sensitive detection of minimal residual disease in …...2020/03/13  · 6 mark fleharty2, greg...

38
1 TITLE: Sensitive detection of minimal residual disease in patients treated for early- 1 stage breast cancer 2 3 AUTHORS: Heather A. Parsons 1 *, Justin Rhoades 2 *, Sarah C. Reed 2 *, Gregory 4 Gydush 2 , Priyanka Ram 1 , Pedro Exman 1 , Kan Xiong 2 , Christopher C. Lo 2,3 , Tianyu Li 4 , 5 Mark Fleharty 2 , Greg Kirkner 1 , Denisse Rotem 2 , Ofir Cohen 1,2 , Fangyan Yu 5 , Mariana 6 Fitarelli-Kiehl 5 , Ka Wai Leong 5 , Melissa Hughes 1 , Shoshana M. Rosenberg 1 , Laura C. 7 Collins 6 , Kathy D. Miller 7 , Brendan Blumenstiel 2 , Lorenzo Trippa 4 , Carrie Cibulskis 2 , 8 Donna Neuberg 4 , Matthew DeFelice 2 , Samuel S. Freeman 2 , Niall J. Lennon 2 , Nikhil 9 Wagle 1 , Gavin Ha 8 , Daniel G. Stover 9 , Atish D. Choudhury 1 , Gad Getz 2 , Eric P. Winer 1 , 10 Matthew Meyerson 1,2 , Nancy U. Lin 1 , Ian E. Krop 1 , J. Christopher Love 2,10 , G. Mike 11 Makrigiorgos 2,5 , Ann Partridge 1 , Erica Mayer 1 , Todd R. Golub 2,11,12 , Viktor A. 12 Adalsteinsson 2,10 13 14 1 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA 15 2 Broad Institute of MIT and Harvard, Cambridge, MA, USA 16 3 Boston University School of Public Health, Boston, MA, USA 17 4 Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, 18 Boston, MA, USA 19 5 Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and 20 Women’s Hospital, Harvard Medical School, Boston, MA 21 6 Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical 22 School, Boston, MA, USA 23 Research. on May 26, 2020. © 2020 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Upload: others

Post on 24-May-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

1

TITLE: Sensitive detection of minimal residual disease in patients treated for early-1

stage breast cancer 2

3

AUTHORS: Heather A. Parsons1*, Justin Rhoades2*, Sarah C. Reed2*, Gregory 4

Gydush2, Priyanka Ram1, Pedro Exman1, Kan Xiong2, Christopher C. Lo2,3, Tianyu Li4, 5

Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 6

Fitarelli-Kiehl5, Ka Wai Leong5, Melissa Hughes1, Shoshana M. Rosenberg1, Laura C. 7

Collins6, Kathy D. Miller7, Brendan Blumenstiel2, Lorenzo Trippa4, Carrie Cibulskis2, 8

Donna Neuberg4, Matthew DeFelice2, Samuel S. Freeman2, Niall J. Lennon2, Nikhil 9

Wagle1, Gavin Ha8, Daniel G. Stover9, Atish D. Choudhury1, Gad Getz2, Eric P. Winer1, 10

Matthew Meyerson1,2, Nancy U. Lin1, Ian E. Krop1, J. Christopher Love2,10, G. Mike 11

Makrigiorgos2,5, Ann Partridge1, Erica Mayer1, Todd R. Golub2,11,12, Viktor A. 12

Adalsteinsson2,10 13

14

1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA 15

2Broad Institute of MIT and Harvard, Cambridge, MA, USA 16

3Boston University School of Public Health, Boston, MA, USA 17

4Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, 18

Boston, MA, USA 19

5Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and 20

Women’s Hospital, Harvard Medical School, Boston, MA 21

6Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical 22

School, Boston, MA, USA 23

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 2: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

2

7Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA 24

8Division of Public Health Services, Fred Hutchinson Cancer Research Center, Seattle, 25

WA, USA 26

9Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, 27

OH, USA 28

10Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA 29

11Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA 30

12Howard Hughes Medical Institute, Chevy Chase, MD, USA 31

* These authors contributed equally 32

33

Running title: Sensitive detection of minimal residual disease 34

35

Keywords: minimal residual disease; breast cancer, circulating tumor DNA (ctDNA), 36

molecular profiling, liquid biopsy 37

38

Financial support: The authors acknowledge the Gerstner Family Foundation for its 39

generous support. This study was also supported in part by the NBTII Foundation and 40

The Bridge Project, a partnership between the Koch Institute for Integrative Cancer 41

Research at MIT and the Dana-Farber/Harvard Cancer Center (DF/HCC), The Terri 42

Brodeur Breast Cancer Foundation, the Friends of Dana-Farber, the Cheryl Tessler 43

Solit Foundation, Breast Cancer Research Foundation (to N.U.L.), National 44

Comprehensive Cancer Network/Pfizer Collaborative Grant Program (to N.U.L.), 45

Fashion Footwear Association of New York, National Cancer Institute Specialized 46

Program of Research Excellence in Breast Cancer (NCI P50CA168504), and 47

R01CA221874 (to GMM). DFCI trial 05-055 was supported by Genentech/Roche. 48

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 3: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

3

49

Co-Corresponding authors: 50

Heather Parsons 51

Dana-Farber Cancer Institute 52

450 Brookline Avenue 53

Boston, MA 02215 54

Tel: 617-632-3800 55

[email protected] 56

57

Viktor A. Adalsteinsson 58

Eli and Edythe L. Broad Institute of MIT and Harvard 59

415 Main Street 60

Cambridge, Massachusetts 02142 61

Tel: 617-714-7971 62

[email protected] 63

64

Conflict of Interest statement: H.A. Parsons reports advisory role for Foundation 65

Medicine and institutional research support from Puma Biotechnology. D. Rotem reports 66

a consulting role and equity with Celsuis Pharmaceuticals (spouse) and has filed 67

patents (US2018 0051319A1, WO2017136508A1, WO2018183908, WO2018183921) 68

with Dana-Farber Cancer Institute and Broad Institute (spouse). L. Trippa reports 69

consulting for Galera Therapeutics. D. Neuberg reports stock ownership in Madrigal 70

Pharmaceuticals; research support from Pharmacyclics; and consulting (no fee) for 71

Clear Creek Bio. S. Freeman reports a patent application filed with Broad Institute. N.J. 72

Lennon reports advisory roles at Genturi, Inc. and New England Biolabs, Inc.. G. Ha 73

reports a patent application filed with Broad Institute. A.D. Choudhury reports research 74

funding from Bayer. M. Meyerson reports consulting for OrigiMed; royalties from 75

LabCorp; research support and royalties from Bayer; and research support from Ono 76

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 4: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

4

and Janssen. N.U. Lin reports research funding from Genentech, Seattle Genetics, 77

Pfizer and Merck; and consulting/advisory board roles at Seattle Genetics, Puma and 78

Daichii. E.L. Mayer reports consulting for Eisai, Pfizer, Lilly, Novartis, Roche, and 79

Context Therapeutics; and institutional research support from Pfizer and Myriad. T.R. 80

Golub reports advisor role (paid) at Foundation Medicine, GlaxoSmithKline and 81

Sherlock Biosciences. V. Adalsteinsson reports a patent application filed with Broad 82

Institute and is a member of the scientific advisory board of AGCT GmbH, which was 83

not involved in this study. The remaining authors report no conflicts of interest. 84

85

Word count: (excluding references and figure legends): 4890/5000 86

References: 22 87

Figures: 4 88

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 5: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

5

STATEMENT OF TRANSLATIONAL RELEVANCE 89

Optimizing systemic therapy for patients with early-stage breast cancer remains 90

challenging. An ultrasensitive blood test for minimal residual disease (MRD) could 91

identify survivors who might benefit from additional systemic treatment versus de-92

escalation. Here we present an ultrasensitive blood test for MRD and evaluate its 93

clinical validity in breast cancer. We show that it is 100-fold more sensitive than ddPCR 94

at limiting dilution when tracking 488 mutations but its sensitivity depends on the 95

number of mutations available to track in cfDNA. We examined MRD detection in 142 96

patients treated for early-stage breast cancer at post-op and one year after surgery. 97

MRD detection was strongly associated with distant recurrence and provided significant 98

lead time to recurrence. Clinical sensitivity was highest in patients who had most 99

mutations identified from their tumor biopsies to track in cfDNA. Our approach could 100

inform future testing of earlier therapeutic intervention in patients who may otherwise 101

develop metastatic recurrence. 102

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 6: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

6

ABSTRACT 103

Purpose: Existing cell-free DNA (cfDNA) methods lack the sensitivity needed for 104

detecting minimal residual disease (MRD) following therapy. We developed a test for 105

tracking hundreds of patient-specific mutations to detect MRD with a 1000-fold lower 106

error rate than conventional sequencing. 107

Experimental Design: We compared the sensitivity of our approach to digital droplet 108

PCR (ddPCR) in a dilution series, then retrospectively identified two cohorts of patients 109

who had undergone prospective plasma sampling and clinical data collection: 16 110

patients with ER+/HER2- metastatic breast cancer (MBC) sampled within six months 111

following metastatic diagnosis and 142 patients with stage 0-III breast cancer who 112

received curative-intent treatment with most sampled at surgery and one year post-op. 113

We performed whole-exome sequencing of tumors and designed individualized MRD 114

tests, which we applied to serial cfDNA samples. 115

Results: Our approach was 100-fold more sensitive than ddPCR when tracking 488 116

mutations, but most patients had fewer identifiable tumor mutations to track in cfDNA 117

(median 57, range 2-346). Clinical sensitivity was 81% (n=13/16) in newly diagnosed 118

MBC, 23% (n=7/30) at post-op and 19% (n=6/32) at one year in early-stage disease, 119

and highest in patients with the most tumor mutations available to track. MRD detection 120

at one year was strongly associated with distant recurrence (HR=20.8 [95%CI: 7.3-121

58.9]). Median lead time from first positive sample to recurrence was 18.9 months 122

(range: 3.4-39.2 months). 123

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 7: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

7

Conclusions: Tracking large numbers of individualized tumor mutations in cfDNA can 124

improve MRD detection, but its sensitivity is driven by the number of tumor mutations 125

available to track. 126

127

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 8: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

8

INTRODUCTION 128

Each year, more than 600,000 people worldwide die of breast cancer, largely due to 129

metastatic recurrence. Although these recurrences can occur early, the majority in 130

patients with hormone receptor-positive breast cancer occur more than 5 years after 131

initial diagnosis, with risk extending for decades.1,2 Systemic recurrence likely arises 132

from micrometastatic disease present at initial diagnosis, but undetectable by imaging 133

or conventional blood tests. Once overt metastatic breast cancer develops, it is 134

generally incurable. Appropriate adjuvant systemic therapy can substantially reduce the 135

risk of cancer recurrence3,4; however, current clinical tools are imperfect in identifying 136

which patients require adjuvant systemic therapy and assessing in real-time which 137

therapies are achieving their intended therapeutic effects. The current American Society 138

of Clinical Oncology guidelines recommend 10 years of adjuvant endocrine therapy for 139

women presenting with stage II or III, estrogen receptor-positive (ER+) breast cancer, 140

based upon randomized trial data demonstrating, on average, an absolute improvement 141

in distant disease-free survival of 1.4-1.9% compared to 5 years of endocrine therapy.5-7 142

More sensitive methods to detect micrometastatic disease may allow therapy to be 143

escalated in a subset of higher risk patients, while sparing other patients potentially 144

adverse effects of unnecessary treatments. 145

146

Recent studies have shown that liquid biopsies can detect minimal residual disease 147

(MRD) in multiple cancer types8-12 including breast,13-15 by tracking tumor mutations in 148

circulating cell-free DNA (cfDNA). However, in these previous studies, clinical sensitivity 149

has been limited at the early post-operative time points, at which treatment decisions 150

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 9: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

9

are typically made, and the lead time prior to clinical presentation of overt metastatic 151

disease has been relatively short. There is a need for more sensitive liquid biopsies, 152

with greater dynamic range, to identify patients with MRD sooner, with sufficient lead 153

time prior to overt metastatic disease such that additional curative-intent therapy could 154

be effective. 155

156

One major challenge in detecting MRD from peripheral blood is that a typical blood draw 157

samples only a few thousand copies of each gene.16 This means that existing methods, 158

which track one or few mutations, will be unable to detect MRD when the fraction of 159

cancerous cfDNA in the bloodstream is lower than twice the inverse of the number of 160

copies of each gene in a given sample, which we call the genomic equivalent (GE) limit 161

(Fig. 1A). Collecting more blood is not always feasible; however, tracking many 162

mutations per patient may increase the likelihood that cfDNA fragments containing the 163

desired targeted mutations are captured when tumor fraction (i.e. the proportion of 164

cfDNA derived from tumor rather than non-cancerous tissues)17-19 in blood is lower than 165

the GE limit (Figs. 1A,B).20 166

167

One practical limitation for tracking more mutations is that each patient’s cancer is 168

unique and harbors few mutations in common with other patients.16 Either a substantial 169

number of digital droplet PCR (ddPCR) assays or deep sequencing of very large gene 170

panels would be required to afford broad patient coverage, and even then, would only 171

cover a limited number of mutations per patient (Fig. 1C, Supplementary Fig. 1). 172

Individualized assays are therefore needed to track many tumor mutations in most 173

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 10: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

10

patients, and groups are beginning to do this, but existing methods lack sufficient 174

specificity10,21. We estimate that error rates need to be 100-fold lower (e.g. ~1 x 10-6) to 175

reliably track hundreds of individualized mutations, because this involves scanning 176

hundreds of thousands to millions of bases for single mutations. 177

178

Beyond limiting the false detection of MRD, we reason that it will be important to 179

calibrate differences in detection power among individualized MRD tests, particularly 180

when the test result is negative for MRD. Existing liquid biopsy assays do not compute 181

detection power, which obscures whether the test was powered to detect MRD in a 182

patient if the tumor fraction in cfDNA is low. As multiple biological and technical factors 183

affect detection power from a liquid biopsy, we hypothesized that creating a single 184

metric for detection power, which encompasses the potential variation, would help to 185

better understand the significance of a negative MRD test. 186

187

In this study, we established that tracking hundreds of patient-specific tumor mutations 188

enables reliable detection of MRD at tumor fractions significantly lower than the GE 189

limit. We also developed a power calculation for each individual MRD test. We 190

hypothesized that tracking multiple mutations would enable earlier detection of 191

metastatic disease. In the first part of our study, we applied our newly developed assay 192

to patients with estrogen receptor-positive (ER+) breast cancer within six months 193

following metastatic diagnosis. The primary objective was to determine the sensitivity of 194

the assay in patients at time of metastatic recurrence. In the second part of our study, 195

we applied the assay to track mutation fingerprints in plasma and identified breast 196

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 11: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

11

cancer recurrence following surgery and systemic therapy. The primary objectives were 197

to determine the clinical sensitivity at post-op and one year following surgery (which 198

reflect key points in patient care at which treatment decisions are made) and the lead 199

interval between detection of ctDNA in plasma and clinical detection of overt metastatic 200

disease. 201

202

METHODS 203

204

Patients and Samples 205

All patients provided written informed consent to allow the collection of blood and/or 206

tumor tissue and analysis of clinical and genetic data for research purposes. Patients 207

with MBC were prospectively identified for enrollment into tissue analysis and banking 208

cohorts (Dana-Farber Cancer Institute [DFCI] protocol identifiers 05-246, 09-204). From 209

within this cohort we retrospectively identified patients with ER-positive, HER2-negative 210

MBC with plasma isolated from 20cc blood in Streck tubes and tissue sampling within 211

six months of diagnosis. In the curative-intent cohort, we included patients participating 212

in two clinical studies (DFCI IRB-approved protocols 05-055 and 06-169) given receipt 213

of curative-intent treatment and available biospecimens (Supplementary Figure 2B). 214

Plasma was derived from 10-20cc whole blood in EDTA tubes. From protocol 06-169, 215

we included the first 99 patients enrolled with breast cancer diagnosis at age <35. From 216

05-055, all study patients were included, if tissue and plasma samples were available. 217

At the time of analysis and data cut off, all patients had completed adjuvant local and 218

systemic therapy aside from hormonal therapy, and median follow up had reached 7.1 219

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 12: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

12

years. Fresh whole blood (10–20 cc) from appropriately consented healthy donors was 220

obtained through Research Blood Components and Boston Biosciences. HapMap cells, 221

NA12878 and NA19238 were purchased from Coriell. This research was conducted in 222

accordance with the provisions of the Declaration of Helsinki, and the U.S. Common 223

Rule. 224

225

Sample Processing 226

Collection and processing of blood samples followed the same protocol as previously 227

described.17 Germline DNA (gDNA) was extracted from buffy coat or whole blood and 228

sheared using a Covaris Ultrasonicator. cfDNA and gDNA libraries were constructed 229

using duplex UMI adapters (IDT) with up to 20ng of DNA input. Hybrid selection (HS) 230

was performed using patient specific probe panels (Twist Biosciences and IDT, 231

Supplementary Methods). 232

233

Sequencing and Data Analysis 234

Samples were sequenced on either a HiSeq 4000 or HiSeq X (Illumina). Exome 235

sequencing was used to identify patient-specific single nucleotide variants (SNVs) as 236

previously described.17 Patient specific SNVs were used to design custom MRD tests, 237

which were subsequently applied to cfDNA and gDNA libraries. MRD detection, 238

including detection limit and tumor fraction, was determined using a custom pipeline 239

(Supplementary Methods). 240

241

Statistical Analysis 242

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 13: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

13

This study was designed to assess primarily the association between MRD status and 243

distant disease recurrence and secondarily the time between MRD detection and 244

clinical metastasis. We determined to identify at least 120 patients to give 80% power to 245

detect a hazard ratio of 3 for distant recurrence-free survival (DRFS). The associations 246

between MRD overall status and demographic/clinical factors were evaluated via Chi-247

square test for categorical variables and Wilcoxon rank-sum test for continuous 248

variables. Both the univariate and multivariate analysis (MVA) of DRFS were conducted 249

using the Cox proportional hazard model. MRD status was measured at 3 time points 250

post-operatively. To cope with the time-variant nature of MRD status (a patient’s MRD 251

status may vary across time), univariate analysis of DRFS was done in the frame of 252

landmark analysis at post-operative and year 1 blood sample draw. Patients who had 253

distant recurrence prior to the landmark time point, post-operation or year 1, were 254

removed from the analysis. Multivariate analysis of DRFS was conducted using MRD 255

status as a time-dependent covariate. A patient was coded as in the MRD-negative 256

group until the patient tested positive at one time point and then was switched to the 257

positive group. MRD status, stage, grade, and HR status were included in the MVA. All 258

analyses were conducted using SAS 9.2. Kaplan Meier curves were made using the 259

Survival package of R (version R2.15). 260

RESULTS 261

Establishment of a novel, scalable assay for detection of circulating tumor DNA 262

We have developed an ultrasensitive blood test for MRD. Our approach involved 263

tracking hundreds of individualized tumor mutations, identified from a patient’s tumor 264

tissue, in the cfDNA from a blood draw. We applied whole-exome sequencing to define 265

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 14: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

14

up to several hundred mutations from each patient’s tumor. To limit potential errors, we 266

employed strict criteria to select somatic single nucleotide variants (SNVs) to track using 267

duplex sequencing22 in cfDNA. We further required detection of ≥ 2 mutations for a 268

cfDNA sample to be called MRD-positive and excluded any mutations also found in a 269

patient’s own germline DNA (Supplementary Methods, Supplementary Fig. 3). 270

271

We first investigated whether tracking many mutations enabled detection of MRD at 272

tumor fractions lower than the GE limit. To model blood draws from cancer patients, we 273

created large volume dilutions of sheared genomic DNA from two well-characterized 274

cancer cell lines and obtained 20 ng DNA samples comprising ~6k GEs. We applied a 275

single mutation ddPCR test and observed detection down to tumor fraction 1/1k as 276

expected for a GE limit of 1/3k (Fig. 2A, Supplementary Fig. 4). Whereas, MRD testing 277

using a fingerprint comprised of 97 and 488 mutations exhibited reliable detection down 278

to tumor fraction 1/10k and 1/100k, respectively, with all lower dilutions and negative 279

controls being MRD-negative. The proportion of MRD-positive samples at each dilution 280

aligned with the expected MRD-positive samples based on calculated detection power. 281

We computed the median detection power for the 1/100k samples with 488 mutation 282

fingerprints to be 82% and found 17/20 (85%) to be MRD-positive (Fig. 2A-B, 283

Supplementary Fig. 5). Observed tumor fractions also correlated with expected values 284

(r2=0.94, Fig. 2C, Supplementary Fig. 6, see Methods), including for another dilution 285

series of cancer patient and healthy donor cfDNA (Supplementary Fig. 7), and further 286

testing confirmed high specificity in healthy donors (Supplementary Fig. 8). These 287

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 15: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

15

results demonstrate that tracking of many mutations enables reliable detection of MRD 288

at tumor fractions that are up to 100-fold lower than the GE limit. 289

290

We then evaluated the reproducibility and cost effectiveness of patient-specific assays 291

by examining the performance of 142 patient-specific panels applied to 370 samples. 292

We found patient-specific assays with duplex sequencing technology provide high 293

efficiency (87.4% +/- 15.5% bases on bait) and uniformity (coefficient of variation 294

[CV]=0.23 +/- 0.12) in target capture; exhibit error rates, which are on average 1000-fold 295

lower than conventional sequencing (1.3E-06 +/- 2.3E-06); detect the same DNA 296

duplexes in replicate capture (92% shared) and sequencing (89% shared); and saturate 297

recovery of DNA duplexes in many samples with a median of 1.46M reads per mutation 298

site (Supplementary Figs. 9, 10). Of note, we found our MRD test results to be 299

reproducible in benchmarking data sets (Fig. 2, Supplementary Fig. 7, Supplementary 300

Fig. 10) but did not perform repeated testing of patient samples in our study. 301

Considering these parameters and the mutational diversity in breast cancer, de novo 302

assays are feasible for most patients and require less sequencing than predefined 303

panels of recurrently mutated genes in breast cancer (due to less genomic territory 304

targeted) to achieve higher detection power for MRD (Fig. 1C). 305

306

MRD detection in newly diagnosed, hormone receptor-positive MBC 307

Given our intent to detect micrometastatic disease, we first explored the clinical 308

sensitivity of our MRD assay in patients with radiologically-overt newly diagnosed MBC. 309

We focused on patients with hormone receptor-positive (HR+) and human epidermal 310

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 16: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

16

growth factor receptor 2-negative (HER2-) breast cancer, in which recurrences may 311

occur years to decades after initial treatment. We reasoned that understanding the 312

detectability of MRD and corresponding tumor fraction in cfDNA would help to establish 313

upper bounds for detection of MRD at earlier stages in care. 314

315

We applied our assay to 35 samples (range 1-4 per patient) from 16 patients with newly 316

diagnosed, HR+ MBC, who had undergone tumor biopsy and cfDNA blood draw within 317

six months following diagnosis (Supplementary Fig. 11A). Of note, we used patients’ 318

metastatic tumor biopsies to define mutation fingerprints, tracking a median of 51.5 319

SNVs (range 6 – 120). Overall, 30/35 (86%) samples from 13/16 (81%) patients had 320

detectable cancer cfDNA (or MRD-positive), including 7/7 (100%) samples taken prior to 321

initiation of therapy, 11/11 (100%) samples taken from patients not benefiting from 322

therapy, and 12/17 (69%) samples taken on therapy from patients experiencing clinical 323

benefit, defined as continuation of treatment for at least 6 months from time of initiation 324

(Fig. 3A-B, Supplementary Fig. 11B). Application of the same MRD tests to cfDNA from 325

healthy donors yielded MRD-negative results in all cases (Supplementary Fig. 11A). 326

Tumor fractions were highest when patients were sampled prior to therapy (median 327

0.044, range 0.002-0.17) and remained high when patients were on ineffective therapy 328

(median 0.028, range 0.0012-0.35, Supplementary Fig. 11B). By contrast, tumor 329

fractions were lower in patients sampled when on effective therapy (median 0.00025, 330

range 0-0.0015). Notably, we observed significant declines in tumor fraction when 331

patients were sampled prior to, and then again while on effective therapy 332

(Supplementary Fig. 11C) as has previously been seen in metastatic prostate cancer.2 333

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 17: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

17

334

We also investigated whether we could have tracked fewer mutations and found that 335

MRD could have been detected using fewer mutations in the cfDNA samples which had 336

the highest tumor fractions: those which were taken prior to therapy or on ineffective 337

therapy. However, tracking fewer mutations would have failed to detect MRD in many of 338

the cfDNA samples which had the lowest tumor fractions: those which were collected on 339

effective therapy (Supplementary Fig. 11D). This suggests that our large fingerprint 340

approach is most useful when seeking to detect trace levels of ctDNA, at tumor fractions 341

below the GE limit. 342

343

Our assay failed to detect MRD in 5 samples from 3 patients with known metastatic 344

disease. As these samples were taken while on effective therapy, when tumor fraction is 345

expected to be low, we explored whether limited detection power could explain these 346

results. We computed the tumor fraction at which detection power is 90% (which we call 347

the detection limit) for all samples from patients experiencing clinical benefit (Fig. 3B, 348

Methods). We found the 5 false-negative samples to exhibit a median detection limit of 349

0.00029 (range 0.00014-0.00082), which is 3-fold higher – i.e. worse – than the 12 350

MRD-positive samples from the rest of patients experiencing clinical benefit (median 351

0.0000848, range 0.0000299-0.000265). We cannot rule out the possibility that these 352

samples actually had no tumor DNA in them, but our calculated detection limits suggest 353

that it would have been less likely to detect MRD below ~1/1k in these 5 samples. In all, 354

our results indicate that detection limits should be in the range of 1/10k or better to 355

detect MRD prior to clinical metastatic recurrence. 356

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 18: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

18

357

MRD detection after definitive treatment for breast cancer 358

To determine the predictive power of MRD testing and associated lead time to 359

recurrence in patients treated for early-stage breast cancer, we identified a patient 360

cohort representative of the major breast cancer subtypes (HR+/HER2-, HER2+, and 361

HR-/HER2- [triple-negative breast cancer, TNBC]), with appropriately long-term follow 362

up given the potential for late recurrence (Supplementary Table 1). Our cohort included 363

142 patients who had been treated for stage 0-III breast cancer, had postoperative 364

blood and plasma samples available, and were monitored for distant recurrences for up 365

to 13 years (Supplementary Fig. 2A-B). All patients had biopsy-proven breast cancer: 366

86 (61%) with HR+/HER2- disease, 31 (22%) with TNBC, and 25 (18%) with HER2+ 367

disease. Overall, 130 (92%) received either neoadjuvant or adjuvant chemotherapy, and 368

108 (76%) received adjuvant endocrine therapy (Supplementary Table 1); 104 (73%) 369

received adjuvant radiation treatment. In terms of stage, 3 (2%) patients presented with 370

stage 0 disease, 32 (23%) with stage I, 68 (48%) with stage II, and 39 (27%) with stage 371

III breast cancer at diagnosis. All patients were treated with curative intent surgery. 372

Thirty-seven (26%) patients (24 [65%] ER+/HER2-, 7 [19%] HER2+, 6 [16%] TNBC) 373

experienced distant recurrence, at a median follow up across the cohort from surgery to 374

time of death or last contact of 7.1 years. Median time to distant recurrence was 32 375

months for those patients with recurrent disease. 376

377

From these 142 patients’ primary tumors, we selected a median of 53 SNVs (range 2 – 378

346) to track. We identified 271 postoperative plasma samples from these patients, 379

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 19: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

19

collected at a median of 3.53 months (range 0.23-8.43) after surgery and at 380

approximately year 1 (median 14.2 months, range 6.77-21.7) and year 4 (median 54.97 381

months, range 42.43-71.27) from surgery (Supplementary Table 2). Most patients had a 382

postoperative (111, 78%) or year 1 (122, 86%) sample available but far fewer had a 383

year 4 (38, 27%) sample. Forty-seven (33%) and 20 (14%) patients with postoperative 384

and year 1 samples, respectively, had their samples collected while on active 385

chemotherapy. Of the 37 patients who experienced distant recurrence, 30 and 32 had a 386

postoperative and year 1 sample available, respectively, but only 3 patients had a year 387

4 sample. Median plasma volume was 3.4 mL (range 0.4-5.3), and a median of 19.9 ng 388

cfDNA (range 2.6-21.6) was allocated for MRD testing. We then created individualized 389

assays targeting a median of 57 mutations (range 2-346) per patient, identified via 390

whole-exome sequencing of primary tumor tissue and germline DNA from whole blood. 391

Of note, the number of identifiable tumor mutations to track in each patient’s cfDNA was 392

much lower than we had used in our benchmarking (n=488 mutations) to achieve 393

1/100k detection limit from 20ng DNA (Fig 2A, Supplementary Fig. 2C). Nonetheless, 394

we applied the assays to all plasma, and germline DNA samples when available. 395

396

We first examined the relationship between postoperative MRD status and distant 397

recurrence via a post-operative landmark analysis, and found that detection of MRD 398

soon after curative surgery was highly predictive of distant relapse (hazard ratio=5.1 399

[95% confidence interval, CI: 2.0-12.7], Fig. 4A, Supplementary Fig. 12). However, we 400

also hypothesized that because adjuvant systemic therapy reduces the risk of breast 401

cancer recurrence,3 we would expect to detect MRD in some patients in whom cancer 402

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 20: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

20

did not recur due to effective adjuvant treatment. Indeed, 3 patients had a single, 403

postoperative MRD-positive sample, but remained recurrence free at last follow up. Two 404

of these patients were undergoing adjuvant therapy at the time of blood sampling. The 405

third patient sample was drawn 7 days after surgery. In all 3 of these patients, 406

subsequent plasma samples were MRD-negative (Supplementary Fig. 13). Based on 407

the post-op blood test, the positive predictive value (PPV) for distant recurrence was 408

0.70 (n=7/10) and clinical specificity was 0.96 (n=78/81). The negative predictive value 409

(NPV) was 0.77 (n=78/101) and clinical sensitivity was 0.23 (n=7/30). Of note, MRD 410

was not detected in the 6 patients who experienced local-only recurrence 411

(Supplementary Fig. 14). 412

413

Reasoning that MRD status after completion of all local therapy and chemotherapy 414

might better predict for distant recurrence, we conducted a landmark analysis at one 415

year following surgery and found all patients (6/6) with detectable MRD experienced 416

recurrence (hazard ratio=20.8 [95% CI: 7.3-58.9]) at a median of 21.7 months (range 417

10.1–32.2) (Fig. 4B, Supplementary Fig. 12). Based on the year 1 sample, PPV for 418

distant recurrence was 1.00 (n=6/6) and clinical specificity was 1.0 (n=90/90). NPV was 419

0.78 (n=90/116) and clinical sensitivity was 0.19 (n=6/32). We then evaluated the 420

subset of patients with available year 4 samples (38, 27%) to assess for MRD status 421

and risk of late recurrence. Of the 38 patients who underwent year 4 sampling, only 3 422

went on to experience recurrence. We detected MRD in one of three patients with 423

recurrence 18.9 months prior to clinical metastatic diagnosis (the other two samples 424

were underpowered with poor detection limits of 0.0015 and 0.0023), and in no patients 425

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 21: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

21

without breast cancer recurrence. Finally, in a multivariate model, MRD remained highly 426

statistically significant for risk of distant recurrence (Supplementary Table 2), 427

independent of stage, subtype, and age at diagnosis. 428

429

We next explored potential factors which could have affected our clinical sensitivity for 430

MRD detection across the cohort. We first reasoned that duration of time between MRD 431

test and time of recurrence could impact MRD detection, as patients in our study 432

experienced recurrence up to 10 years later. Indeed, the median interval from last blood 433

test to recurrence was 11.5 months for patients in whom we detected MRD, versus 23.8 434

months for those in whom we failed to detect MRD (Fig. 4C-i). 435

436

Given that the technical sensitivity of our approach was driven by number of mutations 437

tracked (Fig 2A, Supplementary Fig. 15A), and that most patients in our study had 438

limited mutations identified from whole-exome sequencing of their tumor biopsies, we 439

also explored the association between number of mutations tracked and clinical 440

sensitivity. Expectedly, clinical sensitivity was highest among patients in whom we 441

tracked the most mutations (Fig 4C-ii) and whose MRD tests yielded the best detection 442

limits (Fig 4C-iii). Reassuringly, tracking more mutations was not associated with 443

increased MRD detection in the 105 patients who did not experience distant recurrence 444

(Supplementary Fig. 15B). We were also more likely to detect mutations in the cfDNA 445

that had higher allele fractions and cancer cell fractions (CCF) in the tumor 446

(Supplementary Fig. 16A-B). Had we tracked fewer mutations per patient, we would 447

have failed to detect MRD in 36% (n=5/14) of samples (Supplementary Fig. 15C). 448

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 22: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

22

However, tumor fractions in cfDNA of patients who experienced recurrence (median 449

0.00042, range 0.000056 - 0.041) were close to the detection limits, suggesting that we 450

may need to track even more mutations per patient in future studies. 451

452

Interestingly, among the patients with most mutations tracked, we observed a long 453

median lead time to clinical recurrence of 18.9 months (range 3.4 – 39.2, Fig 4D, 454

Supplementary Table 3). Such lead times could enable early intervention, long before 455

patients develop metastatic recurrence. 456

457

DISCUSSION 458

In this study we present a novel approach to MRD tracking, based on the premise that 459

tracking many, rather than one or a handful of mutations, inherently increases assay 460

sensitivity, and assay personalization maximizes sensitivity in a disease without multiple 461

recurrent mutations. We applied the assay to a group of breast cancer patients treated 462

with curative intent, and detected MRD in many patients prior to presentation of 463

metastatic disease. 464

465

The concept of tracking more mutations to improve sensitivity is not new, but existing 466

methods have had insufficient specificity to track many individualized mutations. To our 467

knowledge, this is the first study to track hundreds of personalized mutations 468

simultaneously in patients with cancer, using an approach with sufficient analytical 469

specificity (~1x10-6). Our approach enables reliable detection well below the GE limit, as 470

compared to following a single mutation via ddPCR. Tracking MRD from blood in cancer 471

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 23: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

23

patients is challenging from a diagnostic perspective, as cancer recurrence happens in 472

the future, and no current standard metric exists against which to measure a true 473

positive or negative test. Here, we quantify two key aspects: the detection limit, and 474

tumor fraction. Detection limit determination allows for a clear understanding of whether 475

a specific test is more likely negative because of assay performance or as a result of a 476

lack of MRD. 477

478

We followed patients for up to 13 years from definitive surgery. This is specifically 479

important in the context of HR+/HER2- breast cancer, where recurrence may occur 480

years to decades from initial treatment. Interestingly, our assay had a median lead time 481

(the time from a positive test to metastatic diagnosis) of 18.9 months, and up to 39 482

months, without compromising clinical specificity. This is significantly longer than what 483

has been seen in prior studies such as Garcia-Murillas et al (median 10.7 months) and 484

Coombes et al (median 8.9 months) which tracked 1 - 16 mutations per patient, but is 485

based on a small number of patients and could be a function of longer follow up.13,14 If 486

additional prospective evaluation confirms a lead time of this magnitude, our assay 487

could offer an opportunity to detect and treat MRD long before the development of overt 488

metastatic disease, with the goal of offering curative treatment to patients for whom it is 489

not currently an option. Testing whether treatment of MRD can, in fact, be curative will 490

be an important next step. 491

492

One limitation of our study was related to the infrequent sampling of blood relative to the 493

duration of clinical follow up. Whereas prior studies have collected blood samples every 494

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 24: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

24

6 months (or more frequently) and reported high rates of MRD detection based on any 495

of many time points tested over a multi-year period being positive13,14, we sampled 496

patients’ blood relatively infrequently and focused MRD testing on key time points at 497

which treatment decisions are typically made. For instance, most patients in our study 498

had a single post-op and year one blood sample following completion of all local therapy 499

and adjuvant chemotherapy, and only 3 patients who experienced recurrence had a 500

year 4 time point. When restricted to the first post-op time point, our clinical sensitivity 501

(23%, n=7/30) was similar to Garcia-Murillas et al 2015 (19%, n=7/37).14 Garcia-Murillas 502

et al 2019 did not compute clinical sensitivity at post-op but stated that most patients 503

who experienced recurrence did not have an initial MRD+ test at post-op.15 Meanwhile, 504

Coombes et al reported higher sensitivity in the first post-op sample tested per patient 505

but this was later for most patients, up to 3 years after completion of adjuvant 506

chemotherapy.13 Comparison of clinical sensitivity and lead time among studies is 507

challenging, however, due to numerous potential confounders such as differences in 508

patient populations, treatments, timing of sampling, and duration of follow-up. 509

510

Another limitation of our study was that most patients had too few mutations identified 511

via whole-exome sequencing of their tumor biopsies, to fully leverage the large 512

fingerprint approach in cfDNA. Whereas our benchmarking had shown that tracking 488 513

mutations achieved reliable detection of 1/100k dilution from 20ng DNA (Fig 2), in 514

contrast to 1/1k for ddPCR, our detection limits for most MRD tests were constrained to 515

1/1k – 1/10k due to insufficient numbers of mutations having been identified from 516

patients’ tumor biopsies (Fig 3C, 4C-iii). Indeed, clinical sensitivity was highest in 517

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 25: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

25

patients with most mutations tracked (Fig 4C-ii). We reason that future efforts will 518

require whole-genome sequencing (instead of whole-exome sequencing) to identify 519

more mutations to track in all patients. 520

521

Other limitations of our study were that (i) most patients in our cohort had HR+/HER2- 522

disease, the most common breast cancer subtype, which meant we were unable to 523

draw meaningful conclusions about the variations of MRD detection by subtype, (ii) for 524

some patients, the last blood sample was taken nearly a decade before metastatic 525

recurrence, (iii) volumes of plasma processed were small, meaning the number of GEs 526

sampled was lower, and (iv) blood samples were collected and processed for plasma 527

many years ago, before cfDNA-specific protocols existed, and variable processing may 528

have led to cfDNA degradation. 529

530

Breast cancer is a uniquely challenging disease. Widespread use of screening and 531

effective systemic therapies have led to improved rates of cure. However, most breast 532

cancer-related deaths are due to recurrence of breast cancers that arise from 533

microscopic residual disease. Current clinico-pathologic risk assessments for patients 534

with breast cancer are imperfect and result in both undertreatment of patients in whom 535

MRD exists but is not detected, and overtreatment of patients who may have been 536

cured with more limited therapy. An accurate MRD test could offer clinicians a more 537

precise method for differentiating between patients who need further treatment and 538

those who do not. Furthermore, clinicians treating patients with other solid tumors face 539

similar challenges, and our findings might be generalizable beyond breast cancer. 540

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 26: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

26

541

Of note, modern adjuvant breast cancer clinical trials often enroll thousands of patients, 542

are expensive, and take many years to complete. An accurate MRD test could 543

potentially transform this approach. A validated MRD test could be used to select 544

eligible participants for clinical trials by identifying patients at highest risk of recurrence, 545

in whom the study of new therapies would be appropriate. This approach would have a 546

number of benefits. It would limit toxicity of additional therapies to those truly at risk. 547

Trials could include substantially fewer participants, putting fewer patients at risk and 548

costing much less to complete. MRD clearance could be tested as a surrogate clinical 549

trial endpoint. If validated, clinical trials could be completed in a shorter time. Finally, an 550

MRD test could potentially address overtreatment, though we acknowledge that doing 551

this would require an assay with exceptional sensitivity. Otherwise, an insufficiently 552

sensitive assay could lead to inappropriate treatment de-escalation in patients who 553

remain at risk of recurrence. 554

555

In conclusion, we found that tracking a customized fingerprint of up to hundreds of 556

mutations is a promising approach to identifying MRD in breast cancer patients, but 557

leveraging the power of this approach requires identifying enough tumor mutations to 558

track in cfDNA. We anticipate that tracking even more mutations may increase assay 559

sensitivity without compromising specificity, with studies currently underway. We look 560

forward to applying this assay to prospective studies in breast cancer to prove clinical 561

utility of MRD testing. 562

563

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 27: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

27

DATA AVAILABILITY STATEMENT 564

Data have been deposited into dbGaP under accession code phs001977.v1.p1. 565

ACKNOWLEDGEMENTS 566

The authors would like to first and foremost acknowledge the courageous patients and 567

their families for their contributions to this study. The authors acknowledge the Gerstner 568

Family Foundation for its generous support. This study was also supported in part by 569

the NBTII Foundation and The Bridge Project, a partnership between the Koch Institute 570

for Integrative Cancer Research at MIT and the Dana-Farber/Harvard Cancer Center 571

(DF/HCC), The Terri Brodeur Breast Cancer Foundation, the Friends of Dana-Farber, 572

the Cheryl Tessler Solit Foundation, Breast Cancer Research Foundation (to N.U.L.), 573

National Comprehensive Cancer Network/Pfizer Collaborative Grant Program (to 574

N.U.L.), Fashion Footwear Association of New York, National Cancer Institute 575

Specialized Program of Research Excellence in Breast Cancer (NCI P50CA168504). 576

DFCI trial 05-055 was supported by Genentech/Roche. This study used cell line 577

samples from the NINDS Repository; NINDS Repository sample numbers 578

corresponding to the samples used are: NA12878 and NA19238. 579

580

The prevalence of mutations in various cancer types was determined using data 581

generated by the TCGA Research Network: https://www.cancer.gov/tcga. 582

583

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 28: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

28

REFERENCES 584

1. Ferlay J, Colombet M, Soerjomataram I, et al. Estimating the global cancer 585

incidence and mortality in 2018: GLOBOCAN sources and methods. Int J 586

Cancer. 2019;144(8):1941-1953. 587

2. Pan H, Gray R, Braybrooke J, et al. 20-Year Risks of Breast-Cancer Recurrence 588

after Stopping Endocrine Therapy at 5 Years. N Engl J Med. 2017;377(19):1836-589

1846. 590

3. Early Breast Cancer Trialists' Collaborative G, Peto R, Davies C, et al. 591

Comparisons between different polychemotherapy regimens for early breast 592

cancer: meta-analyses of long-term outcome among 100,000 women in 123 593

randomised trials. Lancet. 2012;379(9814):432-444. 594

4. Early Breast Cancer Trialists' Collaborative G, Dowsett M, Forbes JF, et al. 595

Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-596

analysis of the randomised trials. Lancet. 2015;386(10001):1341-1352. 597

5. Goss PE, Ingle JN, Pritchard KI, et al. Extending Aromatase-Inhibitor Adjuvant 598

Therapy to 10 Years. N Engl J Med. 2016;375(3):209-219. 599

6. Mamounas EP, Bandos H, Lembersky BC, et al. Abstract S1-05: A randomized, 600

double-blinded, placebo-controlled clinical trial of extended adjuvant endocrine 601

therapy (tx) with letrozole (L) in postmenopausal women with hormone-receptor 602

(+) breast cancer (BC) who have completed previous adjuvant tx with an 603

aromatase inhibitor (AI): Results from NRG Oncology/NSABP B-42. Cancer Res. 604

2017;77(4_Suppl):S1-05. 605

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 29: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

29

7. Burstein HJ, Lacchetti C, Anderson H, et al. Adjuvant Endocrine Therapy for 606

Women With Hormone Receptor-Positive Breast Cancer: ASCO Clinical Practice 607

Guideline Focused Update. J Clin Oncol. 2019;37(5):423-438. 608

8. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects 609

minimal residual disease and predicts recurrence in patients with stage II colon 610

cancer. Sci Transl Med. 2016;8(346):346ra392. 611

9. Chaudhuri AA, Chabon JJ, Lovejoy AF, et al. Early Detection of Molecular 612

Residual Disease in Localized Lung Cancer by Circulating Tumor DNA Profiling. 613

Cancer Discov. 2017;7(12):1394-1403. 614

10. Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts 615

early-stage lung cancer evolution. Nature. 2017;545(7655):446-451. 616

11. Christensen E, Birkenkamp-Demtroder K, Sethi H, et al. Early Detection of 617

Metastatic Relapse and Monitoring of Therapeutic Efficacy by Ultra-Deep 618

Sequencing of Plasma Cell-Free DNA in Patients With Urothelial Bladder 619

Carcinoma. J Clin Oncol. 2019;37(18):1547-1557. 620

12. Reinert T, Henriksen TV, Christensen E, et al. Analysis of Plasma Cell-Free DNA 621

by Ultradeep Sequencing in Patients With Stages I to III Colorectal Cancer. 622

JAMA Oncol. 2019;5(8):1124-1131. 623

13. Coombes RC, Page K, Salari R, et al. Personalized Detection of Circulating 624

Tumor DNA Antedates Breast Cancer Metastatic Recurrence. Clin Cancer Res. 625

2019;25(14):4255-4263. 626

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 30: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

30

14. Garcia-Murillas I, Schiavon G, Weigelt B, et al. Mutation tracking in circulating 627

tumor DNA predicts relapse in early breast cancer. Sci Transl Med. 628

2015;7(302):302ra133. 629

15. Garcia-Murillas I, Chopra N, Comino-Mendez I, et al. Assessment of Molecular 630

Relapse Detection in Early-Stage Breast Cancer. JAMA Oncol. 2019 Aug 1 631

[Epub ahead of print]. DOI: 10.1001/jamaoncol.2019.1838. 632

16. Volik S, Alcaide M, Morin RD, Collins C. Cell-free DNA (cfDNA): Clinical 633

Significance and Utility in Cancer Shaped By Emerging Technologies. Mol 634

Cancer Res. 2016;14(10):898-908. 635

17. Adalsteinsson VA, Ha G, Freeman SS, et al. Scalable whole-exome sequencing 636

of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun. 637

2017;8(1):1324. 638

18. Choudhury AD, Werner L, Francini E, et al. Tumor fraction in cell-free DNA as a 639

biomarker in prostate cancer. JCI Insight. 2018;3(21). 640

19. Stover DG, Parsons HA, Ha G, et al. Association of Cell-Free DNA Tumor 641

Fraction and Somatic Copy Number Alterations With Survival in Metastatic 642

Triple-Negative Breast Cancer. J Clin Oncol. 2018;36(6):543-553. 643

20. Newman AM, Lovejoy AF, Klass DM, et al. Integrated digital error suppression 644

for improved detection of circulating tumor DNA. Nat Biotechnol. 2016;34(5):547-645

555. 646

21. McDonald BR, Contente-Cuomo T, Sammut SJ, et al. Personalized circulating 647

tumor DNA analysis to detect residual disease after neoadjuvant therapy in 648

breast cancer. Sci Transl Med. 2019;11(504). 649

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 31: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

31

22. Schmitt MW, Kennedy SR, Salk JJ, Fox EJ, Hiatt JB, Loeb LA. Detection of ultra-650

rare mutations by next-generation sequencing. Proc Natl Acad Sci U S A. 651

2012;109(36):14508-14513. 652

653

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 32: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

32

FIGURE CAPTIONS 654

655

Figure 1. A) Overview of MRD testing including why tracking many mutations is 656

important, particularly when there is a low fraction of cancerous cfDNA in the 657

bloodstream, B) Theoretical detection power for varied numbers of mutations tracked 658

per patient, C) Cumulative fraction of patients harboring varied numbers of mutations 659

(and in turn, the predicted detection limits based on Fig. 1B) using breast cancer gene 660

sequencing panels versus individualized MRD tests. See methods. 661

662

Figure 2. Serial dilution benchmarking of our MRD test for 100 and 500 mutations, 663

versus digital droplet PCR for a single mutation, using 20ng admixtures of sheared 664

genomic DNA from two well-characterized cell lines. A) Detection power for a range of 665

tumor fractions using our MRD assay and an equivalent ddPCR single mutation assay. 666

B) Fraction of replicate dilutions detected by our MRD assay and an equivalent ddPCR 667

assay as well as the median detection power for those samples. C) Tumor fraction 668

estimation benchmarking for several replicates across a range of tumor fractions. 669

670

Figure 3. MRD testing in patients with newly diagnosed, HR+ MBC. A) Patient timelines 671

with samples collected less than six months after metastatic recurrence. B) Number of 672

MRD-positive and MRD-negative samples including corresponding tumor fractions. 673

Many patients had multiple blood draws, both pre-treatment and on-treatment. C) 674

Comparison of detection limit for MRD- vs MRD+ samples from patients on treatment 675

and experiencing clinical benefit. 676

677

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 33: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

33

Figure 4. MRD testing in early stage breast cancer. A-B) Kaplan-Meier curves showing 678

distant recurrence-free survival for patients treated for early stage breast cancer and in 679

whom MRD was detected or not at the post-op and year 1 (Y1) time points. Post-op and 680

Y1 blood samples were drawn at a median of 3.6 months (0.23 - 8.43) and 14.3 months 681

(6.77 - 21.70) after surgery respectively. C) Comparison of patients with distant 682

recurrence in which MRD was detected or not at the post-op or Y1 time points. D) 683

Timelines for patients with distant recurrence that were detected at post-op or Y1 time 684

points. End points represent distant recurrence. 685

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 34: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 35: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 36: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 37: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005

Page 38: Sensitive detection of minimal residual disease in …...2020/03/13  · 6 Mark Fleharty2, Greg Kirkner1, Denisse Rotem2, Ofir Cohen1,2, Fangyan Yu5, Mariana 7 Fitarelli-Kiehl 5 ,

Published OnlineFirst March 13, 2020.Clin Cancer Res   Heather A Parsons, Justin Rhoades, Sarah C Reed, et al.   treated for early-stage breast cancerSensitive detection of minimal residual disease in patients

  Updated version

  10.1158/1078-0432.CCR-19-3005doi:

Access the most recent version of this article at:

  Manuscript

Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/early/2020/03/13/1078-0432.CCR-19-3005To request permission to re-use all or part of this article, use this link

Research. on May 26, 2020. © 2020 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 13, 2020; DOI: 10.1158/1078-0432.CCR-19-3005