33
Page 1 of 31 Detection of rare drug resistance mutations by digital PCR in a human influenza A 1 virus model system and clinical samples 2 3 Alexandra S. Whale a# , Claire Bushell a , Paul R. Grant b , Simon Cowen c , Ion Gutierrez- 4 Aguirre d , Denise M. O’Sullivan a , Jana Žel d , Mojca Milavec d , Carole A. Foy a , Eleni 5 Nastouli b , Jeremy A. Garson e , Jim F. Huggett a,e . 6 7 Molecular and Cell Biology Team, LGC Ltd, Teddington, UK a , Virology Laboratory, 8 Clinical Microbiology and Virology, University College London Hospital NHS 9 Foundation Trust, London, UK b , Statistics Team, LGC Ltd, Teddington, UK c , National 10 Institute of Biology, Ljubljana, Slovenia d , Department of Infection, Division of 11 Infection and Immunity, University College London, London, UK e . 12 13 Running Title: Evaluation of digital PCR for SNP detection. 14 15 #Address correspondence to Alexandra S. Whale, [email protected] 16 17 JCM Accepted Manuscript Posted Online 9 December 2015 J. Clin. Microbiol. doi:10.1128/JCM.02611-15 Copyright © 2015 Whale et al. This is an open-access article distributed under the terms of the Creative Commons Attribution-Noncommercial-ShareAlike 3.0 Unported license, which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original author and source are credited. on May 26, 2019 by guest http://jcm.asm.org/ Downloaded from

Downloaded from //jcm.asm.org/content/jcm/early/2015/12/07/JCM.02611-15.full.pdf · JCM Accepted Manuscript Posted Online 9 December 2015 ... 19 Digital PCR (dPCR) is being increasingly

  • Upload
    buikien

  • View
    220

  • Download
    0

Embed Size (px)

Citation preview

Page 1 of 31

Detection of rare drug resistance mutations by digital PCR in a human influenza A 1

virus model system and clinical samples 2

3

Alexandra S. Whalea#, Claire Bushella, Paul R. Grantb, Simon Cowenc, Ion Gutierrez-4

Aguirred, Denise M. O’Sullivana, Jana Želd, Mojca Milavecd, Carole A. Foya, Eleni 5

Nastoulib, Jeremy A. Garsone, Jim F. Huggetta,e. 6

7

Molecular and Cell Biology Team, LGC Ltd, Teddington, UKa, Virology Laboratory, 8

Clinical Microbiology and Virology, University College London Hospital NHS 9

Foundation Trust, London, UKb, Statistics Team, LGC Ltd, Teddington, UKc, National 10

Institute of Biology, Ljubljana, Sloveniad, Department of Infection, Division of 11

Infection and Immunity, University College London, London, UKe. 12

13

Running Title: Evaluation of digital PCR for SNP detection. 14

15

#Address correspondence to Alexandra S. Whale, [email protected] 16

17

JCM Accepted Manuscript Posted Online 9 December 2015J. Clin. Microbiol. doi:10.1128/JCM.02611-15Copyright © 2015 Whale et al.This is an open-access article distributed under the terms of the Creative Commons Attribution-Noncommercial-ShareAlike3.0 Unported license, which permits unrestricted noncommercial use, distribution, and reproduction in any medium,provided the original author and source are credited.

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 2 of 31

ABSTRACT 18

Digital PCR (dPCR) is being increasingly used for the quantification of sequence 19

variations including single nucleotide polymorphisms (SNPs) due to its high accuracy 20

and precision in comparison with techniques such as quantitative PCR (qPCR) and 21

melt curve analysis. To develop and evaluate dPCR for SNP detection using DNA, RNA 22

and clinical samples, an influenza model of resistance to oseltamivir (Tamiflu) was 23

used. Firstly, this study was able to recognise and reduce off-target amplification in 24

dPCR quantification thereby enabling technical sensitivities down to 0.1% SNP 25

abundancy at a range of template concentrations; a 50-fold improvement on the 26

qPCR assay used routinely in the clinic. Secondly, a method was developed for 27

determining the false positive rate (background) signal. Finally, comparison of dPCR 28

with qPCR on clinical samples demonstrated the potential impact dPCR could have 29

on clinical research and patient management by earlier (trace) detection of rare drug 30

resistant sequence variants. Ultimately this could reduce the quantity of ineffective 31

drugs taken and facilitate early switching to alternative medication when available. 32

In the short term such methods could advance our understanding of microbial 33

dynamics and therapeutic responses in a range of infectious diseases such as HIV, 34

viral hepatitis and tuberculosis. Furthermore, the findings presented here are 35

directly relevant to other diagnostic areas such as the detection of rare SNPs in 36

malignancy, monitoring of graft rejection and foetal screening. 37

38

INTRODUCTION 39

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 3 of 31

In 2001 the World Health Organisation (WHO) identified the threat of 40

antimicrobial resistance as requiring immediate action with the need for worldwide 41

cooperation (1). The development of new molecular methods for early diagnosis and 42

monitoring of drug resistance is one such action (2, 3). In this study, digital PCR 43

(dPCR) was employed to detect a clinically relevant SNP in a human influenza A 44

(H1N1) model involving resistance to the neuraminidase inhibitor oseltamivir 45

(Tamiflu). Resistance is acquired by a de novo SNP mutation (p.H275Y) encoded in 46

segment 6 of the viral genome (4) that changes the structure of the neuraminidase 47

protein such that oseltamivir is unable to bind (5). This resistance conveys no loss of 48

fitness to the virus thus permitting transmission between humans and enabling 49

resistance to spread (6). Between 1999 and 2002, oseltamivir resistance was present 50

at a background rate of 0.33% in influenza A N1 isolates (4). However, since 2007 the 51

spread of the resistant A (H1N1) virus increased and in 2008 the resistance rates 52

were estimated to be up to 70% in some European countries (6). Subsequently, the 53

WHO recommended vigilant monitoring for the emergence of oseltamivir resistance 54

(7, 8). Since the disappearance of the 2009 pandemic A (H1N1), the vast majority of 55

circulating viruses are sensitive to oseltamivir (~ 99 %) (9). 56

dPCR has been reported to enable rare SNP detection with technical sensitivities 57

(also referred to as fractional abundance) down to 0.001% of the wild type (WT) in 58

genomic DNA extracts (10, 11). To achieve this, dPCR subdivides a PCR into a large 59

number of partitions so that a proportion of them contain no template molecules 60

(12, 13). While this partitioning may increase the accuracy and precision of dPCR 61

over the more widely used quantitative PCR (qPCR) (14-17), it may also improve the 62

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 4 of 31

sensitivity when measuring rare mutations within a high abundance WT background. 63

Rare SNP detection by dPCR is being used in an increasing range of clinical 64

applications including cancer stratification (18, 19), foetal screening (20, 21), 65

monitoring of organ transplant rejection (22) and detection of antimicrobial 66

resistance (23, 24). 67

However, in order for such methods to be effectively applied in research and 68

ultimately be translated into routine clinical analysis, validation of assay sensitivity is 69

essential along with additional considerations such as cost, speed and throughput. 70

To achieve a given sensitivity, a PCR assay must firstly have sufficient specificity to 71

allow confident discrimination between the SNP and WT molecules within a sample. 72

Secondly, a very small number of mutant molecules must be detectable in the 73

presence of a large excess of WT molecules. 74

In this study, we addressed the issue of technical sensitivity, for which we 75

evaluated the ability of dPCR to detect the H275Y SNP at abundancies down to 0.1% 76

of the WT, in a range of nucleic acid concentrations using an in vitro transcribed RNA 77

material. We then validated our dPCR method by quantifying the H275Y SNP in 78

clinical samples from patients who received oseltamivir treatment with comparison 79

to a qPCR method that is used routinely in the clinic that has a SNP detection 80

sensitivity of 5%. 81

82

MATERIALS AND METHODS 83

Unless otherwise stated experiments were performed in a single laboratory (LGC 84

Ltd, UK) and all kits and instruments were used following the manufacturer’s 85

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 5 of 31

instructions. RNA/DNA LoBind micro-centrifuge tubes (Eppendorf) were used 86

throughout the study. 87

Neuraminidase constructs and production of synthetic RNA transcripts. 88

Segment 6 encompassing the full neuraminidase sequence for the human influenza A 89

virus pandemic 2009 (H1N1) strain, containing either the sensitive wild type 90

sequence (WT: p.H275) or the oseltamivir drug-resistance mutation (SNP: p.H275Y; C 91

to T transition), was cloned into the pEX-K plasmid. To generate biologically relevant 92

negative strand RNA, in vitro transcription (IVT) of linearised plasmids was 93

performed. IVTs were diluted to ~1 x 109 copies/µL in carrier (15 ng/μl RNA extracted 94

from human lung cells (Ambion™)) and stored in aliquots to reduce freeze-thawing 95

effects. Full details are given in the supplemental material (section 1 and Figure S1). 96

Clinical samples. Clinical samples were used in accordance with local ethical 97

guidelines. Residual material originating from throat swabs (TS), nasal swabs (NS), 98

combined throat and nasal swabs (CTNS) and endotracheal aspirates (ETA), as part of 99

routine diagnostic laboratory practice in the Department of Clinical Microbiology and 100

Virology at UCLH, were inactivated by mixing with an equal volume of Buffer AL 101

(QIAGEN) prior to storage at 4 °C for several weeks before transferring to -20 °C for 102

long term storage. A positive SNP control (POS) was extracted from a cell culture 103

(provided by R. Gunson, West of Scotland Specialist Virology Centre, Glasgow) 104

propagating a pandemic H1N1 virus encoding the SNP. All viral RNA samples were 105

extracted from 200 µL clinical sample/Buffer AL mix using the EZ1 Virus Mini Kit v2.0 106

on a BioRobot EZ1 and eluted in 90 µL AVE buffer (all QIAGEN). These extracts were 107

provided blind to LGC. 108

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 6 of 31

Design of the H275Y genotyping assay. 733 pandemic influenza H1N1 109

neuraminidase gene sequences were downloaded from the NCBI influenza virus 110

sequence database (25). These were aligned and viewed in the multiple sequence 111

alignment program Clustal X (26). The location of the oseltamivir drug-resistance 112

mutation was identified. Primer express v2.0 software (Applied Biosystems™) was 113

used to design two TaqMan® MGB probes to the region containing the SNP; one 114

probe to target the wild type (WT) sequence and the other to target the resistant 115

(SNP) sequence. Universal primers were designed to flank the probe target sequence 116

to generate a 74 bp amplicon (Table S1). 117

Specificity to the pandemic influenza H1N1 serotype was determined by both in 118

silico alignments (http://www.ncbi.nlm.nih.gov/genomes/FLU/FLU.html) and RT-119

qPCR. Pre-pandemic seasonal influenza A strains (H1N1 and H3N2), the avian 120

influenza A strain H5N1, Influenza B and other common respiratory viruses 121

(respiratory syncytial virus, parainfluenza viruses, human metapneumovirus, and 122

adenovirus) were analysed with the H275Y assay; none of these viruses were 123

detected demonstrating that the assay was specific to pandemic influenza A H1N1 124

virus only. Details of the RT-qPCR method are given in the relevant section below 125

and supplemental material (section 5). 126

Reverse transcription. All reverse transcription (RT) experiments were 127

performed in accordance with the dMIQE guidelines (Table S2) (27). The Maxima 128

cDNA synthesis kit (Thermo Scientific) was used according to the manufacturer’s 129

instructions with the addition of 200 nM of H275Y reverse primer to each 20 µL 130

reaction. For all experiments, triplicate RT reactions were performed with a single 131

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 7 of 31

dPCR analysis per RT reaction. To monitor DNA contamination, reactions containing 132

1 x 105 IVT RNA copies per reaction with the reverse transcriptase omitted were 133

performed; in all cases no amplification occurred (Figure S2F). No template controls 134

(NTCs) were performed using carrier in place of template in every experiment; in all 135

cases no DNA was detected (Figure S2A). RT was perfumed with an initial incubation 136

at 25 °C for 10 minutes followed by RT at 50 °C for 15 minutes. The enzyme was 137

inactivated at 85 °C for 5 minutes. The cDNA was either stored at -80 °C or 138

immediately analysed by dPCR. 139

Digital PCR. dPCR experiments were implemented in accordance with the 140

dMIQE guidelines (Table S2) (27). dPCR experiments were performed using the 141

QX100/QX200™ Droplet Digital PCR System (Bio-Rad): 20 µl reactions containing 1 X 142

ddPCR™ Supermix for Probes with no dUTP (Bio-Rad), H275Y assay (Table S1; 143

LGC_dPCR) and 2 µl cDNA were pipetted into the sample well of a DG8 cartridge. 144

Droplet formation was performed as described previously (28). Thermocycling 145

conditions were 95 °C for 10 minutes, 40 cycles of 94 °C for 30 seconds and 60 °C for 146

1 minute, followed by 98 °C for 10 minutes and a 4 °C hold. The ramp rate for each 147

step was set to 2 °C/second. Droplets were read using the QX100/QX200™ Droplet 148

Reader and the data was analysed using QuantaSoft™ version 1.6.6.0320. Thresholds 149

were set to define the positive and negative droplets and the data was exported as a 150

.csv file. No template controls (NTCs) were performed using water in place of 151

template in every experiment; in all cases no amplification occurred (Figure S2A). 152

Optimisation of the H275Y assay for dPCR was performed at two laboratories; LGC, 153

UK and NIB, Slovenia (supplemental material, section 2). 154

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 8 of 31

One-step reverse transcription quantitative PCR. All RT-qPCR experiments were 155

performed in accordance with the MIQE guidelines (Table S3) (29). For H1N1 typing 156

of the clinical samples at UCLH, the Superscript® III Platinum one step qRT-PCR kit 157

(Invitrogen™) was used with an N1 typing assay published previously (FluA H1N1 158

assay) (30). For the initial genotyping of the H275Y SNP in clinical samples at UCLH, 159

the Superscript® III Platinum one step qRT-PCR kit (Invitrogen™) was used with the 160

UCLH_H275Y assay. Comparison of RT-qPCR with dPCR on the clinical samples at LGC 161

was performed at LGC with the AgPath-ID™ One-step RT-PCR Reagents kit (Applied 162

Biosystems™). Cycling and data capture was performed using the 7500 Fast or Prism 163

7900HT Real Time PCR systems (both Applied Biosystems™). Analysis was performed 164

using the SDS software v2.4 (Applied Biosystems™) to calculate the quantification 165

cycle (Cq) values and the data was exported as a .csv file. Full protocol conditions 166

and experimental setups for all RT-qPCR methods are given in the supplemental 167

material (section 5). 168

Data Analysis. Exported .csv files from dPCR and qPCR experiments were 169

imported into MS Excel 2010 or R statistical programming environment 170

(http://www.r-project.org/) for statistical analysis. For dPCR experiments, the 171

average number of copies per partition (λ) with the associated confidence intervals 172

were calculated in MS Excel 2010 as described previously (27, 31, 32). The mean 173

values for template concentration and % SNP (fractional abundance = λSNP/(λSNP + 174

λWT)) were calculated in MS Excel 2010 along with the associated standard deviation, 175

coefficient of variance, regression analysis (R2), T tests and ANOVA. The RT efficiency 176

was defined as the % cDNA yield that was calculated as (λSNP/λexp), where λexp is the 177

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 9 of 31

expected λ based on the concentration estimate using the Qubit® 2.0 fluorimeter 178

with the dsDNA HS Assay Kit (Invitrogen). Where necessary, data was transformed by 179

taking the natural logarithm as described previously (15). Figures were generated 180

either directly from the software programs or using Prism 6 (GraphPad). 181

For the calculations of the false positive rate (FPR), data was log transformed (λ 182

and % SNP) using the R statistical programming environment as described previously 183

(15). A standard analytical chemistry method based on the dispersion of samples 184

containing WT only molecules was employed to compute the detection limits (33). 185

The decision limits were calculated using two approaches: using the maximum 186

observed value, and an estimate based on Chebyshev's inequality (34). Full details 187

are given in the supplemental material (section 4). 188

189

RESULTS 190

Evaluation of the sensitivity of digital PCR in quantification of rare single 191

nucleotide polymorphisms. Initial experiments were performed to optimise and 192

evaluate the ability of the QX100/200™ dPCR system to detect the H275Y SNP using 193

the linearised plasmid DNA encoding either the WT or SNP sequence. The following 194

parameters were investigated to optimise the specificity of the H275Y assay: primer 195

and probe concentrations, MGB probe fluorophore dye swaps, uniplex and duplex 196

reactions and the annealing temperature (supplemental material, section 2 and 197

Figure S2). Duplex reactions containing 900 nM of each primer and 250 nM of each 198

probe (SNP probe conjugated with FAM and WT probe with VIC) with an annealing 199

temperature of 60 °C were selected for optimum specificity. 200

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 10 of 31

To evaluate the sensitivity of the method, a range of SNP abundancies (between 201

100% and 0.1%) at different IVT RNA concentrations corresponding to different viral 202

titres were generated: [high] 1.2 x 105 copies/RT (expected cDNA λ ~ 5.1, assuming 203

100% conversion rate of RNA to cDNA), [medium] 4 x 104 copies/RT (λ ~ 1.7) and 204

[low] 2 x 104 copies/RT (λ ~ 0.85) (supplemental material, section 3, Table S4 and 205

Table S5). The range of these viral loads represented those obtained from 206

respiratory samples of patients with pandemic H1N1 influenza A (35). The Maxima 207

cDNA synthesis kit was selected due to the high conversion rate of RNA to cDNA (RT 208

efficiency) obtained following reverse transcription (Figure S3). Each % SNP at each 209

concentration was measured with triplicate RTs (and one dPCR for each RT) in three 210

independent experiments (9 data points per % SNP at each concentration). For 211

measurement of the false positive rate (FPR) of the SNP assay (commonly referred to 212

as the signal to noise ratio or ‘blank’), WT only (0% SNP) controls were also prepared 213

at the three concentrations with quadruplicate measurements made in each of the 214

three experiments. 215

Strong linearity between the expected (as estimated by Qubit® 2.0 fluorimeter) 216

and observed WT and SNP molecules per partition (λ) was measured for all 217

experimental replicates (slope = 1.028, y-intercept = – 0.076, R2 = 0.9962) with no 218

significant difference between the replicates (p > 0.65) (Figure S3A). The RT 219

efficiencies were normally distributed with an average of 88% (CV = 9%) and 76% (CV 220

= 23%) for the WT and SNP assays respectively (Figure S3B). 221

The smallest SNP abundancy that could be measured as significantly different 222

from the WT only controls across all three RNA concentrations was 0.5% (p < 0.036). 223

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 11 of 31

For both the [high] and [medium] samples the SNP molecule could also be resolved 224

at 0.1% (p < 0.024 and p < 0.013 respectively) (Figure 1A). The CV for each SNP 225

abundancy increased with reduced sample concentration; measurements of the 226

[high] and [medium] sample had a CV of < 20% for the 100% to 0.5% SNP 227

abundancies while the 0.1% SNP had a CV or 17 and 21%, respectively. The [low] 228

sample had a CV > 29% for all SNP abundancies of 1% or less (Figure 1B). 229

Defining the detection capability of dPCR. The detection of SNP molecules in 230

the 0% SNP controls represents the FPR of the SNP assay and ranged from 0.06 to < 231

0.001% (average 0.012%) across the three different sample concentrations (Figure 232

1A). The observed variance associated with these controls was heteroscedastic with 233

a CV > 90% (Figure 1B). The limit of detection (LOD) of an assay is linked to the FPR 234

and its associated variance; the lower the FPR, the smaller the abundancy of the SNP 235

that can be measured as significantly different from the WT only control. Therefore, 236

it was hypothesised that the theoretical limit of detection for a given sample 237

concentration could be extrapolated from the experimental FPR data. 238

Using two methods: the maximum observed value and Chebyshev’s inequality 239

(34), the decision limit and the detection capability were calculated (Table 1 and 240

Figure S4). The detection capability, that defines the LOD in this context, was lower 241

than those tested experimentally for the [high] and [medium] samples (Table 1). 242

Chebyshev’s inequality, which generates more conservative estimates, defined the 243

limit of detection as 0.13% for the [low] sample (Table 1) and is consistent with the 244

inability to detect the 0.1% SNP abundancy in the [low] sample as significantly 245

different from the WT only controls. 246

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 12 of 31

Identification of oseltamivir resistance mutants in patients with influenza A 247

H1N1. Following the 2009 influenza A H1N1 pandemic, the H1N1 type was detected 248

in patients undergoing oseltamivir treatment at UCLH with the one-step RT-qPCR 249

method using the FluA H1N1 assay (30). Patient samples were screened for 250

oseltamivir resistance using a one-step RT-qPCR method (UCLH_RT-qPCR) to identify 251

the presence of WT (oseltamivir sensitive) and/or SNP (oseltamivir resistant) 252

molecules (Table 2 and Figure 2). 253

Absolute quantification of the clinical samples by RT-qPCR was performed using 254

standard curves generated from the IVT RNA (LGC_RT-qPCR). The limit of detection 255

was defined as 75 RNA copies/reaction (equivalent viral load to 2.65 log10 copies/mL 256

extract) (Figure S6B) with a specificity for the WT and SNP probes of 90% and 87%, 257

respectively, regardless of sample concentration (Figure S6C). Due to the number of 258

samples that were quantified by dPCR only, comparison of LGC_RT-qPCR and dPCR 259

measurements identified a poor linear correlation. Analysis of the six samples that 260

were quantified by both dPCR and LGC_RT-qPCR, identified a good linear correlation; 261

R2 = 0.9223. The gradient of the slope (0.5429) was significantly different from 1 (p = 262

0.04) indicating a measurement bias (Figure 2A). The CV was on average lower for 263

the dPCR measurements compared with qPCR (13% and 23%, respectively). Spike-in 264

experiments confirmed the absence of inhibitors carried over from the sampling or 265

extraction process disrupting the qPCR or dPCR reactions (Figure S6D). 266

Fairly good concordance for detection of WT and SNP molecules was observed 267

between the UCLH_RT-qPCR, LGC_RT-qPCR and dPCR results (Table 2; light grey 268

shaded). Five samples had very low concentrations as defined by dPCR (samples 3.2, 269

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 13 of 31

3.3, 3.4, 4.2 and 5.2) and were below the limit of sensitivity for qPCR. Furthermore, 270

only one of these five clinical samples was identified as positive for H1N1 by 271

UCLH_RT-qPCR (sample 3.2) with the detection of both WT and SNP sequences 272

(Table 2; dark grey shaded). WT sequences were detected in one sample (sample 273

4.1) by LGC_RT-qPCR at LGC while no sequences were detected by UCLH_RT-qPCR or 274

by dPCR. 275

Both WT and SNP molecules were identified in two clinical samples by LGC_RT-276

qPCR and dPCR (samples P1.3 and P1.4) (Figure 2B). SNP molecules were measured 277

in a further two clinical samples by LGC_RT-qPCR but not by dPCR (Figure 2B); the 278

proportion of SNP molecules as measured by LGC_RT-qPCR were 13.53% and 11.86% 279

for samples 1.1 and 5.1, respectively (Figure 2B; asterisk). As the limit of specificity of 280

the assay was defined as 87%, these samples were identified as potential false 281

positives for the presence of SNP molecule as the uncertainty in their quantification 282

was above the limit of the specificity for the SNP assay (Table 2). Absence of 283

amplification of either molecule in the experimental negative controls (NEG) 284

indicated that the SNP signal was unlikely to be caused by contaminating molecules 285

(Figure 2B). The limitations of the specificity of the LGC_RT-qPCR assay were further 286

highlighted in the analysis of the SNP only sample (POS), where the SNP abundancy 287

was measured as 95.82% (Figure 2B). 288

Five samples were taken from patient 1 over the course of a month during 289

treatment with oseltamivir (samples 1.1 to 1.5). Longitudinal analysis of the 290

concentration of the WT and SNP molecules revealed that the SNP molecule became 291

predominant by day 28. From days 28 to 29, the amount of SNP molecules quantified 292

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 14 of 31

by both qPCR and dPCR increased nearly two-fold (Figure 2C). While the 293

concentration of the WT molecule stayed constant over the duration as measured by 294

dPCR, it significantly increased over 1.5-fold (p = 0.003) when measured by qPCR 295

(Figure 2D). 296

297

DISCUSSION 298

Using a human influenza A model system, we have developed a dPCR method 299

for rare drug resistance mutations by dPCR. We have investigated a number of 300

challenges associated with SNP detection, namely the specificity, sensitivity and the 301

false positive (FPR) of an assay. We then validated our method using clinical samples. 302

Our study has confirmed the ability to recognise and reduce off-target 303

amplification (mismatch binding of WT probe with the SNP sequence and vice versa), 304

thereby improving the specificity of the assay and so tackles one of the great 305

challenges of SNP detection. We have demonstrated the ability of dPCR to detect 306

rare SNPs down to 0.1% abundancy (Figure 1), although it should be acknowledged 307

that this was using material generated from IVTs in a background of human lung RNA 308

and so does not consider the effects more complex respiratory matrices may have on 309

the reaction. 310

While there is currently no clinical data demonstrating the necessity of detecting 311

SNPs down to 0.1% abundancy, by developing a method with increased levels of 312

sensitivity we provide the means by which future studies could determine an 313

appropriate cut-off for clinical use and by which the development of resistance can 314

be more accurately monitored during the treatment. Indeed, there is a need to 315

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 15 of 31

consistently measure the 1% population abundance that is the current background 316

rate of the mutation (4, 6, 9) in order to follow the recommended guidelines by the 317

WHO (7, 8). The same assay (primers and probes) in qPCR has a cut-off at 318

approximately 5% abundancy due to the off-target amplification of the WT 319

molecules with the SNP probe (36) and therefore our study represents a 50-fold 320

increase in sensitivity by dPCR. This is consistent with the reported limits of other 321

qPCR assays for SNP detection (37, 38) and with the identification of false positive 322

results for two clinical samples (P1.1 and P5.1) that were identified as containing SNP 323

sequences by qPCR only. 324

The increased sensitivity of dPCR over qPCR was shown by the proportion of 325

clinical samples that were detectable by dPCR only (Figures 2A and 2B). A recent 326

study using a different assay that targets the same H275Y SNP observed comparable 327

differences in the sensitivity between qPCR and dPCR (24). While improved 328

quantification and precision of dPCR over qPCR has also been observed in other RNA 329

viral models (39, 40), studies using infectious agents with DNA genomes have shown 330

further improvement in the quantification and precision by both dPCR and qPCR (41, 331

42). This suggests that the RT step could have a greater effect on the sensitivity of 332

qPCR, although dPCR is not immune from variability in the RT efficiency (Figure S3B). 333

However, it is acknowledged that without a third method to arbitrate the 334

discrepancies between qPCR and dPCR, we cannot be certain that the dPCR result is 335

the correct result. 336

In addition to the off-target amplification, the greater specificity and sensitivity 337

afforded by dPCR, with respect to qPCR, is related to both the FPR and precision of 338

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 16 of 31

the measurement. Here we developed a method to define the FPR that is associated 339

with the sample concentration. When sample concentration is not limiting and a 340

DNA template is used, it is possible to detect abundancies down to 0.001% using 341

dPCR as demonstrated in two genomic DNA cancer models (10, 11). While this is 342

impressive, there are a number of other factors that prevent sensitivities of this 343

magnitude from being routinely achieved in clinical samples. 344

The sample concentration limits are self-explanatory; to detect 0.001% 345

sequence abundance, substantially more than 100,000 sequences must be measured 346

(equates to a viral titre > 8 log10 copies/mL). However, a sample is influenced not 347

only by the sample concentration, but also by the quality of the sample, for example 348

sputum-containing samples can be difficulty to homogenise prior to extraction (43, 349

44). The type of respiratory samples used in this study involved RNA extracted from 350

TS, NS, CTNS and ETA, which could also impact on the results and may be a 351

contributing factor to the difference in performance between qPCR and dPCR. It has 352

previously been shown that inhibitors carried over from the extraction process in a 353

CMV model affect qPCR and dPCR to different extents (45). Furthermore, the RNA 354

yield obtained from extraction of RNA from influenza viruses from sputum can vary 355

from 20-100% depending on the extraction method used (43). These are important 356

areas for future investigation into the ability of dPCR to measure rare sequences in 357

RNA viruses. 358

Additional considerations include the sample concentration, as this is reduced, 359

the associated precision of dPCR is also reduced, although not to the same extent as 360

qPCR, and so smaller differences between samples could be harder to measure with 361

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 17 of 31

confidence (46). While these factors may have implications for clinical samples such 362

as those used in this study, it is also of importance for the longitudinal monitoring of 363

patients whose viral loads are expected to reduce in response to treatment over 364

time. 365

This is clearly demonstrated by the analysis of longitudinal samples from a single 366

patient (Figures 2C and 2D); dPCR measured the WT molecules as remaining at a 367

constant level while there was an increase in the abundance of the SNP molecule 368

during treatment. In contrast, LGC_RT-qPCR measured an increase in both the WT 369

and SNP molecules and so implied that despite treatment, the overall viral load of 370

the patient was increasing. Recently, a similar longitudinal analysis of a patient 371

infected with oseltamivir resistant H1N1 Influenza A demonstrated that dPCR is less 372

prone to operator effects and has reduced inter-assay and intra-assay variability in 373

SNP quantification than qPCR (24). Together these findings show the potential 374

advantages of dPCR for monitoring and surveillance of antimicrobial drug resistance. 375

In this study, we demonstrated that the FPR determined the technical sensitivity 376

of the [low] sample rather than the precision of dPCR and therefore demonstrates 377

that the FPR is linked with the sample concentration. A separate study investigating 378

SNP mutations in a cfDNA model identified the FPR as being independent of sample 379

concentration (18), however, this study was investigating the genotype of cell free 380

DNA extracted from plasma and so the WT sequences were very low in 381

concentration indicating that the FPR can be difficult to define at very low 382

concentration samples. The FPR of an assay can be split into two sources; 383

experimental and technical. Experimental FPR can be attributed to laboratory 384

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 18 of 31

contamination that was absent in this study (Figures S2, Table S5) and is reduced by 385

appropriate optimisation and good laboratory practice. 386

This leaves the technical sources contributing the FPR that are harder to define 387

but could include off-target amplification and enzymatic error, such as those that 388

arise from reverse transcriptases and/or polymerases (47). By defining the limit of 389

detection mathematically and relating it the concentration of the sample as 390

described, experimental constraints that limit the number of negative controls (for 391

example, different dPCR platforms have different maximum sample numbers per 392

experiment) could be overcome (46). This is in sharp contrast to other methods, such 393

as qPCR, that are limited by the quality of the calibration curve or next generation 394

sequencing that can be limited by technical error rates (48). 395

Finally, the robustness of dPCR allowed a qPCR assay that is used routinely in the 396

clinic to be transferred on to a dPCR platform with minimal optimisation. 397

Furthermore, the reproducibility of dPCR compared with qPCR (24) demonstrates 398

the potential of dPCR for clinical use. Various other methods such as CAST 399

(competitive allele-specific TaqMan) PCR (38, 49, 50), ARMS (amplification-refractory 400

mutation system) (51) and the use of LNA (locked nucleic acid) probes (52, 53) have 401

been developed to improve the 5-10 % SNP abundancy limit of qPCR. While these 402

methods are capable of detecting down to 0.1-1 % SNP, they require extensive 403

optimisation, including in some cases a total re-design of the assay. 404

Despite all these advantages, in the short term the non-reliance of dPCR on 405

calibration materials for quantification could be exploited as an alternative value 406

assignment method that is independent of mass or fluorescent measurements (54). 407

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 19 of 31

Previous studies have shown that UV spectrometry methods can over quantify 408

nucleic acid abundance compared with dPCR (14, 16). Therefore the apparently 409

higher concentrations of the clinical samples as measured by qPCR compared with 410

dPCR (Figure 2A) could be attributed to the inherent limitations of the UV 411

quantification of the IVT RNA used for the calibration curve rather than bias within 412

the qPCR method itself. 413

The ability of dPCR to detect rare drug resistance SNP mutations could enable 414

earlier detection, facilitating earlier switching to alternative therapeutic agents and 415

resulting in an overall reduction in the amount of ineffective drugs used. This should 416

result in a reduction in both drug resistance and cost. Other infectious diseases such 417

as HIV, viral hepatitis and bacterial infections including tuberculosis and gonorrhoea 418

could also benefit from this approach to antimicrobial drug resistance monitoring. 419

Finally, the findings of this study should be applicable in a wide range of other 420

diagnostic areas including the monitoring of cell free nucleic acid in malignancy and 421

foetal screening. 422

423

FUNDING INFORMATION 424

The work described in this manuscript was partially funded by the UK National 425

Measurement System (NMS) and the European Metrology Research Programme 426

(EMRP) joint research project [HLT08] “Infect-Met” (http://infectmet.lgcgroup.com) 427

which is jointly funded by the EMRP participating countries within EURAMET and the 428

European Union. 429

430

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 20 of 31

ACKNOWLEDGMENT 431

The authors would like to acknowledge George Karlin-Neumann and Svilen 432

Tzonev from Bio-Rad for extensive assistance in the development of this study and 433

Dr Rory Gunson of the West of Scotland Specialist Virology Centre in Glasgow who 434

kindly donated the cell culture propagating a pandemic H1N1 virus encoding the 435

H275Y mutation. 436

437

REFERENCES 438

1. WHO. 2001. WHO global strategy for containment of antimicrobial resistance 439

http://www.who.int/entity/csr/resources/publications/drugresist/en/EGlobal_440

Strat.pdf. Accessed 18/07/2015. 441

2. van der Vries E, Anber J, van der Linden A, Wu Y, Maaskant J, Stadhouders R, 442

van Beek R, Rimmelzwaan G, Osterhaus A, Boucher C, Schutten M. 2013. 443

Molecular assays for quantitative and qualitative detection of influenza virus 444

and oseltamivir resistance mutations. J Mol Diagn 15:347-354. 445

3. van der Vries E, Schutten M, Fraaij P, Boucher C, Osterhaus A. 2013. Influenza 446

virus resistance to antiviral therapy. Adv Pharmacol 67:217-246. 447

4. Monto AS, McKimm-Breschkin JL, Macken C, Hampson AW, Hay A, Klimov A, 448

Tashiro M, Webster RG, Aymard M, Hayden FG, Zambon M. 2006. Detection 449

of influenza viruses resistant to neuraminidase inhibitors in global surveillance 450

during the first 3 years of their use. Antimicrob Agents Chemother 50:2395-451

2402. 452

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 21 of 31

5. Moscona A. 2005. Oseltamivir resistance--disabling our influenza defenses. N 453

Engl J Med 353:2633-2636. 454

6. Lackenby A, Hungnes O, Dudman SG, Meijer A, Paget WJ, Hay AJ, Zambon 455

MC. 2008. Emergence of resistance to oseltamivir among influenza A(H1N1) 456

viruses in Europe. Euro Surveill 13. 457

7. WHO. 2009. Oseltamivir resistance in immunocompromised hospital patients; 458

pandemic (H1N1) 2009 briefing note 18. 459

http://www.who.int/csr/disease/swineflu/notes/briefing_20091202/en/index.460

html. Accessed 26/06/2015. 461

8. WHO. 2009. Viruses resistant to oseltamivir (Tamiflu) identified; pandemic 462

(H1N1) 2009 briefing note 1. 463

http://www.who.int/csr/disease/swineflu/notes/h1n1_antiviral_resistance_20464

090708/en/index.html. Accessed 26/06/2015. 465

9. Meijer A JM, van Beek P, Swaan CM, Osterhaus AD, Daniels RS, Hurt AC, 466

Koopmans MP. 2012. Oseltamivir-resistant influenza A(H1N1)pdm09 virus in 467

Dutch travellers returning from Spain. 468

http://www.eurosurveillance.org/ViewArticle.aspx?ArticleId=20266 Accessed 469

10. Pekin D, Skhiri Y, Baret JC, Le Corre D, Mazutis L, Salem CB, Millot F, El Harrak 470

A, Hutchison JB, Larson JW, Link DR, Laurent-Puig P, Griffiths AD, Taly V. 471

2011. Quantitative and sensitive detection of rare mutations using droplet-472

based microfluidics. Lab Chip 11:2156-2166. 473

11. Hindson BJ, Ness KD, Masquelier DA, Belgrader P, Heredia NJ, Makarewicz AJ, 474

Bright IJ, Lucero MY, Hiddessen AL, Legler TC, Kitano TK, Hodel MR, Petersen 475

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 22 of 31

JF, Wyatt PW, Steenblock ER, Shah PH, Bousse LJ, Troup CB, Mellen JC, 476

Wittmann DK, Erndt NG, Cauley TH, Koehler RT, So AP, Dube S, Rose KA, 477

Montesclaros L, Wang S, Stumbo DP, Hodges SP, Romine S, Milanovich FP, 478

White HE, Regan JF, Karlin-Neumann GA, Hindson CM, Saxonov S, Colston 479

BW. 2011. High-throughput droplet digital PCR system for absolute 480

quantitation of DNA copy number. Anal Chem 83:8604-8610. 481

12. Sykes PJ, Neoh SH, Brisco MJ, Hughes E, Condon J, Morley AA. 1992. 482

Quantitation of targets for PCR by use of limiting dilution. Biotechniques 483

13:444-449. 484

13. Vogelstein B, Kinzler KW. 1999. Digital PCR. Proc Natl Acad Sci U S A 96:9236-485

9241. 486

14. Sanders R, Huggett JF, Bushell CA, Cowen S, Scott DJ, Foy CA. 2011. Evaluation 487

of digital PCR for absolute DNA quantification. Anal Chem 83:6474-6484. 488

15. Whale AS, Huggett JF, Cowen S, Speirs V, Shaw J, Ellison S, Foy CA, Scott DJ. 489

2012. Comparison of microfluidic digital PCR and conventional quantitative PCR 490

for measuring copy number variation. Nucleic Acids Res 40:e82. 491

16. Sanders R, Mason DJ, Foy CA, Huggett JF. 2013. Evaluation of digital PCR for 492

absolute RNA quantification. PLoS One 8:e75296. 493

17. Hindson CM, Chevillet JR, Briggs HA, Gallichotte EN, Ruf IK, Hindson BJ, 494

Vessella RL, Tewari M. 2013. Absolute quantification by droplet digital PCR 495

versus analog real-time PCR. Nat Methods 10:1003-1005. 496

18. Taly V, Pekin D, Benhaim L, Kotsopoulos SK, Le Corre D, Li X, Atochin I, Link 497

DR, Griffiths AD, Pallier K, Blons H, Bouche O, Landi B, Hutchison JB, Laurent-498

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 23 of 31

Puig P. 2013. Multiplex Picodroplet Digital PCR to Detect KRAS Mutations in 499

Circulating DNA from the Plasma of Colorectal Cancer Patients. Clin Chem 500

59:1722-1731. 501

19. Laurent-Puig P, Pekin D, Normand C, Kotsopoulos SK, Nizard P, Perez-Toralla 502

K, Rowell R, Olson J, Srinivasan P, Le Corre D, Hor T, El Harrak Z, Li X, Link DR, 503

Bouche O, Emile JF, Landi B, Boige V, Hutchison JB, Taly V. 2015. Clinical 504

Relevance of KRAS-Mutated Subclones Detected with Picodroplet Digital PCR in 505

Advanced Colorectal Cancer Treated with Anti-EGFR Therapy. Clin Cancer Res 506

21:1087-1097. 507

20. Barrett AN, McDonnell TCR, Chan KCA, Chitty LS. 2012. Digital PCR Analysis of 508

Maternal Plasma for Noninvasive Detection of Sickle Cell Anemia. Clin Chem 509

58:1026-1032. 510

21. Lench N, Barrett A, Fielding S, McKay F, Hill M, Jenkins L, White H, Chitty LS. 511

2013. The clinical implementation of non-invasive prenatal diagnosis for single-512

gene disorders: challenges and progress made. Prenatal Diagnosis 33:555-562. 513

22. Beck J, Bierau S, Balzer S, Andag R, Kanzow P, Schmitz J, Gaedcke J, Moerer O, 514

Slotta JE, Walson P, Kollmar O, Oellerich M, Schutz E. 2013. Digital Droplet 515

PCR for Rapid Quantification of Donor DNA in the Circulation of Transplant 516

Recipients as a Potential Universal Biomarker of Graft Injury. Clin Chem 517

59:1732-1741. 518

23. Pholwat S, Stroup S, Foongladda S, Houpt E. 2013. Digital PCR to Detect and 519

Quantify Heteroresistance in Drug Resistant Mycobacterium tuberculosis. PLoS 520

One 8:e57238. 521

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 24 of 31

24. Taylor SC, Carbonneau J, Shelton DN, Boivin G. 2015. Optimization of Droplet 522

Digital PCR from RNA and DNA extracts with direct comparison to RT-qPCR: 523

Clinical implications for quantification of Oseltamivir-resistant subpopulations. 524

J Virol Methods 224:58-66. 525

25. Bao Y, Bolotov P, Dernovoy D, Kiryutin B, Zaslavsky L, Tatusova T, Ostell J, 526

Lipman D. 2008. The influenza virus resource at the National Center for 527

Biotechnology Information. J Virol 82:596-601. 528

26. Thompson JD, Gibson TJ, Plewniak F, Jeanmougin F, Higgins DG. 1997. The 529

CLUSTAL_X windows interface: flexible strategies for multiple sequence 530

alignment aided by quality analysis tools. Nucleic Acids Res 25:4876-4882. 531

27. Huggett JF, Foy CA, Benes V, Emslie K, Garson JA, Haynes R, Hellemans J, 532

Kubista M, Mueller RD, Nolan T, Pfaffl MW, Shipley GL, Vandesompele J, 533

Wittwer CT, Bustin SA. 2013. The Digital MIQE Guidelines: Minimum 534

Information for Publication of Quantitative Digital PCR Experiments. Clin Chem 535

59:892-902. 536

28. Devonshire AS, Honeyborne I, Gutteridge A, Whale AS, Nixon G, Wilson P, 537

Jones G, McHugh TD, Foy CA, Huggett JF. 2015. Highly reproducible absolute 538

quantification of Mycobacterium tuberculosis complex by digital PCR. 539

Analytical Chemistry 87:3706-3713. 540

29. Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, Mueller R, 541

Nolan T, Pfaffl MW, Shipley GL, Vandesompele J, Wittwer CT. 2009. The MIQE 542

guidelines: minimum information for publication of quantitative real-time PCR 543

experiments. Clin Chem 55:611-622. 544

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 25 of 31

30. Carr MJ, Gunson R, Maclean A, Coughlan S, Fitzgerald M, Scully M, O'Herlihy 545

B, Ryan J, O'Flanagan D, Connell J, Carman WF, Hall WW. 2009. Development 546

of a real-time RT-PCR for the detection of swine-lineage influenza A (H1N1) 547

virus infections. J Clin Virol 45:196-199. 548

31. Dube S, Qin J, Ramakrishnan R. 2008. Mathematical analysis of copy number 549

variation in a DNA sample using digital PCR on a nanofluidic device. PLoS One 550

3:e2876. 551

32. Whale AS, Cowen S, Foy CA, Huggett JF. 2013. Methods for applying accurate 552

digital PCR analysis on low copy DNA samples. PLoS One 8:e58177. 553

33. Ellison SLR, Barwick VJ, Farrant TJD. 2009. Practical Statistics for the Analytical 554

Scientist – A Bench Guide, p 153, Practical Statistics for the Analytical Scientist 555

– A Bench Guide, 2nd ed. RSC Publishing. 556

34. Abramowitz M, Stegun IA. 1972. Handbook of Mathematical Functions with 557

Formulas, Graphs, and Mathematical Tables., 9th ed. New York: Dover. 558

35. Ngaosuwankul N, Noisumdaeng P, Komolsiri P, Pooruk P, Chokephaibulkit K, 559

Chotpitayasunondh T, Sangsajja C, Chuchottaworn C, Farrar J, Puthavathana 560

P. 2010. Influenza A viral loads in respiratory samples collected from patients 561

infected with pandemic H1N1, seasonal H1N1 and H3N2 viruses. Virol J 7:75. 562

36. Grant PR, Kidd IM. 2011. Rapid real-time PCR for detection of oseltamivir 563

resistant pandemic influenza, abstr Winter Meeting of the European Society for 564

Clinical Virology (ESCV), London, 565

37. Huggett JF, Cowen S, Foy CA. 2015. Considerations for Digital PCR as an 566

Accurate Molecular Diagnostic Tool. Clin Chem 61:79-88. 567

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 26 of 31

38. Roma C, Esposito C, Rachiglio AM, Pasquale R, Iannaccone A, Chicchinelli N, 568

Franco R, Mancini R, Pisconti S, De Luca A, Botti G, Morabito A, Normanno N. 569

2013. Detection of EGFR mutations by TaqMan mutation detection assays 570

powered by competitive allele-specific TaqMan PCR technology. Biomed Res 571

Int 2013:385087. 572

39. Kiselinova M, Pasternak AO, De Spiegelaere W, Vogelaers D, Berkhout B, 573

Vandekerckhove L. 2014. Comparison of Droplet Digital PCR and Seminested 574

Real-Time PCR for Quantification of Cell-Associated HIV-1 RNA. PLoS One 575

9:e85999. 576

40. White RA, 3rd, Quake SR, Curr K. 2012. Digital PCR provides absolute 577

quantitation of viral load for an occult RNA virus. J Virol Methods 179:45-50. 578

41. Hayden RT, Gu Z, Ingersoll J, Abdul-Ali D, Shi L, Pounds S, Caliendo AM. 2013. 579

Comparison of droplet digital PCR to real-time PCR for quantitative detection 580

of cytomegalovirus. J Clin Microbiol 51:540-546. 581

42. Roberts CH, Last A, Molina-Gonzalez S, Cassama E, Butcher R, Nabicassa M, 582

McCarthy E, Burr SE, Mabey DC, Bailey RL, Holland MJ. 2013. Development 583

and Evaluation of a Next-Generation Digital PCR Diagnostic Assay for Ocular 584

Chlamydia trachomatis Infections. J Clin Microbiol 51:2195-2203. 585

43. Vanspauwen MJ, Wolffs PF, Franssen FM, Bruggeman CA, Wouters EF, 586

Linssen CF. 2014. Comparison of three different techniques for the isolation of 587

viral RNA in sputum. J Clin Virol 61:265-269. 588

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 27 of 31

44. Falsey AR, Formica MA, Walsh EE. 2012. Yield of sputum for viral detection by 589

reverse transcriptase PCR in adults hospitalized with respiratory illness. J Clin 590

Microbiol 50:21-24. 591

45. Nixon G, Garson JA, Grant P, Nastouli E, Foy CA, Huggett JF. 2014. A 592

comparative study of sensitivity, linearity and resistance to inhibition of digital 593

and non- digital PCR and LAMP assays for quantification of human 594

cytomegalovirus. Anal Chem 86:4387-4394. 595

46. Huggett JF, Garson J, Whale AS. 2015. Digital PCR and its potential application 596

to microbiology., Molecular Microbiology: Diagnostic Principles and Practice, 597

3rd ed. 598

47. Rittié L, Perbal B. 2008. Enzymes used in molecular biology: a useful guide. 599

Journal of Cell Communication and Signaling 2:25-45. 600

48. Robasky K, Lewis NE, Church GM. 2014. The role of replicates for error 601

mitigation in next-generation sequencing. Nat Rev Genet 15:56-62. 602

49. Didelot A, Le Corre D, Luscan A, Cazes A, Pallier K, Emile JF, Laurent-Puig P, 603

Blons H. 2012. Competitive allele specific TaqMan PCR for KRAS, BRAF and 604

EGFR mutation detection in clinical formalin fixed paraffin embedded samples. 605

Exp Mol Pathol 92:275-280. 606

50. Worwa G, Andrade CC, Thiemann TC, Park B, Maharaj PD, Anishchenko M, 607

Brault AC, Reisen WK. 2014. Allele-specific qRT-PCR demonstrates superior 608

detection of single nucleotide polymorphisms as genetic markers for West Nile 609

virus compared to Luminex(R) and quantitative sequencing. J Virol Methods 610

195:76-85. 611

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 28 of 31

51. Little S. 2001. Amplification-refractory mutation system (ARMS) analysis of 612

point mutations. Curr Protoc Hum Genet Chapter 9:Unit 9.8. 613

52. Morandi L, de Biase D, Visani M, Cesari V, De Maglio G, Pizzolitto S, Pession A, 614

Tallini G. 2012. Allele specific locked nucleic acid quantitative PCR 615

(ASLNAqPCR): an accurate and cost-effective assay to diagnose and quantify 616

KRAS and BRAF mutation. PLoS One 7:e36084. 617

53. Ghedin E, Holmes EC, DePasse JV, Pinilla LT, Fitch A, Hamelin ME, Papenburg 618

J, Boivin G. 2012. Presence of oseltamivir-resistant pandemic A/H1N1 minor 619

variants before drug therapy with subsequent selection and transmission. J 620

Infect Dis 206:1504-1511. 621

54. Pavsic J, Devonshire AS, Parkes H, Schimmel H, Foy CA, Karczmarczyk M, 622

Gutierrez-Aguirre I, Honeyborne I, Huggett JF, McHugh TD, Milavec M, 623

Zeichhardt H, Zel J. 2015. Standardization of Nucleic Acid Tests for Clinical 624

Measurements of Bacteria and Viruses. J Clin Microbiol 53:2008-2014. 625

626

FIGURE LEGENDS 627

Figure 1: Measurement of mutant SNPs by digital PCR is dependent on 628

template concentration and background signal. (A) Graph showing expected % SNP 629

of each sample (x-axis) against the observed % SNP abundance of the SNP molecule 630

(y-axis with log scale) for three concentrations. Each data point shows the mean and 631

95% confidence interval (CI) for the three experiments (performed on different days; 632

3 measurements), with the exception of the 0% SNP (WT only) control that was 633

performed in quadruplicate in all three experiments (12 measurements). The solid 634

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 29 of 31

and dashed horizontal lines represent the mean and 95% CI of the [low] 635

concentration 0% SNP control. There was no significant (ns; p > 0.05) difference 636

between the 0.1% and 0% SNP for the [low] sample. All other SNP abundancies and 637

sample concentrations were significantly different (p < 0.05) from their respective 638

0% SNP controls. (B) Line graph showing coefficient of variance (precision) of the 639

measurements for the three sample concentrations. Horizontal dashed line indicates 640

a CV of 20%. 641

Figure 2: Identification of oseltamivir resistant mutants in patients with 642

Influenza A H1N1. (A) Linear regression for the 18 clinical samples. Data is plotted as 643

the log10 total RNA copies (WT and SNP) per mL of clinical sample extracts between 644

two-step dPCR and one-step RT-qPCR. The regression line (solid line) with its 645

associated 95% CI (dashed lines) is shown. (B) % SNP abundance measured in each 646

clinical sample (x-axis) by two-step LGC_dPCR (red) one-step LGC_RT-qPCR (blue). A 647

single mean value (horizontal dash in red or blue) at 0% indicates a sample in which 648

only WT sequences were measured. Error bars represent the 95% confidence 649

interval in the measurement of the % SNP. The solid and dashed horizontal lines 650

represent the limit of detection of the SNP assay by LGC_RT-qPCR. Two samples 651

were identified as false positives (*). (C and D) Longitudinal quantification of SNP (C) 652

and WT (D) sequences in patient 1 over one month by dPCR (red) and qPCR (blue). 653

Key: POS; SNP only control, NEG; extraction buffer only. 654

655

TABLES 656

Table 1. Calculation of the detection capability. 657

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 30 of 31

Method Sample

name

dPCR

input

copies/rxn

Decision

Limit Detection Capability

Ln λ Ln λ Ln % SNP % SNP

Maximum

observed

value

[high] 120,000 -6.349 -5.965 -7.54 0.05%

[medium] 40,000 -8.078 -7.694 -8.11 0.03%

[low] 20,000 -7.645 -7.261 -6.96 0.10%

Chebyshev's

inequality α =

0.05

[high] 120,000 -5.784 -5.400 -6.96 0.09%

[medium] 40,000 -7.505 -7.121 -7.53 0.05%

[low] 20,000 -7.300 -6.916 -6.61 0.13%

For full details for calculation of these values, see text and supplementary materials 658

and methods. 659

660

Table 2: Analysis of clinical samples by qPCR and dPCR. 661

Patient

Code Day

Sample

Type

Flu A

H1N1

Result

UCLH_R

T-qPCR

Result

LGC_RT-qPCR LGC_dPCR

Total

viral

load

Result

Total

viral

load

Result

P1.1 0 TS POS Sen 4.35 FP 3.92 Sen

P1.2 26 TS n/d n/d n/d n/d

P1.3 28 CTNS POS Res 5.34 Res 4.53 Res

P1.4 29 CTNS POS Res 5.54 Res 4.96 Res

P1.5 32 TS LLP Res n/d n/d

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

Page 31 of 31

P2 0 TS POS Res n/d n/d

P3.1 0 CTNS POS Sen 3.49 Sen 3.51 Sen

P3.2 5 CTNS POS Res n/d 3.78 Sen

P3.3 11 NS n/d n/d n/d 3.36 Sen

P3.4 15 CTNS n/d n/d n/d 3.51 Sen

P4.1 0 ETA POS n/d 4.59 Sen n/d

P4.2 0 CTNS n/d n/d n/d 3.04 Sen

P4.3 5 CTNS n/d n/d n/d n/d

P5.1 0 ETA POS Sen 5.48 FP 4.56 Sen

P5.2 12 CTNS n/d n/d n/d 3.69 Sen

P6 0 CTNS LLP n/d n/d n/d

P7 0 CTNS LLP n/d n/d n/d

P8 0 TS POS Sen n/d 3.03 Sen

POS

POS Res 3.23 Res 3.61 Res

NEG n/d n/d n/d n/d

Total viral load is given as Log10 copies/mL extract. Key. Sample type: TS = Throat 662

Swab, NS = Nasal swab, CTNS = combined Throat and Nasal swab, ETA = 663

Endotracheal aspirate. POS = positive by PCR for Flu A H1N1, n/d = not 664

detected/below limit of detection; LLP = Low Level Positive, Res = resistant 665

sequences (SNP) detected in sample, Sen = only sensitive sequences (WT) detected; 666

FP = false positive, see text for details; Grey shaded = concordance between LGC and 667

UCLH results. 668

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from

on May 26, 2019 by guest

http://jcm.asm

.org/D

ownloaded from