CLB-09002; No. of pages: 5; 4C: Clinical Biochemistry xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Clinical Biochemistry journal homepage: www.elsevier.com/locate/clinbiochem

Review

2Q1

The translational potential of circulating tumour DNA in oncology

3Q2

K.M. Patel a,b, D.W.Y. Tsui a,⁎

4 5

a

6

a r t i c l e

7 8 9 10 11

Article history: Received 1 December 2014 Received in revised form 2 April 2015 Accepted 3 April 2015 Available online xxxx

12 13 14 15 16 30 17 18 19 20

Keywords: Circulating nucleic acids Cancer DNA Oncology Urine Plasma Blood Next generation sequencing

a b s t r a c t

R O

i n f o

O

Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK Department of Academic Urology, University of Cambridge Hospitals, Box 243, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ , UK

C

34 32 31 33

45

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

E

. . . . . . . .

. . . . . . . .

R

Introduction . . . . . . . . . . . . De novo mutation detection in blood . Understanding resistance mechanisms Other bodily fluids . . . . . . . . . Low volume disease . . . . . . . . . Discussion . . . . . . . . . . . . . Acknowledgements . . . . . . . . . References . . . . . . . . . . . . .

R

37 38 39 40 41 42 43 44

Contents

N C O

36 35

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

Introduction

47 48

In 1977, Leon et al. discovered that serum circulating cell-free DNA (cfDNA) levels were higher in patients with cancer [1]. This initial work, focusing on total levels of cfDNA, eventually encouraged the investigation of circulating nucleic acids as a biomarker of cancer. However, total levels of cfDNA were found to be insensitive [2–4] and nonspecific [2,5,6], in part, due to great variability between individuals and, that cfDNA levels are raised in a number of conditions including pregnancy [7] and myocardial infarction [8].

51 52 53 54

U

46

49 50

21 22 23 24 25 26 27 28 29

E

D

P

The recent understanding of tumour heterogeneity and cancer evolution in response to therapy has raised questions about the value of historical or single site biopsies for guiding treatment decisions. The ability of ctDNA analysis to reveal de novo mutations (i.e., without prior knowledge), allows monitoring of clonal heterogeneity without the need for multiple tumour biopsies. Additionally, ctDNA monitoring of such heterogeneity and novel mutation detection will allow clinicians to detect resistant mechanisms early and tailor treatment therapies accordingly. If ctDNA can be used to detect low volume cancerous states, it will have important applications in treatment stratification post-surgery/radical radiotherapy and may have a role in patient screening. Mutant cfDNA can also be detected in other bodily fluids that are easily accessible and may aid detection of rare mutant alleles in certain cancer types. This article outlines recent advances in these areas. © 2015 The Canadian Society of Clinical Chemists. Published by Elsevier Inc. All rights reserved.

T

b

F

1

⁎ Corresponding author. E-mail address: [email protected] (D.W.Y. Tsui).

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

0 0 0 0 0 0 0 0

Tumour DNA contains specific somatic alterations. DNA fragments which contain these tumour-specific somatic mutations can be detected in the blood, and hence are called circulating tumour DNA (ctDNA) [9]. It is challenging to identify ctDNA fragments, because they are surrounded by multiple copies of normal genomic DNA. One strategy is to use a personalised approached, where one could first identify mutations in tumour and subsequently design mutation specific assays to detect ctDNA in plasma [10,11]. A number of studies have demonstrated that the presence of ctDNA and, the dynamics of ctDNA in plasma, reflect tumour burden. For example, in 2008, Diehl et al. evaluated 162 plasma samples from 18 colorectal cancer patients, to demonstrate that levels of ctDNA increased with tumour burden, and that ctDNA kinetics were more sensitive than serum carcino-embroyonic antigen (CEA) for monitoring disease

http://dx.doi.org/10.1016/j.clinbiochem.2015.04.005 0009-9120/© 2015 The Canadian Society of Clinical Chemists. Published by Elsevier Inc. All rights reserved.

Please cite this article as: Patel KM, Tsui DWY, The translational potential of circulating tumour DNA in oncology, Clin Biochem (2015), http:// dx.doi.org/10.1016/j.clinbiochem.2015.04.005

55 56 57 58 59 60 61 62 63 64 65 66 67 68

2

81

De novo mutation detection in blood

82

Substantial work in the field has demonstrated that ctDNA can be detected by tracking tumour-specific point mutations or structural rearrangements, in many cancer types [14,16–20]. However, it is now apparent that the mutational characteristics of tumours are not static and continually evolve in response to various selection pressures (e.g. initiation of targeted therapy) [21]. In order for assays to remain relevant as tumour clones evolve, a personalised approach would ideally require multi-site biopsies repeated sequentially. Furthermore, due to the extent of intra-tumour heterogeneity as demonstrated by several studies [22–24], a single site biopsy risks missing clinically important mutations from a heterogeneous tumour. For these reasons, a personalised ctDNA detection approach based on the detection of mutations found in tumour samples is challenging. There is therefore a need for non-invasive detection of mutations without prior knowledge of their existence directly from the blood, we term this de novo mutation detection. In 2012 Forshew et al. demonstrated that mutations can noninvasively be detected de novo through targeted plasma re-sequencing [15]. They proposed a targeted sequencing approach termed TAm-Seq, which allowed the re-sequencing of approximately 6000 nucleotides whilst maintaining high depth analysis. In a proof-of-concept example, the authors re-sequenced tumour tissue from a right oophorectomy specimen, taken from a patient with ovarian cancer at the time of debulking surgery. The authors identified a TP53 mutation and tracked its ctDNA levels using TAm-Seq. As the cancer progressed, TAm-Seq analysis revealed the emergence of an EGFR mutation in plasma samples. This mutation was not found in the original oophorectomy specimen. However, on further investigation an identical EGFR mutation was present at low frequencies in samples taken from the omentum at the time of the initial debulking surgery. Forshew et al. hypothesised that as chemotherapy regimes restrained the growth of other clones, the resistant EGFR clone, which was initially present only at low frequency, gained in dominance. They demonstrate that plasma analysis can identify heterogeneous clones from different sites of the body. Though TAm-Seq was able to detect de novo mutations directly from the plasma, Forshew et al. only targeted b 6 Kb of the genome [15]. To detect unselected genetic events that span across the genome, Leary et al. used massively paralleled sequencing (MPS) of the whole genome in 7 patients with colorectal cancer and 3 patients with breast cancer [25]. They developed a method termed personalised analysis of rearranged ends (PARE) [16] and detected at least one chromosomal rearrangement in the plasma of every cancer patient but not in the plasma of healthy volunteers. Similarly, in 2013 Chan et al. used “Shotgun” MPS of the whole genome to identify copy number variations and single nucleotide variants (SN) from the plasma of 4 patients with hepatocellular carcinoma [26]. Furthermore, they demonstrated the ability of MPS to track ctDNA level changes pre- and post-surgery. Interestingly, shotgun MPS of the plasma was also able to distinguish between tumour types in a patient with synchronous breast and ovarian tumours [26]. The above studies illustrate that ctDNA analysis, through de novo mutation

93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131

Understanding resistance mechanisms

165

Due to needle biopsy sampling bias and the limited availability of “research biopsies” in advanced cancer patients, the investigation of tumour resistance mechanisms have been challenging. However, its investigation can be of great clinical value as treatment regimens can be tailored to target resistant mechanisms or, treatment intervals introduced as resistant clones are detected and gain dominance. The hypothesis is that plasma is able to capture DNA from all tumour clones. If that holds true, its analysis may demonstrate the evolutionary process tumours undergo to evade targeted therapies and possibly traditional chemotherapies. In 2012, Misale et al. observed the emergence of resistance conferring KRAS mutations in plasma [29]. They analysed ctDNA from the plasma of 10 colorectal cancer patients taking cetuximab before and after progression. Using BEAMing (bead, emulsion, amplification and magnetics), they found KRAS mutations or amplifications in the plasma of 60% of the patients who progressed. Importantly the emergence of resistant clones occurred up to 10 months prior to radiographic confirmation of disease progression. Mutations in the plasma were concordant with KRAS mutations found in subsequent metastatic tumour biopsies. In a parallel study, Diaz et al. described the ctDNA analysis of 28 patients with chemorefractory metastatic colorectal cancer receiving panitumumab. 38% of patients whose initial tumours had wild-type KRAS developed mutant KRAS over the next 6 months. Through ctDNA levels and tumour growth analysis Diaz et al. calculated that KRAS mutants were likely to be present at low quantities prior to initiation of panitumumab [20] and lend weight to the omental EGFR mutant findings of Forshew et al. [15]. In another example, Oxnard et al. used droplet dPCR [30] to analyse plasma ctDNA in 9 patients with NSCLC who were treated with erlotinib therapy [31]. Plasma levels of EGFR mutation T790M were undetectable prior to erlotinib therapy in this group. Initiation of erlotinib led to a

166

O

R O

P

D

91 92

132 133

T

89 90

C

87 88

E

85 86

R

83 84

R

78 79

O

76 77

C

75

N

73 74

U

71 72

detection, can continue to track disease burden as tumours evolve, without the need for re-biopsy. However, in the studies outlined above the expense of whole genome MPS would limit analysis to a small number of samples. For this reason, whole-genome sequencing (WGS) approaches for routinely detecting ctDNA in patient populations are currently, prohibitively expensive. Although low depth, and therefore reduced cost, WGS approaches have been successful at detecting copy number variations [26,27], a higher depth of coverage is often required to detect rearrangements at high resolution or SNVs directly from plasma DNA. Furthermore, where low mutant:wild type allele frequencies exist, e.g. in early stage disease, an even higher depth of coverage would be necessary to detect ctDNA fragments. In addition, WGS approaches detect a higher ratio of intronic or passenger mutations than targeted re-sequencing [26]. The clinical significance of passenger mutations is currently unknown and often not targetable. Another approach demonstrated by Murtaza et al. is to perform whole exome sequencing (WES) of the plasma to track tumour evolution in response to therapy [28]. This proof-of-concept study involved 6 patients with metastatic tumours with plasma collected at the beginning of treatment and at the time of relapse. Subsequent re-sequencing and variant analysis revealed that by comparing the relative representation of mutations in pre- and post-relapse samples, one could identify enrichment of mutations that may drive resistance. This work demonstrated that a WES approach would allow the detection of clinically relevant mutations at a lower cost than WGS [28]. WGS can screen a larger spectrum of the genome but is currently too expensive for routine use to detect SNVs, whereas WES approaches allow more in-depth interrogation of multiple regions but is less sensitive to identifying copy number changes. As the cost of sequencing continues to fall, the price of WGS of plasma is likely to become more palatable. At such a time, WGS may become routine for the analysis of de novo mutations in serial plasma samples.

F

80

burden in colorectal cancer [12]. In 2013, Dawson et al. investigated women with metastatic breast cancer using a similar personalised approach. Tumour tissue mutations were identified in 30 women using next generation re-sequencing and subsequently, digital PCR (dPCR) [13,14] and tagged-amplicon deep sequencing (TAm-Seq) [15] were used to detect ctDNA in 29 out of the 30 women. Computed tomograms were compared with levels of ctDNA, cancer antigen 15-3 (CA15-3) and circulating tumour cells (CTCs) taken sequentially. Overall, ctDNA was able to detect changes in tumour burden earlier and with greater sensitivity than the standard biomarker (CA15-3) or CTCs [11]. These results suggest that ctDNA has the potential to be used as a measure of tumour response in a non-invasive way.

E

69 70

K.M. Patel, D.W.Y. Tsui / Clinical Biochemistry xxx (2015) xxx–xxx

Please cite this article as: Patel KM, Tsui DWY, The translational potential of circulating tumour DNA in oncology, Clin Biochem (2015), http:// dx.doi.org/10.1016/j.clinbiochem.2015.04.005

134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164

167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195

K.M. Patel, D.W.Y. Tsui / Clinical Biochemistry xxx (2015) xxx–xxx

220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 Q3 251 252 Q4 253 254 255 256 257 258 259

C

218 219

E

213 214

R

Low volume disease

284

F

211 212

R

209 210

N C O

207 208

U

205 206

O

Much ctDNA research to-date focuses on plasma and serum derived samples. However, frequent blood sampling in patients who are already prone to anaemia of chronic disease is not ideal. Interestingly, tumourspecific nucleic acids have been detected in other bodily fluids, including stool [13], urine [33], saliva [34], cerebrospinal fluid [35] and pleural fluid [36]. Particular fluids may concentrate mutant cfDNA from regional drainage and may facilitate low volume disease mutant cfDNA detection. For example, mutated DNA fragments from localised oral cancers are hypothesised to be more concentrated in saliva than in the circulation. Additionally, many bodily fluids are freely available and found in abundance, particularly urine. This may allow more frequent and more voluminous sampling, thus aiding low volume disease detection. The presence of genetic material in the urine has long been established, though initially from urinary cells [37]. In 1999, Zhang et al. demonstrated the presence of SRY gene (found on the Y chromosome) in the urine supernatant of female recipients of male donor renal transplants [38]. By 2000, Botezatu et al. demonstrated that some genomic DNA escaped into the urine through renal filtration [39]. They demonstrated this by subcutaneously injecting mice with radiolabelled human DNA and collecting urine over the next 3 days. Collected urine was determined to contain human cfDNA through its radioactivity and PCR amplification of human specific Alu repeat sequences Subsequently, by performing nested PCR to detect highly repetitive regions located on the Y chromosome, Botezatu et al. demonstrated the presence of male DNA in the urine of pregnant women carrying male foetuses [39]. In 2004, Su et al. demonstrated the presence of mutant cfDNA in the urine of colorectal carcinoma patients [40]. Using restriction enriched PCR with primer pairs targeting a known mutation in KRAS codon 12, they were able to selectively amplify mutant cfDNA in the fractionated urine of colorectal cancer patients. Interestingly, they also performed size analysis on urinary DNA using polyacrylamide gels and found two main bands of cfDNA. The first was a band approximately 150–250 nucleotides long, which they hypothesised was derived from the circulation through renal filtration, the second and much larger band represented DNA N 1 kilobase long, which they hypothesised originated in the lining of the urinary tract [40]. In a subsequent study by Tsui et al., next generation sequencing approaches were used to assess the size of DNA present in the urine of pregnant women. They found that maternal urinary cfDNA had a predominant peak at 29 bp and, that the majority of maternal cfDNA was below 100 bp in length [41]. In addition, next generation sequencing approaches have been used to detect mutant cfDNA derived from the urine of cancer patients. In

203 204

R O

217

202

260 261

P

Other bodily fluids

200 201

2012, Millholland et al. targeted the known hotspots in FGFR3 exons 7, 10 and 15 to detect FGFR3 mutations in urinary DNA [33]. They reported a concordance of 91% by detecting mutant urinary cfDNA in 10 out of 11 patients with FGFR3 mutations in their primary tumours. In a separate group, the investigators assessed the urine of 43 known bladder cancer patients and found mutated FGFR3 in 24 of 43 cases [33]. As the mutation status of the primary tumour was not reported, no comment can be made on the concordance of their urinary assay with tumour mutations, though it appears proportional to the prevalence of FGFR3 mutations in bladder cancer populations detected in other studies [42,43]. Similar findings were also presented in previous studies relying on molecular assays to analyse FGFR3 mutations [44,45]. The ability of these assays to detect bladder cancer mutations as a whole was, however, low due to the sole focus on FGFR3, despite some studies selecting patients who had FGFR3 aberrations in their initial tumour sample. The findings of Botezatu and Su demonstrate that cfDNA filtered through the kidney is available for mutational analysis in urine, a bodily fluid that is readily available and abundant. However, quantification of peripheral fluid mutant cfDNA may prove difficult to interpret, with additional factors such as the hydration state, renal disease and presence of bladder outflow obstruction in the case of urine, likely to affect mutant cfDNA levels. Further studies would need to take into consideration these factors to systematically evaluate the potential of urine for mutant cfDNA analysis.

D

216

198 199

T

215

drop in ctDNA levels of the sensitising EGFR mutations in all patients. However, in 6 of the 9 patients, T790M EGFR mutant levels began to increase, 4–24 weeks prior to progression being confirmed by RECIST criteria [31]. As part of the largest ctDNA study reported to date, Bettegowda et al. analysed plasma samples pre- and post-EGFR blockade to detect a broad range of resistance mechanisms in a subgroup of 24 colorectal cancer patients [32]. They showed that the ctDNA of 23 patients had at least one new mutation in mitogen-activated protein kinase pathway genes and that 50% of these new mutations occurred in KRAS codon 12. Currently, only a small number of cases in only a few cancer types have been investigated using methods with high analytical sensitivity (e.g. dPCR, Safe-Seq, and TAm-Seq) and further large-scale studies are required. However, the above studies demonstrate that ctDNA can be utilised to detect tumour resistance mechanisms. Furthermore, the work by Murtaza et al. indicate that less biased, WES approaches for ctDNA analysis could be employed to detect unexpected resistance mechanisms. When coupled with the growing number of targeted therapeutic options, the potential of using ctDNA to guide sequential treatment is promising.

As the sensitivity of ctDNA analysis improves, the detection of ctDNA at lower levels will become feasible. Many cancer patients still present too late for curative therapies and our best chance of improving cancer mortality rates lies in the early detection of cancer [46]. However at present, published studies have largely focused on ctDNA analysis in the advanced cancer setting, where levels of mutant:wild type allele fractions are around 10–50% [10,15,28]. For earlier stages, the mutant:wild type allele fractions are likely to be lower. Despite this, ctDNA has been detected in the plasma of multiple cancers types at an early stage. In Diehl et al.'s seminal work, mutant:wild type allele frequencies ranging from 0.001% to 0.12% were detected in patients with localised colorectal cancer. Of note, ctDNA was also detected from plasma samples in their control group of patients with colorectal adenomas, where mutant:wild type allele frequencies of 0.001–0.02% were reported [10]. The ability to detect ctDNA in patients with adenomatous colorectal polyps has important implications. Colorectal adenomas are precursors to adenocarcinoma and as such, often require further treatment. That ctDNA was detected at this pre-cancerous stage suggests a potential role of ctDNA in early diagnosis or even for population screening. In more recent work, Bettegowda et al. reported a range of 0 (notdetected) to 135,000 mutant fragments/5 mL of blood in 223 patients with 12 tumour types, all with localised disease [32]. By detecting mutations in tumour tissue and subsequently searching for these in matched plasma samples, they detected ctDNA in 55% (122/223) of patients [32]. Furthermore, by using a droplet dPCR assay, Beaver et al. were able to detect PIK3CA mutations in the pre-operative plasma samples of 14/ 15 patients who had low volume breast cancer and PIK3CA mutations in their tumour samples [47]. For 10 patients, blood samples were collected post-operatively and 5 had persistently detectable ctDNA. Indeed, one of these patients went on to have cancer recurrence. If applicable to other cancers, low volume ctDNA analysis could monitor the emergence of early recurrence following surgical resection or other radical therapies and help distinguish between patients who require further adjuvant therapy and patients who are essentially cured. Presently, ctDNA has only been detected in early disease in a small number of cancers [18,32,47]. Early evidence suggests that detecting levels in early disease in certain cancers e.g. glioma, will be extremely challenging [32]. Additionally, it is uncertain whether alerting

E

196 197

3

Please cite this article as: Patel KM, Tsui DWY, The translational potential of circulating tumour DNA in oncology, Clin Biochem (2015), http:// dx.doi.org/10.1016/j.clinbiochem.2015.04.005

262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283

285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323

354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387

C

352 353

E

345 346

R

343 344

R

341 342

O

339 340

C

337 338

N

335 336

U

333 334

Acknowledgements

442

The authors wish to thank the Addenbrooke's Charitable Trust, the Royal College of Surgeons and the Cambridge Cancer Centre for their support. We wish also to thank Dr. Nitzan Rosenfeld, Dr. Tim Forshew and Dr. Florent Mouliere for comments and suggestions.

443

References

447

F

The analysis of ctDNA presents many opportunities to improve cancer management. Early detection of ctDNA could alert clinicians to the presence of sinister disease and prompt curative treatment. Indeed, in several cancer types there are known mutations in precancerous states (e.g. APC in colorectal adenomas [51] and BRAF in benign naevi of the skin [52]). However, there is a risk that early detection of some mutations may lead to over-investigation (particularly to localise the source of the mutation) and overtreatment of patients, who may have never progressed to invasive cancer. At present, the focus of ctDNA research remains on advanced cancers where ctDNA fractions in plasma are higher. The ability of ctDNA to track de novo mutations will deliver more precise cancer monitoring and uncover resistant mechanisms. When metastatic tumour biopsy is not available, ctDNA would provide valuable information for therapy stratification. The limitations of tumour re-biopsy were highlighted in a recent study by Andre et al. who from 2011 to 2012, conducted a large-scale multi-centred trial (SAFIR01/UNICANCER) to investigate the feasibility of using genomic information to stratify patients to targeted therapies. Biopsy samples from metastases were sent to 5 centres for analysis [53]. The primary endpoint was to stratify 30% of patients to targeted therapies (including trial drugs) using array CGH and Sanger sequencing. This was not achieved, with only 13% (55 out of 423) of enrolled patients being suitable to receive targeted therapy. Several important limitations for this were highlighted: Firstly, array CGH analysis required a large amount of DNA and low yield from biopsies prevented analysis in 91 cases. Secondly, of 195 patients with targetable results only 55 could receive targeted therapies. Several of the challenges encountered by the SAFIR01 team might be addressed by integrating ctDNA analysis into the trial: firstly, plasma can be collected more regularly and easily. Secondly, methods for ctDNA analysis require less input DNA than traditional Sanger sequencing or array CGH. Thirdly, the ability of ctDNA to display clonal heterogeneity and track clonal progression, could potentially overcome spatial and temporal heterogeneity issues encountered when relying on tumour biopsies. Lastly, blood sampling is safer, and has fewer complications when compared to tumour biopsy and the use of sequential sampling could track response to treatment more closely or may have alerted clinicians to the development of resistant mechanisms. However, several aspects of ctDNA

331 332

O

350 351

330

R O

Discussion

328 329

388 389

P

349

326 327

analysis require addressing prior to its adoption in the clinic. Plasma processing and data analysis require standardisation to allow largescale, multi-institutional clinical trials. Such trials would need to clearly demonstrate patient benefit before ctDNA analysis can be used to guide treatment decisions. Despite the above, Rothé et al. performed hotspot mutation analysis on 17 breast cancer patients to ascertain whether ctDNA can be used to guide therapeutic decisions [54]. Metastatic tumour and plasma samples were taken at the same time-point and Rothé et al. reported a 76% concordance (13/17 patients) between these samples. Two patients had mutations detected in plasma at relatively high frequencies (12% and 14.3%) but not the tumour samples. The remaining two patients had mutations detected in the tumour but not the plasma. Further analysis of these plasma samples by targeted illumina sequencing revealed mutations identical to that found in the tumour, although present at low frequency. Interestingly, for 9 out of 69 tumour samples, the investigators were unable to perform NGS analysis where as they were able to for all 31 plasma samples received. The investigators conclude that plasma ctDNA analysis is a suitable alternative to metastatic biopsy for molecular screening. Furthermore, in 2014, Douillard et al. published the results of a comparison between ctDNA analysis and tumour biopsy DNA analysis of EGFR mutation status in 1060 advanced lung cancer patients. They found the concordance of EGFR mutations between matched tumour and plasma to be 94.3%, with the test yielding a sensitivity of 65.7% and specificity of 99.8% [55]. Furthermore, patients who have activating EGFR mutations detected in tumour or plasma samples demonstrated similar progression free survival (9.7 vs 10.2 months) after gefitinib treatment [55], supporting the use of ctDNA analysis to initiate targeted therapies. In addition, cell-free tumour DNA is present in alternative bodily fluids, some of which are of much higher abundance than tumour biopsy or plasma, such as urine. These bodily fluids may be more abundant than plasma and maybe more easily collected. Analysis of these fluids may facilitate the detection of rare mutated DNA fragments. The choice of specific fluids to analyse would differ across cancer types, depending on the proximity of the fluid to the tumour sites and clinical context. In summary, there is increasing evidence that ctDNA analysis may have an important role at several stages of disease. Due to their short half-life, ctDNA profiles can present a snapshot of current clonal burden and rapidly assess tumour response to therapies. Indeed, many of the protein based tumour markers currently used to asses treatment response in the clinic (e.g. PSA, CA125, CEA, αFP) have a half-life measured in days [56], whereas the half-life of cfDNA is measured in minutes to hours [12,57]. We envision that, the ability of ctDNA to detect de novo mutations could overcome the problem of mutational profiling in the presence of continual tumour evolution, thus improving our understanding resistance mechanisms and allow us to monitor tumour response in real-time. Furthermore, improvements in the detection of ctDNA present at low volumes in plasma and other bodily fluids, may lead to an earlier diagnosis, where intervention can still lead to cure or improve our ability to stratify patients to adjuvant therapy following radical treatments. What is certain is that as the cost of massively paralleled sequencing reduces, ctDNA analysis will become more readily available and has the potential to improve cancer care.

T

347 348

physicians to the early presence of cancer through ctDNA analysis will translate to improved outcomes. Indeed, the use of prostate specific antigen (PSA) to detect early prostate cancer has resulted in overtreatment, with disease specific mortality improved in only high-risk groups [48]. Furthermore in the future, ctDNA detection may be “too sensitive” and detect mutations from cancers before they can be localised. Indeed by using restriction enriched PCR Gormally et al. were able to detect KRAS mutations in 13 out of 1098 “healthy volunteers”. However, in this instance, 5 “healthy volunteers” were subsequently diagnosed with bladder cancer and a further volunteer diagnosed with upper aero-digestive tract cancer within 25 months [49], conceivably these tumours may have been present at microscopic level at the time of investigation and had superior localisation techniques been available, may have been detected and treated early. Furthermore, Chan et al. were able to detect Epstein Barr viral DNA in plasma, a known risk factor for nasopharyngeal carcinoma, when screening 1318 asymptomatic volunteers [50]. They detected viral DNA in 69 volunteers and on further investigation found nasopharyngeal cancer in 3 previously asymptomatic individuals, demonstrating the utility of plasma-based cancer screening [50]. It is however, likely that robust ctDNA detection will only be possible in certain cancers and, that early detection may be possible in only a subset of these. Although, where ctDNA is detectable at an early stage, it has the potential to reduce mortality through more patients being amenable to curative surgery.

D

324 325

K.M. Patel, D.W.Y. Tsui / Clinical Biochemistry xxx (2015) xxx–xxx

E

4

390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441

444 445 446 Q5

[1] Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients 448 and the effect of therapy. Cancer Res 1977;37:646–50. 449

Please cite this article as: Patel KM, Tsui DWY, The translational potential of circulating tumour DNA in oncology, Clin Biochem (2015), http:// dx.doi.org/10.1016/j.clinbiochem.2015.04.005

K.M. Patel, D.W.Y. Tsui / Clinical Biochemistry xxx (2015) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[31] Oxnard GR, Paweletz CP, Kuang Y, Mach SL, O'Connell A, Messineo MM, et al. Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin Cancer Res 2014;20:1698–705. [32] Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang YX, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6. [33] Millholland JML S, Fernandez CA, Shuber AP. Detection of low frequency FGFR3 mutations in the urine of bladder cancer patients using next-generation deep sequencing. Res Rep Urol 2012;4:33–40. [34] Li Y, Zhou X, St John MA, Wong DT. RNA profiling of cell-free saliva using microarray technology. J Dent Res 2004;83:199–203. [35] Pan W, Gu W, Nagpal S, Gephart MH, Quake SR. Brain tumor mutations detected in cerebral spinal fluid. Clin Chem 2015;61:514–22. [36] Soh J, Toyooka S, Aoe K, Asano H, Ichihara S, Katayama H, et al. Usefulness of EGFR mutation screening in pleural fluid to predict the clinical outcome of gefitinib treated patients with lung cancer. Int J Cancer 2006;119:2353–8. [37] Sidransky D, Von Eschenbach A, Tsai YC, Jones P, Summerhayes I, Marshall F, et al. Identification of p53 gene mutations in bladder cancers and urine samples. Science 1991;252:706–9. [38] Zhang J, Tong KL, Li PK, Chan AY, Yeung CK, Pang CC, et al. Presence of donor- and recipient-derived DNA in cell-free urine samples of renal transplantation recipients: urinary DNA chimerism. Clin Chem 1999;45:1741–6. [39] Botezatu I, Serdyuk O, Potapova G, Shelepov V, Alechina R, Molyaka Y, et al. Genetic analysis of DNA excreted in urine: a new approach for detecting specific genomic DNA sequences from cells dying in an organism. Clin Chem 2000;46:1078–84. [40] Su YH, Wang M, Brenner DE, Ng A, Melkonyan H, Umansky S, et al. Human urine contains small, 150 to 250 nucleotide-sized, soluble DNA derived from the circulation and may be useful in the detection of colorectal cancer. J Mol Diagn 2004;6: 101–7. [41] Tsui NB, Jiang P, Chow KC, Su X, Leung TY, Sun H, et al. High resolution size analysis of fetal DNA in the urine of pregnant women by paired-end massively parallel sequencing. PLoS One 2012;7:e48319. [42] Lamy A, Gobet F, Laurent M, Blanchard F, Varin C, Moulin C, et al. Molecular profiling of bladder tumors based on the detection of FGFR3 and TP53 mutations. J Urol 2006; 176:2686–9. [43] Sjodahl G, Lauss M, Gudjonsson S, Liedberg F, Hallden C, Chebil G, et al. A systematic study of gene mutations in urothelial carcinoma; inactivating mutations in TSC2 and PIK3R1. PLoS One 2011;6:e18583. [44] van Kessel KEM, Kompier LC, de Bekker-Grob EW, Zuiverloon TCM, Vergouwe Y, Zwarthoff EC, et al. FGFR3 mutation analysis on voided urine samples to reduce cystoscopies and cost in non-muscle invasive bladder cancer surveillance: a comparison of three different strategies. J Urol 2012. [45] Zuiverloon TC, van der Aa MN, van der Kwast TH, Steyerberg EW, Lingsma HF, Bangma CH, et al. Fibroblast growth factor receptor 3 mutation analysis on voided urine for surveillance of patients with low-grade non-muscle-invasive bladder cancer. Clin Cancer Res 2010;16:3011–8. [46] Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz Jr LA, Kinzler KW. Cancer genome landscapes. Science 2013;339:1546–58. [47] Beaver JA, Jelovac D, Balukrishna S, Cochran RL, Croessmann S, Zabransky DJ, et al. Detection of cancer DNA in plasma of patients with early-stage breast cancer. Clin Cancer Res 2014;20:2643–50. [48] Wilt TJ, Brawer MK, Jones KM, Barry MJ, Aronson WJ, Fox S, et al. Radical prostatectomy versus observation for localized prostate cancer. N Engl J Med 2012;367: 203–13. [49] Gormally E, Vineis P, Matullo G, Veglia F, Caboux E, Le Roux E, et al. Tp53 and KRAS2 mutations in plasma DNA of healthy subjects and subsequent cancer occurrence: a prospective study. Cancer Res 2006;66:6871–6. [50] Chan KC, Hung EC, Woo JK, Chan PK, Leung SF, Lai FP, et al. Early detection of nasopharyngeal carcinoma by plasma Epstein–Barr virus DNA analysis in a surveillance program. Cancer 2013;119:1838–44. [51] Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, et al. Apc mutations occur early during colorectal tumorigenesis. Nature 1992;359:235–7. [52] Miller AJ, Mihm MC. Melanoma. N Engl J Med 2006;355:51–65. [53] Andre F, Bachelot T, Commo F, Campone M, Arnedos M, Dieras V, et al. Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol 2014;15:267–74. [54] Rothé F, Laes J-F, Lambrechts D, Smeets D, Vincent D, Maetens M, et al. Plasma circulating tumor DNA as an alternative to metastatic biopsies for mutational analysis in breast cancer. Ann Oncol 2014;25:1959–65. [55] Douillard JY, Ostoros G, Cobo M, Ciuleanu T, Cole R, McWalter G, et al. Gefitinib treatment in EGFR mutated caucasian nsclc: circulating-free tumor DNA as a surrogate for determination of EGFR status. J Thorac Oncol 2014;9:1345–53. [56] Bidart J-M, Thuillier F, Augereau C, Chalas J, Daver A, Jacob N, et al. Kinetics of serum tumor marker concentrations and usefulness in clinical monitoring. Clin Chem 1999; 45:1695–707. [57] Thierry AR, Mouliere F, Gongora C, Ollier J, Robert B, Ychou M, et al. Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res 2010;38:6159–75.

E

T

[2] Jung K, Fleischhacker M, Rabien A. Cell-free DNA in the blood as a solid tumor biomarker-a critical appraisal of the literature. Clin Chim Acta 2010;411:1611–24. [3] Zhang R, Shao F, Wu X, Ying K. Value of quantitative analysis of circulating cell free DNA as a screening tool for lung cancer: a meta-analysis. Lung Cancer 2010;69: 225–31. [4] Gonzalez-Masia JA, Garcia-Olmo D, Garcia-Olmo DC. Circulating nucleic acids in plasma and serum (cnaps): applications in oncology. Onco Targets Ther 2013;6:819–32. [5] Bartoloni E, Ludovini V, Alunno A, Pistola L, Bistoni O, Crino L, et al. Increased levels of circulating DNA in patients with systemic autoimmune diseases: a possible marker of disease activity in Sjogren's syndrome. Lupus 2011;20:928–35. [6] Ha TT, Huy NT, Murao LA, Lan NT, Thuy TT, Tuan HM, et al. Elevated levels of cell-free circulating DNA in patients with acute dengue virus infection. PLoS One 2011;6: e25969. [7] Lui YY, Dennis YM. Circulating DNA in plasma and serum: biology, preanalytical issues and diagnostic applications. Clin Chem Lab Med 2002;40:962–8. [8] Antonatos D, Patsilinakos S, Spanodimos S, Korkonikitas P, Tsigas D. Cell-free DNA levels as a prognostic marker in acute myocardial infarction. Ann N Y Acad Sci 2006;1075:278–81. [9] Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, Yao SL. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol Biomarkers Prev 1994;3:67–71. [10] Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A 2005;102:16368–73. [11] Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, Chin SF, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med 2013;368:1199–209. [12] Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008;14:985–90. [13] Vogelstein B, Kinzler KW. Digital PCR. Proc Natl Acad Sci U S A 1999;96:9236–41. [14] Yung TK, Chan KC, Mok TS, Tong J, To KF, Lo YM. Single-molecule detection of epidermal growth factor receptor mutations in plasma by microfluidics digital PCR in non-small cell lung cancer patients. Clin Cancer Res 2009;15:2076–84. [15] Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DWY, Kaper F, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 2012;4:136ra68. [16] Leary RJ, Kinde I, Diehl F, Schmidt K, Clouser C, Duncan C, et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med 2010;2:20ra14. [17] McBride DJ, Orpana AK, Sotiriou C, Joensuu H, Stephens PJ, Mudie LJ, et al. Use of cancer specific genomic rearrangements to quantify disease burden in plasma from patients with solid tumors. Genes Chromosomes Cancer 2010;49:1062–9. [18] Chen X, Bonnefoi H, Diebold-Berger S, Lyautey J, Lederrey C, Faltin-Traub E, et al. Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin Cancer Res 1999;5:2297–303. [19] Otsuka J, Okuda T, Sekizawa A, Amemiya S, Saito H, Okai T, et al. Detection of p53 mutations in the plasma DNA of patients with ovarian cancer. Int J Gynecol Cancer 2004;14:459–64. [20] Diaz Jr LA, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012;486:537–40. [21] Van Allen EM, Wagle N, Sucker A, Treacy DJ, Johannessen CM, Goetz EM, et al. The genetic landscape of clinical resistance to raf inhibition in metastatic melanoma. Cancer Discov 2014;4:94–109. [22] Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012;366:883–92. [23] de Bruin EC, McGranahan N, Mitter R, Salm M, Wedge DC, Yates L, et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 2014;346:251–6. [24] Zhang J, Fujimoto J, Zhang J, Wedge DC, Song X, Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science 2014;346:256–9. [25] Leary RJ, Sausen M, Kinde I, Papadopoulos N, Carpten JD, Craig D, et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci Transl Med 2012;4:162ra54. [26] Chan KC, Jiang P, Zheng YW, Liao GJ, Sun H, Wong J, et al. Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencing. Clin Chem 2013;59:211–24. [27] Heitzer E, Ulz P, Belic J, Gutschi S, Quehenberger F, Fischereder K, et al. Tumor associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med 2013;5:30. [28] Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013;497:108–12. [29] Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012;486:532–6. [30] Taly V, Pekin D, El Abed A, Laurent-Puig P. Detecting biomarkers with microdroplet technology. Trends Mol Med 2012;18:405–16.

U

450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529

5

610

Please cite this article as: Patel KM, Tsui DWY, The translational potential of circulating tumour DNA in oncology, Clin Biochem (2015), http:// dx.doi.org/10.1016/j.clinbiochem.2015.04.005

530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 Q6 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609

The translational potential of circulating tumour DNA in oncology.

The recent understanding of tumour heterogeneity and cancer evolution in response to therapy has raised questions about the value of historical or sin...
333KB Sizes 1 Downloads 10 Views