Open Access

A technical application of quantitative next generation sequencing for chimerism evaluation

  • Authors:
    • Michelangelo Aloisio
    • Danilo Licastro
    • Luciana Caenazzo
    • Valentina Torboli
    • Angela D'Eustacchio
    • Giovanni Maria Severini
    • Emmanouil Athanasakis
  • View Affiliations

  • Published online on: August 4, 2016     https://doi.org/10.3892/mmr.2016.5593
  • Pages: 2967-2974
  • Copyright: © Aloisio et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

At present, the most common genetic diagnostic method for chimerism evaluation following hematopoietic stem cell transplantation is microsatellite analysis by capillary electrophoresis. The main objective was to establish, through repeated analysis over time, if a complete chimerism was present, or if the mixed chimerism was stable, increasing or decreasing over time. Considering the recent introduction of next generation sequencing (NGS) in clinical diagnostics, a detailed study evaluating an NGS protocol was conducted, coupled with a custom bioinformatics pipeline, for chimerism quantification. Based on the technology of Ion AmpliSeq, a 44‑amplicon custom chimerism panel was designed, and a custom bioinformatics pipeline dedicated to the genotyping and quantification of NGS data was coded. The custom chimerism panel allowed identification of an average of 16 informative recipient alleles. The limit of detection of the protocol was fixed at 1% due to the NGS background (<1%). The protocol followed the standard Ion AmpliSeq library preparation and Ion Torrent Personal Genome Machine guidelines. Overall, the present study added to the scientific literature, identifying novel technical details for a possible future application of NGS for chimerism quantification.

Introduction

Allogenic hematopoietic stem cell transplantation (HSCT) is the predominant treatment used to cure malignant and non-malignant hematological disorders. The number of HSCTs conducted has increased due to an overall improvement in the safety of the procedure resulting from reduced-intensity conditioning regimens, and the availability of new donor sources, available in the national registries (1,2).

Reduced-intensity conditioning is widely used to avoid the complications of myeloablative conditioning to prepare for HSCT in the adult population. However, this procedure is associated with a high risk of complications that may result in graft loss. To prevent this, it is important to monitor chimerism for early intervention (3). Evaluation of chimerism status at regular intervals is useful to prevent risk of early graft rejection and relapse in patients suffering from malignant diseases. Quantification of the chimerism percentage is also a potential marker of minimal residual disease (MRD) for patients without suitable MRD markers and it provides useful information on graft vs. host disease and graft vs. tumor effects (46).

Chimerism analysis is a tool that allows to determination of the genotypic origin of post-transplantation hematopoiesis. Subsequent to HSCT, a patient presenting with 100.0% donor-origin cells during follow up is considered to have the status of complete chimerism (CC), patients in which the donor- and recipient-origin cells coexist have the status of mixed chimerism (MC) (7). Informative genetic markers are used to discriminate between recipient and donor genomes in order to detect the chimerism status (8).

At present, different approaches based on polymerase chain reaction (PCR) amplification of polymorphic DNA sequences (short tandem repeat, STR; single nucleotide polymorphism, SNP; and insertion/deletion, INDEL) are used for chimerism analysis. In the vast majority of laboratories, semi-quantitative fluorescent PCR of STRs is the procedure of choice for diagnostic purposes. The key advantage offered by this method is the highly polymorphic nature of the STR markers, which allow for a high probability of two-genome discrimination. Laboratories currently use different commercial multiplex kits for forensic identifications or in house assays, however have formed consortiums to standardize procedures and set guidelines for the correct interpretation of results, in order to improve this intrinsically semi-quantitative platform with a sensitivity of 1.0–3.0% (9).

Next generation sequencing (NGS) technologies are an innovation in human and animal genomics research, as they are capable of producing 100-fold more data than the most powerful Sanger based capillary sequencers; thus enabling researchers to investigate the large number of queries that remain to addressed (10).

NGS generates hundreds of giga-bases of nucleotide sequences per instrument run and produces this data at a lower cost, thus motivating researchers to use NGS for various purposes: To identify rare variations on the whole genome or on a target sequence, to analyze transcriptome profiling of cells, tissues and organisms and to identify epigenetic markers for disease diagnosis. Progress in the optimization of procedures, in addition to further reduction of costs, are the key factors that will lead to a more extensive uptake of this technique in diagnosis and for practical clinical applications.

NGS provides qualitative and quantitative data. Quantitative data depends on the depth of sequence data collected on each sample and on the quality of the target to expose. For samples with a lower abundance target, many more sequence reads are required to achieve accurate quantification (11). A previous study demonstrated that NGS exhibits sensitivity comparable to that of quantitative PCR (qPCR) in the evaluation of MRD in B cell disorders (12).

In the present study, an Ion AmpliSeq custom chimerism (ACCh) panel and a custom bioinformatic pipeline was created for chimerism quantification by NGS. The first aim was to detect the existence of cells of two origins in chimera samples and then to evaluate the capability of NGS to determine the percentage of the recipient cells.

Materials and methods

DNA sample preparation

The Ethics Committee of the Institute for Maternal and Child Health, IRCCS 'Burlo Garofolo' approved the present study (approval number: Prot. 18/2015, Cl. M/11). Written informed consent was obtained from all the participants.

Total peripheral blood was collected from 10 volunteer donors (V01-V10; 4 males and 6 females) with ages ranging from 20–50 years, and from 2 pediatric patients that underwent allogeneic HSCT (pR1, male, 5 years; and pR2, male, 12 years) and their donors (pD1, male, 9 years; and pD2, female, 25 years; Table I). Written informed consent was obtained from all the participants.

Table I

List of the patient DNA samples used in the present study.

Table I

List of the patient DNA samples used in the present study.

DNA IDChimera IDChimera informationNotes
pD1pR1Samples used to evaluate panel informativity in consanguinityBrothers, pre-HSCT
pD2pR2pCh1MC by STR-CE analysisa+1 month post-HSCT
pD2pR2pCh2CC by STR-CE analysis+2 months post-HSCT
pD2pR2pCh3MC by STR-CE analysis+3 months post-HSCT
pD2pR2pCh4MC by STR-CE analysis+4 months post-HSCT
pD2pR2pCh5MC by STR-CE analysis+6 months post-HSCT
pD2pR2pCh6CC by STR-CE analysis+10 months post-HSCT

a Chimerism evaluation of all patient samples (pCh1–6) was performed by STR-CE analysis in an external laboratory (Department of Molecular Medicine, University of Padova, Padova, Italy). HSCT, hematopoietic stem cell transplantation; MC, mixed chimerism; CC, complete chimerism; STR-CE, short tandem repeat capillary electrophoresis.

All DNA samples were isolated using the QIAamp DNA Blood kit according to manufacturer's protocol (Qiagen GmbH, Hilden, Germany). The DNA status was evaluated using the NanoDrop 1000 spectrophotometer (Thermo Fisher Scientific, Inc., Waltham, MA, USA) and the Qubit dsDNA HS Assay kit and the Qubit fluorometer (Thermo Fisher Scientific, Inc.). A DNA stock solution was prepared for all DNA samples at 20.0 ng/μl.

DNA samples from the 10 volunteer donors were randomly paired. A total of 5 artificial chimeric DNA mixtures, as the donor/recipient chimera, were created by diluting DNA with its paired DNA at several percentages of the first DNA for each artificial DNA mixture (aCh).

Ion AmpliSeq custom chimerism panel design

A multi phase strategy was employed to evaluate the main characteristics of the ACCh panel: i) The panel average heterozygosity was assessed around 0.5 for the European population (HapMap Phase 3 CEU population); ii) two SNPs per somatic chromosome, termed 'main SNPs' (mSNPs), were selected and located in two different regions of the same chromosome; iii) the amplicon composition was evaluated according to the following requirements: a) GC percentage ranging between 40.0 and 60.0%; b) presence of one mSNP inside each amplicon; c) mSNP location preferably in the centre of the amplicon; d) absence of INDEL SNPs; e) absence of homopolymers and potential homopolymer generation from SNP variants and their flanking regions; f) absence of flanking SNPs to the mSNPs.

In total, 44 single-nucleotide, biallelic, polymorphisms were selected from the NCBI dbSNPs database (http://www.ncbi.nlm.nih.gov/SNP/, build 138, last database update 28.03.2014; Table II). A total of 4 base sequences including mSNPs were used as target regions for primer design. The primer pool, intended for DNA library construction through multiplex PCR, was defined by Ion AmpliSeq Designer software, version 3.0.1 (Thermo Fisher Scientific, Inc.). A single-tube, 44 primer pair pool was purchased from Life Technologies (Thermo Fisher Scientific, Inc.).

Table II

List of all main SNPs included in the Ion AmpliSeq custom chimerism panel.

Table II

List of all main SNPs included in the Ion AmpliSeq custom chimerism panel.

SNP IDGenome positionAllelesEuropean heterozygosityInformativity of recipient allelea %
rs12070036chr1:g.227819514A/G0.40741
rs1234315chr1:g.173178463C/T0.51337
rs10496711 chr2:g.134516742C/G0.40740
rs12612347 chr2:g.219057338A/G0.44240
rs1984630 chr3:g.134414219G/T0.52236
rs9831477 chr3:g.30693522A/T0.48338
rs10033900 chr4:g.110659067C/T0.49637
rs5335 chr4:g.148463840C/G0.49237
rs983889 chr5:g.15555486A/C0.48738
rs10038113 chr5:g.25902342C/T0.46938
rs552655 chr6:g.13370488A/G0.50437
rs2077163 chr6:g.33636907C/T0.46039
rs39395 chr7:g.103489729A/G0.42540
rs2270188 chr7:g.116140524G/T0.49638
rs10505477 chr8:g.128407443C/T0.53136
rs532841 chr8:g.12957475C/T0.54935
rs2297313 chr9:g.91669362A/G0.96037
rs424539 chr9:g.14442595C/G0.46738
rs1561570 chr10:g.13155726C/T0.52236
rs619824 chr10:g.104581288G/T0.40741
rs198464 chr11:g.61521621C/T0.50437
rs178503 chr11:g.44082931A/G0.44240
rs1126758 chr12:g.103248924A/G0.41640
rs8608 chr12:g.53294381A/G0.52236
rs1061472 chr13:g.52524488A/G0.50437
rs504544 chr13:g.19735891A/T0.50837
rs10143250 chr14:g.104723433C/T0.43439
rs1957779 chr14:g.63669647C/T0.44939
rs634990 chr15:g.35006073A/G0.49238
rs2117215 chr15:g.94879684C/T0.60332
rs121893 chr16:g.66183995C/T0.41439
rs2191125 chr16:g.7720923T/C0.55034
rs6808 chr17:g.62400575C/G0.45039
rs744166 chr17:g.40514201T/C0.44140
rs620898 chr18:g.48509148A/T0.46738
rs633265 chr18:g.57831468A/C0.49637
rs108295 chr19:g.34224816A/G0.49637
rs892086 chr19:g.10837677C/T0.45139
rs753381 chr20:g.39797465T/C0.45139
rs715147 chr20:g.50055350G/A0.36743
rs225436 chr21:g.43729034A/G0.51736
rs8128316 chr21:g.35721560C/T0.54235
rs4444 chr22:g.31205334A/G0.48338
rs132985 chr22:g.38563471C/T0.51737

a In order to calculate the probability (informativity) to identify an informative recipient allele in a casual donor/recipient pair, the single values of probability of each genotype combination that carry an informative recipient allele were added as follows: Donor homozygous and recipient heterozygous [Donor (AA) and Recipient (Aa); Donor (aa) and Recipient (Aa)]; donor and recipient homozygous but for different alleles [Donor (AA) and Recipient (aa); Donor (aa) and Recipient (AA)]. SNP, single nucleotide polymorphism; A, adenine; G, guanine; C, cytosine; T, thymine.

Ion torrent library preparation and sequencing

DNA sample library preparation was performed according to the AmpliSeq Library Preparation protocol (Life Technologies; Thermo Fisher Scientific, Inc.). For each DNA sample, a library was constructed using 10.0 ng genomic DNA through the Ion AmpliSeq Library kit, version 2.0. Subsequently, according to the library preparation protocol, each DNA library was indexed using the Ion Xpress Barcode Adapters kit (Thermo Fisher Scientific, Inc.) and was purified using AMPure XP magnetic beads (Beckman Coulter, Inc., Brea, CA, USA). Each DNA library was then quantified by qPCR using the thermo-cycler 7900HT Fast Real-Time PCR system with the Ion Library TQMN Quantification kit (Thermo Fisher Scientific, Inc.). Template Ion Sphere Particles were arranged using the Ion Personal Genome Machine (PGM) Template OT2 200 kit (Thermo Fisher Scientific, Inc.) and a single end 200 base-read sequencing run was conducted using the Ion Torrent PGM system. Libraries were pooled at 8 pM using the following rates: Donor/recipient, 1:1; and chimera/chimera, 1:1. The recipient/chimera rate was fixed at 1:40 in order to obtain an average coverage of the above libraries around 250X:10,000X. Library pools were sequenced on ion 314 and 316 chip (Table III).

Table III

List of the library pools sequenced by next generation sequencing.

Table III

List of the library pools sequenced by next generation sequencing.

DNA IDChimera IDChimera notesIon chip
V01, V02 aCh1Chimera: 1.0% of V01; 99.0% of V02314
V03, V04 aCh2Chimera: 1.25% of V03; 98.75% of V04314
V03, V04 aCh3Chimera: 2.5% of V03; 97.5% of V04314
V05, V06 aCh4Chimera: 5.0% of V05; 95.0% of V06314
V05, V06 aCh5Chimera: 10.0% of V05; 90.0% of V06314
V07, V08 aCh6,7,8,9,10,11Chimeras: 0.5%, 1.0%, 4.0%, 8.0%, 12.0%, 20.0% of V07; 99.5%, 99.0%, 96.0%, 92.0%, 88%, 80.0% of V08316
V09, V10 aCh12,13Chimeras: 40.0% of V09 and 100% of V09; 60.0% of V10 and 0.0% of V10316
pD2, pR2 pCh1,2,3,4,5,6Chimeras: MC, CC, MC, MC, MC, CC of pR2a316
pD2, pR2 pCh2Technical replication314
pD1, pR1 pCh6Technical replication314

a All patient samples were previously analyzed by short tandem repeat capillary electrophoresis in an external laboratory (Department of Molecular Medicine, University of Padova, Padova, Italy).

Hotspot panel bed file

A hotspot panel bed file was created using the UCSC Genome Browser (https://genome.ucsc.edu). All SNPs located in the central region of each amplicon were included (NCBI dbSNPs build 138; 'Common SNPs' =286). All the INDELs present across the amplicons, and the SNPs near the 5′ and 3′ ends of the amplicons were excluded from the file. All the above 44 selected SNPs were marked as 'mSNP' and the SNPs belonging to the same amplicon were indexed with the same chromosome/amplicon ID number. Finally, a hotspot panel bed file was created: 'HP286SNPs'.

Genotyping and quantification

Genotyping of all DNA was performed automatically, together with the quantification of all the chimeras, using a custom bioinformatics tool. The code of our tool was written using the Shiny package in R, a web framework to build interactive web applications (https://cran.r-project.org/web/packages/shiny/index.html). A functional diagram of the code is presented in Fig. 1 and the full code is available on request. The code is based on dependencies of the Bioconductor package that must be pre-installed for proper tool functionality.

The custom tool requires as input the sequencing bam files of 'Donor', ' Recipient' and 'Chimeric' patient. Briefly, it uses readGAlignments and pileLettersAt functions, from the GenomicAlignments package (13), to read bam files and extracts the letters/nucleotides into a set of individual genomic positions defined from the bed file. Thresholds for 'Donor' and 'Recipient', homozygous and heterozygous genotyping calls, are settled in base counts frequency ranges of 90.0–100.0% and 30.0–60.0%, respectively. Genotyping calls not included in the thresholds ranges were excluded as unreliable; users can modify the thresholds according to their needs from the user interface. Genotypes from each library were crosschecked to select only SNPs comparable in all conditions. Selected SNPs from donors and recipients were labelled as informative recipient alleles (IRA) according to the following schema: Donor homozygous and recipient heterozygous [Donor (AA) and Recipient (Aa); Donor (aa) and Recipient (Aa)]; donor and recipient homozygous for different alleles [Donor (AA) and Recipient (aa); Donor (aa) and Recipient (AA)].

Only the IRA SNPs tagged as informative were used to calculate the chimera's donor:recipient ratio as median of the allele frequency ratio, while standard error was used to calculate confidence intervals of prediction at 95.0%.

To cross validate the tool, genotyping of all donor and recipient samples was also performed manually obtaining the variant data from the Ion Torrent plugin Variant Caller, version 4.4 using the 'Generic-PGM-Germ Line-Low Stringency' configuration coupled by the HP286SNPs hotspot bed file.

Microsatellite analysis and patient data validation

Multiplex PCR amplification of V01-V10 and aCh1–13 samples, in addition to the patient samples pD2, pR2 and pCh1–6, was performed according to the manufacturer's instructions of the AmpFlSTR Identifiler Plus PCR Amplification kit (Thermo Fisher Scientific, Inc.). Amplicons were resolved on a Genetic Analyzer 3130 and analyzed with GeneMapper software, version 4.1 (Life Technologies; Thermo Fisher Scientific, Inc.).

Patient samples (pCh1–6) were also analyzed by qPCR (data not shown), as previously investigated by Bai et al (14). This analysis was performed as an additional validation method of NGS data, where a discrepancy between NGS and STR data was present.

Results

Ion chips and ACCh panel performance

A total of 7 library pools were loaded and sequenced on ion chip 314. Each pool was comprised of one donor, one recipient and one chimera. In addition, three additional pools were run on ion 316 chip (Table III). The mean values of performance of the 10 runs and of all samples are summarized in Table IV.

Table IV

In-house Ion Torrent Personal Genome Machine analysis observed average performances using the Ion AmpliSeq custom chimerism panel on ion 314 and 316 chips.

Table IV

In-house Ion Torrent Personal Genome Machine analysis observed average performances using the Ion AmpliSeq custom chimerism panel on ion 314 and 316 chips.

A, In-house observed performanceIon 314 chipIon 316 chip
Ion sphere particles loading84.3%70.0%
Total bases (Mb)105.1677.0
Total reads533,5353,474,065
Reads on-target99.7%99.3%
Panel uniformity97.6%96.0%
B, Coverage performanceIon 314 chipIon 316 chip
Chimera samples - amplicons over 2,500X43/44 range 4,069X–23,944X)43/44 (range 4,777X–28,314X)
Donor & recipient samples - amplicons over 50X44/44 (range 105X–952X)44/44 (range 75X–2328X)
NGS genotyping performances using the ACCh panel

A total of 14 DNA samples were genotyped on Ion Torrent PGM using the ACCh panel with the HP286SNPs bed file. Regarding the mSNPs, the Variant Caller output identified that 2 mSNPs (rs121893 and rs12612347) were assigned as 'No Call' in over 50.0% of the genotyping runs due to low quality. The remaining 42 mSNPs were successfully genotyped. Concerning the remaining 242 SNPs, 27 SNPs were assigned as 'No Call', with an average of 10 SNPs per patient. The call of these SNPs failed in two Variant Caller filtering steps: 'Maximum common signal shift' and 'minimum coverage on either strand'.

Genotyping of all samples was additionally performed using our tool with the HP286SNPs bed file (Table V). To identify the IRAs, the data of the donor was compared with the recipient using the tool and manually cross-validated with the Variant Caller genotypes. Inside the genotyping calls of 242 SNPs, a small bias was present between these 2 tools; this is due to the high conserved filters of variant caller, dedicated predominantly for standard sequencing applications, and due to the absence of these filters in our custom tool.

Table V

List of the number of IRAs identified in each DNA pair using the custom pipeline.

Table V

List of the number of IRAs identified in each DNA pair using the custom pipeline.

DNA pair 'Donor'/'Recipient'Custom pipeline
IRA genotypes
Total IRAs (%)a
44 mSNPs242 SNPsHeterozygousHomozygous
V01/V0215310818 (43)
V03/V0418419322 (52)
V05/V0614414418 (43)
V07/V0813510818 (43)
V09/V10838311 (26)
V02/V0115512820 (48)
V04/V0372639 (21)
V06/V0514212416 (38)
V08/V0716412820 (48)
V10/V0919319322 (52)
pD1/pR1929211 (26)
pD2/pR217418321 (50)

a The percentage of each IRA was calculated on 42–44 amplicons, due to the low quality of the remaining two, considering that each amplicon could contain at least one potential IRA. IRA, informative recipient allele; mSNP, main single nucleotide polymorphism.

NGS linearity, detection limit and accuracy with the ACCh panel

In order to test the linearity of Ion Torrent PGM with the ACCh panel in a fixed detection range (0.5–100.0%), a series of DNA mixtures was developed, diluting a DNA with its paired DNA at several percentages of the original. In order to increase the genetic marker variability in addition to the biological variability, a total of 12 artificial chimeras (aCh1–12) were prepared from 5 different DNA pairs. Finally a pure DNA (V09) was run as 100.0% DNA (aCh13). Subsequent to Ion Torrent sequencing, using the custom tool, quantitative data for all IRAs of each artificial chimera were obtained.

In addition, to increase the putative points in the dynamic range, the informative alleles of both DNA in the chimeras aCh12 (40.0% of V09 and 60.0% of V10) and aCh11 (20.0% of V07 and 80.0% of V08) were calculated and quantified.

Least-squares analysis of the above putative points identified a clear linearity (R2=0.999; Y=1.008X-0.005) between NGS and the reference values (Fig. 2).

Analyzing the artificial chimeras aCh6–13 by capillary electrophoresis using the STRs markers, chimerism ranging from 4.0–100.0% was detected. Least-squares analysis identified a clear linearity (R2=0.999; Y=1.012X-0.009) between NGS and STRs values (Fig. 3).

In addition, the background of Ion Torrent generated by the ACCh panel was estimated. In this case, the custom tool was used, considering the 'donor' samples as chimera. The average background value at each SNP was estimated at 0.3% (range, 0.0–0.8%) and with a 95.0% confidence interval between 0.1 and 0.5%. Considering the background values and the reported literature on the error rates at each base of NGS technologies (range from 0.04–1.0%) (15), the detection limit of the NGS protocol with the ACCh panel was set at 1.0%, although an artificial chimera was detected at 0.5%.

Finally, considering that the method determined each chimera, calculating the average value of all IRA, the average standard error was used as an indirect marker of accuracy, using the data of all artificial chimeras ranging from 1.0–99.0% (excluding 0.5 and 100.0%). For the dynamic range of 1.0–20.0% of chimeras, the average standard error was calculated at 0.3% with a deviation at 0.2%. For higher values of chimeras, up to 99.0%, the standard error increased up to 1.8% with a maximum deviation at 2.0%.

Patient chimerism evaluation on the NGS platform

Considering the linearity between NGS and reference values and between NGS and STR values using the standard NGS workflow and the custom tool, our workflow was tested in 6 samples of the same patient (pCh1–6) in which the chimerism quantification report, previously performed by microsatellite analysis at different times in an external laboratory (Department of Molecular Medicine, University of Padova, Padova, Italy), presented at least one CC between two MCs (Table I).

NGS analysis detected a mixed chimerism in all samples (pCh1, pCh2, pCh3, pCh4, pCh5 and pCh6) while the microsatellite analysis only in 4 of them (pCh1, pCh3, pCh4 and pCh5). For these 4 MC samples, the percentage of predicted chimerism was equal between the 2 methods of analysis (Fig. 4). In regard to the pCh2 and pCh6 samples, NGS analysis evaluated a mixed chimerism at 3.0 and 2.0%, respectively (Fig. 4).

To confirm the obtained NGS data for the pCh2 and pCh6 samples, the NGS analysis was repeated, and qPCR was additionally perfomed in all patient samples. The results of qPCR analysis were in agreement with that of the NGS and microsatellite data for the pCh1, pCh3, pCh4 and pCh5, and confirmed the NGS data for the pCh2 and pCh6 samples.

Discussion

NGS technologies have revolutionized the field of genomics, and its application has been extended to different fields such as clinical diagnostics and forensic science (1517). As a result of the continuous development of NGS, several applications previously performed on Sanger sequencing with capillary electrophoresis have been transferred onto the NGS platform, enabling fast and cost-effective generation sequence data with high resolution and accuracy. For this reason, different panels are being developed for the sequencing of genetic mutations involved in human diseases (e.g. MiSeqDx Cystic Fibrosis Clinical Sequencing Assay; Illumina, Inc., San Diego, CA, USA) or in cancer (Ion AmpliSeq BRCA1 and BRCA2 Panel; Life Technologies; Thermo Fisher Scientific, Inc.).

In the field of chimerism quantification by NGS platforms, Debeljak et al (18) reported an innovative and well performed study, using haplotype counting. In addition, Kim et al (19) briefly reported a relative quantification analysis of SNP markers by NGS in one human bone marrow chimerism sample. However, the study by Kim et al (19) was conducted in a 4.9% chimerism sample without any detail concerning the limit of detection, the technical error or additional important technical information and validation data of NGS application in chimerism quantification.

In the present study, a full workflow was designed, and the proposed protocols and a bioinformatics tool were tested for chimerism quantification by NGS. A 44-amplicon custom chimerism panel based on Ion AmpliSeq technology was designed, and in addition a bioinformatics tool dedicated to the genotyping and quantification of NGS data was coded. These resources were created in order to provide a novel tool for the evaluation of the chimerism following allogenic HSCT, thus potentially increasing the number of clinical analyses supported on NGS platforms.

The ACCh proposed panel is composed of 44 amplicons, containing 44 selected mSNPs, of which 2 mSNPs are located in different regions of each somatic chromosome. It is suggested that the different mSNP locations in all somatic chromosome may be useful to avoid predominantly false negatives results caused by chromosomal deletions characteristic of certain malignancies (20). In addition, the bed file uploaded in the custom tool, containing all targeted SNPs, can be modified in order to exclude the SNPs present in chromosome target regions subjected to deletions in a specific patient.

The panel average heterozygosity was assessed around 0.5 for the European population in order to obtain different informative markers for each transplanting pair, for a more precise and robust quantification. The theoretical panel informativity for unrelated donor:recipient values, calculated according to the data present on the NCBI dbSNPs database, was estimated to be approximately 16/42 mSNPs, while for siblings the informativity was estimated at 50.0% (approximately 8/42 mSNPs). In order to increase the informativity of the ACCh panel, an additional 242 selected SNPs present in the targeted regions were included in the bed file. This addition of SNPs experimentally increased the average informativity (Table V).

The ACCh panel reached the limit of detection on the Ion Torrent PGM platform of 0.5%, however, this was updated to the conservative value of 1.0% for two reasons: i) The Ion Torrent error is defined to be between 0.04 and 1.0% (21); and ii) the background of the ACCh panel, based on the IRA data of our experiments, ranges between 0.1 and 0.5%.

Regarding the timing of chimerism analysis, the UK NEQAS Consortium has recommended that results should be assessed in 5 working days from the reception of the sample and in 3 working days for urgent requests (6). The protocol suggested in the current study is feasible in 2 days; the first day for library preparation and quantification and the second for template preparation, run sequence and data analysis. In addition, due to the fact the ACCh protocol suggested in the current study does not present any differences from the standard AmpliSeq Library Preparation and Ion Torrent PGM Run Sequence protocols, it is possible to introduce it to a standard manual library preparation workflow or in a library preparation workstation.

Considering the AmpliSeq library construction protocol (based on multiplex PCR) from Life Technologies (Thermo Fisher Scientific, Inc.), the same concept could be tested on additional NGS platforms, such as Illumina or Roche, according to their library preparation protocols. Concerning the custom bioinformatics tool, any bam and bai file coupled by a bed file, generated from any platform could be used.

At present, the cost of NGS analysis, compared with microsatellite methods, remains high, however considering the continuously reducing cost per NGS run, an NGS-based method for chimerism quantification could be evaluated in the future for its adoption in laboratories with a high volume of activity, and with NGS platforms already in use for other purposes. Although the present study reported a clear correlation between NGS and STR methods and identifed important technical details, further experimental replications are required in order for the NGS protocol to be validated for future laboratory use.

Acknowledgments

The current study was supported by a grant from the Institute of Maternal and Child Health-IRCCS 'Burlo Garofolo' (grant no. RC 16-2014). The authors would like to thank Dr Erika Ferrari for assistance with SNP data crosschecking.

References

1 

Gratwohl A, Baldomero H, Gratwohl M, Aljurf M, Bouzas LF, Horowitz M, Kodera Y, Lipton J, Iida M, Pasquini MC, et al: Quantitative and qualitative differences in use and trends of hematopoietic stem cell transplantation: A Global Observational Study. Haematologica. 98:1282–1290. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Passweg JR, Baldomero H, Gratwohl A, Bregni M, Cesaro S, Dreger P, de Witte T, Farge-Bancel D, Gaspar B, Marsh J, et al: The EBMT activity survey: 1990–2010. Bone Marrow Transplant. 47:906–923. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Oshrine BR, Olson TS and Bunin N: Mixed chimerism and graft loss in pediatric recipients of an alemtuzumab-based reduced-intensity conditioning regimen for non-malignant disease. Pediatr Blood Cancer. 61:1852–1859. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Lawler M, McCann SR, Marsh JC, Ljungman P, Hows J, Vandenberghe E, O'Riordan J, Locasciulli A, Socié G, Kelly A, et al: Serial chimerism analyses indicate that mixed haemopoietic chimerism influences the probability of graft rejection and disease recurrence following allogeneic stem cell transplantation (SCT) for severe aplastic anaemia (SAA): Indication for routine assessment of chimerism post SCT for SAA. Br J Haematol. 144:933–945. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Terwey TH, Hemmati PG, Nagy M, Pfeifer H, Gökbuget N, Brüggemann M, Le Duc TM, le Coutre P, Dörken B and Arnold R: Comparison of chimerism and minimal residual disease monitoring for relapse prediction after allogeneic stem cell transplantation for adult acute lymphoblastic leukemia. Biol Blood Marrow Transplant. 20:1522–1529. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Clark JR, Scott SD, Jack AL, Lee H, Mason J, Carter GI, Pearce L, Jackson T, Clouston H, Sproul A, et al: Monitoring of chimerism following allogeneic haematopoietic stem cell transplantation (HSCT): Technical recommendations for the use of short tandem repeat (STR) based techniques, on behalf of the United Kingdom national external quality assessment service for leucocyte immunophenotyping chimerism working group. Br J Haematol. 168:26–37. 2015. View Article : Google Scholar

7 

Bader P, Kreyenberg H, Hoelle W, Dueckers G, Handgretinger R, Lang P, Kremens B, Dilloo D, Sykora KW, Schrappe M, et al: Increasing mixed chimerism is an important prognostic factor for unfavorable outcome in children with acute lymphoblastic leukemia after allogeneic stem-cell transplantation: Possible role for pre-emptive immunotherapy? J Clin Oncol. 22:1696–1705. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Gineikiene E, Stoskus M and Griskevicius L: Single nucleotide polymorphism-based system improves the applicability of quantitative PCR for chimerism monitoring. J Mol Diagn. 11:66–74. 2009. View Article : Google Scholar :

9 

Lion T, Watzinger F, Preuner S, Kreyenberg H, Tilanus M, de Weger R, van Loon J, de Vries L, Cavé H, Acquaviva C, et al: The EuroChimerism concept for a standardized approach to chimerism analysis after allogeneic stem cell transplantation. Leukemia. 26:1821–1828. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Pareek CS, Smoczynski R and Tretyn A: Sequencing technologies and genome sequencing. J Appl Genet. 52:413–435. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Xuan J, Yu Y, Qing T, Guo L and Shi L: Next-generation sequencing in the clinic: Promises and challenges. Cancer Lett. 340:284–295. 2013. View Article : Google Scholar

12 

Ladetto M, Brüggemann M, Monitillo L, Ferrero S, Pepin F, Drandi D, Barbero D, Palumbo A, Passera R, Boccadoro M, et al: Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders. Leukemia. 28:1299–1307. 2014. View Article : Google Scholar

13 

Lawrence M, Huber W, Pagès H, Aboyoun P, Carlson M, Gentleman R, Morgan MT and Carey VJ: Software for computing and annotating genomic ranges. PLoS Comput Biol. 9:e10031182013. View Article : Google Scholar : PubMed/NCBI

14 

Bai L, Deng YM, Dodds AJ, Milliken S, Moore J and Ma DD: A SYBR green-based real-time PCR method for detection of haemopoietic chimerism in allogeneic haemopoietic stem cell transplant recipients. Eur J Haematol. 77:425–431. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Mwaigwisya S, Assiri RA and O'Grady J: Emerging commercial molecular tests for the diagnosis of bloodstream infection. Expert Rev Mol Diagn. 15:681–692. 2015. View Article : Google Scholar : PubMed/NCBI

16 

D'Argenio V, Esposito MV, Telese A, Precone V, Starnone F, Nunziato M, Cantiello P, Iorio M, Evangelista E, D'Aiuto M, et al: The molecular analysis of BRCA1 and BRCA2: Next-generation sequencing supersedes conventional approaches. Clin Chim Acta. 446:221–225. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Børsting C and Morling N: Next generation sequencing and its applications in forensic genetics. Forensic Sci Int Genet. 18:78–89. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Debeljak M, Freed DN, Welch AJ, Haley L, Beierl K, Iglehart BS, Pallavajjala A, Gocke CD, Leffell MS, Lin MT, et al: Haplotype counting by next-generation sequencing for ultrasensitive human DNA detection. J Mol Diagn. 16:495–503. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Kim J, Hwang IS, Kim HS, Joo DJ, Hong KR and Choi JR: Bone marrow chimerism detection using next generation sequencing based on single nucleotide polymorphisms following liver transplantation: Comparison with short tandem repeat-PCR. Ann Lab Med. 36:82–84. 2016. View Article : Google Scholar :

20 

Chen DP, Tsai SH, Tseng CP, Wu TL, Chang PY and Sun CF: Bone marrow transplant relapse with loss of an allele. Clin Chim Acta. 387:161–164. 2008. View Article : Google Scholar

21 

Bragg LM, Stone G, Butler MK, Hugenholtz P and Tyson GW: Shining a light on dark sequencing: Characterizing errors in Ion Torrent PGM data. PLoS Comput Biol. 9:e10030312013. View Article : Google Scholar

Related Articles

Journal Cover

October-2016
Volume 14 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Aloisio M, Licastro D, Caenazzo L, Torboli V, D'Eustacchio A, Severini GM and Athanasakis E: A technical application of quantitative next generation sequencing for chimerism evaluation. Mol Med Rep 14: 2967-2974, 2016
APA
Aloisio, M., Licastro, D., Caenazzo, L., Torboli, V., D'Eustacchio, A., Severini, G.M., & Athanasakis, E. (2016). A technical application of quantitative next generation sequencing for chimerism evaluation. Molecular Medicine Reports, 14, 2967-2974. https://doi.org/10.3892/mmr.2016.5593
MLA
Aloisio, M., Licastro, D., Caenazzo, L., Torboli, V., D'Eustacchio, A., Severini, G. M., Athanasakis, E."A technical application of quantitative next generation sequencing for chimerism evaluation". Molecular Medicine Reports 14.4 (2016): 2967-2974.
Chicago
Aloisio, M., Licastro, D., Caenazzo, L., Torboli, V., D'Eustacchio, A., Severini, G. M., Athanasakis, E."A technical application of quantitative next generation sequencing for chimerism evaluation". Molecular Medicine Reports 14, no. 4 (2016): 2967-2974. https://doi.org/10.3892/mmr.2016.5593