23
1 Plasma Cell-free DNA (cfDNA) Assays for Early Multi-cancer Detection: the Circulating Cell-free Genome Atlas (CCGA) Study Minetta C. Liu, MD 1 ; Eric A. Klein, MD 2 ; Earl Hubbell, PhD 3 ; Tara Maddala, PhD 3 ; Alexander M. Aravanis, MD, PhD 3 ; John F. Beausang, PhD 3 ; Darya Filippova, PhD 3 ; Samuel Gross, PhD 3 ; Arash Jamshidi, PhD 3 ; Kathryn Kurtzman, MD 3 ; Ling Shen, PhD 3 ; Nan Zhang, PhD 3 ; Oliver Venn, DPhil 3 ; Jessica Yecies, PhD 3 ; Shilpen Patel, MD 3 ; David A. Smith, MD 4 ; Timothy Yeatman, MD 5 ; Michael Seiden, MD 6 ; Anne-Renee Hartman, MD 3 ; Geoffrey R. Oxnard, MD 7 1 Mayo Clinic, Rochester, MN. 2 Cleveland Clinic, Cleveland, OH. 3 GRAIL, Inc., Menlo Park, CA. 4 Compass Oncology, Vancouver, WA. 5 Gibbs Cancer Center and Research Institute, Spartanburg, SC. 6 US Oncology Research, The Woodlands, TX. 7 Dana Farber Cancer Institute, Boston, MA.

GRAIL - Plasma Cell-free DNA (cfDNA) Assays for Early Multi … · 2018-10-20 · 2 The Mayo Clinic received grant or other funding from Eisai, Genentech, GRAIL, Inc, Janssen, Merck,

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

1

Plasma Cell-free DNA (cfDNA) Assays for Early Multi-cancer Detection: the Circulating

Cell-free Genome Atlas (CCGA) Study

Minetta C. Liu, MD1; Eric A. Klein, MD2; Earl Hubbell, PhD3; Tara Maddala, PhD3; Alexander M. Aravanis, MD, PhD3; John F. Beausang, PhD3; Darya

Filippova, PhD3; Samuel Gross, PhD3; Arash Jamshidi, PhD3; Kathryn Kurtzman, MD3; Ling Shen, PhD3; Nan Zhang, PhD3; Oliver Venn, DPhil3; Jessica Yecies, PhD3; Shilpen Patel, MD3; David A. Smith, MD4; Timothy

Yeatman, MD5; Michael Seiden, MD6; Anne-Renee Hartman, MD3; Geoffrey R. Oxnard, MD7

1Mayo Clinic, Rochester, MN. 2Cleveland Clinic, Cleveland, OH. 3GRAIL, Inc., Menlo Park, CA. 4Compass Oncology, Vancouver, WA. 5Gibbs Cancer Center and Research Institute, Spartanburg,

SC. 6US Oncology Research, The Woodlands, TX. 7Dana Farber Cancer Institute, Boston, MA.

2

The Mayo Clinic received grant or other funding from Eisai, Genentech, GRAIL, Inc, Janssen, Merck, Novartis, Pfizer, Seattle Genetics, and Tesaro for Minetta C. Liu’s research activities. Eric A. Klein is a consultant for GRAIL, Inc., Genomic Health, and GenomeDx. Earl Hubbell, Tara Maddala, Alexander M. Aravanis, John F. Beausang, Darya Filippova, Samuel Gross, Arash Jamshidi, Kathryn Kurtzman, Ling Shen, Nan Zhang, Oliver Venn, Jessica Yecies, Shilpen Patel, and Anne-Renee Hartman are employees of GRAIL, Inc., with options to hold stock in the company. Michael Seiden is an employee of, and shareholder in, McKesson Corporation. Geoffrey R. Oxnard is an advisory board member and consultant for Inivata, an honorarium recipient from Guardant Health, Sysmex, and BioRad, and a consultant for DropWorks, AstraZeneca, and GRAIL, Inc.

Disclosures

3

Cancer Mortality Increases with Later Stage

*Cancer specific survival data from SEER18 ages 50+ diagnosed 2006-2015. Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database: Incidence - SEER 18 Regs Research Data, Nov 2017 Sub. **Includes intrahepatic bile duct.

4

CCGA is a Prospective, Longitudinal Cohort Study Designed for Cancer Detection

Blood samples(all participants)

Tissue samples(cancer only)

~15,000 participants

70% with cancer30% without

142 Sites

Targeted sequencing cfDNA, WBCs

Whole-genome sequencing cfDNA, WBCsTargeted & whole-genome bisulfite sequencing cfDNA

Follow-up for 5 yrsVital status & cancer status

Whole-genome sequencing of tumor tissue

People with cancer: Data on treatment, recurrence, mortalityPeople without cancer: Data on cancer diagnosis + treatment, recurrence, mortality; or remain cancer-free

cfDNA, cell-free deoxyribonucleic acid; WBC, white blood cell.

5

CCGA: Discovery, Training, and Validation for a Multi-Cancer Test

Training Set: 1,785Clinically Locked*

Test Set: 1,010Clinically Locked*

~15,000 planned participants

70% cancer : 30% non-cancer

12,200 reserved for future studies

2,800 participants:Prespecified case-control

substudy1,628 cancer; 1,172 non-cancer

*5 participants not clinically locked were excluded.

FPI: 08/2016

6

CCGA: Prespecified Case-Control Substudy of 2,800 Participants

Training Set: 1,785Clinically Locked*

Test Set: 1,010Clinically Locked*

1,733 Clinically Evaluable● 984 Cancer

● 878 stage I-IV● 580 Non-Cancer● 169 Non-Cancer Assay Controls

980 Clinically Evaluable● 576 Cancer

● 478 stage I-IV● 368 Non-Cancer● 36 Non-Cancer Assay Controls

1,406 Analyzable with Assay Data ● 845 Cancer

● 539 with Tumor Tissue ● 561 Non-Cancer

834 Analyzable with Assay Data ● 472 Cancer

● 220 with Tumor Tissue ● 362 Non-Cancer

● Eligibility criteria (3%)

● Stage 0 or missing stage (6%)● Other clinical reasons (<1%)● Unevaluable assay data for

≥1 assays (3%)● Non-cancer assay controls

(10%)

● Eligibility criteria (3%)

● Stage 0 or missing stage (10%)● Other clinical reasons (0%)● Unevaluable assay data for ≥1

assays (1%)● Non-cancer assay controls (4%)

Samples excluded due to:Samples excluded due to:

*5 participants not clinically locked were excluded.

7

Comparable Cancer and Non-Cancer Groups

Training TestCancer* Non-Cancer Cancer* Non-Cancer

Total, n (%) 984 580 576 368Age, Mean ± SD (years) 61 ± 12 60 ± 13 62 ± 12 59 ± 14Sex (%)

Female 697 (71%) 452 (78%) 363 (63%) 238 (65%)Race/Ethnicity (%)

White, Non-Hispanic 846 (86%) 489 (84%) 475 (82%) 312 (85%)African American 67 (7%) 47 (8%) 40 (7%) 25 (7%)

Hispanic, Asian, Other 71 (7%) 44 (8%) 61 (11%) 31 (8%)Smoking Status (%)

Never-smoker 484 (49%) 330 (57%) 290 (50%) 185 (50%)BMI

Normal/Underweight 266 (27%) 156 (27%) 162 (28%) 86 (23%)Overweight 319 (32%) 184 (32%) 190 (33%) 126 (34%)

Obese 398 (40%) 240 (41%) 224 (39%) 155 (42%)

*Cancer types by training/test: Breast (410/201), lung (127/47), prostate (74/58), colorectal (51/46), renal (29/18), uterine (28/9), pancreas (27/23), esophageal (25/8), lymphoma (25/22), head & neck (21/12), ovarian (21/7), hepatobiliary (15/16), melanoma (15/12), cervical (14/11), multiple myeloma (14/21), leukemia (13/16), thyroid (13/10), bladder (12/3), gastric (12/15), anorectal (7/3), and unknown primary/other (22/18).

Clinically evaluable cancer and non-cancer groups were comparable with respect to age, sex, race/ethnicity, and BMI

8

Stage Distribution and Method of Diagnosis were Consistent in Training and Test Sets

Cancer,Training Set (n=984)

Cancer, Test Set (n=576)

Overall Clinical Stage (n, %)0* 56 (6%) 34 (6%)

I 300 (30%) 165 (29%)II 249 (25%) 142 (25%)

III 165 (17%) 76 (13%)IV 164 (17%) 95 (16%)

Non-Informative** 50 (5%) 64 (11%)Method of Dx (n, %)

Diagnosed by Screening§ 354 (36%) 202 (35%)Diagnosed by Clinical Presentation¶ 630 (64%) 373 (65%)

*DCIS/CIS. **Staging information not available. §Percent screen-detected in training/test sets for breast cancer: 58%/58%, colorectal cancer: 29%/37%, lung cancer: 18%/15%, prostate cancer: 91%/90%, and other cancers 4%/4%. ¶Clinical presentation includes all cancers not detected by screening (ie, detected symptomatically or as incidental findings).

Broad distribution of stages in training and in test sets

9

Multiple Tumor Types Represented in the CCGA Cohort

Training Testing

Num

ber o

f Par

ticip

ants

400

250

200

100

0

Tumor Type

150

10

Prototype Sequencing Assays Used to Comprehensively Characterize Cancer-Specific cfDNA Signals

● All major somatic and epigenetic cfDNA features characterized

WGS

Targeted

WGBS

Input Interference Final Features Classifierscf

DN

Acf

DN

Acf

DN

AW

BC g

DNA

WBC

gDN

A

● 30X depth

● 507 gene panel

● 60,000X depth

● 3,000X unique coverage

● Bisulfite sequencing

● 30X depth

● Aging● Biological

variation

● Variants derived from WBCs

● SCNA signals derived from WBCs

● Fragment-level CpG methylation status

● Non-synonymous SNVs/indels

● cfDNA Somatic copy number

WGS Classifier

TargetedClassifier

cfDNA, cell-free deoxyribonucleic acid; WBC, white blood cell; WGS, whole-genome sequencing; WGBS, whole-genome bisulfite sequencing.

WGBSClassifier

11CONFIDENTIAL & PROPRIETARY

● Non-tumor WBC-matched cfDNA somatic variants (SNVs/indels) accounted for, on average:○ 98% of all variants in non-cancer group○ 71% in cancer group

Majority of cfDNA Variants Are WBC-Matched Clonal Hematopoiesis

Age (years)

● Number of WBC variants is positively associated with age in cancer and non-cancer groups

● Majority of CH variants were low variant allele frequency (<1.0%) and private1

● Accounting for CH using targeted methods is critical for high specificity

1Swanton C et al. J Clin Oncol 36, 2018 (suppl; abstr 12003).cfDNA, cell-free DNA; WBC, white blood cell; SNV, single-nucleotide variant; indel, insertion or deletion; CH, clonal hematopoiesis.

Cancer Non-Cancer

12

High Specificity (>99%) is Feasible5-year follow-up will enable identification of participants who are subsequently diagnosed

575No notable cancer-like signal* (test negative or

absent)

2No cancer identifiedFollow-up ongoing

*Notable cancer-like signal defined as ≥2 assays with significant abnormalities compared to the typical non-cancer population, or known cancer drivers present with ≥1 significant assay abnormality.

Training:580

Control Participants

5 (<1%) Notable cancer-like

signal* present

3 Confirmed cancer

Endometrial IIOvarian III Lung IV

Test:368

Control Participants

365No notable cancer-like signal* (test negative or

absent)

3No cancer identifiedFollow-up ongoing

3 (<1%) Notable cancer-like

signal* present

13

Consistent Results Across Assays and Between Training and Test Sets

Training

Stage IV

117

245

Targeted

Methylation

WGS

Sensitivity Reported at 98% Specificity

● Depicted here: cancers with >50% signal in training: HR-negative breast, colorectal, esophageal, head & neck, hepatobiliary, lung, lymphoma, ovarian, and pancreatic cancers, and multiple myeloma.

131

64

Stage I-IIITest

Training

Test

0% 20% 40% 60% 80% 100%

NSubgroup

Signal was also consistent in low-signal cancers (<10% in training on any of the three assays: prostate, thyroid, gastric, melanoma).cfDNA, cell-free deoxyribonucleic acid; WBC, white blood cell; WGS, whole-genome sequencing; WGBS, whole-genome bisulfite sequencing.

14

Early- and Late-Stage Cancers Detected in the Test Set

0% 20% 40% 60% 80% 100%

Sensitivity (Methylation Score) Reported at 98% Specificity

Cancer Type NBreast (HR-)

Colorectal

Lung

193

2910

2719

1 Pham D et al. J Clin Oncol 36, 2018 (suppl; abstr 6504).

● Assays detected later-stage cancers (Stage IV) in cancers with screening paradigms

● Signal also observed in participants with early-stage cancers (Stage I-III)

Stage I-III Stage IV

15

Early- and Late-Stage Cancers Detected in the Test Set

0% 20% 40% 60% 80% 100%

Sensitivity (Methylation Score) Reported at 98% Specificity

Cancer Type NBreast (HR-)

Colorectal

Esophageal

Hepatobiliary

Ovarian

Lung

Pancreas

Head & Neck

Lymphoma

Multiple Myeloma*

193

2910

86

52

2719

814

135

8

7

*Multiple myeloma onlyincludes stages I-III

1 Pham D et al. J Clin Oncol 36, 2018 (suppl; abstr 6504).

● Assays detected later-stage cancers (Stage IV) in cancers with screening paradigms

● Signal also observed in participants with early-stage cancers (Stage I-III)

○ 45% of cancers were stage I-II

● Many of these cancers do not have a screening paradigm, or the screening paradigm is not well-adopted (eg, lung1)

Stage I-III Stage IV

75

16

Early- and Late-Stage Cancers Detected in the Test Set

0% 20% 40% 60% 80% 100%

Sensitivity (Methylation Score) Reported at 98% Specificity

Cancer Type NBreast (HR-)

Colorectal

Esophageal

Hepatobiliary

Ovarian

Lung

Pancreas

Head & Neck

Lymphoma

Multiple Myeloma*

193

2910

86

52

2719

814

135

8

7

*Multiple myeloma onlyincludes stages I-III

● Assays detected later-stage cancers (Stage IV) in cancers with screening paradigms

● Signal also observed in participants with early-stage cancers (Stage I-III)

○ 45% of cancers were stage I-II

● Many of these cancers do not have a screening paradigm, or the screening paradigm is not well-adopted (eg, lung1)

● Many of these cancers are associated with a >50% 5-year cancer-specific mortality2

Stage I-III Stage IV

75

1 Pham D et al. J Clin Oncol 36, 2018 (suppl; abstr 6504).25-year cancer-specific mortality rates for persons aged 50-79 from SEER18, 2010-2014; https://seer.cancer.gov.

17

HR-Negative Breast Cancer Detection by Stage in the Test Set

Targeted MethylationWGS

Sensitivity Reported at 98% Specificity

Overall*

Stage I-II

NSubgroup

22

18

*Of the 4 stage III-IV HR- breast cancers, 2 were detected by each assay. Of the 2 stage III-IV TNBC breast cancers, 1 was detected by all 3 assays.

0% 20% 40% 60% 80% 100%

17

15

0% 20% 40% 60% 80% 100%

NHR-Negative Breast Cancer Triple-Negative Breast Cancer

● Signal was detected in early-stage (Stage I-II) as well as later-stage (Stage III-IV) HR-negative breast cancers, including TNBC

● 68% (15/22) of HR-negative breast cancers were Stage I or II TNBC

WGS, whole-genome sequencing; TNBC, triple-negative breast cancer.

18

Lung Cancer Detection by Smoking Status and Histologic Subtype in the Test Set

● 93% (43/46) of participants with lung cancer were ever-smokers● Signal was detected in ever-smokers, as well as in never-smokers

○ Of 3 never-smokers, 2 were detected by the methylation assay, 1 by the WGS assay, and 3 by the targeted assay

0% 20% 40% 60% 80% 100%

Sensitivity Reported at 98% Specificity

43Ever-smoker,

Stage I-IV

NSubgroup Smoking StatusTargeted

Methylation

WGS

WGS, whole-genome sequencing.

19

Lung Cancer Detection by Smoking Status and Histologic Subtype in the Test Set

● 93% (43/46) of participants with lung cancer were ever-smokers● Signal was detected in ever-smokers, as well as in never-smokers

○ Of 3 never-smokers, 2 were detected by the methylation assay, 1 by the WGS assay, and 3 by the targeted assay

0% 20% 40% 60% 80% 100%

Sensitivity Reported at 98% Specificity

43Ever-smoker,

Stage I-IV

NSubgroup Smoking StatusTargeted

Methylation

WGS

● Signal was also detected consistently across histologic subtypes (Stage I-IV methylation assay reported):○ 100% (5/5) of SCLC cases were detected○ 65% (11/17) of SCC cases were detected○ 60% (12/20) of adenocarcinoma cases were detected

WGS, whole-genome sequencing.

20

Colorectal Cancer Detection by Stage and Location

● Signal was detected in early-stage (Stage I-III) as well as later-stage (Stage IV) colorectal cancers

0% 20% 40% 60% 80% 100%

Sensitivity Reported at 98% Specificity

Targeted MethylationWGS

39

29

10

Overall

Stage I-III

Stage IV

NSubgroupColorectal Cancer Overall ● Signal was also consistently detected

across locations in colorectal cancer (methylation assay reported):

○ Rectum: 63% (10/16)

○ Left: 63% (5/8)

○ Right: 60% (6/10)

○ Other: 60% (3/5)

WGS, whole-genome sequencing.

21

Signal Observed in Participants with Screen-Detected Cancers and Cancers Detected by Clinical Presentation*

● Signal was generally stronger in cancers detected by clinical presentation

Colorectal

Overall I-III IV

HR- Breast Lung

0% 20% 40% 60% 80% 100%

Sensitivity (Methylation Score) Reported at 98% Specificity

0% 20% 40% 60% 80% 100% 0% 20% 40% 60% 80% 100%

1211

1

2718

9

9

9

13

103

7

7

39

2019

N N N

Screen

ClinicalPres.

*Cancers detected by “clinical presentation” include all cancers not detected by screening (ie, detected symptomatically or as incidental findings).

22

● A cfDNA-based blood test detected multiple cancers across all stages, including at early stages when treatment may be more effective

○ Test set confirmed the signal observed in the training set

○ >99% specificity is feasible

■ Targeted methods require accounting for clonal hematopoiesis

○ High detection of cancers with high mortality and that lack screening paradigms

● This approach is thus promising as a multi-cancer detection test, including for early-stage cancers

● Further assay and clinical development in large-scale clinical studies, including CCGA, is ongoing

Conclusions

cfDNA, cell-free DNA.

23

● Study participants who graciously donated their time, energy, and specimens

● CCGA investigators and collaborators for advice, enrolling participants, and collecting data and specimens○ Principal Investigators from sites enrolling >35 participants in this preplanned substudy: Minetta C. Liu (Mayo

Clinic, MN); David Thiel (Mayo Clinic, FL); Rosanna Lapham, MD (Spartanburg Regional Health Services, SC); Donald Richards, MD, PhD (TOPA Tyler, TX, US Oncology Network); Nicholas Lopez, MD (Baptist Health Paducah, KY); Daron G. Davis, MD (Baptist Health Lexington, KY); Mohan Tummala, MD (Mercy Springfield, MO); Peter Yu, MD (Hartford Hospital, CT); Wangjian Zhong, MD (Baptist Health, Louisville, KY); Alexander Parker, MD (Mayo Clinic Jacksonville, FL); Kristi McIntyre, MD (TOPA Dallas Presbyterian, TX, US Oncology Network); Fergus Couch (Mayo Clinic Rochester, MN); Robert Seigel (Bon Secours Greenville, SC); Allen L. Cohn, MD (Rocky Mountain Cancer Center Hale Parkway Denver, CO, US Oncology Network); Alan H. Bryce (Mayo Clinic Phoenix, AZ).

● Advisors and Scientific Advisory Board members for their helpful feedback and advice

● The many GRAIL teams who have worked and continue to work on this study

Acknowledgements