You are on page 1of 10

PNAS PLUS

Second generation noninvasive fetal genome analysis


reveals de novo mutations, single-base parental
inheritance, and preferred DNA ends

SEE COMMENTARY
K. C. Allen Chana,b,1, Peiyong Jianga,b,1, Kun Suna,b,1, Yvonne K. Y. Chengc, Yu K. Tonga,b, Suk Hang Chenga,b,
Ada I. C. Wonga,b, Irena Hudecovaa,b, Tak Y. Leungc, Rossa W. K. Chiua,b,2, and Yuk Ming Dennis Loa,b,2
a
Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong; bDepartment of Chemical Pathology,
The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong; and cDepartment of Obstetrics and Gynaecology, The
Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong

Contributed by Yuk Ming Dennis Lo, October 4, 2016 (sent for review August 24, 2016; reviewed by Mary E. Norton and Erik A. Sistermans)

Plasma DNA obtained from a pregnant woman was sequenced to a the PPV to 0.438%, although the sensitivity was reduced to 38.6%.
depth of 270× haploid genome coverage. Comparing the maternal These data, thus, indicate the enormous challenge of detecting fetal
plasma DNA sequencing data with the parental genomic DNA data de novo mutations on a genome-wide scale using NIPT. In par-
and using a series of bioinformatics filters, fetal de novo mutations ticular, dramatic improvement in the PPV would be needed for
were detected at a sensitivity of 85% and a positive predictive value such an approach to be clinically practical.
of 74%. These results represent a 169-fold improvement in the pos- A second area that needs improvement concerns the detection
itive predictive value over previous attempts. Improvements in the of sequences that the fetus has inherited from its mother. Previous
interpretation of the sequence information of every base position efforts in elucidating the maternal inheritance of the fetus on a
in the genome allowed us to interrogate the maternal inheritance genome-wide scale have generally used a haplotype-based strat-
of the fetus for 618,271 of 656,676 (94.2%) heterozygous SNPs within egy, which has been referred to as the relative haplotype dosage
the maternal genome. The fetal genotype at each of these sites was
(RHDO) approach (8–10). Hence, for a pregnant woman who has
deduced individually, unlike previously, where the inheritance was
two haplotypes in a particular chromosomal region, she would
determined for a collection of sites within a haplotype. These results
represent a 90-fold enhancement in the resolution in determining the
pass one of these onto her fetus. Because her plasma contains a
fetus’s maternal inheritance. Selected genomic locations were more
likely to be found at the ends of plasma DNA molecules. We found Significance
that a subset of such preferred ends exhibited selectivity for fetal- or
maternal-derived DNA in maternal plasma. The ratio of the number of We explored the limit of noninvasive prenatal testing by per-
maternal plasma DNA molecules with fetal preferred ends to those forming genome-wide sequencing of maternal plasma DNA at
with maternal preferred ends showed a correlation with the fetal DNA 195× and 270× haploid genome coverages. Combined with the
fraction. Finally, this second generation approach for noninvasive fetal use of a series of bioinformatics filters, fetal de novo mutations
whole-genome analysis was validated in a pregnancy diagnosed with could be detected with a positive predictive value that was two
cardiofaciocutaneous syndrome with maternal plasma DNA sequenced orders of magnitude higher than previously reported. A de novo

MEDICAL SCIENCES
to 195× coverage. The causative de novo BRAF mutation was success- BRAF mutation was noninvasively detected in a case with car-
fully detected through the maternal plasma DNA analysis. diofaciocutaneous syndrome. The maternal inheritance of the
fetus could be ascertained on a genome-wide level without the
|
noninvasive prenatal testing massively parallel sequencing | use of maternal haplotypes, hence greatly increasing the reso-
DNA fragmentation patterns lution of such analysis. Finally, we showed that certain genomic
locations were overrepresented at the ends of plasma DNA
fragments with fetal or maternal selectivity.
T he discovery of cell-free fetal DNA in maternal plasma has
enabled the development of noninvasive prenatal testing
(NIPT) (1). Over the last few years, NIPT has been implemented
Author contributions: K.C.A.C., R.W.K.C., and Y.M.D.L. designed research; K.C.A.C., P.J., K.S.,
Y.K.Y.C., Y.K.T., S.H.C., A.I.C.W., I.H., and T.Y.L. performed research; K.C.A.C., P.J., K.S.,
globally for the noninvasive prenatal investigation of fetal chro- R.W.K.C., and Y.M.D.L. analyzed data; Y.K.Y.C. and T.Y.L. recruited subjects and ana-
mosomal aneuploidies (2–5). With higher depth of sequencing and lyzed clinical data; and K.C.A.C., P.J., R.W.K.C., and Y.M.D.L. wrote the paper.
improved bioinformatics analyses, NIPT has now been extended to Reviewers: M.E.N., University of California, San Francisco; and E.A.S., VU University
the detection of a variety of subchromosomal aberrations (6, 7). Medical Center.
Further expanding the applications of NIPT, we showed in 2010 Conflict of interest statement: R.W.K.C. and Y.M.D.L. received research support from
that it was possible to deduce the fetal genome by deep sequencing Sequenom, Inc. R.W.K.C. and Y.M.D.L. were consultants to Sequenom, Inc. K.C.A.C., R.W.K.C.,
and Y.M.D.L. hold equities in Sequenom, Inc. K.C.A.C., R.W.K.C., and Y.M.D.L. are founders
of maternal plasma (8). Work by Fan et al. (9) and Kitzman et al. of Xcelom and Cirina. K.C.A.C., P.J., and R.W.K.C. are consultants to Xcelom. P.J. is a
(10) confirmed these results. In these previous efforts, the depths consultant to Cirina. K.C.A.C., P.J., R.W.K.C., and Y.M.D.L. have filed patent applications
of maternal plasma DNA sequencing ranged from 52.7× to 78× (PCT/CN2016/073753 and PCT/CN2016/091531) based on the data generated from this
haploid human genome coverages (8–10). work, which have been licensed to Cirina.

There are a number of limitations in these previous studies. For Freely available online through the PNAS open access option.

example, Kitzman et al. (10) explored the possibility of detecting Data deposition: The sequence data for the subjects studied in this work who had con-
sented to data archiving have been deposited in the European Genome-Phenome Archive
fetal de novo mutations on a genome-wide level from the maternal (EGA), https://www.ebi.ac.uk/ega/, hosted by the European Bioinformatics Institute (EBI;
plasma DNA sequencing data. In one variation of bioinformatics accession no. EGAS00001001882).
analysis, they found 2.5 × 107 candidate fetal de novo mutation See Commentary on page 14173.
sites in the plasma DNA sequencing data. Only 39 of these were 1
K.C.A.C., P.J., and K.S. contributed equally to this work.
true fetal de novo mutations. Because the studied fetus had a total 2
To whom correspondence may be addressed. Email: rossachiu@cuhk.edu.hk or loym@
of 44 de novo mutations, the positive predictive value (PPV) cuhk.edu.hk.
was 0.000156%, and the sensitivity was 88.6%. With additional
Downloaded by guest on July 8, 2021

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.


refinement in bioinformatics analysis, Kitzman et al. (10) improved 1073/pnas.1615800113/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1615800113 PNAS | Published online October 31, 2016 | E8159–E8168


mixture of her own DNA and that from the fetus, there would be a bioinformatics analysis that takes into account the sequence,
slight overrepresentation of the haplotype shared by both the concentration, and size of the circulating DNA fragments would
pregnant mother and her fetus. This haplotype-based approach allow one to significantly improve the detection of fetal de novo
has imposed two limitations to NIPT. First, it requires the eluci- mutations, interrogate fetal inheritance at single-base resolution,
dation of the maternal haplotype using a direct haplotyping ap- and study the end characteristics of plasma DNA on a molecule by
proach (11–13), via pedigree analysis (8, 14), or via founder molecule manner. We further illustrated how such detailed level of
haplotype analysis in selected populations (15). Second, this fetal genomic information would be of clinical utility.
haplotype-based approach has limited the resolution in which the
maternal inheritance of the fetus can be determined. In this Results
regard, the mean length of the haplotype blocks that had been Clinical Samples and DNA Sequencing. Pregnant women were
used in previous efforts ranged from 300 kb to over 1 Mb (8–10). recruited from the Department of Obstetrics and Gynecology,
Recently, there is a lot of interest in the fragmentation patterns Prince of Wales Hospital with informed consent. Hypotheses were
of plasma DNA (16–18). Studies showed that plasma DNA frag- first explored and methodologies were developed based on the
mentation sites are located in clusters across the genome that bore analysis of a plasma sample from a pregnant woman at 38 wk of
relationships with positions of nucleosome arrays and open chro- gestation. These “second generation” fetal genome methodologies
matin domains. Based on these data, researchers have concluded
were then applied to the analysis of plasma collected from a sec-
that the fragmentation process of plasma DNA is nonrandom. One
ond trimester pregnancy affected by cardiofaciocutaneous syn-
interpretation of such observations is that plasma DNA fragments
drome. Plasma samples from 26 first trimester pregnant women
are cleaved at the accessible parts of the genome. We have gone
further in this study to explore whether the actual ending sites of were analyzed to further investigate the phenomenon of preferred
plasma DNA were nonrandom down to a single-base level. In ending sites among plasma DNA molecules.
other words, are plasma DNA molecules repeatedly cut at a se-
Key Maternal Plasma DNA Sequencing Metrics. Maternal plasma
lected set of genome coordinates? Previous studies could not ad-
DNA of the third trimester case was sequenced using the Illumina
dress this question, because the sequencing depths of individual
samples were not high enough, and PCR duplicates were discarded TruSeq PCR-free library preparation protocol to a depth of 270×
before additional data analysis. We hypothesized that some of such coverage of a haploid human genome. The maternal blood cells,
“duplicates” were not created by PCR. Instead, they were plasma paternal blood cells, and umbilical cord blood cells were se-
DNA molecules that had originated from different cells and were quenced to 40×, 45×, and 50× haploid human genome coverages,
cleaved at the same genome coordinates. To investigate this hy- respectively, using the same sequencing protocol. We analyzed
pothesis, we processed some of the studied plasma samples using a SNPs where both parents were homozygous but for different al-
PCR-free library preparation protocol. leles and identified 239,816 of such SNPs. For these SNPs, the
In this study, we aimed to develop a second generation approach fetus would be an obligatory heterozygote for the paternal and
to noninvasively decipher the fetal genome from maternal plasma maternal alleles. Among these SNPs, the paternal alleles were
using sequencing depth that had hitherto not been achieved in detectable in the maternal plasma of all of 239,816 SNPs. The
previous studies (Fig. 1). We hypothesized that the use of ultra- fetal DNA fraction (F) in maternal plasma was deduced as 31.3%
deep genome-wide plasma DNA sequencing and multiparametric from the aggregated numbers of paternally inherited fetal alleles

Noninvasive fetal whole genome analysis


1st generaona 2nd generaon
Sequencing Depth 53× − 78× 195× − 270×
De Novo Mutaon
Sensivity 38.6% 81% − 85%
PPV 0.438% 62% − 74%

Maternal Inheritance
Principle Haplotype-based by RHDO* SNP-based by GRAD*
Resoluon 300k − over 1M Single nucleode
1. Nucleosomal paern Plasma DNA preferred end sites
2. Size difference between maternal- Genomic coordinates
and fetal-derived fragments
Fetal-
derived
DNA
166 fragments
Frequency (%)

Plasma DNA Maternal-


Fragmentaon 143 derived
DNA
Paern fragments
Maternal preferred
end site
Size (bp) Fetal preferred end site

Fig. 1. Summary of the key differences between the first and second generation approaches for noninvasive fetal whole-genome analysis. aThe per-
Downloaded by guest on July 8, 2021

formance characteristics quoted for first generation fetal whole-genome analysis were from refs. 8–10. *RHDO represents RHDO analysis, and GRAD
represents GRAD analysis.

E8160 | www.pnas.org/cgi/doi/10.1073/pnas.1615800113 Chan et al.


PNAS PLUS
(p) and the maternal alleles (m) at these SNPs using the formula plasma was more likely to have originated from the fetus or se-
F = 2p/(p + m). quencing errors. In this algorithm, a variant would need to be
We next measured the difference in the sizes of fetally and observed in a threshold number of sequenced reads so as to qualify
maternally derived DNA in the maternal plasma at individual as a candidate de novo mutation (Table S1). This threshold was
SNPs. We compared the difference in size distribution of fragments determined from a binomial probability function based on the total

SEE COMMENTARY
carrying the fetal-specific allele and the allele shared by the fetus number of times that a particular nucleotide was actually se-
and the mother at a particular SNP (Fig. S1). Such size information quenced and the sequencing error rate, so that the theoretical
would be useful for determining the likelihood of any variant allele probability of a putative de novo mutation being a sequencing
detected in maternal plasma being a fetal de novo mutation. The error would be less than 1 in 3 × 109. Using this dynamic cutoff
median difference between fragments carrying the fetal-specific algorithm, 60,111 candidate de novo mutations were identified,
alleles and the shared alleles was 22 bp (Fig. S2A). At 90% of these and these candidate de novo mutations included 54 of 56 (i.e.,
SNP loci, the difference was larger than 10 bp (Fig. S2A). 96%) de novo mutations present in the cord blood (Fig. 2).
Then, we realigned the sequenced reads covering the 60,111
Detection of Fetal de Novo Mutations from Maternal Plasma. Fifty-six candidate de novo mutation sites to the reference human genome
single-nucleotide variants were found to be present in the umbilical using the BOWTIE2 software (19). The initial alignment software,
cord blood cells and absent in the parents. These 56 single- Short Oligonucleotide Alignment Program 2 (SOAP2) (20), and
nucleotide variants were thus deemed de novo mutations possessed the realignment software, BOWTIE2, were based on two different
by the fetus. Deep genome-wide sequencing is needed for the matching algorithms, namely the Burrows–Wheeler transform al-
screening of fetal de novo mutations from maternal plasma. At this gorithm and Smith–Waterman-like algorithm, respectively (19, 20).
level of sequencing, a vast number of sequencing errors would be A variant would be removed from being considered as a candidate
generated and hinder the sensitive and specific detection of true de novo mutation if the two alignment algorithms do not map the
fetal de novo mutations. To tackle the challenge, we designed a variant to the same genomic location. This realignment procedure
bioinformatics protocol that used a series of steps to filter out the significantly reduced the number of false-positive results caused by
false-positive mutations (Fig. 2). The bioinformatics protocol made alignment errors. After this realignment process, the number of
use of steps that identified high-quality base sequences in combi- candidate de novo mutations was reduced to 148 and contained
nation with steps that took into account the known biological 52 (93%) of the de novo mutations detected in the cord blood
characteristics of cell-free fetal DNA. (Fig. 2). This result is equivalent to a PPV of 35%.
First, we identified genomic locations where the maternal To further reduce the number of false positives, we applied a
plasma DNA sequencing showed at least one sequence variant, filter based on the fractional concentration of the variants. Because
whereas the parental DNA analysis showed that both parents the fetal DNA fraction in the sample was 31.3%, 95% of the var-
shared the same sequence. Second, we applied a dynamic cutoff iants would be expected to be present at a level of ≥10% of the
algorithm to determine if the variant detected in the maternal sequenced reads covering a particular nucleotide based on the
Poisson distribution. Hence, we filtered out the candidate muta-
tions that were present at a fraction of <10%. Eighty-five candidate
de novo mutations were present at ≥10% of the sequenced reads
Dynamic cutoff covering the nucleotide and included 51 (91%) of the mutations
Variant frequency higher than sequencing error rate detected in the cord blood (Fig. 2). This result represents a PPV
60,111 of 60%.
Next, we included a filter based on the size of the plasma DNA

MEDICAL SCIENCES
Detection rate: 54/56 (96%); PPV: 0.089%
molecules carrying the candidate de novo mutations. As illustrated
in Fig. S2A, the difference between the mean fragment size for
fragments carrying the fetal-specific alleles and the shared alleles
was larger than 10 bp for 90% of loci at which the fetus was
Realignment heterozygous and the mother was homozygous. Thus, we applied a
Realign all reads covering putative mutation sites size filter to the candidate de novo mutations, requiring the mean
148 size of the plasma DNA fragments carrying the mutants to be at
Detection rate: 52/56(93%); PPV: 35% least 10 bp shorter than that of the fragments carrying the parental
alleles. Sixty-five candidates passed this size-filtering criterion that
included 48 (85%) of the mutations detected in the cord blood
(Fig. 2). This result represents a PPV of 74%.
Fractional concentration
Variant frequency compatible with fetal DNA fraction Paternal Inheritance Analysis. For paternal inheritance analysis, we
85 focused on SNPs that were heterozygous in the father and homo-
Detection rate: 51/56 (91%); PPV: 60% zygous in the mother. In principle, the presence or absence of the
paternal-specific allele in the maternal plasma would indicate the
inheritance of paternal-specific allele or the shared parental allele
by the fetus, respectively. To enhance the accuracy of deducing the
paternal inheritance, we used a binomial mixture model, which
Plasma DNA fragment size took into account the fetal DNA concentration, the sequencing
Mean size of fragments carrying the variant 10 bp shorter than error rate, and the number of times that the paternal-specific allele
the mean size of fragments carrying the parental alleles was observed in the maternal plasma (details are in Materials and
65 Methods) (21). For 667,586 SNPs for which the mother was ho-
Detection rate: 48/56 (85%); PPV: 74% mozygous and the father was heterozygous, the overall accuracy
was 99.99% for the deduction of the paternal inheritance.
Fig. 2. Detection of the fetal de novo mutations through the analysis of the
sequencing data of maternal plasma DNA. The number in bold in each step Genome-Wide Relative Allele Dosage Analysis. Previous efforts in
prenatally elucidating the maternal inheritance of the fetus on a
Downloaded by guest on July 8, 2021

represents the number of candidate de novo mutations identified after the


particular process. genome-wide level had all been based on the RHDO approach

Chan et al. PNAS | Published online October 31, 2016 | E8161


(8–10). Enabled by the high-quality genome-wide base information, by the mother. These peaks represent the hotspots for the end
we showed that the maternal inheritance of these heterozygous positions of fetal- and maternal-derived DNA in maternal plasma,
SNPs could be resolved without the use of the haplotype in- respectively. In contrast, the fragmentation pattern of the soni-
formation of the mother. The dosage of the two maternal alleles in cated DNA seems to be random without such clusters, and the
maternal plasma would be compared to determine, statistically, fragment end probability is similar across the region (Fig. 3).
whether the two alleles are present at the same or different con- We further studied the coordinates that had an increased
centrations. For SNP loci at which one allele was present at signif- probability of being an ending position for plasma DNA fragments.
icantly higher concentration and at which the two alleles were We focused our search based on fragments covering the in-
present at statistically nondifferentiable concentrations, the fetus formative SNPs, so that the fragments carrying fetal-specific alleles
would be deduced to be homozygous and heterozygous, respectively. and alleles shared by the mother and the fetus could be evaluated
We denote this method as genome-wide relative allelic dosage separately. We determined if certain locations within the human
(GRAD) analysis. This method is a genome-wide and sequencing- genome had a significantly increased probability of being an ending
based version of our previously described relative mutation dosage position of plasma DNA fragments using a Poisson probability
method (22). There were a total of 656,676 heterozygous SNPs in function (Materials and Methods). A P value of <0.01 had been
the mother’s genome. Using GRAD analysis, the maternal in- chosen to indicate statistical significance. Statistically significant
heritance of the fetus was resolved with statistically significant results ending positions were determined for DNA fragments carrying the
in 618,271 SNPs (i.e., 94.2%). The allelic counts were insufficient to shared allele and the fetal-specific allele independently (Fig. 4A).
give statistically significant results in 38,405 SNPs (i.e., 5.8%). The We identified a total of 8,242 (Set A) and 23,857 (Set B) nu-
fetus was deduced to be homozygous at 335,988 (54%) SNPs and cleotide positions with a significantly increased chance of being an
heterozygous at 282,283 (46%) SNPs. Among 618,271 SNPs with end for plasma DNA fragments carrying fetal-specific alleles and
statistically significant results, the deduction of maternal inheritance shared alleles, respectively, covering 10,233 SNPs; 8,909 of the
was correct for 610,084 (98.7%) SNPs compared with the genotypes nucleotide positions were observed to be overrepresented among
from the analysis of the cord blood cells. the plasma DNA molecules with the fetal-specific allele (Set A) as
The resolution of GRAD analysis would be affected by the se- well as molecules with the shared allele (Set B). We called this
quencing depth and the fetal DNA fraction in the maternal plasma overlapping set of ends Set C (Fig. 5A). There were 48,707, 53,901,
sample. We performed computer simulation analyses to determine and 65,205 plasma DNA fragments carrying fetal-specific alleles
the number of SNPs required for achieving one classification of ending on Set A, Set B, and Set C positions, respectively. In other
maternal inheritance with an overall accuracy of over 95% (Fig. words, multiple fetal-specific DNA molecules showed identical
S3). With a sequencing depth of 250×, we would be able to achieve ending positions. A median of 6 (range = 6–41) plasma DNA
close to single-nucleotide resolution for the deduction of maternal fragments carrying fetal-specific alleles terminated at a Set A
inheritance when the fetal DNA fraction was above 20%. When ending position. Based on a sequencing depth of 270×, fetal DNA
the fetal DNA fraction was 10% and a sequencing depth was at fraction of 31.3%, and the size distribution of fetal DNA frag-
300×, maternal inheritance could be deduced in 1 of every 5 SNPs ments, it was expected that only 0.29 fragment would end at each
with an accuracy of over 95%, but the resolution of maternal in- site if the fragmentation of plasma DNA had been random. Thus,
heritance would drop to 1 classification per 30 SNPs with a se- the probability of ending at these sites was 20 times higher than
quencing depth of 150×. expected. There were 54,541, 376,343, and 182,791 plasma DNA
fragments carrying shared alleles ending at Set A, Set B, and Set C
Preferred End Sequences in Maternal Plasma DNA. Facilitated by the positions, respectively. The genomic coordinates of Set A, Set B,
high-sequencing depth of a non-PCR–amplified library of the and Set C positions are shown in Dataset S1.
maternal plasma DNA sample, we investigated if certain base Using the same principle, we analyzed the ending positions for
positions in the genome would be preferentially represented at the maternal-specific plasma DNA fragments. SNPs that were het-
ends of plasma DNA fragments. In this regard, we showed that erozygous in the mother (genotype AB) and homozygous in the
plasma DNA fragments carrying the most prevalent 0.5% of fetus (genotype AA) were noted, and plasma DNA molecules that
plasma DNA ends represented 3.5% of all DNA fragments. Across carried any one of the maternal-specific alleles (B allele) were
the genome, 25% of the fragments had at least one identical deemed to be maternally derived. Among the maternal-specific
fragment that shared the same ending sites at both ends. If the plasma DNA molecules, 7,527 (Set X) and 18,829 (Set Y) nucle-
cleavage or breakage of plasma DNA was completely random, only otide positions showed increased occurrence as an ending position
1.45% of the DNA molecules would be expected to have at least for plasma DNA fragments carrying maternal-specific alleles and
one counterpart sharing common ends (details are in Materials and shared alleles, respectively, covering 9,489 SNPs (Fig. 4B); 10,534
Methods). Had the sequencing been performed using a PCR- positions were observed to be overrepresented among the plasma
amplified library, then 14% of the fragments would have been wrongly DNA molecules with the maternal-specific allele (Set X) as well as
assumed to be PCR duplicates and would have been filtered off. molecules with the shared allele (Set Y). We called this over-
We further investigated if there would be differences in the lapping set of ends Set Z (Fig. 5B). There were 69,136, 82,413, and
preferred ending sites for maternal and fetal DNA in the same cell- 121,607 plasma DNA fragments carrying maternal-specific alleles
free DNA sample. To show this phenomenon, informative SNP ending at Set X, Set Y, and Set Z positions, respectively. A median
loci where the mother was homozygous (genotype denoted as AA) of 10 (range = 9–54) plasma DNA fragments carrying maternal-
and the fetus was heterozygous (genotype denoted as AB) were specific alleles terminated at a Set X base position. If the frag-
identified. In this illustrative example, the B allele would be fetal- mentation of plasma DNA had been random, it was expected that
specific, and the A allele would be shared by the mother and the only 0.56 fragment would terminate at each site. Thus, the prob-
fetus. The fetal-specific and shared reads covering one such in- ability for ending at these sites was 18 times higher than expected.
formative SNP are shown in Fig. 3. For comparison, the se- There were 46,554, 245,037, and 181,709 plasma DNA fragments
quencing results of a DNA sample obtained from blood cells and carrying shared alleles ending on Set X, Set Y, and Set Z positions,
artificially fragmented using sonication are also shown. Clusters of respectively. The genomic coordinates of the Set X, Set Y, and Set
preferred ending positions could be observed among the plasma Z positions of this case are shown in Dataset S1.
DNA data. For the plot of the probability of a genomic location
being an end of DNA fragments (also referred to as the fragment Using Preferred End Positions to Deduce Fetal DNA Fractions in Maternal
end probability), three peaks were observed for each of two groups Plasma in First Trimester Pregnancies. After having identified sets of
Downloaded by guest on July 8, 2021

of fragments carrying the fetal-specific allele and the allele shared maternal plasma DNA preferred end positions from one third

E8162 | www.pnas.org/cgi/doi/10.1073/pnas.1615800113 Chan et al.


PNAS PLUS
Maternal plasma DNA Sonicated maternal blood cell DNA
Informative SNP Informative SNP

SEE COMMENTARY
Shared allele
Fetal-specific allele

8
4

Fraction of
fragments ending
8

on the nucleotide

8
position (%)
4

4
-300 -200 -100 0 100 200 300 -300 -200 -100 0 100 200 300
Relative genomic coordinates (bases) Relative genomic coordinates (bases)

Fig. 3. Left shows an illustrative example of the nonrandom fragmentation patterns of plasma DNA carrying a fetal-specific allele and an allele shared by the
mother and the fetus. In Upper, each horizontal line represents one sequenced DNA fragment. The ends of the DNA fragments represent the ending position of
the sequenced read. The fragments are sorted according to the coordinate of the left outermost nucleotide (genomic coordinate with the lower numerical value).
In Lower, the percentage of fragments ending on a particular position is shown. Upper Right shows the span of all of the sequenced DNA fragments aligned to
the same SNP obtained from the sonicated blood cell DNA of the same pregnant woman. Lower Right shows the percentage of fragments ending on a particular
position. The x axes show the relative genomic coordinates. The SNP of interest is located in the middle (marked by a dotted line).

trimester case, we explored if such ends would be detectable in fetus. Sequencing libraries were prepared from these maternal
samples of other pregnancies and whether the relative abundance plasma samples using the KAPA Library Preparation Kits (Kapa
of plasma DNA ending at these sets of nucleotide positions would Biosystems), which included a PCR amplification step to enrich the
reflect the fetal DNA fraction. We sequenced the plasma DNA of adaptor-ligated molecules. The median number of mapped reads
26 first trimester (10–13 wk) pregnancies, each involving a male per sample was 16 million (range = 12–22 million). The proportion

A Informative SNP that mother was homozygous B Informative SNP that mother was heterozygous
and fetus was heterozygous and fetus was homozygous

MEDICAL SCIENCES
Sequencing reads carrying Sequencing reads carrying
8

shared allele
8

Significant ending shared allele


Proportion of plasma DNA ending at

Proportion of plasma DNA ending at

Significant ending
position specific for
Significant ending position specific for
fragments carrying Significant ending
position common for fragments carrying
a genomic coordinate (%)

shared alleles
a genomic coordinate (%)

shared alleles position common for


fragments carrying
fragments carrying
shared and fetal-specific
4

shared and maternal-


alleles
specific alleles
0

0
4

Significant ending Significant ending


position specific for position specific for
fragments carrying fragments carrying
fetal-specific alleles maternal-specific alleles
Sequencing reads carrying Sequencing reads carrying
8

fetal-specific allele maternal-specific allele

18878200 18878400 18878600 18878800


17053400 17053600 17053800 17054000 17054200
Genomic coordinates (chr22) (bases) Genomic coordinates (chr1) (bases)
Fig. 4. Plot of probability of a genomic coordinate being an ending position of maternal plasma DNA fragments across a region with an informative SNP at
which (A) the mother was homozygous and the fetus was heterozygous and (B) the mother was heterozygous and the fetus was homozygous. (A) Results for
nucleotide positions with a significantly increased probability of being an end of plasma DNA fragments carrying a shared allele and a fetal-specific allele are
shown in red and blue, respectively. (B) Results for nucleotide positions with a significantly increased probability of being an end of plasma DNA fragments
Downloaded by guest on July 8, 2021

carrying a shared allele and a maternal-specific allele are shown in red and blue, respectively. The x axes of the graphs show the relative genomic coordinates.
The position of the SNP of interest is marked by a dotted line.

Chan et al. PNAS | Published online October 31, 2016 | E8163


A Ending positions for plasma DNA fragments To investigate the specificity of these positions, we analyzed the
across a SNP that was homozygous in the mother size distribution of DNA fragments ending on the Set A or Set X
and heterozygous in the fetus positions when the genomic coordinates of these positions were
shifted by one to five nucleotides. To quantify the size difference,
Preferred ending positions Preferred ending positions cumulative frequencies for fragments ending at the two sets of
for fragments carrying for fragments carrying
shared alleles
positions were plotted against size. The difference in the two cu-
fetal-specific alleles
mulative frequency curves (denoted as ΔS) would reflect the
magnitude of the size difference (Figs. 7B and 8B). For the third
trimester sample, the difference in size distribution diminished
8,242 8,909 23,857 when the number of shifted nucleotides was increased and no
longer observable when the coordinates were shifted by four nu-
Set A Set C Set B cleotides (Fig. 7 C and D). For the pooled sequenced reads from 26
first trimester cases, a reduction in the difference in size distribu-
tion with nucleotide shift was similarly observed (Fig. 8 C and D).

Fetal Genome Analysis of a Second Trimester Pregnancy Affected by


Ending positions for plasma DNA fragments
B across a SNP that was heterozygous in the mother Cardiofaciocutaneous Syndrome. To validate the above-mentioned
and homozygous in the fetus methodologies, we sequenced a maternal plasma sample collected
at 18 wk of gestation to 195× coverage. This case presented with
Preferred ending positions Preferred ending positions increased nuchal translucency in the first trimester. A chorionic
for fragments carrying for fragments carrying villus sample was obtained at 11 wk of gestation, which showed a
maternal-specific alleles shared alleles normal karyotype. At 14 wk of gestation, a cystic hygroma and
mild club foot deformity were detected on ultrasound scan, and
hence, microarray analysis was performed on the chorionic villus
sample to detect gene mutations that were associated with
7,527 10,534 18,829 Noonan syndrome and related conditions. The microarray analysis
Set X Set Z Set Y detected a mutation (c770A > G) on the BRAF (B-Raf proto-
oncogene, serine/threonine kinase) gene that resulted in car-
diofaciocutaneous syndrome. Because the mutation was absent in
the mother’s and father’s genomes, it was deemed a de novo
mutation. The fetus was aborted, and placental tissues were col-
Fig. 5. Analysis of ending positions for plasma DNA fragments across
(A) SNPs that were homozygous in the mother and heterozygous in the fetus
lected after the procedure.
and (B) SNPs that were homozygous in the fetus and heterozygous in the DNA from the maternal buffy coat, paternal buffy coat, and
mother. (A) Set A included preferred ending positions for fragments carrying placental tissues collected after termination was sequenced to 40×,
fetal-specific alleles. Set B included preferred ending positions for fragments 60×, and 60× human genome coverages, respectively. The fetal
carrying shared alleles. Set C included preferred ending positions for both DNA fraction in the maternal plasma was 24%. A difference of at
types of plasma DNA fragments. (B) Set X included preferred ending posi- least 8 bp was observed between the size of fragments carrying
tions for fragments carrying maternal-specific alleles. Set Y included pre- fetal-specific alleles and alleles shared by the fetus and the mother
ferred ending positions for fragments carrying shared alleles. Set Z included at over 90% of informative SNPs (Fig. S2B). Because the size
preferred ending positions for both types of plasma DNA fragments.

of sequenced reads aligning to chromosome Y was used to calcu- 1.1


late the actual fetal DNA fraction in each plasma sample. A pos-
itive correlation could be observed between the relative abundance Pearson’s r = 0.66
[denoted as the fetal/maternal (F/M) ratio] of plasma DNA with P-value < 0.001
recurrent fetal (Set A) and maternal (Set X) ends and the fetal
1.0
DNA fraction (R = 0.66, P < 0.001, Pearson correlation) (Fig. 6). A
Ratio (F/M)

median of 248 Set A ends was observed among 26 samples. A


median of 286 Set X ends was observed among those samples.

Size Distribution of Plasma DNA Fragments Terminating on the Fetal- 0.9


Specific End Positions. Because fetal DNA in maternal plasma is
shorter than the maternal-derived DNA in maternal plasma, we
compared the size distributions of plasma DNA ending on the Set
A and Set X positions. For the deeply sequenced third trimester 0.8
case, the size distribution for fragments ending at Set A positions
was shorter than those ending at Set X positions (Fig. 7 A and B).
Then, we analyzed the pooled sequenced reads from 26 first tri-
mester plasma samples used for fetal DNA fraction analysis. A 0.7
shorter size distribution was observed for fragments ending 0 5 10 15 20 25
at fetal-preferred Set A positions compared with those ending at
maternal-preferred Set X positions (Fig. 8 A and B). These results
were consistent with the fact that fetal DNA was shorter than Fetal DNA fraction (%)
maternal-derived DNA and further support the hypothesis that Fig. 6. Correlation between the relative abundance (ratio F/M) of plasma
the end positions derived from one pregnant woman can poten-
Downloaded by guest on July 8, 2021

DNA molecules with preferred fetal (Set A) and maternal (Set X) ends, and
tially be generalized to other pregnant cases. fetal DNA fraction.

E8164 | www.pnas.org/cgi/doi/10.1073/pnas.1615800113 Chan et al.


PNAS PLUS
A B maternal-preferred (Set X) ends and the fetal DNA fraction (R =
0.66, P < 0.001, Pearson correlation) (Fig. S6).

Cumulave frequency (%)


Fetal- Fetal-
preferred preferred
Discussion
Frequency (%)

Maternal- Maternal-
preferred preferred In this work, we performed ultradeep genome-wide sequencing of

SEE COMMENTARY
the plasma DNA obtained from two pregnant women to 195× and
270× haploid genome coverages. We believe that these are the
deepest genome-wide sequencings yet reported for plasma DNA
ΔS obtained from any one pregnancy. Previous efforts in the genome-
wide sequencing of plasma DNA obtained from pregnant women
had attempted sequencing depths from 52.7× to 78× haploid ge-
nome coverages (8–10). The depth of sequencing achieved in our
Size (bp) Size (bp) analyzed case has allowed us to push forward the limit and expand
C D the applications of NIPT.
First, we investigated the possibility of searching for fetal de
novo mutations on a genome-wide scale using the maternal
plasma DNA sequencing data. It has been reported that there are
∆S (%)
∆S (%)

∼74 de novo point mutations per person and that the mutation
rate increases with paternal age (23). It is now increasingly rec-
ognized that de novo mutations play an important role in human
genetic diseases and that diseases associated with de novo muta-
tions are not rare collectively (23). Thus, it is clinically relevant to
be able to screen for de novo mutations prenatally. Efforts in the
noninvasive prenatal detection of fetal de novo mutations are
Size (bp) Size (bp) limited to diseases associated with structural abnormalities de-
tectable on ultrasound and where those diseases are known to be
Fig. 7. (A) Plasma DNA size distributions for fragments ending on fetal-pre-
associated with hotspot de novo mutations (24). For example,
ferred ending positions (Set A; blue) and fragments ending on maternal-pre-
ferred ending positions (Set X; red). A shorter size distribution was observed
for fragments ending on Set A positions compared with those ending on Set X
positions. (B) Cumulative plot for the size distributions for the two sets of
fragments. The difference in the cumulative frequencies of the two sets of A B

Cumulative frequency (%)


fragments (ΔS) against fragment size is shown. (C) A plot of ΔS against size Fetal- Fetal-
when the end coordinates are shifted by 0 to +5 bp with regard to the ge- preferred preferred
nomic coordinates of the preferred end. (D) A plot of ΔS against size when the
Frequency (%)

Maternal- Maternal-
end coordinates are shifted by 0 to −5 bp with regard to the genomic coor- preferred preferred
dinates of the preferred end.

MEDICAL SCIENCES
distribution of plasma DNA fragments carrying the alleles shared ΔS
by the fetus and the mother is dependent on the fractional fetal
DNA concentration in plasma, the size difference required for
filtering de novo mutation would need to be determined on a case
by case basis. In this case, at least 8-bp size difference between the Size (bp) Size (bp)
variant and the parental alleles was used as the criterion for C D
qualifying the variant as a candidate de novo mutation; 75 can-
didate de novo mutations were identified in the plasma, of which
47 were confirmed to be present in the placenta. Because a total of
ΔS (%)

58 de novo mutations were detected in the placenta, the plasma


ΔS (%)

analysis had a detection rate of 81% and a PPV of 62% (Fig. S4).
The de novo BRAF mutation was among 47 variants detected by
the plasma DNA analysis. Using GRAD analysis, the maternal
inheritance of the fetus was deduced in 528,008 (68% of 775,456
SNPs where the mother was heterozygous) SNPs with an accuracy
of 96.8%. Regarding preferred end sites, plasma DNA fragments
carrying the most prevalent 0.5% of plasma DNA ends repre-
Size (bp) Size (bp)
sented 2.4% of all DNA fragments. Strikingly, 59% of the most
prevalent 0.5% plasma DNA end sites overlapped with those of Fig. 8. (A) Plasma DNA size distributions in a pooled plasma DNA sample from
the third trimester case. Based on the analysis of informative 26 first trimester pregnant women for fragments ending on fetal-preferred
SNPs, 10,401 fetal-preferred (Set A) (Fig. S5A) and 6,562 ma- ending positions (Set A; blue) and fragments ending on maternal-preferred
ternal-preferred (Set X) (Fig. S5B) end sites were identified. The ending positions (Set X; red). A shorter size distribution was observed for
preferred end sites for fragments carrying alleles shared between fragments ending on Set A positions compared with those ending on Set X
the mother and the fetus were also identified (Set B, Set C, Set Y, positions. (B) Cumulative plot for the size distributions for the two sets of
fragments. The difference in the cumulative frequencies of the two sets of
and Set Z) (Fig. S5). The genomic coordinates of each set of
fragments (ΔS) against fragment size is shown. (C) A plot of ΔS against size
preferred positions of this case are shown in Dataset S2. In 26 first when the end coordinates are shifted by 0 to +5 bp with regard to the genomic
trimester maternal plasma samples, a positive correlation could be coordinates of the preferred end. (D) A plot of ΔS against size when the end
observed between the relative abundance (i.e., the F/M ratio) of coordinates are shifted by 0 to −5 bp with regard to the genomic coordinates of
Downloaded by guest on July 8, 2021

the plasma DNA molecules with fetal-preferred (Set A) and the preferred end.

Chan et al. PNAS | Published online October 31, 2016 | E8165


>90% of achondroplasia cases are caused by a de novo mutation to a relatively shallow depth. These observations indicate that the
in the FGFR3 (fibroblast growth factor receptor 3) gene with no ending sites that we have identified are indeed preferential plasma
prior familial history of skeletal dysplasia (25). A clinical suspicion DNA fragmentation sites that are detectable recurrently across
for achondroplasia could, therefore, be followed by maternal different samples at different gestational ages and with more than
plasma DNA analysis with the aim to specifically detect fetal one library preparation protocol.
FGFR3 mutations (26). Furthermore, we have found that there were subsets of such
Genome-wide scanning for fetal de novo mutations without a recurrent plasma DNA ends that were preferentially associated
priori information on potentially affected genes or diseases is a with fetal or maternal DNA (Figs. 3, 4, and 5). Using such fetal- or
huge technological challenge. Previous efforts in this area had maternal-preferred ending sites, one could estimate the fetal DNA
reported a PPV of 0.438% at a sensitivity of 38.6% (10). Hence, fraction in a particular sample (Fig. 6 and Fig. S6). This latter
using the depth of sequencing as a foundation, we used a series of correlation is particularly striking, because the fetal- and maternal-
bioinformatics filters taking into account the sequencing error rate, preferred ending sites were originally identified in the third and
minimizing the realignment errors, and considering the plasma second trimester cases. However, the abundance of such fetal-
fraction of a putative mutation as well as the size difference be- specific ending sites correlated with the fetal DNA fractions in the
tween fetal and maternal DNA in maternal plasma. Using such series of independent first trimester cases.
filters, we were able to detect fetal de novo mutations with a PPV For two cases in the study that were subjected to ultradeep
of 74% at a sensitivity of 85% for the third trimester case. For the sequencing (270× and 195× haploid genome coverages), it was
second trimester case, we achieved a PPV of 62% and a sensitivity desirable to use PCR-free library preparations, because the
of 81%. The PPVs, in particular, represent a significant step for- probability of encountering PCR duplicates increased with the
ward over previous efforts and are at a level that suggests that, with depth of sequencing. Furthermore, the use of PCR-free libraries
additional improvements, such an approach might eventually have was also the most convincing approach to answer the fundamental
clinical impact. It is noteworthy that the approach successfully question as to whether there were preferred ending sites for
detected the de novo BRAF mutation that caused the craniofa- plasma DNA, because one could positively identify them without
ciocutaneous syndrome in the second trimester case. For the the worry that one was seeing an artefactual result (i.e., PCR
analysis of cases with earlier gestational ages and lower fetal DNA duplicates).
fractions, higher sequencing depths would be required to achieve For using the recurrent ends to deduce the fetal DNA fractions
similar levels of performance. We should perhaps add a cautionary for the first trimester samples, we had used PCR-amplified se-
note that, although we have shown the technological feasibility of quencing libraries. However, the depth of sequencing was very
detecting fetal de novo mutations from maternal plasma, the in- shallow (median of 15 million reads per case; i.e., 0.8× haploid
terpretation of the clinical implications of each of these mutations genome coverage). Hence, one would not expect any of the pre-
is a great challenge that would require a well-supported clinical ferred end sites to appear more than once in a particular sample.
genetics infrastructure and continual improvements in our un- Consequently, the filtering step that one normally performed to
derstanding of the human genome. remove PCR duplicates, leaving one read for subsequent bio-
Second, the depth of maternal plasma DNA sequencing and informatic analysis, would not be expected to bias the results. Our
refined bioinformatics strategies used in this study allow one to demonstration of a good correlation between the F/M ratio (based
determine the maternal inheritance of the fetus on a genome-wide on preferred ends) and the fetal DNA fraction is a testimony to
level without resorting to maternal haplotype dosage analysis (8). our belief.
Using GRAD analysis, the maternal inheritance of the fetus could The plasma DNA fragments with the fetal-preferred ending
be resolved in 94.2% of 656,676 heterozygous SNPs in the ma- sites had a shorter size distribution than those with the maternal-
ternal genome of the third trimester pregnancy and 68% of preferred ending sites (Fig. 7). Most importantly, this size differ-
775,456 heterozygous SNPs in the genome of the second trimester ence was observed in not only the third trimester case in which the
pregnant woman. In our previous study on noninvasive fetal ge- recurrent ends were identified but also, 26 independent first tri-
nome analysis, the maternal inheritance was deduced using mester cases (Fig. 8). It is also interesting to note that, as we
RHDO analysis (8). The mean size of the haplotype blocks was shifted the end coordinates by one to five nucleotides on both
409 kb. For other studies involving the noninvasive fetal genome sides of the fetal- and maternal-preferred end sites, the size dif-
analysis, the mean size of haplotype blocks with maternal in- ference rapidly disappeared (Fig. 7 C and D). A similar finding
heritance determined ranged from 300 kb to over 1 Mb (9, 10, 27). was also observed from 26 first trimester cases (Fig. 8 C and D).
For our previous study (8), the maternal inheritance of 7,332 These data suggest that there is a high level of specificity in the
haplotype blocks was successfully determined with an accuracy of process of plasma DNA fragmentation to the extent where, in
99.1%. The present work, thus, represents an ∼90-fold (i.e., some parts of the genome, the cleavage or breakage occurs pref-
656,676/7,332 and 716,646/7,332 for the third and second trimester erentially at specific base locations.
cases, respectively) increase in resolution in elucidating the ma- There are active recent discussions on the nucleosomal origin of
ternal inheritance of the fetus. plasma DNA. One clue to such an origin came from work on the
Third, the deep sequencing of maternal plasma DNA samples high-resolution size profiling of maternal plasma DNA (8, 28).
prepared using non–PCR-amplified libraries has allowed us to Such work has revealed that the size profile of the total DNA in
investigate if there might be genomic locations that would be maternal plasma, in which a predominant proportion is from the
preferentially represented at the ends of plasma DNA fragments pregnant mother, contains a dominant population of plasma DNA
and whether such ends would exhibit differences depending on molecules with a size of 166 bp. Such data have also shown that,
their tissue of origin (i.e., from the placenta of the fetus or the when one focuses on the placentally derived fetal DNA in ma-
mother). Our data indicate that such recurrent ends are indeed ternal plasma, one would observe a predominant population with
present in plasma DNA. For example, our data from the third a size of 143 bp. The 166-bp molecules have been postulated to
trimester case revealed that plasma DNA fragments carrying the represent molecules containing the nucleosome core plus the
most prevalent 0.5% of plasma DNA ends represented 3.5% of all linker (8). The 143-bp molecules, however, have been postulated
DNA fragments and that 25% of the fragments had at least one to represent molecules containing the nucleosomal core without
additional fragment sharing identical ends at both ends. In- the linker. Straver et al. (17) created an artificial “nucleosome
terestingly, the preferred ending positions identified in the third track” by pooling maternal plasma DNA sequencing data from
trimester case were also detectable in 26 first trimester cases, even multiple cases. They found that the frequency of reads with
Downloaded by guest on July 8, 2021

when conventional PCR-amplified DNA libraries were sequenced starting sequences within regions 73 bp upstream and downstream

E8166 | www.pnas.org/cgi/doi/10.1073/pnas.1615800113 Chan et al.


PNAS PLUS
of the deduced nucleosome center showed a positive correlation DNA Library Construction. For the two cases subjected to deep sequencing, DNA
with the fetal DNA fraction. Outside of the context of pregnancy, libraries for the genomic DNA samples and the maternal plasma sample were
constructed with the TruSeq DNA PCR-Free Library Preparation Kit (Illumina)
Snyder et al. (16) also explored the nucleosomal footprint of
according to the manufacturer’s protocol, except that one-fifth of the indexed
plasma DNA. They developed a metric that they called the win- adapter was used for plasma DNA library construction. For the construction of
dowed protection score, which has been defined as the number of plasma DNA libraries, DNA extracted from 8 mL plasma was used. For the

SEE COMMENTARY
molecules spanning a particular genomic window minus those genomic DNA samples, including the mother’s buffy coat DNA, the father’s
molecules with an end point within the window. Conclusions buffy coat DNA, and the cord blood buffy coat DNA, 1 μg sonicated DNA was
drawn from those studies are that plasma DNA cleavages or used for library construction. The library concentrations ranged from 34 to
breakages cluster around locations in the genome in relation to 58 nM in a 20-μL library. For the first trimester, plasma DNA sequencing libraries
positions of nucleosome arrays and open chromatin domains. were constructed using a KAPA Library Preparation Kit (Kapa Biosystems).
However, as shown in Figs. 7 and 8, the preferred plasma DNA
Sequencing of DNA Libraries. The libraries for maternal plasma DNA and ge-
ending sites identified by this study are exquisitely sensitive to the
nomic DNA were sequenced using the HiSeq2500 and the HiSeq1500 Se-
genomic location. We have, therefore, shown that there is another quencing Systems (Illumina), respectively, using a paired end sequencing
layer of complexity associated with the nonrandom nature of protocol. Seventy-five nucleotides were sequenced from each end.
plasma DNA fragmentation that is beyond factors just related to
genome architecture or structural domains. Additional studies are Alignment of Sequencing Data. The paired end sequencing data were analyzed
needed to understand the relationship between the various factors by means of the SOAP2 in the paired end mode (20). The paired end reads were
and mechanisms resulting in the highly orchestrated and location- aligned to the nonrepeat-masked reference human genome (hg19). Up to two
specific patterns of plasma DNA fragmentation. We also believe nucleotide mismatches were allowed for the alignment of each end. The ge-
that a catalog of such tissue-specific end sites would provide an nomic coordinates of these potential alignments for the two ends were then
analyzed to determine whether any combination would allow the two ends to
alternative approach for elucidating the origin of plasma DNA in
be aligned to the same chromosome with the correct orientation spanning an
addition to methods based on methylation analysis (29) and nu- insert size ≤600 bp and mapping to a single location in the reference human
cleosome foot printing (16). genome. On average, 86% of the reads were aligned to the reference human
In summary, we have developed a second generation approach genome (hg19). For the detection of fetal de novo mutations from maternal
that produces noninvasive fetal genomes at high resolution using plasma, an additional step of realignment using the BOWTIE2 software was
maternal plasma DNA sequencing. We believe that this work has performed (19). All sequenced reads covering the candidate mutation sites
significantly pushed forward the limit of NIPT by showing the and exhibiting the variant alleles were realigned using BOWTIE2.
feasibility of detecting fetal de novo mutations on a genome-wide
level from maternal plasma. We have also substantially increased Binomial Mixture Model Analysis for Paternal Inheritance. To deduce the pa-
ternal inheritance of the fetus using maternal plasma DNA, we focused on SNPs
the resolution in which one could determine the maternal in-
at which the mother was homozygous (genotype AA) and the father was
heritance of the fetus using a nonhaplotype-based approach. heterozygous (genotype AB). At these SNPs, the genotype of the fetus would be
Currently, the potential clinical implementation of this approach is either AA or AB. Hence, we used a two-component binomial mixture model to
limited by the costs involved in sequencing maternal plasma DNA fit the observed allelic counts at each SNP locus (21). For each SNP locus i, the
to the depth reported here (195× and above; e.g., over US $40,000 counts for the A and B alleles are denoted as ai and bi, respectively, and Ni is
in our center). However, with the continual reduction in the costs the total number of reads covering the locus. Binomial mixture model was
of massively parallel sequencing, it is envisioned that this cost then used to determine if the fetus would be homozygous (AA) or hetero-
barrier would eventually be removed. Finally, our ΔS demonstra- zygous (AB) for each locus i based on the observed allelic counts ai and bi, the
sequencing error rate, and the fetal DNA fraction (21).
tion of preferred end sites in plasma DNA that exhibit specificity of

MEDICAL SCIENCES
their origin (e.g., the placenta of the fetus) has opened up nu-
GRAD Analysis for Maternal Inheritance. For SNPs in which the mother was
merous avenues of investigation. In the context of NIPT, it would heterozygous, we tested whether the two alleles were present in maternal
allow a relatively simple approach for estimating the fetal DNA plasma at the same or different concentrations using the sequential probability
fraction. Additional work would need to determine the relative ratio test (SPRT). An odds ratio of 20 was used for the calculation of the
merit between this approach and others previously reported [e.g., threshold for accepting the null or the alternative hypothesis. The null hy-
using plasma DNA size (28) and nucleosomal profiles (17)]. Ac- pothesis for each SPRT analysis was the absence of dosage imbalance between
tually, these methods could potentially be used in combination for the read counts for the two maternal alleles. The alternative hypothesis was the
NIPT. It would also be of great interest to explore the presence of overrepresentation of one allele. The calculation of the upper and lower
boundaries of the SPRT curves was as previously described (8).
such recurrent end positions in clinical and pathological scenarios
outside of the pregnancy context (e.g., cancer).
Dynamic Cutoff Analysis for de Novo Mutation. Dynamic cutoff filtering criteria
were developed to distinguish de novo mutations of the fetus from sequencing
Materials and Methods
errors. The sequencing error rate was assumed to be 0.3% (30). The probability
Clinical Samples. The study was approved by the Joint Chinese University of that the same variant would be observed in a number of sequenced reads
Hong Kong and Hospital Authority New Territories East Cluster Clinical Re- because of sequencing errors (Perr) would be calculated as
search Ethics Committee. For each of the two cases whose plasma DNA samples
   e ðbi −1Þ
were sequenced to high depths, 20 mL maternal venous peripheral blood was Ni
collected; 10 mL paternal venous peripheral blood was also collected. For the Perr = ×e× ,
bi 3
third trimester pregnant case, 3 mL cord blood was collected after delivery. For  
Ni
the case with cardiofaciocutaneous syndrome, the placenta was collected where represents a mathematical combination function Ni !=bi !ðNi − bi Þ!, e
bi
after therapeutic abortion. Twenty-six first trimester pregnant women were represents the sequencing error rate, Ni represents the total number of se-
recruited for fetal DNA fraction analysis. quenced reads covering the locus i, and bi represents the number of sequenced
reads carrying the variant. The dynamic cutoff values were determined based
Sample Processing. Blood samples were centrifuged at 1,600 × g for 10 min at on a theoretical false-positive rate of <1 in 3 × 109. In other words, the the-
4 °C. The plasma portion was harvested and recentrifuged at 16,000 × g for oretical number of false-positive sites caused by sequencing errors would be
10 min at 4 °C to remove the blood cells. The blood cell portion was recentri- less than one for a haploid genome.
fuged at 2,500 × g, and any residual plasma was removed. DNA from the blood
cells and that from maternal plasma were extracted with the blood and body Plasma DNA End Position Analysis. To screen for genomic coordinates that were
fluid protocol of the QIAamp DNA Blood Mini Kit and the QIAamp DSP DNA preferred ending positions for maternally and fetally derived DNA fragments,
Blood Mini Kit (Qiagen), respectively. DNA from the placenta was extracted fetal-specific informative SNP sites (i.e., at which the mother was homozygous
Downloaded by guest on July 8, 2021

with the QIAamp DNA Mini Kit (Qiagen) according to the manufacturer’s and the fetus was heterozygous) and maternal-specific informative SNP sites
tissue protocol. (i.e., at which the fetus was homozygous and the mother was heterozygous)

Chan et al. PNAS | Published online October 31, 2016 | E8167


were identified. For each informative SNP site, all of the sequencing reads Expected Proportion of Fragments Sharing Two Identical Ends. The probability
covering the informative SNP sites were analyzed. of having n fragments sharing the same ending position on one side, P(n), was
For the analysis of SNPs where the mother was homozygous (genotype AA) calculated using a Poisson probability function assuming that the average size
and the fetus was heterozygous (genotype AB), the A allele would be the of plasma DNA fragments was 166 bp and that the average sequencing depth
was 270×:
“shared allele,” and the B allele would be the fetal-specific allele. The number
of sequenced reads carrying the shared allele and the fetal-specific allele  
270
would be counted. In the size distribution of plasma DNA, a peak would be PðnÞ = Poisson n, .
166
observed at 166 bp for both the fetally and maternally derived DNA. If the
fragmentation of the plasma DNA had been random, the two ends would be The probability of two fragments having the same size (Psize) can be calcu-
evenly distributed across a region 166 bp upstream and 166 bp downstream of lated from the size distribution of the plasma DNA fragments:
the informative SNP. A P value would be calculated to determine if a particular
X
600
position has a significantly increased probability for being an end for the reads Psize = ½%ðsizeÞ2 ,
carrying the shared allele or the fetal-specific allele based on a Poisson prob- size=20

ability function:
where %(size) is the proportion of fragments having the particular size.
  Therefore, the overall probability of finding two fragments with identical
P  value = Poisson Nactual , Npredict ,
ends on both sides (Pboth) can be calculated as
where Poisson() is the Poisson probability function, Nactual is the actual number
X
∞  
of reads ending at the particular nucleotide, and Npredict is the total number of n
Pboth = PðnÞ × × Psize ,
2
reads divided by 166. n=2

A P value of <0.01 was used as a cutoff to define preferred ending positions  


n
where represents a mathematical combination function n!=2!ðn − 2Þ!.
for the reads carrying the fetal-specific allele or the shared allele. 2

For the SNPs where the mother was heterozygous (AB) and the fetus was
ACKNOWLEDGMENTS. This work was supported by the Hong Kong Research
homozygous, the reads carrying the maternal-specific allele (B allele) and the Grants Council Theme-Based Research Scheme T12-403/15. Y.M.D.L. was
shared alleles were analyzed. supported by an endowed chair from the Li Ka Shing Foundation.

1. Lo YM, et al. (1997) Presence of fetal DNA in maternal plasma and serum. Lancet 17. Straver R, Oudejans CB, Sistermans EA, Reinders MJ (2016) Calculating the fetal
350(9076):485–487. fraction for noninvasive prenatal testing based on genome-wide nucleosome profiles.
2. Chiu RW, et al. (2008) Noninvasive prenatal diagnosis of fetal chromosomal aneu- Prenat Diagn 36(7):614–621.
ploidy by massively parallel genomic sequencing of DNA in maternal plasma. Proc 18. Chandrananda D, Thorne NP, Bahlo M (2015) High-resolution characterization of
Natl Acad Sci USA 105(51):20458–20463. sequence signatures due to non-random cleavage of cell-free DNA. BMC Med
3. Chiu RW, et al. (2011) Non-invasive prenatal assessment of trisomy 21 by multiplexed Genomics 8:29.
maternal plasma DNA sequencing: Large scale validity study. BMJ 342:c7401. 19. Langmead B, Salzberg SL (2012) Fast gapped-read alignment with Bowtie 2. Nat
4. Bianchi DW, et al.; CARE Study Group (2014) DNA sequencing versus standard pre- Methods 9(4):357–359.
natal aneuploidy screening. N Engl J Med 370(9):799–808. 20. Li R, et al. (2009) SOAP2: An improved ultrafast tool for short read alignment.
5. Norton ME, et al. (2015) Cell-free DNA analysis for noninvasive examination of tri- Bioinformatics 25(15):1966–1967.
somy. N Engl J Med 372(17):1589–1597. 21. Jiang P, et al. (2012) FetalQuant: Deducing fractional fetal DNA concentration from
6. Srinivasan A, Bianchi DW, Huang H, Sehnert AJ, Rava RP (2013) Noninvasive detection massively parallel sequencing of DNA in maternal plasma. Bioinformatics 28(22):
of fetal subchromosome abnormalities via deep sequencing of maternal plasma. Am J 2883–2890.
Hum Genet 92(2):167–176. 22. Lun FM, et al. (2008) Noninvasive prenatal diagnosis of monogenic diseases by digital
7. Yu SC, et al. (2013) Noninvasive prenatal molecular karyotyping from maternal size selection and relative mutation dosage on DNA in maternal plasma. Proc Natl
plasma. PLoS One 8(4):e60968. Acad Sci USA 105(50):19920–19925.
8. Lo YM, et al. (2010) Maternal plasma DNA sequencing reveals the genome-wide 23. Veltman JA, Brunner HG (2012) De novo mutations in human genetic disease. Nat Rev
genetic and mutational profile of the fetus. Sci Transl Med 2(61):61ra91. Genet 13(8):565–575.
9. Fan HC, et al. (2012) Non-invasive prenatal measurement of the fetal genome. Nature 24. Chitty LS, et al. (2015) Non-invasive prenatal diagnosis of achondroplasia and tha-
487(7407):320–324. natophoric dysplasia: Next-generation sequencing allows for a safer, more accurate,
10. Kitzman JO, et al. (2012) Noninvasive whole-genome sequencing of a human fetus. and comprehensive approach. Prenat Diagn 35(7):656–662.
Sci Transl Med 4(137):137ra76. 25. Vajo Z, Francomano CA, Wilkin DJ (2000) The molecular and genetic basis of fibro-
11. Fan HC, Wang J, Potanina A, Quake SR (2011) Whole-genome molecular haplotyping blast growth factor receptor 3 disorders: The achondroplasia family of skeletal dys-
of single cells. Nat Biotechnol 29(1):51–57. plasias, Muenke craniosynostosis, and Crouzon syndrome with acanthosis nigricans.
12. Kitzman JO, et al. (2011) Haplotype-resolved genome sequencing of a Gujarati Indian Endocr Rev 21(1):23–39.
individual. Nat Biotechnol 29(1):59–63. 26. Saito H, Sekizawa A, Morimoto T, Suzuki M, Yanaihara T (2000) Prenatal DNA di-
13. Lam KW, et al. (2012) Noninvasive prenatal diagnosis of monogenic diseases by targeted agnosis of a single-gene disorder from maternal plasma. Lancet 356(9236):1170.
massively parallel sequencing of maternal plasma: Application to β-thalassemia. Clin 27. Chen S, et al. (2013) Haplotype-assisted accurate non-invasive fetal whole genome
Chem 58(10):1467–1475. recovery through maternal plasma sequencing. Genome Med 5(2):18.
14. New MI, et al. (2014) Noninvasive prenatal diagnosis of congenital adrenal hyper- 28. Yu SC, et al. (2014) Size-based molecular diagnostics using plasma DNA for non-
plasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab 99(6): invasive prenatal testing. Proc Natl Acad Sci USA 111(23):8583–8588.
E1022–E1030. 29. Sun K, et al. (2015) Plasma DNA tissue mapping by genome-wide methylation se-
15. Zeevi DA, et al. (2015) Proof-of-principle rapid noninvasive prenatal diagnosis of quencing for noninvasive prenatal, cancer, and transplantation assessments. Proc Natl
autosomal recessive founder mutations. J Clin Invest 125(10):3757–3765. Acad Sci USA 112(40):E5503–E5512.
16. Snyder MW, Kircher M, Hill AJ, Daza RM, Shendure J (2016) Cell-free DNA Comprises 30. Ross MG, et al. (2013) Characterizing and measuring bias in sequence data.
an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 164(1-2):57–68. Genome Biol 14(5):R51.
Downloaded by guest on July 8, 2021

E8168 | www.pnas.org/cgi/doi/10.1073/pnas.1615800113 Chan et al.

You might also like