throbber
REVIEWS
`
`Liquid biopsy: monitoring cancer-genetics
`in the blood
`
`Emily Crowley, Federica Di Nicolantonio, Fotios Loupakis and Alberto Bardelli
`
`Abstract | Cancer is associated with mutated genes, and analysis of tumour-linked genetic alterations is
`increasingly used for diagnostic, prognostic and treatment purposes. The genetic profile of solid tumours
`is currently obtained from surgical or biopsy specimens; however, the latter procedure cannot always be
`performed routinely owing to its invasive nature. Information acquired from a single biopsy provides a spatially
`and temporally limited snap-shot of a tumour and might fail to reflect its heterogeneity. Tumour cells release
`circulating free DNA (cfDNA) into the blood, but the majority of circulating DNA is often not of cancerous origin,
`and detection of cancer-associated alleles in the blood has long been impossible to achieve. Technological
`advances have overcome these restrictions, making it possible to identify both genetic and epigenetic
`aberrations. A liquid biopsy, or blood sample, can provide the genetic landscape of all cancerous lesions
`(primary and metastases) as well as offering the opportunity to systematically track genomic evolution.
`This Review will explore how tumour-associated mutations detectable in the blood can be used in the clinic
`after diagnosis, including the assessment of prognosis, early detection of disease recurrence, and as
`surrogates for traditional biopsies with the purpose of predicting response to treatments and the development
`of acquired resistance.
`
`Crowley, E. et al. Nat. Rev. Clin. Oncol. 10, 472–484 (2013); published online 9 July 2013; doi:10.1038/nrclinonc.2013.110
`
`Introduction
`Biopsies have been used by clinicians to diagnose and
`manage disease for 1,000 years.1 In patients with cancer,
`biopsies allow the histological definition of the disease
`and, more recently, have revealed details of the genetic
`profile of the tumour enabling prediction of disease pro-
`gression and response to therapies. As the techniques that
`have enabled us to analyse a biopsy become ever more
`sophisticated, we have realised the limitations of looking
`at this single snap-shot of the tumour. This single-biopsy
`bias was highlighted in a study by Gerlinger et al.2 in which
`it was demonstrated that a portion taken from different
`parts of a primary tumour and its metastases showed
`extensive intertumoural and intratumoural evolution.
`This tumoural heterogeneity highlights the diffi culty of
`dictating a therapeutic course of action based on a single
`biopsy, as it is likely to underestimate the c omplexity of the
`genomic landscape of the tumour.
`Having established that there is considerable tumour
`heterogeneity, taking multiple biopsies from the patients’
`primary tumour and metastases would seem to be the most
`obvious next step, so why is it not routinely done? There are
`many difficulties in obtaining a tissue biopsy—i ncluding
`the discomfort suffered by the patient, inherent clinical
`risks to the patient, potential surgical complications and
`economic considerations—meaning that multiple or serial
`biopsies are often impractical. In addition, some tumours
`are not accessible for biopsy, the procedure itself might
`
`Competing interests
`The authors declare no competing interests.
`
`increase the risk of the cancer ‘seeding’ to other sites,3 and
`the procedure might not be recommended for patients
`receiving antiangiogenic treatment.4 Even in an ideal
`situation where several meta static sites can be biopsied
`simultaneously, the analysis of the samples can delay the
`initiation of treatment, and might irremediably jeopardise
`it. These limitations are particular restraints in the setting
`of acquired resistance to therapy, as the ability of a clinician
`to detect thera peutic biomarkers at an early stage would
`allow a potentially successful change in treatment course.
`In light of these limitations on the use of single biopsies,
`new ways to observe tumour genetics and tumour dynam-
`ics have evolved. In 1948, the publication of a manuscript
`that described circulating free DNA (cfDNA) and RNA
`in the blood of humans was, without knowing it, the first
`step towards the ‘liquid biopsy’.5 Furthermore, the levels of
`cfDNA were higher in diseased than healthy individuals,
`indicating that it is possible to screen for the presence of
`disease through a simple blood test.6 Indeed, the specific
`detection of tumour-derived cfDNA has been shown to
`correlate with tumour burden, to a change in response
`to treatment or surgery, and to indicate that subpopula-
`tions of tumour cells that are resistant to treatment can
`prolifer ate in response to therapy.7,8 With the development
`of sensitive techniques that enable the detection of rare
`mutations, it is now possible to understand the hetero-
`geneous landscape of the tumour using a blood sample.9
`Nevertheless, key challenges lie ahead and not all results
`consistently support the application of c irculating tumour
`DNA (ctDNA) in the clinic.
`
`Department of
`Oncology, University
`of Turin, Institute for
`Cancer Research
`and Treatment,
`Strada Provinciale
`142 Km 3.95,
`10060 Candiolo,
`Turin, Italy (E. Crowley,
`F. Di Nicolantonio,
`A. Bardelli). Unit of
`Medical Oncology 2,
`Azienda Ospedaliero-
`Universitaria Pisana,
`Via Roma 67,
`56126 Pisa, Italy
`(F. Loupakis).
`
`Correspondence to:
`A. Bardelli
`alberto.bardelli@
`unito.it
`
`472 | AUGUST 2013 | VOLUME 10
`
`www.nature.com/nrclinonc
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`00001
`
`EX1040
`
`

`

`The early diagnosis of cancer is one application for
`cfDNA assessment and has been covered extensively
`in previous reviews.10,11 This Review will explore how
`tumour-associated genetic alterations detectable in the
`blood can be used in the clinic, including the assessment
`of prognosis, early detection of disease recurrence, and
`as surro gates for traditional biopsies with the purpose of
`predicting response to treatments or the development
`of acquired resistance. It will also cover technical and
`logistical challenges that might represent hurdles for
`clinical implementation.
`
`Discovery and detection of ctDNA
`There are at least two potential, but not mutually exclu-
`sive, mechanisms by which DNA can enter into the cir-
`culation. These mechanisms can be broadly categorized
`as passive and active. The passive mechanism suggests
`that apoptotic and necrotic cells release nuclear and
`mitochondrial DNA into the circulation in the process
`of cellular destruction.12 Under normal physiological
`conditions, infiltrating phagocytes clear apoptotic and
`necrotic debris and, therefore, levels of cfDNA in healthy
`individuals are low.13 However, certain conditions, such
`as within a tumoural mass or in the instance of exhaus-
`tive exercise or inflammation, clearance does not happen
`efficiently,13 leading to an accumulation of cellular debris,
`including DNA, that is then released into the blood
`(Figure 1).14,15 Alternatively, active DNA release has been
`reported in studies with cultured cell lines of different
`origins and is believed to involve the spon taneous release
`of DNA fragments into the circulation.16–19 Multiple
`hypotheses have been formulated to explain why living
`cancer cells would actively release DNA into the circu-
`lation, including the possibility that cancer cells release
`oncogenic DNA to affect the transformation of suscep-
`tible cells at distant sites.20,21 Several reviews are available
`that detail the biology and mechanisms of DNA release
`into circulation.10,12,22
`ctDNA might also be released by circulating tumour
`cells (CTCs) present in the blood.23 Definitive evidence
`for this mechanism has not been reported, and there is a
`discrepancy between the number of CTCs and the quan-
`tity of cfDNA in the blood. A single human cell contains
`6 pg of DNA24 and there is an average of 17 ng of DNA
`per ml of plasma in advanced-stage cancers;25 therefore,
`if CTCs were the primary source of ctDNA it would
`require over 2,000 cells per ml of plasma. In reality, there
`are, on average, less than 10 CTCs per 7.5 ml blood.26
`Presently, the most commonly used protocols to obtain
`cfDNA require approximately 1 ml of serum or plasma
`(3 ml of blood) and preparation should not exceed 4–5 h
`following the blood draw. For plasma preparation, blood
`must be collected in a tube treated with an anti coagulant,
`preferably EDTA (ethylenediaminetetraacetic acid). Cells
`are then removed by centrifugation and the super natant,
`or plasma, is removed.27 Serum is collected after the
`blood is allowed to clot and following centrifugation
`the supernatant, or serum, is removed.28 Circulating
`DNA is then extracted from the plasma or serum using
`c ommercially available kits.
`
`REVIEWS
`
`Key points
`
`■ Under representation of the heterogeneity of a tumour and poor sample
`availability means tissue biopsies are of limited value for the assessment
`of tumour dynamics in the advanced stages of disease
`■ Extended periods between sampling and clinical application of the results,
`as well as additional lines of treatment between sampling, might result in
`an altered genetic composition of the tumour
`■ Circulating free DNA can be extracted from the blood and tumour-specific
`aberrations assessed to provide a genetic landscape of the cancerous lesions
`in a patient
`■ Tracking tumour-associated genetic aberrations in the blood can be used to
`assess the presence of residual disease, recurrence, relapse and resistance
`■ Monitoring the emergence of tumour-associated genetic aberrations in
`the blood can be used to detect the emergence of resistant cancer cells
`5–10 months before conventional methods
`■ To implement circulating tumour DNA testing in the clinic, standardization of
`techniques, assessment of reproducibility and cost-effectiveness is required
`as well as prospective validation in clinical trials
`
`The analysis of ctDNA from the blood offers an excit-
`ing clinical application in patients, that is, the detection
`of tumour-specific genetic aberrations (Figure 1). This
`approach has greater dynamic range compared to total
`circulating DNA,29 is more specific, and has many poten-
`tial applications in the clinic. However, it is also tech-
`nically more challenging owing to the presence of high
`levels of DNA originating from tissue of non-tumour
`origin. A proportion of cfDNA is derived directly from
`the tumour (ctDNA) and this fraction can be quanti-
`fied.30–35 Furthermore, an in vivo study has shown a direct
`correlation between tumour burden and the quantity of
`ctDNA released.33
`High-analytical sensitivity and specialized equip-
`ment are required for detection of ctDNA because the
`quantity and quality of tumour-derived DNA can vary
`dramatically. Techniques are available that allow reliable
`monitoring of tumour-associated genetic aberrations,
`including somatic mutations, loss of heterozygosity and
`chromosomal aberrations in the blood (Table 1), at fre-
`quencies as low as 0.01%.8,9,36–38 The fraction of circu-
`lating DNA that is derived from the tumour can range
`between 0.01% and 93%.36,39 Tumour-specific genetic
`aberrations in cfDNA might offer intriguing insights
`into tumour dynamics. Nevertheless, there are clini-
`cal situations, including early stage disease, where it is
`unlikely to have broad applications as well as clinical
`settings where cfDNA levels are below optimal levels for
`the detection of mutations.25,40,41 According to a study by
`Perkins et al.,25 the optimal DNA concentration for the
`detection of mutations (using a Sequenom® MassARRAY®
`platform) is approximately 30 ng/ml in the plasma. As
`certain cancers, including melanoma, have a low average
`total cfDNA level (7 ng/ml in the plasma) the accuracy of
`detection could be suboptimal. However, with improving
`technique sensitivities, the ability to detect genetic aber-
`rations at lower frequencies will undoubtedly improve,
`as exemplified by the improvement in PIK3CA mutation
`detection in studies by Board et al.40 and Higgins et al.,42
`which showed a concordance between primary tumour
`tissue and plasma DNA of 95% and 100%, respectively.
`
`NATURE REVIEWS | CLINICAL ONCOLOGY
`
`VOLUME 10 | AUGUST 2013 | 473
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`00002
`
`

`

`REVIEWS
`
`Circulating
` tumour cell
`
`Healthy
`tissue
`
`Apoptosis
`or necrosis
`
`Blood plasma or
`serum sample
`containing ctDNA
`
`Apoptosis
`
`In(cid:10)amed
`tissue
`
`Healthy cell
`Phagocyte
`Tumour cell
`Mutation
`Red blood cell
`Endothelial cell
`Chromosome
`
`Figure 1 | Release and extraction of cfDNA from the blood. cfDNA is released from healthy, inflamed or diseased
`(cancerous) tissue from cells undergoing apoptosis or necrosis. cfDNA can be extracted from a blood sample and
`genetic aberrations in the DNA released from cancerous tissue detected and quantified. Tumour-derived genetic alterations
`that can be detected in the blood include point mutations (consecutive purple, red, green and blue DNA strands), copy
`number fluctuations (red portion of chromosomes) and structural rearrangements (green and red DNA strands).
`Abbreviations: cfDNA, circulating free DNA; ctDNA, circulating tumour DNA.
`
`Liquid biopsies in the clinic
`In the past few years there has been considerable focus
`on the need for ‘biomarkers’. These biomarkers should
`be surrogate indicators for a future event, such as disease
`recurrence, disease progression, or death, and should
`indicate if a specific treatment will reduce that risk.43
`Despite extensive investigations, there are no currently
`approved applications for liquid biopsies in the clinical
`setting. Nevertheless, recently published data demon-
`strate that ongoing research holds considerable promise
`for the future of molecular testing of ctDNA—one of the
`most promising future tools for clinical application.
`
`Assessment of prognosis
`Prognosis
`Assessing prognosis for an individual patient involves
`a combination of clinical observations, staging, and
`histopathological and biomolecular characterization
`of different tumour types. This information, derived
`from imaging and biopsy specimens, allows a clini-
`cian to appraise the tumour biology, precisely stage the
`patient’s tumour and differentiate between those patients
`with more-aggressive or less-aggressive disease. In this
`context, liquid biopsies are unlikely to supersede current
`
`methods, but their use might be important in circum-
`stances where a biopsy is not available or genetic analysis
`of archived tumour samples is not possible.44,45
`Assessment of disease stage is one of the most-
`reliable predictors of prognosis and the relationship
`between levels of tumour-specific genetic aberrations
`and stage requires thorough evaluation for all cancer
`types. Studies have revealed a statistically significant
`correlation between disease stage and the presence
`of tumour- associated genetic aberrations (including
`mutations in TP53, KRAS, and APC and allelic imbal-
`ances) in the blood of patients with resectable breast,
`ovarian, pancreatic and colorectal cancer and oral
`squamous-cell carcinoma.27,28,46–53
`Furthermore, following mastectomy as treatment for
`breast cancer, it was reported that patients with tumours
`with vascular invasion, more than three lymph-node
`metastases, and high histological grade at diagnosis
`had persistent tumour-associated microsatellite DNA
`alterations as detected by PCR post-surgery in plasma-
`extracted DNA.46 Moreover, the presence of tumour-
`associated genetic aberrations in the blood, including
`TP53 mutations47 and loss of hetero zygosity,48 correlated
`with overall survival or disease-free survival as assessed
`
`474 | AUGUST 2013 | VOLUME 10
`
`www.nature.com/nrclinonc
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`00003
`
`

`

`Table 1 | Tumour-associated genetic aberrations in circulating free DNA
`
`Tumour type
`
`Stage
`
`n
`
`Tumour-specific
`aberration
`
`Tumour
`burden
`or stage*
`
`Source
`
`Technique
`
`Reference‡
`
`REVIEWS
`
`APC
`
`No/Yes
`
`Plasma
`
`BEAMing
`
`Colorectal
`cancer
`
`Breast cancer
`
`Ovarian
`cancer
`
`Early to
`advanced
`Advanced
`
`Early to
`advanced
`Early to
`advanced
`
`Early to
`advanced
`Early to
`advanced
`
`Advanced
`
`Advanced
`
`Early to
`advanced
`
`33
`
`18
`
`104
`
`70
`
`72
`
`34 (retrospective)
`and 51
`(prospective)
`30
`
`38
`
`63
`
`APC, KRAS,
`PIK3CA, TP53
`APC, KRAS, TP53
`
`KRAS
`
`PIK3CA
`
`PIK3CA
`
`PIK3CA, TP53,
`structural variation
`
`TP53, PTEN, EGFR,
`BRAF, KRAS,
`PIK3CA
`
`Yes
`
`NA
`
`NA
`
`Yes
`
`NA
`
`Yes
`
`Yes
`
`Yes
`
`Plasma
`
`BEAMing
`
`Serum
`
`PCR-SSCP
`
`Plasma
`
`ME-PCR
`
`Plasma
`and serum
`Plasma
`
`ARMS-
`Scorpion PCR
`BEAMing
`
`Diehl et al.
`(2005)36
`Diehl et al.
`(2008)27
`Wang et al.
`(2004)28
`Frattini et al.
`(2008)34
`
`Board et al.
`(2010)40
`Higgins et al.
`(2012)42
`
`Plasma
`
`Plasma
`
`Serum
`
`TAm-Seq and
`digital PCR
`
`Dawson et al.
`(2013)29
`
`TAm-Seq
`Digital PCR
`Fluorescent-
`PCR
`
`WGS
`
`Forshew et al.
`(2012)38
`Kuhlmann
`et al. (2012)52
`
`4
`
`21
`
`44
`
`64
`
`20
`
`SNV
`
`KRAS
`
`KRAS
`
`Yes
`
`Yes
`
`Plasma
`
`Plasma
`
`MASA PCR
`
`No/Yes
`
`Plasma
`
`RFLP-PCR
`
`Microsatellite loci
`
`Yes
`
`Microsatellite loci
`
`No
`
`Serum
`
`Serum
`
`PCR
`
`PCR
`
`Chan et al.
`(2013)9
`
`Yamada et al.
`(1998)54
`Castells et al.
`(1999)53
`
`Hamana et al.
`(2005)50
`Kakimoto et al.
`(2008)157
`
`Nygaard et al.
`(2013)59
`
`Hepatocellular
`carcinoma
`
`Early
`
`Early to
`advanced
`Early to
`advanced
`
`Early to
`advanced
`Early to
`advanced
`
`Pancreatic
`cancer
`
`Oral
`squamous-cell
`carcinoma
`
`Non-small-cell
`lung cancer
`
`Breast and
`osteosarcoma
`
`Colorectal and
`breast cancer
`
`Advanced
`
`246
`
`KRAS
`
`Advanced
`
`3
`
`Advanced
`
`10
`
`Genomic
`alterations
`
`Chromosomal
`alterations
`
`Yes
`
`Yes
`
`Yes
`
`Plasma
`
`ARMS-qPCR
`
`Plasma
`and serum
`
`Nested-real
`time PCR
`
`McBride et al.
`(2010)154
`
`Plasma
`
`WGS
`
`Leary et al.
`(2012)37
`
`*This column indicates if the study observed a correlation between tumour-associated genetic aberrations and tumour burden or disease stage. ‡The table
`includes studies in which different tumour-associated genetic aberrations have been detected using a variety of techniques, with different cancer types and at
`different stages. Abbreviations: ARMS, amplification refractory mutation system; BEAMing, beads, emulsion, amplification, magnetics; MASA, mutant allele
`specific amplification; ME-PCR, mutant enriched PCR; NA, not applicable; PCR-SSCP, single-strand conformation polymorphism PCR; qPCR, quantitative PCR;
`RFLP-PCR, restriction fragment length polymorphism PCR; SNV, single nucleotide variants; WGS, whole-genome sequencing.
`
`by multivariate analysis. There are, however, conflict-
`ing studies in which a correlation between stage and
`levels of tumour-associated genetic aberrations (includ-
`ing KRAS) are not observed.34,54 Controversy associ-
`ated with such studies is often due to limited sample
`sizes as well as some of the studies not being designed
`to specific ally address this issue,34 in addition to the
`obvious technical differences that will be discussed later
`in this Review.
`What are the possible applications of liquid biopsies
`in the clinic for the assessment of patient prognosis?
`In the context of resectable tumours, the utility of
`liquid biopsies is likely to be limited. In this setting,
`a sample of the tumour itself is usually available and
`aside from conventional histological, immunohisto-
`chemical and molecular analysis, the development of
`gene- expression classifier assays provide additional
`prognostic information.55,56 In the context of breast and
`colorectal cancer, there are FDA-approved assays that
`
`can discriminate, in specific settings, between high-risk
`and low-risk groups.55–58 Equivalent panels of genetic
`aberrations that can be tested using cfDNA samples to
`accurately stratify patients are not currently available.
`Therefore, liquid biopsies in the resectable setting are
`currently unlikely to have significant clinical utility for
`assessing prognosis.
`What could be the utility of liquid biopsies for
`advanced-stage, unresectable diseases? In a multi-
`variate analysis, KRAS mutations present in the plasma
`of 246 patients with advanced-stage non-small-cell lung
`cancer (NSCLC) was shown to predict poor prognosis
`in patients receiving first-line chemotherapy.59 KRAS
`mutations in circulation have also been assessed in a
`cohort of 44 patients with pancreatic cancer to deter-
`mine patient prognosis and, despite the poor sensitiv-
`ity of the test (27%), it was shown that patients with
`KRAS mutations had a significantly reduced probability
`of survival compared to those patients without KRAS
`
`NATURE REVIEWS | CLINICAL ONCOLOGY
`
`VOLUME 10 | AUGUST 2013 | 475
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`00004
`
`

`

`A different approach involves the quantification of
`the level of multiple tumour-associated genetic muta-
`tions present in the blood—this approach might be
`more sensi tive than the assessment of total cfDNA or
`measuring single tumour-specific aberrations for the
`stratification of patients with cancer according to their
`probability of survival.29 For example, the number of
`copies of DNA harbouring specific somatic muta-
`tions when detected in the plasma were calculated in
`a cohort of 30 women with metastatic breast cancer,
`and an inverse relationship (P <0.001) was identified
`between plasma copy number and survival.29 Whether
`this method can be used to accurately stratify patients
`of all cancer types25 and at different disease stages40,72
`remains to be validated.
`
`Detection of recurrence
`A promising clinical application for liquid biopsies is the
`early detection of relapse after potentially curative treat-
`ments (Figure 2a). After treatment with curative intent,
`patients are monitored for signs of residual disease and
`local or distant recurrences using radiological imaging
`during post-treatment follow-up.73–75 These methods are
`costly, often requiring contrast media (exposing patients
`to doses of radiation), and thus cannot be used for fre-
`quent monitoring. Another important consideration is
`that these imaging techniques have limited sensitivity for
`the detection of micrometastases.76,77
`In a landmark paper, Diehl et al.27 showed that by
`monitoring tumour-specific aberrations (including APC,
`TP53 and KRAS) in the plasma of patients with colorectal
`cancer post-surgery it was possible to identify disease
`recurrence with almost 100% sensitivity and specifi-
`city. Patients with residual disease were also identified
`based on the persistence of tumour-associated genetic
`aberrations in cfDNA immediately after surgery in each
`case of incomplete resection. Other reports, including
`studies in breast and lung cancer, and oral squamous-
`cell carcinoma, demonstrated a consistent relationship
`between disease recurrence and the reappearance of
`certain tumour aberrations, including KRAS, APC and
`TP53 mutations as well as allelic imbalances.34,50,51,78
`Monitoring single tumour-associated aberrations
`in the blood is not only effective for the stratification
`of patients with minimal residual disease and to iden-
`tify patients likely to relapse, but also to identify those
`with dormant disease. Dormancy is a common feature of
`many cancers, including breast, melanoma, renal cancer
`and non-Hodgkin lymphoma, and cannot be detected
`by standard methods.79 In 50 patients with breast cancer,
`tracking tumour-specific copy-number aberrations in
`the plasma pre-operatively and post-operatively demon-
`strated that tumour-specific copy-number aberrations
`persisted up to 12 years post diagnosis.80 As DNA is
`cleared from circulation within 30 min,81 the presence
`of DNA might reflect persistent dormant cells cycling
`between replication and cell death; however, relapse was
`not detected in any of the 50 patients. Nevertheless, pro-
`spective validation in a larger cohort and in other cancer
`models is warranted.
`
`REVIEWS
`
`Surgery
`
`Early relapse
`
`b
`
`Treatment
`
`Resistance
`
`A
`
`A
`
`Time
`
`Time
`
`a
`
`genetic aberation
`Tumour-associated
`
`Figure 2 | Monitoring tumour-specific aberrations to detect recurrence and
`resistance. The figure represents two hypothetical clinical scenarios in which
`genetic alterations in the blood plasma or serum can be tracked following
`a | surgery, to monitor signs of early relapse and b | treatment with a targeted
`agent (such as anti-EGFR monoclonal antibodies in CRCs) to monitor the
`emergence of resistant clones. ‘A’ represents the time of clinically-detectable
`recurrence or relapse. The blue line represents a ‘founder’ (early occurring)
`mutation that is present throughout the bulk of the tumour and reflects overall
`tumour burden. The red and green lines represent genetic aberrations associated
`with the growth and expansion of resistant clones.
`
`mutations (17% versus 41% at 6 months, and 0% versus
`24% at 12 months in KRAS mutant and wild type, respec-
`tively).53 However, a parallel study, conducted in 308
`patients with advanced-stage NSCLC, showed no corre-
`lation between KRAS mutations in the plasma and prog-
`nosis.60 BRAF mutations as assessed in serum samples
`have also been shown to effectively stratify 103 patients
`with melanoma in both early stage and advanced-stage
`settings.61 Nevertheless, results remain contradictory in
`these small patient populations.
`Neuroblastoma is a special case where amplification
`of the gene MYC-related oncogene (MYCN) has been
`identified as a genetic hallmark of aggressive disease,
`and this abnormality provides important information
`for patient treatment.62,63 Patient stratification according
`to MYCN status is important because improved treat-
`ment approaches, such as immunotherapy,64 were shown
`to significantly increase event-free and overall survival
`compared to standard treatments and might improve cur-
`rently unsatisfactory long-term survival rates in patients
`with high risk MYCN amplified neuroblastomas.65
`It is estimated that for 29% of all patients with
`neuro blastoma,65,66 MYCN status is unknown despite
`guidelines recommending genetic testing for patient
`stratification and treatment tailoring.67,68 Liquid biop-
`sies are able to accurately detect MYCN amplification
`in the serum of stage III to IV patients with neuro-
`blastoma with a sensitivity and specificity of 75–85% and
`100%, respectively.69–71
`
`476 | AUGUST 2013 | VOLUME 10
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`www.nature.com/nrclinonc
`
`00005
`
`

`

`REVIEWS
`
`The sensitivity of detecting single tumour- specific
`genetic aberrations in serum or plasma is very vari-
`able, particularly in settings of low tumour burden.30,40
`Monitoring tumour-specific genetic aberrations using
`next-generation sequencing (NGS) techno logy might
`offer the opportunity to increase such sensitivity.
`Genome-wide sequencing of plasma cfDNA has been
`carried out in patients diagnosed with hepato cellular
`carcinoma,9 and by calculating the fractional concentra-
`tion of tumour-derived DNA (mutations or copy-number
`aberrations) with respect to total DNA, the research-
`ers were able to demonstrate a significant reduction in
`tumour-derived DNA following surgery. This finding
`highlights one of the inherent technical difficulties of
`using NGS platforms in that the study demonstrated
`a reduction (threefold to 60-fold), not eradication, of
`tumour-derived DNA following surgery. As none of the
`patients relapsed during the course of the study it was
`assumed that surgery was curative and persistent tumour-
`associated aberrations were related to sequencing errors.
`Although residual disease is a possible explanation,
`tumour-related mutations have also been observed in
`healthy individuals and smokers suggesting that genetic
`aberrations might be present at low frequencies even in
`the absence of cancer.82–84 This possibility will be clarified
`with the use of increasingly sensitive techniques.
`
`Difficult-to-diagnose cancers
`Liquid biopsies can be used to assist in the clinical
`manage ment of difficult-to-diagnose patients with
`advanced-stage cancer, as is the case for bone metasta-
`ses,29 some pancreatic cancers and deep pelvic masses.38
`For example, in a study in which targeted deep sequenc-
`ing of cancer-related genes (including TP53, PIK3CA
`and KRAS) was carried out on cfDNA in a patient who
`had previously undergone surgery to resect synchro-
`nous cancers of the bowel and ovary, it was shown that
`on relapse the meta stasis was derived from the original
`ovarian cancer (owing to the presence of a R273H TP53
`mutation and absence of other mutations).38 A biopsy
`in this case was not possible and had the informa-
`tion derived from ctDNA been available immediately
`unneces sary delay or uncertainty over treatments might
`have been avoided.
`
`Open issues
`In theory, the use of liquid biopsies to identify early
`relapse or progression is very attractive, but there are still
`some open issues. The most intriguing is the possibility
`that tumour-associated aberrations can be lost or gained
`over the monitoring period or in response to drug pres-
`sure.29,42,85 This variation has been ascribed to changes in
`clonal sub-populations and might occur in response to
`treatment, adaptation to environmental changes or an
`increase in metastatic potential.86 This observation is also
`interesting in cancers that have long latency periods and
`liquid biopsies could be used to establish genetic markers
`of relapse.
`Attempts to monitor early relapse or progression
`require the selection of appropriate ‘founder’ genetic
`
`aberrations (such as APC in colorectal cancer or KRAS in
`pancreatic cancer) that are likely to be present from the
`initiation of tumori genesis, but provide little to no selec-
`tive advantage (so-called ‘passive’ mutations).87–90 For
`example, EGFR mutation frequency decreases follow-
`ing chemotherapy treatment and would, therefore, not
`represent a suitable marker of overall tumour burden in
`a patient receiving chemotherapy.85 The less responsive
`the tumour-specific aberration is to therapy the more
`suitable the marker. The plethora of passive and active
`mutations that accumulate throughout the lifetime of
`a tumour means that selecting multiple candid ates is
`not simple.91 Monitoring relapse or progression using
`tumour-associated genetic aberrations in the blood
`requires detailed knowledge of the genes most frequently
`mutated in each type of cancer. These are now available
`from large cancer genomics projects such as The Cancer
`Genome Atlas92 and the International Cancer Genome
`Consortium.93 A rational way to guide the selection of
`marker lesions might be to choose those that occur early
`during tumour progression, as they would presumably
`be present in the whole population as opposed to a
`clonal sub-population.2
`
`Prediction of response to treatment
`The presence or absence of a single genetic alteration
`in tumour DNA is currently employed to guide clini-
`cal decision making for a number of targeted agents (for
`example, EGFR mutations for gefitinib in NSCLC, BRAF
`mutations for vemurafenib in melanoma, KRAS muta-
`tions for cetuximab or panitumumab in colorectal cancer,
`ALK rearrangements for crizotinib in NSCLC).94–96
`Ever-increasing numbers of genomic alterations are
`being tested as putative predictive biomarkers in clinical
`trials of novel anticancer therapies.97
`Targeted agents are often used or tested in patients
`with advanced-stage disease (often with multiple metas-
`tases). However, the associated molecular test is often
`performed on archived tumour tissues collected years
`before. This situation is suboptimal as it might not reflect
`the genomic landscape of current disease. In the context
`where a new biopsy cannot be obtained, ctDNA might
`provide superior molecular information compared to
`archival tissue DNA for determining the current cancer
`molecular status. Potentially, a ‘liquid biopsy’ could
`obviate the need for tumour tissue DNA in metastatic
`patients. Gevensleben et al.98 have recently described the
`optimization of a digital PCR method to detect HER2
`amplification in ctDNA of patients with metastatic breast
`cancer using a development cohort (65 patients) and an
`independent validation cohort (58 patients). The poten-
`tial clinical use of such a tool would be in the analysis of
`HER2 status in patients who might not have received a
`biopsy of recurrent breast cancer as part of their routine
`care, as is often the case. Such patients with metastatic
`disease that are identified as acquiring HER2-positive
`status might subsequently benefit from HER2 targeted
`therapeutics, such as trastuzumab. The researchers also
`showed that three patients with metastatic breast cancer
`acquired a HER2-positive status on disease recurrence
`
`NATURE REVIEWS | CLINICAL ONCOLOGY
`
`VOLUME 10

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