You are on page 1of 14

SPECIAL REPORT

SC

Circulating Tumor DNA for Early Cancer


Detection

Clare Fiala,1 Vathany Kulasingam,2,3 and Eleftherios P. Diamandis1,2,3*

Background: Cancer cells release circulating tumor DNA (ctDNA) into the bloodstream, which can now
be quantified and examined using novel high-throughput sequencing technologies. This has led to the
emergence of the “liquid biopsy,” which proposes to analyze this genetic material and extract informa-
tion on a patient's cancer using a simple blood draw.
Content: ctDNA has been detected in many advanced cancers. It has also been proven to be a highly
sensitive indicator of relapse and prognosis. Sequencing the genetic material has also led to the
discovery of mutations targetable by existing therapies. Although ctDNA screening is more expensive,
it is showing promise against circulating tumor cells and traditional cancer biomarkers. ctDNA has also
been detected in other bodily fluids, including cerebrospinal fluid, urine, saliva, and stool.
The utility of ctDNA for early cancer detection is being studied. However, a blood test for cancer faces
heavy obstacles, such as extremely low ctDNA concentrations in early-stage disease and benign mu-
tations caused by clonal hematopoiesis, causing both sensitivity and specificity concerns. Nonetheless,
companies and academic laboratories are highly active in developing such a test.
Conclusion: Currently, ctDNA is unlikely to perform at the high level of sensitivity and specificity
required for early diagnosis and population screening. However, ctDNA in blood and other fluids has
important clinical applications for cancer monitoring, prognosis, and selection of therapy that require
further investigation.

IMPACT STATEMENT
Circulating tumor DNA (ctDNA) testing is poised to usher in a new era of precision, individualized
cancer treatment. Many promising results have been reported in monitoring for disease relapse,
mutation profiling, and treatment response. Nevertheless, problems with sensitivity, specificity,
and clonal hematopoiesis have plagued the development of ctDNA tests to test asymptomatic
patients for cancer.

1
Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada; 2Department of Laboratory Medicine and Patho-
biology, University of Toronto, Toronto, Ontario, Canada; 3Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada.
*Address correspondence to this author at: Mount Sinai Hospital and University Health Network, 60 Murray St., Box 32, Floor 6, Rm. L6-201,
Toronto, ON, MST 3L9, Canada. Fax 416-586-8628; e-mail Eleftherios.diamandis@sinaihealthsystem.ca.
DOI: 10.1373/jalm.2018.026393
© 2018 American Association for Clinical Chemistry
4
Nonstandard abbreviations: ctDNA, circulating tumor DNA; cfDNA, cell-free DNA; NSCLC, non–small cell lung cancer; CSF, cerebrospinal fluid;
CTC, circulating tumor cell; SCLC, small-cell lung cancer.
5
Human genes: PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; TP53, tumor protein p53; KRAS, KRAS proto-
oncogene, GTPase; APC, APC, WNT signaling pathway regulator; DNMT3A, DNA methyltransferase 3 alpha; EGFR, epidermal growth factor receptor;

...............................................................................................................

300 JALM | 300 –313 | 03:02 | September 2018


Circulating Tumor DNA for Early Cancer Detection SPECIAL REPORT

In the current cancer diagnostic pathway, a tu- metastases) release DNA into the circulation, known
mor is detected by imaging, prompted by a screen- as ctDNA (4, 5). The mechanisms of this release are
ing program, clinical presentation, or sometimes still uncertain; however, tumor cells are thought to
just chance. Next, the patient usually undergoes a secrete their DNA during necrosis or apoptosis (6).
tissue biopsy. The sample is analyzed by a pathol- ctDNA is extremely fragmented and disordered,
ogist to determine whether it is cancerous. If it is often into a laddering pattern of 170 to 180 base
malignant and accessible, the tumor is usually re- pairs (7). Tissue-damaging therapies to treat can-
sected surgically. The pathology analysis, com- cer, such as surgery, radiation, and chemotherapy,
bined with targeted DNA sequencing, is used to further disfigure the ctDNA (8). Renal dysfunction
determine the need for, and type of, adjuvant can further confound data (9), as ctDNA is cleared
chemotherapy. through the kidneys and liver (10). In the past de-
Although tumor biopsies in some shape or form cade, the advent of sensitive, high-throughput
have been used for the past 100 years and form screening technologies such as beads, emulsion,
the basis of modern cancer treatment, they are by amplification, magnetics, safe sequencing system,
no means a perfect tool. Even with more sophisti- tagged-amplicon deep sequencing, and digital
droplet PCR means it is possible to isolate, quan-
cated technology, such as needle biopsies, to re-
tify, and sequence this DNA, bringing ctDNA appli-
move tiny amounts of tissue, these procedures are
cations even closer to the clinic.
still uncomfortable and invasive. Tumors are ex-
tremely heterogeneous, so sequencing only small
CLINICAL APPLICATIONS OF ctDNA
lesions, as in the current paradigm, will not pro-
LIQUID BIOPSIES
duce a comprehensive picture of all the mutations.
Moreover, some tumors such as brain or lung neo-
ctDNA can be harvested from a simple blood
plasms are not easily accessible, so a biopsy can-
draw. It has a half-life from 15 min to up to 3 h,
not be performed. In these cases, there is no
meaning it must be processed rapidly (11). There is
knowledge of mutations to inform the optimum 2 to 4 times more ctDNA in plasma than serum, so
course of treatment. Finally, while biopsies cannot generally EDTA whole blood is centrifuged im-
be repeated often because of their high morbidity, mediately after collection (12). The ctDNA in the
cancer is continuously mutating and evolving, plasma is then analyzed/sequenced, forming a
meaning treatment can be based on data that are liquid biopsy.
not timely or even relevant. This new biomarker is poised to confer many
Recently, much attention has been focused on advantages over conventional biomarkers (13, 14).
the emergence of a new cancer biomarker: circu- Assuming all regions of the tumor secrete ctDNA at
lating tumor DNA (ctDNA)4. Since 1946, it has been the same rate, sequencing the genetic material will
known that normal cells naturally release DNA provide a far more comprehensive overview of all
fragments, resulting in cell-free DNA (cfDNA) circu- the mutations present in the tumor than just se-
lating in the bloodstream (1). In 1994, it was discov- quencing small lesions. This information can then
ered that some cfDNA had mutations that be used to inform more targeted treatment. Be-
originated from neoplasms (2, 3). Tumor cells (from cause liquid biopsies involve only drawing blood,
primary tumors, micrometastases, and overt they can be repeated far more often to provide an

ERBB2, erb-b2 receptor tyrosine kinase 2; FGFR3, fibroblast growth factor receptor 3; SEPT9, septin 9; CDKN2A, cyclin dependent kinase inhibitor 2A;
SMAD4, SMAD family member 4; p53, long intergenic non-protein coding RNA, p53 induced transcript; TET2, tet methylcytosine dioxygenase 2;
PPM1D, protein phosphatase, Mg2+/Mn2+ dependent 1D.

...............................................................................................................

September 2018 | 03:02 | 300 –313 | JALM 301


SPECIAL REPORT Circulating Tumor DNA for Early Cancer Detection

up-to-date overview of the mutations, ensuring the plasma of individuals who relapsed before the
consistent personalized treatment. tumors were visible by computerized tomography,
A groundbreaking study by Bettegowda et al. providing a significant lead time to introduce new
showed that ctDNA is detectable in >75% of ad- treatments. Similar results were observed by Tie et al.,
vanced pancreatic, ovarian, colorectal, bladder, whose team used massively parallel sequencing-
liver, gastroesophageal, breast, melanoma, liver, based assays to sequence plasma samples from
and head and neck cancers (15). Many more inves- 164 postoperative stage II colon cancer patients.
tigations have been performed, and ctDNA has Only 16 (9.8%) patients with negative ctDNA after
shown promise in managing many different can- resection relapsed. In patients treated with adju-
cers. Thus, the literature about the utility of ctDNA vant chemotherapy, the presence of ctDNA after
in monitoring for relapse, treatment progress, and completion was also associated with lower recur-
disease burden is rapidly expanding.
rence-free survival rates (17).

Early detection of relapsing disease


Liquid biopsies
The accurate and early detection of metastases
Another study of colorectal cancer used matched
is crucial to ensure the assessment of response to
tumor samples to create personalized liquid biop-
treatment. Monitoring treatment response is also
imperative, as it enables clinicians to discontinue sies. They used up to 6 assays per individual to
ineffective therapies, thus minimizing side effects detect specific somatic variants in 11 mid- to late-
and unnecessary costs. It also provides an oppor- stage cancer patients. The specific somatic vari-
tunity to introduce novel therapeutics to target the ants were both 100% sensitive and 100% specific
new mutations detected in the tumor. ctDNA is in detecting postsurgical relapse. This approach
showing a distinct advantage over conventional provided 2 to 15 months (average, 10 months) of
imaging methods, as it provides longer lead times lead time on detection of metastatic recurrence
by detecting chemotherapy resistance or residual compared with typical postsurgical screening, pro-
disease earlier. ctDNA screening to identify pa- viding compelling evidence of the ability of ctDNA
tients at high risk of relapse would provide high to closely monitor patients' responses to treat-
translational value in creating personalized adju- ment (18).
vant therapy protocols. A similar investigation was recently performed
with a subcohort of 24 early-stage non–small-cell
Prediction of relapse lung cancer (NSCLC) patients. In summary, Abbosh
et al. sequenced and compared tumor and healthy
There have been extensive investigations about
the utility of ctDNA for predicting relapse in individ- tissue from the primary surgical resections to iden-
uals diagnosed with colorectal cancer. In a 2017 tify the single-nucleotide variants associated with
longitudinal study of 27 colorectal cancer patients, cancer. They then used this a priori knowledge of
the presence of ctDNA immediately after surgical mutations to design personalized ctDNA panels to
resection was strongly correlated with relapse. All check for relapse by scanning each patient's blood
14 of the patients who eventually relapsed had for mutated ctDNA. These liquid biopsies could de-
ctDNA detectable after their resections, whereas tect relapse and therapy resistance in patients an
all those who were ctDNA-negative post surgery average of 70 days before tumors were observable
remained free of cancer (16). Furthermore, ctDNA through computed tomography scans, although in
was detected an average of 9.4 months earlier in 4 cases, the lead time was >6 months (19).

...............................................................................................................

302 JALM | 300 –313 | 03:02 | September 2018


Circulating Tumor DNA for Early Cancer Detection SPECIAL REPORT

Treatment response sample. Significantly, 53% of patients had mutation


profiles targetable by immune checkpoint block-
A 2013 ground-breaking proof-of-concept study
ade or tyrosine kinase inhibitors (32).
of 52 women with metastatic breast cancer could
KRAS and PIK3CA mutations were identified in
correlate ctDNA with treatment response to sys-
the ctDNA of 69% and 17% of patients with colo-
temic therapy. Dawson et al. used targeted se-
rectal cancer, respectively. These mutations are
quencing to detect mutations in PIK3CA5 and TP53.
actionable by the multikinase inhibitor rego-
Although cancer-associated mutations were de-
rafenib, showing that ctDNA can help clinicians se-
tected in only 57% of patients' tumors, these mu-
lect personalized targeted chemotherapies (33).
tations were in turn detected in the plasma of all
More recently, a similar study was performed
but 1 (97%) of this subcohort. ctDNA levels were
including 605 mid- to late-stage cancer patients
further shown to be proportional with tumor size,
with 28 different solid tumor types (61% lung can-
as detected on radiographic imaging, and a signif-
cer). Shu et al. sequenced tumor specimens from
icant correlation between the number of ctDNA
biopsies and then used targeted next-generation
copies in the blood and patient outcomes was ob-
sequencing to sequence matched plasma sam-
served (20).
ples. Somatic mutations were detected in the
Garcillas-Murillas et al. showed the utility of
ctDNA of 87% of patients. The most frequently mu-
ctDNA monitoring in early-stage breast cancer.
tated tumor suppressor genes were TP53 (18.1%),
The group created personalized tumor-specific
APC (3.3%), and DNMT3A (2.5%), and EGFR (11.9%),
digital droplet PCR assays for 55 patients who re-
KRAS (3.7%), and PIK3CA (3.0%) were the most com-
ceived neoadjuvant chemotherapy with curative
monly mutated oncogenes. In total, 71% of pa-
intent and predicted relapse far more sensitively
than conventional clinical imaging methods, pro- tients had at least 1 clinically actionable mutation
viding an average lead time of 7.9 months (21). (either with a drug approved or in clinical trials)
The utility of ctDNA in monitoring disease prog- (34).
nosis, tumor burden, and treatment response has In 2016, therapeutically targetable and driver re-
been shown for several other malignancies, includ- sistance mutations were detected in the ctDNA of
ing melanoma (22, 23), head and neck (24), lym- advanced NSCLC patients using massively parallel
phoma (25), prostate (26), liver (27), gastric (28), digital exome sequencing. Thompson et al. de-
gynecologic (29), kidney (30), and pancreatic can- tected 275 variants (single-nucleotide variants, in-
cers (31). dels, and fusions) in 102 patients without previous
knowledge of primary tumor mutations. Mutations
were detected in 45 genes, with EGFR mutations
Detection of mutations
making up 20% of total variants. The group detected
Although the technology is still in its infancy, 11 KRAS mutations associated with resistance to
ctDNA analysis has shown translational value in EGFR inhibitors while also finding 10 actionable
detecting mutations that can be targeted by exist- ERBB2 mutations shown to be sensitive to targeted
ing treatments. Chaudhuri et al. used ctDNA deep therapies. Furthermore, the variants were cross-
sequencing (CAPP-seq) analysis to create person- referenced against available treatments with auspi-
alized profiles of 54 healthy controls and 40 lung cious results: 70% of patients had a relevant clinical
cancer (stages I–III) patients who received chemo- trial, 55% patients had a potential off-label targeted
therapy with curative intent. In total, 94% of pa- therapy, and 31% of patients had variants targetable
tients who experienced recurrence had detectable by a Food and Drug Administration–approved treat-
ctDNA in the first postchemotherapy blood ment. This study further compared the utility of

...............................................................................................................

September 2018 | 03:02 | 300 –313 | JALM 303


SPECIAL REPORT Circulating Tumor DNA for Early Cancer Detection

Table 1. Potential applications of ctDNA in cancer diagnostics and management.

Potential for
Application Summary clinical utility References
Prognosis • Undetectable ctDNA in the bloodstream Excellent 13–31, 70, 88, 89
after surgery are correlated with improved
prognosis and smaller chances of relapse
• Strength and type of chemotherapy can be
informed by ctDNA analysis showing l
ikelihood of relapse
Monitoring treatment • Increased number of mutations or rising Excellent, maybe in 13–20, 23–29
efficacy and early ctDNA concentration can indicate combination
relapse detection treatment failure, resistance, and relapse with protein markers
Selection of treatment • Knowledge of mutations in the ctDNA Excellent 17, 28, 30–33, 73
informs choice of therapy (personalized
treatments)
• ctDNA avoids tumor heterogeneity issues
by providing overview of all the mutations
in the tumor (assuming all tumor cells
secrete DNA at the same rate)
Tumor size/disease • Larger amount of ctDNA in blood denotes Excellent, especially in 15, 17, 18, 20–29,
burden advanced tumor stage and volume combination with 88, 89
• Blood testing does not carry the risk of imaging
radiation exposure or poor accuracy of
imaging and can be repeated more often
than traditional biopsies
Detection of cancer in • Most studies show poor sensitivity, Under intense 13, 30, 70, 88, 89
asymptomatic especially for early-stage tumors investigation; could be
individuals/population • For small tumors, there is not enough used in combination
screening ctDNA present to allow for an accurate test with protein biomarkers
result
• Threshold appears to be 1-cm-diameter
tumors

sequencing tissue vs plasma samples for detecting The description and clinical utility of ctDNA tech-
variants in a 50-patient subgroup. ctDNA plasma se- nologies for prognosis, detection of relapse, selec-
quencing detected a mean number of 2.8 variants tion of treatment, disease/tumor burden, and
per patient, whereas only 1.5 variants were detected early detection can be found in Table 1.
per patient in tumor DNA screening, showing the ad-
vantage of ctDNA in encompassing tumor heteroge- ctDNA COMPARED WITH BLOOD-BASED
neity (35). PROTEIN MARKERS
Only 1 ctDNA liquid biopsy test has been ap-
proved so far by the Food and Drug Administra- ctDNA sequencing for monitoring disease pro-
tion. The Cobas EGFR Mutation Test v2 (Roche gression is showing advantages over traditional
Molecular Systems) was approved in June 2016 biomarkers. Although conventional biomarker
and is intended to help clinicians decide whether tests are far less expensive and much less labor-
patients may benefit from erlotinib by screening intensive than ctDNA testing, they have many
for exon 19 deletion or exon 21 (L858R) substitu- weaknesses, mainly low sensitivity and specificity.
tion mutations in EGFR (https://www.fda.gov/News Commonly used biomarkers such as cancer anti-
Events/Newsroom/PressAnnouncements/ucm gen 125, carcinoembryonic antigen, and prostate-
504488.htm). Consequently, ctDNA-based diagnos- specific antigen are also found in the serum of
tics are currently rarely used in the clinic. patients without cancer, although usually in lower

...............................................................................................................

304 JALM | 300 –313 | 03:02 | September 2018


Circulating Tumor DNA for Early Cancer Detection SPECIAL REPORT

concentrations (36–38). Furthermore, they are not useful to treat patients with primary tumors of the
elevated in a significant portion of patients with brain or spinal cord.
advanced cancer (39, 40). By its nature, ctDNA ctDNA in urine has also been found to be a use-
could not be found unless there is a tumor pres- ful biomarker in urologic malignancies (45). ctDNA
ent, and levels of ctDNA have been strongly corre- was studied in the urine of a 68-patient subcohort
lated with disease burden. Improvements in with noninvasive bladder transitional cell cancer
sequencing would increase ctDNA testing sensitiv- who had undergone transurethral resection. Uri-
ity and specificity, as well as further decrease the nary cellular FGFR3 mutation was found to have a
cost. From 2009 to 2014, the cost of sequencing 73% sensitivity and 87% specificity in the diagnosis
per genome dropped from $100000 to $5000 (in- of cancer recurrence after resection (46).
cluding labor and reagents) (41). ctDNA has a Pu et al. detected EFGR mutations in saliva and
shorter half-life compared with some traditional plasma samples of 17 patients with lung adenocar-
biomarkers that can stay in the circulation for cinoma, finding a 100% concordance between sa-
weeks, meaning sequencing this material provides liva and tumor samples (47). Saliva is also being
a highly current snapshot of the disease (42). investigated as a potentially valuable biomarker for
head and neck cancers (48).
Gathering ctDNA from stool has also been
ctDNA IN OTHER BIOLOGICAL FLUIDS
shown as a viable alternative to blood testing for
those with colorectal cancer. A comparison be-
ctDNA is not just limited to blood-based applica-
tween a plasma methylated test and a multimarker
tions. It has been detected in other readily avail-
able biological fluids that have the advantage of stool test for SEPT9 in colorectal cancer patients
being closer to the tumor and, thus, are likely to demonstrated that the stool DNA test had signifi-
contain more ctDNA. cantly greater sensitivity than the ctDNA test (87%
Sequencing ctDNA from cerebrospinal fluid vs 60% sensitivity) (49).
(CSF) is promising for managing brain cancer. Al- In the discussion of alternatives to ctDNA, it is
though lumbar punctures are more invasive than integral to note continuing research in circulating
venipunctures, the blood– brain barrier dramati- tumor cells (CTCs). CTCs enter the bloodstream
cally reduces the amount of ctDNA in the blood, from primary tumors to eventually form metasta-
making the detection of brain cancer in the blood ses (50, 51). They have been detected in many can-
infeasible. However, ctDNA CSF biopsies for brain cers (52), including bladder (53), gastric (54),
cancer have far lower morbidity than performing a prostrate (55, 56), ovarian (57), colon (58), breast
traditional needle or open biopsy, which benefits (59), and lung carcinomas (60).
patients. In a 2017 study, ctDNA CSF biopsy could Extremely sensitive methods are required to de-
detect histone H3 mutations in 8 pediatric patients tect these cells, as there is 1 circulating cancer cell
with diffuse midline glioma. The test sensitivity was per 1 billion of normal cells (61). The Food and Drug
87.5%, and specificity (100%) was validated through Administration has approved only 1 test for the
immunohistochemical staining and Sanger sequenc- quantification of CTCs. The CELLSEARCH CTC
ing of available concordant tumor tissue specimens (Menarini-Silicon Biosystems) test is designed for pa-
(43). Another study identified ctDNA in 100% (n = 21) tients with metastatic breast, prostate, or colorectal
of all medulloblastomas, ependymomas, and high- cancer. The test enumerates CTCs of epithelial origin
grade gliomas that were adjacent to the CSF space in whole blood using immunofluorescence (https://
(44). This suggests that cancer DNA in CSF could be www.cellsearchctc.com/).

...............................................................................................................

September 2018 | 03:02 | 300 –313 | JALM 305


SPECIAL REPORT Circulating Tumor DNA for Early Cancer Detection

An investigation of 19 patients with early breast have the opposite problem: low specificity. This
cancer indicated that both CTC counts and ctDNA leads to unnecessary overtreatment and distress.
(EGFR amplification) were predictors of metastatic Consequently, both cancers could benefit from im-
disease (62). In 2013, a study of metastatic colon proved screening technologies (65–67).
cancer found a significant correlation between the Demonstrating that any ctDNA test for early can-
presence of CTCs and the detection of circulating cer detection has nearly 100% specificity and sen-
DNA fragments containing tumor-specific muta- sitivity is fundamental because of the perils
tions such as those in KRAS (63). associated with false-negative and false-positive
The Dawson et al. study also compared quanti- findings (68). Not only would patients and clinicians
ties of CTCs and cancer antigen 15.3 with imaging should be deceptively reassured by a false-
results and found that ctDNA provided the great- negative blood test result but also the patient
est correlation with disease burden, as well as the would likely not be treated until the carcinoma be-
earliest indicator of response to treatment (20). came obvious on imaging or caused physical
ctDNA was often present in patients without symptoms. Moreover, at that time, the tumor
detectable CTCs, indicating that more research would be much larger, with more complex muta-
needs to be conducted to understand the relation- tions and possibly even metastases. False-positive
ship between these biomarkers (15). findings could cause distress and expensive addi-
tional tests to correct the result and could lead to
ctDNA TESTING FOR EARLY DETECTION overtreatment. Finally, even if clinicians receive a
true-positive result for an early tumor, it can be
Early cancer detection is often viewed as a new difficult to determine its location, as the same mu-
frontier in ctDNA technology. The paradigm be- tations can occur across several cancers.
hind using ctDNA to detect cancer is that tumors Despite these challenges, several cancers have
will secrete genetic material into the circulation be- been identified as attractive targets for early diag-
fore they are visible on imaging or produce symp- nosis via ctDNA screening. Pancreatic cancer has
toms. The development of this technology could an extremely high mortality rate, and surgical re-
eliminate the need for frequent imaging, which section is the only curative treatment. However, it
carries the risk of radiation exposure. Detecting is generally detected in its late stages, and most
cancer early means that the tumor is not only patients (80%– 85%) present with inoperable ad-
smaller but also less complex, so it can be effec- vanced or metastatic cancer, causing a dismal 4%
tively managed with a milder therapy (64). It is to 7% 5-year survival rate (69). However, pancreatic
also far less likely to have metastasized. Thus, cancer is extremely slow developing. There is an
the knowledge that early diagnosis leads to in- average 11.7-year period from the acquisition of
creased survival is the backstay of current the initiating mutation to a full tumor and another
screening programs. 6.8 years needed to develop the first metastatic
Routine mammograms and prostate-specific subclone (70). Its mutations have been widely
antigen tests are stalwarts of the efforts to diag- studied; KRAS, CDKN2A, TP53, and SMAD4 are
nose cancer early. However, both methods have known to be commonly altered (71). A liquid biopsy
serious flaws. Mammography can miss small tu- test to detect pancreatic cancer in asymptomatic
mors, and breast cancer propagates rapidly. This individuals could save thousands of lives.
means that a woman who is reassured by a nega- Lung cancers such as small-cell lung cancer
tive result 1 year can return the next to find she has (SCLC) are also good targets. Lung cancer has the
a metastatic tumor. Prostate-specific antigen tests highest mortality rates of all cancers, and there are

...............................................................................................................

306 JALM | 300 –313 | 03:02 | September 2018


Circulating Tumor DNA for Early Cancer Detection SPECIAL REPORT

no good treatment options at late stages. SCLC Murtaza et al. performed an unguided search
patients have almost universal inactivation of TP53, for deleterious mutations in blood samples by se-
a mutation potentially targetable with existing quencing the exome of a small cohort (n = 6) of
therapies. A 2016 study of 51 SCLC patients and patients with advanced ovarian, lung, or breast
123 healthy controls detected TP53 mutations in cancers. In 5 of the cases, exome sequencing re-
the plasma of 59.1% of late-stage patients but in vealed specific results, such as an activating muta-
only 35.7% of early-stage patients (72). tion in PIK3CA after treatment with paclitaxel or a
Slightly more promising results were reported resistance-conferring mutation in EGFR following
by Bettegowda et al. in 2014. The group used digital treatment with gefitinib (75).
droplet PCR and found ctDNA in <50% of patients Even if exome sequencing methods become
with primary brain, renal, prostate, and thyroid can- less prohibitively expensive, fundamental prob-
cers. In patients with localized tumors, the detection lems remain in using ctDNA for early detection.
rate was 50% (breast), 48% (pancreatic), 53% (gastro- Cancer is an extremely heterogeneous disease
esophageal), and 73% (colorectal) (15).
with layers of mutations. However, new discoveries
In NSCLC, ctDNA was detected in 100% of patients
about clonal hematopoiesis have shown that not
with stage II to IV disease and in 50% of patients with
all mutations are cancer-causing or lead to tumors.
stage I cancer, using CAPP-Seq, 1 of the most sensi-
Spontaneous mutations occur in a person's cells
tive sequencing methods available because of its
throughout their lifetime, and most do not have
combined approach of isolating ctDNA, enhanced
deleterious or even noticeable effects. However,
sample preparation, and extremely targeted deep
mutations during gestation can cause develop-
sequencing (73). This approach is economically via-
mental disabilities, and accumulating somatic mu-
ble, with a cost around $500 to process each sample,
tations can progressively cause aging and cancer.
including materials and labor (74).
The rate of mutation is positively correlated with
age (76). In a study of 26136 cancer-free individu-
CHALLENGES OF USING ctDNA FOR
als, the frequency of somatic mutations increased
EARLY DIAGNOSIS
with age, from 0.23% in those <50 years to 1.91%
in those between 75 and 79 years (77). In those
A priori knowledge of the tumor mutations is es-
<50 years of age, the frequency of detectable
sential for the functioning of the most sensitive
clonal mosaicism in peripheral blood is low (<0.5%)
ctDNA sequencing techniques. Most studies gar-
ner this knowledge by sequencing tissue biopsy but rapidly increases to 2% to 3% in the elderly
samples and designing personalized tests. This population (78).
labor-intensive approach is required for each pa- Genovese et al. performed a massive study of
tient. Thus, these methods could not be used in 12380 individuals of 19 to 93 years of age. They
detection of disease; by the nature of this process, used whole exome sequencing on peripheral
there would be no previous biopsy sample to work blood cells and followed subjects for 2 to 7 years.
off. They observed clonal hematopoiesis in 10% of
Currently, exome sequencing is the only method people >65 years of age and only 1% in those <50
for sequencing ctDNA without knowing the tumor years. Significantly, they found that some of the
mutations. This method is extremely expensive, genes that are mutated in patients with myeloid
especially to sequence at the depth required for cancers are also mutated in healthy individuals
primary tumors. Consequently, there are limited and do not cause cancer. Subsequently, they
studies using this technique in the literature. found that the risk of an individual with clonal

...............................................................................................................

September 2018 | 03:02 | 300 –313 | JALM 307


SPECIAL REPORT Circulating Tumor DNA for Early Cancer Detection

hematopoiesis developing cancer was only 1.0% of Clinical Oncology (http://www.globenewswire.


per year (79). com/news-release/2017/06/03/1007917/0/en/GRAIL-
Another study reported mutations in individuals Announces-Data-From-Early-Research-Analyzing-its-
who remained free of cancer over a 6-year follow-up High-Intensity-Sequencing-Approach.html). They se-
period. KRAS2 was mutated with a frequency of 1.2%, quenced tumor tissue and ctDNA from blinded
and p53 was mutated 3.6% in healthy volunteers (80). blood and tissue from 124 patients with meta-
In 2016, Fernandez-Cuesta reported an even higher static breast, NSCLC, and castration-resistant
mutation rate: 11% of healthy controls had detect- prostate cancer. The main novelty is their high-
able cfDNA p53 mutations (72). Mutations of p53 in throughput approach; sequencing the cfDNA at
normal individuals were also reported by Newman 60 000× raw depth using a 508-gene panel. They
et al. (81). detected at least 1 variant in plasma that was
previously detected in tumor tissue in 90% of the
LOOKING FORWARD patients. In total, 72% of the actionable muta-
tions that were detected in tumor tissue were
Despite all these challenges, at least 1 company also detected in plasma (82).
(GRAIL) was founded in 2016 to develop a blood GRAIL also acknowledges the challenge of clonal
test for early-stage cancer in asymptomatic individ- hematopoiesis in its twin abstract studying muta-
uals to detect tumors so tiny that they cannot be tions in 151 metastatic cancer patients and 47
viewed on imaging. The company's current plan healthy volunteers. It found that both groups had
is to sequence ctDNA from thousands of blood mutations in their blood, most commonly in
samples at an incredible depth (https://grail.com/ DNMT3A, TET2, PPM1D, and TP53, and the rate of
about). mutations increased with age. This makes devel-
In August 2016, GRAIL launched the Circulating oping their proposed ctDNA test even more chal-
Cell-Free Genome Atlas study to characterize the lenging (83).
mutational heterogeneity of cancer, as well as to The development of a plasma test for early-
distinguish benign and cancerous mutations. They stage cancer detection is a noble cause and could
aim to collect contemporaneous blood and tumor save millions of lives. However, more research is
samples from 10500 patients with early-stage ma- required to investigate the challenges that this test
lignancies and blood samples from 4500 healthy must surmount. A standardized process, e.g., re-
controls. The study's estimated completion date is agents, equipment, and procedure, must be estab-
September 2023 (https://clinicaltrials.gov/show/ lished to homogenize the collection of ctDNA (42).
NCT02889978). Other challenges may be more difficult to over-
Since February 2017, GRAIL has also been en- come. Regardless of the sensitivity of the sequenc-
rolling participants in their STRIVE breast cancer ing technology, there simply may not be enough
study, an observational longitudinal investigation ctDNA in a sample from an early-stage cancer pa-
of 120 000 women undergoing routine mam- tient to allow detection.
mography. Blood samples will be collected and Very small tumors, e.g., those with 5-mm diam-
the cfDNA sequenced, and the women will be eter, are unlikely to cause clinical symptoms or be
followed for 5 years to see whether they receive visible on imaging. Although the detection of
a cancer diagnosis (https://clinicaltrials.gov/ct2/ smaller tumors, e.g., 1-mm diameter, is the goal of
show/NCT03085888). some institutions, these tumors are more likely to
GRAIL released its first abstracts in June 2017 be benign and may be overtreated (84). Tumors
during the annual meeting of the American Society with a 5-mm diameter have only a 6% chance of

...............................................................................................................

308 JALM | 300 –313 | 03:02 | September 2018


Circulating Tumor DNA for Early Cancer Detection SPECIAL REPORT

Fig. 1. Likelihood of detecting tumors of various sizes through ctDNA quantification.

progressing and are also localized, less complex, These predictions were recently verified by 2
and easier to cure (85). However, only 26% of these high-profile reports on the use of ctDNA for early
tiny malignancies are detected on imaging (86), cancer diagnosis. Phallen et al. and Cohen et al.
e.g., mammography, which has a detection limit of reported good sensitivities and specificities for de-
4-mm lesions (87). tecting early-stage cancers, but all their patients
Assuming a spherical neoplasm and based on had fractional ctDNA >0.01% and all patients were
published reports (88), we recently calculated that symptomatic (90, 91).
a tumor volume of 1 cm3 or 12.5 mm in diameter Based on current data, it is unlikely that ctDNA
(89) will release into the circulation enough ctDNA has high enough sensitivity or specificity for use in
to represent 0.01% of all circulating DNA (90). We early detection in asymptomatic patients. Clearly,
have further shown that this fraction of ctDNA is this is an active area of investigation that promises
equivalent to 1 mutant genome equivalent per 4 to produce data that are superior to those deliv-
mL of plasma (10-mL blood draw). These data led ered by the classical cancer biomarkers. Advances
us to conclude that ctDNA could be used to detect in assay sensitivity, the bioinformatic pipeline, data
tumors of approximately 10 mm in diameter but analysis/interpretation, and the utility of panel pro-
not smaller (89). A visual representation of these filing may lead to the improved performance of
data can be found in Fig. 1. ctDNA for early cancer detection.

...............................................................................................................

September 2018 | 03:02 | 300 –313 | JALM 309


SPECIAL REPORT Circulating Tumor DNA for Early Cancer Detection

Additional Content on this Topic

Circulating Tumor DNA as a Cancer Biomarker: Fact or Fiction?


Felix Leung, Vathany Kulasingam, Eleftherios P. Diamandis, Dave S.B. Hoon, et al. Clin Chem 2016;62:
1054 – 60

Author Contributions: All authors confirmed they have contributed to the intellectual content of this paper and have met the following
4 requirements: (a) significant contributions to the conception and design, acquisition of data, or analysis and interpretation of data; (b)
drafting or revising the article for intellectual content; (c) final approval of the published article; and (d) agreement to be accountable for
all aspects of the article thus ensuring that questions related to the accuracy or integrity of any part of the article are appropriately
investigated and resolved.

Authors’ Disclosures or Potential Conflicts of Interest: Upon manuscript submission, all authors completed the author disclosure
form. Employment or Leadership: None declared. Consultant or Advisory Role: E. Diamandis, Abbott Diagnostics. Stock
Ownership: None declared. Honoraria: None declared. Research Funding: None declared. Expert Testimony: None
declared. Patents: None declared.

Role of Sponsor: No sponsor was declared.

REFERENCES
1. Mandel P, Métais P. Les acides nucléiques du plasma plasma DNA and cell-free DNA from other body fluids.
sanguin chez l'homme. C R Seances Soc Biol Fil 1948; Ann N Y Acad Sci 2008;1137:135–9.
142:241–3. 10. Elshimali YI, Khaddour H, Sarkissyan M, Wu Y, Vadgama
2. Vasioukhin V, Anker P, Maruice P, Lyautey J, Lederrey C, JV. The clinical utilization of circulating cell free DNA
Stroun M. Point mutations of the N-ras gene in the blood (CCFDNA) in blood of cancer patients. Int J Mol Sci 2013;
plasma DNA of patients with myelodysplastic syndrome 14:18925–58.
or acute myelogenous leukaemia. Br J Haematol 1994; 11. Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth
86:774 –9. JF, et al. Circulating tumor cells in patients with breast
3. Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, cancer dormancy. Clin Cancer Res 2004;10:8152– 62.
Yao SL. Soluble normal and mutated DNA sequences 12. Jung M, Klotzek S, Lewandowski M, Fleischhacker M, Jung
from single-copy genes in human blood. Cancer K. Changes in concentration of DNA in serum and
Epidemiol Biomarkers Prev 1994;3:67–71. plasma during storage of blood samples. Clin Chem
4. Crowley E, Di Nicolantonio E, Loupakis F, Bardelli A. 2003;49:1028 –9.
Liquid biopsy: monitoring cancer-genetics in the blood. 13. Jiang P, Lo YM. The long and short of circulating cell-free
Nat Rev Clin Oncol 2013;10:472– 84. DNA and the ins and outs of molecular diagnostics.
5. Heitzer E, Ulz P, Gegil JB. Circulating tumor DNA as a Trends Genet 2016;32:360 –71.
liquid biopsy for cancer. Clin Chem 2015; 14. Chan KC, Lo YM. Circulating tumour-derived nucleic acids
61:112–23. in cancer patients: potential applications as tumour
6. Choi JJ, Reich CF III, Pietsky DS. The role of macrophages markers. Br J Cancer 2016;96:681–5.
in the in vitro generation of extracellular DNA from 15. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y,
apoptotic and necrotic cells. Immunology 2005;115: Agrawal N, et al. Detection of circulating tumor DNA in
55– 62. early- and late-stage human malignancies. Sci Transl
7. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Med 2014;6:224ra24.
Hesch RD, et al. DNA fragments in the blood plasma of 16. Scholer LV, Reinert T, Orntoff MW, Kassentoft CG,
cancer patients: quantitations and evidence for their Arnadottir SS, Vang S, et al. Clinical implications of
origin from apoptotic and necrotic cells. Cancer Res monitoring circulating tumor DNA in patients with
2001;61:1659 – 65. colorectal cancer. Clin Cancer Res 2017;23:5437– 45.
8. Alix-Panabieres C, Pantel K. Clinical applications of 17. Tie J, Wang Y, Tomasetti C, Springer S, Kinde I, Silliman N,
circulating tumor cells and circulating tumor DNA as et al. Circulating tumor DNA analysis detects minimal
liquid biopsy. Cancer Discov 2016;6:479 –91. residual disease and predicts recurrence in patients with
9. Kirsch C, Weickmann S, Schmidt B, Fleischhacker M. An stage II colon cancer. Sci Transl Med 2016;8:356ra92.
improved method for the isolation of free-circulating 18. Reinert T, Scholer LV, Thompsen R, Tobiasen H, Vang S,

...............................................................................................................

310 JALM | 300 –313 | 03:02 | September 2018


Circulating Tumor DNA for Early Cancer Detection SPECIAL REPORT

Nordentoft I, et al. Analysis of circulating tumour DNA to Ychou M, et al. Analysis of circulating DNA and protein
monitor disease burden following colorectal cancer biomarkers to predict the clinical activity of regorafenib
surgery. Gut 2016;65:625–34. and assess prognosis in patients with metastatic
19. Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, colorectal cancer: a retrospective, exploratory analysis of
Constantin T, Salari R, et al. Phylogenetic ctDNA analysis the CORRECT trial. Lancet Oncol 2015;16:937– 48.
depicts early-stage lung cancer evolution. Nature 2017; 34. Shu Y, Wu X, Tong X, Wang X, Chang Z, Mao Y, et al.
545:446 –51. Circulating tumor DNA mutation profiling by targeted
20. Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, next generation sequencing provides guidance for
Chin SF, et al. Analysis of circulating tumor DNA to personalized treatments in multiple cancer types. Sci
monitor metastatic breast cancer. N Engl J Med 2013; Rep 2017;7:583.
368:1199 –201. 35. Thompson JC, Yee SS, Troxel AL, Savitch SL, Fan R, Balli D,
21. Garcillas-Murillas I, Schiavon G, Weigelt B, Ng C, Hrebien et al. Detection of therapeutically targetable driver and
S, Cutts RJ, et al. Mutation tracking in circulating tumor resistance mutations in lung cancer patients by next
DNA predicts relapse in early breast cancer. Sci Transl generation sequencing of cell-free circulating tumor
Med 2015;7:302ra133. DNA. Clin Cancer Res 2016;22:5772– 82.
22. Gray ES, Rizos H, Reid AL, Boyd SC, Pereira MR, Lo J. 36. Mazzucchelli R, Colanzi P, Pomante R, Muzzonigro G,
Circulating tumor DNA to monitor treatment response Montironi R. Prostate tissue and serum markers. Adv Clin
and detect acquired resistance in patients with Pathol 2000;4:111–20.
metastatic melanoma. Oncotarget 2015;6:420000 –18. 37. Ruibal Morell A. CEA serum levels in non-neoplastic
23. Lee RJ, Gremel G, Marshall A, Myers KA, Fisher N, Dunn disease. Int J Biol Markers 1992;7:160 – 6.
JA, et al. Circulating tumor DNA predicts survival in 38. Sikaris KA. CA125—a test with a change of heart. Heart
patients with resected high risk stage II/III melanoma. Lung Circ 2011;20:634 – 40.
Ann Oncol 2017;29:490 – 6. 39. Ballehaninna UK, Chamberlain RS. Serum CA. 19 –9 as a
24. Van Ginkel JW, Huibers MMH, Van Es RJJ, de Bree R, biomarker for pancreatic cancer—a comprehensive
Willems SM. Droplet digital PCR for detection and review. Indian J Surg Oncol 2011;2:88 –100.
quantification of circulating tumor DNA in plasma of 40. Wanebo HJ, Rao B, Pinsky CM, Hoffman RG, Stearns M,
head and neck cancer patients. BMC Cancer 2017;17:428. Schwartz MK, Oettgen HF. Preoperative
25. Scherer F, Kurtz DM, Newman AM, Stehr H, Craig AF, carcinoembryonic antigen level as a prognostic indicator
Esfahani MS, et al. Distinct biological subtypes and in colorectal cancer. N Engl J Med 1978;299:448 –51.
patterns of genome evolution in lymphoma revealed by 41. Leung F, Kulasingam V, Diamandis EP, Hoon DS, Kinzler
circulating tumor DNA. Sci Transl Med 2016;8:364ra24. K, Pantel K, Alix-Panabieres C. Circulating tumor DNA as
26. He W, Bishop KS. The potential use of cell-free- a cancer biomarker: fact or fiction? Clin Chem 2016;62:
circulating-tumor DNA as a biomarker for prostate 1054 – 60.
cancer. Expert Rev Mol Diagn 2016;8:839 –52. 42. Wang R, Li X, Zhang H, He J. Cell-free circulating tumor
27. Ono A, Fujimoto A, Yamamoto Y, Akamatsu S, Hiraga N, DNA analysis for breast cancer and its clinical utilization
Imamura M, et al. Circulating tumor DNA analysis for liver as a biomarker. Oncotarget 2017;8:75742–55.
cancers and its usefulness as a liquid biopsy. Cell Mol 43. Huang TY, Piunti A, Lulla R, Qi J, Horbinski CM, Tomita, et
Gastroenterol Hepatol 2015;1:516 –34. al.. Detection of histone H3 mutations in cerebrospinal
28. Hamakawa T, Kukita Y, Kurokawa Y, Miyazaki Y, Takahashi fluid-derived tumor DNA from children with diffuse
T, Yamasaki M, et al. Monitoring gastric cancer midline glioma. Acta Neuropathol Commun 2017;5:28.
progression with circulating tumour DNA. Br J Cancer 44. Wang Y, Springer S, Zhang M, McMahon KW, Kinde I,
2015;1:352– 6. Dobbyn L, et al. Detection of tumor-derived DNA in
29. Pereira E, Camacho-Vanegas O, Anand S, Sebra R, cerebrospinal fluid of patients with primary tumors of
Catalina Camacho S, Garnar-Wortzel L, et al. the brain and spinal cord. Proc Natl Acad Sci U S A 2015;
Personalized circulating tumor DNA biomarkers 112:9704 –9.
dynamically predict treatment response and survival in 45. Di Meo A, Bartlett J, Cheng Y, Pasic MD, Yousef GM.
gynecologic cancers. PLoS One 2015;10:e0145754. Liquid biopsy: a step forward towards precision medicine
30. Fenner A. Kidney cancer: tracking and therapy selection in urologic malignancies. Mol Cancer 2017;16:80.
using ctDNA. Nat Rev Urol 2017;14:389. 46. Couffignal C, Desgrandchamps F, Mongiat-Artus P,
31. Cheng H, Liu C, Jiang J, Luo G, Lu Y, Jin K, et al. Analysis of Ravery V, Ouzaid I, Roupret M, et al. The diagnostic and
ctDNA to predict prognosis and monitor treatment prognostic performance of urinary FGFR3 mutation
responses in metastatic pancreatic cancer patients. Int J analysis in bladder cancer surveillance: a prospective
Cancer 2017;140:2344 –50. multicenter study. Urology 2015;86:1185–90.
32. Chaudhuri HH, Chabon JJ, Lovejoy AF, Newman AM, Stehr 47. Pu D, Liang H, Wei F, Akin D, Feng Z, Yan QX, et al.
H, Azad TD, et al. Early detection of molecular residual Evaluation of a novel saliva-based epidermal growth
disease in localized lung cancer by circulating tumor DNA factor receptor mutation detection for lung cancer: a
profiling. Cancer Discov 2017;7:1394 – 403. pilot study. Thorac Cancer 2016;7:428 –36.
33. Tabernero J, Lenz HJ, Siena S, Sobrero A, Falcone A, 48. Van Ginkel JH, Slieker FJB, de Bree R, van Es RJJ, Van Cann

...............................................................................................................

September 2018 | 03:02 | 300 –313 | JALM 311


SPECIAL REPORT Circulating Tumor DNA for Early Cancer Detection

EM, Willems SM. Cell-free nucleic acids in body fluids as et al. Establishment of tumor-specific copy number
biomarkers for the prediction and early detection of alterations from plasma DNA of patients with cancer. Int
recurrent head and neck cancer: a systematic review of J Cancer 2013;133:346 –56.
the literature. Oral Oncol 2017;75:8 –15. 64. Aravanis AM, Lee M, Klausner RD. Next-generation
49. Berger BM, Ahlquist DA. Stool DNA screening for sequencing of circulating tumor DNA for early cancer
colorectal neoplasia: biological and technical basis for detection. Cell 2017;168:571– 4.
high detection rates. Pathology 2012;44:80 – 8. 65. Diamandis EP. Cancer dynamics and the success of
50. Ignatiadas M, Lee M, Jeffrey SS. Circulating tumor cells cancer screening programs. Clin Chem Lab Med 2016;
and circulating tumor DNA: challenges and opportunities 54:e211–2.
on the path to clinical utility. Clin Cancer Res 2015;21: 66. Diamandis EP, Li M. The side effects of translational
4786 – 800. omics: overtesting, overdiagnosis, overtreatment. Clin
51. Millner LM, Linder MW, Valdes R Jr. Circulating tumor Chem Lab Med 2016;54:389 –96.
cells: a review of present methods and the need to 67. Esserman L, Shieh Y, Thompson I. Rethinking screening
identify heterogeneous phenotypes. Ann Clin Lab Sci for breast cancer and prostrate cancer. JAMA 2009;302:
2013;43:295–304. 1685–92.
52. Lianidou ES. Gene expression profiling and DNA 68. Wentzensen N, Wacholder S. From differences in means
methylation analyses of CTCs. Mol Oncol 2016;10:431– 42. between cases and controls to risk stratification: a
53. Naoe M, Ogawa Y, Morita J, Omori K, Takeshita K, business plan for biomarker development. Cancer
Shichijyo T, et al. Detection of circulating urothelial Discov 2013;3:148 –57.
cancer cells in the blood using the CellSearch System. 69. Vincent A, Herman J, Schulick R, Hruban RH, Goggins M.
Cancer 2007;109:1439 – 45. Pancreatic cancer. Lancet 2011;378:607–20.
54. Wolfrum F, Vogel I, Fändrich F, Kalthoff H. Detection and
70. Takai E, Yachida SD. ctDNA as a liquid biopsy target for
clinical implications of minimal residual disease in gastro-
detection of pancreatic cancer. World J Gastroenterol
intestinal cancer. Langenbecks Arch Surg 2005;390:430–41.
2016;22:8480 – 8.
55. Markou A, Lazaridou M, Paraskevopoulos P, Chen S,
71. Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras
Swierszewska M, Budna J, et al. Multiplex gene
MC, et al. Genomic analyses identify molecular subtypes
expression profiling of in vivo isolated circulating tumor
of pancreatic cancer. Nature 2016;531:47–52.
cells in high-risk prostate cancer patients. Clin Chem
72. Fernandez-Cuesta F, Perdomo S, Avogbe PH, Leblay N,
2018;64:297–306.
Delhomme TM, Gaborieau V, et al. Identification of
56. Makarovskiy AN, Ackerley W 3rd, Wojcik L, Halpert GK,
circulating tumor DNA for the early detection of small-
Stein BS, Carreiro MP, Hixson DC. Application of
cell lung cancer. EBioMedicine 2016;10:117–23.
immunomagnetic beads in combination with RT-PCR for
the detection of circulating prostate cancer cells. J Clin 73. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC,
Lab Anal 1997;11:346 –50. Modlin LA, et al. An ultrasensitive method for
quantitating circulating tumor DNA with broad patient
57. Choy A, McCulloch PG. Detection and enumeration of
coverage. Nat Med 2014;20:548 –54.
circulating tumour cells in colorectal cancer. Br J Surg
1993;80:1490. 74. Spellman PT, Gray JW. Detecting cancer by monitoring
58. Giannopoulou L, Kasimir-Bauer S, Lianidou ES. Liquid circulating tumor DNA. Nat Med 2014;20:474 –5.
biopsy in ovarian cancer: recent advances on circulating 75. Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T,
tumor cells and circulating tumor DNA. Clin Chem Lab Piskorz AM, et al. Non-invasive analysis of acquired
Med 2018;56:186 –97. resistance to cancer therapy by sequencing of plasma
59. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, DNA. Nature 2013;497:108 –12.
Miller MC, et al. Circulating tumor cells, disease 76. Martincorena I, Campbell PJ. Somatic mutation in cancer
progression, and survival in metastatic breast cancer. and normal cells. Science 2015;349:1483–9.
N Engl J Med 2004;351:781–91. 77. Jacobs KB, Yeager M, Zhou W, Wacholder S, Wang Z,
60. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rodriguez-Santiago B, et al. Detectable clonal mosaicism
Rao C, et al. Tumor cells circulate in the peripheral blood and its relationship to aging and cancer. Nat Genet 2012;
of all major carcinomas but not in healthy subjects or 44:651– 8.
patients with nonmalignant diseases. Clin Cancer Res 78. Laurie CC, Laurie CA, Rice K, Doheny KF, Zelnick LR,
2004;10:6897–904. McHugh CP, et al. Detectable clonal mosaicism from
61. Zhang Z, Ramnath N, Nagrath S. Current status of CTCs birth to old age and its relationship to cancer. Nat Genet
as liquid biopsy in lung cancer and future directions. 2012;6:642–50.
Front Oncol 2015;5:209. 79. Genovese G, Kahler AK, Handsaker RE, McCarroll SA.
62. Shaw JA, Brown J, Coombes RC, Jacob J, Payne R, Lee B, Clonal hematopoiesis and blood-cancer risk inferred
et al. Circulating tumor cells and plasma DNA analysis in from blood DNA sequence. N Engl J Med
patients with indeterminate early or metastatic breast 2014;371:2477– 87.
cancer. Biomark Med 2011;5:87–91. 80. Gormally E, Vineis E, Matullo G, Veglia F, Caboux E, Le
63. Heitzer E, Auer M, Hoffman EM, Pichler M, Gasch C, Ulz P, Roux E, et al. TP53 and KRAS2 mutations in plasma DNA

...............................................................................................................

312 JALM | 300 –313 | 03:02 | September 2018


Circulating Tumor DNA for Early Cancer Detection SPECIAL REPORT

of healthy subjects and subsequent cancer occurrence: Tretli S. Breast cancer tumor growth estimated through
a prospective study. Cancer Res 2006;66:6871– 6. mammography screening data. Breast Cancer Res 2008;
81. Newman AM, Lovejoy AF, Klass DM, Kurtz DM, Chabon JJ, 10:R4.
Scherer F, et al. Integrated digital error suppression for 87. Schwaederle M, Husain H, Fanta PT, Piccioni DE, Kesari S,
improved detection of circulating tumor DNA. Nat Schwab RB, et al. Detection rate of actionable mutations
Biotechnol 2016;34:547–55. in diverse cancers using a biopsy-free (blood) circulating
82. Razavi R, Li BT, Abida W, Aravanis A, Jung B, Shen R, et al. tumor cell DNA assay. Oncotarget 2016;7:9707–17.
Performance of a high-intensity 508-gene circulating- 88. Del Monte U. Does the cell number 109 still really fit one
tumor DNA (ctDNA) assay in patients with metastatic gram of tumor tissue? Cell Cycle 2009;8:505– 6.
breast, lung, and prostate cancer. J Clin Oncol 2017;35:
89. Diamandis EP, Fiala C. Can circulating tumor DNA be
LBA11516.
used for direct and early stage cancer detection? F1000
83. Rasavi P, Li BT, Hou C, Shen R, Venn O, Lim RS, et al. Cell-
Res 2017;6:2129.
free DNA (cfDNA) mutations from clonal hematopoiesis:
implications for interpretation of liquid biopsy tests. J Clin 90. Phallen J, Sausen M, Adleff V, Leal A, Hruban C, White J,
Oncol 2017;35:11526. et al. Direct detection of early-stage cancers using
84. Garber K. Ontario institute offers new model of cancer circulating tumor DNA. Sci Transl Med 2017;9:eaan2415.
research. J Natl Cancer Inst 2008;100:980 –2. 91. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L,
85. Countercurrents Series, Narod SA. Disappearing breast et al. Detection and localization of surgically resectable
cancers. Curr Oncol 2012;19:59 – 60. cancers with a multi-analyte blood test. Science 2018;
86. Weedon-Fekjaer H, Lindqvist BH, Vatten LJ, Aalen OO, 359:926 –30.

...............................................................................................................

September 2018 | 03:02 | 300 –313 | JALM 313

You might also like