingtheAgilentCytoGenomicssoftware(Agilent). of multiplex PCR pre-amplification is available in the Additional file 1: Supplementary Material. Briefly, PCR Sequencinglibrarypreparationandsequencing reactions were carried out on a C1000 Thermal Cycler Purified WGA products were sheared to generate DNA (Bio-Rad) with the Multiplex PCR Plus Kit (Qiagen) fragments of 350 bps using the Covaris sonicator using total volumes of 17 uL per reaction. The reaction (Covaris). Followingcleanupof the sonicated DNA, end- conditions were as follows: 95 °C for 15 min, denatur- repair and ligation were performed using the KAPA ation at 94 °C for 30 s, annealing at 64 °C for 30 s, and DNA Library Preparation Kit (KAPA Biosystems) ac- extension at 72 °C for 30 s for a total of 10cycles, with a cording to the manufacturer’s protocol, followed by final stepof72°Cfor 10min. library amplification by PCR. Sequencing was performed Following the pre-amplification step by multiplex on an Illumina HiSeq 2000 using random primers and PCR, the resulting amplified products were analyzed and pair-endreads of75bps(2X75bps). detected using qPCR. Reactions took place on BioRad CFX-96 real time system (BioRad) using the QuantiTect Sequencinganalysisandvisualization SYBR Green PCR Kit (Qiagen). A 25-ul reaction mix- The sequencing read data was analyzed using Ginkgo tures were prepared, which contained 12.5 ul of the (http://qb.cshl.edu/ginkgo), an open-source platform for SYBR Green PCR Master Mix (Qiagen), 9.5 ul of RNA- the analysis of single-cell copy-number variations (CNV). grade water, 1 ul of individual primers sets (10uM) and This analysis platform was developed specifically to 2ul of the multiplex PCR product. The reaction condi- address the unique challenges of single-cell sequen- tion were as follows: 95 °C for 15 min, followed by 35 - cing data, and incorporates built-in computational cycles of 94 °C for 15 s, 64 °C for 20 s, and 72 °C for tools for data optimization. Specifically, limitations as- 20 s. The plate was read following the extension step at sociated with low depth of sequencing coverage, amp- 72 °C. Melting curve analysis was performed between 70 lification bias and inflated read counts from poorly Shoetal.BMCCancer (2017) 17:457 Page5of12 assembled genomic regions were addressed in Ginkgo’s in amplified DNA quality was noted for MALBAC, with analysis. Detailed description of the computational meth- QC-scores ranging between 2 and 6. Although Picoplex odologyandcorrectiontoolsareprovidedbyGarvinetal. resulted in less quality disparity, majority of samples [21]. Mapped reads were binned using variable length- achieved scores of only 3. QC-score profiles for the binning and underwent GC bias normalization prior to MDA and MALBAC methods were overall similar, but segmentation using circular binary segmentation. Quality the WGA product that passed our QC criteria (QC- metrics data, including Lorenz curve, histogram of read score = 8) was only found in the single-cell sample count distribution and index of dispersion were obtained amplifiedwith usingtheMDAmethod. as a part of the Ginkgo analysis pipeline. Sequencing data visualization was performed using Nexus soft- Determinationofnumberofcellsneededtoachieve ware (Biodiscovery, inc.). reproducibleWGAproductquality We noted significant random variations in WGA prod- KRASPCRandsangersequencing uct quality when using single-cells as starting template PCR amplification of KRAS exon 2 was performed using (Fig. 2). Increasing the amount of starting genomic the following primer (Integrated DNA Technologies): template has been shown to improve WGA product Forward 5′ – AAG GTA CTG GTG GAG TAT TTG – quality [19, 22]. Thus, we hypothesized that there would 3′ and Reverse 5′ – GTA CTC ATG AAA ATG GTC be a certain “threshold” number of cellular input above AGA G – 3′, with expected amplicon length of 295 bps. which a reliable and reproducible WGA process is PCR reactions were carried out on a C1000 Thermal possible, i.e. a number of cells which will achieve a Cycler (Bio-Rad) with Platinum PCR SuperMix High “pass”inourQC steponaconsistentbasis. Fidelity Kit (Invitrogen) using total volumes of 50 μLper In order to test this hypothesis, 5-cell, 10-cell and 20- reaction according to the manufacturer’s protocol. The cell samples ofHPAF-II cells were cut and isolated using reaction conditions were as follows: denaturation at a laser micro-dissector. WGA was performed using the 94 °C for 30 s, annealing at 55 °C for 30 s, and exten- MDAmethod, followed bythe QC assay and assignment sion at 68 °C for 45 s for a total of 40 cycles. The PCR of the QC-scores (Fig. 3a). Less variability in amplified products were purified using the QIAquick PCR Purifi- DNA quality was noted when 5 or more cells were used cation Kit (Qiagen) and eluted into 50 uL of nuclease- as starting genomic template. All samples persistently free water (Qiagen). DNA was diluted to a concentra- achieved a QC-score of ≥5 (5-cell) or ≥6 (10-cell), as tion of 10 ng/uL based on Nanodrop quantification of opposed to single-cell WGA reactions with scores ran- the PCR product. Automated dideoxy terminator se- ging from 1 to 8. Furthermore, at least one of the tripli- quencing was performed by capillary electrophoresis by cates in 5-cell and 10-cell group passed the QC-step the UCLA GenoSeq Core on an ABI 3730 DNA (QC-score =8). However, ahighly reliable WGA process analyzer using Big Dye Terminator chemistry (Applied withalloftheWGAproductspassingtheQCstepcould Biosystems). All sequences wereanalyzed by manual in- not be achieved until 20 cells were used as starting gen- spection of the individual trace files using Four Peaks omictemplate input. (Nucleobytes). ModifiedMDAreactiontoimprovequalityofWGA Results productfromlimitedtemplatesamples. WGAproductvariabilityinsingle-cells With the existing MDA protocol, multiple single-cell We applied our 8-gene QC assay to single-cell WGA- WGA reactions must be performed in order to obtain DNA samples. We tested three different WGA reaction one high-quality WGA product suitable for downstream methods: MDA, MALBAC and Picoplex. Total of 18 analysis (Fig. 2). Even when more than a single-cell was individual HPAF-II cells were isolated using laser micro- used, a reliable WGA reaction with its amplified DNA dissection and used in the respective WGA methods. product passing the QC step on a consistent basis could The experiment was repeated 6 times in order to ac- not be achieved until 20 cells were used (Fig. 3a). For count for the variability expected with the single-cell many GI cancers that are known to generate only few WGA process. QC assay was performed and the QC- CTCs (i.e. pancreas ductal adenocarcinoma), obtaining score wasassignedforeachWGA product. 20 CTCs from a single peripheral blood draw may be Despite using a clonally expanded cell line and per- unrealistic. forming WGA reactions in a parallel fashion under the Given these problems, we sought to develop a mo- same condition, we noted significant variability in dified WGA protocol that would reduce the sample- sample-to-sample WGA-DNA quality as assessed by the to-sample variability in DNA quality and lower the 8-gene QC assay (Fig. 2). QC-scores for MDA amplified number of starting cells needed to achieve a reliable single-cells ranged between 1 and 8. Similar variability WGA process. Multiple prior reports have described Shoetal.BMCCancer (2017) 17:457 Page6of12 Fig.2QC-scoresforsingle-cellWGAproducts.Single-cellWGAwasperformedusingMDA,MALBACandPicoPLEX.Significantsample-to-sample variabilityinamplifiedDNAqualitywasnoted,asmeasuredbyour8-geneQCscore the benefit of performing MDA by partitioning the reac- For single-cells, 2 of 3 WGA reactions resulted in a tion into parallel smaller volume reactions [16, 18, 22]. product with the perfect QC-score of 8, compared to 1 The small reaction volumes and template DNA partition- in6WGA reactionsusing the conventional MDAproto- ing restricts the degree of aberrant preferential amplifica- col. Importantly, for 5-cell and 10-cell samples, all WGA tion, leading to a more uniform WGA process overall. reactions generated productsthat passed our QC criteria Based on this principle, we developed a modified MDA (Fig. 3b). The modified MDA protocol in our hands protocol with key changes in two aspects: (1) increasing effectively reduced the “threshold” number of cells the cell lies step from 10 min to a minimum of neededforareliableWGAreactiondownto5cellsfrom 30 min to ensure adequate release of genomic mate- 20cells,wellwithinthenumberofCTCsattainablefrom rials from cells, and (2) partitioning the final MDA a singleperipheral blooddraw. reaction into 16 individual reactions containing ~3ul each on a 96-well plate, prior to the final isothermal WGAproductQCassayusing8-vs.16-genemultiplex amplification step at 30 °C for 8 h. PCR WGA products obtained using this modified MDA Our proposed 8-gene QC assay evaluates 8 genomic protocol resulted in improved reproducibility and higher locations within a WGA product. Evaluating more loci in QC-scores overall (Fig. 3b). Amplification reaction gain theory provides more comprehensive evaluation of the was overall lower for the modified MDA products com- amplifiedDNAproduct.Wetestedwhethertherewasany pared to the conventional MDA, with reduction of ap- benefit to evaluating more genetic loci beyond the 8- proximately 50% on average for each sample undergoing cancer genes during the QC process. To test for this, we the modified MDAprotocol (Additional file 2: Table S1). performed a secondary QC assay using a different set of Fig.3NumberofcellsusedforgenomictemplateinputandWGA-DNAquality.aConventionalMDA.OverallQC-scoresimproveasthenumber ofcellsusedforstartingcellularinputincreases.20-cellsarerequiredinordertoattainapassingQC-scoreof8reproducibly.bModifiedMDA.A passingQC-scoreof8isachievedreproduciblyfrom5-cells(asopposedto20-cells)usingthemodifiedMDAprotocol Shoetal.BMCCancer (2017) 17:457 Page7of12 8-housekeeping genes on the same single-cell conven- The same two single-cell WGA products were also tional MDA products, as well as the 1/5/10-cell modified analyzed by massive multiplex short read sequencing. MDA products (Additional file 2: Table S2A and B). The Figure 5 shows the quality metrics of sequencing data original (8-cancer gene) and the secondary (8-housekeep- associatedwitheachWGA-DNA sampleand unamplified ing genes) QC assays generated highly concordant QC- batch gDNA. When we compare the two single-cell scoresforallofthesamplestested.Allofthesamplesthat WGA products(WGA-QCpass and WGA-QCfail), Lorenz passed the QC-step based on the original QC assay also curves, histograms of read count frequency and indexes of passed using the secondary QC assay. For the sole modi- dispersion all indicated a superior NGS data quality with fied MDA sample with the low QC-score (sample 1-C), highercoverageuniformityforWGA-QCpasscomparedto the secondary QC also resulted in a similarly low QC- WGA-QCfail. The Lorenz curve provides information on score indicating poor quality WGA-DNA. Thus, evalu- theuniformityofthesequencingreadsdistribution.Perfect ation of more genetic loci beyond the original 8-cancer coverage results in a straight line with slope of 1 (y = x). genes did not provide any additional QC information and The wider the curve below the line of y = x,the lower the didnotchangeQCresultsforanyofthesamplestested. coverage uniformity and greater the amplification bias. As canbeseeninFig.5a,WGA-QCpassresultedinmoreuni- Using8-cancergeneQCassaytoselectsamplesfor formdistributionofreaddepthcomparedtoWGA-QCfail. downstreamaCGHandNGSapplications Thehistogramofreadcountfrequencyalsoprovidesinfor- Weperformedpointmutationdetection,aCGHandNGS mationoncoveragedispersion(Fig.5b).Histogramswitha using single-cell WGA products that either passed wide range of distribution, as seen in the sequencing data (WGA-QCpass) or failed (WGA-QCfail) the QC-step. obtained using WGA-QCfail, indicates a greater degree of Figures 4 and 5 illustrate the relationship between amplification bias. Genomic profiles generated from NGS QC results and performances in various downstream sequencing data are as shown in Fig. 5c. Comparison be- applications. tweenWGA-QCpassandWGA-QCfailrevealssignificantly Point mutation detection within the KRAS gene was more“noise”inthegenomicprofilederivedfromthelatter, successful in both QCpass and QCfail WGA products withagreaterindexofdispersioncomparedtothatderived (Fig. 4a). However, while the QCpass WGA sample fromWGA-QCpass. exhibited a mutant to wild-type allelic ratio closely matching that of the batch gDNA (HPAF-II cells are Determinationofthresholdnumberofcellsneededfor known to have 4 to 1 mutant:wild-type allelic ratio), the optimalperformancesinaCGHandNGSanalysis QCfail WGA sample exhibited evidence of amplification Although the single-cell WGA product passing our QC bias,demonstratedbyanalteredallelic ratio. criteria (WGA-QCpass) performed well in point muta- When we compared aCGH performances between the tion detection, aCGH and NGS, room for improvement two single-cell WGA products, WGA-QCpass resulted still existed when compared to the performance metrics in a notably better derivative log2 ratio spread (DLRS) of unamplified batch gDNA (Figs. 4 and 5). Increasing and signal-to-noise ratio compared to WGA-QCfail the number of starting cells has been shown to improve (Fig. 4d). DLRS is a key quality metric for aCGH data WGA product quality [15, 19, 22]. However, the number measuringthepoint-to-pointconsistencyor“noisiness”in ofcellsneededforgeneratingaWGAproductthatfaith- data,withhighvaluesindicatingpoorsignal-to-noiserela- fully represents the unamplified, original DNA in down- tionshipanddifficultyinassessingtruecopynumbervari- stream molecular analysisremainstobedefined. ation (CNV) status. The poor quality metrics (DLRS: 1.3, Using our modified MDA protocol, we lowered the signal-to-noise ratio: 3.9) associated with WGA-QCfail numbers of cells needed to reliably pass our QC-step renders its aCGH data unfit for meaningful analysis and down to 5 cells from 20 cells. However, it remained to interpretation. On the other hand, DLRS of 0.72 and be answered how closely 5-cell and 10-cell WGA prod- signal-to-noise ratio of 53.6 associated with WGA- ucts approximated the unamplified DNA in aCGH and QCpassmeetsthequalitythresholdforsingle-cellderived NGS analysis, and whether obtaining 5 to 10 CTCs from aCGH set forth by Agilent Technologies [23]. Gain and a single liquid biopsy is truly sufficient for sequencing loss profiles generated from aCGH data are as shown in applications within precision oncology strategies. Figure Fig. 4c. Comparison between WGA-QCpass and WGA- 4b and 5 illustrate the performances of 5-cell and 10-cell QCfail reveals disparities in single-cell aCGH profiles, modified MDA products (QC score = 8 for both) in even though clonally expanded HPAF-II cells were used aCGH and NGS. As the number of cells used for starting for both. When compared to the unamplified batch genomic template increased from 1 to 5 to 10 cells, pro- gDNA, we found multiple areas of alterations that were gressive improvement in all quality metrics of aCGH and detected with WGA-QCpass, but not with WGA-QCfail NGS were noted. Notably, the Lorenz curves for WGA- (Fig.4a,redarrows). DNA and unamplified batch gDNA nearly overlapped by Shoetal.BMCCancer (2017) 17:457 Page8of12 Fig.4PointmutationdetectionandaCGHanalysisusingasingle-cellWGAproductsthatpassed(QC-pass)orfailed(QC-fail)thequalitycontrol step,b)5and10-cellWGAproductswithpassingQC,andcunamplified,batchgenomicDNA.Single-cellWGAproducts(QC-failvs.QC-pass) exhibitsignificantlydifferentdownstreamanalysesresultsdespiteusingcellsisolatedfromaclonallyexpandedcellline.Thesingle-cellWGA productwiththepassingQC(QC-pass)generatedSangersequencingandaCGHresultsthatmorecloselyresembledthatoftheunamplified batchgDNAcomparedtothesingle-cellWGAproductfailingtheQC(QC-fail).Redarrowssignifytheareasofalterationthatweredetectedin QC-passandbatchgDNA,butnotinQC-fail.daCGHqualitymetrics(DLRSandsignal-to-noiseratio)for1,5and10-cellWGAproductsand batchgDNA.ImprovedDLRSvalueswereassociatedwithWGAproductsthatpassedtheQCstepaswellasusinganincreasingnumberof startingcellularinput 10-cells (Fig. 5a). Furthermore, by 10-cells, WGA-DNA dispersion as the number of cells used increased (Figs. 4b derivedaCGHandNGSgenomicprofilesappearedhighly and 5c). Lastly, the resolution limits of 1, 5 and 10-cell concordantcomparedtothatofunamplifiedbatchgDNA, amplified products applied to the NGS analysis were with progressive reduction in signal “noise” and index of assessed.Byevaluating for the accuracyofCNVcallingat Shoetal.BMCCancer (2017) 17:457 Page9of12 Fig.5NGS application using 1, 5 and 10-cell WGA products and batch gDNA. a Lorenz curve illustrating the amplification bias in read coverage. Lorenz curve provides information on the uniformity of the sequencing reads distribution. Perfect coverage results in a straight line with slope of 1 (y = x) as shown by the dotted line. The wider the curve below the line of y = x, the lower the coverage uniformity and greater the amplification bias. Single-cell WGA product passing the QC-step (QC-pass) is observed to have less amplification bias compared to the single-cell WGA product that failed the QC-step (QC-fail). As the number of starting cellular input increases, the degree of amplification bias becomes even less pronounced, approaching that of unamplified batch gDNA by 10-cells. b Histogram of read count distribution. Wider range of distribution without a distinct peak (as seen for single-cell WGA: QC-fail) signifies worse coverage dispersion. c Genomic profiles generated from NGS sequencing data. Significantly less “noise” is observed with single-cell QC-pass when compared to single-cell QC-fail. Increasing the number of starting cellular input further improves the data quality, as reflected in the decreasing index of dispersion Shoetal.BMCCancer (2017) 17:457 Page10of12 various bin sizes (250 kb, 500 kb, 1 Mb and 2.5 Mb), we cancer therapy. The eight genes included in our QC found 5 and 10-cells to require a minimum bin size of assay are all considered important molecular targets for approximately250kbtoreproducethebatchDNAresult. cancer therapy, and under active investigation in the For single-cell, bin size of approximately 2.5 Mb was National Cancer Institute – Molecular Analysis for needed(Additionalfile3:Fig.S1).Insummary,using5–10 Therapy CHoice (NCI-MATCH) trial [28–31]. Thus, cells as the starting template for our modified MDA reac- evaluation for WGA-DNA quality using this QC assay tion generated WGA-DNA that closely approximated the helps identify samples suitable for potential use in preci- unamplifiedgDNAinbothaCGHandNGSperformances. siononcology strategies. Along with the 8-cancer gene QC-assay, we have also Discussion developed a modified MDA protocol in order to help CTC analysis offers unprecedented potentials for fur- reduce the number of cells needed for a reliable WGA thering precision oncology. Realization of a “liquid bi- reaction. Our modified MDA protocol based on the opsy” through CTC sequencing helps avoid invasive and principle of small volume MDA required only a 96-well costly traditional biopsy procedures. Moreover, it allows plate and negated the need for labor-intensive protocols for a dynamic monitoring of evolution in tumor genome and costly special equipment described in previous in response to cancer therapy [24]. However, most works. Although this meant that we could not achieve downstream applications (i.e. aCGH, NGS) necessitate a the nanoliter reaction volumes described in prior stud- whole genome amplification (WGA) step prior to the ies, we still observed a meaningful improvement in analysis of single/few-cell, which is known to introduce sample-to-sample reproducibility and overall improve- errors and biases [11–14, 25]. In the current study, we ment in our defined QC-scores. Importantly, we noted describedour strategic approach for selectingand gener- reduced amplification reaction gains associated with our ating optimal WGA products for analysis using aCGH modified MDA protocol, consistent with observations and NGS. We presented and validated a quality control made inpriorliteratures onsmallvolumeMDA[18, 22]. assay for WGA product quality, and introduced a modi- As previously described, volume restriction limits ampli- fied MDA protocol that helped improve the reproduci- fication reaction and decreases the overall reaction gain. bility and reliability of the existing WGA process. However, this process can also restrict the degree of Finally, we showed that by combining our QC-criteria, aberrant amplification of certain preferred sequences, modified MDA protocol and using as little as 5–10 cells resulting in a more uniform product overall. Although as a starting template, a WGA product with high fidelity an excessively low DNA yield may signify either a poor totheunamplifiedtemplate DNAcouldbeobtained. starting DNA quality or an ineffective amplification Multiple groups have published on performance com- process, an exceedingly high amplification gain is also parison of different WGA methods over the recent associated with an increased amplification bias [22]. Par- years, with significant differences in results and conclu- ticularly in MDA, reaction gain >107 has been shown to sions existing between these studies [14, 16, 26]. How- correlate with poor amplification quality [16]. These ever, the majority of these studies lack analysis of the findings suggest that the degree of amplification gain, or quality of WGA samples being used. Given the known DNA yield, is likely an important parameter of amplified high sample-to-sample variability in single-cell WGA DNAquality. quality, no meaningful interpretation and comparison of In addition to improving the WGA process itself, WGA-DNA derived data can be made without a well bioinformatic computational tools are also available to defined quality standard. Our proposed 8-cancer gene improve the quality of sequencing data. A number of QC assay has the potential to fulfill this gap, as it suc- algorithms have been described to date, each with estab- cessfully predicts the performances of WGA-DNA in lished efficacy in optimizing the sequencing data from downstream analysis by aCGH and NGS, in addition to single/few-cell amplified DNA [21, 32–35]. We utilized providinglimitedsequencinginformation. the Ginkgo (http://qb.cshl.edu/ginkgo) platform in our Prior reports have described multiplex PCR based QC current analysis, which incorporates built-in algorithms assay for formalin-fixed paraffin embedded (FFPE) DNA designed specifically to address the challenges associated and Ampli1 WGA-DNA [20, 27]. The Ampli1 WGA is a with single/few-cell amplified DNA [21]. Notably, des- PCR-based amplification process using non-random pite the data optimization achieved by the Ginkgo algo- primers, which is fundamentally dissimilar to the MDA rithm, the failed QC sample remained highly biased and method of non-PCR based isothermal amplification was unsuitable for accurate interpretation. Although process using random primers. To date, a QC assay statistical correction algorithms remain highly useful in specific to the MDA method remains to be defined. Fur- single/few-cell amplified DNA analysis, generation and thermore, no study to date has described a QC assay selection of optimal amplified DNA likely remains a key designed specifically to evaluate for genes implicated in factor insuccessfuldownstream sequencing analysis.

Description:
Keywords: Precision oncology, Whole genome amplification, Single-cell sequencing, Next generation sequencing,. Multiple displacement molecular analysis has been described, major obstacles still remain prior to its clinical .. Based on this principle, we developed a modified MDA protocol with key
See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.