ArticlePDF Available

Differential strand separation at critical temperature: A minimally disruptive enrichment method for low-abundance unknown DNA mutations

Authors:

Abstract and Figures

Detection of low-level DNA variations in the presence of wild-type DNA is important in several fields of medicine, including cancer, prenatal diagnosis and infectious diseases. PCR-based methods to enrich mutations during amplification have limited multiplexing capability, are mostly restricted to known mutations and are prone to polymerase or mis-priming errors. Here, we present Differential Strand Separation at Critical Temperature (DISSECT), a method that enriches unknown mutations of targeted DNA sequences purely based on thermal denaturation of DNA heteroduplexes without the need for enzymatic reactions. Target DNA is pre-amplified in a multiplex reaction and hybridized onto complementary probes immobilized on magnetic beads that correspond to wild-type DNA sequences. Presence of any mutation on the target DNA forms heteroduplexes that are subsequently denatured from the beads at a critical temperature and selectively separated from wild-type DNA. We demonstrate multiplexed enrichment by 100- to 400-fold for KRAS and TP53 mutations at multiple positions of the targeted sequence using two to four successive cycles of DISSECT. Cancer and plasma-circulating DNA samples containing traces of mutations undergo mutation enrichment allowing detection via Sanger sequencing or high-resolution melting. The simplicity, scalability and reliability of DISSECT make it a powerful method for mutation enrichment that integrates well with existing downstream detection methods.
Content may be subject to copyright.
Differential strand separation at critical temperature:
A minimally disruptive enrichment method for
low-abundance unknown DNA mutations
Minakshi Guha
1
, Elena Castellanos-Rizaldos
1
, Pingfang Liu
1
, Harvey Mamon
2
and
G. Mike Makrigiorgos
1,2,
*
1
Division of DNA Repair and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer
Institute, Harvard Medical School, Boston, MA and
2
Department of Radiation Oncology, Dana-Farber Cancer
Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
Received October 4, 2012; Revised October 31, 2012; Accepted November 3, 2012
ABSTRACT
Detection of low-level DNA variations in the
presence of wild-type DNA is important in several
fields of medicine, including cancer, prenatal diag-
nosis and infectious diseases. PCR-based methods
to enrich mutations during amplification have
limited multiplexing capability, are mostly restricted
to known mutations and are prone to polymerase or
mis-priming errors. Here, we present Di
fferential
S
trand Separation at Critical Temperature
(DISSECT), a method that enriches unknown muta-
tions of targeted DNA sequences purely based on
thermal denaturation of DNA heteroduplexes
without the need for enzymatic reactions. Target
DNA is pre-amplified in a multiplex reaction and
hybridized onto complementary probes immobilized
on magnetic beads that correspond to wild-type
DNA sequences. Presence of any mutation on the
target DNA forms heteroduplexes that are subse-
quently denatured from the beads at a critical tem-
perature and selectively separated from wild-type
DNA. We demonstrate multiplexed enrichment by
100- to 400-fold for KRAS and TP53 mutations at
multiple positions of the targeted sequence using
two to four successive cycles of DISSECT.
Cancer and plasma-circulating DNA samples
containing traces of mutations undergo mutation
enrichment allowing detection via Sanger
sequencing or high-resolution melting. The simpli-
city, scalability and reliability of DISSECT make it a
powerful method for mutation enrichment that inte-
grates well with existing downstream detection
methods.
INTRODUCTION
Cancer treatment is gradually moving toward
personalized therapy, allowing patients to avoid unneces-
sary or ineffective treatments (1). Tailoring therapies to
the genetic profile of tumors requires effective diagnostic
tools that would accurately detect clinically relevant
genetic alterations. The detection of mutated DNA is
often masked by an abundance of wild-type DNA
present in stromal tissue or in bodily fluids such as
plasma, urine or sputum (2–4). These low abundance mu-
tations are particularly important for early cancer detec-
tion, assessment of residual disease post treatment, disease
staging and monitoring of therapy following remission/
relapse ( 2 ). To date, several PCR-based technologies
have been developed for enrichment of low-abundance
mutations at known sequence positions including
methods that modify the targeted sequence (e.g. restriction
fragment length polymorphism or PCR-RFLP, restriction
endonuclease-mediated selective PCR or REMS-PCR
and allele-specific PCR), peptide/locked nucleic acid
approaches and co-amplification at lower denaturation
temperature-PCR or COLD-PCR (2,5–7). COLD-PCR
enables enrichment of any mutation on the sequence,
without prior knowledge of the mutation type or
position within the PCR amplicon. Accordingly, COLD-
PCR can be used in conjunction with downstream
sequencing methods to reveal low-level unknown muta-
tions (5,7,8). This is achieved by using preferential de-
naturation of mismatch-forming mutations at critical
denaturation temperature. Upon two consecutive
COLD-PCRs, mutations can be enriched by 100-fold or
more (7–11). Further mutation enrichment can be a chal-
lenge since polymerase errors and mis-priming events
increase upon additional PCR cycling.
As an alternative to PCR-based approaches, a bead-
based method known as DNA enrichment by allele-
*To whom correspondence should be addressed. Tel: +1 617 525 7122; Fax: +1 617 582 6037; Email: mmakrigiorgos@lroc.harvard.edu
Nucleic Acids Research, 2012, 1–9
doi:10.1093/nar/gks1250
ß The Author(s) 2012. Published by Oxford University Press.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by-nc/3.0/), which
permits non-commercial reuse, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact
journals.permissions@oup.com.
Nucleic Acids Research Advance Access published December 20, 2012
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
specific hybridization (DEASH) has been described to
enrich for low-abundance mutations (12). DEASH uses
allele-specific biotinylated oligonucleotides that hybridize
competitively to the location of the targeted base substi-
tution followed by capture on streptavidin-coated
paramagnetic beads. The simplicity of DEASH is
powerful, as it circumvents the use of enzymatic steps
and the potential generation of artifacts in the course of
genotypic selection. Nevertheless, the approach is limited
to the enrichment of known mutations since it requires
two carefully designed probes that match both the
mutant and the wild-type alleles.
Here, we describe a novel method based on Di
fferential
S
trand Separation at Critical Temperature (DISSECT) to
enrich unknown mutations within mixed DNA popula-
tions. Similar to COLD-PCR, DISSECT uses differential
denaturation of DNA heteroduplexes and can therefore
enrich mutations at any position on the sequence,
enabling mutation scanning and discovery via down-
stream sequencing. At the same time DISSECT avoids
polymerase extension or other enzymatic steps, as it is
entirely based on repeated cycles of hybridization and
preferential denaturation on solid support (streptavidin-
coated magnetic beads). Since the target sequence
remains unmodified during DISSECT, the resulting
mutation-enriched DNA pool can be combined with any
existing downstream detection method. Thus, DISSECT
can generate DNA template enriched for mutations by
200-fold or more, resulting in radically enhanced ability
to identify low-level nucleic acid alterations. Here, we
validate DISSECT for a variety of mutated DNA
targets of clinical interest (KRAS and TP53) and demon-
strate its simplicity, scalability and application to low-level
mutation enrichment in clinical cancer samples.
MATERIALS AND METHODS
Cell lines and tumor specimens
Human cancer cell lines SW480 and PFSK-1 were
purchased from American Type Culture Collection
(ATCC), and genomic DNA was extracted from
cultured cells using a DNeasy
TM
Blood and Tissue kit
(Qiagen, Valencia, CA, USA) as per the manufacturer’s
protocol. Genomic DNA for A549, NCI-H69, SNU-182
and HCC1008 were purchased directly from ATCC. To
demonstrate mutation enrichment, genomic DNA from
human cancer cell lines SW480, NCI-H69, SNU-182,
HCC1008, A549 and PFSK-1 were serially diluted into
WT DNA as a test panel for various mutations
(Supplementary Table S1). Human male genomic DNA
(Promega Corporation, Madison, WI, USA) served as
the wild-type control for all experiments. DNA mutation
abundances examined were 10, 5, 3, 1, 0.1 and 0.05%
mutant-to-WT ratios. For the capture of multiple muta-
tions in a single-tube, genomic DNA of SW480, NCI-H69
and SNU-182 were combined to generate a 5 and 1%
mixed mutant DNA dilution. All experiments were
carried out in parallel with wild-type controls and
mutant mixtures and were replicated at least three times
for validation.
Clinical colorectal tumor and lung tumor
samples containing low-level mutations previously
identified using COLD-PCR-sequencing, dHPLC and
REMS-PCR-sequencing (7,9,13,14) were also used to val-
idate DISSECT. Additionally, plasma-circulating
DNA donated by a radiation therapy patient following
informed consent and institutional review board
approval was used to demonstrate mutation enrichment
using DISSECT. Plasma-circulating DNA was previously
screened and validated to contain a TP53 mutation (15).
DNA obtained from both plasma and clinical samples
were pre-amplified using 20 cycles of multiplex PCR
prior to DISSECT, as described below.
Multiplex pre-amplification
Multiplex PCR primers (Supplementary Table S2) were
designed for 50 most commonly mutated exonic regions
identified in lung and esophageal cancers according to the
COSMIC database (16). These regions contain a mutant
count of greater than 1 for any single point mutation.
Multiplex pre-amplification from 20 ng genomic DNA
or 10 ng plasma-circulating DNA was performed in a
total volume of 25 ml using a mixture of 100 primers
(50 paired sets) at a final concentration of 0.3 mM for
each primer with 0.3 mM dNTPs, 3 mM MgCl
2
,1
Kapa HiFi buffer and 0.5 U of Kapa HiFi HotStart
DNA polymerase (Kapa Biosystems, Woburn, MA,
USA) reported to have an error rate of 2.8 10
7
mis-incorporations/bp. Multiplex PCR cycling was per-
formed according to the manufacturer’s recommendations
(Kapa Biosystems) for a total of 15–25 PCR cycles using
63
C as optimal annealing temperature. Amplicon lengths
ranged from 120 to 190 bp in size depending on the
amplicon (Supplementary Table S2). Following multiplex
cycling, 1 ml of exonuclease I (New England Biolabs,
Ipswich, MA, USA) was added to each reaction and
incubated at 37
C for 30 min and 80
C for 15 min to
remove unincorporated primers.
DISSECT procedure and downstream analysis
Probe design and preparation. Streptavidin-coated
Dynabeads (Life Technologies, Grand Island, NY,
USA) were coupled to biotinylated probe sequences of
30–95 nt long matching the wild-type target sequences of
TP53, KRAS and EGFR genes (Supplementary Table S3).
Probes for TP53 exons 5–9 and EGFR exons 19–21 were
synthesized by Integrated DNA Technologies Inc.
(Coralville, IA, USA). The probe for KRAS exon 2 was
synthesized to contain three locked nucleic acid bases,
LNA (Exiqon, Woburn, MA, USA). The LNA design
for KRAS exon 2 was based on previous reports for
LNA-assisted detection of KRAS mutation hotspots
(17,18). ‘Dual-biotin’ was used for the design of capture
probes to strengthen the bond between the streptavidin-
coated beads and the immobilized oligonucleotides during
the thermal cycling procedure (19). Dual-biotinylated
probes were conjugated to streptavidin beads as per the
manufacturer’s protocol (Invitrogen Life Technologies,
Grand Island, NY). Dynabead probes were finally
resuspended in a stock buffer containing 6 SSPE
2 Nucleic Acids Research, 2012
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
(0.9 M NaCl, 60 mM Na
2
HPO
4
, 6 mM EDTA) to a final
concentration of 10 mg/ml and kept at 4
C.
DISSECT protocol. 5–10 ml of Dynabead probes were
incubated with a 1:50 dilution of pre-amplified DNA in a
total volume of 50 mlof1 Phusion Buffer (New England
Biolabs). To hybridize DNA onto beads, DNA was
denatured at 95
C for 2 min, then annealed at 60
C for
2 min, 58
C for 2 min, 56
C for 2 min, 54
C for 1 min and
25
C for 5 min using an Eppendorf Mastercycler EP
machine (Eppendorf Inc., Haupage, NY, USA). Beads
were then separated using a Dynamag-PCR magnet
(Invitrogen Life Technologies, Grand Island, NY) and
washed twice with 50 mlof1 Phusion buffer to remove
unbound DNA fragments. Washed beads were resus-
pended in 55 ml1 Phusion buffer, and a 5-ml aliquot
was collected for testing DNA bound to beads.
Single-stranded mutant DNA was preferentially
denatured by heating the bead solution at a critical
denaturation temperature (e.g. 75
C) for 2 min followed
by magnetization, thereby collecting enriched mutated
DNA sequences in the supernatant. Critical denaturation
temperature (T
c
) of the shorter biotinylated probe se-
quences was predicted using IDT’s Oligo Analyzer that
predicts the melting temperature (T
m
) of the matched
(wild-type) sequence at the buffer conditions used for
DISSECT. A range of three to four potential T
c
values
were then experimentally tested to derive the optimal T
c
for the probe tested, ranging from 3
C below the predicted
T
m
and increasing temperature in 0.5
C steps until T
m-1
.
We found that adequate mutation enrichments are
obtained for denaturation temperature within 2
C for
most 30–45 nt probes, and optimal values for T
c
can be
arrived at using the approximation T
c
= T
m
(wild-type)-2
(data not shown). For longer 75–95 nt probes, the publicly
available UMelt software developed by the Wittwer la-
boratory (http://dna.utah.edu/umelt/umelt.html) was ini-
tially used to predict T
m
. The optimal T
c
was
experimentally derived using high-resolution melting
(HRM) analysis on the LightScanner HR96 system
(Idaho Technologies Inc.). Following elution from
beads, a 5-ml aliquot of the supernatant enriched for
mutant DNA was saved for analysis, while the remaining
supernatant was used for additional rounds of DISSECT
to increase the mutation enrichment as described in
Figure 1. These additional rounds of DISSECT required
a single 50 ml1 Phusion buffer replacement following the
step-down anneal procedure to eliminate unbound DNA
material in the supernatant. The same T
c
was used for
each denaturation step following capture of target DNA
from supernatant. For the multiplexed DISSECT
(combined bead capture), the same DISSECT procedure
was carried out, but with 2–4 ml of each kind of Dynabead
probe in a total bead volume of 8–16 ml per sample. The
capture probes used for multiplexed DISSECT were
designed to have similar T
m
so that a common critical
denaturation temperature could be used for multiplexed
mutation enrichment.
Downstream analysis . DNA eluted from beads following
DISSECT was amplified by conventional PCR or
COLD-PCR (5,14) followed by HRM analysis (9) and
Sanger Sequencing (Eton Bioscience, Cambridge, MA,
USA) (15). For post DISSECT amplification, nested
primers were used for each target region amplified in the
original pre-amplification reaction (Supplementary
Table S4).
RESULTS
The principle of DISSECT
The DISSECT procedure presented in Figure 1 illustrates
a bead-based method by which low-level mutations can be
enriched using repeated cycles of annealing and denatur-
ation at a critical temperature. Following multiplex
pre-amplification (Step 1) from genomic DNA, amplicons
are captured on magnetic beads using sequence-specific
binding to bead-immobilized DNA probes (Step 2).
Target DNA sequences are captured on beads by denatur-
ation of the double-stranded DNA at 95
C, followed by
step-down annealing procedure that allows single-
stranded DNA to anneal to complementary oligonucleo-
tides (probes) attached to streptavidin-coated magnetic
beads. These probes contain the wild-type sequence of
the targeted DNA regions (Supplementary Table S3),
and therefore the presence of a mutation at any
sequence position of the target DNA leads to a
mismatch that lowers the melting temperature (T
m
)of
the duplex. In Step 3 of Figure 1, mutation-containing
DNA is preferentially released from beads by denatur-
ation at a critical denaturation temperature (T
c
) corres-
ponding to the probe-target duplex. T
c
was defined as
detailed in the ‘Materials and Methods’ section. The
critical denaturation step preferentially elutes mutant
DNA strands into the supernatant, while a fairly large
fraction of the wild-type sequences remains substantially
bound to the beads and are subsequently removed by
magnetization. Steps 2 and 3 can be repeated multiple
times to enable further enrichment of mutation-containing
DNA regions. In the final step, the released mutation-
enriched sequences are PCR-amplified and analyzed.
HRM and Sanger sequencing were used for downstream
analysis. It should also be noted that although the
amplicons generated from the pre-amplification reaction
range from 120 to 190 bp, only the region of the amplicons
that corresponds to the immobilized probes actually
becomes enriched for mutations during DISSECT.
KRAS and TP53 mutation enrichment
As a large portion of lung cancer mutations occur in
KRAS (20) and TP53 genes (21,22), we implemented
DISSECT to analyze and enrich point mutations within
these two genes. Three successive rounds of DISSECT
were performed from a 1% mutation abundance of both
SW480 and A549 cell lines using a KRAS exon 2 probe at
a critical denaturation temperature of 75
C. Figure 2A
shows a 40- to 50-fold enrichment of two KRAS mutations
(c.35C>A, p.G12V and c.C>T, p.G12S) after DNA
elution following three rounds of DISSECT. Similarly,
tumor sample TL119 containing a double KRAS
mutation (c.34_35CC>AA, p.G12F) showed the same
degree of enrichment (Figure 2A). PCR-amplified
DISSECT products were also analyzed by HRM
Nucleic Acids Research, 2012 3
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
analysis showing incremental mutant enrichment for every
additional round of DISSECT for both SW480 and A549
mutation-containing DNA (Figure 2B). Beads containing
a non-LNA probe designed for TP53 exon 8 were equally
effective in enriching for low-level mutations from an
original 1 and 0.1% mutant abundance (Supplementary
Figures S1–S4). DISSECT was able to successfully
enrich low-level TP53 exon 8 T
m
-reducing mutations
(c.818C>T, Supplementary Figure S1), T
m
-retaining mu-
tations (c.841C>G, Supplementary Figure S2) and T
m
-
increasing mutations (c.823A>C, Supplementary Figure
S3). All three mutations were enriched with a single
bead-bound oligonucleotide probe, indicating the ability
to enrich different types of single-base substitutions within
the length of the DNA sequence covered by the probe.
These experiments yielded up to 100- to 200-fold
mutation enrichment after three rounds of DISSECT,
equivalent to mutant enrichments of 5-fold/cycle, as
inferred by comparing the apparent mutation abundance
in the Sanger chromatographs to the initial abundance of
0.1%. The gradual increase in mutation enrichment of
TP53 exon 8 after each round of DISSECT was
demonstrated via HRM analysis following DISSECT,
using 5, 1 or 0.1% original mutation abundances
(Supplementary Figure S4).
Next, to evaluate longer probes for use with DISSECT,
a 90-nt long TP53 exon 8 probe was used. In
Supplementary Figure S5A, serial dilutions of SW480
DNA into wild-type DNA (0.3, 1, 5 and 10% abundance)
were used to enrich for a c.818G>A mutation using two
rounds of DISSECT. Furthermore, a 1% mutation abun-
dance using DNA with a C>G mutation (HCC1008
DNA) within the same exon 8 target region resulted in
similar mutation enrichment (Supplementary Figure
S5B). Finally, a single-tube, multiplexed mutation enrich-
ment tested using 76, 95 and 90 nt probes for exons 5, 7
and 8, respectively, are depicted in Supplementary Figure
S5C. The data demonstrated that two rounds of
DISSECT in conjunction with the 76- to 95-nt probes
result in 20- to 60-fold mutation enrichments,
depending also on the mutation type and position.
The longer probes provide a broader ‘footprint’ for the
use of DISSECT in the enrichment of unknown
mutations.
Limits of mutation detection via Sanger sequencing and
HRM following DISSECT
Using an initial mutation abundance of 0.1%, conven-
tional PCR or single-round COLD-PCR-Sanger
sequencing failed to detect the mutation (Figure 3A).
Figure 1. DISSECT flowchart. Following multiplexed pre-amplification from genomic DNA (Step 1), enriched DNA target regions are captured on
complementary probes immobilized to magnetic beads resembling the wild-type form of the target sequence (Step 2). Mutation-containing target
DNA forms heteroduplexes that are subsequently denatured at a critical temperature leaving preferentially wild-type DNA bound to beads (Step 3).
Eluted DNA is separated and re-bound to a fresh aliquot of beads. Steps 2 and 3 are repeated to increase the mutation enrichment. Aliquots of
eluted DNA from each round are analyzed by downstream assays (Sanger sequencing and HRM were used in this work).
4 Nucleic Acids Research, 2012
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
In contrast, four rounds of DISSECT followed by conven-
tional PCR resulted to an apparent mutation abundance
of 26% for TP53 exon 8 and 16% for KRAS exon 2,
corresponding to 260- and 160-fold mutation enrichment,
respectively, or 4- to 5-fold mutation enrichment
per DISSECT cycle (Figure 3B, left). The final mutation
enrichment was higher when COLD-PCR was applied to
the eluted DNA following four rounds of DISSECT,
resulting in a mutation enrichment of 350- to 400-fold
(Figure 3B, right). The increased overall enrichment
reflects the additional enrichment provided by
COLD-PCR itself. Three or four rounds of DISSECT
followed by conventional PCR altered the HRM profiles
for both 0.1 and 0.05% mutant KRAS compared with
wild-type DNA (Figure 3C, middle), and these HRM
differences were greater when COLD-PCR was adopted
following DISSECT (Figure 3C, bottom). Similar results
were shown for mutations in TP53 exon 8 and KRAS exon
2 after four rounds of DISSECT from an initial mutation
of 0.05% (Supplementary Figure S6). In summary, three
or four rounds of DISSECT enrich low-level mutations,
originally present at 0.05–0.1% mutation abundance, to
levels of about 15–25%, allowing their subsequent
identification via COLD-PCR, HRM and Sanger
sequencing.
Multiplexing DISSECT
Bead-based processes are inherently scalable and
amenable to multiplexing. For a proof of principle in
multiplexing DISSECT, we prepared a 5 and 1%
mixtures of genomic DNA from cell lines SW480,
SNU-182 and NCI-H69, containing mutations in four dif-
ferent exon regions (Supplementary Table S1) diluted in a
wild-type DNA background. A 50-plex pre-amplification
was performed using this DNA mixture and processed for
the simultaneous mutation enrichment of four mutated
DNA targets using a combination of beads in a single
tube. The oligonucleotide probes attached to the beads
were designed to have similar melting temperatures,
within a ±0.5
C temperature range. During the single-
tube multiplexed DISSECT, a single denaturation
Figure 2. KRAS mutation enrichment. KRAS exon 2 enrichment following three rounds of DISSECT at a critical denaturation temperature of 75
C,
using immobilized KRAS oligonucleotide (30 nt, wild-type sequence). (A) Sanger sequencing chromatographs following conventional PCR amplifi-
cation of DISSECT products for three different KRAS mutations from an initial mutation abundance of 1% (c.35C>A, p.G12V and c.C>T, p.G12S
from SW480 and A549, respectively, and c.34_35CC>AA, p.G12F from a lung tumor sample, TL119). The DNA remaining on beads (left) is
compared with DNA eluted from beads following three rounds of DISSECT. (B) HRM analysis showing increasing mutation enrichment after every
round of DISSECT (R1-R3) for SW480 and A549 samples with 1% input mutation abundance.
Nucleic Acids Research, 2012 5
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
temperature of 75
C was used for the enrichment of all
four mutated DNA targets.
Figure 4 shows a combined mutation enrichment of
60-fold following three rounds of DISSECT for KRAS
exon 2, TP53 exon 5, TP53 exon 6 and TP53 exon 8
amplicons, using initial mutation abundances of 1–5%.
HRM analysis also depicts melting curves with increasing
differences for all four mutated amplicons (KRAS exon 2
and TP53 exons 5, 6 and 8) following each round of
DISSECT (Supplementary Figure S7). These results illus-
trate that DISSECT can be readily multiplexed provided
the T
m
of the bead-bound probes are matched.
Analysis of clinical tumor samples and circulating
DNA from plasma
To validate the use of DISSECT with clinical specimens,
we used previously tested DNA from lung and colorectal
tumor samples containing mid- to low-abundance TP53
and KRAS mutations (7, 14). Figure 5A shows strong
mutation enrichment following three rounds of
DISSECT for low-abundance TP53 exon 8 mutations in
tumor samples CT20, TL6 and TL121. HRM analysis also
demonstrates differences between the original DNA
captured on TP53 exon 8 beads versus DNA eluted after
three rounds of DISSECT followed by conventional
PCR-HRM (Figure 5B). Additional tumor specimens
were tested for enrichment of mutations from other
hotspot regions including TP53 exons 7 and 9 and
KRAS exon 2 (Supplementary Figure S8 ). A list of the
clinical samples, mutation positions and enrichment
following DISSECT is listed in Supplementary Table S5.
As an additional test, we used DISSECT to enrich mu-
tations from plasma-circulating DNA obtained from an
esophageal cancer patient undergoing radiation therapy.
The circulating DNA was purified from plasma and
pre-amplified using 20 cycles of multiplex PCR.
A biotinylated probe for DISSECT, p53Ex8-FC39
(Supplementary Table S3), was designed to enrich for a
previously identified mutation at an abundance of 0.5%
on TP53 exon 8 (c.847C>T, p.R823C) (15). Three rounds
of DISSECT resulted in a final mutation abundance of
36% (G>A, antisense) (Figure 5C). Alternatively, a
single round of COLD-PCR following two rounds of
DISSECT was performed, which resulted in mutation
abundance of 65% according to the chromatograph. In
summary, DISSECT can readily be applied to enrich
low-abundance mutations from clinical tumor specimens
and circulating tumor DNA prior to downstream
mutation detection.
Figure 3. Mutation enrichment following four rounds of DISSECT. (A) Sanger sequencing following direct conventional PCR or COLD-PCR in the
absence of DISSECT for TP53 exon 8 (C>T mutation) and KRAS exon 2 (C>A mutation) from an original 0.1% mutation abundance (from
SW480 mutant DNA diluted into wild-type DNA). (B) Sanger sequencing following direct conventional PCR or COLD-PCR after four rounds (R4)
of DISSECT. Resulting mutation abundance of 26% for TP53 and 16% for KRAS (corresponding to a final 160- to 260-fold mutation enrichment)
are depicted. DISSECT in combination with COLD-PCR increased the enrichment to 35% for TP53 and 42% for KRAS (corresponding to a final
400-fold mutation enrichment). (C) HRM analysis shows differential melting curves for KRAS exon 2, before and after DISSECT, from original
input mutation abundances of 0.05–0.1%. Data are representative of at least three independent experiments.
6 Nucleic Acids Research, 2012
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
DISCUSSION
DISSECT is a novel method for enriching DNA samples
for low-level unknown mutations that is purely based on
the thermal denaturation properties of DNA, without the
need for enzymatic reactions during the enrichment
process. The process is ‘minimally disruptive’ to the
input DNA material since no enzymatic modification
such as RFLP or sequence alterations by PCR primers
and polymerase extension occur during the mutation
enrichment, thus minimizing the probability for artifacts.
By using selective denaturation of mismatch-forming
target sequences bound to immobilized wild-type
sequence at critical temperature, DISSECT enriches for
most types of mutations, enabling mutation scanning
over the entire length of the probe. This was not
possible using the previously described DEASH competi-
tive hybridization method (12) because (a) special oligo-
nucleotides matching the specific targeted mutation were
needed and (b) selective denaturation confers superior se-
lectivity and faster kinetics over selective hybridization, as
our experience with COLD-PCR has demonstrated (5).
To date, few methods have been able to provide multi-
plexing capability in enriching low-level mutations in
diverse targets (2). Therefore, an advantage of DISSECT
lies in the ability to enrich multiple mutant targets in a
single tube without compromising accuracy or selectivity.
This can inherently enable multiplexed downstream
analysis platforms to be used with DISSECT, such as
matrix-assisted laser desorption/ionization time-of-flight
mass spectrometry, pyrosequencing and next-generation
sequencing, in addition to Sanger sequencing and HRM
implemented here.
In the present work, a pre-amplification step using a
50-plex PCR from genomic DNA was used to generate
target pre-amplification prior to applying DISSECT.
While this step combines conveniently with DISSECT,
the pre-amplification can potentially be omitted by per-
forming direct target capture from genomic DNA, as per-
formed with DEASH (12). This would eliminate potential
artifacts introduced during the initial 15–25 cycles of
pre-amplification. A limitation of DISSECT is that the
original DNA content is gradually reduced following
each round since part of the bead-bound DNA is
removed after the critical denaturation step. Thus, follow-
ing three to four consecutive rounds of DISSECT that
produced up to 100- to 400-fold mutation enrichment,
an amplification was applied prior to downstream analysis
by Sanger sequencing (or, potentially, prior to additional
rounds of DISSECT to increase mutation enrichment
further).
Here, we have shown that DISSECT can simultan-
eously enrich diverse targets for multiple mutations in
the same tube using a single denaturation temperature.
The mutation enrichment varies somewhat since the dif-
ferent DNA duplexes may not have the same optimal
critical denaturation temperature and also because
Figure 4. Multiplexed, single-tube DISSECT. Sanger sequencing analysis from a combined bead capture experiment showing simultaneous mutation
enrichment for four independent regions, KRAS exon 2 and TP53 exons 5, 6 and 8 following three rounds of DISSECT, from an original mutation
abundance of 1 and 5% (mixture of SW480, SNU-182 and NCI-H69 mutant DNA diluted into wild-type DNA, MP3). Data are representative of at
least three independent experiments.
Nucleic Acids Research, 2012 7
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
mutation enrichment depends on the specific mismatches
and the sequence context.
DISSECT is simple and amenable to scaling and auto-
mation, e.g. using liquid-handling robots, microfluidics or
micro-arrays. Accordingly, we anticipate using DISSECT
in routine genetic screening, especially in cancer-related
applications where it is important to identify low-level
mutations, such as EGFR resistance mutations (23) or pre-
dictive KRAS mutations (24,25). KRAS mutations at an
initial abundance of 0.05–0.1% are capable of being
directly detected via Sanger sequencing following
DISSECT (Figure 3), which illustrates a detection limit
improvement over previously reported methodologies
used to improve KRAS Sanger sequencing limits (13,26).
In the notable reports by Misale et al.(27) and Diaz et al.
(28), BEAM-ing was used to identify KRAS mutations at
the 0.1% level in plasma, as an indicator of developing
resistance to cetuximab treatment in colorectal cancer
patients. The BEAM-ing detection capability is similar
to that provided by DISSECT when followed by
PCR-Sanger sequencing (Figure 3). In addition,
DISSECT coupled with sequencing provides information
on adjacent positions on the same amplicon, thus enabling
‘unknown’ mutation detection and surveying of multiple
‘hotspots’ along the sequence. Accordingly, DISSECT
may also be used for detecting biomarker mutations in
tumor suppressor genes with multiple hotspot mutations
present in bodily fluids such as plasma-circulating DNA,
as was shown for the TP53 gene mutation in Figure 5.
In addition to cancer, DISSECT applications in prenatal
diagnosis or infectious diseases can also be envisioned.
In summary, DISSECT is a novel method for enriching
DNA target sequences for low-level unknown mutations
based on thermal denaturation properties of DNA
heteroduplexes, without the need for excessive amplifica-
tion or other enzymatic reactions. The process is readily
amenable to scaling and automation and can be combined
with existing downstream DNA sequencing platforms.
The simplicity of DISSECT makes it a powerful method
to screen for multiple mutation targets in a single tube
without compromising accuracy or sensitivity.
SUPPLEMENTARY DATA
Supplementary Data are available at NAR Online:
Supplementary Tables 1–5 and Supplementary Figures
1–8.
Figure 5. Clinical specimen analysis using DISSECT. Conventional PCR-Sanger sequencing (A) and conventional PCR-HRM (B) showing antisense
TP53 exon 8 mutations from lung (c.830C>A, TL6 and c.818C>T, TL121) and colorectal (c.818C>T, CT20) clinical tumor samples, after three
rounds of DISSECT. Results from DNA remaining on beads versus DNA eluted from beads are depicted (C) Plasma-circulating DNA from an
esophageal cancer patient with a low-level mutation (c.847G>A, R283C) was analyzed by DISSECT followed by conventional or
COLD-PCR-Sanger sequencing.
8 Nucleic Acids Research, 2012
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
FUNDING
NCI [R21 CA-111994 and R21 CA-155615, in part]. The
contents of this article do not necessarily represent
the official views of the National Cancer Institute or the
National Institutes of Health. Funding for open access
charge: Departmental funds.
Conflict of interest statement. None declared.
REFERENCES
1. Sadowska,A.M., Nowe,V., Janssens,A., Boeykens,E., De
Backer,W.A. and Germonpre,P.R. (2011) Customizing systemic
therapy in patients with advanced non-small cell lung cancer.
Ther. Adv. Med. Oncol., 3, 207–218.
2. Milbury,C.A., Li,J. and Makrigiorgos,G.M. (2009) PCR-based
methods for the enrichment of minority alleles and mutations.
Clin. Chem., 55, 632–640.
3. Sidransky,D., Von Eschenbach,A., Tsai,Y.C., Jones,P.,
Summerhayes,I., Marshall,F., Paul,M., Green,P., Hamilton,S.R.,
Frost,P. et al. (1991) Identification of p53 gene mutations in
bladder cancers and urine samples. Science, 252, 706–709.
4. Tada,M., Omata,M., Kawai,S., Saisho,H., Ohto,M., Saiki,R.K.
and Sninsky,J.J. (1993) Detection of ras gene mutations in
pancreatic juice and peripheral blood of patients with pancreatic
adenocarcinoma. Cancer Res., 53, 2472–2474.
5. Li,J., Wang,L., Mamon,H., Kulke,M.H., Berbeco,R. and
Makrigiorgos,G.M. (2008) Replacing PCR with COLD-PCR
enriches variant DNA sequences and redefines the sensitivity of
genetic testing. Nat. Med., 14, 579–584.
6. Milbury,C.A., Li,J. and Makrigiorgos,G.M. (2010) Ice-COLD-
PCR enables rapid amplification and robust enrichment for
low-abundance unknown DNA mutations. Nucleic Acids Res., 39,
e2.
7. Milbury,C.A., Correll,M., Quackenbush,J., Rubio,R. and
Makrigiorgos,G.M. (2011) COLD-PCR enrichment of rare cancer
mutations prior to targeted amplicon resequencing. Clin. Chem.,
58, 580–589.
8. Li,J., Milbury,C.A., Li,C. and Makrigiorgos,G.M. (2009)
Two-round coamplification at lower denaturation
temperature-PCR (COLD-PCR)-based sanger sequencing identifies
a novel spectrum of low-level mutations in lung adenocarcinoma.
Hum. Mutat., 30, 1583–1590.
9. Milbury,C.A., Li,J. and Makrigiorgos,G.M. (2009)
COLD-PCR-enhanced high-resolution melting enables rapid and
selective identification of low-level unknown mutations. Clin.
Chem., 55, 2130–2143.
10. Li,J., Wang,L., Janne,P.A. and Makrigiorgos,G.M. (2009)
Coamplification at lower denaturation temperature-PCR increases
mutation-detection selectivity of TaqMan-based real-time PCR.
Clin. Chem., 55, 748–756.
11. Boisselier,B., Marie,Y., Labussiere,M., Ciccarino,P., Desestret,V.,
Wang,X., Capelle,L., Delattre,J.Y. and Sanson,M. (2010) COLD
PCR HRM: a highly sensitive detection method for IDH1
mutations. Hum. Mutat., 31, 1360–1365.
12. Jeffreys,A.J. and May,C.A. (2003) DNA enrichment by
allele-specific hybridization (DEASH): a novel method for
haplotyping and for detecting low-frequency base substitutional
variants and recombinant DNA molecules. Genome Res., 13,
2316–2324.
13. Song,C., Milbury,C.A., Li,J., Liu,P., Zhao,M. and
Makrigiorgos,G.M. (2011) Rapid and sensitive detection of
KRAS mutation after fast-COLD-PCR enrichment and
high-resolution melting analysis. Diagn. Mol. Pathol., 20, 81–89.
14. Milbury,C.A., Chen,C.C., Mamon,H., Liu,P., Santagata,S. and
Makrigiorgos,G.M. (2011) Multiplex amplification coupled with
COLD-PCR and high resolution melting enables identification of
low-abundance mutations in cancer samples with low DNA
content. J. Mol. Diagn., 13, 220–232.
15. Castellanos-Rizaldos,E., Liu,P., Milbury,C.A., Guha,M., Brisci,A.,
Cremonesi,L., Ferrari,M., Mamon,H. and Makrigiorgos,G.M.
(2012) Temperature-tolerant COLD-PCr reduces temperature
stringency and enables robust mutation enrichment. Clin. Chem.,
58, 1130–1138.
16. Forbes,S.A., Bhamra,G., Bamford,S., Dawson,E., Kok,C.,
Clements,J., Menzies,A., Teague,J.W., Futreal,P.A. and
Stratton,M.R. (2008) The Catalogue of Somatic Mutations in
Cancer (COSMIC). Curr. Protoc. Hum. Genet., Chapter 10 , Unit
10.11.
17. Efrati,E., Elkin,H., Peerless,Y., Sabo,E., Ben-Izhak,O. and
Hershkovitz,D. (2010) LNA-based PCR clamping enrichment
assay for the identification of KRAS mutations. Cancer Biomark.,
8, 89–94.
18. Huang,Q., Wang,G.Y., Huang,J.F., Zhang,B. and Fu,W.L. (2010)
High sensitive mutation analysis on KRAS gene using LNA/
DNA chimeras as PCR amplification blockers of wild-type alleles.
Mol. Cell. Probes, 24, 376–380.
19. Diehl,F., Li,M., He,Y., Kinzler,K.W., Vogelstein,B. and
Dressman,D. (2006) BEAMing: single-molecule PCR on
microparticles in water-in-oil emulsions. Nat. Methods, 3,
551–559.
20. Suzuki,Y., Orita,M., Shiraishi,M., Hayashi,K. and Sekiya,T.
(1990) Detection of ras gene mutations in human lung cancers by
single-strand conformation polymorphism analysis of polymerase
chain reaction products. Oncogene, 5, 1037–1043.
21. Wistuba,I.I., Gazdar,A.F. and Minna,J.D. (2001) Molecular
genetics of small cell lung carcinoma. Semin. Oncol. , 28, 3–13.
22. Ding,L., Getz,G., Wheeler,D.A., Mardis,E.R., McLellan,M.D.,
Cibulskis,K., Sougnez,C., Greulich,H., Muzny,D.M.,
Morgan,M.B. et al. (2008) Somatic mutations affect key pathways
in lung adenocarcinoma. Nature, 455, 1069–1075.
23. Kuang,Y., Rogers,A., Yeap,B.Y., Wang,L., Makrigiorgos,M.,
Vetrand,K., Thiede,S., Distel,R.J. and Janne,P.A. (2009)
Noninvasive detection of EGFR T790M in gefitinib or erlotinib
resistant non-small cell lung cancer. Clin. Cancer Res., 15,
2630–2636.
24. Molinari,F., Felicioni,L., Buscarino,M., De Dosso,S., Buttitta,F.,
Malatesta,S., Movilia,A., Luoni,M., Boldorini,R., Alabiso,O.
et al. (2011) Increased detection sensitivity for KRAS mutations
enhances the prediction of anti-EGFR monoclonal antibody
resistance in metastatic colorectal cancer. Clin. Cancer Res., 17,
4901–4914.
25. Molinari,F. and Frattini,M. (2012) KRAS mutational test for
metastatic colorectal cancer patients: not just a technical problem.
Expert Rev. Mol. Diagn., 12, 123–126.
26. Araki,T., Shimizu,K., Nakamura,K., Nakamura,T., Mitani,Y.,
Obayashi,K., Fujita,Y., Kakegawa,S., Miyamae,Y., Kaira,K.
et al. (2010) Usefulness of peptide nucleic acid (PNA)-clamp
smart amplification process version 2 (SmartAmp2) for clinical
diagnosis of KRAS codon 12 mutations in lung adenocarcinoma:
comparison of PNA-clamp SmartAmp2 and PCR-related
methods. J. Mol. Diagn., 12, 118–124.
27. Misale,S., Yaeger,R., Hobor,S., Scala,E., Janakiraman,M.,
Liska,D., Valtorta,E., Schiavo,R., Buscarino,M., Siravegna,G.
et al. (2012) Emergence of KRAS mutations and acquired
resistance to anti-EGFR therapy in colorectal cancer. Nature,
486, 532–536.
28. Diaz,L.A. Jr, Williams,R.T., Wu,J., Kinde,I., Hecht,J.R.,
Berlin,J., Allen,B., Bozic,I., Reiter,J.G., Nowak,M.A. et al. (2012)
The molecular evolution of acquired resistance to targeted EGFR
blockade in colorectal cancers. Nature
, 486, 537–540.
Nucleic Acids Research, 2012 9
at Ernst Mayr Library of the Museum Comp Zoology, Harvard University on December 20, 2012http://nar.oxfordjournals.org/Downloaded from
... However, considering the need to identify mutations at very low subclonal levels (<1%), greater enrichment would have to be performed before most of the methods mentioned above, except the deep NGS. The COLD-PCR (coagulation at a lower denaturation temperature -PCR) 55 and the DISSECT (differential strand separation at a critical temperature) 56 are methods that allow the enrichment of the mutations by approximately 100-200 times. The former is based on preferential denaturation of the mutated DNA during PCR reactions in which the denaturation temperature is reduced. ...
... Most of the wild-type DNA will remain bound to the beads, which are removed from the solution with the help of a magnet. 56 It is important to note that these methods enrich only mutations that promote a reduction in the melting tem-perature of the amplicon. Furthermore, both require a pre-amplification step of the total genome. ...
... Furthermore, both require a pre-amplification step of the total genome. 55,56 ...
Article
Full-text available
Chronic lymphocytic leukemia is the most common hematologic malignancy among adults in Western countries. Several studies show that somatic mutations in the TP53 gene are present in up to 50% of patients with relapsed or refractory chronic lymphocytic leukemia. This study aims to review and compare the methods used to detect somatic TP53 mutations and/or 17p deletions and analyze their importance in the chronic lymphocytic leukemia diagnosis and follow-up. In chronic lymphocytic leukemia patients with refractory or recurrent disease, the probability of clonal expansion of cells with the TP53 mutation and/or 17p deletion is very high. The studies assessed showed several methodologies able to detect these changes. For the 17p deletion, the chromosome G-banding (karyotype) and interphase fluorescence in situ hybridization are the most sensitive. For somatic mutations involving the TP53 gene, moderate or high-coverage read next-generation sequencing and Sanger sequencing are the most recommended ones. The TP53 gene mutations represent a strong adverse prognostic factor for patient survival and treatment resistance in chronic lymphocytic leukemia. Patients carrying low-proportion TP53 mutation (less than 20–25% of all alleles) remain a challenge to these tests. Thus, for any of the methods employed, it is essential that the laboratory conduct its analytical validation, documenting its accuracy, precision and sensitivity/limit of detection.
... The use of bead-based hybrid capturing has also led to the development of enrichment technologies. The differential strand separation at critical temperature (DISSECT) method is a non-disruptive enrichment approach based on the thermal denaturation of DNA heteroduplexes and magnetic bead separation (32). Initially, target DNA is pre-amplified in a multiplex reaction. ...
Article
Background Presence of excess unaltered, wild-type DNA (wtDNA) providing information of little clinical value may often mask low-level mutations containing important diagnostic or therapeutic clues. This is a recurring hurdle in biotechnology and medicine, including cancer, prenatal diagnosis, infectious diseases, and organ transplantation. Mutation enrichment techniques that allow reduction of unwanted DNA to enable the detection of low-level mutations have emerged since the early 1990s. They are continuously being refined and updated with new technologies. The burgeoning interest in liquid biopsies for residual cancer monitoring, detection of resistance to therapy, and early cancer detection has driven an expanded interest in new and improved methodologies for practical and effective mutation enrichment and detection of low-level mutations of clinical relevance. Content Newly developed mutation enrichment technologies are described and grouped according to the main principle of operation, PCR-blocking technologies, enzymatic methods, and physicochemical approaches. Special emphasis is given to technologies enabling pre-PCR blockage of wtDNA to bypass PCR errors [nuclease-assisted minor-allele enrichment assay with overlapping probes (NaME-PrO) and UV-mediated cross-linking minor allele enrichment (UVME)] or providing high multiplexity followed by next-generation sequencing [Minor allele enriched sequencing through recognition oligonucleotides (MAESTRO)]. Summary This review summarizes technological developments in rare mutation enrichment over the last 12 years, complementing pre-2010 reviews on this topic. The expanding field of liquid biopsy calls for improved limits of detection (LOD) and highly parallel applications, along with the traditional requirements for accuracy, speed, and cost-effectiveness. The current technologies are reviewed with regards to these new requirements.
... ARSMS and PNA fit for the identification of single nucleotide polymorphisms, as specifically designed for detecting each mutation based on single base changes or small deletions [14,15]. Eventually, DISSECT has been implemented to detect low-level DNA variations in the presence of wild-type DNA [16]. The characteristic of DISSECT is the capacity to enrich unknown mutations of targeted DNA sequences based on the thermal denaturation of DNA heteroduplexes, thus with no need for enzymatic reactions. ...
Article
Circulating tumour DNA (ctDNA) is a novel tool that has being investigated in several types of tumours, includ-ing colorectal cancer (CRC). In fact, the techniques based on liquid biopsies are proposed as appealing non-invasive alter-natives to tissue biopsy, adding more insights into tumour molecular profile, heterogeneity and for cancer detection and monitoring. Additionally, some analysis showed that in CRC patients ctDNA seems to act as biomarker able to predict the outcome (prognostic role) and the response to treatments (predictive role). In particular, in the early stage CRC (stage I-III) it could represent a time marker of adjuvant therapy benefit as well as a marker of minimal residual disease and recurrence risk in addition to the already recognized risk factors. In metastatic CRC, the analysis of molecular tumour profile by ctDNA has shown to have high concordance with the tissue biopsy at diagnosis. Additionally, some studies demonstrated that ctDNA level during the treatment was linked with early response to treatment and prognosis. Finally, the quantitative anal-ysis of ctDNA and copy number alterations may be useful in order to detect resistance to therapy at the time of progression of disease and to help in finding new therapeutic targets
... The current technologies in testing ctDNA have two obstacles for early cancer diagnosis: possibly false positives and prior knowledge of the size of the ctDNA for primer design. [8][9][10][11][12] For early cancer genetic diagnosis, efficient sequence analysis of ctDNA would be more important with the recent surge of high-throughput technologies. Either Sanger's sequencing or the next generation sequencing requires amplification of the samples to be decoded, such as PCR amplification. ...
Article
Full-text available
A primerless amplification suitable for enrichment of particular genotype cfDNA which is a one-dimensional material has been developed. This primerless amplification coordinated by two thermostable enzymes of endonuclease and proofreading polymerase, functions as a genotype switch in analyzing cfDNA. The endonuclease digests the wild-typed fragments into mega-primer and discriminately destroys the wild-type DNA alleles. The DNA polymerase proofreads the mega-primer and then extends the mega-primer using the mutant DNA as the template. The prototypes of this technology were applied to two hotspot mutations of APC andEGFR with confirmed by DNA sequencing analysis. Genotype switch was then employed to clinical cfDNA assay targeting PIK3CA. Data from the clinical application suggest its potential in early cancer diagnosis.
... By suppressing the wild-type alleles with allele-specific blockers, amplification of the underrepresented mutant alleles is favored and sensitivities down to 0.1% can be achieved. 116 In addition, a variety of other methods have been developed, including bidirectional pyrophosphorolysis-activated polymerization (bi-PAP), 117,118 melting curve analysis using mutant specific hybridization probes and wild-type specific peptide nucleic acids (PNAs), 119 COLD-PCR, 120-122 DISSECT (Differential Strand Separation at Critical Temperature), 123,124 high-resolution melting analysis (HRM), 125,126 the use of LNAs (locked nucleic acids), 127,128 DHPLC, 129 mass spectrometry genotyping, 130 and UltraSEEK-a multiplex PCR, followed by mutation-specific, single-base extension using chain terminators labeled with a moiety for solid phase capture. 131 Although these methods can reach a high sensitivity, none of these methods seems to prevail. ...
Chapter
In recent years real-time monitoring using liquid biopsies has become a reality in cancer treatment. A liquid biopsy is defined as the sampling and analysis of circulating components from blood and other body fluids. The liquid biopsy holds great promise for precision and personalized medicine and in particular circulating cell-free dna (cfDNA) has been demonstrated to be a valuable tool to monitor recurrence, resistance, metastasis, and minimal residual disease. Although a plethora of options for the analysis of cfDNA exist-starting from the evaluation of single variants to gene panels and untargeted, genome-wide approaches-these assays are not yet ready for diagnosis or early detection of cancer. In this chapter, we discuss the state of the art of cell-free circulating DNA, present clinical applications for the most common cancer types and provide perspective on future developments.
Article
Point mutations can be used as biomarkers to perform diagnosis for diseases. In this study, a nanorobot for low-abundance point mutation enrichment was constructed using DNA origami. The novel design achieved limits of detection of 0.1% and 1% for synthesized DNA samples and clinical gene samples, respectively. Resettability was a key property of this method, which also involved a simpler process, lower cost and shorter detection duration than traditional enrichment methods. This novel DNA nanorobot may enable the detection of tumor markers, potentially facilitating early cancer diagnosis.
Article
Because cancer is caused by an accumulation of genetic mutations, mutant DNA released by tumors can be used as a highly specific biomarker for cancer. Although this principle was described decades ago, the advent and falling costs of next-generation sequencing have made the use of tumor DNA as a biomarker increasingly practical. This review surveys the use of cellular and cell-free DNA for the detection of cancer, with a focus on recent technological developments and applications to solid tumors. It covers ( a) key principles and technology enabling the highly sensitive detection of tumor DNA; ( b) assessment of tumor DNA in plasma, including for genotyping, minimal residual disease detection, and early detection of localized cancer; ( c) detection of tumor DNA in body cavity fluids, such as urine or cerebrospinal fluid; and ( d) challenges posed to the use of tumor DNA as a biomarker by the phenomenon of benign clonal expansions. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 16 is January 25, 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Liquid biopsies that analyse ctDNA hold great promise in the guidance of clinical treatment for various cancers. However, the innate characteristics of ctDNA make it a difficult target: ctDNA is highly fragmented, and found at very low concentrations, both in absolute terms and relative to wildtype species. Clinically-relevant target sequences often differ from the wildtype species by a single DNA base pair. These characteristics make analysing mutant ctDNA a uniquely difficult process. Despite this, techniques have recently emerged for analysing ctDNA, and have been used in pilot studies that showed promising results. These techniques each have various drawbacks, either in their analytical capabilities or practical considerations, which restrict their application to many clinical situations. Many of the most promising potential applications of ctDNA require assay characteristics that are not currently available, and new techniques with these properties could have benefits in companion diagnostics, monitoring response to treatment and early detection. Here we review the current state of the art in ctDNA detection, with critical comparison of the analytical techniques themselves. We also examine the improvements required to expand ctDNA diagnostics to more advanced applications and discuss the most likely pathways for these improvements.
Article
Full-text available
Colorectal cancer (CRC) is among the most commonly diagnosed cancers worldwide. Point mutations on the Kisten rat sarcoma viral oncogene homologue (KRAS) have been identified as a crucial diagnostic and prognostic biomarker for response to cancer therapy targeting the epidermal growth factor receptor. A rapid label-free colorimetric assay for detection of point mutations in codon 12 and 13 of the KRAS gene using peptide nucleic acid (PNA) and unmodified gold nanoparticles (AuNPs) was presented in this study. Charge neutral PNA induces the AuNPs aggregation in the absence of complementary DNA; when the complementary DNA is present, PNA–DNA hybridization retains the dispersion of AuNPs as the negative charges of the DNA strand adsorbed on the AuNPs surface ensure sufficient charge repulsion. Based on this principle, the developed assay was able to detect the specific sequence in the presence of at least 20 times the amount of interference oligonucleotides, and the limit of detection was low as DNA/PNA ratio of 0.02. The PNA induced colorimetric changes in the presence of the complementary DNA at different ratios of DNA/PNA yielded a standard curve with a linear coefficient of R² = 0.9371. Furthermore, this assay was validated in order that it could be used to detect the seven known mutant alleles in codon 12 and 13 of the KRAS gene by the naked eye with higher concentration of PNA. This simple colorimetric assay is rapid, sensitive and easily performed at room temperature without additional stringent conditions, which promises the method great potential utility for clinical sample screening and point-of-care testing.
Article
Full-text available
Colorectal tumours that are wild type for KRAS are often sensitive to EGFR blockade, but almost always develop resistance within several months of initiating therapy. The mechanisms underlying this acquired resistance to anti-EGFR antibodies are largely unknown. This situation is in marked contrast to that of small-molecule targeted agents, such as inhibitors of ABL, EGFR, BRAF and MEK, in which mutations in the genes encoding the protein targets render the tumours resistant to the effects of the drugs. The simplest hypothesis to account for the development of resistance to EGFR blockade is that rare cells with KRAS mutations pre-exist at low levels in tumours with ostensibly wild-type KRAS genes. Although this hypothesis would seem readily testable, there is no evidence in pre-clinical models to support it, nor is there data from patients. To test this hypothesis, we determined whether mutant KRAS DNA could be detected in the circulation of 28 patients receiving monotherapy with panitumumab, a therapeutic anti-EGFR antibody. We found that 9 out of 24 (38%) patients whose tumours were initially KRAS wild type developed detectable mutations in KRAS in their sera, three of which developed multiple different KRAS mutations. The appearance of these mutations was very consistent, generally occurring between 5 and 6 months following treatment. Mathematical modelling indicated that the mutations were present in expanded subclones before the initiation of panitumumab treatment. These results suggest that the emergence of KRAS mutations is a mediator of acquired resistance to EGFR blockade and that these mutations can be detected in a non-invasive manner. They explain why solid tumours develop resistance to targeted therapies in a highly reproducible fashion.
Article
Full-text available
A main limitation of therapies that selectively target kinase signalling pathways is the emergence of secondary drug resistance. Cetuximab, a monoclonal antibody that binds the extracellular domain of epidermal growth factor receptor (EGFR), is effective in a subset of KRAS wild-type metastatic colorectal cancers. After an initial response, secondary resistance invariably ensues, thereby limiting the clinical benefit of this drug. The molecular bases of secondary resistance to cetuximab in colorectal cancer are poorly understood. Here we show that molecular alterations (in most instances point mutations) of KRAS are causally associated with the onset of acquired resistance to anti-EGFR treatment in colorectal cancers. Expression of mutant KRAS under the control of its endogenous gene promoter was sufficient to confer cetuximab resistance, but resistant cells remained sensitive to combinatorial inhibition of EGFR and mitogen-activated protein-kinase kinase (MEK). Analysis of metastases from patients who developed resistance to cetuximab or panitumumab showed the emergence of KRAS amplification in one sample and acquisition of secondary KRAS mutations in 60% (6 out of 10) of the cases. KRAS mutant alleles were detectable in the blood of cetuximab-treated patients as early as 10 months before radiographic documentation of disease progression. In summary, the results identify KRAS mutations as frequent drivers of acquired resistance to cetuximab in colorectal cancers, indicate that the emergence of KRAS mutant clones can be detected non-invasively months before radiographic progression and suggest early initiation of a MEK inhibitor as a rational strategy for delaying or reversing drug resistance.
Article
Full-text available
Low-level mutations in clinical tumor samples often reside below mutation detection limits, thus leading to false negatives that may impact clinical diagnosis and patient management. COLD-PCR (coamplification at lower denaturation temperature PCR) is a technology that magnifies unknown mutations during PCR, thus enabling downstream mutation detection. However, a practical difficulty in applying COLD-PCR has been the requirement for strict control of the denaturation temperature for a given sequence, to within ±0.3 °C. This requirement precludes simultaneous mutation enrichment in sequences of substantially different melting temperature (T(m)) and limits the technique to a single sequence at a time. We present a temperature-tolerant (TT) approach (TT-COLD-PCR) that reduces this obstacle. We describe thermocycling programs featuring a gradual increase of the denaturation temperature during COLD-PCR. This approach enabled enrichment of mutations when the cycling achieves the appropriate critical denaturation temperature of each DNA amplicon that is being amplified. Validation was provided for KRAS (v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog) and TP53 (tumor protein p53) exons 6-9 by use of dilutions of mutated DNA, clinical cancer samples, and plasma-circulating DNA. A single thermocycling program with a denaturation-temperature window of 2.5-3.0 °C enriches mutations in all DNA amplicons simultaneously, despite their different T(m)s. Mutation enrichments of 6-9-fold were obtained with TT-full-COLD-PCR. Higher mutation enrichments were obtained for the other 2 forms of COLD-PCR, fast-COLD-PCR, and ice-COLD-PCR. Low-level mutations in diverse amplicons with different T(m)s can be mutation enriched via TT-COLD-PCR provided that their T(m)s fall within the denaturation-temperature window applied during amplification. This approach enables simultaneous enrichment of mutations in several amplicons and increases significantly the versatility of COLD-PCR.
Article
Full-text available
Despite widespread interest in next-generation sequencing (NGS), the adoption of personalized clinical genomics and mutation profiling of cancer specimens is lagging, in part because of technical limitations. Tumors are genetically heterogeneous and often contain normal/stromal cells, features that lead to low-abundance somatic mutations that generate ambiguous results or reside below NGS detection limits, thus hindering the clinical sensitivity/specificity standards of mutation calling. We applied COLD-PCR (coamplification at lower denaturation temperature PCR), a PCR methodology that selectively enriches variants, to improve the detection of unknown mutations before NGS-based amplicon resequencing. We used both COLD-PCR and conventional PCR (for comparison) to amplify serially diluted mutation-containing cell-line DNA diluted into wild-type DNA, as well as DNA from lung adenocarcinoma and colorectal cancer samples. After amplification of TP53 (tumor protein p53), KRAS (v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog), IDH1 [isocitrate dehydrogenase 1 (NADP(+)), soluble], and EGFR (epidermal growth factor receptor) gene regions, PCR products were pooled for library preparation, bar-coded, and sequenced on the Illumina HiSeq 2000. In agreement with recent findings, sequencing errors by conventional targeted-amplicon approaches dictated a mutation-detection limit of approximately 1%-2%. Conversely, COLD-PCR amplicons enriched mutations above the error-related noise, enabling reliable identification of mutation abundances of approximately 0.04%. Sequencing depth was not a large factor in the identification of COLD-PCR-enriched mutations. For the clinical samples, several missense mutations were not called with conventional amplicons, yet they were clearly detectable with COLD-PCR amplicons. Tumor heterogeneity for the TP53 gene was apparent. As cancer care shifts toward personalized intervention based on each patient's unique genetic abnormalities and tumor genome, we anticipate that COLD-PCR combined with NGS will elucidate the role of mutations in tumor progression, enabling NGS-based analysis of diverse clinical specimens within clinical practice.
Article
Full-text available
Lung cancer is the leading cause of cancer deaths worldwide. Standard chemotherapy has been shown to improve quality of life and has a modest influence on overall survival. This modest improvement in survival is partly due to the choice of chemotherapy regimens that have been based on prognostic factors such as age, performance status and comorbidities of the patient. This underlines the importance of developing a more personalized therapy for patients with non-small cell lung cancer. Such an approach may reduce the variation in how individual patients respond to medications by tailoring therapies to their genetic profile. In this review we focus on several aspects of customized therapy, looking not only at patient characteristics but also to tumor histology and specific tumor biomarkers.
Article
The status of epidermal growth factor receptor (EGFR) and Kirsten ras (KRAS) mutations has been used widely in management of patients with non-small cell lung cancer (NSCLC). However, it may be difficult to get tumor tissues for analyzing the status of EGFR and KRAS mutation in large proportion of patients with advanced disease. We obtained pairs of tumor and serum samples from 57 patients with advanced NSCLC, between March 2006 and January 2009. EGFR mutation status from tumor samples and KRAS mutation status from serum samples were analyzed by genomic polymerase chain reaction and direct sequence, and EGFR mutation status from serum samples was determined by the peptide nucleic acid-locked nucleic acid PCR clamp. EGFR mutations were detected in the serum samples of 11 patients and in the tumor samples of 12 patients. Fourteen patients revealed (?) KRAS mutation in the serum sample. EGFR mutation status in the serum and tumor samples was consistent in 50 (87.7 %) of the 57 pairs (correlation index 0.62, p < 0.001). Only 5 of 57 (8.7 %) patients showed mutation of both EGFR and KRAS in serum sample. Twenty-two of 57 patients (38.5 %) received EGFR-TKIs as any line therapy. The response for EGFR-TKIs was significantly associated with EGFR mutations in both tumor samples and serum samples (p < 0.05). The status of KRAS mutation in serum was not predictive for the response of EGFR-TKI (p > 0.05). There was no significant difference in OS according to the status of EGFR mutations in both serum and tumor samples (p > 0.05) and KRAS mutations in serum samples (p > 0.05). The status of EGFR and KRAS mutation in serum was not prognostic in patients with advanced NSCLC. However, the clinical usefulness of EGFR mutation of serum as a selection marker for EGFR-TKIs sensitivity in NSCLC might be allowed, not KRAS mutation.
Article
The identification of KRAS and BRAF mutations as predictive molecular alterations of resistance to EGF receptor monoclonal antibody therapy in metastatic colorectal cancer have significantly improved the selection of patients more likely to be eligible for the treatment with these targeted agents. Several methods are available for KRAS and BRAF mutation detection but few studies have compared different techniques, especially in the clinical setting. In this article, we contextualize the wobble-enhanced amplification refractory mutation sequencing method for the identification of KRAS and BRAF mutations with the other methodologies frequently used for the assessment of these alterations in colorectal cancer, discussing advantages and limitations over other frequently used diagnostic methods.