Blood Based Tests For Multicancer Early Detection

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Articles

Blood-based tests for multicancer early detection


(PATHFINDER): a prospective cohort study
Deb Schrag, Tomasz M Beer, Charles H McDonnell III, Lincoln Nadauld, Christina A Dilaveri, Robert Reid, Catherine R Marinac, Karen C Chung,
Margarita Lopatin, Eric T Fung, Eric A Klein

Summary
Background Multicancer early detection (MCED) blood tests can detect a cancer signal from circulating cell-free DNA Lancet 2023; 402: 1251–60
(cfDNA). PATHFINDER was a prospective cohort study investigating the feasibility of MCED testing for cancer See Comment page 1213
screening. Memorial Sloan Kettering
Cancer Center, New York, NY,
USA (Prof D Schrag MD); Exact
Methods In this prospective cohort study done in oncology and primary care outpatient clinics at seven US health
Sciences, Madison, WI, USA
networks, a convenience sample of adults aged 50 years or older without signs or symptoms of cancer consented to (Prof T M Beer MD); Sutter
MCED testing. We collected blood, analysed cfDNA, and returned results to participants’ doctors. If a methylation Health, Sacramento, CA, USA
signature indicative of cancer was detected, predicted cancer signal origin(s) informed diagnostic assessment. The (C H McDonnell III MD);
Intermountain Healthcare,
primary outcome was time to, and extent of, diagnostic testing required to confirm the presence or absence of cancer.
St George, UT, USA
This trial is registered at ClinicalTrials.gov, NCT04241796, and is completed. (L Nadauld MD); Mayo Clinic,
Rochester, MN, USA
Findings Between Dec 12, 2019, and Dec 4, 2020, we recruited 6662 participants. 4204 (63·5%) of 6621 participants (C A Dilaveri MD); US Oncology
Research, VA Cancer Specialists,
with analysable results were women, 2417 (36·5%) were men, and 6071 (91·7%) were White. A cancer signal was
Fairfax, VA, USA (R Reid MD);
detected in 92 (1·4%) of 6621 participants with analysable results. 35 (38%) participants were diagnosed with cancer Dana-Farber Cancer Institute,
(true positives) and 57 (62%) had no cancer diagnosis (false positives). Excluding two participants whose diagnostic Boston, MA, USA
assessments began before MCED test results were reported, median time to diagnostic resolution was 79 days (C R Marinac PhD); GRAIL,
Menlo Park, CA, USA
(IQR 37–219): 57 days (33–143) in true-positive and 162 days (44–248) in false-positive participants. Most participants
(K C Chung MS,
had both laboratory tests (26 [79%] of 33 with true-positive results and 50 [88%] of 57 with false-positive results) and E T Fung MD PhD,
imaging (30 [91%] of 33 with true-positive results and 53 [93%] of 57 with false-positive results). Fewer procedures Prof E A Klein MD, M Lopatin MS)
were done in participants with false-positive results (17 [30%] of 57) than true-positive results (27 [82%] of 33) and Correspondence to:
few had surgery (one with a false-positive result and three with a true-positive result). Prof Deb Schrag, Memorial Sloan
Kettering Cancer Center,
New York, NY 10065, USA
Interpretation This study supports the feasibility of MCED screening for cancer and underscores the need for further schragd@mskcc.org
research investigating the test’s clinical utility.

Funding GRAIL.

Copyright © 2023 Elsevier Ltd. All rights reserved.

Introduction these tests are new and insight about how doctors and
Cancer is the second most common cause of death in patients respond to a positive MCED test is limited.7–10
Europe and the USA and, globally, more than 1·9 million The PATHFINDER study was designed to address this
new cases are expected to be diagnosed in 2023.1 knowledge gap.
Recommended cancer screening tests have reduced The MCED test investigated in this study detects
cancer-related mortality, but are only available for a few cancer-specific DNA methylation patterns from cell-free
cancer types and represent less than 25% of cancer DNA (cfDNA) shed by tumours into circulating blood. If
deaths in the USA.2 The US Preventive Services Task a cancer signal is detected, the test also predicts the
Force advises screening (grade A/B) for breast, lung, cancer signal origin (CSO; ie, the cancer or tissue type
colorectal, and cervical cancers. For many other cancer where the tumour arose).7,8 This test was developed and
types, screening strategies either lack well established validated in the Circulating Cell-Free Genome Atlas
net benefit or do not exist. As a result, many cancers are (CCGA) study (NCT02889978, US Food and Drug
identified at late stages, when treatment is less Administration Investigational Device Exemptions
effective.3 G190251). In the CCGA study, the test detected a
Advances in genomics and machine learning have methylation-based signal for more than 50 distinct
enabled development of blood tests for cancer screening, cancer types from a single blood draw and accurately
monitoring, and treatment selection.4–10 Multicancer early predicted CSO in 1273 (89%) of 1435 participants.7,8 We
detection (MCED) blood tests have the potential to aimed to investigate the feasibility of MCED testing in
improve the efficiency and convenience of screening and outpatient settings for adults older than 50 years without
do not carry the risks of whole-body imaging. However, symptoms of cancer.

www.thelancet.com Vol 402 October 7, 2023 1251


Articles

Research in context
Evidence before this study Added value of this study
Previous evidence has established that cancers shed cell-free DNA PATHFINDER assessed clinician-guided diagnostic
(cfDNA) into the bloodstream that is detectable before the onset assessments for adults who received positive results from a
of clinical signs and symptoms, creating a window of preclinical targeted-methylation blood-based multicancer early
detectability. We searched PubMed using the terms “multicancer detection (MCED) test as well as the extent of imaging and
early detection” OR “multicancer detection” OR “liquid biopsy diagnostic testing needed to reach diagnostic resolution.
AND cancer screening” for publications in English published The study measured performance characteristics of this test
between Jan 1, 1991, and July 29, 2023, and found 2415 results. applied among adults aged 50 years or older with and
After excluding non-human studies, reviews, commentaries, without increased cancer risk seen in outpatient medical
health economics-focused articles, and single-cancer studies, practice. These findings add to emerging evidence supporting
we identified 12 analytic or case-control studies using various the feasibility of MCED testing to screen for multiple types of
technologies to detect cancer signals of multiple tumour types in cancer simultaneously from a single blood specimen and
blood. Collectively, these studies have established the feasibility characterise the diagnostic pathways that follow receipt of a
and putative performance characteristics of genomic-based tools positive MCED test.
to detect cancers and predict their tissue of origin that could serve
Implications of all the available evidence
to establish a new blood-based screening framework. To date,
These results support further development of MCED testing
one published cohort study has investigated the feasibility and
for cancer screening and provide preliminary estimates of the
safety of a blood-based assay for multicancer detection, but this
consequences and diagnostic yield of this emerging
study was limited to women in a single health system and relied
technology. Further research is needed to investigate the
on whole-body imaging rather than a blood test for tumour
clinical utility of MCED testing to screen for cancer in diverse
localisation. Little evidence exists about the diagnostic
populations.
assessments that ensue following the use of this technology, the
test performance characteristics, or how patients and clinicians
deal with the results.

Methods Procedures
Study design and participants Participants contributed a blood specimen, obtained at
In the PATHFINDER study, we assessed use of MCED outpatient ambulatory care clinics, typically by
testing in a prospective cohort study done in oncology and phlebotomists. Plasma isolation, followed by cfDNA
primary care outpatient clinics at seven US health extraction, was done as previously described.7,8,11 Results
networks, including hospitals and community-based of the MCED test were returned to the ordering doctor
clinics. Eligible participants, a non-consecutive within approximately 15 days and then shared with
convenience sample, consented to MCED testing and participants. The MCED test report provided a binary
1 year of follow-up surveillance of their electronic health result (cancer signal detected or not detected), including
records. No specific diagnostic or screening assessments prediction of the most likely CSOs if a signal was
were mandated. All study procedures were approved by a detected. If a cancer signal was detected, participants had
central or local institutional review board. Additional diagnostic assessments coordinated by, and at the
details regarding study design have been previously discretion of, their doctor. Doctors determined when the
published.11 diagnostic work-up was considered complete. Parameters
Eligible participants were adults aged 50 years or older, for this diagnostic assessment were not stipulated by the
with or without additional cancer risk factors. Participants protocol, and the study sponsor supported all costs. This
with additional risk met at least one of the following design enabled insight into the diagnostic pathways
criteria: a history of smoking at least 100 cigarettes within followed in response to MCED test results and CSO
the participant’s lifetime, cancer predisposition based on predictions. All participants, including those with no
meeting criteria for germline testing from the National cancer signal detected, were advised to continue usual
Comprehensive Cancer Network guidelines12 or having medical care and guideline-recommended cancer
known hereditary cancer syndrome, or personal history screening.
of cancer with definitive treatment completed at least We did an end-of-study cancer-status assessment
3 years before enrolment.11 Participants were excluded if 12 months after enrolment, which included a review of
they were undergoing diagnostic assessment for electronic health records to confirm the presence or
suspicion of cancer or cancer recurrence, had a history of absence of cancer. Record review was supplemented with
untreated cancer, or had been treated for cancer within telephone contact as needed. Cancer status assessment
3 years of enrolment. Participants provided written was considered complete if a cancer diagnosis was
informed consent. reported at any time during the follow-up period or there

1252 www.thelancet.com Vol 402 October 7, 2023


Articles

was no cancer diagnosis recorded in the electronic health undetermined significance [MGUS]) were not considered
records or through patient contact 12 months after the cancer.
MCED blood draw. A 12-month follow-up period has traditionally been used
The MCED test used in PATHFINDER uses next- to assess the performance characteristics of single cancer-
generation sequencing to interrogate cfDNA in peripheral screening modalities, including mammography,16–18 low-
blood to measure the extent and location of genomic dose CT screening for lung cancer19–21 and ovarian cancer
DNA methylation patterns, which are characteristic of screening.22 The same interval was chosen for this study to
cancers13 and specific to cancer type. A computational facilitate assessment of MCED test performance compared
algorithm identifies whether a cancer signal is present with these other cancer screening tests. This arbitrary
and, if so, a second algorithm predicts the most likely reference was not informed by biological considerations.
tumour type, labelled the CSO. This prediction might Within 12 months of enrolment, participants with an
help to direct diagnostic assessment.8,14 We assigned CSO MCED cancer signal detected and a clinically confirmed
predictions from a set of 21 predetermined categories that cancer were classified as true positive and those without
correspond to major tumour types described (appendix confirmed cancer as false positive. Participants with no See Online for appendix
p 5),7,8,11 and are ranked in order of likelihood. We MCED cancer signal detected but who had a confirmed
calculated CSO prediction accuracy as the number of cancer diagnosis within 12 months were classified as
patients with a correctly predicted tumour type among all false negative. False negatives were defined as those
patients with a confirmed cancer diagnosis following a participants with cancers detected by means other than
positive MCED assay. Technical aspects of the assay and the diagnostic work-up prompted by a positive MCED
clinical validation have been described previously.7,8 test, which included those detected by routine screening
assessments that occurred during the 12-month study
Outcomes period or those detected because the patient developed
The primary objective was to investigate the time signs or symptoms. Screening was defined as standard if
required to achieve diagnostic resolution following it was consistent with US Preventive Services Task Force
receipt of a positive MCED test and to assess the extent of (USPSTF) grade A, B, or C recommendations (breast,
testing pursued. Time to diagnostic resolution for each colorectal, cervical, prostate, or high-risk lung), although
participant was defined as the number of days between not all institutions follow USPSTF guidelines
the date that test results were available to the ordering specifically.23 Cancers detected by non-standard screening
doctor through the reporting portal and the date of included cancers of the ovary, testis, thyroid, and skin
diagnostic resolution as determined by the ordering without USPSTF recommendations. Clinically detected
doctor. cancers were defined as those diagnosed on the basis of a
For each participant, the extent of diagnostic testing patient’s presentation with clinical signs or symptoms
included all clinic visits, laboratory and imaging tests, such as pain, unexplained weight loss, or a palpable
and procedures recorded between the date a positive test mass. More detail on the definitions of cancers detected
result was available in the portal and the date of diagnostic by standard screening, cancers detected by non-standard
resolution or end of study, whichever came first. screening, and clinically detected cancers is provided in
Procedures were categorised as either non-surgical the appendix (p 30).
(eg, endoscopy, bone marrow biopsy, core biopsy, fine
needle aspiration, and pap smear) or surgical. Safety
Safety assessments included a summary of adverse
Cancer diagnosis events reported and defined by site investigators at any
A cancer diagnosis was established by pathological, time during the study, including at phlebotomy, results
laboratory, or radiographic confirmation. 113 (93%) of disclosure, or diagnostic assessments in those with a
122 cancers were pathologically confirmed. Malignancies cancer signal detected. Adverse events were reported to
were categorised as either invasive solid tumours the study sponsor and the relevant institutional review
(excluding non-metastatic basal cell carcinoma and board or independent ethics committee. An independent
squamous cell carcinoma of the skin) or haematological. Data Safety Monitoring Board provided oversight of the
Cancer diagnosis and staging for solid tumours was study conduct.
based on the American Joint Committee on Cancer
(AJCC) Staging Manual (version 8) and on AJCC or the Test performance
Ann Arbor staging systems for haematological As a secondary objective, MCED test performance was
malignancies.15 For participants with a cancer history of assessed with the following measures: resolution (true
the same tumour type identified from MCED testing, positive or false positive), positive and negative predictive
staging was categorised as either recurrent locoregional value, specificity, and CSO accuracy. Participants with an
or metastatic for solid tumours and as recurrent for indeterminate CSO were excluded from CSO accuracy
haematological malignancies. Premalignant conditions calculations. Additional performance endpoints were the
(eg, cervical dysplasia or monoclonal gammopathy of cancer yield rate defined as the number of true positives

www.thelancet.com Vol 402 October 7, 2023 1253


Articles

among all participants tested and the number needed to in 55 (0·8%; appendix p 7). This study is registered with
screen to detect one cancer. ClinicalTrials.gov, NCT04241796.
We collected participant-reported outcomes relating to
anxiety, distress, uncertainty, and MCED test satisfaction Development of a refined MCED test
at the time of initial blood draw, when test results were After PATHFINDER was launched, a refined MCED test
returned, when diagnostic resolution was achieved, and version was developed, using a larger CCGA training
at the end of the study, and these will be reported in a dataset. All elements of this refined test were locked
separate manuscript. We did a post hoc exploratory before sample processing, validation was done in a new
analysis of circulating tumour allele fraction (cTAF) for independent test set, and the statistical analysis plan was
all cancers detected during the study. The detection finalised before evaluation. MCED test refinements
threshold was defined as 3·1 × 10–⁴ cTAF at included: (1) an increased cancer signal detection
99·3% specificity.24 All outcome measures are defined in threshold for haematological signals to reduce false
the appendix (p 6). positives from non-malignant hematological conditions
such as MGUS, and decreased threshold for solid tumour
Statistical analysis signals to increase the detection of solid cancers;
Analyses were descriptive with no pre-specified (2) elimination of the “indeterminate” label for CSO to
hypothesis testing. The analysis plan assumed that generate a prediction for all test-positive samples; and
70% of enrolled participants would have at least one (3) restriction of CSO predictions to the two most likely
additional cancer risk factor. Simulations estimated that tumour types. Results of testing using the refined MCED
a target enrolment of 6200 participants would generate test were not returned to either doctors or participants,
106–141 cancer signals detected.11 The primary analysis and therefore did not influence diagnostic assessments.
was based on participants with a diagnostic assessment
triggered by a cancer signal detected. Two-sided Role of the funding source
Wilcoxon 95% CIs are reported for all test performance The study sponsor contributed to the study design, data
measures. Analyses were done using R (version 4.1.2). analysis, interpretation, and drafting, review, and
Protocol deviations occurred in 106 (1·6%) of 6662 approval of the manuscript. The sponsor used a medical
participants, including important protocol deviations writing agency to assist with manuscript preparation.

Outcome analysis sets—true positives

6662 participants enrolled


36 cancers

32 retrospectively ineligible
1 true positive had 2 cancers

6630 clinically eligible


35 true positives

5 withdrew consent

2 true positives began 1 true positive had


diagnostic workup indeterminate CSO 6625 clinically evaluable
before return of
MCED test results

4 assay result not evaluable

33 evaluated for extent of 34 assessed for CSO


diagnostic testing prediction accuracy
6621 with evaluable MCED results

92 cancer signal detected 6529 no cancer signal detected

35 true positive 57 false positive 6235 true negative 86 false negative 208 no end-of-study
assessment

Figure 1: Trial profile


Participant flow diagram, including true-positive analysis sets for outcomes with variable participant numbers. CSO=cancer signal origin. MCED=multicancer early
detection.

1254 www.thelancet.com Vol 402 October 7, 2023


Articles

Results
Aged ≥50 years Aged ≥50 years Total
Between Dec 12, 2019, and Dec 4, 2020, we enrolled with additional without additional (n=6621)
6662 participants, 6625 (99·4%) of whom were risk (n=3681) risk (n=2940)
clinically evaluable, and 6621 (99·4%) had analysable Age, years* 64·0 (58·0–71·0) 61·0 (55·0–67·0) 63·0 (56·0–70·0)
MCED results (figure 1). Of these 6621 participants, Age group, years
3681 (55·6%) were considered to have additional risk 50–64 1858 (50·5%) 1933 (65·7%) 3791 (57·3%)
factors. Participants were predominately White 65–79 1637 (44·5%) 931 (31·7%) 2568 (38·8%)
(6071 [91·7%]) and highly educated (4276 [64·6%] with ≥80 186 (5·1%) 76 (2·6%) 262 (4·0%)
college degrees), and a smaller proportion were current
Sex
smokers (268 [4·0%]) compared with the general
Female 2393 (65·0%) 1811 (61·6%) 4204 (63·5%)
population (table 1). 1622 (24·5%) participants had a
Male 1288 (35·0%) 1129 (38·4%) 2417 (36·5%)
previous cancer history; demographic details and
Race or ethnicity
MCED test performance characteristics in this subset
Asian† 39 (1·1%) 90 (3·1%) 129 (1·9%)
are given in the appendix (p 8). At enrolment, most
Hispanic 66 (1·8%) 68 (2·3%) 134 (2·0%)
participants were up to date with cancer screening,
Non-Hispanic Black 44 (1·2%) 46 (1·6%) 90 (1·4%)
with 4492 (91·9%) of 4888 reporting recent colorectal
Non-Hispanic White 3441 (93·5%) 2630 (89·5%) 6071 (91·7%)
cancer screening and 2854 (80·5%) of 3547 women
Other‡ 28 (0·7%) 38 (1·3%) 66 (1·0%)
reporting up-to-date breast cancer screening (according
Missing 63 (1·7%) 68 (2·3%) 131 (2·0%)
to USPSTF guidelines) among eligible participants
BMI, kg/m²
with complete data (table 1).
A cancer signal was detected in 92 (1·4%) of <18·5 32 (0·9%) 18 (0·6%) 50 (0·8%)

6621 participants (figure 1), with 56 (1·5%) of 3681 in 18·5 to <25·0 1045 (28·4%) 956 (32·5%) 2001 (30·2%)
those with additional risk and 36 (1·2%) of 2940 in those 25·0 to <30·0 1297 (35·2%) 1039 (35·3%) 2336 (35·3%)
without additional risk (table 2). Of those with a cancer ≥30·0 1264 (34·3%) 887 (30·2%) 2151 (32·5%)
signal detected, 35 (38%) of 92 were diagnosed with Other or missing 43 (1·2%) 40 (1·4%) 83 (1·3%)
cancer (true positives) and 57 (62%) had no cancer Education
diagnosis (false positives; figure 1). Most true positives Less than high school 50 (1·4%) 15 (0·5%) 65 (1·0%)
(27 [77%] of 35) had a cTAF above 3·1 × 10–⁴, the MCED High school graduate 345 (9·4%) 150 (5·1%) 495 (7·5%)
clinical detection threshold (appendix p 31). The number Some college 1060 (28·8%) 645 (21·9%) 1705 (25·8%)
of positive MCED tests (n=92) was lower than the College graduate 2176 (59·1%) 2100 (71·4%) 4276 (64·6%)
projected 106–141 in the sample size calculations. Other or missing 50 (1·4%) 30 (1·0%) 80 (1·2%)
Of those without a cancer signal detected, Smoking status
6235 (95·5%) of 6529 were true negatives, 86 (1·3%) were Current smoker 268 (7·3%) 0 268 (4·0%)
false negatives, and 208 (3·2%) did not have a cancer- Former smoker 2229 (60·6%) 0 2229 (33·7%)
status assessment at the end of the study. 82 (95%) of Non-smoker 1184 (32·3%) 2940 (100%) 4124 (62·3%)
86 false negatives had a cTAF below 3·1 × 10–⁴ (appendix Eligible for lung cancer screening§ 223 (6·1%) 0 223 (3·4%)
p 31). Previous cancer history 1622 (44·1%) 0 1622 (24·5%)
Of the 92 participants with a cancer signal detected, Cancer predisposition 425 (11·5%) 0 425 (6·4%)
two (2%) began their diagnostic assessment because of Up to date with standard cancer screening before MCED testing
symptoms or physical examination finding after the Colorectal cancer¶ 2404/2628 (91·5%) 2088/2260 (92·4%) 4492/4888 (91·9%)
blood draw and before the MCED test results were Breast cancer|| 1504/1930 (77·9%) 1350/1617 (83·5%) 2854/3547 (80·5%)
returned and are not included in the analysis of the
Data are n (%), n/N (%), or median (IQR). Total includes all clinically evaluable patients with analysable MCED results.
extent of diagnostic testing (appendix p 10). Median MCED=multicancer early detection. USPSTF=US Preventive Services Task Force. *Age was truncated at 85 years to
numbers of clinic visits, laboratory tests, and imaging protect confidentiality. †Non-Hispanic Asian, Native Hawaiian, or Pacific Islander. ‡Includes unknown,
tests were similar for true-positive and false-positive American Indian, or Alaska Native. §Satisfy approved USPSTF criteria for lung cancer screening using low-dose CT.
¶Participants aged 75 years or younger, up to date with USPSTF colorectal cancer screening recommendations
participants, whereas true positives were more likely (n=4888 eligible with complete information). ||Women aged 50–74 years up to date with breast cancer screening
than false positives to undergo both non-surgical and (USPSTF recommendations, MRI, or ultrasound; n=3547 eligible with complete information).
surgical procedures (appendix p 10).
Table 1: PATHFINDER participant demographics and baseline characteristics
Median observed time to diagnostic resolution was
79 days (IQR 37–219). True positives had a shorter
median time to resolution (57 days [33–143]) compared count with differential (48 [53%]), and protein tumour
with false positives (162 days [44–248]; figure 2). Most markers (39 [43%]; appendix p 10).
true positives (24 [73%] of 33) had diagnostic resolution At least one imaging test was done in
within 3 months (figure 2). 83 (92%) of 90 participants with a cancer signal detected
76 of 90 participants with positive MCED results had (figure 3), and 44 (53%) of those had more than one
laboratory tests (figure 3). The most common laboratory imaging study. PET-CT (55 [61%] of 90), CT (35 [39%]),
tests were a metabolic panel (51 [57%]), complete blood and MRI (19 [21%]) were the most common imaging

www.thelancet.com Vol 402 October 7, 2023 1255


Articles

Age ≥50 years with additional Age ≥50 years without Total
cancer risk (n=3681) additional cancer risk (n=2940) (n=6621)
Resolution
All 56 (1·5%) 36 (1·2%) 92 (1.4%)
True positive 24 (0·7%) 11 (0·4%) 35 (0·5%)
False positive 32 (0·9%) 25 (0·9%) 57 (0·9%)
Positive predictive value 24/56; 43% (30·8–55·9) 11/36; 31% (18·0–46·9) 35/92; 38% (28·8–48·3)
Negative predictive value 3449/3502; 98·5% (98·0–98·8) 2786/2819; 98·8% (98·4–99·2) 6235/6321; 98·6% (98·3–98·9)
Specificity 3449/3480; 99·1% (98·7–99·4) 2786/2810; 99·1% (98·7–99·4) 6235/6290; 99·1% (98·9–99·3)
Yield rate 24/3681; 0·65% (0·41–0·92) 11/2940; 0·37% (0·17–0·61) 35/6621; 0·53% (0·36–0·71)
Number needed to screen 3681/24; 153 (108–245) 2940/11; 267 (163–588) 6621/35; 189 (141–276)
Predicted origin accuracy*
First CSO correct 20/23; 87% (67·9–95·5) 9/11; 82% (52·3–94·9) 29/34; 85% (69·9–93·6)
First or second CSO correct 23/23; 100% (85·7–100) 10/11; 91% (62·3–99·5) 33/34; 97% (85·1–99·8)
Data are n (%), n/N, or % (95% CI). CSO=cancer signal origin. *Excludes one participant with indeterminate CSO from the true-positive set.

Table 2: Multicancer early detection test performance

tests (appendix p 10). Diagnostic tests done in fewer than those with false-negative MCED results are shown in the
three participants are listed in the appendix (p 22). appendix (p 18).
32 (73%) of 44 participants with a haematological CSO Overall positive predictive value was 38% (35/92) and
had a PET-CT scan as their first imaging test, consistent was higher in those with additional risk (24 [43%] of 56)
with diagnostic guidelines.25 and lower in those without additional risk (11 [31%] of 36;
Of the 90 participants with a diagnostic assessment table 2). Negative predictive value was 98·6% (6235/6321;
triggered by MCED testing, 44 (49%) of 90 had at least table 2). Specificity was 99·1% (6235/6290). The cancer
one procedure (figure 3; appendix p 10). More true- yield rate was 0·53% (35/6621), corresponding to a
positive than false-positive participants had procedures number needed to screen to detect one cancer of 189.
(figure 3). In most false-positive participants, procedures A correct first or second CSO prediction was returned
were prompted by abnormal imaging. Three true-positive for 33 (97%) of 34 true positives, excluding one participant
participants had surgical procedures (lymph node with an indeterminate CSO (table 2). The one participant
mesenteric excision, mesenteric mass and mesenteric with an inaccurate first (colon or rectum) and second
lymph node biopsy, and base of tongue biopsy). Only (upper gastrointestinal tract) CSO prediction was
one participant with a false-positive result had a surgical diagnosed with adenocarcinoma of the small intestine. Of
procedure (inguinal orchiectomy), prompted by the 57 false positives, 35 (61%) had a haematological CSO
abnormal imaging. prediction; of these, 12 (34%) had a haematological
Within 12 months after enrolment, 122 cancers were precursor condition, such as MGUS (appendix p 23).
diagnosed in 121 participants (appendix p 14). Of these, Adverse events were reported for four participants,
35 (29%) had a cancer signal detected by MCED, two with events related to phlebotomy (anxiety and
38 (31%) were detected through screening (29 in bruising at the venipuncture site) and two with anxiety
accordance with USPSTF guidelines and nine by non- reported before the MCED test results were returned. All
standard screening tests; appendix p 32) and the remaining events were mild in severity.
48 (40%) were clinically detected. Of the 35 true positives, Results of the refined MCED test are given in
24 (69%) of 35 were in the additional-risk cohort; the appendix (pp 25, 27, 28, 34). 6578 (98·7%) of
28 (80%) of 35 had a new cancer, six (17%) had recurrent 6662 participants had analysable results for the refined
cancer, and one (3%) had both, for a total of 36 cancers MCED test. The refined test version improved
(19 [53%] solid tumours and 17 [47%] haematological specificity (99·5% [95% CI 99·3–99·6]) and had a
malignancies (figure 4). In true positives with a new cancer positive predictive value (43·1% [31·2–55·9]) similar to
diagnosis, 14 (48%) of 29 cancers were stage I–II (figure 4; that of the study test version (table 2; appendix p 27).
appendix p 14): six solid tumours and eight haematological Characteristics of cancers detected with the refined
malignancies. MCED identified a tumour type that does MCED test are summarised in the appendix (p 28).
not have a USPSTF screening recommendation in Concordance between the study and refined versions of
26 (74%) of 35 participants (figure 4; appendix p 14). Most the MCED test is shown in the appendix (p 34). The
new cancers diagnosed in false negatives were stage I–II refined MCED test reduced the proportion of participants
(55 [73%] of 75; appendix p 16). Recurrent cancers were with a haematological CSO prediction among false
identified in 11 (13%) of 86 participants with false-negative positives from 35 (61%) of 57 to four (12%) of 33. With
results. The histology results for cancers diagnosed in the refined MCED test version, three of the

1256 www.thelancet.com Vol 402 October 7, 2023


Articles

Median 57 days True positive False positive


(IQR 33–143)

Any laboratory test 26/33 (79%) 50/57 (88%)


73% (24/33) <3 months

Any imaging test 30/33 (91%) 53/57 (93%)


True positive

Any procedure 27/33 (82%) 17/57 (30%)

Any non-surgical procedure 26/33 (79%) 16/57 (28%)

Any surgical procedure 3/33 (9%) 1/57 (2%)


Median 162 days
(IQR 44–248) 100 50 0 50 100
Participants (%)
42% (24/57) <3 months

Figure 3: Extent of diagnostic testing in participants with cancer signal detected (n=90)

Importantly, the MCED assay detected many cancer


types for which screening tests do not exist, including
some found at early stages. For example, cancers of the
bile duct, small intestine, pancreas, and a spindle-cell
False positive

neoplasm were detected at early stages amenable to


surgical resection. These lethal malignancies are
unlikely to have been identified upon routine physical
examination or screening. Haematological malignancies,
particularly follicular lymphomas, were also detected at
early stages, although the clinical significance of these
more indolent neoplasms is less certain. Nearly half of
the newly diagnosed cancers were identified at an early
stage.
At the study’s inception, participants were highly
adherent to established cancer screening and some
0 100 200 300 400
received forms of screening that extend beyond USPSTF
Days to diagnostic resolution
recommendations. This healthy volunteer effect might
Figure 2: Time to diagnostic resolution for participants with cancer signal help explain why the number of cancers diagnosed by
detected (n=90) MCED testing was less than expected. The lower-than-
projected rate of enrolment of individuals with cancer
four participants with false-positive results predicted to risk factors and uncertainty in assumptions regarding
have a haematological malignancy had a precursor cancer dwell times might also explain the lower-than-
condition such as MGUS. expected rate of cancer detection.
Because this study represented a prevalence round of
Discussion screening, indolent haematological conditions were
The PATHFINDER study provides early evidence of the likely to have been overrepresented compared with what
feasibility of blood testing to screen for multiple cancers would be detected in later rounds of repeated screening.
with a single test, a departure from the prevailing The high rate of screening participation in the study
framework requiring distinct screening tests for each population might also explain both the number and
cancer type. When the MCED assay was positive and types of cancers diagnosed in the absence of an MCED
cancer was present, the test accurately predicted the signal. For example, eight of the 11 total lung cancers
tumour origin, and half of all cancer diagnoses were diagnosed in this study arose in individuals who did not
made in less than 2 months. When cancer could not be meet USPSTF 2021 criteria for low-dose CT screening.26
confirmed, diagnostic assessments, often with serial Most of these lung cancers were early stage, and all had
scans, continued for a longer duration, typically extending cTAF below the MCED detection threshold. By contrast,
for 4 months. Blood tests and advanced imaging studies the one case of lung cancer detected by MCED testing
were commonly used following a signal detected result. presented at stage III.

www.thelancet.com Vol 402 October 7, 2023 1257


Articles

Oropharyngeal cancer (n=2) Lymphoma (n=12)


Squamous cell carcinoma stage II Follicular lymphoma stage I
Squamous cell carcinoma stage IV Follicular lymphoma stage I
Marginal-zone lymphoma stage I
Lung (n=1)
Hodgkin lymphoma stage I
Adenocarcinoma stage III
Follicular lymphoma stage II
Breast (n=5) Follicular lymphoma stage II
Carcinoma consistent with breast primary RM Follicular lymphoma stage II
Carcinoma consistent with breast primary RM Follicular lymphoma stage II
Carcinoma consistent with breast primary RM Diffuse large B-cell lymphoma stage III
Carcinoma consistent with breast primary RM Mature B-cell neoplasm stage IV
Carcinoma consistent with breast primary RM Diffuse large B-cell lymphoma stage IV
Liver (n=1) Recurrent local cancer, no pathology
No pathology stage I
Intrahepatic bile ducts (n=1)
Adenocarcinoma stage III
Pancreas (n=1)
Adenocarcinoma stage II Waldenstrom macroglobulinaemia (n=2)
Small intestine (n=1) Waldenstrom macroglobulinaemia
Adenocarcinoma stage I Waldenstrom macroglobulinaemia
Colon and rectum (n=2)
Adenocarcinoma stage IV Lymphoid leukaemia (n=2)
No pathology stage IV Chronic B-cell lymphocytic leukaemia
Chronic B-cell lymphocytic leukaemia
Uterus (n=1)
Endometrial adenocarcinoma stage I
Ovary (n=1) Plasma-cell myeloma (n=1)
Serous adenocarcinoma stage III Plasma-cell neoplasm
Of the 29 participants with new cancers,
Prostate (n=2)
14 (48%) were stage I or II
Adenocarcinoma stage IV
Biochemical recurrence, no pathology
Bone (n=1) USPSTF screening
Spindle-cell neoplasm stage II No USPSTF screening

Figure 4: Cancer diagnosed after a positive multicancer early detection result


The 36 cancers diagnosed in the 35 true-positive participants are depicted by a dot in the organ or system in which the cancer was found. Histology details are given
when available. Overall, 26 (74%) of 35 participants had cancers diagnosed that did not have USPSTF recommendations for screening tests. RM=recurrent metastatic
cancer. USPSTF=US Preventive Services Task Force.

The distribution of cancers in participants with a Study strengths include usage of a validated fixed
false-negative result did not have a clear pattern, other assay and algorithm to predict both cancer signal
than the predictable outcome of being enriched for presence and CSO, and recruitment of participants
cancers that are commonly screen-detected such as from various health-care settings in US ambulatory care
breast and colorectal cancers. Most false negatives had practice. The absence of protocol-mandated diagnostic
cTAF below the MCED test limit of detection.24,27–29 algorithms allowed insight into clinician judgement
Because MCED testing technologies are still in early regarding clinical management of a positive MCED test
phases of development, rely on different approaches to result.
cohort ascertainment, and do not have a consistent The study had several limitations. First, access to
gold-standard definition of a true positive, it is infeasible advanced imaging and laboratory testing in the USA is
to compare the sensitivity of different MCED test often limited by insurance coverage, but in this study
methods. was fully supported by the sponsor. Second, the
False-positive results and the potential for generalisability of the cancer detection rate is
overdiagnosis are inherent risks of all screening constrained by the little ethnic, racial, and socioeconomic
methods. In this study, fewer than 1% of participants diversity of the study cohort and the high rate of baseline
had false-positive results, which compares favourably adherence to cancer screening. Similarly, volunteer bias
with recommended single-cancer screening tests.18,30,31 In could have influenced participants’ risk or their
the CCGA study,7,32 we found that cancers with detectable preferences for undergoing comprehensive diagnostic
circulating tumour DNA were more likely to be lethal assessments. The number, types, and stages of cancers
than those with undetectable levels of circulating tumour detected reflect results of an initial screen of undetected
DNA. This finding suggests that MCED testing on the cancers and might not be representative of the results of
basis of circulating tumour DNA technology might long-term screening. Third, the study was done during
minimise the risk of overdiagnosis by preferentially the COVID-19 pandemic, which hindered the ability to
detecting more lethal tumours. However, larger trials obtain diagnostic assessments promptly and might have
with longer-term follow-up are needed to quantify rates lengthened the diagnostic resolution intervals. Fourth,
of overdiagnosis. there was no structured evaluation system to elicit

1258 www.thelancet.com Vol 402 October 7, 2023


Articles

adverse event reports, which might have led to Data sharing


underestimation. Fifth, the study was not designed to The datasets analysed for this study are available upon reasonable
request by email to the corresponding author.
assess acceptability or cost-effectiveness. Finally, the
12-month follow-up period was short, and rates of later Acknowledgments
We acknowledge the contributions to this effort of the patients and their
cancer diagnoses are uncertain. families and the research coordinators and data managers at the
Although this study establishes clinical feasibility of investigational sites. We would like to acknowledge Anne-Renee Hartman
MCED testing, forthcoming results of larger studies for her contributions to the PATHFINDER study concept and design, and
will be required to show clinical utility and effects on Jafi Lipson for her contributions to the PATHFINDER study concept,
design, and implementation. We acknowledge Sana Raoof for her
cancer mortality. Three other studies are evaluating the contribution to the manuscript drafting. This study was sponsored by
refined version of the MCED test developed in GRAIL. We acknowledge Sarah A Prins (GRAIL) for manuscript editing
PATHFINDER in different populations: (1) the and management. We acknowledge Jennifer Hepker (Prescott Medical
PATHFINDER2 cohort study (NCT05155605) is Communications Group, Chicago, IL, USA) and Neva West (NeuroWest
Solutions, Seattle, WA, USA) for medical writing and editorial and
investigating test performance in a larger and more administrative support that was funded by GRAIL. DS was affiliated with
diverse US adult population, (2) the UK’s National Dana Farber Cancer Institute and Harvard Medical School, Boston, MA,
Health Service NHS Galleri study (ISRCTN 91431511) is USA, during the conduct of the study. TMB was affiliated with OHSU
investigating test performance in 140 000 adults Knight Cancer Institute, Portland, OR, USA, during the conduct of the
study. EAK was affiliated with Cleveland Clinic, Cleveland, OH, USA,
randomly assigned to usual cancer screening versus during the conduct of the study.
usual screening supplemented by annual MCED
References
testing,33 and (3) the University of Oxford’s SYMPLIFY 1 Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023.
study (ISRCTN10226380) investigated the use of MCED CA Cancer J Clin 2023; 73: 17–48.
testing for individuals presenting to primary care 2 Hackshaw A, Cohen SS, Reichert H, Kansal AR, Chung KC,
Ofman JJ. Estimating the population health impact of a multi-
doctors with non-specific symptoms such as weight cancer early detection genomic blood test to complement existing
loss or fatigue.34 Results of these studies will contribute screening in the US and UK. Br J Cancer 2021; 125: 1432–42.
evidence regarding the utility of this MCED test in 3 Clarke CA, Hubbell E, Kurian AW, Colditz GA, Hartman AR,
Gomez SL. Projected reductions in absolute cancer-related deaths
specific clinical contexts. from diagnosing cancers before metastasis, 2006–2015.
In conclusion, PATHFINDER establishes the feasibility Cancer Epidemiol Biomarkers Prev 2020; 29: 895–902.
of MCED testing in ambulatory practice and describes 4 Beer TM. Novel blood-based early cancer detection: diagnostics in
development. Am J Manag Care 2020; 26 (suppl): S292–99.
the diagnostic assessments undertaken in response to
5 Liu MC. Transforming the landscape of early cancer detection using
receipt of a positive test suggesting a possible cancer blood tests—commentary on current methodologies and future
diagnosis. This study also provides preliminary estimates prospects. Br J Cancer 2021; 124: 1475–77.
of test performance characteristics and the ultimate 6 Hackshaw A, Clarke CA, Hartman AR. New genomic technologies
for multi-cancer early detection: rethinking the scope of cancer
diagnostic yield of this emerging technology. This screening. Cancer Cell 2022; 40: 109–13.
experience lays the foundation for larger-scale studies to 7 Klein EA, Richards D, Cohn A, et al. Clinical validation of a
investigate the safety, utility, and clinical effectiveness of targeted methylation-based multi-cancer early detection test using
an independent validation set. Ann Oncol 2021; 32: 1167–77.
MCED testing as a cancer screening strategy.
8 Liu MC, Oxnard GR, Klein EA, et al. Sensitive and specific multi-
Contributors cancer detection and localization using methylation signatures in
EAK, CRM, KCC, ML, ETF, and DS conceptualised the study. ML and cell-free DNA. Ann Oncol 2020; 31: 745–59.
ETF provided data curation. EAK, TMB, CHM, ML, ETF, and DS did the 9 Lennon AM, Buchanan AH, Kinde I, et al. Feasibility of blood
formal analysis. EAK, TMB, CHM, LN, CAD, RR, CRM, and DS were testing combined with PET-CT to screen for cancer and guide
study investigators. EAK, TMB, ML, ETF, and DS contributed to intervention. Science 2020; 369: eabb9601.
visualisation of the data and writing of the original draft. All authors 10 Cristiano S, Leal A, Phallen J, et al. Genome-wide cell-free DNA
were involved in project administration, resourcing, and final review and fragmentation in patients with cancer. Nature 2019; 570: 385–89.
editing of the manuscript. EAK, KCC, ML, ETF, and DS accessed and 11 Nadauld LD, McDonnell CH 3rd, Beer TM, et al.
verified the data. All authors had access to the data, reviewed and The PATHFINDER study: assessment of the implementation of an
approved the final version of the manuscript, and were responsible for investigational multi-cancer early detection test into clinical
practice. Cancers (Basel) 2021; 13: 3501.
the decision to submit for publication.
12 National Comprehensive Cancer Network. NCCN clinical
Declaration of interests practice guidelines, detection, prevention, and risk reduction. https://
DS reports an uncompensated advisory role and serving as a study www.nccn.org/guidelines/category_2 (accessed July 11, 2023).
investigator with research funding to Dana Farber Cancer Institute 13 Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov
(GRAIL) and personal fees for editorial service 2022; 12: 31–46.
(Journal of the American Medical Association). TMB reports a consultant 14 Hubbell E, Venn O. Shared cancer signal: evidence from cross-
agreement with GRAIL, AbbVie, Amgen, Arvinas, Astellas Pharma, training. The Early Detection of Cancer Conference (EDCC);
AstraZeneca, Bayer, Bristol Myers Squib, Constellation, Dantari Oct 18–20, 2022.
Pharmaceuticals, GlaxoSmithKline, Janssen, Myovant Sciences, Pfizer, 15 Amin MB, Edge SB, Greene FL, et al. LAJCC cancer staging
Sanofi, and Sapience Therapeutics; and reports stock ownership in manual, 8th edn. New York, NY, USA: Springer Cham, 2017.
Arvinas, Salarius Pharmaceuticals, and Exact Sciences. CHM reports a 16 Chamberlain J, Clifford RE, Nathan BE, Price JL, Burn I. Repeated
consultant agreement with GRAIL. LN reports stock ownership in screening for breast cancer. J Epidemiol Community Health 1984;
Culmination Bio. KCC reports stock ownership in Illumina, Bristol 38: 54–57.
Myers Squib, Gilead, Baxter, and Bayer, and is an employee of GRAIL. 17 Sprague BL, Arao RF, Miglioretti DL, et al. National performance
ETF and ML report stock ownership of Illumina and are employees of benchmarks for modern diagnostic digital mammography: update
GRAIL. EAK is an employee of GRAIL. All other authors declare no from the Breast Cancer Surveillance Consortium. Radiology 2017;
competing interests. 283: 59–69.

www.thelancet.com Vol 402 October 7, 2023 1259


Articles

18 Lehman CD, Arao RF, Sprague BL, et al. National performance 27 Chen X, Dong Z, Hubbell E, et al. Prognostic significance of blood-
benchmarks for modern screening digital mammography: update based multi-cancer detection in plasma cell-free DNA.
from the Breast Cancer Surveillance Consortium. Radiology 2017; Clin Cancer Res 2021; 27: 4221–29.
283: 49–58. 28 Bredno J, Venn O, Chen X, Freese P, Ofman JJ. Circulating tumor
19 Church TR, Black WC, Aberle DR, et al. Results of initial low-dose DNA allele fraction: a candidate biological signal for multicancer
computed tomographic screening for lung cancer. N Engl J Med early detection tests to assess the clinical significance of cancers.
2013; 368: 1980–91. Am J Pathol 2022; 192: 1368–78.
20 Horeweg N, Scholten ET, de Jong PA, et al. Detection of lung cancer 29 Bredno J, Lipson J, Venn O, et al. Tumor area and microscopic
through low-dose CT screening (NELSON): a prespecified analysis extent of invasion to determine circulating tumor DNA fraction in
of screening test performance and interval cancers. Lancet Oncol plasma and detectability of colorectal cancer (CRC). J Clin Oncol
2014; 15: 1342–50. 2020; 38 (suppl): 243.
21 Bartlett EC, Silva M, Callister ME, Devaraj A. False-negative results 30 Pinsky PF, Gierada DS, Black W, et al. Performance of lung-RADS
in lung cancer screening—evidence and controversies. in the National Lung Screening Trial: a retrospective assessment.
J Thorac Oncol 2021; 16: 912–21. Ann Intern Med 2015; 162: 485–91.
22 Menon U, Gentry-Maharaj A, Hallett R, et al. Sensitivity and 31 Alsayid M, Singh MH, Issaka R, et al. Yield of colonoscopy after a
specificity of multimodal and ultrasound screening for ovarian positive result from a fecal immunochemical test OC-Light.
cancer, and stage distribution of detected cancers: results of the Clin Gastroenterol Hepatol 2018; 16: 1593–97.
prevalence screen of the UK Collaborative Trial of Ovarian Cancer 32 Bredno J, Lipson J, Venn O, Aravanis AM, Jamshidi A. Clinical
Screening (UKCTOCS). Lancet Oncol 2009; 10: 327–40. correlates of circulating cell-free DNA tumor fraction. PLoS One
23 US Preventive Services Taskforce. Search results: Published A, B, C 2021; 16: e0256436.
recommendations cancer screening, 2023. https://www. 33 Neal RD, Johnson P, Clarke CA, et al. Cell-free DNA-based multi-
uspreventiveservicestaskforce.org/uspstf/topic_search_ cancer early detection test in an asymptomatic screening population
results?topic_ status=P&grades%5B%5D=A&grades%5B%5D= (NHS-Galleri): design of a pragmatic, prospective randomised
B&grades%5B%5D=C&category%5B%5D=15&type%5B%5D= controlled trial. Cancers (Basel) 2022; 14: 4818.
5&searchterm= (accessed July 11, 2023). 34 Nicholson BD, Oke J, Virdee PS, et al. Multi-cancer early detection
24 Jamshidi A, Liu MC, Klein EA, et al. Evaluation of cell-free DNA test in symptomatic patients referred for cancer investigation in
approaches for multi-cancer early detection. Cancer Cell 2022; England and Wales (SYMPLIFY): a large-scale, observational cohort
40: 1537–1549.e12. study. Lancet Oncol 2023; 24: 733–43.
25 National Comprehensive Cancer Network. NCCN clinical practice
guidelines, treatment by cancer type. https://www.nccn.org/
guidelines/category_1 (accessed July 11, 2023).
26 Krist AH, Davidson KW, Mangione CM, et al. Screening for lung
cancer: US Preventive Services Task Force recommendation
statement. JAMA 2021; 325: 962–70.

1260 www.thelancet.com Vol 402 October 7, 2023

You might also like