throbber
Downloaded from http://aacrjournals.org/cancerdiscovery/article-pdf/11/12/2968/3013378/2968.pdf by guest on 07 September 2023
`
`REVIEW
`
`Detecting Liquid Remnants of Solid Tumors:
`Circulating Tumor DNA Minimal Residual
`Disease
`
`Everett J. Moding1,2, Barzin Y. Nabet1,2,3, Ash A. Alizadeh1,3,4, and Maximilian Diehn1,2,3
`
`ABSTRACT
`
`Growing evidence demonstrates that circulating tumor DNA (ctDNA) minimal resid-
`ual disease (MRD) following treatment for solid tumors predicts relapse. These
`results suggest that ctDNA MRD could identify candidates for adjuvant therapy and measure response
`to such treatment. Importantly, factors such as assay type, amount of ctDNA release, and technical and
`biological background can affect ctDNA MRD results. Furthermore, the clinical utility of ctDNA MRD
`for treatment personalization remains to be fully established. Here, we review the evidence support-
`ing the value of ctDNA MRD in solid cancers and highlight key considerations in the application of this
`potentially transformative biomarker.
`Significance: ctDNA analysis enables detection of MRD and predicts relapse after definitive treatment
`for solid cancers, thereby promising to revolutionize personalization of adjuvant and consolidation
`therapies.
`
`INTRODUCTION
`Nearly two thirds of patients with solid tumors present
`with locoregional disease and are amenable to curative thera-
`pies (1). Surgery, radiotherapy, systemic therapy, or a combi-
`nation of these approaches can achieve disease remission in
`the majority of these cases when using conventional measures
`of response, such as functional body imaging. However, in
`a significant subset of patients, small numbers of remnant
`tumor cells, termed minimal residual disease (MRD), can
`persist at levels below the detection threshold of imaging or
`physical exam and ultimately lead to disease relapse (2, 3).
`Systemic therapy delivered after surgery (i.e., adjuvant ther-
`apy) or radiotherapy (i.e., consolidation therapy) has been shown
`to improve long-term survival in multiple cancer types (4–7),
`
`1Stanford Cancer Institute, Stanford University School of Medicine,
`Stanford, California. 2Department of Radiation Oncology, Stanford
`University School of Medicine, Stanford, California. 3Institute for Stem
`Cell Biology and Regenerative Medicine, Stanford University School of
`Medicine, Stanford, California. 4Division of Oncology, Department of
`Medicine, Stanford University, Stanford, California.
`Current address for B.Y. Nabet: Department of Oncology Biomarker Devel-
`opment, Genentech, South San Francisco, California.
`Corresponding Authors: Maximilian Diehn, Department of Radiation
`Oncology, Stanford University, 875 Blake Wilbur Drive, Stanford, CA
`94305-5847. Phone: 650-721-1550; E-mail: diehn@stanford.edu; and
`Ash A. Alizadeh, Division of Oncology, Department of Medicine, Stanford
`University, 875 Blake Wilbur Drive, Stanford, CA 94305-5847. Phone:
`650-725-0120; E-mail: arasha@stanford.edu
`Cancer Discov 2021;11:2968–86
`doi: 10.1158/2159-8290.CD-21-0634
`©2021 American Association for Cancer Research
`
`providing strong clinical evidence that eradication of MRD can
`improve rates of cure. However, for most solid cancers where
`adjuvant/consolidation therapy is currently part of the standard
`of care, the magnitude of benefit from adjuvant therapies is
`modest. This is likely in part because a significant subset (and
`sometimes the majority) of patients who receive adjuvant thera-
`pies are already cured by the preceding local therapy (4, 6, 8).
`For several hematologic malignancies, detection of MRD
`via flow cytometry for tumor cells or quantitative molecular
`techniques for patient-specific aberrations has long been
`established as a poor prognostic factor following induction
`therapy. Accordingly, modification of therapy based on the
`presence of MRD has become a standard of care for acute
`lymphoblastic leukemia, acute promyelocytic leukemia, and
`chronic myelogenous leukemia (9, 10). Given such actionabil-
`ity of MRD in blood neoplasms, biomarkers that can simi-
`larly identify which patients with solid tumors harbor MRD
`could also have significant utility in personalizing adjuvant/
`consolidation therapy. However, until recently, approaches to
`detect MRD in solid cancers have lacked the sensitivity and
`specificity required for clinical application (11).
`Recent work focused on circulating tumor DNA (ctDNA)
`has produced promising results, suggesting that this analyte
`could serve as a generalizable biomarker for MRD in solid
`cancers. Tumors release DNA into the blood that can be iso-
`lated from plasma collected via routine blood draws (12–14).
`Despite generally representing a small fraction of cell-free DNA
`(cfDNA) in blood plasma, ctDNA can be detected via polymer-
`ase chain reaction (PCR) or next-generation sequencing (NGS)
`assays targeting tumor-specific mutations, structural variants,
`copy-number alterations, and epigenetic features (15, 16).
`
`2968 | CANCER DISCOVERY DECEMBER 2021
`
`AACRJournals.org
`
`Personalis EX2023
`
`

`

`Downloaded from http://aacrjournals.org/cancerdiscovery/article-pdf/11/12/2968/3013378/2968.pdf by guest on 07 September 2023
`
`REVIEW
`
`Multiple commercial platforms have been developed based on
`these approaches, including Natera Inc.’s Signatera (34–36),
`ArcherDX’s personalized cancer monitoring assay (37), and
`Inivata’s RaDaR assay (38). Although beyond the scope of
`this review, numerous other PCR amplicon-based NGS meth-
`ods have been used for ctDNA analysis in the past and have
`been reviewed in detail elsewhere (39–41).
`
`Hybridization Capture–Based NGS
`DNA enrichment using hybrid capture with biotinylated
`oligonucleotides allows sequencing of larger targeted panels
`with better uniformity of coverage (42, 43). Capture-based
`NGS approaches such as CAncer Personalized Profiling by
`deep sequencing (CAPP-Seq) preserve cfDNA fragment size
`information and can also incorporate UMIs to minimize
`technical background (18). Capture panels can be used as “off-
`the-shelf” tools designed to target frequently mutated genes
`in one or more cancers of interest (e.g., AVENIO assay from
`Roche Diagnostics; ref. 44). Alternatively, personalized capture
`panels can be designed for each patient to enrich for patient-
`specific mutations identified from tumor sequencing (refs. 45,
`46; Fig. 1A). Recently, we developed a novel capture-based
`ctDNA MRD assay called Phased variant Enrichment and
`Detection Sequencing (PhasED-Seq), which leverages multi-
`ple somatic mutations within individual DNA fragments to
`decrease both technical and biological error rates (see below)
`and improves ctDNA MRD detection limits down to 1 part
`per million, 30–100-fold lower than previous approaches (47).
`
`Whole-Genome Sequencing
`In theory, application of whole-genome sequencing (WGS)
`to posttreatment plasma could allow detection of an even
`greater number of mutations than the aforementioned NGS
`approaches and not require custom panel design. However, due
`to assay background error rates, there are diminishing returns of
`increasing the number of tracked mutations past a certain point
`(see below). Therefore, recent work suggests that when combined
`with customized bioinformatics, direct WGS of plasma can
`potentially achieve similar but not superior limits of MRD detec-
`tion as the other NGS approaches (48). Additionally, the amount
`of sequencing required for the much larger targeted genomic
`space makes the per sample costs of WGS significantly higher.
`
`Emerging Techniques
`Although not a focus of this review, other analysis
`approaches based on DNA methylation or other epigenetic
`features reflecting chromatin state of tumor cells (termed
`“fragmentomics”) have been used to detect ctDNA (49).
`Although these emerging techniques have not been exten-
`sively evaluated in the context of MRD, epigenetic features
`may be complementary to somatic mutation tracking and
`may enable tumor genotype–naïve MRD detection. For exam-
`ple, the Guardant Reveal assay integrates somatic muta-
`tion and “epigenomic” (i.e., methylation and fragmentomic)
`approaches to detect ctDNA without prior sequencing of
`tumor DNA (50, 51). A recent study using this assay for
`detecting MRD in colorectal cancer demonstrated that incor-
`porating epigenomic analysis improved sensitivity compared
`with tumor-naïve somatic alterations alone (52). However,
`methylation-based approaches have been reported to have
`
`DECEMBER 2021 CANCER DISCOVERY | 2969
`
`Recent advances in molecular and computational biology
`have significantly improved the limit of detection (LOD) for
`ctDNA using somatic alterations (17, 18). These improve-
`ments have raised the possibility that ctDNA analysis could
`be used to identify patients harboring MRD following cura-
`tive therapy of solid cancers and to guide the administration
`of adjuvant or consolidation therapies (19). Here, we review
`the current evidence that detection of ctDNA following defin-
`itive therapy is prognostic in solid tumors and discuss the
`promise and limitations of ctDNA MRD testing.
`
`APPROACHES FOR ctDNA MRD ANALYSIS
`Digital PCR
`Several approaches have been utilized to identify known or
`common tumor mutations in plasma cfDNA samples. Digi-
`tal PCR (dPCR) improves on conventional allele-specific PCR
`amplification by partitioning a DNA sample into a large num-
`ber of smaller reactions to provide an absolute quantifica-
`tion that improves sensitivity (20). Digital PCR primers and
`probes can be designed to achieve very high specificity, and
`droplet-based approaches such as BEAMing (beads, emulsion,
`amplification, and magnetics) have maximized the number of
`individual DNA molecules that can be analyzed from a single
`sample (21). As a result, the detection limit of such digital PCR
`assays is limited in practice by the amount of cfDNA that can
`be isolated from a blood draw (22). With DNA inputs routinely
`achievable from patient plasma samples (∼30 ng), dPCR has
`been shown to have a detection limit of approximately 0.1%
`(23–25). However, due to differences in DNA input, sample
`quality, and analysis approaches, reported LODs vary substan-
`tially between studies. dPCR is very effective for tracking a small
`number of mutations identified from sequencing of tumor tis-
`sue or hotspot mutations with a high prevalence in the cancer
`of interest, such as KRAS mutations in pancreatic cancer (26).
`Due to complexities of multiplexing a large number of dPCR
`assays, the extent of multiplexing varies widely between studies
`and dPCR generally has inferior clinical sensitivity for MRD
`than highly parallel NGS methods monitoring multiple muta-
`tions (27, 28). Accordingly, dPCR is not a commonly preferred
`approach for solid tumor MRD detection in most contexts (29).
`
`PCR Amplicon–Based NGS
`NGS, also known as massively parallel sequencing, has
`been incorporated into several different ctDNA analysis
`approaches, enabling interrogation of millions to billions of
`DNA molecules from a biological sample. One approach to
`achieve sufficient sensitivity to detect rare ctDNA molecules
`uses gene-specific PCR amplicons to amplify one or more
`genomic regions expected to harbor tumor-derived muta-
`tions prior to NGS. Several approaches, including Safe-SeqS,
`introduce unique molecular identifier (UMI) sequences dur-
`ing preparation of DNA libraries for sequencing to reduce
`technical errors (17). Personalized multiplex PCR can be
`utilized to monitor multiple patient-specific mutations
`identified from sequencing of tumor tissue (30), and NGS
`sequencing of these lesions in cfDNA can achieve remarkable
`sensitivity levels (31). A related set of approaches uses a
`combination of ligation- and gene-specific PCR primers to
`partially preserve cfDNA fragment end information (32, 33).
`
`ctDNA Minimal Residual Disease in Solid Tumors
`
`Personalis EX2023
`
`

`

`REVIEW
`
`A
`
`Off-the-shelf panel
`Historical patients
`Individual patient
`
`Personalized panel
`Individual patient
`
`D
`
`Diagnosis
`
`Tumor genotype−naïve
`
`Tumor genotype−informed
`Variant identification in
`high allele fraction sample
`
`and/or
`
`KRASG12C
`GCCACCAGCT
`GCCACAAGCT
`
`WT:
`MUT:
`
`Positions analyzed
`↓
`GCCACCAGCT
`GCCACCAGCT
`GCCACAAGCT
`GCCACCAGCT
`GCCACCAGCT
`Monitor for patient-
`specific variants
`
`Tumor genotype–informed
`1.0
`
`6 mutations
`5,000× depth
`
`LOD95:
`0.01%
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`No baseline tumor
`or
`ctDNA testing
`
`Curative-intent treatment
`
`MRD testing
`
`Cell-free DNA
`
`+
`
`Leukocyte DNA
`
`Positions analyzed
`
`GCCACCAGCT
`GCCACCAGCT
`GCCACAAGCT
`GCCACCAGCT
`GCCACCAGCT
`Multiple hypothesis testing
`limits sensitivity
`
`E
`
`Tumor genotype-naive
`
`Mutation
`
`123456
`
`LOD95:
`0.2%
`
`Cell-free DNA
`
`Patient mutations
`CAGTTGCG
`GGTTCACT
`
`Cell-free DNA
`
`Recurrent mutations
`GGTTCAGT
`CAATTGCG
`CAATAGCG
`GGTTCAGT
`CAATAGCG
`GGTTCAGT
`CAATAGCG
`GGTTCAGT
`GGTTCAGT
`CAATTGCG
`CAATTGCG
`GGTTCAGT
`
`Same panel
`for all patients
`
`Target
`enrichment
`
`Personalized
`panel
`
`Target
`enrichment
`
`Background
`error rate: 0.001%
`LOD95:
`0.001%
`
`LOD95:
`0.1%
`
`LOD95:
`0.01%
`
`Undetectable
`VAFs because
`below error rate
`
`B
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`
`
`M utationscfD N A InputDepth
`
`+
`+ +
`+ + +
`
`+
`+ +
`+ + +
`
`Downloaded from http://aacrjournals.org/cancerdiscovery/article-pdf/11/12/2968/3013378/2968.pdf by guest on 07 September 2023
`
`F
`
`1
`0.1
`0.01
`0.001
`Allele fraction (%)
`Result: MRD not detected
`Assay
`type
`Plasma
`genotyping
`
`cfDNA
`methylation
`
`SNV
`ctDNA MRD
`
`Phased variant
`ctDNA MRD
`
`Tumor
`genotype
`
`Naïve
`
`Naïve
`
`Informed
`
`Informed
`
`Panel
`size
`~30 Mb
`
`355 kb
`
`~1.6 Mb
`
`2.1 Mb
`
`Sequencing
`depth
`95×
`4,075×
`380×
`4,400×
`
`Reference
`
`75
`
`46
`
`76
`
`77
`
`1
`
`0.01
`0.1
`Approximate limit of detection (%)
`
`0.0001
`0.001
`
`0.1
`0.01
`0.001
`0.0001
`Allele fraction (%)
`
`<40
`
`50−59
`40−49
`Age (years)
`
`>59
`
`0.0
`
`1
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`Probability of detection
`
`Clonal hematopoiesis (%)
`
`C
`
`Probability of detection
`
`MRD plasma sample
`6 mutations in tumor
`ctDNA = 0.01%
`
`
`+
`+ +
`+ + +
`
`0.0
`0.1
`
`0.0001
`
`0.01
`0.001
`Allele fraction (%)
`Result: MRD detected
`Clinically/commercially available
`example(s) [reference]
`FoundationOne Liquid CD× [*], Guardant 360 CD× [^],
`MSK-ACCESS [105], TruSight Oncology 500 [106]
`
`Adela [54], GRAIL [53]
`
`ArcherD× [37], C2i Genomics [48], Inivata [38],
`Natera Signatera [31], Roche AVENIO [44]
`
`Foresight Diagnostics [47]
`
`Figure 1.  Technical approaches for ctDNA MRD detection and factors affecting assay sensitivity and specificity. A, Schematic comparing assays
`using off-the-shelf versus personalized sequencing panels. Off-the-shelf panels are designed to cover recurrently mutated genes in the cancer type(s)
`of interest. The same panel is applied to tumor tissue and plasma of every patient, and personalization is achieved by bioinformatically considering only
`the positions mutated in the matched tumor. Personalized panels are designed to cover patient-specific mutations identified through sequencing of their
`tumor DNA. In this approach, personalization is achieved by every patient having a unique panel. B, Factors affecting the probability of ctDNA detection.
`Increasing the number of mutations tracked, the sequencing depth at mutant positions, or the cfDNA input can increase the probability of detection.
`Technical background from sources such as polymerase errors and oxidative damage sets the lower LOD due to an inability to distinguish artifacts from
`true tumor variants. LOD with 95% probability = LOD95. C, Prevalence of at least one clonal hematopoiesis variant detected in plasma as a function of
`the sequencing panel size and sequencing depth in published studies (46, 75–77). Error bars represent binomial 95% confidence intervals. D, Schematic
`of tumor genotype–informed versus tumor genotype–naïve ctDNA analysis. In genotype-naïve analysis, the MRD sample is interrogated at all sequenced
`genomic positions, leading to reduced sensitivity due to multiple hypothesis testing. In tumor genotype–informed analysis, variants are identified from
`tumor tissue or pretreatment plasma samples with high tumor allele fraction. Only patient-specific variants are monitored in the MRD sample. In both
`cases, genotyping DNA from leukocytes in the peripheral blood can improve specificity by identifying variants stemming from clonal hematopoiesis.
`E, Comparison of the LODs of tumor genotype–naïve and tumor genotype–informed ctDNA analysis at a median sequencing depth of 5,000× for a patient
`with six tumor mutations and a ctDNA allele fraction of 0.01%. Due to multiple hypothesis testing with tumor genotype–naïve analysis, the LOD95 is
`0.2%, and no mutations are detected above background despite mutations with allele fractions of 0.02% (1/5,000 molecules) and 0.04% (2/5,000
`molecules) being present in the sample. In contrast, in the same sample, tumor genotype–informed analysis at 5,000× depth is associated with an LOD95
`of 0.01% (approximated by a binomial distribution), and therefore ctDNA MRD has a 95% chance of being detected. F, Summary of assay types, tumor
`genotyping requirements, and approximate LODs (i.e., analytic sensitivity) for commercially/clinically available ctDNA analysis tests. Because no data
`comparing these methods on the same samples are available, the approximate LOD at which 95% of samples would be expected to be called positive for
`each group of assays is estimated from published studies and/or conference abstracts and rounded to the nearest log (31, 37, 38, 44, 47, 48, 53, 54, 105,
`106). *, http://www.accessdata.fda.gov/cdrh_docs/pdf19/P190032B.pdf; ∧, https://www.accessdata.fda.gov/cdrh_docs/pdf20/P200010B.pdf
`
`LODs of approximately 0.1% (53, 54), and rigorous LOD
`analyses for fragmentomic-based approaches in the context
`of ctDNA MRD have not been published to date. Therefore,
`although more studies are clearly needed, it seems unlikely
`that these approaches will be able to match the LODs of the
`most sensitive tumor genotype–informed, somatic mutation–
`based ctDNA MRD approaches.
`
`TECHNICAL ASPECTS OF ctDNA MRD
`DETECTION USING NGS
`Physical Limits of ctDNA Analysis
`The LOD for an assay is the lowest quantity of an analyte
`that can be reliably detected (55). Unfortunately, most studies
`in the ctDNA field have not rigorously defined or established
`
`2970 | CANCER DISCOVERY DECEMBER 2021
`
`AACRJournals.org
`
`Moding et al.
`
`Personalis EX2023
`
`

`

`Downloaded from http://aacrjournals.org/cancerdiscovery/article-pdf/11/12/2968/3013378/2968.pdf by guest on 07 September 2023
`
`REVIEW
`
`for anemia limit the amount of blood that can be collected for
`ctDNA analysis (69). Although greater cfDNA input improves
`sensitivity across different methods, increasing input cannot
`increase the LOD beyond an assay’s background error rate.
`
`Technical and Biological Sources of Error
`Even if ctDNA molecules are efficiently recovered by an
`NGS MRD assay, their subsequent detection relies on the
`successful identification and quantitation by the associated
`bioinformatics pipeline. Within these pipelines, a key chal-
`lenge is resolving the desired ctDNA biological signal from
`the noise or background errors arising from various techni-
`cal or biological sources. In this context, there are two major
`components that together determine the overall background
`error rate of ctDNA MRD assays: (i) technical errors leading
`to artifactual mutations occurring ex vivo during the various
`molecular biology steps and (ii) bona fide somatic variants
`stemming from nontumor tissues that harbor mutations.
`Technical errors can be introduced during sample process-
`ing from sources such as unrepaired DNA polymerase errors
`arising during PCR (17) and oxidative DNA damage (70, 71).
`Because true tumor variants cannot be resolved below the
`error noise floor, these errors serve to limit the lowest pos-
`sible ctDNA concentration that can be detected. To address
`this issue, multiple strategies have been developed to decrease
`technical errors. These strategies include the use “barcoding”
`techniques that use UMIs, in silico elimination of stereotypi-
`cal background artifacts (i.e., “polishing”), and inclusion of
`free radical scavengers during library preparation to decrease
`oxidative damage (17, 18, 31, 46, 68). Collectively, these error-
`suppression strategies can dramatically reduce the technical
`errors arising ex vivo during cfDNA profiling.
`Biological background secondary to somatic mutations
`found in cfDNA but not originating from tumor cells rep-
`resents a second important source of potential false-posi-
`tive mutations. Through a process called age-related clonal
`hematopoiesis (CH), hematopoietic stem cells can acquire
`mutations that can be found in both circulating peripheral
`blood cells and cfDNA (72). When considering peripheral
`blood leukocytes, patients with clonal mutations above a
`variant allele fraction of 2% in genes that are canonically
`associated with hematologic malignancies, but who do not
`meet the criteria for diagnosis of leukemia, are considered to
`have clonal hematopoiesis of indeterminate potential (CHIP;
`ref. 73). Because the majority of cfDNA arises from hemat-
`opoietic sources (74), CH represents a major contributor to
`biological background in cfDNA profiling exercises, includ-
`ing detection of ctDNA MRD. Importantly, the prevalence of
`CH variants increases with patient age, broader panels, and
`increasing sequencing depth (Fig. 1C) and has been found to
`be as high as 100% in patients 60 years or older (46, 75–77).
`CH represents the dominant biological source of false-
`positive mutations when identifying somatic mutations in
`cfDNA. Importantly, sequencing peripheral blood leukocytes
`can help to identify cfDNA mutations due to CH. In support
`of this notion, a recent ctDNA study of localized non–small
`cell lung cancer (NSCLC) found that up to 15% of TP53
`mutations in the cfDNA were attributable to CH and found
`in matched leukocytes but not in matched tumor (78). In a
`separate study, deeper sequencing of matched leukocytes in
`
`DECEMBER 2021 CANCER DISCOVERY | 2971
`
`the LOD. To conform to clinical laboratory testing guide-
`lines, the LOD should ideally be defined as the ctDNA
`concentration at which 95% of clinical samples will be called
`positive (56). For mutation-based NGS ctDNA MRD assays,
`the LOD depends on both biological and technical factors.
`In practice, reliable LODs have not been published for most
`ctDNA analysis approaches, and multiple definitions have
`been used, making it challenging to compare across studies.
`In order to be meaningful, LODs should be determined in
`settings that match clinical samples as closely as possible,
`including in the amount of cfDNA input (57).
`Regardless of the detection limit, for noninvasive detection
`to be feasible using circulating nucleic acids in blood plasma,
`tumor DNA must first be released into the blood and collected
`in a blood draw. Although ctDNA is generally thought to be
`released from necrotic or apoptotic tumor cells (58–60), the
`exact mechanisms of ctDNA release into the bloodstream and
`the relative contributions of different cell death mechanisms,
`phagocytosis, exocytosis, active secretion, and other cellular
`processes have not been well characterized in human cancers
`(61). Across detection platforms, ctDNA levels generally cor-
`relate with tumor burden on imaging (28, 31, 46, 62, 63).
`Accordingly, the number of residual cells after therapy likely
`also correlates with ctDNA MRD levels. Prior studies have also
`demonstrated that ctDNA release varies within and between
`tumor types and histologies, with a recurring observation that
`squamous cell carcinomas tend to shed higher ctDNA levels
`than adenocarcinomas (31, 46, 64, 65). Furthermore, levels of
`normal cfDNA increase with tissue injury from inflammation
`and ischemia related to nonmalignant conditions, surgery, or
`even vigorous exercise (66), which can lower the allele fraction
`of ctDNA molecules below the LOD of an assay. Indeed, post-
`surgical inflammatory changes have been shown to induce a
`significant increase in cfDNA levels postoperatively for up to
`three to four weeks (67), suggesting that ctDNA MRD should
`not be measured immediately following surgery. Collectively
`and irrespective of the assay used, these biological factors can
`substantially affect the sensitivity of ctDNA MRD techniques
`for accurately detecting residual disease and predicting relapse.
`Assuming that ctDNA molecules are present in a blood
`sample, detection of these molecules relies on their efficient
`profiling. Each of the ctDNA MRD assay approaches includes
`multiple molecular biology steps that have imperfect mol-
`ecule recovery and at any of which rare mutant ctDNA
`molecules could potentially be lost. For instance, when inter-
`rogated by NGS profiling, mutant ctDNA molecules need to
`be incorporated into the sequencing library, and this library
`needs to be sequenced deeply enough to observe these rare
`molecules in the final result. Due to losses of molecules dur-
`ing library preparation and sequencing, NGS methods gener-
`ally have cfDNA molecule recovery efficiencies ≤50% (44, 68).
`Lastly, the probability of detecting ctDNA MRD is a func-
`tion of the ctDNA concentration, the number of mutations
`tracked, and the number of unique cfDNA molecules inter-
`rogated (Fig. 1B; ref. 18). For NGS approaches, tracking mul-
`tiple mutations, using more cfDNA input, and sequencing
`to a greater depth can improve the likelihood of identifying
`mutant ctDNA molecules in a given blood sample. cfDNA
`input is ultimately limited by the amount of plasma that can
`be collected and analyzed. In patients with cancer, concerns
`
`ctDNA Minimal Residual Disease in Solid Tumors
`
`Personalis EX2023
`
`

`

`Downloaded from http://aacrjournals.org/cancerdiscovery/article-pdf/11/12/2968/3013378/2968.pdf by guest on 07 September 2023
`
`REVIEW
`
`patients with NSCLC found that 40.6% of TP53 mutations
`present in cfDNA were also detected in matched leukocytes
`(46). TP53 mutations in cfDNA that were also found in leu-
`kocytes displayed less evidence of the smoking mutational
`signature than TP53 mutations also found in matched tumor
`tissue, supporting their disparate biological sources.
`Although sequencing peripheral blood leukocytes is help-
`ful for distinguishing tumor-derived mutations from CH,
`approximately 10% of mutations found in cfDNA of noncan-
`cer controls are not found in matched leukocytes (46). This
`observation suggests that some CH mutations can be missed
`due to low prevalence of CH subclones in the peripheral
`blood, perhaps reflecting CH in noncirculating hematopoi-
`etic precursors. Additionally, it is possible that mutations
`from nonmalignant, nonhematopoietic cells can be found in
`cfDNA. In support of this, recent studies have demonstrated
`that somatic mutations can be present in diverse nonmalig-
`nant cell types, including epithelial (79, 80), endothelial (81),
`stromal (82), and others (83). As detailed below, incorporat-
`ing sequencing of tumor tissue to identify somatic mutations
`that are then monitored in plasma samples can guard against
`these sources of biological background.
`
`Tumor Genotype–Informed versus Tumor
`Genotype–Naïve ctDNA Analysis
`Due to the complications posed by technical and biological
`background mutations described above, most ctDNA MRD
`studies have performed “tumor genotype–informed” analyses
`to monitor known tumor variants in posttreatment plasma
`(Fig. 1D). This approach includes the genotyping of tumor
`tissue to identify mutations that are then tracked in plasma,
`which lowers the risk of false positives due to technical and
`biological background sources of error. Additionally, such a
`tumor genotype–informed approach limits the number of
`genomic positions interrogated in cfDNA, therefore decreas-
`ing multiple hypothesis testing. Accordingly, the tumor geno-
`type–informed approach can be less demanding for blood
`sample volumes, because fewer unique mutant cfDNA mol-
`ecules are required for detecting ctDNA as compared with a
`genotype-naïve approach (Fig. 1E).
`Several commercially available ctDNA platforms support
`tumor genotype–informed MRD detection, including assays
`for solid tumors from Natera, Roche Diagnostics, Invitae
`(ArcherDx), Inivata, and Foresight Diagnostics, among oth-
`ers. Notably, these platforms are technically distinct from
`liquid biopsy panels such as Guardant360 (https://www.
`accessdata.fda.gov/cdrh_docs/pdf20/P200010A.pdf) and
`FoundationOne Liquid CDx (http://www.accessdata.fda.gov/
`cdrh_docs/pdf19/P190032A.pdf) that have been developed
`for noninvasive genotyping in the setting of advanced disease
`(84). Although such noninvasive genotyping assays are very
`useful for identifying actionable tumor mutations in patients
`with metastatic disease (85), a recent analysis by the SEQC2
`Working Group led by the FDA found that detection of
`variants was less reliable below an allele fraction of 0.5% (86).
`Therefore, currently available assays designed for the primary
`purpose of noninvasive genotyping in advanced disease are
`not optimal for ctDNA MRD detection. This is mainly due
`to the low ctDNA allele fractions typically observed following
`definitive treatment of localized solid cancers in the absence
`
`of radiographic disease burden as well as the confounding
`effects of CH. In contrast, tumor genotype–informed MRD
`detection approaches can attain LODs of ≤0.01%, which
`makes them preferrable for detection of minute amounts of
`MRD (refs. 18, 37, 47, 87; Fig. 1F).
`
`EVIDENCE SUPPORTING THE PROGNOSTIC
`VALUE OF ctDNA MRD IN SOLID CANCERS
`Landmark versus Surveillance ctDNA
`MRD Analysis
`In ctDNA MRD studies focused on solid tumors, two main
`types of analysis have generally been reported: (i) MRD land-
`mark analysis and (ii) surveillance analysis (Fig. 2A). Although
`these two types of analysis are related, their distinct features
`are relevant for the clinical application of ctDNA MRD. MRD
`landmark analysis determines the ctDNA status of patients
`at a single, prespecified time point, which is typically shortly
`after completion of standard-of-care treatment (e.g., surgery
`and radiotherapy). In contrast, ctDNA surveillance analysis
`involves evaluating longitudinal blood draws at multiple time
`points during follow-up, with ctDNA status determined by
`whether any blood draw (irrespective of time point) is positive.
`From a clinical perspective, determining MRD status at
`an early posttreatment landmark time is attractive because it
`would facilitate immediate decision-making about adjuvant
`or consolidation therapies, while minimizing the costs from
`testing serial blood samples. However, one could also envision
`performing repeat MRD surveillance over time and initiating
`adjuvant or consolidation therapy at the first positive blood
`draw. This approach has the potential advantage of decreas-
`ing false negatives. For either the landmark or surveillance
`approaches to offer the potential of meaningful improvement
`over current standard of care, MRD must be detectable with
`a significant lead time over conventional imaging. Given the
`differences in these two approaches, the type of analysis must
`be considered when comparing across studies and techniques.
`
`Clinical Sensitivity and Specificity of
`Current Approaches
`More than 20 studies to date have demonstrated the prog-
`nostic value of ctDNA MRD detection in multiple solid can-
`cers with a variety of technical approaches (Table 1). We
`attempted to summarize the performance of current ctDNA
`analysis approaches from some of these seminal studies, which
`we selected from the literature based on inclusion of at least
`10 patients with ctDNA analysis performed after completion
`of curative-intent therapy for nonmetastatic solid tumors (Fig.
`2B). Studies and/or patients were excluded if adjuvant therapy
`was given after ctDNA analysis, but this information was not
`available for every study. It is important to note that different
`methodologies for ctDNA MRD analysis have not been tested
`side-by-side on the same set of samples, and these analyses
`would be challenging due to the large amounts of plasma
`that would be required. Additionally, the same method may
`perform differently across laboratories or in samples with dif-
`ferent quality. Our goal in this review is to synthesize the pub-
`lished data rather than to rigorously compare diverse methods.
`Despite variable study designs, different tumor types, and
`inconsistent study endpoints, detection of ctDNA MRD at
`
`2972 | CANCER DISCOVERY DECEMBER 2021
`
`AACRJournals.org
`
`Moding et al.
`
`Personalis EX2023
`
`

`

`Downloaded from http://aacrjournals.org/cancerdiscovery/article-pdf/11/12/2968/3013378/2968.pdf by guest on 07 Sept

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket