You are on page 1of 12

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Frontiers in Medicine

Application of Cell-free DNA Analysis


to Cancer Treatment
Ryan B. Corcoran, M.D., Ph.D., and Bruce A. Chabner, M.D.​​

T
From the Massachusetts General Hos­ umor biopsies represent the standard for cancer diagnosis
pital Cancer Center and the Department and the primary method for molecular testing to guide the selection of
of Medicine, Harvard Medical School,
Boston. Address reprint requests to Dr. precision therapies. Liquid biopsies, particularly those involving cell-free
Corcoran at Massachusetts General Hos­ DNA (cfDNA) from plasma, are rapidly emerging as an important and minimally
pital Cancer Center, 149 13th St., 7th Fl., invasive adjunct to standard tumor biopsies and, in some cases, even a potential
Boston, MA 02129, or at ­rbcorcoran@​
­partners​.­org. alternative approach. Liquid biopsy is becoming a valuable tool for molecular test-
ing, for new insights into tumor heterogeneity, and for cancer detection and
N Engl J Med 2018;379:1754-65.
DOI: 10.1056/NEJMra1706174 monitoring. Here, we review the current and potential clinical applications of
Copyright © 2018 Massachusetts Medical Society. cfDNA analysis in patients with cancer (see video).

L iquid Biopsie s
Although liquid biopsy has most often referred to the analysis of cfDNA from
peripheral blood, this term also encompasses the isolation and analysis of tumor-
derived material (e.g., DNA, RNA, or even intact cells) from blood or other bodily
An illustrated fluids1,2 (Fig. 1). For example, intact circulating tumor cells intravasate into the
glossary and a bloodstream at low frequency (often <10 circulating tumor cells per milliliter of
video overview of blood in patients with metastatic cancer). With specialized technology, circulating
cfDNA analysis
tumor cells can be detected and isolated from a background of normal blood cells,
in cancer are
available at facilitating molecular analysis and even implantation and growth of these cells in
NEJM.org immunocompromised mice.1-3 Subcellular particles called exosomes, or extracellular
membrane-encased vesicles, are also released by tumor cells into the bloodstream
and contain tumor-specific proteins and nucleic acids.4 Cell-free nucleic acids, in-
cluding cell-free RNA (which is less stable than DNA)5 and cfDNA (the focus of
this review), are also released into the circulation.
Blood is not the only bodily fluid that can be used for liquid-biopsy approaches.
Urine, stool, cerebrospinal fluid (CSF), saliva, pleural fluid, and ascites are all
potential sources of tumor-derived material, including cfDNA.2,6-8 Although this
review will focus on the analysis of cfDNA from blood, detection and analysis of
cfDNA from other bodily fluids may have applications for specific cancer types
(e.g., CSF for cancers of the central nervous system) or for the detection of cancers
arising in defined organ systems (e.g., stool for colorectal cancer or saliva for head
and neck cancers).

Pl a sm a-Der i v ed cf DNA
The term “cfDNA” refers to fragmented DNA found in the noncellular component
of the blood, as first reported by Mandel and Metais in 1948.9 It is thought that
cfDNA is released into the bloodstream through apoptosis or necrosis, and cfDNA
is typically found as double-stranded fragments of approximately 150 to 200 base
pairs in length, corresponding to nucleosome-associated DNA.10 Molecules of
cfDNA are rapidly cleared from the circulation, with a half-life of an hour or less.

1754 n engl j med 379;18 nejm.org  November 1, 2018

The New England Journal of Medicine


Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Cell-free DNA Analysis and Cancer Treatment

Liquid-Biopsy Sources

Cerebrospinal fluid
Tumors of the central nervous system

Saliva
Head and neck tumors

Pleural fluid
• Thoracic cancers
• Metastatic cancers

Peripheral blood

BLOOD
VESSEL
Tumor

Circulating
tumor cells

Exosomes
Apoptotic
tumor cell
cfDNA

Ascites
Metastatic cancers

Stool
Gastrointestinal tract cancers

Urine
• Urinary tract cancers
• cfDNA filtered from blood

Figure 1. Overview of Liquid-Biopsy Approaches.


Liquid­biopsy approaches involving peripheral blood include isolation of circulating tumor cells, which intravasate from tumors into the
bloodstream; exosomes, which are membrane­bound vesicles released by tumor cells; and cell­free DNA (cfDNA), which is released by
apoptotic or necrotic tumor cells. Although peripheral blood is the most common source of liquid biopsy, several other bodily fluids have
been used for specific liquid­biopsy applications, including isolation and analysis of cfDNA, as shown.

The cfDNA from normal cells is found in plas- DNA (ctDNA) and constitutes only a portion of
ma at low levels in healthy persons (approxi- the overall cfDNA. The fraction of ctDNA in
mately 10 to 15 ng per milliliter, on average),11 overall cfDNA in patients with cancer can vary
and the level can increase under conditions of greatly, from less than 0.1% to more than
tissue stress, including exercise, inflammation, 90%.12,13 Although the fraction of ctDNA tends
surgery, or tissue injury.12 to parallel tumor burden within an individual
More than 40 years ago, it was observed that patient, substantial variability has been observed
patients with cancer have higher overall levels of among patients with the same cancer type, possi-
cfDNA than persons without cancer.11 In patients bly reflecting biologic differences or differences
with cancer, cfDNA that is released from tumor in rates of cell death in the individual tumors.13
cells is often referred to as circulating tumor Moreover, patients with different tumor types

n engl j med 379;18 nejm.org November 1, 2018 1755


The New England Journal of Medicine
Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

show considerable variation in the frequency of nucleotide barcode), and error-suppression algo-
detectable ctDNA. Thus, the detection and analy- rithms have improved the limits of detection.23-25
sis of ctDNA amidst a background of normal
germline cfDNA presents a considerable challenge. Cl inic a l A ppl ic at ions
The ability to analyze tumor-derived DNA from
Isol at ion a nd A na lysis of cf DNA
a routine blood draw without the need for an
Owing to the low levels and short half-life of invasive tumor biopsy represents a critical ad-
tumor-derived cfDNA, specialized approaches are vance with potentially transformative clinical ap-
needed for both isolation and analysis of cfDNA. plications (Fig. 3). In particular, the minimally
Two key issues are the stability of the cfDNA it- invasive nature of cfDNA analysis provides a
self and the potential for lysis of normal blood means of molecular profiling for tumors that
cells, leading to contamination with normal DNA. are difficult or unsafe to biopsy and allows a
To limit these effects, when cfDNA is isolated practical means for monitoring tumor DNA seri-
from blood collected in standard phlebotomy ally over time without the risk and potential
tubes, plasma must typically be centrifuged and complications of standard tumor biopsy. In ad-
separated within 1 to 4 hours after collection. dition, cfDNA analysis may better capture the
This need for rapid processing creates logistic molecular heterogeneity harbored by multiple dis-
challenges and the potential for preanalytic vari- tinct clonal populations in a patient’s tumor, as
ability caused by fluctuations of cfDNA concen- compared with a needle biopsy of a single tumor
tration and purity due to differences in process- lesion. Finally, cfDNA analysis offers the poten-
ing times.14,15 Alternatively, specialized cfDNA tial for tumor detection or monitoring in patients
collection tubes containing fixatives can stabilize without clinically evident disease.
both cfDNA and intact cells for up to 7 to 14
days at room temperature, allowing for easy Diagnosis and Molecular Profiling
shipping, storage, and batched or centralized Tumor molecular profiling for the selection of
processing.16 therapy has become a fundamental practice in
The fraction of ctDNA within the background cancer medicine.26 The potential to assess the
of normal cfDNA in patients with cancer is typi- molecular profile of a patient’s tumor from a
cally small and highly variable from patient to simple blood draw, without the need for an inva-
patient. Thus, ultrasensitive methods are required sive biopsy, makes cfDNA analysis an attractive
to detect mutations, copy-number changes, or other tool. However, a key initial question is whether
alterations that are present in cfDNA at very low the mutational profile established through cfDNA
variant-allele frequencies (i.e., the percentage testing reliably reproduces the mutational pro-
of variant alleles present among all alleles, in- file derived from a direct tumor biopsy, which re-
cluding wild-type alleles) (Fig. 2). For detection mains the standard of care. Early studies, based
of individual point mutations, mutation-specific on small numbers of patient samples, suggest-
techniques based on polymerase-chain-reaction ed low concordance between DNA alterations
(PCR) analysis — such as BEAMing (beads, detected in tumor and plasma samples from the
emulsion, amplification, and magnetics) or drop- same patient.27,28 However, the validity of these
let digital PCR (ddPCR) analysis — can identify studies was compromised by the shortcoming
and quantify alterations present at allele frequen- that tumor and plasma samples were often not
cies of 0.01% or less in cfDNA.17,18 Next-genera- collected at the same time, yielding potential
tion sequencing methods have also been tailored differences due to molecular evolution of the
for cfDNA, ranging from whole-genome or whole- tumor. In addition, many plasma samples were
exome sequencing to targeted sequencing of a collected at suboptimal times, such as during
limited gene panel.19-22 However, the sensitivity therapy, when ctDNA levels are at their lowest.
and specificity of these approaches are limited by Nonconcordance of key alterations is most
the error rate of DNA polymerase and the se- often observed in patients with low ctDNA levels,
quencing reaction. Thus, modified approaches in- and low levels make detection of alterations more
corporating deep sequencing coverage, molecular challenging. For example, one recent study com-
barcoding methods (in which individual input pared the performance of two commercial cfDNA
template DNA fragments are tagged with a unique sequencing assays on parallel pairs of blood

1756 n engl j med 379;18 nejm.org  November 1, 2018

The New England Journal of Medicine


Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Cell-free DNA Analysis and Cancer Treatment

Mutations Nucleotide coverage


10%
• Whole-genome sequencing
• Whole-exome sequencing
1%

Targeted next-generation sequencing

Copy-number alterations 0.1%


Next-generation sequencing with molecular
barcodes
0.01%
• BEAMing
• Droplet digital PCR analysis
Plasma 0.001%
Limit of detection

Next-generation sequencing with molecular barcodes


Gene fusions Wild type Mutant Wild type
Buffy coat
(source of
germline DNA) Molecular Each DNA molecule
barcode tags tagged with barcode

Redundant sequencing; true


mutations represented in all
reads with given barcode

DNA methylation

CH3
Normal cfDNA
ctDNA
CH3
True mutation Polymerase error

Figure 2. Isolation and Analysis of cfDNA.


As shown on the left, cfDNA is isolated from plasma after centrifugation of peripheral blood to separate the cellular component of the
blood, including white and red cells. Circulating tumor DNA (ctDNA) is found (often at low fractions) among a background of normal
germline cfDNA released by normal cells throughout the body. The cfDNA can be analyzed to identify several common DNA­based altera­
tions observed in tumors, including mutations, copy­number alterations, gene fusions, and DNA methylation changes. As shown at the
upper right, owing to the frequently low fraction of ctDNA in a background of normal cfDNA, specialized methods for cfDNA analysis
have been developed, including sequencing and methods based on polymerase­chain­reaction (PCR) analysis (BEAMing [beads, emulsion,
amplification, and magnetics] and droplet digital PCR). Typically, those analyses providing the greatest breadth or nucleotide coverage
have lesser sensitivity and require higher fractions of ctDNA in overall cfDNA (percentages in red) for analysis. Conversely, methods that
are directed against targeted mutation panels or single mutations offer improved limits of detection. As shown at the lower right, molecu­
lar barcoding to tag individual molecules of DNA has allowed next­generation sequencing approaches for cfDNA to achieve better limits
of detection, allowing discrimination of true mutations present in the original template DNA molecule (and thus present in all sequenc­
ing reads corresponding to a specific molecular barcode) from mutations introduced by polymerase error during the sequencing reaction.

samples drawn from 40 patients with metastatic to low cfDNA levels at or below the limit of de-
prostate cancer.29 Low concordance rates were tection of these assays. In addition, the way in
observed, raising questions about the reproduc- which true positive or negative results are scored
ibility of cfDNA analysis. However, the study in patients with low levels of cfDNA remains an
had major limitations, including the facts that important challenge.
at least half the patients had normal levels of Larger and more carefully controlled studies
prostate-specific antigen and that participants have shown high concordance rates of 80 to 90%
were not restricted to those not receiving therapy. between plasma and tissue samples obtained
Thus, much of the variability may be attributable simultaneously, particularly for alterations in key

n engl j med 379;18 nejm.org November 1, 2018 1757


The New England Journal of Medicine
Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Localized Disease Metastatic Disease

Clinically Clinically Minimal Metastatic Treatment Disease Subsequent


undetectable detectable residual disease response progression therapy
disease

Surgical resection Systemic therapy 1 Systemic therapy 2

Resection
location

Clinical Identification
Residual-disease Molecular Response Monitoring of
Early detection of resistance
Application: detection profiling monitoring mechanisms clonal dynamics

Normal germline Baseline clonal alteration Subclonal resistance Subclonal resistance


cfDNA in ctDNA alteration A alteration B
Abundance of ctDNA

driver genes.20,30-32 Similarly, analyses of large to standard-of-care tumor biopsy testing, for now
cfDNA sequencing databases have yielded mo- tumor biopsy remains the standard for initial
lecular landscapes that closely match mutation pathological diagnosis and molecular testing.
frequencies produced by large-scale tumor tissue Tumor biopsies allow for histologic interpreta-
sequencing compendia, such as the Cancer Ge- tion and the assessment of non–DNA-based altera-
nome Atlas.33 tions, such as in the expression of hormone re-
Although these data suggest that cfDNA test- ceptors or other proteins, which can be important
ing could be a potential surrogate for or adjunct for diagnosis and treatment decisions. Further-

1758 n engl j med 379;18 nejm.org November 1, 2018

The New England Journal of Medicine


Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Cell-free DNA Analysis and Cancer Treatment

Nonetheless, cfDNA testing can play an im-


Figure 3 (facing page). Clinical Applications of cfDNA
Analysis. portant role in initial molecular testing, par-
ticularly for patients in whom standard tumor
Analysis of cfDNA has potential applications at multi­
biopsies yield insufficient material for clinical
ple points throughout the natural course of cancer de­
sequencing, which can occur in as many as 20 to
velopment, diagnosis, and treatment. Early-detection
methods that screen for evidence of nascent tumors in
25% of needle biopsies.30,34 In such circumstances,
cfDNA are currently under development. A liquid-biopsy
cfDNA testing for treatment selection is increas-
test capable of identifying early-stage cancers in asymp­
ingly used as an alternative to repeat invasive
tomatic persons may allow cancers to be identified at
biopsy and may reveal actionable mutations that
a stage when they are more likely to be curable. After
guide treatment decisions in these patients.30,35
surgery with curative intent, cfDNA from postoperative
plasma drawn during the weeks after surgery can be
Technological advances are likely to allow more
analyzed for the persistent presence of mutations or
rapid and inexpensive testing of cfDNA for rela-
other alterations known to exist in the patient’s resected
tively rare abnormalities that could identify ac-
tumor. Since the half-life of cfDNA is very short (an hour
tionable mutations and predict the likelihood of
or less), any evidence of persistent tumor-derived muta­
response (e.g., the presence of microsatellite in-
tions in cfDNA from postoperative plasma can provide
direct evidence of residual disease that may ultimately
stability as a predictor of response to T-cell check-
lead to tumor relapse. Detection of residual disease in
point inhibition).
cfDNA shortly after surgery may allow patients to be
For repeat or serial testing after one or more
stratified according to risk of recurrence and may offer
lines of therapy, minimally invasive cfDNA analy-
an opportunity for early intervention to salvage cure. In
sis offers many apparent advantages over repeat
the context of metastatic disease, clinical sequencing
of cfDNA can identify potentially targetable genetic al­
invasive tumor biopsies, which are less practical,
terations to select precision therapies. Sequencing of
less safe, and less cost-effective. In particular,
cfDNA can identify many of the same target alterations
liquid biopsy may identify emergent genetic al-
identified by tumor sequencing, which can be particu­
terations driving acquired resistance to therapy
larly useful when insufficient tumor material is available
that can be targetable with newer-generation
for clinical sequencing. Studies have suggested that
ctDNA levels closely parallel overall tumor burden and
therapies.22,36-43 One of the earliest and best
can be used as an accurate means of monitoring treat­
examples is the use of cfDNA testing to identify
ment response and the development of resistance. On
the emergence of the epidermal growth factor
disease progression, cfDNA analysis has proved effec­
receptor (EGFR) T790M gatekeeper mutation
tive in identifying emergent genetic alterations that drive
after EGFR inhibitor therapy in EGFR-mutated
therapeutic resistance, which can guide subsequent
therapy choice. Owing to the potential for extensive tu­
non–small-cell lung cancer. Key studies have
mor heterogeneity in the context of acquired resistance,
shown a high degree of concordance of cfDNA
cfDNA analysis may identify multiple concurrent resis­
analysis with tissue testing in identifying the
tance alterations residing in distinct tumor metastases
presence of the T790M mutation, which responds
that would not be captured by a single tumor biopsy.
to third-generation EGFR inhibitors, such as
Analysis of cfDNA has been used to monitor the clonal
dynamics of multiple tumor subclones during subse­
osimertinib.35,44 Indeed, the cobas EGFR Muta-
quent therapy, an approach that can provide a molecu­
tion Test can identify T790M and other EGFR
lar basis for mixed clinical responses to therapy.
driver mutations in plasma and has been ap-
proved by the Food and Drug Administration as
more, large series have shown that approxi- a companion diagnostic for choosing specific
mately 15% of patients with metastatic can- EGFR therapies.45
cer may not have sufficient ctDNA levels to allow
for mutational profiling from plasma, and these Tracking of Therapeutic Response
numbers vary depending on tumor type and tu- Although ctDNA levels can vary greatly among
mor burden.13,31,33 Although improving the tim- patients, ctDNA levels in an individual patient
ing of blood draws for cfDNA analysis (e.g., over time correlate well with changes in tumor
before the initiation of therapy) may increase the burden and treatment response. The short half-
yield of cfDNA testing, confidence in the pres- life of cfDNA in circulation (approximately 1 hour)
ence or absence of key alterations decreases as can be advantageous for measuring real-time
the ctDNA fraction approaches the limit of de- tumor burden in response to therapy, in contrast
tection of current technologies. This factor must to many standard serum tumor markers in cur-
be taken into account when interpreting clinical rent clinical use (e.g., carcinoembryonic antigen
cfDNA tests. [CEA] and cancer antigen 125 [CA-125]), which

n engl j med 379;18 nejm.org  November 1, 2018 1759


The New England Journal of Medicine
Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

have half-lives of days to weeks.38 Many standard vidual patient but can be detected collectively in
clinical tumor markers also have limited sensi- cfDNA from a single plasma sample.39,40,49 A
tivity and specificity, whereas tumor-specific study involving patients with colorectal cancer
clonal alterations (i.e., those alterations present in after anti-EGFR antibody therapy showed that
the original tumor clone and thus present in all patients may harbor as many as 13 different re-
tumor cells) can be monitored in plasma with sistance alterations, as detected in cfDNA, with
high sensitivity and are unique to a patient’s fewer than 10% of patients having only a single
tumor. Some studies have suggested that ctDNA resistance alteration.33 Studies comparing matched
levels may actually increase transiently after the tumor biopsies and cfDNA tests at the time of
initiation of therapy, as tumor-cell death leads to disease progression have suggested that cfDNA
increased release of ctDNA.6 However, within 1 to testing may reveal additional alterations not
2 weeks after the initiation of therapy, ctDNA identified by a single tumor biopsy in as many
levels drop dramatically in patients who have a as two thirds of cases.50 Thus, targeting a sin-
response to therapy. gle resistance mechanism on the basis of the
Indeed, some studies have suggested that results of a single tumor biopsy may lead to
changes in ctDNA can outperform standard mixed clinical responses as a result of the out-
tumor markers in the prediction of treatment growth of distinct resistant subclones not ob-
response.38,42 Moreover, a rise in ctDNA levels tained in the biopsy specimen,39 and cfDNA test-
may precede radiographic progression by weeks ing may help guide treatment decisions.
to months.36-38,46 Thus, as technologies for cfDNA In addition to emerging as an important dis-
monitoring become increasingly available and covery tool, cfDNA analysis can be applied clini-
cost-effective, their potential to detect early evi- cally for the management of therapeutic resis-
dence of response or progression may become tance. The use of cfDNA to identify the presence
important for clinical management. of the T790M resistance mutation in EGFR-mutated
lung cancer was discussed above. Other studies
Monitoring of Resistance and Tumor have shown that cfDNA analysis can identify the
Heterogeneity coexistence of T790M with other resistance altera-
The clinical benefit of precision cancer therapies tions, such as MET amplification, and that pa-
is limited by the eventual emergence of acquired tients with such coexisting alterations may de-
resistance.26 In general, acquired resistance arises rive less benefit from subsequent therapy with
from one or more tumor subclones that harbor third-generation EGFR inhibitors, such as osimer­
preexisting resistance alterations, which emerge tinib.44 Similarly, detection of one or more ESR1
under the selective pressure of therapy. Analysis mutations in the cfDNA of patients with estrogen-
of cfDNA has become an effective tool for the receptor–positive breast cancer predicted poorer
early detection and identification of molecular outcome of subsequent hormonal therapy.51 Other
alterations leading to clinical resistance to ther- studies show that resistance to various targeted
apy. A key advantage of cfDNA is the ability to therapies can be observed in cfDNA before the
capture molecular heterogeneity associated with clinical detection of progression by standard
resistance (Fig. 4). Acquired resistance is often imaging or tumor markers.36,37,40
characterized by the clonal outgrowth of mul- Analysis of cfDNA has also been used to
tiple resistant subclones in an individual pa- track the clonal dynamics of distinct resistant
tient.36,39,40,43,47,48 These resistant subclones may subclones during sequential therapy and even
coexist in the same lesion or in distinct meta- after the discontinuation of therapy. Siravegna
static sites. Thus, a single-lesion tumor biopsy et al. found that the RAS mutations emerging in
may dramatically underestimate the molecular patients with colorectal cancer during EGFR
heterogeneity present, whereas the presence of blockade can fall to undetectable levels in cfDNA
multiple resistance mechanisms can often be after withdrawal of EGFR-directed therapy, and
captured by cfDNA, which is shed from tumor many of these patients may then benefit from
cells throughout the body. reinstitution of anti-EGFR therapy.46 Integration
Indeed, studies incorporating multiple tumor of real-time cfDNA analysis into clinical trials
biopsies or autopsy specimens have shown that and eventually into standard clinical manage-
multiple unique resistance alterations frequently ment has the potential to become a valuable tool
coexist in different metastatic sites in an indi- for precision medicine.

1760 n engl j med 379;18 nejm.org  November 1, 2018

The New England Journal of Medicine


Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Cell-free DNA Analysis and Cancer Treatment

First-line therapy Second-line therapy

Tumor
biopsy

Clonal target Resistance alteration A Resistance alteration C


alteration Resistance alteration B Resistance alteration D
Clonal abundance
Abundance of ctDNA

Figure 4. Tumor Heterogeneity and cfDNA.


Acquired resistance to therapy is often thought to arise from rare tumor subclones that harbor preexisting resistance
alterations. Although all cells may harbor the original clonal target alteration, preexisting subclonal alterations that
provide a fitness advantage under the selective pressure of therapy may exist in some cells. As therapy is initiated,
it may exert a cytotoxic effect on most tumor cells, but an outgrowth of resistant subpopulations may occur, leading
to dynamic shifts in clonal abundance and eventual disease progression. Because preexisting resistant subclones
may reside in different metastatic lesions or in different subpopulations within a single lesion, the emergence of re­
sistance can be characterized by extensive molecular heterogeneity. Thus, a single tumor biopsy specimen that is
obtained during disease progression may reveal only a subset (i.e., green only) of the resistant clones present, and
subsequent therapies that are directed against this resistance mechanism only may lead to mixed clinical response
and treatment failure due to outgrowth of other coexistent clones. Analysis of cfDNA has the potential to identify
multiple concurrent mechanisms of resistance and can monitor clonal dynamics during therapy. Clonal alterations
(present in the original tumor clone and thus in all cells) are represented in gray, and resistant subclonal alterations
in other colors.

Detection of Postsurgical Residual Disease resection. If any tumor cells remain postsurgi-
One of the most transformative potential appli- cally, they can lead to eventual tumor relapse. In
cations of cfDNA analysis may be its ability to high-risk patients, adjuvant chemotherapy can
detect the presence of tumor in patients with no reduce the risk of relapse. However, it is cur-
clinically evident disease — for example, as a rently not possible to determine which patients
screening tool in the early detection of new can- harbor residual disease immediately after sur-
cers or for detection of relapse after surgery or gery and which patients have been cured of their
adjuvant therapy. In general, the primary means disease. In particular contexts, such as patients
by which solid tumors can be cured is surgical with stage II colorectal cancer who have low

n engl j med 379;18 nejm.org November 1, 2018 1761


The New England Journal of Medicine
Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

clinical risk and no evidence of nodal or distant Figure 5 (facing page). Use of cfDNA for the Detection
metastases, adjuvant therapy is not routinely of Residual Disease and Management of Postoperative
given, although approximately 15% of these pa- Therapy.
tients may eventually have a recurrence. An ef- After surgery with curative intent, there is currently no
fective method for the detection of postoperative way to determine which patients are cured of their dis­
residual disease could spare patients who have ease and which have subclinical residual disease that
may ultimately lead to recurrence. Thus, decisions about
been cured the need to undergo potentially toxic adjuvant chemotherapy are based on clinical-risk strati­
adjuvant chemotherapy or could identify those fication. In the example of colon cancer, patients with
patients who have residual disease and might stage II disease who are at low clinical risk undergo
benefit from adjuvant therapy (Fig. 5). ­observation alone and do not receive adjuvant chemo­
In a landmark study, Diehl et al. found that therapy, although approximately 10 to 15% will eventu­
ally have recurrent disease. Conversely, patients with
cfDNA analysis just a few weeks after surgery for stage III disease are treated with adjuvant chemother­
colorectal cancer could accurately predict those apy, even though more than half are cured by surgery
patients who had residual disease and would alone. Current efforts are focused on using postoper­
eventually relapse.12 First, tumor-specific muta- ative cfDNA to identify patients with residual disease,
tions were identified in each patient by standard which may allow more precise decisions about adju­
vant treatment. Briefly, tumor-specific mutations or
sequencing of resected tumor. Next, highly sen- other alterations are identified in the resected tumor
sitive methods (BEAMing), capable of detecting specimen of each patient, and plasma specimens ob­
mutant-allele frequencies as low as 0.01%, were tained a few weeks after surgery are assessed for the
used to determine whether one or more of these persistent presence of these alterations as direct evi­
tumor-specific mutations persisted in that patient’s dence of residual tumor. With further improvement of
this approach, it may be possible in future studies to
plasma when collected approximately 4 weeks assign patients who would not have received adjuvant
after surgery. Detection of even trace levels of therapy as the standard of care (i.e., with stage II dis­
these mutations would provide direct evidence ease) to receive adjuvant therapy if evidence of residual
of residual tumor cells. In a follow-up study in- disease is detected in cfDNA. Similarly, as the sensitiv­
volving 178 patients with stage II colon cancer ity and reliability of these tests improve, it may even be
possible to assign patients who would receive adjuvant
who did not receive adjuvant chemotherapy, the therapy as the standard of care (i.e., those with stage
detection of residual tumor-specific mutations III disease) to a low-risk group on the basis of cfDNA
in postoperative plasma was associated with a testing; active surveillance could be considered in lieu
risk of tumor recurrence that was 18 times as of adjuvant therapy, thus sparing many patients who are
high as that among patients with undetectable cured by surgery alone the toxic effects of chemother­
apy. Studies are currently ongoing (e.g., the DYNAMIC
ctDNA (P<0.001). In addition, ctDNA markedly trial [Australian New Zealand Clinical Trials Registry
outperformed clinical risk factors and a standard number, ACTRN12615000381583]).
tumor marker (CEA) for predicting relapse.52
Similarly, several key studies have shown that
the postoperative detection of tumor-specific
mutations in cfDNA can predict residual disease would require a highly sensitive method — to
and tumor relapse in breast cancer, lung cancer, detect trace amounts of cfDNA or other material
and pancreatic cancer.53-56 This approach has the released by precancerous lesions or early-stage
potential to become a critical tool in the postop- cancers — and would also require high specific-
erative management of the care of patients with ity to minimize false positive results in the large
cancer and is currently being tested in prospec- unaffected population undergoing screening.
tive clinical trials that will assess the usefulness Additional challenges complicate this undertak-
of residual postoperative ctDNA detection to guide ing. First, since many cancer types share com-
adjuvant chemotherapy (Fig. 5). mon mutations in genes such as KRAS, BRAF, or
TP53, localizing a cancer to a specific organ after
Early Cancer Detection a positive liquid-biopsy test may be difficult.57 In
The holy grail of liquid-biopsy applications is the addition, benign lesions may harbor the same
potential for early cancer detection through a mutations seen in cancer and have the potential
simple blood test in otherwise healthy, asymptom- to shed cfDNA into the circulation. Indeed,
atic persons. At present, no mature technology benign nevi may harbor the same BRAF muta-
exists to achieve that goal, but such an approach tions observed in advanced cancers.58 Mutations

1762 n engl j med 379;18 nejm.org  November 1, 2018

The New England Journal of Medicine


Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Cell-free DNA Analysis and Cancer Treatment

Current standard of care Potential cfDNA-guided postoperative management

Stage II disease and low clinical risk


No adjuvant
chemotherapy

Stage II disease and low clinical risk Active


(~10–15% with residual disease) surveillance
No adjuvant
chemotherapy Serial cfDNA
for all monitoring

~10–15% have
recurrence and Normal germline cfDNA
may have
Tumor-specific mutation in ctDNA
benefited from
chemotherapy No evidence of residual disease in post-
operative cfDNA
Evidence of residual disease in post-
Cure by surgery alone operative cfDNA

Residual disease after surgery


Adjuvant
chemotherapy

Sequence resected
tumor, identify
clonal mutations
Detection of tumor-specific
mutations in postoperative plasma

Stage III disease No adjuvant


Adjuvant chemotherapy
chemotherapy
for all Stage III disease
Active
(~45% with residual disease)
>50% cured surveillance
by surgery
alone; Serial cfDNA
chemotherapy monitoring
unnecessary

~30% have
recurrence
despite
chemotherapy
and ~15%
cured by
chemotherapy
Adjuvant
chemotherapy

n engl j med 379;18 nejm.org November 1, 2018 1763


The New England Journal of Medicine
Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

detectable in cfDNA can also originate from 1005 patients with nonmetastatic, clinically de-
aberrant and often benign clonal populations in tectable tumors across eight common tumor
the bone marrow, through a process referred to types, the test was positive in a median of 70%
as clonal hematopoiesis of indeterminate poten- of patients, with sensitivities ranging from 69 to
tial (CHIP). The frequency of CHIP increases 98%. Overall specificity was more than 99%,
exponentially with age, with a rate of more than with only 7 of 812 healthy controls testing posi-
10% among persons older than 70 years of age.59 tive. Moreover, using supervised machine learn-
CHIP is thus a common source of diverse muta- ing, the investigators used the profile of detected
tions detectable in cfDNA, posing a major chal- mutations and proteins to localize the cancer to
lenge for this approach. For this reason, methods its tissue of origin in 83% of cases.
other than simple mutation detection in cfDNA Though these results are encouraging, evalu-
— including tumor-associated viral sequences, ation in a more representative screening popula-
such as in tumors associated with Epstein–Barr tion of asymptomatic patients will be critical.
virus (EBV) infection and human papillomavirus Although further improvement of liquid-biopsy
infection, and DNA methylation changes — are screening approaches and validation in prospec-
being explored for early detection.7,60,61 tive clinical trials are needed, cfDNA analysis as
Two recent studies have illustrated the poten- an early-detection tool offers a potentially trans-
tial of liquid biopsy for the early detection of formative advance for cancer medicine.
cancer. Chan et al. used detection of EBV DNA in
plasma to screen for nasopharyngeal carcinoma Sum m a r y a nd F u t ur e Dir ec t ions
in 20,174 Chinese patients.60 A total of 309 pa-
tients (1.5%) had detectable EBV DNA in plasma, Analysis of cfDNA has rapidly emerged as a tech-
as confirmed on two sequential tests, and 34 of nology with many promising clinical applications
these patients (0.17% of the original population) in oncology. Effective clinical integration of cfDNA
had nasopharyngeal carcinoma on endoscopic analysis will require a careful understanding of
evaluation. Conversely, only 1 of the EBV DNA– the advantages and limitations of this approach
negative patients presented with nasopharyngeal for proper interpretation of results to guide clini-
carcinoma within a year after screening. Overall, cal decision making. Although further prospec-
the sensitivity and specificity of this approach tive study is needed, cfDNA analysis harbors the
An audio were 97.1% and 98.6%, respectively. Recently, potential to have a transformative effect on can-
interview with
Dr. Corcoran
Cohen et al. developed a screening method inte- cer medicine.
is available grating mutation detection in cfDNA with circu- Disclosure forms provided by the authors are available with
at NEJM.org lating protein markers, termed CancerSEEK.57 In the full text of this article at NEJM.org.

References
1. Haber DA, Velculescu VE. Blood-based 7. Wang Y, Springer S, Mulvey CL, et al. mor dynamics. Nat Med 2008;​ 14:​
985-
analyses of cancer: circulating tumor Detection of somatic mutations and HPV 90.
cells and circulating tumor DNA. Cancer in the saliva and plasma of patients with 13. Bettegowda C, Sausen M, Leary RJ,
Discov 2014;​4:​650-61. head and neck squamous cell carcinomas. et al. Detection of circulating tumor DNA
2. Siravegna G, Marsoni S, Siena S, Sci Transl Med 2015;​7:​293ra104. in early- and late-stage human malignan-
Bardelli A. Integrating liquid biopsies into 8. De Mattos-Arruda L, Mayor R, Ng CK, cies. Sci Transl Med 2014;​6:​224ra24.
the management of cancer. Nat Rev Clin et al. Cerebrospinal fluid-derived circulat- 14. El Messaoudi S, Rolet F, Mouliere F,
Oncol 2017;​14:​531-48. ing tumour DNA better represents the Thierry AR. Circulating cell free DNA:
3. Alix-Panabières C, Pantel K. Circulat- genomic alterations of brain tumours than preanalytical considerations. Clin Chim
ing tumor cells: liquid biopsy of cancer. plasma. Nat Commun 2015;​6:​8839. Acta 2013;​424:​222-30.
Clin Chem 2013;​59:​110-8. 9. Mandel P, Metais P. Les acides nu­ 15. Swinkels DW, Wiegerinck E, Steegers
4. Ruivo CF, Adem B, Silva M, Melo SA. cléiques du plasma sanguin chez l’homme. EA, de Kok JB. Effects of blood-process-
The biology of cancer exosomes: insights C R Seances Soc Biol Fil 1948;​142:​241-3. ing protocols on cell-free DNA quantifica-
and new perspectives. Cancer Res 2017;​ 10. Fan HC, Blumenfeld YJ, Chitkara U, tion in plasma. Clin Chem 2003;​49:​525-6.
77:​6480-8. Hudgins L, Quake SR. Analysis of the size 16. Alidousty C, Brandes D, Heydt C, et al.
5. Schwarzenbach H, Nishida N, Calin distributions of fetal and maternal cell- Comparison of blood collection tubes from
GA, Pantel K. Clinical relevance of circu- free DNA by paired-end sequencing. Clin three different manufacturers for the col-
lating cell-free microRNAs in cancer. Nat Chem 2010;​56:​1279-86. lection of cell-free DNA for liquid biopsy
Rev Clin Oncol 2014;​11:​145-56. 11. Leon SA, Shapiro B, Sklaroff DM, mutation testing. J Mol Diagn 2017;​19:​
6. Husain H, Melnikova VO, Kosco K, Yaros MJ. Free DNA in the serum of can- 801-4.
et al. Monitoring daily dynamics of early cer patients and the effect of therapy. 17. Diehl F, Li M, He Y, Kinzler KW, Vogel-
tumor response to targeted therapy by de- Cancer Res 1977;​37:​646-50. stein B, Dressman D. BEAMing: single-
tecting circulating tumor DNA in urine. 12. Diehl F, Schmidt K, Choti MA, et al. molecule PCR on microparticles in water-in-
Clin Cancer Res 2017;​23:​4716-23. Circulating mutant DNA to assess tu- oil emulsions. Nat Methods 2006;​3:​551-9.

1764 n engl j med 379;18 nejm.org  November 1, 2018

The New England Journal of Medicine


Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Cell-free DNA Analysis and Cancer Treatment

18. Hindson BJ, Ness KD, Masquelier DA, in patients with colorectal cancer. Cancer 48. Piotrowska Z, Niederst MJ, Karlovich
et al. High-throughput droplet digital PCR Discov 2018;​8:​164-73. CA, et al. Heterogeneity underlies the
system for absolute quantitation of DNA 34. Meric-Bernstam F, Brusco L, Shaw K, emergence of EGFRT790 wild-type clones
copy number. Anal Chem 2011;​83:​8604-10. et al. Feasibility of large-scale genomic following treatment of T790M-positive
19. Forshew T, Murtaza M, Parkinson C, testing to facilitate enrollment onto ge- cancers with a third-generation EGFR in-
et al. Noninvasive identification and moni- nomically matched clinical trials. J Clin hibitor. Cancer Discov 2015;​5:​713-22.
toring of cancer mutations by targeted deep Oncol 2015;​33:​2753-62. 49. Blakely CM, Watkins TBK, Wu W, et al.
sequencing of plasma DNA. Sci Transl Med 35. Oxnard GR, Thress KS, Alden RS, et al. Evolution and clinical impact of co-occur-
2012;​4:​136ra68. Association between plasma genotyping ring genetic alterations in advanced-stage
20. Adalsteinsson VA, Ha G, Freeman SS, and outcomes of treatment with osimer- EGFR-mutant lung cancers. Nat Genet
et al. Scalable whole-exome sequencing of tinib (AZD9291) in advanced non-small- 2017;​49:​1693-704.
cell-free DNA reveals high concordance cell lung cancer. J Clin Oncol 2016;​ 34:​ 50. Parikh A, Goyal L, Hazar-Rethinam M,
with metastatic tumors. Nat Commun 3375-82. et al. Systematic liquid biopsy identifies
2017;​8:​1324. 36. Diaz LA Jr, Williams RT, Wu J, et al. novel and heterogeneous mechanisms of
21. Leary RJ, Kinde I, Diehl F, et al. Devel- The molecular evolution of acquired resis- acquired resistance in gastrointestinal (GI)
opment of personalized tumor biomarkers tance to targeted EGFR blockade in colo­ cancer patients. Ann Oncol 2017;​28:​Suppl 3:​
using massively parallel sequencing. Sci rectal cancers. Nature 2012;​486:​537-40. mdx262. abstract.
Transl Med 2010;​2:​20ra14. 37. Misale S, Yaeger R, Hobor S, et al. 51. Chandarlapaty S, Chen D, He W, et al.
22. Murtaza M, Dawson SJ, Tsui DW, et al. Emergence of KRAS mutations and ac- Prevalence of ESR1 mutations in cell-free
Non-invasive analysis of acquired resis- quired resistance to anti-EGFR therapy in DNA and outcomes in metastatic breast
tance to cancer therapy by sequencing of colorectal cancer. Nature 2012;​486:​532-6. cancer: a secondary analysis of the
plasma DNA. Nature 2013;​497:​108-12. 38. Dawson SJ, Tsui DW, Murtaza M, ­BOLERO-2 clinical trial. JAMA Oncol 2016;​
23. Kinde I, Wu J, Papadopoulos N, Kinzler et al. Analysis of circulating tumor DNA 2:​1310-5.
KW, Vogelstein B. Detection and quantifi- to monitor metastatic breast cancer. N Engl 52. Tie J, Wang Y, Tomasetti C, et al. Cir-
cation of rare mutations with massively J Med 2013;​368:​1199-209. culating tumor DNA analysis detects
parallel sequencing. Proc Natl Acad Sci 39. Russo M, Siravegna G, Blaszkowsky minimal residual disease and predicts re-
U S A 2011;​108:​9530-5. LS, et al. Tumor heterogeneity and lesion- currence in patients with stage II colon
24. Newman AM, Bratman SV, To J, et al. specific response to targeted therapy in cancer. Sci Transl Med 2016;​8:​346ra92.
An ultrasensitive method for quantitating colorectal cancer. Cancer Discov 2016;​6:​ 53. Garcia-Murillas I, Schiavon G, Weigelt
circulating tumor DNA with broad patient 147-53. B, et al. Mutation tracking in circulating
coverage. Nat Med 2014;​20:​548-54. 40. Goyal L, Saha SK, Liu LY, et al. Poly- tumor DNA predicts relapse in early breast
25. Newman AM, Lovejoy AF, Klass DM, clonal secondary FGFR2 mutations drive cancer. Sci Transl Med 2015;​7:​302ra133.
et al. Integrated digital error suppression acquired resistance to FGFR inhibition in 54. Chaudhuri AA, Chabon JJ, Lovejoy AF,
for improved detection of circulating tu- patients with FGFR2 fusion-positive chol- et al. Early detection of molecular residual
mor DNA. Nat Biotechnol 2016;​34:​547-55. angiocarcinoma. Cancer Discov 2017;​ 7:​ disease in localized lung cancer by circu-
26. Garraway LA, Jänne PA. Circumvent- 252-63. lating tumor DNA profiling. Cancer Dis-
ing cancer drug resistance in the era of 41. Schiavon G, Hrebien S, Garcia-Muril- cov 2017;​7:​1394-403.
personalized medicine. Cancer Discov las I, et al. Analysis of ESR1 mutation in 55. Sausen M, Phallen J, Adleff V, et al.
2012;​2:​214-26. circulating tumor DNA demonstrates evo- Clinical implications of genomic altera-
27. Kuderer NM, Burton KA, Blau S, et al. lution during therapy for metastatic breast tions in the tumour and circulation of
Comparison of 2 commercially available cancer. Sci Transl Med 2015;​7:​313ra182. pancreatic cancer patients. Nat Commun
next-generation sequencing platforms in 42. Corcoran RB, André T, Atreya CE, 2015;​6:​7686.
oncology. JAMA Oncol 2017;​3:​996-8. et al. Combined BRAF, EGFR, and MEK in- 56. Beaver JA, Jelovac D, Balukrishna S,
28. Chae YK, Davis AA, Carneiro BA, et al. hibition in patients with BRAFV600E-mutant et al. Detection of cancer DNA in plasma
Concordance between genomic alterations colorectal cancer. Cancer Discov 2018;​8:​ of patients with early-stage breast cancer.
assessed by next-generation sequencing in 428-43. Clin Cancer Res 2014;​20:​2643-50.
tumor tissue or circulating cell-free DNA. 43. Hazar-Rethinam M, Kleyman M, Han 57. Cohen JD, Li L, Wang Y, et al. Detec-
Oncotarget 2016;​7:​65364-73. GC, et al. Convergent therapeutic strate- tion and localization of surgically resect-
29. Torga G, Pienta KJ. Patient-paired sam- gies to overcome the heterogeneity of ac- able cancers with a multi-analyte blood
ple congruence between 2 commercial quired resistance in BRAFV600E colorec- test. Science 2018;​359:​926-30.
liquid biopsy tests. JAMA Oncol 2017 De- tal cancer. Cancer Discov 2018;​8:​417-27. 58. Shain AH, Yeh I, Kovalyshyn I, et al.
cember 14 (Epub ahead of print). 44. Chabon JJ, Simmons AD, Lovejoy AF, The genetic evolution of melanoma from
30. Zill OA, Greene C, Sebisanovic D, et al. Circulating tumour DNA profiling precursor lesions. N Engl J Med 2015;​373:​
et al. Cell-free DNA next-generation se- reveals heterogeneity of EGFR inhibitor 1926-36.
quencing in pancreatobiliary carcinomas. resistance mechanisms in lung cancer pa- 59. Jaiswal S, Fontanillas P, Flannick J,
Cancer Discov 2015;​5:​1040-8. tients. Nat Commun 2016;​7:​11815. et al. Age-related clonal hematopoiesis
31. Schrock AB, Pavlick DC, Klempner SJ, 45. Malapelle U, Sirera R, Jantus-Lewintre associated with adverse outcomes. N Engl
et al. Hybrid capture-based genomic pro- E, et al. Profile of the Roche cobas EGFR J Med 2014;​371:​2488-98.
filing of circulating tumor DNA from pa- mutation test v2 for non-small cell lung 60. Chan KCA, Woo JKS, King A, et al.
tients with advanced cancers of the gastro- cancer. Expert Rev Mol Diagn 2017;​ 17:​ Analysis of plasma Epstein–Barr virus
intestinal tract or anus. Clin Cancer Res 209-15. DNA to screen for nasopharyngeal cancer.
2018;​24:​1881-90. 46. Siravegna G, Mussolin B, Buscarino M, N Engl J Med 2017;​377:​513-22.
32. Lanman RB, Mortimer SA, Zill OA, et al. Clonal evolution and resistance to 61. Chan KC, Jiang P, Chan CW, et al.
et al. Analytical and clinical validation of EGFR blockade in the blood of colorectal Noninvasive detection of cancer-associated
a digital sequencing panel for quantita- cancer patients. Nat Med 2015;​21:​795-801. genome-wide hypomethylation and copy
tive, highly accurate evaluation of cell-free 47. Gerlinger M, Rowan AJ, Horswell S, number aberrations by plasma DNA bisul-
circulating tumor DNA. PLoS One 2015;​ et al. Intratumor heterogeneity and fite sequencing. Proc Natl Acad Sci U S A
10(10):​e0140712. branched evolution revealed by multi­ 2013;​110:​18761-8.
33. Strickler JH, Loree JM, Ahronian LG, region sequencing. N Engl J Med 2012;​ Copyright © 2018 Massachusetts Medical Society.
et al. Genomic landscape of cell-free DNA 366:​883-92.

n engl j med 379;18 nejm.org  November 1, 2018 1765


The New England Journal of Medicine
Downloaded from nejm.org at GOTEBORGS UNIVERSITETSBIBL. on November 29, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.

You might also like