168
1-1 University of Portsmouth MD Thesis Dr. R. J. Mead Queen Alexandra Hospital, Portsmouth. December 2012 Circulating DNA markers of Gastro-intestinal Cancers The thesis is submitted in partial fulfilment of the requirements for the award of the degree of Doctor of Medicine of the University of Portsmouth

December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-1

University of Portsmouth

MD Thesis

Dr. R. J. Mead

Queen Alexandra Hospital, Portsmouth.

December 2012

Circulating DNA markers

of Gastro-intestinal

Cancers

The thesis is submitted in partial fulfilment of the

requirements for the award of the degree of Doctor

of Medicine of the University of Portsmouth

Page 2: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-2

Abstract.

Introduction: Early diagnosis represents the best opportunity for cure of gastro-intestinal

cancers; however current gastro-intestinal cancer screening programmes have low test

sensitivity and low patient acceptability. It is hoped that a better understanding of

carcinogenesis and the development of new biomarkers will provide answers to these

clinical problems.

Hypothesis and aims: This thesis examines the current understanding of gastro-intestinal

carcinogenesis focusing on colorectal and oesophageal cancers. It reviews the circulating

gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and

successes of current approaches.

The study tests the hypothesis that control patients and patients with colorectal and

oesophageal neoplasia have differing plasma levels and fragmentation patterns of cell free

nuclear and mitochondrial DNA, and that endoscopic removal of these lesions returns the

levels and patterns to normal.

We aim to show how new techniques of DNA processing can improve results, and how the

application of these results could form part of a diagnostic approach in an at risk

population. We compare our results to and including carcinoembryonic antigen levels.

Results: Cell free DNA was isolated from 164 patients, including 71 patients with

oesophageal neoplasia, 50 patients with colorectal neoplasia, 35 patients without

endoscopic abnormality and 8 patients with Barrett’s oesophagus.

This is the first report of statistically significant differences in circulating DNA quantities and

patterns in patients with early oesophageal and colorectal neoplasia (p≤ 0.005). Logistic

regression of the best DNA marker for colorectal neoplasia demonstrated a ROC of 0.888,

with a sensitivity of 81.1% and specificity of 75.8%. Logistic regression of the best DNA

marker for oesophageal neoplasia demonstrated a ROC of 0.778 with a sensitivity of 81.3%

and specificity of 60.5%. Carcinoembryonic antigen performed poorly with a ROC of 0.547

and did not add diagnostically. There were no significant changes in markers from patients

resected at endoscopy.

Conclusions: These circulating markers in combination with other markers offer the

prospect of a simple blood test as a possible screen for colorectal and oesophageal

dysplasia and cancers in an at risk population.

Page 3: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-3

Table of Contents.

Abstract. ................................................................................................................................ 1-2

Table of Contents. ................................................................................................................. 1-3

Declaration. ......................................................................................................................... 1-10

List of Figures. ..................................................................................................................... 1-11

List of Tables. ...................................................................................................................... 1-13

Acknowledgements. ............................................................................................................ 1-15

Dissemination. .................................................................................................................... 1-15

Chapter 1 Gastro-intestinal cancers in the UK. ................................................................... 1-16

1.1 Introduction. ....................................................................................................... 1-16

1.2 Carcinogenesis theory. ........................................................................................ 1-17

1.2.1 Gene sequence changes. ............................................................................ 1-17

1.2.2 Epigenetic changes. ..................................................................................... 1-18

1.2.3 Extra-genetic changes. ................................................................................ 1-18

1.2.4 The metaplasia, dysplasia, carcinoma sequence. ....................................... 1-19

1.3 The Colon. ........................................................................................................... 1-19

1.3.1 Polyp adenoma and carcinoma pathogenesis. ........................................... 1-19

1.3.2 Serrated adenomas and Lynch syndrome. ................................................. 1-21

1.4 The oesophagus. ................................................................................................. 1-21

1.4.1 Barrett’s oesophagus pathogenesis. ........................................................... 1-21

1.4.2 Barrett’s dysplasia and carcinoma pathogenesis. ....................................... 1-22

1.5 Genetic hallmarks in colon and oesophageal adenocarcinomas. ....................... 1-25

Page 4: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-4

1.6 Clinical diagnosis of oesophageal and colonic carcinomas. ................................ 1-28

1.6.1 UK Colorectal cancer screening guidelines. ................................................ 1-28

1.6.2 UK Barrett’s oesophagus surveillance guidelines. ...................................... 1-30

1.7 Conclusions. ........................................................................................................ 1-31

Chapter 2 Circulating markers of gastro-intestinal cancers................................................ 2-33

2.1 Introduction. ....................................................................................................... 2-33

2.2 Circulating DNA tumour markers – genomics. .................................................... 2-34

2.2.1 Origins of circulating DNA. .......................................................................... 2-34

2.2.2 Circulating cell free DNA Studies. ............................................................... 2-35

2.2.2.1 Nuclear DNA studies. .............................................................................. 2-36

2.2.2.1.1 DNA quantification............................................................................ 2-36

2.2.2.1.2 DNA hyper-methylation. ................................................................... 2-38

2.2.2.1.3 DNA hypo-methylation. .................................................................... 2-39

2.2.2.1.4 DNA Mutation. .................................................................................. 2-39

2.2.2.1.5 Loss of heterozygosity, allelic imbalance, and aneuploidy. .............. 2-40

2.2.2.1.6 DNA sequencing. ............................................................................... 2-41

2.2.2.2 Mitochondrial DNA studies. .................................................................... 2-42

2.3 Circulating RNA tumour markers – transcriptomics. .......................................... 2-42

2.3.1 RNA marker studies. ................................................................................... 2-43

2.4 Circulating protein tumour markers- proteomics. .............................................. 2-44

2.4.1 Origins of circulating proteins. .................................................................... 2-44

2.4.2 Circulating protein studies .......................................................................... 2-45

Page 5: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-5

2.4.2.1 Protein signature studies ........................................................................ 2-46

2.4.2.2 Circulating Tumour antigen studies ........................................................ 2-47

2.4.2.3 Autologous T-cell activating antigen studies .......................................... 2-49

2.4.2.4 Auto-antibody studies ............................................................................. 2-49

2.4.2.5 Miscellaneous protein studies ................................................................ 2-50

2.4.2.5.1 MMP - matrix metalloproteinases .................................................... 2-50

2.4.2.5.2 TIMP 1 (Tissue inhibitor of metalloproteinases)............................... 2-50

2.4.2.5.3 Tissue Polypeptide antigen - cytokeratins 8, 18 and 19 ................... 2-51

2.4.2.5.4 Specific Tissue Polypeptide antigen – cytokeratin 18 ....................... 2-51

2.4.2.5.5 CYFRA 21-1 – cytokeratin 19 ............................................................. 2-51

2.4.2.5.6 Nuclear matrix protein antigens ....................................................... 2-51

2.4.2.5.7 VEGF – vascular endothelial growth factor ...................................... 2-52

2.4.2.5.8 CD 44 ................................................................................................. 2-52

2.4.2.5.9 Kallikrein ........................................................................................... 2-52

2.4.2.5.10 Laminin .............................................................................................. 2-53

2.4.2.5.11 βHCG - beta human chorionic gonadotrophin .................................. 2-53

2.5 Circulating tumour cells. ..................................................................................... 2-53

2.6 Circulating DNA markers in colonic and oesophageal adenocarcinoma. ........... 2-53

2.7 Conclusions. ........................................................................................................... 56

Experimental Hypothesis and Aims .................................................................................... 2-57

Chapter 3 Methods. ............................................................................................................ 3-58

3.1 Introduction. ....................................................................................................... 3-58

Page 6: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-6

3.1.1 Plasma processing. ...................................................................................... 3-58

3.1.2 DNA isolation and purification. ................................................................... 3-58

3.1.2.1 High Yield Plasma-XS nucleospin modification. ...................................... 3-59

3.1.3 DNA quantification. ..................................................................................... 3-60

3.1.4 Absolute qPCR. ............................................................................................ 3-63

3.1.4.1 Primer selection and optimization. ......................................................... 3-63

3.1.4.2 Amplicons and cycling conditions. .......................................................... 3-65

3.1.4.3 DNA quantity calculation. ....................................................................... 3-67

3.1.4.4 DNA dilution correction factor. ............................................................... 3-69

3.1.4.5 PCR assay co-efficient of variation results. ............................................. 3-69

3.1.5 CEA analysis. ................................................................................................ 3-70

3.1.6 Patient choice, pathological grading and disease staging. ......................... 3-71

3.1.7 Statistical analysis. ...................................................................................... 3-71

3.2 Experimental protocols. ...................................................................................... 3-73

3.2.1 Venepuncture. ............................................................................................ 3-73

3.2.2 Cell and plasma separation. ........................................................................ 3-73

3.2.3 Nucleospin DNA purification. ...................................................................... 3-74

3.2.4 qPCR. ........................................................................................................... 3-75

3.2.5 CEA analysis. ................................................................................................ 3-76

Chapter 4 Colon results. ...................................................................................................... 4-77

4.1 Demographics. .................................................................................................... 4-77

4.2 Patient groups. .................................................................................................... 4-78

Page 7: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-7

4.3 Mean and median marker values. ...................................................................... 4-78

4.4 Statistical significance - Mann Whitney U-tests. ................................................ 4-79

4.5 Logistic regression and ROC curve values. .......................................................... 4-80

4.6 Box plots and whisker analysis............................................................................ 4-82

4.7 Correlation of Total cfDNA with age. .................................................................. 4-86

4.8 Conclusions. ........................................................................................................ 4-86

Chapter 5 Oesophago-gastric results. ................................................................................. 5-88

5.1 Demographics. .................................................................................................... 5-88

5.2 Patient groups. .................................................................................................... 5-89

5.3 Mean and median marker values. ...................................................................... 5-90

5.4 Statistical significance - Mann Whitney U-tests. ................................................ 5-90

5.5 ROC curve values and Logistic regression. .......................................................... 5-91

5.6 Box plots and whisker analysis............................................................................ 5-93

5.7 Conclusions. ........................................................................................................ 5-96

Chapter 6 Paired colonic and oesophageal results. ............................................................ 6-98

6.1 Introduction. ....................................................................................................... 6-98

6.2 Colon results. ...................................................................................................... 6-98

6.2.1 Demographics . ........................................................................................... 6-98

6.2.2 Lesions. ........................................................................................................ 6-98

6.2.3 Mean results. .............................................................................................. 6-98

6.2.4 Significance testing. .................................................................................... 6-99

6.2.5 Individual markers. .................................................................................... 6-100

Page 8: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-8

6.2.5.1 CEA analysis. .......................................................................................... 6-100

6.2.5.2 Line 1 79bp - total cfDNA. ..................................................................... 6-100

6.2.5.3 Mitochondrial cfDNA. ........................................................................... 6-101

6.2.5.4 Alu 115bp fragment. ............................................................................ 6-102

6.2.5.5 Line 1 300bp fragment. ......................................................................... 6-103

6.2.5.6 Alu 247bp fragment. ............................................................................. 6-103

6.2.6 Marker ratios............................................................................................. 6-104

6.2.6.1 Line 1 79:300. ........................................................................................ 6-104

6.2.6.2 Alu 115:247. .......................................................................................... 6-105

6.3 Oesophageal results. ......................................................................................... 6-107

6.3.1 Demographics . ......................................................................................... 6-107

6.3.2 Lesions. ...................................................................................................... 6-107

6.3.3 Mean results. ............................................................................................ 6-107

6.3.4 Significance testing. .................................................................................. 6-108

6.3.5 Individual markers. .................................................................................... 6-108

6.3.5.1 Line 1 79bp - total cfDNA. ..................................................................... 6-108

6.3.5.2 Mitochondrial cfDNA. ........................................................................... 6-109

6.3.5.3 Alu 115 fragment. ................................................................................. 6-110

6.3.5.4 Line 1 300bp fragment. ......................................................................... 6-110

6.3.5.5 Alu 247bp fragment. ............................................................................ 6-111

6.3.6 Marker ratios............................................................................................. 6-111

6.3.6.1 Line 1 79:300. ........................................................................................ 6-111

Page 9: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-9

6.3.6.2 Alu 115:247. .......................................................................................... 6-112

6.4 Conclusions. ...................................................................................................... 6-113

Chapter 7 Discussion and conclusions. ................................................................................ 114

7.1 Control population results. .................................................................................. 114

7.2 Colonic results. ..................................................................................................... 116

7.3 Oesophageal results. ............................................................................................ 120

7.4 Paired results. ...................................................................................................... 123

7.5 Sample processing. .............................................................................................. 125

7.6 Implication for the origins of circulating DNA. .................................................... 125

7.7 Diagnostic testing. ................................................................................................ 127

7.8 Conclusions. ......................................................................................................... 128

Addendum ........................................................................................................................... 129

Reflections after Viva Voce examination ............................................................................. 129

Example of experimental amplification curves - Line 1 79bp .............................................. 131

Raw data - sample set up line 1 79bp ............................................................................... 7-132

Raw data - results Line 1 79bp .......................................................................................... 7-141

References ........................................................................................................................... 153

Page 10: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-10

Declaration.

Whilst registered as a candidate for the above degree, I have not been registered for any

other research award. Results and conclusions embodied in this thesis are my work and

have not been submitted for any other academic award.

Word count = 34,030

Page 11: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-11

List of Figures.

Figure 1 -Top 20 cancers by incidence in the UK 2009. ...................................................... 1-16

Figure 2 Adenoma pathways to cancer. ............................................................................. 1-20

Figure 3 COX II pathways in Barrett’s oesophagus. ............................................................ 1-24

Figure 4 Modified DNA loading results. .............................................................................. 3-60

Figure 5 Melt curve for Line 1 79 bp 400/400 primer amplification. ................................. 3-64

Figure 6 Line 1 79 bp standard curve. ................................................................................. 3-65

Figure 7 Plasma XS spin protocol. ....................................................................................... 3-75

Figure 8 Combined DNA marker ROC curve. ...................................................................... 4-81

Figure 9 Combined DNA marker plus CEA ROC curve. ........................................................ 4-82

Figure 10 CEA box plot analysis. ......................................................................................... 4-82

Figure 11 Total Nuclear DNA box plot analysis. .................................................................. 4-83

Figure 12 Mitochondrial DNA box plot analysis. ................................................................. 4-83

Figure 13 Alu 155bp DNA box plot analysis. ....................................................................... 4-84

Figure 14 Alu 247bp DNA box plot analysis. ...................................................................... 4-84

Figure 15 Line 1 300bp DNA box plot analysis. ................................................................... 4-85

Figure 16 Line 1 79:300 DNA ratio box plot analysis. ......................................................... 4-85

Figure 17 Alu 115:247 DNA ratio box plot analysis. ........................................................... 4-86

Figure 18 Correlation of total cfDNA with age. ................................................................... 4-86

Figure 19 Normal group vs. all invasive cancer group. ....................................................... 5-92

Figure 20 Normal group vs. all abnormal oesophagus group. ............................................ 5-92

Figure 21 Total Nuclear DNA box plot analysis. .................................................................. 5-93

Figure 22 Mitochondrial DNA box plot analysis. ................................................................. 5-94

Figure 23 Alu115bp DNA box plot analysis. ........................................................................ 5-94

Figure 24 Alu 247bp DNA box plot analysis. ....................................................................... 5-95

Figure 25 Line 1 300bp DNA box plot analysis. ................................................................... 5-95

Page 12: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-12

Figure 26 Line 1 79:300 DNA fragment ratio box plot analysis. ......................................... 5-96

Figure 27 Alu 115:247 DNA fragment ratio box plot analysis. ............................................ 5-96

Figure 28 Paired CEA values pre and post resection. ....................................................... 6-100

Figure 29 Paired total cfDNA values pre and post resection. ........................................... 6-101

Figure 30 Paired mitochondrial cfDNA values pre and post resection. ............................ 6-102

Figure 31 Paired Alu 115bp values pre and post resection. ............................................. 6-102

Figure 32 Paired Line 300bp values pre and post resection. ............................................ 6-103

Figure 33 Paired Alu 247bp values pre and post resection. ............................................. 6-104

Figure 34 Paired Line 1 79:300 ratio values pre and post resection. ................................ 6-105

Figure 35 Paired Alu 115:247 ratio values pre and post resection. .................................. 6-106

Figure 36 Paired total cfDNA values pre and post resection. ........................................... 6-109

Figure 37 Paired mitochondrial cfDNA values pre and post resection. ............................ 6-109

Figure 38 Paired Alu 115bp values pre and post resection. ............................................. 6-110

Figure 39 Paired Line 1 300bp values pre and post resection. ......................................... 6-110

Figure 40 Paired Alu 247bp values pre and post resection. ............................................. 6-111

Figure 41 Paired Line 1 79:300 ratio values pre and post resection. ................................ 6-112

Figure 42 Paired Alu 115:247 ratio values pre and post resection. .................................. 6-112

Figure 43 Paired Line 1 79:300 ratio values pre and post resection. ................................... 124

Figure 44 Paired Line 1 300bp values pre and post resection. ............................................ 124

Figure 45. DNA quantities in a patient with progressive oesophageal cancer. ................... 126

Page 13: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-13

List of Tables.

Table 1 Hallmarks of colorectal cancers. ............................................................................ 1-26

Table 2 Hallmarks of oesophageal cancers. ........................................................................ 1-27

Table 3 Circulating tumour antigens. .................................................................................. 2-48

Table 4 Colorectal circulating DNA papers............................................................................ 55

Table 5 Oesophageal circulating DNA papers. ....................................................................... 55

Table 6 Optimization experimental layout. ........................................................................ 3-63

Table 7 Optimization experimental results for line 1 79bp primer. ................................... 3-64

Table 8 PCR amplicons. ....................................................................................................... 3-65

Table 9 Plate layout for all experiments. ............................................................................ 3-66

Table 10 PCR cycling conditions for each amplicon. ........................................................... 3-66

Table 11 Mean standard curve values for each primer. ..................................................... 3-67

Table 12 Intra-assay coefficient of variation experimental results. ................................... 3-70

Table 13 Normal population indication for endoscopy. ..................................................... 4-77

Table 14 Mean values for all markers. ................................................................................ 4-78

Table 15 Median values for all markers. ............................................................................. 4-79

Table 16 Statistical significance testing. ............................................................................. 4-79

Table 17 ROC curve characteristics. .................................................................................... 4-80

Table 18 Normal population indication for endoscopy. ..................................................... 5-88

Table 19 Mean value for all markers. ................................................................................. 5-90

Table 20 Median value for all markers. .............................................................................. 5-90

Table 21 Statistical significance testing. ............................................................................. 5-90

Table 22 ROC curve characteristics. .................................................................................... 5-91

Table 23 Paired means pre and post resection, and normal and polyps. .......................... 6-99

Table 24 Significant P values. .............................................................................................. 6-99

Table 25 Paired means pre and post resection, and control and mucosal neoplasia. ..... 6-108

Page 14: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-14

Table 26 Significant P values. ............................................................................................ 6-108

Page 15: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

1-15

Acknowledgements.

This project would not have been possible without the support of my wife Zoe, and the

understanding of my children Ben, Alex and Jamie.

It also took the belief and vision of Pradeep, the experience and understanding of Ian, and

the support of many who encouraged and cajoled me into finishing this. They know who

they all are.

Dissemination.

R.J.Mead, M.D.Duku, P.Bhandari, I.Cree. Circulating tumour markers can define patients

with normal colons, benign polyps, and cancers. Br J Cancer. 2011 Jul 12;105(2):239-

45.Epub 2011 Jun.

G.R.Longcroft-Wheaton, M.Duku, R.J.Mead, D.Poller, P.Bhandari. Acetic acid spray is an

effective tool for the endoscopic detection of neoplasia in patients with Barrett's

esophagus. Clin Gastroenterol Hepatol. 2010 Oct;8(10):843-7. Epub 2010 Jun 30.

Digestive diseases Federation 2012.

Circulating tumour markers can discriminate between patients with and without

oesophageal neoplasia. (Oral presentation)

British Society of Gastroenterology Meeting 2011.

Circulating tumour markers can define patients with normal colons,

benign polyps and cancers. (Oral presentation)

World Congress of Gastroenterology 2009.

Impact of EMR on the management of Barrett’s neoplasia in a UK setting. (Poster)

British Society of Gastroenterology Meeting 2007.

Endoscopic mucosal resection techniques for upper gastrointestinal neoplasms: A video

review. (Oral presentation)

Page 16: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-16

Chapter 1 Gastro-intestinal cancers in the UK.

1.1 Introduction.

Gastro-intestinal carcinomas are some of the most common and rapidly increasing cancers

in the United Kingdom (UK) and the western world1-3 (Figure 1). Societal costs are

considerable with emotional, structural, and financial losses.

The epithelial cells lining the bowel wall are the most usual sites affected, developing

typically into squamous or adeno-carcinomas.

Colorectal adenocarcinoma, the most common gastro-intestinal cancer and oesophageal

adenocarcinoma, the most rapidly increasing cancer in the UK, are perhaps the two most

important gastro-intestinal cancers of modern times. Twenty-three thousand six hundred

and twenty-three people die from these two conditions in the UK each year (2010)2, and

these are the focus of this thesis.

Figure 1 -Top 20 cancers by incidence in the UK 2009.

Page 17: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-17

1.2 Carcinogenesis theory.

Current theory suggests that slow accumulation of random genetic and epigenetic

aberrations, catalysed by carcinogens and inherited deoxy-ribonucleic acid (DNA) repair

enzyme defects, lead to pro-cancerous expression of key genes. Eventually this results in a

common cancer phenotype.

Inherited genetic coding, environmental factors, inflammation (driving increased cellular

replication), and the build up of errors from repeated genetic replication (ageing) effect the

speed at which cancers develop, and reflect the age related incidence.

1.2.1 Gene sequence changes.

Genes promoting and supporting cellular growth (oncogenes), genes involved in cellular

differentiation and shut down (tumour suppressors), and genes controlling cellular death

(apoptosis) are present in all cells. Pro-cancerous mutations to these key genes and/ or

epigenetic changes may produce uncontrolled proliferation signals. Similarly failure of

tumour suppression and apoptosis pathways also result in uncontrolled proliferation.

Neoplastic transformation often requires both.

Primary genetic damage usually occurs through oxidation of DNA bases and itself may lead

to failure of genetic expression. Secondary inaccurate repair of the damaged bases leads to

mutations in the genetic sequence which may or may not alter gene expression. These

changes occur throughout our lives and largely result in ageing, but at each stage neoplasia

may result.

Page 18: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-18

Increasing age and genetic damage may result in chromosomal instability (CIN). Loss of

chromosomal areas (deletion) and multiplication of others (amplification) may occur

increasing the rate of genetic change and increasing the risk of carcinoma development.

Abnormal copy numbers of complete chromosomes (aneuploidy) may be seen in advanced

neoplasia.

1.2.2 Epigenetic changes.

Changes promoting cancer are not restricted to genetic coding sequences. Epigenetic DNA

changes allow modification of gene expression without change to the coding (genetic)

region sequence. The most studied effect is at islands of relatively high Cytosine and

Guanine base content (CpG islands) close to gene promoter regions. High levels of cytosine

methylation (5-methylcytosine) in these CpG islands acts as a physiological gene silencing

mechanism. Aberrant hypermethylation, and therefore silencing, of tumour suppressor and

apoptotic genes are a frequently reported change in neoplasia progression.

Hypomethylation of CpG sites is also seen in neoplasia, potentially releasing control of

oncogenes, and by a different mechanism increasing susceptibility to DNA breaks (CIN)4.

A less well studied epigenetic phenomenon, genetic imprinting, occurs when DNA

methylation fixes genetic expression to a single predetermined parental allele. Loss of

methylation and therefore imprinting of oncogenes may allow uncontrolled expression of

the usually silent allele.

1.2.3 Extra-genetic changes.

Histone proteins act as structural scaffolding for DNA. They dictate the folding of DNA

within chromatin influencing access to and expression of genes.

Page 19: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-19

Epigenetic methylation, acetylation, and substitution of key amino acids in these proteins

alter their structure changing gene accessibility and regulation which may favour

expression of pro-cancerous genes5 (or repression of tumour suppressors).

Stromal cells and the local extracellular matrix are also extra-genetic co-factors supporting

the development of cancer6. Co-evolution of supporting cells7, and co-option of

inflammatory cells to provide further local growth stimulus8 demonstrate the range of

possible contributory factors combining to create the neoplastic environment.

1.2.4 The metaplasia, dysplasia, carcinoma sequence.

With increasing genetic damage gastro-intestinal lesions develop through common

pathological stages: Benign metaplasia (oesophageal cancer only – Barrett’s oesophagus),

pre-cancerous dysplasia; carcinoma in situ; and finally invasive cancer. A carcinogenic

process is required for neoplastic changes to develop but once invasive cancer has formed

it is self sustaining.

1.3 The Colon.

The colon is lined by columnar-type epithelium, measuring approximately 150cm long,

joining the small intestine to the anus. Its main function is to reabsorb water and control

waste excretion. Anatomically it is divided into six parts; caecum and ileo-caecal valve;

ascending colon; transverse colon; descending colon; sigmoid colon; and rectum.

1.3.1 Polyp adenoma and carcinoma pathogenesis.

A sequential morphological and pathological change from normal mucosa to adenoma with

low (LGD) then high grade dysplasia (HGD), and finally carcinoma is recognised as the most

common route for disease progression explaining 80% of cancers in the normal population9,

10.

Page 20: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-20

The proposed genetic model for this follows on from work by Fearon and Vogelstein11, with

growth of abnormal epithelial cells from the surface of the bowel mucosa into the

glandular crypts.

The initiating event is loss of the Wnt (Wingless and Int drosophila gene) pathway through

APC dysfunction, often due to mutation, followed by further genetic loss and damage to

onco- and tumour-suppressor genes eventually leading to cancer.

Three central combinations have been associated with adenomas that progress to

carcinoma: 17p loss (P53) and Kras mutation, 8q (C-MYC) and 13q (mir 17-92 cluster) gain,

and 18q (DCC, SMAD4) loss and 20q (MMP9, SRC, TNFRSF6B) gain 12. However, although

present they are not sufficient to initiate cancer, with further genetic change required.

Figure 2 Adenoma pathways to cancer.

CIMP= CpG hypermethylation phenotype, MMR = mis-match repair gene, APC = adenoma Polyposis coli, DCC= deleted in colon cancer, P53 = protein 53, MSS = microsatellite stable, MSI = microsatellite instability, SMAD = mothers against decapentaplegic homolog, hMLH1 = human mutL homolog 1, P16(INK4) = cyclin dependent kinase inhibitor 2A, PTEN = phosphatase and tensin homolog.

13

Page 21: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-21

1.3.2 Serrated adenomas and Lynch syndrome.

A subset of colorectal cancers has been characterized with deficient DNA mismatch repair

linked to mutations of repair genes such as MSH2 (MutS Homolog 2), MSH6 (MutS

Homolog 6), MLH1 (MutL Homolog 1), and PMS2 (post-meiotic segregation increased 2)14, 15.

These mutations result in high frequency microsatellite instability (H-MSI), instability in the

repeat number of usually highly conserved 1-6 bp DNA sequences present throughout the

normal genome, reflecting defective DNA mis-match repair.

Inherited defects of these genes lead to H-MSI Hereditary Non-Polyposis Colorectal Cancer

or Lynch syndrome and account for 6% of all colon cancers16.

Sporadic H-MSI is also found in 15-20% of colon cancers17, and sessile serrated adenomas

are likely precursors. High levels of BRAF mutations, an oncogene similar to K-Ras, and

aberrant DNA methylation are found in serrated adenomas and mirror the genotypical

changes seen in sporadic H-MSI cancers18.

1.4 The oesophagus.

The oesophagus is lined by stratified squamous epithelium. Measuring 25 cm in length, the

oesophagus joins the pharynx to the stomach, with an anatomical sphincter at its lower

end. Its main function is to transmit food from the pharynx to the stomach, and prevent

backwash of acid and stomach contents.

1.4.1 Barrett’s oesophagus pathogenesis.

Barrett’s oesophagus is a benign condition, involving metaplastic change of the epithelium.

However, once Barrett’s oesophagus has developed the risk of adenocarcinoma increases

by 30-150 folds. Metaplastic change is not seen in colonic neoplasia.

Page 22: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-22

Barrett’s oesophagus was first described in 195019 and involves replacement of squamous

epithelium with a specialised columnar epithelium, including intestinal-type mucosa. The

development of Barrett’s oesophagus and adenocarcinoma is considered to be driven by

reflux of acid and bile (refluxate) into the lower oesophagus. However, evidence for this is

mainly epidemiological in humans, with gastro-oesophageal reflux linked to obesity.

At a molecular level, caudal type homeobox 1 and 2 (CDX1, CDX2) expression are perhaps

the only recognised pathogenetic molecular changes in Barrett’s oesophagus. They are not

usually expressed in squamous or gastric mucosa, and are involved only in the

development of normal intestinal-type mucosa.

Conjugated bile acids and the inflammatory cytokines Tumour necrosis Factor α (TNF-α)

and Interleukin 1 β (IL-1β) have been shown to increase CDX1 mRNA expression in vitro in

colorectal cancer cell lines, but only when the CDX1 promoter is un-methylated or partially

methylated. Study of bile salt exposure in rat squamous cell lines and in metaplastic cells

from the rat OGJ have also shown increased CDX2 expression.

Although the promoter methylation status in squamous epithelium prior to Barrett’s

oesophagus formation is uncertain, these studies at a molecular level link refluxate with

increased expression of genes involved development of normal intestinal type mucosa and

possibly with development of Barrett’s oesophagus. Perhaps this is key to further

understanding the molecular pathway and explaining the well known epidemiological

associations20-22.

1.4.2 Barrett’s dysplasia and carcinoma pathogenesis.

Once Barrett’s oesophagus is established it remains unclear which changes lead to

adenocarcinoma, but inflammatory pathways are key.

Page 23: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-23

The healing of oesophagitis with acid suppression and experiments with bile salts causing

acid disassociation constant (pka) dependent mucosal injury demonstrate the role of acid

and bile salts in inflammation. More specifically Taurine-conjugated bile salts cause chronic

mucosal injury when the oesophageal reflux is acidic (pH 4); glycine-conjugated bile salts

when the pH is 4–6; and un-conjugated bile salts when the pH is neutral or alkaline23.

Clinical studies demonstrating prolonged episodes of acid reflux and higher levels of

oesophageal taurine-conjugated and un-conjugated bile salts in patients with Barrett’s

oesophagus, tie the principles together. Acid and bile as the causes of inflammation in

Barrett’s seem certain24-28.

At a molecular level the causes are less certain. Chronic acid and bile salt injury induce

reactive oxygen species and free radicals, depleting antioxidants and increasing the

expression of oxidative-stress-related genes29. Reduced levels of glutathione and vitamin C

in the metaplastic epithelium indicate that anti-oxidant defences are compromised, and

high levels of oxygen free radicals and lipid-peroxidation products confirm this29.

Primary free radical DNA damage results in genetic change which may include key cell

regulatory genes, and therefore a molecular beginning for the neoplastic process. Cells

with such mutations would usually undergo forced cell-cycle arrest or apoptosis by a P53

dependent mechanism. However, bile salts and inflammation prevent this through

proteosome mediated P53 degradation and migration inhibition factor by-pass of P53

function allowing genetic abnormalities to accumulate8, 30. Multiple other pathways are

likely to contribute. Inflammatory stimulus of prostaglandin synthesis through the

arachidonic acid pathway (Figure 3 below) is also important. Inducible cyclo-oxygenase-2

(COX-2) is found in increasing amounts through dysplasia and carcinoma31 with

prostaglandin E2, one of the main product types from the COX-2 pathway, increasing cell

proliferation32.

Page 24: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-24

Protein kinase C and P38 mitogen activated protein kinase (MAPK*) pathways are

proposed as further growth promoting mediators of the increased COX-2 products 33, 34.

Lastly experiments treating cell lines with pulsed acidified bile show an important role for

the C-myc oncogene protein, contributing to uncontrolled cellular growth35

Figure 3 COX II pathways in Barrett’s oesophagus.

* Previously known as Extra-cellular regulated kinase (ERK).

Page 25: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-25

Together these findings provide evidence for bile and acid reflux driving genetic and

molecular change, leading to the accumulation of genetically abnormal cells, with a pro-

proliferative drive able to escape the normal cell cycle controls. Cells become dysplastic

and finally cancerous.

Clinical observations demonstrate a more complex understanding is needed, with neoplasia

development despite healing of oesophagitis and also following acid suppression with a

proton pump inhibitor (PPI).

Some evidence to explain this follows from prolonged proton pump inhibitor (PPI - acid

suppressing medication) usage itself. Secondary bacterial colonization of the gullet at

neutral pH can produce inflammatory de-conjugated bile salts which continue to inflame

the gullet36, and secondary hyper-gastrinaemia acting at cholecystikinin 2 (CCK2) receptors

drives cellular proliferation37. Perhaps unsurprisingly even on a PPI up to 80% of patients

continue to reflux some acid and up to one third bile salts 38-40.

Human study substantiating the pathological mechanisms in Barrett’s oesophagus is not

available, with evidence mainly from cell line studies, animal models and case reports30, 41-43.

This may not accurately reflect human pathophysiology, and therefore molecular

confirmation for the role of acid and bile reflux in human neoplasia remains unproven.

1.5 Genetic hallmarks in colon and oesophageal adenocarcinomas.

Over the last 50 years there have been many studies of genetic change in cancers.

Observation of genetic, epigenetic, and extra-genetic changes have demonstrated multiple

pathways to cancer, however an incomplete and confusing picture has emerged. To gather

and organise this new information a conceptual framework, “hallmarks of cancer”, was

developed and published in 200044 and updated in 201145.

Page 26: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-26

This serves to provide common understanding across cancers whilst allowing work to

continue searching for further understanding of cancer pathogenesis.

These “hallmarks of cancer” are: Self-sufficiency in growth signalling; insensitivity to

growth-inhibitory (antigrowth) signalling; evasion of programmed cell death (apoptosis);

limitless replicative potential; sustained angiogenesis; and tissue invasion and metastasis44.

They serves as a useful way to review the main genetic changes found in oesophageal and

colonic cancer and these are tabulated below.

Table 1 Hallmarks of colorectal cancers.

HALLMARKS of CANCER (DNA)

Colorectal cancer

Genetic change Prevalence Reference

Self-sufficiency in growth signalling

SRC up regulation 90+% Talamonti 1993, Frame 2004.

C-Myc up regulation 66% Smith 1993, Sansom 2007.

K-Ras up regulation 37% Salbe 2000, DeReynies 2008.

Insensitivity to growth-

inhibitory (antigrowth)

signalling

P53 down regulation 90+% Veloso 2000

SMAD-4 down regulation 40% Woodford-Richens 2001

APC down regulation 90+% Sansom 2007

PTEN down regulation 20% Nassif 2004, Goel 2004.

Evasion of programmed cell

death (apoptosis)

P53 down regulation 50% Jansson 2001

DCC down regulation 50-70% Forcet 2001

mir 17-92 up-regulation 100% Diosdado 2009, He 2005

TNFRSF6B up regulation 53 - 73% Pitti 1998, Mild 2002.

SMAD 4 down regulation 40% Woodford-Richens 2001

Limitless replicative potential Telomerase up regulation 90% Engelhardt 1997.

Sustained angiogenesis

HIF up regulation 44-55% Cao 2009, Wu 2010.

VEGF up regulation 37-100% Okuno 2001, Xiong 2002

MMP 9 up regulation 63-74% Xeng 1999

TGF-β up regulation 38% Xiong 2002

FGF down regulation - Mathonnet 2006.

Tissue invasion and metastasis

E-cadherin down regulation 19 - 33% Dorudi 1993, Rosivatz 2004

MMP-9up regulation 20-75% Zeng 1999, Fan 2006.

TIMP up regulation 42 - 100% Zeng 1995, Murashighe 1996

nm 23 H(1) down regulation 62% Fan 2006.

Page 27: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-27

Table 2 Hallmarks of oesophageal cancers.

HALLMARKS of CANCER (DNA)

Oesophageal cancer

Genetic change Prevalence Reference

Self-sufficiency in growth signalling

SRC up regulation 20% Kumble 1997.

β-Catenin up regulation 60% Bailey 1998, Seery 1999.

COX II up regulation 80 - 97% Wilson 1998, Lagorce 2003.

EGFR / ERBb2 / HER2neu up regulation 10 - 70%

Kim 1997, Al-Kasspooles 1993, Hardwick 1997.

Insensitivity to growth-

inhibitory (antigrowth)

signalling

P53 down regulation 95 -100% Barrett 1999.

SMAD-4 down regulation 20 - 35% Barrett 1996, Van Dekken 2008.

P16 down regulation 12 - 100% Van Dekken 2008, Shi 2008.

Cyclin D up regulation 30 - 92% Bani-Hani 2000, Murray 2006.

Cyclin E up regulation 14% Sarbia 1999.

P27 down regulation 83% Singh 1998

Evasion of programmed cell death (apoptosis)

P53 down regulation 64 - 87% Younes 1993, Van Dekken 2008.

P16 down regulation 68 - 75% Wong1997, Klump 1998.

15 LOX-1 (13-S-HODE) down regulation - Shureiqui 2001

Fas ligand up regulation 100% Bennett 1998, Younes 1999.

Limitless replicative potential Telomerase up regulation 100% Morales 2001, Shammas 2008.

Sustained angiogenesis

HIF-1, and 2 up regulation 51 - 60% Stein 2004, Griffiths 2007.

VEGF up regulation 64 - 100% Saad 2005, Von Rahden 2005.

MMP up regulation 60- 95% Salmela 2001, Grimm 2010.

Tissue invasion and metastasis

E-cadherin down regulation 100% Bailey 1998, Washington 1998.

MMP-1, 7, 12 up regulation 60-95% Salmella 2001, Grimm 2010.

TIMP-1, &3 up regulation 53 - 73% Salmella 2001.

CD-44 up regulation 70 - 75% Castella 1996, Lagorce 1998.

β-cathepsin up regulation 73% Hughes 1998.

Page 28: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-28

Clearly some changes appear in both cancers but there is temporal heterogeneity in the

dysplasia carcinoma sequence. From a diagnostic point of view, without knowing the stage

of disease this makes the diagnostic use of specific genetic changes less attractive. Some of

the changes can also be found in “normal” patients46. It seems unlikely that a genetic

signature diagnosing early cancers or dysplasia will emerge, although patterns may suggest

risk of cancer and subtype.

1.6 Clinical diagnosis of oesophageal and colonic carcinomas.

Current clinical diagnosis of gastro-intestinal cancer is based around a clinical history,

examination, and routine blood tests. The final diagnosis is predominantly made at

endoscopy with confirmation on histological examination47-50. There are no diagnostic

blood or stool tests recommended for early or late gastro-intestinal cancers 51.

The current clinical approach is certainly effective, but early diagnosis of gastro-intestinal

cancers is unlikely as symptomatic change often occurs late in disease. Clinical symptoms

and blood tests are not specific criteria and are often found in the normal population

without cancer52, 53.

Clinically this is a well recognised problem. There are clear costs involved in investigating

normal patients, but there are also substantial outcome advantages gained from early

diagnosis54. Fortunately in the UK we have programmes screening for colorectal cancer and

providing surveillance in Barrett’s oesophagus patients for oesophageal adenocarcinoma48,

55.

1.6.1 UK Colorectal cancer screening guidelines.

Average risk, asymptomatic patients aged 60-70 years of age are offered screening with

faecal occult blood testing (fOBT) 55.

Page 29: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-29

For every 100 participants, the programme is expected to find 20 positive faecal occult

bloods, detect 2 patients with colorectal cancer and 6 patients with polyps55.

While practical and affordable, this system leaves room for improvement 56, 57, with a low

estimated point sensitivity for cancer (40.58%) , and even lower sensitivity for significant

dysplastic polyps (5.00%)55. Many cancers and pre-cancerous polyps will be missed. Gains in

sensitivity are made by offering a programme of fOBT testing, but compliance is likely to

fall over the program timescale. Initial UK compliance rates of 56.8%55 are unlikely to

improve much, as demonstrated by American studies, which have a similar patient group

and which share common health values. Worldwide some programmes only achieve 18% 58-

61 compliance, and put the UK achievement into perspective. It seems unlikely that further

gains will arise from improved compliance, and opportunities to prevent colon cancer will

be missed.

Immuno-histochemical testing for faecal occult blood (iFoBT) will offer some gains62-65 and

it is likely this will replace fOBT in the near future. The test utilises a specific antibody

reaction to human globin, labelled with a fluorescing compound. This allows more specific

and sensitive testing for faecal human blood, and with a numerical fluorescence result

allows a variable test sensitivity and specificity. In combination with other risk factors such

as age weight, sex, and smoking history a tailored risk assessment for the individual could

be produced. However, the need for faecal sampling remains, and in the longer term

investment into better screening approaches may be needed to tackle the relatively low

uptake.

Page 30: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-30

Recently a large clinical trial looking at the role of flexible sigmoidoscopy in colorectal

cancer screening has concluded 66. This demonstrated reduced incidence and mortality

from colorectal cancer, with improved sensitivity for cancer at 66%, and for polyps of 97%.

However, the sensitivity of flexible sigmoidoscopy for detecting cancer is limited to the left

side of the colon, and quality issues regarding completion of the test remain. Compliance

with the intervention is approximately 1/3 of those invited (although in smaller trials this

has been as high as 46%)67. This is undergoing national roll out as part of the bowel cancer

screening programme (BCSP).

Biomarkers from blood and urine are active areas of research looking at alternative or

additive strategies to improve the screening programme avoiding invasive testing. This

would be more acceptable for population screening, and therefore could offer a new gold

standard.

Hospital based endoscopic screening in high and moderate risk cancer groups is also part of

a national approach, advocated by specialist bowel screening guidelines 47.

1.6.2 UK Barrett’s oesophagus surveillance guidelines.

Finding Barrett’s oesophagus over 3 cm in length prompts entry into surveillance

programmes 48. This programme requires upper endoscopy every 2 years. If dysplasia is

discovered, guidelines recommend more frequent endoscopy (yearly) and the number of

biopsy samples taken increase.

This approach to “screening” presents a numbers of difficulties, not least the identification

of patients with Barrett’s oesophagus within the general population. Current practice only

identifies Barrett’s incidentally, and estimation of patients not identified for screening

suggests 20 unidentified : 1 in screening 68.

Page 31: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-31

Ongoing study of techniques to diagnose Barrett’s in high risk populations is ongoing, with

the best so far the cytosponge69.

Even when in surveillance programmes evidence of improvement over a symptomatic

diagnostic approach is limited, with many cases discovered at a late stage and often

inoperable. Outcome data is unimpressive70-77, and worldwide these programmes are areas

of ongoing discussion78. A randomised trial comparing screening to a symptomatic

diagnostic approach in Barrett’s oesophagus is currently under way in the UK (Barrett’s

oesophagus surveillance study - BOSS trial).

Identification of dysplastic change and early cancer is the main reason for the diagnostic

difficulties. Visual abnormalities are subtle and in the low prevalence surveillance

population are easily missed even in expert hands. The current gold standard of systematic

quadrantic biopsies, designed to overcome this problem, however only samples only 2% of

the total surface area and the poor diagnostic outcome is therefore perhaps unsurprising.

Research into improving the detection of abnormal areas in Barrett’s has been ongoing for

many years. In expert hands, in a high prevalence population, 95% of cases can be

identified with 2% acetic acid dye spray79. This is an effective and economical technique,

and if the population prevalence could be enriched this would be a suitable screening

technique.

1.7 Conclusions.

Oesophageal and colon cancer are important gastro-intestinal cancers, with 23,610

deaths a year caused by them.

Despite a great deal of research over many years, a diagnostic molecular picture

remains elusive, and their pathogenesis remains incompletely understood.

The current gold standard invasive diagnostic tests mainly diagnose advanced

cancers, even in screening and surveillance programmes.

Page 32: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Gastro-intestinal cancers in the UK.

1-32

Better diagnostic testing for early and pre-cancerous neoplasia would almost

certainly result in better patient outcome.

The use of biomarkers to improve current testing for colonic and oesophageal

neoplasia, or to enrich the incidence in the Barrett’s surveillance population, is a

desirable area for clinical research.

Page 33: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-33

Chapter 2 Circulating markers of gastro-intestinal cancers.

2.1 Introduction.

Cancer biomarkers have been sought for many years with prostate specific antigen (PSA) in

prostatic adenocarcinoma a good clinical example of what can be achieved80.

The ideal diagnostic marker for cancer is well characterised as a non-invasive test81, 82 which

is socially highly acceptable, with 100% sensitivity and specificity for all stages including

pre-cancerous lesions, and correlating with stage and treatment. It must be inexpensive,

allowing population screening, utilising generally available technology, and be operator

independent.

Unfortunately, in colorectal cancer tests there are problems with social acceptability,

invasiveness, and accuracy 56, 83, and in oesophageal cancer there is no such test.

Studies of circulating DNA, RNA, proteins and circulating tumour cells have generated many

potential cancer markers and will be reviewed below, but none have found a clinical

diagnostic role to date51.

Routine sampling of blood and urine, using polymerase chain reaction (PCR) assays of

nucleic acids and enzyme-linked immuno-sorbent assays of proteins (ELISA) are already

part of accepted clinical practice84-87. Therefore new circulating DNA, ribonucleic acid (RNA)

and protein markers hold great hope for diagnostic and screening purposes, and research

in these areas may quickly advance from bench to bedside.

Circulating cell free DNA (cfDNA) was one of the earliest targets for researchers studying

cancer88 and is the most established. With established DNA protocols and extensive PCR

expertise in our laboratory, circulating cfDNA is therefore the focus of this thesis.

Page 34: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-34

2.2 Circulating DNA tumour markers – genomics.

2.2.1 Origins of circulating DNA.

Circulating DNA in the human blood stream was first described in 1948 by P. Mandel88, and

since then has been described in many forms .89-91 The quantity and pattern of circulating

DNA appears to differ in different physiological states including pregnancy92, cancers 90, 91,

93-102, benign diseases 103-105, and after trauma106, 107. Although the potential of cfDNA to

act as a diagnostic marker is clear, the origin of the DNA and the mechanisms leading to its

presence are not clear.

The most commonly cited theory is that increased and abnormal apoptosis in tissue leads

to increased circulating DNA. This theory is based around circulating cfDNA fragmentation

analysis demonstrating a “fragment ladder” phenomenon at multiples of approximately

180bp, similar to the pattern found in cells apoptosing 108, 109.

However, there are five main problems with this theory in cancer. Firstly inhibition of

apoptotic pathways is considered one of the hallmarks of cancer110 so high DNA levels in

blood don’t seem to correlate with this. Secondly, degradation of apoptotic cells generally

occurs without DNA spillage and therefore levels of cfDNA should remain largely

unchanged 111. Thirdly, DNA fragmentation patterns and quantities change as cancer

progresses, however apoptotic processes do not. Fourthly, in pregnancy circulating

maternal DNA length is increased, although there is no obvious reason for an increase in

maternal apoptosis 112, 113.

Lastly circulating DNA quantities in healthy normal patients and cancer patients can be

similar 101, 114.

Page 35: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-35

A dynamic process combining necrosis and apoptosis seems most likely to explain

increased circulating DNA in cancer patients. A study of cell lines (following induced cellular

necrosis, and apoptosis) and further study in patients supports this, demonstrating

fragments > 10,000 bp as well as an “apoptotic ladder” phenomenon in both108, 115.

Circulating white blood cells and ‘free’ nucleases are increased in the blood of cancer

patients 116, 117, and it is possible that these may break larger fragments from necrotic cells

into multiple smaller pieces. These two observations potentially explain different ‘total

‘DNA levels as well as different fragmentation patterns in the cancer and normal patients.

This is an attractive and potentially comprehensive theory.

However, this is likely to be overly simplistic with findings from other studies seeming to

paint a more complicated picture. Unexplained observations of homeostatic DNA

excretion118, 119; cell to cell nucleic acid transfer120-123; cell bound circulating DNA (ccDNA) 95,

124-126; and immune and genetic modulating nucleic acids127, 128 need to be included to

complete the full picture.

Perhaps some processes engendering survival advantage in normal patients are

advantageous to cancer cells possibly helping evasion of immune regulation, and in the co-

opting of local and distant cells which aid the cancerous process.

2.2.2 Circulating cell free DNA Studies.

DNA markers in blood have been researched most frequently reflecting the clinical

familiarity with blood sampling and the interaction of blood with all cells in the body. Urine

has not been studied as much but is promising, especially in the study of urological

cancers129-134. Urine has the advantage of collecting DNA over a period of time, and

contains mainly smaller fragments, a possible source of enriched tumour DNA135-139.

Page 36: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-36

Picking between these two fluids is difficult; the use of urine is advantageous in some

respects as sampling is non-invasive, however perhaps for screening purposes social

embarrassment may impact on patient acceptability. Research in both is underway136, 140-143.

This thesis used blood because of ease of sampling patients at the time of endoscopy and

surgery.

Circulating DNA can be isolated from plasma and serum blood products. It is unclear which

of the two offers the best medium94, 115, 144-149. Although there is more cfDNA in serum than

plasma, the source and tumour specificity of the extra cfDNA is uncertain100, 108, 148. It is

currently felt that contaminating genomic leukocyte DNA may be the source150, 151. Plasma

has become increasingly accepted as a better, purer source than serum148, 150-154.

2.2.2.1 Nuclear DNA studies.

2.2.2.1.1 DNA quantification.

Across multiple cancer types and multiple genetic sequences total cfDNA quantification for

diagnostic purposes is encouraging89, 95, 155-158.

Study results in cancer patients serum show an increase in total DNA quantity, and

“apoptotic” long fragment (>180 bp) DNA115, 149. Short and long fragment ratios change in

cancer patients reflecting relatively more “apoptotic” long fragment circulating DNA, and

providing a more sensitive discriminator between normal and cancer patients.

Many studies look at prognosis as well as diagnosis, with higher levels of cfDNA generally

associated with a poorer prognosis96, 159-161.

Page 37: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-37

A single study in the oesophagus studying squamous carcinoma looked quantitatively at

cyclin D1 oncogene amplification and showed a relative increase in copy number in both

tissue and plasma, with prognostic significance162. Oesophageal adenocarcinoma has not

been studied.

In colorectal cancer a short (115 bp) Alu sequence has been used to accurately quantify

total circulating serum DNA, with a significant difference (P 0.006) between the cancer

and control group. A longer (247 bp) Alu sequence, proposed as another measure of

apoptotic DNA, was also significantly different. However, it was the ratio of long : short

fragments which performed best at discriminating cancer form normal patients with a

receiver operating characteristic curve (ROC) of 0.78 149. Alu sequences are a family of

abundant repetitive DNA elements in the human genome classified as short (<300 bp)

interspersed elements (Sine). Line or Long interspersed elements are a group of similar

genetic elements, also found in large numbers throughout the human genome. A Line 1 79

bp sequence has also been shown to be significantly elevated in colorectal cancer and a

Line 1 300 bp sequence in breast cancer serum147, 163 . Line 1 79 bp may prove to be a more

accurate measure of total DNA with the ability to measure small (<100 bp) DNA fragments

and reflecting total tumour DNA more accurately.

PCR analysis of these interspersed element markers is technically simple and economical

making them an attractive option for diagnostic testing. With the majority of studies above

looking at advanced cancers using serum, the use of plasma may provide more accurate

results and may allow testing in early and pre-cancerous disease. In particular study of

plasma cfDNA in colorectal and oesophageal cancer had not been explored and this forms

the basis for this research project using ALU and LINE 1 markers.

Page 38: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-38

Quantitative study of DNA is not standardised with results affected by purification

protocols and inflammatory disease, which confound interpretation89, 109, 115, 149, 164-176. The

careful selection of purification techniques and control groups in the analysis of early and

pre-cancerous changes is therefore crucial in any future study.

2.2.2.1.2 DNA hyper-methylation.

Diagnostic approaches looking at epigenetic methylation are reported in many cancers and

pre-cancerous conditions90, 177-179.

Early papers looking at cancers and dysplastic tissues180 showed the diagnostic possibilities

studying multiple aberrantly hyper-methylated promoter regions in colonic and

oesophageal adenocarcinomas181, 182. Difficulties translating the technology into use for

much smaller quantities of circulating DNA held back advances in this field, however the

recent and novel use of restriction enzymes 178 and better purification techniques have

begun to overcome these 183. This approach requires complex pre-PCR processing of

samples, and may make interpretation uncertain.

However, the resulting serum and plasma studies do show different patterns depending on

cancer type184 and can even discriminate between patients with colon cancer and intestinal

polyps 185 .

Transmembrane protein containing epidermal growth factor and follistatin domains (TPEF)

186, Aristaless-like Homeobox-4 (ALX4) and Septin 9 (S9)177, 179 in particular showed good

results in colonic neoplasia. S9 has undergone further validation in the only large screening

study to date, presented in abstract form only187. Disappointingly it had a 37% detection

rate for stage I early cancers, and only a 50% sensitivity including more advanced cancers.

The sensitivity for diagnosing adenomas was not reported, and leaves results in 6431/7938

study participants unexplained 187. This represents the only clinically relevant screening

study to date.

Page 39: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-39

With variation in hyper-methylated genes in normal tissue suggesting that change in the

methylation pattern is a marker of neoplasia risk rather than diagnostic of cancer

development188. Further complexity in interpreting results is added by the observations

that differing methylation patterns are found in cancer patients with the same disease, and

even differing patterns in the same patients when comparing pre-cancerous changes185.

Increasing methylation occurs with increasing age and means that control groups must be

corrected for this, further complicating interpretation.

With this level of complexity required for interpretation it seems unlikely a diagnostic

methylation pattern will be found.

2.2.2.1.3 DNA hypo-methylation.

Hypomethylation of key oncogene promoter regions may also contribute to oncogenesis,

allowing increased replication of cancer promoting proteins189. A study of circulating

lymphocyte DNA showed a significant decrease in genomic methylation in patients with

colorectal adenomas190. Gastric and head and neck cancers have been associated with

higher risk in individuals with line 1 hypo-methylation191, 192. These techniques also require

pre-PCR processing and age specific control groups, the same problems with complex

results are also likely to affect diagnostic testing role.

2.2.2.1.4 DNA Mutation.

Genetic mutation is detected in colorectal cancer and adenomas in 23-95% of patients 136,

137, 193-195, K-Ras, APC and P53 being the most common. Key mutations in cancers can also

be picked up in plasma, however to date there is no diagnostic combination available and

the only clinical role is for K-Ras directing choice of chemotherapy196.

Page 40: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-40

Similarly to other techniques, mutation analysis has its problems, with only very low copy

numbers of circulating mutant genes being present (<1% in adenomas and early colon

cancers137). The presence of detectable mutations in normal patients who do not develop

cancer on long term follow up also suggests risk rather than diagnosis and limits its role 46.

2.2.2.1.5 Loss of heterozygosity, allelic imbalance, and aneuploidy.

In patients heterozygous at key regulatory gene sites, demonstrable loss of heterozygosity

(LOH) in cancer DNA has been shown with microsatellite markers in both tissue and

circulating DNA138, 184, 197-200.

A single retrospective cfDNA study in colon cancer identified 59% of cases201 and two

studies in oesophageal adenocarcinoma identify 81-96% of cases199, 202. In keeping with

mutation and methylation, testing is likely to reflect increased risk rather than the cancer

process itself.

Single Nucleotide Polymorphisms (SNP) allowing even more accurate mapping of genetic

changes was hoped to offer a breakthrough with very large micro-array studies conducted

extensively in tissue. However, whilst the results in tissue have been impressive, there are

concerns with the feasibility of using this technique on cfDNA 203.

An initially impressive study in ovarian cancer with SNP markers showed allelic imbalance in

95% of cancer patients, and no loss of heterozygosity in the normal population. This looked

like a breakthrough, however surprisingly high DNA levels were also present in the “control”

population raising the possibility of contaminating genomic DNA. This would significantly

affect the loss of heterozygosity analysis in the normal population and call the study result

into question170. With tumour changes not always reflected in blood samples, and other

doubts over methodology and accuracy 154, 193, 204 microsatellite and SNP results must be

interpreted with caution.

Page 41: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-41

Fluorescence In Situ Hybridization (FISH) techniques looking at larger chromosomal

amplification and deletion have shown excellent results in tissue. Results in an oesophageal

study identified HGD and adenocarcinoma with 84% sensitivity and 93% specificity 205.

Circulating markers have yet to be studied because of technical difficulties with DNA

quantity, and the requirement for tumour DNA specificity.

Similarly flow cytometric studies of aneuploidy in Barrett’s oesophagus and oesophageal

adenocarcinoma tissue have shown excellent results, and represent the single best marker

of disease206, 207. However, no study to date has identified these changes in blood or urine,

and it is difficult to see how this would be possible.

2.2.2.1.6 DNA sequencing.

Sequencing offers the most accurate assessment of circulating DNA, with information on

fragment length, copy number, and gene sequence and mutations. Until recently selection

and gene enrichment were required to analyse particular genomic sequences, but new

platforms are able to sequence all circulating DNA with excellent results. This technique

offers the potential to analyse all of the above markers at the same time.

A new study in breast cancer sequenced all DNA in normal and neoplastic groups, with

validation in a second set. The results were startling with a ROC of 0.92 for stage I disease,

and 70% sensitivity with 100% specificity208. Interestingly both Line and Alu DNA elements

were significant, with Line being more significant. Further work to prove a clinical role is

required because of the complex statistics and use of whole genomic pre-amplification,

both of which may produce undetectable biases 209, 210. Repeat analysis by a different

technique such as PCR, against a clinically relevant non-breast cancer group, could confirm

its significance.

Page 42: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-42

2.2.2.2 Mitochondrial DNA studies.

Mitochondrial DNA has been studied less frequently and never in oesophageal and

colorectal cancer. Results are mixed with some studies showing decreased levels

(MATPase8, ND1)173, 211, and others increased212. Mitochondria lack intrinsic DNA repair

enzymes and are therefore sensitive to oxidative damage, especially important in

oesophageal adenocarcinoma pathogenesis. Quantitative mitochondrial DNA testing may

therefore complement nuclear DNA quantification.

Levels are also raised in traumatic conditions and probably inflammatory conditions, which

are likely to confound previous studies106. The choice of patient and control groups is once

again important.

With no previous study of mitochondrial markers in oesophageal and colorectal cancers the

oxidation sensitive mitochondrial ND1 sequence was chosen to complement study of

nuclear DNA markers in this thesis211.

2.3 Circulating RNA tumour markers – transcriptomics.

The origin of circulating cell free RNA (cfRNA) is uncertain but it is likely to arise from

similar processes suggested for cfDNA.

Although damage to DNA is a key step in carcinogenesis not all change leads to cancer. RNA

levels reflecting cellular and genomic activity was hoped to identify the cancer phenotype

bridging the diagnostic gap. Unfortunately, normal levels of RNA expression may only differ

slightly from that seen in cancer cases, and other diseases, making quantification difficult.

With cfRNA levels in the blood even lower, analysis of circulating RNA is challenging.

Furthermore optimal purification and processing techniques have not been agreed, and

interpreting negative studies is therefore difficult. RNA results will require careful

validation in large populations.

Page 43: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-43

2.3.1 RNA marker studies.

Multiple studies using micro-array gene expression analysis in tissue have identified a wide

range of differing genes which may be helpful, but unfortunately without a consistent

picture. A meta-analysis has compared the results across 25 studies and picked out the

more consistent markers which may be helpful in future studies213.

In colorectal cancer a micro-array and RT-PCR study identified a 5 gene marker set using

circulating white blood cell RNA214. Test characteristics in a validating set identified 88% of

cancers correctly. However, only 67% of the normal group were identified, half of the

cancer patients had advanced disease, and an indeterminate group still remained after

analysis. Improvements to the assay will be needed before further clinical evaluation.

A second micro-array and RT-PCR study used a panel of 7 biomarkers and also tested on

circulating white blood cell RNA samples. This was found to have a sensitivity of 72%, and

specificity of 70%215. Most of the subjects (60%) had early cancers, and the control group

had undergone normal colonoscopy making this one of the best and most clinically relevant

colon studies to date. Further testing in a larger screening population is needed.

Telomerase mRNA shows potential, with two small studies in colorectal cancer showing at

best 82% sensitivity at 100% specificity, although against healthy controls216, 217. Further

evidence of potential is shown in a small clinically relevant hepatocellular carcinoma (HCC)

study including chronic liver disease controls, showing 77% sensitivity and 97%

specificity218. All three studies require validation.

Plasma cytokeratin 19 (CK19) a marker for epithelial cells and CEA RNA analysis in cancer

patients showed a sensitivity of 83%,and specificity of 76%219. However, other groups have

not found similar results220 and the study identified advanced cancers from healthy controls,

suggesting these figures may fall in a clinically relevant validating population.

Page 44: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-44

Most recently in abstract form only, an RNA multi-gene signature identified colon cancer

and adenomas in a validating set analysis using RT-PCR with 82-85% sensitivity221.

Unfortunately there is no comment on specificity or stage of disease studied but this may

be useful in future diagnostic work.

In oesophageal cancer there are 3 studies. Circulating tumour cell survivin mRNA (an anti-

apoptosis gene) has been studied in a small and retrospective study looking at response to

chemo-radio therapy in oesophageal squamous and adenocarcinoma. There was only

significance in non-responders, and RNA was only found in advanced disease 222. More

promising results in bladder cancer with urine sampling showed a difference between

normal and bladder cancer patients that increased with grade of tumour. This finding

requires further validation, but shows some potential for future research134, 223.

In oesophageal squamous cancer, plasma carcinoembryonic antigen (CEA) and squamous

cell carcinoma (SCC) antigen mRNA (markers of circulating tumour cells) 224, showed a

prognostic significance when either SCC or CEA was detected. However, these were only

present in 13% of patients, (8% CEA antigen and 4% SCC) and reflect advanced disease225.

These are unlikely to represent diagnostic targets.

2.4 Circulating protein tumour markers- proteomics.

2.4.1 Origins of circulating proteins.

The presence of proteins in plasma is normal, and consists mainly of albumin (55%). Other

classical plasma proteins include coagulation factors, immunoglobulins, peptide hormones,

cytokines, and paracrine messengers.

Page 45: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-45

Abnormal plasma proteins may be derived from cellular death and damage leading to

release of non-plasma proteins which can be highly specific e.g. troponin I in cases of

myocardial infarction. Abnormal secretion patterns in tumour cells may also be a source of

non-plasma proteins, which whilst being much less controversial than the idea of DNA

secretion 226 , have still not currently been identified.

2.4.2 Circulating protein studies

Approximately 289 plasma proteins have been detected, covering concentrations 10 orders

of magnitude different. This number is expected to increase and does not reflect all the

post translational modifications and precursor variations of each protein including

immunoglobulins with 10,000,000 different sequences.

Whilst the complexity of the plasma proteomics is apparent, the study of circulating

proteins also offers great possibilities 226.

Proteins as the functional products of DNA and RNA are involved in all cell growth and

change. They also offer great promise in bridging the diagnostic gap between DNA coding

and cancer phenotype. However, like DNA and RNA there are problems to overcome.

Protein components of the cellular cancer process can be very subtlety different to ‘normal’

cellular proteins quantitatively and structurally. These changes can affect function. There

are no sensitive and specific assays available at present to detect these subtly different

proteins.

Page 46: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-46

Detection of circulating abnormal proteins can be challenging. Their usual position is

primarily within the intra-cellular compartment, and circulating levels are therefore much

lower than other normal circulating proteins. Changing levels of acute phase inflammatory

proteins, clotting proteins and variation with meals and in other disease states further

confound analysis227. Advanced techniques are required to obtain relevant proteins at

suitable concentrations, which may require prior digestion, or depletion and fractioning.

These approaches are not standardised, and generation of a protein assay can be laborious

with small changes difficult to account for. Identifying possible candidate proteins has been

sped up by using high throughput techniques such as matrix-assisted laser desorption

ionization time of flight (MALDI-TOF), surface enhanced laser desorption ionization time of

flight (SELDI-TOF), and mass spectroscopy (MS).

These offer the possibility to screen very complex proteins mixtures such as urine and

plasma, identifying individual proteins and protein signatures within them228. Statistical

analysis of such data is challenging and can be misleading229, with differing signatures

found in the same cancers, and variability between samples not uncommon230-233. Further

ELISA testing is generally required to validate and test individual markers234, 235.

2.4.2.1 Protein signature studies

An impressive study in colorectal cancer patient’s sera identified six peaks using SELDI-TOF,

with subsequent development of a validating ELISA assay in a second blinded population.

Complement C3a-desArg was the best performing marker in both cancers and adenomas

with approximate sensitivity and specificity of 96%236. However, the final diagnostic results

reflected a partial training set and classification did not include lesions that were

indeterminate. Furthermore the study control group only consisted of healthy volunteers.

Page 47: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-47

A second study initially employing mass spectrometry identified five proteins for antibody

development and further testing in serum. Nicotinamide N-methyltransferase and

proteasome activator complex subunit 3, were the best performing markers but only just

improved on CEA237. Comparison between CEA and another putative protein marker

Macrophage migration inhibitory factor(MMIF) 238 using ELISA also showed little

improvement.

There are no oesophageal cancer studies.

2.4.2.2 Circulating Tumour antigen studies

Multiple circulating tumour antigens have been developed including those in current

clinical practice, CEA, CA 19-9, and CA-125.

Originally they were developed by reacting cancer patients sera with mouse B-lymphocytes,

and then harvesting the antibodies to test against other cancer patients sera.

Over time further reactive antigens have been identified and sequenced, although no

tumour antigens have been identified with any clinical diagnostic role. They represent the

oldest239 and largest group studied (see Table 3).

Page 48: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Circulating markers of gastro-intestinal cancers.

2-48

Table 3 Circulating tumour antigens.

Name Properties and research roles CEA CEA can increase with various GI cancers, allowing follow up and assessment of response to

chemotherapy240. Studies use combination or comparison to show improved clinical utility.241-243 CA 19-9 CA 19-9 is a glyco-lipid often raised in pancreatic cancer. Unfortunately it is also expressed in a range

of other cancers, cystic fibrosis and liver disease244, 245. It can be used to follow up patients and assess treatment response51.

CA 50 (sialylated Lewis (a) antigen) CA 50 is an alternative antigen epitope of CA19-9. It is no better and it’s behaviour and use reflects this245.

CA 242 (sialylated Lewis (a) antigen) CA 242 is the final antigen in a triplet against sialylated Lewis (a) antigen, neither is better than the other, but perhaps in combination they add some extra sensitivity in detection of pancreatic cancer.246

CA 125 (Carbohydrate antigen 125, MUC 16)

Ca 125 (MUC16) is a protein associated with ovarian cancer but may be raised in pancreatic and colorectal cancers, with other benign conditions. It has follow up, and treatment response roles247.

CA 72-4 (Tumour Associated Glycoprotein TAG-72)

Ca-72-4 is a tumour associated glycoprotein, with limited specificity for gastric cancer248.

CA 15-3 (MUC 1) Ca15-3 is a glycoprotein with some specificity for breast cancer at an advanced stage249.

CA 27.29 (MUC 1) CA 27.29 is also an epitope of MUC-1. Elevated CA 27.29 levels are primarily associated with metastatic breast cancer and may be useful for predicting recurrent breast cancer. CA 27.29 levels are not useful for screening or diagnosis of malignant disorders250, 251.

CA 11-19 The EDP Biotech company reports that this protein in a test (Colomarker) is able to pick 99% of patients with colorectal cancer from the normal population. There are no published trials of this marker in pubmed.

Page 49: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Chapter 2 – Circulating markers in gastro-intestinal cancers.

2-49

2.4.2.3 Autologous T-cell activating antigen studies

Autologous T-cell activating class I MHC antigens are a new avenue for investigation with new

markers isolated after investigation into melanomas, oesophageal cancer, astrocytoma, and Hodgkin

lymphoma; NY-ESO-1; MAGE; GAGE; BAGE; tyrosinase; SSX2; TRP-I; CDK4; and ı-catenin.

Tissue expression studies in colon252 and oesophageal cancers253 look promising, but studies of

circulating markers are mostly still awaited.

A study looking at a single patient with circulating antibodies to T cell derived antigen NY-ESO-1 in

metastatic adenocarcinoma found higher titres associated with progressive disease254.

In advanced oesophageal cancer serum NY-ESO-Y1antigen identified only 30% of cases but in

combination with a FAS ligand antibody identified 89% of tumours with 100% specificity255. With

poor availability of any markers in oesophageal cancer this may warrant further study.

2.4.2.4 Auto-antibody studies

Detection of serum antibodies to tumour antigens in a reverse ELISA may provide useful circulating

serum markers for cancer diagnosis and prognosis256. These markers are of course the opposite of

protein antigens above, and testing consists of ELISA with bound peptides selectively binding auto-

antibodies from cancer patient’s sera. Secondary analysis allows identification of the individual

antigens.

In colorectal cancer a combination of five auto-antibodies picked by screening peptides produced

from cancer complementary DNA (cDNA) libraries, show a diagnostic sensitivity of 58%, and

specificity of 92.4%. Addition of CEA analysis to these further increases the sensitivity to 77.6%, a

good clinical test. However, the comparator group were healthy volunteers and no validation

exercise has been performed257.

Breast and lung cancer studies have shown more promising results258, 259, perhaps demonstrating the

difficulties in diagnosing colorectal cancer.

Page 50: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Chapter 2 – Circulating markers in gastro-intestinal cancers.

2-50

In a single study of Barrett’s oesophagus, oesophageal squamous and adenocarcinomas, anti-P53

antibodies were detected in plasma. However, the diagnostic yield at 11% in Barrett’s patients, and

20% in oesophageal adenocarcinoma was very low and is unlikely to help diagnostically 260.

2.4.2.5 Miscellaneous protein studies

2.4.2.5.1 MMP - matrix metalloproteinases

A model to detect colorectal cancers and significant polyps, using gender, smoking history, pain, and

age adjusted ELISA MMP9 levels demonstrated a sensitivity of 78%, and specificity of 77% 261.

However, 45% of patients had advanced disease and the control group included 46 healthy

volunteers. These proteins catalyse the dissolution of the extracellular matrix, and perhaps identify

more advanced aggressive disease moving towards an invasion phenotype. They therefore may not

be the best marker for early disease.

MMP7 has been studied in colorectal cancer, but there was only a statistically significant change in

advanced cancers, suggesting it is less suitable for diagnostic work.262

MMP2 has been studied with CEA but has lower serum sensitivity (11.1% vs 27.8% in Dukes A).

Similar to CEA it is useful if high in the primary tumour, associated with worse prognosis and higher

recurrence263-265. Together the two markers were more sensitive, (28-33%) but not significantly

better than current markers264, 266.

2.4.2.5.2 TIMP 1 (Tissue inhibitor of metalloproteinases)

Impressive results with ELISA kits in a large number of patients show promise in early cancer

diagnosis, with 52% sensitivity at 98% specificity 267 and even higher sensitivity in early cancers (56%

in Dukes A and B268). A pre-operative study showed correlation with stage and outcomes, suggesting

a role in post-operative decision making265. However, a lack of positive findings in adenomas, and a

correlation with age269 may limit its usefulness as a screening tool for colorectal cancer prevention.

Page 51: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Chapter 2 – Circulating markers in gastro-intestinal cancers.

2-51

2.4.2.5.3 Tissue Polypeptide antigen - cytokeratins 8, 18 and 19

This is an antibody to epithelial cytokeratin fragments 8, 18, and 19. It has been trialled as a

circulating marker for colorectal cancer but the results have been mixed with positive and negative

results 270, 271.

2.4.2.5.4 Specific Tissue Polypeptide antigen – cytokeratin 18

This is an antibody to epithelial cytokeratin fragment 18. A trial using it as a circulating marker for

colorectal cancer met with little success271 . Another study in lung cancer also demonstrated a low

sensitivity (40%) and use as a diagnostic marker is therefore unlikely272.

2.4.2.5.5 CYFRA 21-1 – cytokeratin 19

A study in oesophageal squamous and adenocarcinoma showed a low sensitivity and specificity273.

Further study of cytokeratins in gastrointestinal cancers is unlikely to add to diagnostic testing, at

least with ELISA.

2.4.2.5.6 Nuclear matrix protein antigens

Nuclear matrix protein markers in cancer patients serum and urine have been described since

1992274, 275, with an FDA approved diagnostic urinary test in bladder cancer276. Colon cancer specific

proteins 2, 3, and 4 are new nuclear matrix markers initially picked using mass spectrometry analysis

and re-analysed with an ELISA against normal, adenomatous, and colon cancer patients serum277-279.

Control groups were healthy volunteers, patients with hyperplastic polyps, and non-advanced

adenomas. Testing for “significant” polyps and colon cancer patients showed the test appears

sensitive with excellent Receiver Operator Characteristics (area under curve 0.82-0.90). This will

require further evaluation in a larger study.

Nucleosomes which consist of nuclear matrix proteins and associated DNA are unfortunately less

sensitive than cfDNA 280, and so further analysis is unlikely to improve diagnostic testing.

Page 52: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Chapter 2 – Circulating markers in gastro-intestinal cancers.

2-52

2.4.2.5.7 VEGF – vascular endothelial growth factor

Studies looking at serum VEGF levels have shown significant differences between cancer and normal

patients281, 282, however in a small study comparing this with circulating DNA, it was less useful.283

This would seem to preclude a diagnostic role for serum VEGF levels, particularly with bigger more

advanced tumours requiring enhanced vascular supply.

2.4.2.5.8 CD 44

CD44 is a transmembrane glycoprotein family with multiple roles, including cell to cell interactions.

The exact role depends on the particular isoform. It has been suggested as a marker of breast and

prostate cancer stem cells284, and changes in cd44 have been associated with tumour metastasis285.

A clinical trial in different intestinal diseases and cancer patients found significant differences from

normal patients286. However, when 3 antibodies were used to detect early colon cancer only 25-50%

of patients were identified, and only 63% of patients with non-metastatic gastric cancer were

identified. It is therefore unlikely to play a major role in diagnostic testing of cancer in the gastro-

intestinal tract. 287.

2.4.2.5.9 Kallikrein

Kallikreins are a subgroup of serine proteases which cleave proteins. Within cancer tissue Kallikrein

hK3 has a very well recognised role as a cancer biomarker, but by an alternative name, PSA.

Physiologically hK1 releases bradykinin from kininogen, and plasmin from plasminogen. The role in

enzyme pathways for other members of this family is not known however they play a prognostic role

in other types of cancer, including colorectal cancer, in both tissue and serum288-292. There have been

no diagnostic studies to date.

Page 53: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Chapter 2 – Circulating markers in gastro-intestinal cancers.

2-53

2.4.2.5.10 Laminin

In colorectal cancer a serum ELISA assay for laminin showed a sensitivity of 89% and a specificity of

88%. Nearly half of the patients had advanced disease, including metastasis and the control group

were healthy controls. Further studies are needed293.

2.4.2.5.11 βHCG - beta human chorionic gonadotrophin

This was trialled as a circulating marker for colorectal cancer with little success271.

2.5 Circulating tumour cells.

Circulating tumour cells offer hope acting as diagnostic, prognostic and therapeutic markers. They

always signify cancer when detected, but are found mainly in late/ advanced disease and in small

quantities, representing a metastatic phase.

They may play an important role in therapeutics and individualised chemotherapy regimens 294-296,

but their use prognostically shows mixed results 219, 297-300. As they generally detect cancer at a late

stage they are unlikely to play a diagnostic role with late disease generally symptomatic and

accurately diagnosed within currently available practice 301, 302.

Alternative techniques using analysis of cytokeratin mRNA in purified blood220, 224, 303 can detect the

presence of circulating cells at an earlier stage. Unfortunately as RNA and not the cell itself some of

the advantages are lost and the later stage of disease remains difficult to surmount diagnostically.

2.6 Circulating DNA markers in colonic and oesophageal adenocarcinoma.

Worldwide there are 27 papers looking at the diagnostic and prognostic application of cfDNA

markers in colonic and oesophageal adenocarcinoma. All are nuclear DNA studies. The papers

report quantitative DNA fragmentation changes, tumour suppressor gene promoter

hypermethylation, patient lymphocyte DNA tumour suppressor gene promoter hypermethylation 304,

oncogene mutations, and loss of heterozygosity at tumour suppressor genes.

Page 54: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Chapter 2 – Circulating markers in gastro-intestinal cancers.

2-54

The main studies in colonic cancer are tabulated in Table 4 and consist of 22 papers in English, 1

paper in Japanese (English abstract, Hibi et.al.305), and 1 in abstract form only (Church et.al.187).

There are 3 papers looking at oesophageal adenocarcinoma, these are tabulated in Table 5.

Most studies involve patients with advanced cancer (stages III-IV), commonly representing half of

the study population. These may not represent patients with curable disease.

There are only 5 studies looking to identify colonic adenomas136, 137, 140, 187, and no studies look to

identify oesophageal dysplasia.

There are only 4 papers with clinically relevant control groups137, 187, 304, 306. Eight papers don’t report

findings in control groups, and fifteen use ‘healthy volunteers’, which may not represent a good

control population. There are no studies of oesophageal adenocarcinoma using clinically relevant

control groups such as patients with risk factors for cancer or other diseases.

These markers hold great promise for future study, but none are used clinically yet, and most don’t

identify early disease.

Page 55: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

Chapter 2 – Circulating markers in gastro-intestinal cancers.

55

Table 4 Colorectal circulating DNA papers.

* = only tested in positive tissue patients. I-IV = staging grade of cancer, early (I) to advanced(IV).

Table 5 Oesophageal circulating DNA papers.

Author Year DNA marker Serum / plasma / urine Control group (n=) Cancer / adenoma (n=) ValidationPrognostic / diagnostic /

therapeuticSensitivity Specificity

Hibi 1998 LOH at 8 microsatellite markers serum N/A I-IV cancers (44) N diagnostic 0% XXX

Anker 1997 K-ras mutation plasma healthy volunteer (6) adv cancer (14) N diagnostic 43% XXX

De Kok 1997 K-ras mutation serum N/A adv cancer (14) N diagnostoc 43% XXX

Kopreski 1997 K-ras mutation serum or plasma healthy volunteer (28) adv cancer (31) N diagnostic 39% XXX

Ryan 2003 K-ras mutation serum N/A I-IV cancers (78) N prognostic 41% XXX

Su 2005 K-ras mutation urine N/A cancer/adenoma (20) N diagnostic 83%* XXX

su 2008 K-ras mutation urine N/A cancer/adenoma (20) N diagnostic 95%* XXX

Hibi 1998 P53 and K-ras mutation serum N/A I-IV cancers (44) N diagnostic 23% XXX

Wang 2004 APC, TP53, K-ras mutation serum healthy volunteer I-IV cancers (104) N diagnsotic 35% XXX

Diehl 2005 APC mutation plasma age matched volunteer (10) I-IV cancer (22) (and polyps(11)) N diagnostic 72% *(39%) XXX

Diehl 2008 APC, TP53, PIK3CA mutation plasma N/A I-IV cancers (25) N diagnostic 50% XXX

Frattinin 2008 K-ras mutation, P16 methylation. plasma healthy volunteers (20) I-IV cancers (70) N diagnostic 25% XXX

Lecomte 2003 K-ras mutation, P16 methylation plasma N/A I-IV cancers (58) N prognostic 64% XXX

Ally 2005 oestrogen receptor methylation leukocytes disease free control group (57) cancer (57) N diagnostic non significant non significant

Grutzman 2008 Sept9 methylation plasma healthy volunteer (183) I-IV cancer (126) Y diagnostic 72% 90%

De Vos 2009 Sept9 methylation plasma healthy control (97) I-IV cancer (172) Y diagnostic 56% 95%

Church 2010 Sept9 methylation plasma ave risk group (930) I-IV cancer and polyps (53) Y diagnostic 50% 91%

Tanzer 2010 Sept9 and ALX4 methylation plasma health volunteer (22) cancer/adenoma (54) Y diagnostic 40% 95%

Holdenrieder 2001 Nucleosome DNA quantification serum healthy and benign disease (172) various adv cancer (418) N therapeutic N/A N/A

Thijssen 2002 pico green DNA quantification serum or plasma age matched volunteer (28) adv cancer (26) N prognostic XXX XXX

Umetani 2006 Alu 115 +247 DNA quantification and ratio serum healthy volunteer (51) adv cancer (136) N diagnostic 63% 92%

Pucciarelli 2009 Alu 115 +247 DNA quantification plasma healthy volunteer (55) I-IV cancer (136) N diagnostic 79% 86%

Flamini 2006 G-3-PD DNA quantification plasma healthy volunteer (75) I-IV cancer (75) N diagnostic 81% 73%

Boni 2007 Rnase P DNA quantification plasma healthy volunteer (67) cancer (67) N diagnostic 100% XXX

Author Year DNA marker Serum / plasma / urine Control group (n=) Cancer / adenoma (n=) ValidationPrognostic / diagnostic /

therapeuticSensitivity Specificity

Kawakami 2000 APC methylation plasma healthy control (20) I-IV cancer (52) N prognostic 25% 100%

Eisenberger 2006 LOH at 12 microsattelite markers serum healthy volunteer (10) cancer (32) N diagnostic 81% XXX

Banki 2008 β-actin plasma healthy volunteer (44) recurrent cancer (42) N prognostic 100% XXX

Page 56: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

56

2.7 Conclusions.

The characteristics of ‘ideal’ diagnostic markers for the early detection of cancer

have been well described, and circulating markers offer many of these features.

A number of research studies into circulating DNA, RNA, and proteins have identified

potential markers which may be useful diagnostically and prognostically. However,

none have been proven clinically suitable to date.

DNA, with well established sensitive analytical techniques offers a pragmatic area

for further research, with the possibility of rapid, inexpensive translation into clinical

practice.

Alu, Line and mitochondrial DNA quantification in plasma offers the opportunity for

an economical and sensitive diagnostic assay for both oesophageal and colorectal

cancers.

Page 57: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

2-57

Experimental Hypothesis and Aims

Hypothesis

This thesis examines the hypothesis that plasma from patients with pre-cancerous, early cancer and

advanced colorectal and oesophago-gastric cancers contains increasing quantities and changing

fragmentation patterns of cfDNA, and that this may have a diagnostic application as part of simple

blood test.

This thesis also examines the hypothesis that plasma cfDNA levels and fragmentation patterns from

patients with pre-cancerous, and early colorectal and oesophago-gastric cancers return to normal

after endoscopic resection.

Aims:

1. To develop assays to determine the quantities of nuclear and mitochondrial cfDNA.

2. To develop assays to determine the fragmentation patterns of nuclear and mitochondrial

cfDNA.

3. To examine the use of nuclear and mitochondrial cfDNA quantities as a potential diagnostic

test in pre-cancerous, and early colorectal and oesophago-gastric cancers.

4. To examine the use of fragmentation patterns in nuclear and mitochondrial cfDNA as a

potential diagnostic test in pre-cancerous, and early colorectal and oesophago-gastric

cancers.

5. To examine the effect of combining cfDNA with carcinoembryonic antigen as a potential

diagnostic test in pre-cancerous, and early colorectal cancers.

Page 58: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-58

Chapter 3 Methods.

3.1 Introduction.

Previous studies using serum with Alu and Line 1 markers, purified cfDNA fragments >100bp long

and showed significant discrimination between healthy volunteers and cancer patients.

This research project utilised plasma to quantify cfDNA fragments >20bp with Alu, Line 1, and

mitochondrial markers.

Plasma processing however requires further technical choices with no accepted standard for DNA

sampling, processing time, separation, or measurement 152, 154, 307. The available techniques are

discussed below with the reasons for the subsequent protocols described. The full laboratory

protocols are documented at the end of the chapter.

3.1.1 Plasma processing.

Studies show changes in DNA levels and fragmentation when plasma and serum are left for greater

than 2-4 hours before processing. Processing within this period is recommended153, 308. No

differences in quantity or integrity were seen between samples stored at 4°C or room temperature

within this period.

A double centrifuge protocol separating plasma from whole blood has shown improved

reproducibility. Additional cell separation techniques such as histopaque and filtration are probably

not helpful153, 309, 310. Our isolation protocol processed samples within 4 h, utilising a triple spin

technique with histopaque separation. This followed our standardised laboratory protocol, chosen

to reduce cellular contamination, and pre-dating the above research.

3.1.2 DNA isolation and purification.

Common DNA isolation techniques include: binding of DNA to silica membranes followed by alcohol

washing; phenol and sodium acetate washing and precipitation with cold ethanol or iso-propanol

followed by spinning down and drying.

Page 59: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-59

Both methods re-suspend DNA with addition of an aqueous solution. A variation on the silica

membrane technique is to use microscopic silica coated magnetic beads e.g. Dynabeads. These are

mixed with the DNA and body fluid solution outside of a magnetic field and then separated from the

suspensions by attraction of the beads to a magnetic field. The supernatant fluid minus DNA and

beads is removed by pipette. The technique uses the same chaotropic salts and ethanol washes, but

is quicker, much easier to mechanise and can be up-scaled for larger use. It does not use centrifuges

or filters.

Most papers in the medical literature use silica membrane spin column techniques 108, 139, 147, 154, 167.

They offer highly standardized tissue specific procedures, quick and easy use, few toxic or harsh

chemicals, minimise contaminating inhibitors, and are used in NHS laboratories311-313. DNA isolation

relies on the formation of hydrogen bonds between the DNA and the silica membranes in the

presence of high concentrations of chaotropic salts, and relative dehydration. The DNA containing

solution is spun through silica membrane columns which bind the DNA when the conditions above

are in place. The chaotropic salts are then removed with an alcohol-based wash and spin. The DNA is

finally eluted after addition of a low-ionic-strength aqueous solution such as TE (10mM Tris-HCl, 0.1

mM EDTA) buffer or water with a final spin down into a collecting tube.

Newer columns have been developed for purifying small DNA fragments (<100 bp) thought to

contain more tumour DNA. Plasma-XS spin columns (Macherey-Nagel) economically purify

fragments >20 bp long, and are used in this study.

3.1.2.1 High Yield Plasma-XS nucleospin modification.

The spin column manufacturer recommends that up to740 µl of plasma can be loaded onto the spin-

column without compromising quantification. With our low concentration samples, it was desirable

to process larger volumes of plasma. A preliminary study showed that up to 960 μL of our plasma (2x

480 μL aliquots with 2 x 720 μL of binding buffer, loaded in 4 x 600 μL aliquots) can be loaded

without losing DNA (Figure 4).

Page 60: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-60

Figure 4 Modified DNA loading results.

3.1.3 DNA quantification.

Quantitative study of purified cfDNA has been measured in three main ways: absorbance (Ultra

Violet (UV) spectrophotometry)314; fluorescent DNA binding dyes (e.g. ethidium bromide, Pico

green)306; and quantitative PCR reaction101, 149, 315.

UV spectrophotometry is the easiest and most common technique for measuring DNA generally.

DNA and most of the common contaminants found in DNA preparations absorb UV light and

measurements between the 220 nm and 320nm region and are used to determine concentration

and assess purity. Measurement at 260 nm quantifies DNA; 230 nm guanidium salts and phenol

(process contaminants); 280 nm tyrosine and tryptophan (used as an indicator of protein

contamination); and 320 nm turbidity of the sample which is normally subtracted after a background

reading. These separate absorbance readings allow measurement of the DNA concentration and

provide information about the contaminant levels.

0 0.005

0.01 0.015

0.02 0.025

0.03 0.035

0.04 0.045

Sam

ple

1.1

(4

80

μl)

Sam

ple

1.2

(9

60

μl)

Sam

ple

1.3

(1

44

l)

Sam

ple

2.1

(4

80

μl)

Sam

ple

2.2

(9

60

μl)

Sam

ple

2.3

(1

44

l)

Sam

ple

3.1

(4

80

μl)

Sam

ple

3.2

(9

60

μl)

Sam

ple

3.3

(1

44

l)

Sam

ple

4.1

(4

80

μl)

Sam

ple

4.2

(9

60

μl)

Sam

ple

4.3

(1

44

l)

Sam

ple

5.1

(4

80

μl)

Sam

ple

5.2

(9

60

μl)

Sam

ple

5.3

(1

44

l)

Sam

ple

6.1

(4

80

μl)

Sam

ple

6.2

(9

60

μl)

Sam

ple

6.3

(1

44

l)

Sam

ple

7.1

(4

80

μl)

Sam

ple

7.2

(9

60

μl)

Sam

ple

7.3

(1

44

l)

Sam

ple

8.1

(4

80

μl)

Sam

ple

8.2

(9

60

μl)

Sam

ple

8.3

(1

44

l)

Sam

ple

9.1

(4

80

μl)

Sam

ple

9.2

(9

60

μl)

Sam

ple

9.3

(1

44

l)

Sam

ple

10

.1 (

48

l)

Sam

ple

10

.2 (

96

l)

Sam

ple

10

.3 (

14

40

μl)

1 2 3 4 5 6 7 8 9 10

Me

an q

uan

tity

of

DN

A (

ng/μ

l)

Modified protocol - total DNA yield

Page 61: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-61

Absorbance value at 260 nm (A260) is used to calculate DNA concentration with an equation derived

from Beer’s Law:

Concentration (µg/mL) = (A260 reading – A320 reading) x dilution factor x 50 µg/ml.

A260 of 1.0 = 50 µg/mL pure DNA.

Proteins and guanidium salts do contribute to absorbance at A260, therefore if they are present at

high levels in low concentration DNA preparations they will lead to an overestimation of the DNA

concentration312. A good quality DNA sample should have an A260/A280 ratio of 1.7-2.0 and an

A260/A230 ratio of greater than 1.5.

The nanodrop spectrophotometer 1000 (Thermoscientific) is accurate (± 10-15%) when samples are

greater than 10 ng/μL, and is unsuitable for the purified DNA samples we are using at <3 ng/μL.

There is also no information about relative quantities of specific genes or fragmentation patterns

offering a potentially more sensitive test. Therefore this method was not chosen for the main study.

Quantitative fluorescent DNA dyes e.g. ethidium bromide, fluoresce markedly when bound to

double stranded DNA. Isolated DNA is usually loaded onto an agarose gel and separated according to

size and charge by exposure to an electric field. The negative charge associated with the DNA

backbone causes migration towards the anode. The dye is then applied to the gel and the DNA

concentration is calculated by a fluorometer comparing fluorescent intensity to that of a known

concentration and molecular weight marker band. This technique can provide information on

specific genes, fragment size and DNA quantity, however DNA content less than 10 ng is very difficult

to measure on agarose gels. Therefore this technique is also not suitable for our purposes.

Page 62: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-62

Pico Green is a newer double-stranded DNA binding fluorescent dye. It works in solution comparing

the unknown DNA to a prepared standard curve of appropriate DNA and is more sensitive, able to

detect pg of DNA. However, 200 bp fragments are required for successful quantitation316; genomic

and fragment DNA each require their own standard curves; there is no information about relative

quantities of specific genes; and fragmentation pattern analysis is not possible. This technique is also

not suitable for our purposes.

The last and most recent technique uses quantitative real time PCR (qPCR) a development from

standard PCR. It allows the measurement of the target sequence PCR product as it replicates by the

generation of a DNA bound fluorescent signal. The fluorescence signal increases with each

amplification cycle. The PCR cycle number at which the exponentially rising fluorescence first

increases significantly above the background, is called the threshold cycle (Ct). This provides an

inverse quantitative relationship to the original DNA concentration of the amplicon.

Absolute qPCR for DNA quantitation is well established in the forensic science community, as well as

the medical research community317. Research has shown it to be highly accurate and reproducible

with blood and plasma, even at low picogram DNA concentrations149, 318. Absolute qPCR also relies

on a standard curve methodology, but is not reliant on similar fragmentation size.

With the use of differing size amplicons either side of the “apoptotic fragment” size e.g. 79 bp and

300 bp, this technique allows highly accurate and reliable quantitative and fragment analysis of both

nuclear and mitochondrial DNA. This technique was chosen for our analysis and has previously been

used with Alu, Line and mitochondrial markers.

Whichever methods are chosen caution must be used when comparing yields. Contaminants,

accuracy, and reaction efficiency differences may cause variable measurements and results.

Page 63: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-63

3.1.4 Absolute qPCR.

3.1.4.1 Primer selection and optimization.

A sufficient magnesium (Mg2+) concentration is an essential component required for DNA

polymerase function, however too much Mg2+ can cause non-specific amplification. Primer

concentration must also be at a level that allows optimal efficiency of the reaction, but not so high

that non-specific annealing and primer-primer interactions occur (primer-dimers). It is therefore

essential to optimise these two factors specifically for each target.

The primers were tested for optimal conditions initially with 3 master mixes, mix A std

manufacturers double strength master mix, mix B = + 1 mM MgCl2, and mix C = + 2 mM MgCl2.

Forward and reverse primers were tested at concentrations of 100 - 400 mM in a grid formation. A

positive control to confirm PCR reaction, and a 400/400 mM concentration negative control was

added to rule out primer-dimer artefacts (Table 6.). Preliminary Ct results for Line 1 79 bp are shown

in Table 7. The concentration of 400 mM forward and reverse primers without additional

magnesium was chosen for efficiency and ease of preparation.

Table 6 Optimization experimental layout.

1 2 3 4 5 6 7 8 9 10 11 12

A MM A 15ul

MM A 15ul

MM A 15ul

MM B 15ul

MM B 15ul

MM B 15ul

MM C 15ul

MM C 15ul

MM C 15ul

F1 5ul F1 5ul F1 5ul F1 5ul F1 5ul F1 5ul F1 5ul F1 5ul F1 5ul R1 5ul R2 5ul R4 5ul R1 5ul R2 5ul R4 5ul R1 5ul R2 5ul R4 5ul

B MM A 15ul

MM A 15ul

MM A 15ul

MM B 15ul

MM B 15ul

MM B 15ul

MM C 15ul

MM C 15ul

MM C 15ul

MM B+ve 13ul

MM B-ve 13ul

F2 5ul F2 5ul F2 5ul F2 5ul F2 5ul F2 5ul F2 5ul F2 5ul F2 5ul

F2 5ul F4 5ul R1 5ul R2 5ul R4 5ul R1 5ul R2 5ul R4 5ul R1 5ul R2 5ul R4 5ul

R2 5ul R4 5ul

2ul DNA 2ul water

C MM A 15ul

MM A 15ul

MM A 15ul

MM B 15ul

MM B 15ul

MM B 15ul

MM C 15ul

MM C 15ul

MM C 15ul

F4 5ul F4 5ul F4 5ul F4 5ul F4 5ul F4 5ul F4 5ul F4 5ul F4 5ul R1 5ul R2 5ul R4 5ul R1 5ul R2 5ul R4 5ul R1 5ul R2 5ul R4 5ul

Page 64: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-64

Table 7 Optimization experimental results for line 1 79bp primer.

Well Identifier Ct Well Identifier Ct Well Identifier Ct Well Identifier Ct

A01 MMA 100/100 22.4 A05 MMB 100/100 20.7 A07 MMC 100/100 21.3

A02 MMA 100/200 20.6 A04 MMB 100/200 N/A A08 MMC 100/200 20.8 B11 MMB 200/200 18.6

A03 MMA 100/400 20.9 A06 MMB 100/400 20.5 A09 MMC 100/400 20.6

+ve control

B01 MMA 200/100 19.4 B04 MMB 200/100 19.4 B07 MMC 200/100 N/A B12 MMB 400/400 N/A

B02 MMA 200/200 19 B05 MMB 200/200 N/A B08 MMC 200/200 19.6

-ve control

B03 MMA 200/400 18.9 B06 MMB 200/400 18.9 B09 MMC 200/400 19.6

C01 MMA 400/100 19.3 C04 MMB 400/100 19.8 C07 MMC 400/100 20.5

C02 MMA 400/200 18.8 C05 MMB 400/200 19.6 C08 MMC 400/200 19.6

C03 MMA 400/400 18.7 C06 MMB 400/400 19 C09 MMC 400/400 20

Melt curves to look for non-specific amplification and evidence of primer-dimers were analysed, e.g.

Figure 5.

Figure 5 Melt curve for Line 1 79 bp 400/400 primer amplification.

Standard curves were tested in the optimised PCR conditions to ensure appropriate efficiency, range

and correlation (R2). R2 Values greater than 0.995 were achieved confirming accuracy of

experimental standard curve in each case.

Page 65: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-65

Figure 6 Line 1 79 bp standard curve.

R2 = 0.998.

.

3.1.4.2 Amplicons and cycling conditions.

The 5 previously described amplicons are detailed in Table 8.

Table 8 PCR amplicons.

The plate setup for all amplicons and experiments is shown in Table 9.

Name Sequence 5’-3’ (forward and reverse primer) Product length [Primer]nM Source

5'-CCTGAGGTCAGGAGTTCGAG-3' 200

5'-CCCGAGTAGCTGGGATTACA-3' 200

5'-GTGGCTCACGCCTGTAATC-3' 200

5'-CAGGCTGGAGTGCAGTGG-3' 200

5’-AGGGACATGGATGAAATTGG-3' 400

5’-TGAGAATATGCGGTGTTTGG-3' 400

5'-ACACCTATTCCAAAATTGACCAC-3' 400

5'-TTCCCTCTACACACTGCTTTGA-3' 400

5'-CACCCAAGAACAGGGTTTGT-3' 400

5'-TGGCCATGGGATTGTTGTTAA-3' 400Lin, C. S.Interact Cardiovasc Thorac Surg, 2008

Diehl, F. Nat Med 2008

297bp Sunami, E Ann N Y Acad Sci 2008

79bp

108bp

Umetani, N. Clin Chem 2006

247 Umetani, N. Clin Chem 2006

115Alu 115

Alu 247

Line 1 79bp

Line 1 300bp

Mitochondrial ND1

Page 66: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-66

Table 9 Plate layout for all experiments.

1 2 3 4 5 6 7 8 9 10 11 12

A NTC NTC NTC STD 1 STD 1 STD 1 STD 2 STD 2 STD 2 STD 3 STD 3 STD 3

B STD 4 STD 4 STD 4 STD 5 STD 5 STD 5

SAMPLE

1

SAMPLE

1

SAMPLE

1

SAMPLE

2

SAMPLE

2

SAMPLE

2

C

SAMPLE

3

SAMPLE

3

SAMPLE

3

SAMPLE

4

SAMPLE

4

SAMPLE

4

SAMPLE

5

SAMPLE

5

SAMPLE

5

SAMPLE

6

SAMPLE

6

SAMPLE

6

D

SAMPLE

7

SAMPLE

7

SAMPLE

7

SAMPLE

8

SAMPLE

8

SAMPLE

8

SAMPLE

9

SAMPLE

9

SAMPLE

9

SAMPLE

10

SAMPLE

10

SAMPLE

10

E

SAMPLE

11

SAMPLE

11

SAMPLE

11

SAMPLE

12

SAMPLE

12

SAMPLE

12

SAMPLE

13

SAMPLE

13

SAMPLE

13

SAMPLE

14

SAMPLE

14

SAMPLE

14

F

SAMPLE

15

SAMPLE

15

SAMPLE

15

SAMPLE

16

SAMPLE

16

SAMPLE

16

SAMPLE

17

SAMPLE

17

SAMPLE

17

SAMPLE

18

SAMPLE

18

SAMPLE

18

G

SAMPLE

19

SAMPLE

19

SAMPLE

19

SAMPLE

20

SAMPLE

20

SAMPLE

20

SAMPLE

21

SAMPLE

21

SAMPLE

21

SAMPLE

22

SAMPLE

22

SAMPLE

22

H

SAMPLE

23

SAMPLE

23

SAMPLE

23

SAMPLE

24

SAMPLE

24

SAMPLE

24

SAMPLE

25

SAMPLE

25

SAMPLE

25

SAMPLE

26

SAMPLE

26

SAMPLE

26

After experimental optimization of primer concentrations, magnesium concentration and thermal

cycling conditions, qRT-PCR was carried out as described in Table 10.

Table 10 PCR cycling conditions for each amplicon.

Name PCR conditions

Alu 115

Alu 247

Line 1 79bp

Line 1 300bp

Mitochondrial ND1

50⁰C 2 mins, 95⁰C 10mins (30 cycles of: 95⁰C 15s, 51⁰C 30s, 57⁰C 45s).

50⁰C 2 mins, 95⁰C 10mins, (30 cycles of: 95⁰C 15s, 60⁰C 1min).

94⁰C 2 min, (2 cycles of 94⁰C 20s, 67⁰C 30s, 70⁰C 45s), (2 cycles of: 94⁰C 20s, 64⁰C 30s, 70⁰C 45s),

(2 cycles of: 94⁰C 20s, 61⁰C 30s, 70⁰C 45s), (30 cycles of: 95⁰C 15s, 59⁰C 30s, 60⁰C 45s).

50⁰C 2mins 95⁰C 10mins, (30 cycles 95⁰C 15s, 57⁰C 1min).

94⁰C 2 mins, (3 cycles of : 94⁰C 20s, 68⁰C 30s, 72⁰C 30s), (3 cycles of: 94⁰C 20s, 67⁰C 30s, 72⁰C 30s),

(3 cycles of : 94⁰C 20s, 66⁰C 30s, 72⁰C 30s), (30 cycles of: 94⁰C 20s, 64⁰C 30s, 72⁰C 30s).

Page 67: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-67

Each run used either 10 minutes at 95°C (to activate the DNA Taq polymerase), or 2 minutes at 94°C

and 6-9 cycles of a modified touchdown PCR, before 30 amplification cycles. Each PCR cycle

comprised denaturing, annealing, and extension. Fluorescence measurements were automatically

collected at the end of each annealing phase, with baseline measurements collected during cycles 3-

15. At the end of each run a final melt curve cycle was performed to exclude the presence of primer-

dimer artefacts.

The line 1 79 bp and Alu 115 bp fragments have both been used to quantify circulating DNA levels

before137, 149. With the Plasma XS nucleospin collecting DNA fragments > 20bp, the Line 1 79 bp

fragment is referred to as “Total DNA” reflecting the additional <100 bp fragments being collected

and measured with this technique for the first time. Alu 115 is used as a comparator and as part of

the Alu 115/247 bp fragment short : long ratio comparison.

3.1.4.3 DNA quantity calculation.

The qPCR analyser used for all experiments was the AB7500 (Applied Biosystems inc. California).

Manufacturer default analysis settings were retained. The Ct value and standard curves were

calculated automatically on the 7500 software.

To maximise comparative results across all samples, mean values for standard curve intercept and

slope were calculated from the nine experimental runs (Table 11).

Table 11 Mean standard curve values for each primer.

Line 1 79 Line 1 300 Alu 115 Alu 247 Mito

Slope CT slope CT slope CT slope CT slope CT

-3.40 14.77 -3.57 15.27 -3.49 16.78 -4.56 11.34 -3.50 18.92

0.16 0.53 0.09 0.63 0.12 1.08 0.19 0.62 0.15 1.00

Page 68: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-68

These were then used to manually calculate quantities for each individual sample against a single

standard curve for each amplicon following the AB7500 manufacturer calculation below. The

standard linear regression equation Y = mX +c was used.

Ct = m [log (Qty)] + c

The slope of the line is m, X is log (Qty), c is the y intercept, and Y is the Ct value.

Rearranging the equation to solve for DNA quantity:

Qty = 10

Standard deviation for each triplicate sample quantity was calculated according to:

SD =

X = sample value, x~ = the sample mean, and n = number of samples.

Page 69: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-69

3.1.4.4 DNA dilution correction factor.

Previously purified high concentration healthy volunteer lymphocyte DNA was measured in triplicate

using a nanodrop 1000 spectrophotometer following the manufacturer protocol. The mean value

was 140.79 ng/μL. This sample was diluted 1:29 and used to make a 4.69 ng/μL standard curve

solution. This was aliquoted into 10µl samples and stored at -20°C just prior to starting the

experiments.

For the experimental standard curves this solution was further diluted 3:85, and then 1:3, and then

lastly 1:4. The calculated value of 0.1998 ng when compared to the standard curve value used (0.25)

requires application of a 0.79996 correction factor to the DNA quantity calculated by the AB7500

data.

A final dilution factor was calculated incorporating the above standard curve correction. This allows

conversion of the dilute DNA quantity calculated by the AB7500 into an accurate DNA concentration

for each mL of whole plasma.

This factor = 2.083 (correction to millilitres of whole plasma) *40 (1:40 dilution) * 23 (DNA

purification dilution) * 0.79666 (STD curve quantity correction) = 1533.01.

3.1.4.5 PCR assay co-efficient of variation results.

Coefficient of variation % is a measure of assay accuracy, and gives a measure of reliability for the

results.

It was calculated for each sample triplicate by:

The mean of the samples coefficient of variation is also known as the plate inter-assay coefficient of

variation.

Page 70: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-70

To calculate the intra-assay coefficient of variation identical mid-point standard curve triplicate CT

values across all plates were used to calculate DNA quantities as above. These values were then used

in the coefficient of variation equation to calculate the intra-assay coefficient of variation (Table 12).

Table 12 Intra-assay coefficient of variation experimental results.

Line 1 79 Line 1300 Alu 115 Alu247 Mito

inter-assay 5.59% 12.80% 13.76% 7.98% 14.83%

Range 0.26-26.05% 0.86-29.34% 0.76-76.33% 0.38-17.59% 0.01-38.48%

intra-assay 8.60% 9.19% 11.97% 6.23% 10.98%

3.1.5 CEA analysis.

CEA as the only circulating marker with any diagnostic role in gastrointestinal cancers makes an

interesting diagnostic comparator51. Testing for CEA in the colorectal neoplasia populations was

therefore undertaken.

CEA analysis was performed using a Beckman Coulter Unicel DXL 800 machine (CEA2 assay) with a

CEA2 kit (CEA Access, Beckman Coulter ref 33200). This utilised an ELISA assay with Lumi-Phos 530

(Lumigen PPD (4-methoxy-4-(3-phosphatephenyl)spiro[1,2-dioxetane-3,2’-adamantane], disodium

salt) as the fluorescent marker, and alkaline phosphatase as the linked enzyme.

The assay contains two mouse anti-bodies (MAb) with differing epitopes to the CEA protein, one

linked to a magnetic bead, and the other to alkaline phosphatase. The plasma (35 μL) is aspirated

from the sample placed in the machine, and incubated together with the MAb before washing and

separating with a magnetic field. Lumi-Phos 530 substrate is added to the separated magnetic beads

solution, and a fluorescent reading at 530 nm taken. A stored multipoint callibration curve (0, 1, 10,

100, 500 and 1000 ng/mL CEA, re-calibrated every 28 days), and daily quality control testing, allow

accurate “random access” quantification of the CEA level from 0.1 – 1000 ng/mL with 95%

confidence.

Page 71: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-71

3.1.6 Patient choice, pathological grading and disease staging.

Normal patients had undergone normal upper and/or lower endoscopy without biopsies that were

otherwise well. This represented a population with symptomatic gastro-intestinal disease that were

otherwise well.

Barrett’s oesophagus surveillance patients were included in the group when controlling the

oesophageal neoplasia study.

Patients with biopsy proven colorectal or oesophageal neoplasia undergoing endoscopic or surgical

resection had plasma samples taken pre-procedure. Oesophageal cases planned for neo-adjuvant or

palliative chemotherapy had plasma samples taken prior to commencing treatment.

Paired samples were taken from previously consented patients post-resection, when the neoplasia

was thought to have been resected back to normal. The average follow up period prior to sampling

was 330 days in colon cases (range 85-753 days) and in oesophageal cases 276 days (range 81-480

days. All paired samples were endoscopically or biopsy proven to be normal or incompletely

resected.

Radiological computed tomography staging was performed by three experienced gastro-intestinal

radiologists supplemented with endoscopic, laparoscopic and positron emission tomography (PET)

scanning results. It was the multi-disciplinary team accepted staging. Pathological staging and

grading was performed by three experienced gastro-intestinal pathologists, who reported in tandem

for endoscopically resected specimens. The pathological report of the resected specimen was used

as the definitive stage and grade. The staging system used for all cases was the Tumour Node

Metastasis (TNM) system.

3.1.7 Statistical analysis.

SPSS version 19.0 was used for all statistical analysis.

Mean and median values were calculated for all variables.

Page 72: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-72

The population values were analysed for normality of distribution graphically and with a Shapiro-

Wilk test, and this was not found. Therefore statistical tests for non-parametric data were used.

To test the statistical significance of the un-paired normal and neoplasia populations the Mann-

Whitney U test was used.

Using multiple markers increases the probability of statistically significant findings occurring by

chance. To correct for this a Bonferroni correction factor of 10 was applied raising the significant cut-

off to P= 0.005.

To evaluate the possibility of diagnostic testing with these markers logistic regression was

undertaken with receiver operating curves drawn to demonstrate the tests diagnostic possibilities.

Logistic regression predicts a dichotomous categorical variable from a set of predictor variables. It

does not require a normally distributed data set. The predicted dependent variable is a function of

the probability that a particular subject will be in one of the categories, in this thesis that the patient

has neoplasia. Logistic regression calculates the probability that a specific point is a true member of

a population by regression of the data to a best fit linear relationship. Multiple markers can be

analysed and used to improve the model. This analysis uses the default SPSS logistic regression cut-

off, predicting at a probability of ≥ 0.5.

Receiver operating curves (ROC) curves are used to scrutinise diagnostic probability. They plot the

false positive rate on the X axis and 1 - the false negative rate on the Y axis, and show the trade-off

between the two. Effectively this plots sensitivity against specificity at every single cut off. The area

under the curve measures the discrimination, or the ability of the test to correctly classify those with

and without the disease. ROC values 0.90 - 1.0 represent an excellent test, 0.80 - 0.90 a good test,

0.70 - 0.80 a fair test, 0.60 - 0.70 a poor test, and 0.50 - 0.60 a failed test.

Page 73: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-73

3.2 Experimental protocols.

All methods were carried out in accordance with the control of substances hazardous to health

(COSHH) regulations, following good laboratory practice. All blood and DNA processing followed

universal precautions, and were carried out in cleaned class II biological safety cabinets.

All patients were consented for the study following ethical committee approved protocols and

national guidelines. Patient information leaflets were supplied.

3.2.1 Venepuncture.

Venous blood was taken from a peripheral vein after application of a tourniquet using a 20 millilitre

(mL) BD Plastipak syringe and a 21 gauge BD needle. Blood was subsequently inserted into a 4.5 mL

EDTA BD vacutainer. Blood was stored at room temperature for 2-3 hours maximally prior to

processing.

3.2.2 Cell and plasma separation.

A 5 mL aliquot of blood was added to 5 mL of Hanks balanced salt solution (GIBCO 14175, –MgCl2, -

CaCl2. [Potassium Chloride (KCl 5.33mM), Potassium Phosphate monobasic (KH2PO4 0.441mM),

Sodium Bicarbonate (NaHCO3 4.17mM), Sodium Chloride (NaCl 137.93mM, Sodium Phosphate

dibasic (Na2HPO4 anhydrous 0.338mM), D-Glucose (Dextrose) 5.56mM])

in a 30mL polypropylene universal container and inverted 3 times to mix. The mixture was layered

carefully over 10 mL of histopaque 1077 (Sigma Aldrich 10771) in a new 30 mL polypropylene

universal container using a sterile 3 mL polypropylene Pasteur pipette before centrifuging at 600g

for 30 minutes (1830rpm – Harrier 15/80 MSE centrifuge MSB080.CX 1.5).

Page 74: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-74

After centrifuging the blood had been separated into 4 layers: half strength plasma (“plasma”) top

layer; white cells (“buffy coat”) layer; Histopaque 1077 layer; and bottom red cell layer. The top

“plasma” layer was carefully removed from the underlying “buffy” coat layer with a new sterile 3 mL

polypropylene Pasteur pipette, watching for any disturbance to the “buffy” coat layer. The “plasma”

was re-centrifuged at 400g (Harrier 15/80 MSE centrifuge MSB080.CX 1.5) for 7 minutes, before

removal of the top part of the “plasma” using a new sterile 3mL polypropylene Pasteur pipette

without disturbing the bottom pellet.

The “plasma” was stored in a 2 mL nuclease-free polypropylene container at -80°C (Sanyo Ultralow

VIP series -86), before batch processing at the end of the collection period.

3.2.3 Nucleospin DNA purification.

Prior to processing “plasma” was re-centrifuged at 11,050g (Eppendorf microfuge) for 3 minutes in a

final manufacturer recommended step to remove all contaminating cellular debris.

Two aliquots of 480 μL of “plasma” were mixed (inverting 3 times, and briefly vortexed for 3 s with

720 μL of binding buffer in two 1.7 mL nuclease-free polypropylene micro-centrifuge tubes, and

spun down briefly. 600 µl of the mixture was aliquoted onto a nucleospin column before spinning

at 2000g for 30 s, and 5 s at 11,000g (Eppendorf microfuge). The flow through was discarded. The

spin column was re-loaded until all 2400 μL was processed.

The spin column was put into a new collection tube and 500 μL of washing buffer was added, prior

to spinning at 11,050g for 30 s. The spin column was put into another new collecting tube and 250

μL of washing buffer was added, prior to spinning at 11,050g for 3 minutes. Finally the spin column

was put into a 1.7 mL nuclease-free micro-centrifuge tube, and 30 μL of elution buffer added to the

spin column, prior to spinning at 11,050g for 30 s.

Page 75: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-75

Figure 7 Plasma XS spin protocol.

1. Processed Plasma 2. Repeated plasma spins 3. Elution and wash spins 4.PurifedDNA

Excess ethanol was driven off from the eluted purified DNA sample by heating at 90°C for 8 minutes

in a class II cabinet, without laminar airflow.

DNA was stored at -20°C (Liebherr Pro line freezer), in the micro-centrifuge tubes, defrosting only

once for processing. A 1 in 40 dilution of the DNA samples was used to maximise residual DNA

available for further experimentation.

3.2.4 qPCR.

All pipette tips and plastic-ware was certified DNase- and RNase-free by the manufacturer. All

solutions were carefully mixed by vortexing or flicking before pipetting.

In the clean “reagent only” laboratory and class II biological hood, a 10 μM solution of forward and

reverse primers was made for each amplicon (100 μM stock solution). The working solutions were

stored at 4o for up to 2 weeks at which point they were discarded and fresh working primers were

made.

Five master mixes were made containing the following: AB X 2 SYBR Green Master mix™(Applied

Biosystems; S4438- Sigma, UK), 12.5 μL per well; forward and reverse primers (1μL per well of 10

μM primer for reactions using 400 nM concentrations, and 0.5 μL per well for 200 nM

concentrations); and DNase-free water to a total volume of 20 μL per well. The master mixes were

pipetted into each 96 well plate, checked and sealed.

Page 76: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

3-76

In a separate “template only” laboratory and class II biological hood, 5 μL of 1:40 diluted template

was added to each reaction well, making a total of 25 μL in each well .

Control wells were incorporated into the plate design; instead of template, these contained 5 μL

water and 20 μL of master mix only (NTC - no template controls).

A 5 point 1:4 dilution standard curve was constructed using a standardised, batched and previously

prepared aliquot of genomic DNA, with known concentration. This was pipetted in triplicate

sequentially across the plate.

All five amplicon plates were made up at the same time using the same genomic DNA sample for

each standard curve. The plates were stored at 4°C prior to sequential analysis on the ABI 7500

machine.

Continual melt curve fluorescence analysis was carried out by cooling to 60°C, and raising the

temperature to 95°C with a 1% ramp rate.

3.2.5 CEA analysis.

Plasma samples were thawed and mixed thoroughly before being placed in a 35 μL Coulter

autosampler and aspirated. Daily laboratory quality controls checks were confirmed and results

printed off.

Page 77: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-77

Chapter 4 Colon results.

4.1 Demographics.

The study population comprised 85 patients, including 35 patients without endoscopic abnormality,

a group of 26 patients with benign colorectal adenomas, and 24 patients with colorectal carcinomas.

The reference ‘normal’ group of 35 patients, included 15 men, and was chosen as representing a

typical population attending for endoscopic investigation but with no detectable abnormality (Table

13). The mean age of this group was 54.1 years (range 24 - 80 years). Significant intercurrent

diagnoses in this group included: osteoarthritis; ischaemic heart disease; pernicious anaemia; and

peptic ulcer.

Table 13 Normal population indication for endoscopy.

Endoscopy indication Patient

numbers Dysphagia 5

Dyspepsia 10

Ulcer follow up 1

Haematemesis 2

Abdominal pain 8

Change in bowel habit 7

Family history of cancer 1

Anaemia 5

Polyp follow up 1

Melaena 1

Page 78: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-78

4.2 Patient groups.

The benign colorectal polyp group of 26 patients, included 14 men, and had a mean age of 70.2

years, (range 56 - 85 years).

Twenty-nine polyps were removed from these patients, mean size 54.3 mm (range 5 - 200 mm).

Eighteen of these polyps showed low grade dysplasia (LGD), and had a mean size of 56.1 mm (range

5 - 200 mm), while 11 polyps showed high grade dysplasia (HGD) mean size 52.5mm (range 28 - 100

mm). Significant residual polyp remained in 3 patients, and a further blood sample taken from these

patients is included in the analysis.

The group of cancer patients included 19 men and had a mean age of 71.5 years, (range 49 - 87

years). There were four patients with polyp cancers, mean size 41.7 mm (range 25 - 40 mm), but the

majority had biopsy proven colorectal carcinomas, pathological TNM T staging (pT) : pT0 x 5 (4

polyps, 1 post radiotherapy); pT2 x 4; pT3 x 14; pT4 x 1.

In total, both polyp and cancer populations include 50 patients, and 53 plasma samples, with a mean

age of 71.1 years, (49 - 87 years).

4.3 Mean and median marker values.

Table 14 Mean values for all markers.

Mean values

Age years

CEA ng/ml plasma

Total DNA ng /ml plasma

Mito ng/ml plasma

Alu 115 ng/ml plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml plasma 79/300 115/247

normal 54.1 0.5 8.6 0.9 24.6 1.6 2.8 6.4 8.3

Polyps 71.7 0.7 16.1 2.9 74.4 4.4 5.5 6.4 18.0

Cancer 71.4 1.1 29.7 3.8 122.1 4.5 8.7 7.0 14.3

LGD polyps 73.4 0.46 15.8 3.5 78.3 5.0 5.4 7.11 20.1

HGD polyps 69.1 0.94 16.5 1.91 68.4 3.4 5.5 5.27 14.1

Page 79: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-79

Table 15 Median values for all markers.

Median values Age years

CEA ng/ml plamsa

Total DNA ng /ml plasma

Mito ng/ml plasma

Alu 115 ng/ml plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml plasma 79/300 115/247

Normal group 55.0 0.4 7.2 0.7 24.1 1.6 2.7 5.0 8.4

Polyps group 71.0 0.6 12.5 1.4 62.4 3.2 4.9 4.3 16.0

Cancer group 72.5 0.5 14.9 1.6 73.1 2.2 4.8 6.1 11.7

LGD polyps 73 0.6 12.7 1.5 64.3 3.5 4.8 4.7 18.0

HGD polyps 69 1.1 11.9 1.2 34.8 3.1 4.9 4.2 11.0

Each marker was analysed between normal, polyp and cancer populations (Tables 14 and 15). There

was an increase in the mean values of all markers with increasing pathological grade, though not all

these differences were statistically significant from the control population (Table 16).

Looking at the polyp group separately these patterns were not mirrored in the transition from LGD

to HGD, and statistically there were no significant differences (Table 16). There was a single patient

with a LGD polyp represented an extreme outlier, with a total DNA level of 206 ng/ml. This patient

was removed from the mean and median data, but is included in all other analyses.

4.4 Statistical significance - Mann Whitney U-tests.

Applying a 10 fold Bonferroni correction, the significant P value = 0.005.

Table 16 Statistical significance testing.

Mann Whitney-U test P values

CEA ng/ml plamsa

Total DNA ng /ml plasma

Mito ng/ml plasma

Alu 115 ng/ml plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml plasma 79/300 115/247

Polyps vs. normal groups 0.288 <0.001 0.001 <0.001 <0.001 0.003 0.189 0.002

Cancer vs. normal groups 0.425 0.002 0.002 <0.001 0.015 0.001 0.087 0.001

Polyps and cancer vs. normal 0.267 <0.001 <0.001 <0.001 <0.001 <0.001 0.908 <0.001

LGD vs. HGD polyps 0.906 0.269 0.557 0.796 0.981 0.655 0.438 0.906

Page 80: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-80

As compared with normal controls, patients with benign polyps had significantly raised levels of total

DNA (p<0.001), mitochondrial DNA (p=0.001), Alu 115bp (P<0.001), Line 1 300bp (P<0.001) Alu

247bp fragment (p=0.003, and Alu 115/247 ratio (P=0.002) Table 16. Further investigating the polyp

populations there were no significant differences comparing LGD polyps to HGD polyps.

As compared with normal controls, patients with colon cancer had significantly raised levels of total

DNA (P=0.002), Mitochondrial DNA (p=0.002), Alu 115bp (P<0.001), Alu 247bp (p=0.001), and the

Alu 115/247 ratio (P=0.001) Table 16.

Comparison of normal with all neoplasia patients showed that all markers except the Line 1 79/300

ratio were highly significantly different between the populations (P<0.001).

4.5 Logistic regression and ROC curve values.

Table 17 ROC curve characteristics.

ROC values

CEA ng/ml plamsa

Total DNA ng /ml plasma

Mito ng/ml plasma

Alu 115 ng/ml plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml plasma 79/300 115/247

polyps vs normal 0.578 0.767 0.742 0.856 0.756 0.719 0.596 0.732

cancer vs normal 0.562 0.738 0.743 0.838 0.687 0.764 0.632 0.760

polys and cancer 0.571 0.754 0.742 0.848 0.725 0.74 0.507 0.745

The best single marker in the polyp population was the Alu 115 (ROC=0.856), with the combination

DNA marker showing an excellent diagnostic ability (ROC = 0.921) Table 17.

The best single marker in the colon cancer population was the Alu 115 (ROC=0.838), with the

combination DNA marker showing a very good diagnostic ability for distinguishing cancer from

normal in this series (ROC = 0.854).

Page 81: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-81

The best single marker for diagnosis of all neoplasia in this population was Alu 115bp (ROC = 0.848),

with the combination DNA marker showing a very good diagnostic ability (ROC=0.888). This had a

sensitivity of 81.1%, specificity of 75.8%, positive predicitve value of (PPV) of 84.3%, and negative

predictive value (NPV) of 71.4% (Figure 8).

CEA is shown once again to be a poor diagnostic marker for colonic neoplasia, with no significant

difference between the populations. The ROC values indicating failure to discriminate between the

populations (ROC = 0.578).

Its addition with the 4 marker regression did increase the test performance in all categories (ROC =

0.929, 0.875, 0.901). This final test had a sensitivity of 81.1%, specificity of 79.4%, PPV of 86.0%, and

NPV of 72.9% (Figure 9). Perhaps this demonstrates a role for combination marker types in future

testing.

Figure 8 Combined DNA marker ROC curve.

(Sensitivity = 81.1%, specificity = 75.8%, PPV = 84.3%, NPV = 71.4%).

Page 82: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-82

Figure 9 Combined DNA marker plus CEA ROC curve.

(Sensitivity = 81.1%, specificity 79.4%, PPV = 86.0%, NPV = 72.9%).

4.6 Box plots and whisker analysis.

Boxes represent 25th to 75th centiles and the internal black line the median value. The top whisker is

the maximum SPSS accepted population value (< 1.5 Inter-quartile range (IQR) from the edge of the

box). The bottom whisker is the minimum SPSS accepted population value (<1.5 (IQR) ). “O” is an

outlier in population between 1.5 and 3.0 x IQR from the edge of the box. “*” is an extreme outlier

greater than 3.0 x inter-quartile range from edge of box. The scale on the Y axis is logarithmic to

allow easier evaluation of the main populations. There are normal, polyp and cancer groups.

Figure 10 CEA box plot analysis.

Page 83: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-83

Figure 11 Total Nuclear DNA box plot analysis.

Figure 12 Mitochondrial DNA box plot analysis.

Page 84: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-84

Figure 13 Alu 155bp DNA box plot analysis.

Figure 14 Alu 247bp DNA box plot analysis.

Page 85: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-85

Figure 15 Line 1 300bp DNA box plot analysis.

Figure 16 Line 1 79:300 DNA ratio box plot analysis.

Page 86: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-86

Figure 17 Alu 115:247 DNA ratio box plot analysis.

4.7 Correlation of Total cfDNA with age.

Analysing the most uniform population (normal group), there was no significant correlation with age

using both Spearman’s rho (P=0.07), and Kendalls tau b (P=0.09).

Figure 18 Correlation of total cfDNA with age.

4.8 Conclusions.

This study demonstrates highly significant differences between a “normal” population, a

population with adenomatous polyps, and a population with colonic cancer.

Page 87: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

4-87

Quantities and patterns of circulating DNA differ significantly between the groups, with total

quantities of nuclear and mitochondrial DNA increasing with pathological grade, and

fragmentation patterns changing between normal, adenomatous and cancer populations.

Individual markers demonstrate good diagnostic tests, and in combination provide an

improved test for both polyps and cancer (ROC = 0.888).

Although adding CEA levels to the combination DNA marker did improve the final diagnostic

test for polyps and early cancer, ROC curve = 0.901. It did not add greatly and the results are

included largely to show that this marker, even in combination with others, has little

diagnostic value.

Page 88: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-88

Chapter 5 Oesophago-gastric results.

5.1 Demographics.

The study population comprised 115 patients, with 120 samples and includes: 35 patients without

endoscopic abnormality; 9 patients with Barrett’s oesophagus; 13 patients with high grade dysplasia;

12 patients with invasive cancers restricted to the mucosa (IMC); 3 patients with invasive cancers

restricted to the sub-mucosa (SMC); and 43 patients with advanced cancers.

The reference “normal” group consisted of nine biopsy proven benign Barrett’s oesophagus cases,

and 35 patients representing a typical population attending for endoscopic investigation but with no

detectable abnormality (Table 18). The mean age of the combined normal group was 56.1 years

(range 24 - 80 years), and included 22 men (7 Barrett’s, and 15 normal endoscopy patients).

Significant inter-current diagnoses in this group included: osteoarthritis; ischaemic heart disease;

pernicious anaemia; and peptic ulcer.

Table 18 Normal population indication for endoscopy.

Endoscopy indication Patient

numbers Dysphagia 5

Dyspepsia 10

Ulcer follow up 1

Haematemesis 2

Abdominal pain 8

Change in bowel habit 7

Family history of cancer 1

Anaemia 5

Polyp follow up 1

Melaena 1

Barrett’s surveillance 9

Page 89: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-89

5.2 Patient groups.

Due to well documented difficulties accurately identifying Barrett’s dysplasia grades, the groups

were combined into clinically relevant groups. These groups have a higher concordance for accurate

differentiation pathologically319, 320. When pathological samples showed mixed grades the highest

grade of dysplasia was used. The combined groups were “HGD and IMC”, and “SMC and cancer”.

The HGD and IMC group consisted of 25 patients, 20 men, with a mean age of 70.0 years (range 54-

89 years). There were 30 samples, 15 each of HGD and IMC, six from patients with recurrent disease.

The SMC and cancer groups consisted of 43 patients, 18 men, with a mean age of 68.9 years (range

50-82 years). There were 46 samples, 3 from patients with superficial sub-mucosal invasive disease.

Forty samples were from advanced cancers, staged by CT, EUS, and PET scanning as: 5 x T1N0M0, 16

x T2/3N0M0, 4 x T2N1M0, 5 x T3N1M0, 1 x T4N1M1, 10 had metastatic disease and 2 had unclear

staging prior to receiving palliative chemotherapy. The T0 and T1 cancers were deep sub-mucosal

invading tumours.

In total, both dysplasia and cancer populations include 68 patients, and 76 plasma samples, with a

mean age of 69.5 years, (39 - 89 years).

Page 90: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-90

5.3 Mean and median marker values.

Table 19 Mean value for all markers.

Mean values Total Mito 115 300 247 79/300 115/247

"Normal" 10.8 1.1 35.6 1.8 3.1 6.3 11.0

HGD + IMC 14.1 4.2 42.6 3.2 4.6 6.1 10.3

SMC & Cancer 19.2 6.2 76.9 5.3 4.9 8.8 16.3

Table 20 Median value for all markers.

Median values Total Mito 115 300 247 79/300 115/247

"Normal" 7.3 0.7 24.9 1.5 2.7 5.1 9.5

HGD + IMC 11.0 1.5 30.7 2.4 3.2 5.3 10.3

SMC & Cancer 12.2 1.5 39.5 2.8 2.3 6.3 12.4

Each marker was analysed between normal, HGD and IMC, and SMC and cancer populations (Tables

19 and 20). There was an increase in the mean values of all markers with increasing pathological

grade except for the fragment ratios. Not all of these differences were statistically significant (Table

21).

5.4 Statistical significance - Mann Whitney U-tests.

Applying a 10 fold Bonferroni correction, the significant P value = 0.005.

Table 21 Statistical significance testing.

Mann Whiteny U-test P values Total Mito 115 300 247 79/300 115/247

“Normal” vs HGD + IMC 0.010 0.016 0.121 0.014 0.076 0.939 0.779

“Normal” vs SMC & Cancer 0.013 0.000 0.003 0.004 0.448 0.180 0.180

“Normal” vs all abnormal 0.003 0.000 0.006 0.001 0.715 0.121 0.086

As compared with normal controls, patients with HGD and IMC did not have significantly raised

levels of all markers, Table 21.

As compared with normal controls, patients with SMC and oesophageal cancer had significantly

raised levels of mitochondrial DNA (p=0.000), Alu 115bp (P<0.003), Line 1 300bp (P=0.004) Table 21.

Page 91: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-91

Comparison of normal with all neoplasia patients showed that Total DNA (p=0.003), mitochondrial

DNA (p=0.000), and Line 1 300bp (p=0.001) were highly significantly different between the

populations.

5.5 ROC curve values and Logistic regression.

Table 22 ROC curve characteristics.

ROC curve values Total Mito 115 300 247 79/300 115/247

"Normal" vs HGD + IMC 0.678 0.665 0.607 0.67 0.622 0.505 0.481

"Normal" vs SMC & cancer 0.652 0.713 0.68 0.677 0.454 0.646 0.646

"Normal" vs all abnormal 0.662 0.694 0.651 0.674 0.674 0.585 0.595

The best single marker in the HGD and IMC population was Total DNA (ROC=0.678), with the

combination DNA marker showing a fair diagnostic ability (ROC = 0.718) Table 22.

The best single marker in the SMC and oesophageal cancer population was mitochondrial DNA

(ROC=0.713), with the combination DNA marker showing a good diagnostic ability for distinguishing

cancer from normal in this series (ROC = 0.847). The combination test had a PPV for invasive cancer

patients of 81.1% and a NPV of 70.6% (Sensitivity = 61.3%, specificity = 83.7%) (Figure 19).

The best single marker for diagnosis of all neoplasia in this population was Alu 115 mitochondrial

DNA (ROC = 0.694), with the combination DNA marker showing a fair diagnostic ability (ROC=0.778).

The combination test had a PPV for dysplasia and cancer patients of 78.2%, and a NPV of 65.0%

(Sensitivity = 81.3%, specificity = 60.5%) (Figure 20).

Page 92: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-92

Figure 19 Normal group vs. all invasive cancer group.

Sensitivity = 61.3%, specificity = 83.7, PPV = 81.1%, NPV = 70.6%.

Figure 20 Normal group vs. all abnormal oesophagus group.

Sensitivity = 81.3%, specificity = 60.5%, PPV = 78.2%, NPV = 65.0%.

Page 93: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-93

5.6 Box plots and whisker analysis.

Boxes represent 25th to 75th centiles and the internal black line the median value. The top whisker is

the maximum SPSS accepted population value (< 1.5 Interquartile range (IQR) from the edge of the

box). The bottom whisker is the minimum SPSS accepted population value (<1.5 (IQR) ). “O” is an

outlier in population between 1.5 and 3.0 x IQR from the edge of the box. “*” is an extreme outlier

greater than 3.0 x inter-quartile range from edge of box. There are normal, mucosal neoplasia and

cancer groups.

Figure 21 Total Nuclear DNA box plot analysis.

Page 94: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-94

Figure 22 Mitochondrial DNA box plot analysis.

Figure 23 Alu115bp DNA box plot analysis.

Page 95: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-95

Figure 24 Alu 247bp DNA box plot analysis.

Figure 25 Line 1 300bp DNA box plot analysis.

Page 96: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-96

Figure 26 Line 1 79:300 DNA fragment ratio box plot analysis.

Figure 27 Alu 115:247 DNA fragment ratio box plot analysis.

5.7 Conclusions.

This study demonstrates highly significant differences in cfDNA between a “normal”

population, a population with dysplasia and early cancer, and a population with invasive

cancers.

Quantities of circulating DNA differ significantly between the groups, with total quantities of

nuclear and mitochondrial DNA increasing with pathological grade. Mean and median

fragmentation ratios increase through grades, which although not significant, may reflect a

true change in populations.

Page 97: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

5-97

Although most individual markers demonstrate only fair diagnostic tests, in combination

they provide an improved test for both neoplastic groups and in particular provide a good

test for advanced invasive cancers (ROC = 0.847).

Page 98: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-98

Chapter 6 Paired colonic and oesophageal results.

6.1 Introduction.

This chapter looks at the feasibility of using cfDNA markers to judge whether a colonic or

oesophageal lesion has been completely removed endoscopically. Current practice involves

repeating the endoscopic assessment at 3, 6 and 12 monthly intervals.

The markers will be measured before and after resection in pairs from the same patient. The paired

samples will allow the statistically more powerful Wilcoxon signed rank test to be used.

The mean pre and post-resection levels will also be compared to the control population using the

Mann-Whitney test. This will analyse whether pre-resection patient levels and later post-resection

levels are different from background control results.

6.2 Colon results.

6.2.1 Demographics .

The paired samples came from 15 patients with an average age of 72 y, 7 men.

6.2.2 Lesions.

Pre-resection lesions included one polyp cancer, nine HGD polyps, and five LGD polyps. After follow

up over 2 years 14 patients had been resected back to normal, and 1 patient had a small area of

residual LGD polyp at the time of the last sampling.

Only the fourteen samples with complete resection were analysed for statistical purposes. However,

in the line graphs below, results included the sample with residual LGD dysplasia and an

intermediate sampling result in a patient with residual LGD who went on to complete resection.

These are included for comparison purposes.

6.2.3 Mean results.

Page 99: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-99

Table 23 below shows mean values for the patients with polyps before and after resection. To allow

comparison the polyps group (pre-resection equivalent) and normal (post-resection equivalent) are

included.

Although the absolute values are difficult to interpret due to the small numbers, there is a consistent

trend mirroring the differences between normal and polyp groups.

Table 23 Paired means pre and post resection, and normal and polyps.

Mean values

CEA ng/ml plamsa

Total DNA ng /ml plasma

Mito ng/ml

plasma

Alu 115 ng/ml plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml plasma

79/300 115/247

Polyps group 0.7 16.1 2.9 74.4 4.4 5.5 6.4 18.0

Normal group 0.5 8.6 0.9 24.6 1.6 2.8 6.4 8.3

Pre-resection 0.80 33.58 8.27 101.07 26.06 20.53 3.63 14.96

Post-resection 0.75 12.89 6.55 51.49 1.78 5.36 7.09 10.96

6.2.4 Significance testing.

Paired sample values pre and post-resection were tested with the Wilcoxon signed rank test, non-

paired comparisons against the control group and were tested with the Mann-Whitney statistic.

Applying a 10 fold Bonferroni correction, the significant P value = 0.005.

Table 24 Significant P values.

P values

CEA ng/ml plamsa

Total DNA ng /ml plasma

Mito ng/ml

plasma

Alu 115 ng/ml

plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml

plasma 79/300 115/247

pre vs post-resection 0.637 0.551 0.433 0.331 0.005 0.875 0.001 0.272

pre-resection vs normal 0.048 0.025 0.004 0.000 0.001 0.049 0.013 0.046

post-resection vs normal 0.064 0.138 0.347 0.028 0.507 0.020 0.127 0.785

Page 100: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-100

6.2.5 Individual markers.

6.2.5.1 CEA analysis.

There are no significant differences between the polyp CEA levels, before and after resection (P >

0.048). Moreover the trend does not decrease towards normal levels and the illustration below

shows this (black lines represent cases resected back to normal, and red broken lines cases not

resected back to normal). This confirms the opinion that CEA is a poor diagnostic marker.

Figure 28 Paired CEA values pre and post resection.

6.2.5.2 Line 1 79bp - total cfDNA.

There are no significant differences between the polyp total DNA levels, before and after resection

(P > 0.025). However, the trend does decrease towards normal levels and the illustration below

shows this.

Page 101: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-101

Figure 29 Paired total cfDNA values pre and post resection.

6.2.5.3 Mitochondrial cfDNA.

There are no significant differences between the patients’ mitochondrial DNA levels before and after

resection. However, when comparing pre-polyp levels to the control population there is a significant

difference ( p=0.004). Post resection levels are not significantly different to the normal population

(p=0.347). The illustration below shows this, although not all cases follow this pattern.

Page 102: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-102

Figure 30 Paired mitochondrial cfDNA values pre and post resection.

6.2.5.4 Alu 115bp fragment.

There are no significant differences between the patients’ Alu 115bp DNA levels before and after

resection. However, when comparing pre-polyp levels to the normal population there is a significant

difference (p=0.000). Post resection Alu 115bp levels are not significantly different to the normal

population (p=0.028). The illustration below shows this, although not all cases follow this pattern.

Figure 31 Paired Alu 115bp values pre and post resection.

Page 103: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-103

6.2.5.5 Line 1 300bp fragment.

There are significant differences between the patients’ Line1 300bp DNA levels before and after

resection of their polyps (p=0.005). When comparing pre-polyp DNA levels to the normal population

there is also a significant difference (p=0.001). Post resection levels are not significantly different to

the normal population (p=0.507).The illustration below shows this, although not all cases follow this.

Figure 32 Paired Line 300bp values pre and post resection.

6.2.5.6 Alu 247bp fragment.

There are no significant differences between the polyp total DNA levels, before and after resection

(P > 0.020). However, the trend does decrease towards normal levels and the illustration below

shows this.

Page 104: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-104

Figure 33 Paired Alu 247bp values pre and post resection.

6.2.6 Marker ratios.

6.2.6.1 Line 1 79:300.

There are significant differences between the patients’ Line 1 79/300bp ratio levels before and after

resection of their polyps (p=0.001). However, when comparing pre-polyp Line 1 79/300bp ratio

levels to the normal population there is no significant difference (p=0.013), and therefore the

analysis of this marker is uncertain.

Post resection Line 1 79/300bp ratio levels are not significantly different to the normal population

(p=0.127), and therefore the mixed results reflect the poor diagnostic usefulness of this marker

when used alone.

The illustration below does show a clear change in ratio with resection, which would be in keeping

with the theories of circulating DNA origins reflecting the fall in the amount of apoptotic long

fragment DNA. Perhaps this explains in part why this marker is helpful in combination with other

markers.

Page 105: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-105

Figure 34 Paired Line 1 79:300 ratio values pre and post resection.

6.2.6.2 Alu 115:247.

There are no significant differences between the polyp Alu 115/247bp ratio levels, before and after

resection (P > 0.046). However, the trend does decrease towards normal levels and the illustration

below shows this.

The illustration below shows a mixed picture but there are only a few cases that rise. Interestingly,

the fall in ratio is the exact opposite of the Line 1 ratio above, but does mirror the trend in the polyp

and cancer groups compared to normal patients. Looking at the results the large Alu 247bp fragment

levels do fall.

Page 106: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-106

Figure 35 Paired Alu 115:247 ratio values pre and post resection.

Page 107: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-107

6.3 Oesophageal results.

6.3.1 Demographics .

The paired samples came from 14 patients, 12 men, with an average age of 66.3 years.

6.3.2 Lesions.

Pre-resection lesions included 4 SM1 invasive cancers, 3 IMC lesions, and 7 HGD.

After follow up over 2 years 8 patients had been resected back to normal Barrett’s, 1 patient later

developed carcinomatosis of unknown primary, 1 patient had a residual IMC lesion, 3 patients had

residual HGD lesions, and 1 patient developed a LGD lesion.

Post resection diagnoses were made by an experienced endoscopist using specialist dye spray

techniques and gold standard biopsy practice. Biopsy specimens and resection samples were

reviewed by two experienced pathologists.

Only the eight samples with complete resection back to normal Barrett’s were analysed for

statistical purposes. However, in the line graphs below results included the other samples for

comparison purposes. The sample pair with residual LGD dysplasia sample was represented as

normal due to the difficulties discriminating LGD from inflammation in specimens.

6.3.3 Mean results.

Table 25 below shows mean values for patients with neoplastic Barrett’s lesions before and after

resection. To allow comparison the mucosal neoplasia group (pre-resection equivalent) and control

group (post-resection equivalent) are included.

Although the absolute values are difficult to interpret due to the small numbers, there is little if any

pattern or difference visible.

Page 108: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-108

Table 25 Paired means pre and post resection, and control and mucosal neoplasia.

Mean values Total DNA

ng /ml plasma

Mito ng/ml

plasma

Alu 115 ng/ml

plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml

plasma 79/300 115/247

Mucosal neoplasia group 14.1 4.2 42.6 3.2 4.6 6.1 10.3

Control group 9.88 0.99 34 1.7 2.83 6.3 11.1

Pre-resection 12.36 1.57 32.17 2.23 3.57 7.94 9.36

Post-resection 14.04 0.98 37.08 2.57 6.41 6.42 6.35

6.3.4 Significance testing.

Paired sample values pre and post-resection were tested with the Wilcoxon signed rank test, non-

paired comparisons against the control group and were tested with the Mann-Whitney statistic. In

keeping with the mucosal neoplasia group in chapter 5 there are no markers that are significantly

different to the control population results.

Applying a 10 fold Bonferroni correction, the significant P value = 0.005.

Table 26 Significant P values.

P values

Total DNA ng /ml plasma

Mito ng/ml

plasma

Alu 115 ng/ml

plasma

Line 300 ng/ ml plasma

Alu 247 ng/ml

plasma 79/300 115/247

pre vs post-resection 0.859 0.515 0.314 0.260 0.594 0.110 0.678

pre-resection vs normal

0.041 0.251 0.522 0.293 0.339 0.586 0.748

post-resection vs normal

0.032 0.679 0.517 0.422 0.006 0.312 0.101

6.3.5 Individual markers.

6.3.5.1 Line 1 79bp - total cfDNA.

There are no significant differences between the patients’ total DNA levels before and after

resection (P > 0.032). The illustration below shows a general reduction in levels but unfortunately

total DNA is not a good measure of completeness of resection.

(Black lines represent cases resected k to normal, and red broken lines cases not resected to normal)

Page 109: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-109

Figure 36 Paired total cfDNA values pre and post resection.

6.3.5.2 Mitochondrial cfDNA.

There are no significant differences between the patients’ mitochondrial DNA levels before and after

resection (P > 0.251). The illustration below shows a mixed picture.

Figure 37 Paired mitochondrial cfDNA values pre and post resection.

Page 110: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-110

6.3.5.3 Alu 115 fragment.

There are no significant differences between the patients’ Alu 115bp fragment DNA levels before

and after resection (P > 0.314). The illustration below shows a mixed picture.

Figure 38 Paired Alu 115bp values pre and post resection.

6.3.5.4 Line 1 300bp fragment.

There are no significant differences between the patients’ Line 1 300bp fragment DNA levels before

and after resection (P > 0.260). The illustration below shows a mixed picture.

Figure 39 Paired Line 1 300bp values pre and post resection.

Page 111: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-111

6.3.5.5 Alu 247bp fragment.

There are no significant differences between the patients’ Alu 247bp fragment DNA levels before

and after resection, although it is the best test (P > 0.006). The illustration below shows mainly an

increased level of the “apoptotic” fragment post resection, which doesn’t fit the explanations given

for the origin of circulating DNA. This result needs to be treated cautiously.

Figure 40 Paired Alu 247bp values pre and post resection.

6.3.6 Marker ratios.

6.3.6.1 Line 1 79:300.

There are no significant differences between the patients’ Line 1 79/300bp fragment ratio before

and after resection (P > 0.110). However, the illustration below shows a mainly decreased level

regardless of resection, which may illustrate why it adds diagnostically as part of the multi-marker

model.

Page 112: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-112

Figure 41 Paired Line 1 79:300 ratio values pre and post resection.

6.3.6.2 Alu 115:247.

There are no significant differences between the patients’ Alu 155/247bp fragment ratio before and

after resection (P > 0.101). The illustration below shows a mixed picture.

Figure 42 Paired Alu 115:247 ratio values pre and post resection.

Page 113: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

6-113

6.4 Conclusions.

The number of paired samples resected to normal in colonic and oesophageal neoplasia

cases is small, making interpretation difficult. There are occasional trends, and these data

warrant larger studies in both cancer types.

In colonic neoplasia the Line 1 300bp marker is significant different in the paired and non-

paired analysis. This appears to confirm its diagnostic role in colonic neoplasia and suggests

it may be feasible to use this as a measure of endoscopic polyp resection.

The other markers mirror the results in chapter 4, but are not significant. This pattern is

confirmed by the graphical illustrations which suggest that the total DNA, mitochondrial

DNA, Alu 115bp fragment, Alu 247bp fragment, and Line 1 79/300bp fragment ratio may be

helpful in the diagnosis of colonic neoplasia.

In oesophageal neoplasia the best marker is the Alu 247bp marker which approaches

significance (P=0.006), but the result appears to confound current understanding of cfDNA

origins. The graphical illustrations suggest that the Line 1 300bp and Line 1 79/300bp

markers may be helpful in a larger oesophageal neoplasia study.

Overall the oesophageal results reflect those in chapter 6 where no marker was significantly

different when comparing mucosal neoplasia to the control group. It seems unlikely that

these markers will help to judge complete endoscopic removal.

Page 114: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

114

Chapter 7 Discussion and conclusions.

Colorectal and oesophageal cancer carry a high mortality in the UK, and efforts to diagnose the

disease at an earlier stage are regarded as key to reduce associated deaths54. Current diagnostic

approaches have problems with poor sensitivity and low acceptability. This study was undertaken to

try and find markers to identify early cancer and thereby address these issues.

cfDNA shows potential as an early cancer marker with Alu 115bp, Alu 247bp, Line 1 79bp, Line 1

300bp and mitochondrial DNA sequences previously shown to be raised in populations with

advanced cancer.

Using improved techniques, this study tested the hypothesis that patients with pre-cancerous

change and early cancers also have significantly different cfDNA levels and ratios when compared to

control or “normal” populations. As part of this study, levels of the markers were tested before and

after endoscopic resection to act as a marker for completeness of endoscopic resection.

This chapter reviews the individual results and discusses them in the context of the circulating

biomarker literature, the origins of cfDNA, and the potential role as a diagnostic test for colonic and

oesophageal cancers. The current evidence based approaches to the collection and processing of

specimens will also be reviewed.

7.1 Control population results.

The patients in the control group have symptomatic benign gastrointestinal disease, and normal

investigation. With a mean age of 54.1 years they represent an “at risk” group for colorectal and

oesophageal cancer and therefore are a good clinical control group.

The total amount of cfDNA in the control population in this study was 10.8 ng/ml of plasma ,

comparable to a study of normal population baselines reporting 16 ng of cfDNA/ml of whole blood.

Age does not seem to affect cfDNA levels 124.

Page 115: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

115

Included in the oesophageal control group were patients with Barrett’s oesophagus. This is

important if the test is to have a clinical role. Although the number of these patients was small there

was no significant difference in the individual marker results, except the ratio marker Alu 115/247bp.

In the context of non-significant individual markers this is suspicious for a type I error. Further

testing will be needed to confirm this.

Reservations regarding the utility of cfDNA markers in discriminating the normal population have

been expressed. An ovarian cancer study included a population of patients admitted to hospital for

treatment of benign diseases and these formed part of the control group. Results from this control

group showed raised cfDNA levels reaching 16 ng/ml of plasma. It was therefore concluded that

cfDNA has reduced diagnostic accuracy170. However, these ‘benign’ conditions included patients

admitted to hospital with infections, autoimmune disease, post organ transplant, post-trauma, AIDS,

and asthma. These conditions are likely to be associated with active inflammation and increased

apoptosis which cause proven increases in circulating DNA103, 107, 113, 164. Inflammation in a study of

circulating DNA nucleosomes shows a clear correlation with DNA levels174. Such severely ill patients

are unlikely to be represented in large numbers within the population attending screening visits and

would be rapidly removed from the test pool. We therefore believe that in our group of patients

confounding factors such as inflammatory lesions would be picked up through the clinical history

and treated separately and that cfDNA could still be a good marker for differentiating between

normal and cancer groups.

In a single patient with Barrett’s oesophagus the cfDNA level was surprisingly high with a total DNA

of 78ng/ml. The reason for this patient’s results are not clear; the patient was losing weight and had

transfusion dependent anaemia. A similar statistically outlying patient with a high DNA level of

47ng/ml had an unexpected pancreatic cancer found.

Page 116: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

116

It may be that in the 1st patient a similar diagnosis will eventually be made. These cases

demonstrate both the strength of measuring cfDNA as a test for cancer, and the problems that

clinicians may face when a positive cfDNA result leads to a negative clinical test.

For the purposes of this study the first patient was included but the second patient was excluded.

7.2 Colonic results.

Screening for colorectal cancer is challenging with the diagnosis of pre-cancerous adenomatous

polyps regarded as essential for the prevention of cancer, and yet they represent a more difficult

diagnostic target.

This study demonstrated highly significant differences between the control population, the

population with adenomatous polyps, and the population with colonic cancer.

Quantities and patterns of circulating DNA differed significantly between the groups, with total

quantities of nuclear and mitochondrial DNA increasing with pathological grade, and fragmentation

patterns changing between normal, adenomatous and cancer populations. Test results showed that

the markers individually acted as good markers of disease and that in combination they provided an

improved test for both polyps and cancer with a ROC = 0.888. To our knowledge this is the first study

to demonstrate significant differences in circulating DNA in colonic polyps and cancer patients and

offers the prospect of improved diagnosis of neoplasia in colorectal cancer screening with a simple

blood test.

However, overlapping results with false positives in normal patients, and false negatives in

neoplastic patients needs some work. The false positive patients with an excess of cfDNA seem likely

to result from initial storage and processing issues, artefactual haemolysis and white cell lysis, or

confounding sub-clinical disease.

Page 117: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

117

A stricter approach to sampling, a visual and microscopic assessment for haemolysis and careful

patient questioning with prospective CRP testing to exclude patients with inflammation (CRP>5) 321

may help avoid this. False negative testing may prove more difficult but prospective testing with

limited storage and careful processing without cell separation or filtration may help. Tumour sizes,

pathological differentiation, and proliferation measures were not used in this study and may

influence the results e.g. small and well differentiated tumours with low proliferation rates may

produce low cfDNA levels. Further studies looking at oxidised or methylated 98, 182, 211 cfDNA levels

may help sensitivity and specificity for cancer, with assessment of hypermethylated foetal cfDNA in

the maternal blood circulation showing the possibilities322.

Although total cfDNA levels differed significantly between the polyps and cancer groups within the

polyp population neither size, nor estimated surface area showed significant correlation with cfDNA

in both the total polyp population (p=0.28) and the larger category of LGD polyps (p=0.24). Perhaps

surprisingly there was also no significant difference in total cfDNA levels (p=0.926) or size between

the LGD (52.5mm) and HGD (51.9mm) polyps. Accurate sizing and grading of polyps is difficult with

lateral and outward growth contributing to an overall volume, and small foci of HGD dictate polyp

categories. Therefore although we have found no differences in cfDNA with size or grade of polyp it

is difficult to be entirely confident that they are not relevant to cfDNA levels. It seems reasonable to

group polyps together, and it will be difficult to get a clearer answer.

Whilst a larger population would have been desirable, these results arise in a clinically relevant

population. Results from other studies also provide good support for our findings. A study published

in 2009 in abstract form only, confirmed the significance of the 115bp and 247bp Alu marker

changes in early and late colorectal cancer patients in plasma. However, results in patients with

polyps were not significantly different to the “clean-colon healthy subject” control population 323.

Page 118: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

118

No details of sample processing methodology were included and so interpretation of the true

negativity of this study is difficult. Perhaps our results reflects the small fragment cfDNA and spin

protocols used. The polyps in our population were also unusually large and although in our analysis

size was not found to correlate with DNA levels, this may have contributed to our positive findings.

A paper published in 2006 also demonstrated strikingly similar DNA levels to our findings, although

in serum. The study looked at more advanced colon cancers and showed a healthy control group

with an almost identical median total DNA level of 7.7 ng/ml (current study - 6.86 ng/ml). CfDNA

levels in cancer patients was 35.8 ng/ml compared to 14.58 ng/ml) in this study. A combined

diagnostic ROC score discriminating cancer cases was 0.92. 242.

In other cfDNA studies the fragment ratio was the most sensitive diagnostic marker89, 94, 115, 149, 165,

however it is unclear whether DNA integrity of all genes is significant as two studies found negative

results166, 324. In our colonic results there were significant differences in the colonic ALU 115/247bp

fragment ratio, and the ratio results did add to the 4 marker DNA regression analysis. However, the

fragmentation ratio was not the most sensitive marker in our study possibly suggesting that it acts as

a more sensitive marker in advanced cancers.

Mitochondrial DNA, with multiple copies in each cell, has been proposed to provide a more accurate

marker in the analysis of low quantities of circulating DNA. In agreement with this, our colonic

results showed highly significant differences between the groups with a rise in benign and cancer

groups. However, in logistic regression analysis it did not add to the accuracy obtained with nuclear

cfDNA markers, and other studies have reported both high and low levels in cancer325-327. We did not

use it as a marker for our final analysis because of this.

Page 119: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

119

cfDNA quantification and Alu 247 fragment results have now been demonstrated to be consistent

across serum studies in both non-inflammatory normal patients, three types of gastro-intestinal

cancers149, 157, 328 and breast cancer115. We believe that they offer the prospect of a broad neoplasia

screening test in non-inflammatory cancers. This approach would be highly attractive to patients,

although greater specificity for cancer type would be required. Differentiating between cancers

however may be possible from other tests, even using the same blood sample. For example, in our

study the 300bp marker did not perform well, but this fragment did show promise in breast cancer147.

The different DNA patterns may help identify gastrointestinal cancers and differentiate them from

other cancer types e.g when the Alu 247 bp fragment is raised. This interesting finding may relate to

the biological handling of cfDNA from the digestive system before it enters the normal circulation329,

330, and may give the Alu 247 bp fragment a degree of specificity for colorectal neoplasia.

Alternatively specificity may be achieved by using additional PCR markers such as methylated Septin

9, or ELISA based assays with Colon Cancer Specific-2, or TIMP 1 proteins177, 187, 268, 278. This may allow

combinations similar to the CEA results in our study offering the prospect of a more specific blood

test screen for colorectal cancer. In our study, CEA did not add greatly to the ROC, and the results

are included to show that this marker, even in combination with others, has little diagnostic value.

Mitochondrial fragments with highly significant results across both studies and high individual ROC

values (0.742, 0.694) may also offer a broad neoplasia screen. Previous studies have shown

significant findings discriminating testicular and breast cancer 173, 212, 326, and mitochondrial cfDNA

also has promise as a universal neoplasia marker.

Alternatively, cfDNA may be a useful triage test for patients with a positive faecal occult blood test.

Those with cfDNA below a threshold defined by the ROC curve could be spared the risk and cost of

colonoscopy

Page 120: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

120

In 2010 a study sequenced all circulating cfDNA in breast cancer patients. Alu and line were

significant markers which in combination with others gave sensitivity (90%) and specificity (95%) in

breast cancer patients208. This should give a more complete answer and looks a promising approach

for the future.

7.3 Oesophageal results.

Screening for oesophageal cancer is especially challenging even in those diagnosed with Barrett’s

oesophagus. The diagnosis of pre-cancerous dysplasia and early cancer is essential to improve the

prognosis and represents a very challenging diagnostic target.

This study demonstrates highly significant differences between the control population, a population

with invasive cancers, and a combined population with mucosal neoplasia and invasive cancers.

Quantities and patterns of circulating DNA differ significantly between the groups, with total

quantities of nuclear and mitochondrial DNA increasing with pathological grade, and fragmentation

patterns changing between control and invasive cancer populations.

Although most individual markers are only moderately successful as diagnostic tools, in combination

they provide an improved test. In particular they provide a good test for advanced invasive cancers

(ROC = 0.859), although of course the clinical problem is to detect patients before they reach this

stage.

Comparing cfDNA levels from oesophageal neoplasia patients to those form colorectal patients it is

apparent that all sample levels are broadly lower except for mitochondrial DNA. With at least some

of the blood by-passing the liver circulation this is surprising. Why oesophageal cancers should

produce less cfDNA is unclear. Technically some of the plasma samples were stored for over 2 years

reflecting the relative rarity of mucosal Barrett’s neoplasia samples.

Page 121: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

121

A previous study has shown that cfDNA levels decrease with time in storage even at -80 °C331, and

some of the neoplasia samples may have suffered because of this. The Low levels of cfDNA in the

cancer group however remain unexplained. They were collected towards the end of the study and

stored for less than 1 year. As discussed in the colon results above, false positives and false negatives

will need to be addressed. The strategies suggested may also help with the low DNA levels found

with the oesophageal samples.

When the results were analysed without including both normal Barrett’s oesophagus outlier samples,

suspicious for underlying cancers, the mean results for line 1 79 bp, Line 1 300 bp, Alu 115 bp and

Alu 247 bp fragments were all significant at p = 0.001. The ROC value, discriminating controls from

all abnormalities was also 0.809, perhaps showing more clearly the potential for this type of testing.

To our knowledge this is the first study to demonstrate these circulating DNA changes in

oesophageal dysplasia and oesophageal adenocarcinoma, offering the prospect of improved

diagnosis of neoplasia with a simple blood test. While the test lacks specificity for oesophageal

cancer, it could be particularly useful as a triage test for patients with Barrett’ s oesophagus. Those

with cfDNA below a threshold defined by the ROC curve could be spared the risk and cost of

oesophagogastroscopy.

Alternatively specificity for oesophageal adenocarcinoma may be available from other tests, which

could be performed on the same blood sample following an abnormal result. Methylated APC or

LOH at microsatellite markers202, 332 may begin to offer this.

Sequencing and analysis of all cfDNA may be the ultimate diagnostic approach in oesophageal cancer,

giving both sensitivity and specificity. Further studies are required however.

Page 122: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

122

Whilst a larger population would be desirable, these results arise in a clinically relevant population

with other results providing good support for our findings. Similar DNA levels in plasma from a study

of more advanced mainly oesophageal adenocarcinomas show a healthy control group with an

almost identical median total DNA level of 10 ng/ml (current study 7.3 ng/ml), and resectable

cancer patients with 25 ng/ml (current study 12.2 ng/ml) 333. In the above study cfDNA was

compared retrospectively to CEA (cut off level of 5 ng/ml) as a detector of recurrent disease, cfDNA

detecting 100% of recurrences and CEA detecting only 33%. CEA continues to show little clinical use.

Mitochondrial DNA in our study rises in dysplastic and cancer groups showing highly significant

differences in the cancer group. In logistic regression analysis it is the best single marker, although

once again it adds little to the 4 DNA marker in common with colon cancers. Mitochondrial DNA,

Total DNA and Line 1 300 fragments are significant in our colonic and oesophageal results as well as

breast cancer. Results in other cancers and our colonic series with the Alu 247 bp suggests these 4

markers offer the best cfDNA prospect of a broad neoplasia screening test in cancers.

The oesophageal fragment ratio results are not significantly different between the groups, which is

disappointing and surprising. A study of squamous oesophageal cancers showed a significant

difference (P=0.001), albeit against healthy controls, and other studies with adenocarcinoma have

also been positive. Perhaps the long DNA storage once again contributed to the loss of DNA

fragments in particular. It is noteworthy however that on removing the 2 outlier cases of normal

Barrett’s oesophagus disease the results showed significant differences between the groups and this

does need to be examined further.

In conclusion cfDNA markers offer an interesting prospect for a blood test screen in oesophageal

cancer, with our results showing the possibilities in early oesophageal cancer and pre-cancerous

dysplasia. We have identified a combination marker in our population able to discriminate control

patients with common medical problems from patients with dysplasia and invasive cancers, though

this requires validation in a larger independent series.

Page 123: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

123

7.4 Paired results.

The number of cases with paired samples resected to normal in colonic and oesophageal neoplasia

patients is small, making statistical interpretation difficult.

In colon paired cases there is fall in Alu 247 bp fragment quantity and 115 bp / 247 bp ratio which

although non-significant is similar to a study of patients who responded to radiotherapy for rectal

cancer157. In colonic neoplasia the Line 1 300 bp marker is significantly different in the paired and

non-paired analysis. This appears to confirm its changing level in colonic neoplasia and suggests even

without a second validating series that the statistically significant results are correct.

The oesophageal paired data showed little change, and were not significantly different to normal.

Clinically this reflects the fact that only mucosal disease is resectable and mirrors the results from

the larger unpaired mucosal neoplasia data (P > 0.10). Disappointingly the resected samples did not

return to normal values. Whether this is a sample size issue, a reflection of underlying abnormal

tissue remaining, or whether there is ongoing inflammation post resection is unclear. There are no

studies available to clarify this.

Statistically the Alu 247 bp marker is the only marker approaching significance (P=0.006), but with

the levels rising after resection and confounding current understanding of cfDNA origins this

significance should be treated cautiously.

Graphical analysis of the samples resected shows that nearly all of the Line 1 300bp and Line 1

79/300 bp markers change in similar ways, and these may be helpful in a larger colonic neoplasia

study. (Black lines = specimens resected back to normal, red broken lines = not fully resected to

normal)

Page 124: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

124

Figure 43 Paired Line 1 79:300 ratio values pre and post resection.

Figure 44 Paired Line 1 300bp values pre and post resection.

Overall, the colonic and oesophageal paired results are disappointing, and it seems unlikely these

markers will help to judge complete endoscopic removal.

Page 125: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

125

7.5 Sample processing.

Within the study timescale, new research and equipment has been developed. Increasing evidence

of the need to collect smaller DNA fragments137, 334 has led to the industry creating new specialist kits

with Quiagen and nucleospin being the first two. With fast, reproducible, and standardized

techniques these kits should be considered the gold standard.

Studies looking at the effects of storage times on plasma samples have shown increased DNA levels

after 2-6 hours, presumably from haemolysis at the time of sampling. Processing of samples in less

than 2 hours is optimal and should prevent genomic DNA contamination153, 308. Haemolysis of

samples is not entirely time related and it is important to avoid this through careful blood sampling.

A double spin protocol to separate cells from plasma without the use of filtration and histopaque is

optimal 153, 309, 310. A further fast spin on thawing plasma prior to separating cfDNA is recommended

by the manufacturer, making a triple spin protocol comprehensive.

DNA degradation occurs at a fast rate even at -80°C. An extended collection period over 2 years is

not recommended331. Testing prospectively is recommended however where this is not possible

storage in optimal buffers at temperatures < -80°C, with minimal freeze thaw cycles is best 210, 335.

7.6 Implication for the origins of circulating DNA.

In control patients the cfDNA level of the Alu 115 bp sequence is much greater than would be

expected. When we observe the results of the Line 1 79 bp product (8-10ng/ml) in comparison to

the Alu 115 bp product (25-35 ng/ml), both measured against the same genomic standard curve, we

see that the Alu product is 3 times more abundant. If cfDNA was to arise from apoptosis and

necrosis of normal cells then the relative quantities of Alu and Line 1 sequences should be 1:1.

Therefore simple apoptosis and necrosis leading to release of cfDNA cannot explain this

phenomenon.

Page 126: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

126

Previous comparisons of Alu to β-globin gene sequences in cfDNA have shown similar results and

active secretion of Alu sequences was therefore proposed336. DNA excretion has also been observed

in differing in vitro-experiments and the phenomenon seems likely to play a significant role in cfDNA

generation 337, 338.

Quantities of both Line 1 79 bp and Alu 115 bp cfDNA sequences increase in cancer and pre-

cancerous patients but the reason for this unclear. If as traditionally proposed increased apoptosis

and necrosis of neoplastic cells occurs then the ratio of Alu 115 bp to Line 1 79 bp should be lower or

unchanged. Apoptosis or necrosis of normal cells should reduce the ratio. However, further

increases in the Alu 115 bp : Line 1 79 bp sequence (4:1) in both the oesophageal and colorectal

cancer series suggests that the phenomenon leading to this unexpected ratio increases.

Paired results also demonstrate sudden changes in cfDNA levels in early cancers in the same patient.

This seems unlikely to be secondary to increased apoptosis and necrosis of the tumour or

surrounding normal cells (figure 45).

Figure 45. DNA quantities in a patient with progressive oesophageal cancer.

Page 127: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

127

With an oesophageal cancer study showing little change in cfDNA cancer levels until metastatic

disease is present , it is hard to see how increased apoptosis and necrosis can explain cfDNA

levels339.

Perhaps these changes reflect a loss of cellular control as well as active secretion, with increased

retro-transposon activity reflecting and even contributing to the cancer process340, 341 . Line 1 and

Alu sequences are both produced by retro-transposon activity, although only the long Line 1

sequence is able to self replicate. Alu relies on Line 1 activity but is much shorter and therefore

quicker to replicate. Increased retro-transposon activity may explain both the overall increasing

quantities of Alu and Line 1 cfDNA sequences, and the increasing ratio of Alu to Line 1.

Alternatively this may reflect an increased immune response to the cancer cells, with evidence for

white cell activation leading to increasing DNA excretion. However, why the DNA excreted should be

high in Alu sequences is unclear 342.

Finally although a phenomenon such as secretion of DNA is necessary to explain the observed ratios

and types of cfDNA sequences in these experiments, apoptosis and necrosis of neoplastic and

normal cells may still contribute. This complex system would explain the observed increases in

cfDNA, the “ladder phenomenon”, and the changing Alu and Line 1 levels.

7.7 Diagnostic testing.

While there is undoubtedly a rise of cfDNA in cancer patients, current results are not clinically

sensitive or specific enough. Improved processing may help sensitivity and allow a screening

approach to be developed, but the problems raised by lack of specificity for cancer sub-type remain.

Validation of these results in a second population is required. In particular the ability to

prospectively select normal patients from their given test result needs to be proven especially in

light of the confounding presence of inflammatory conditions.

Page 128: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

128

If the results prove reliable then a clinical use for this test may arise. The most obvious role for

quantitative cfDNA testing is in screening for cancers, where a negative result may avoid further

expensive and invasive tests. Importantly our results show changes in cfDNA in dysplasia samples

compared to control groups which is an important and new finding, and a pre-requisite for screening

an asymptomatic population. Barrett’s patients in particular with negative results may be able to

avoid the unpleasant bi-annual endoscopy and biopsy.

Identification of new additional markers, or a combination of old and new markers may provide a

more specific and clinically useful test. Perhaps quantitative study of oxidised or hyper-methylated

cfDNA will add sensitivity, or perhaps next generation sequencing techniques looking at all cfDNA

sequences will provide a breakthrough.

7.8 Conclusions.

In conclusion, this study has shown that nuclear and mitochondrial cfDNA sequences are clearly

raised in colonic and oesophageal dysplasia and cancer, which are new and important findings.

We have identified that a combination of markers used in our population could discriminate control

patients with common medical problems from patients with pre-cancerous changes and operable

cancers. The ROC curves show that these markers present a good to excellent discriminating test,

though this requires validation in a larger independent series.

These results demonstrate the possibility of using cfDNA to screen for asymptomatic and

unrecognized cancers using a simple blood test although further specificity or a negative test

approach will be required.

Our results seem incongruous with the established hypothesis that apoptosis and cellular necrosis

causes escape of cfDNA. The exact mechanism is still unclear but perhaps loss or change in the cells

usual control processes allows increased DNA replication and subsequent overspill into the

circulation.

Page 129: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

129

Addendum

Reflections after Viva Voce examination

If cfDNA testing is to have a diagnostic or prognostic role it is important to understand the intra-

individual test variability in control and cancer patient cfDNA levels over time. The body has clear

diurnal changes in hormone and metabolic profiles and the processes leading to cfDNA are also likely

to be dynamic. Although in a steady state situation the cfDNA level appears homeostatically

controlled118, 119, perhaps reflecting the situation in the normal population. In cancer patients the

cfDNA levels are clearly changed and therefore homeostatic regulation of cfDNA cannot be assumed.

Studies with repeat sampling at the same point in time and hourly sampling over a day are needed

to further understand this. Work is also needed to understand the best time of day to sample cfDNA.

With these results it should be possible to determine the number and timing of samples required for

testing and the possible variation of individual cfDNA levels.

Finding a role for cfDNA in staging cancers and predicting prognosis is hopeful, in what is a clinically

challenging area. There are some promising studies already published98, 106, 114, 159, 178, 273, 343-346.

However, in common with the diagnostic caveats above, further systematic study of cfDNA patterns

in patients with pre-cancerous changes through to invasive and metastatic disease is required.

Analysis of individual cfDNA levels at multiple time points through the disease stages will be needed

to confirm and validate this. The confirmation of these findings will also serve to validate the cfDNA

diagnostic approach.

Oesophageal dysplasia is a relatively rare finding with agreement on pathological grading and

staging being challenging319, 320. Particularly in this disease longitudinal follow up over 5-10 years will

be important to confirm the initial grade and stage which informed the cfDNA studies. The average

follow up in these patients for this thesis is 276 days and this may not be long enough.

Page 130: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

130

Perhaps even before these studies are undertaken work to confirm processing outcomes and

process timings is needed so that standardisation of techniques can occur. There has been some

work towards this153, 309, 310.

Page 131: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

131

Example of experimental amplification curves - Line 1 79bp

Page 132: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-132

Raw data - sample set up line 1 79bp

Block

Type

96fast

Chemistry

SYBR_GREEN

Experiment File Name

C:\Applied Biosystems\7500\experiments\experiments\Line 1 79\experiment results.eds

Experiment Run End Time

2010-03-08 04:58:43 AM GMT

Instrument Type

sds7500fast

Passive Reference

ROX

Page 133: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-133

Well

Sample

Name Sample Color Target Name Target Color Task Quantity

A1

Line 1 79 RGB(244,165,230) NTC

A2

Line 1 79 RGB(244,165,230) NTC

A3

Line 1 79 RGB(244,165,230) NTC

A4

Line 1 79 RGB(244,165,230) STANDARD 0.25

A5

Line 1 79 RGB(244,165,230) STANDARD 0.25

A6

Line 1 79 RGB(244,165,230) STANDARD 0.25

A7

Line 1 79 RGB(244,165,230) STANDARD 0.0625

A8

Line 1 79 RGB(244,165,230) STANDARD 0.0625

A9

Line 1 79 RGB(244,165,230) STANDARD 0.0625

A10

Line 1 79 RGB(244,165,230) STANDARD 0.015625

A11

Line 1 79 RGB(244,165,230) STANDARD 0.015625

Page 134: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-134

A12

Line 1 79 RGB(244,165,230) STANDARD 0.015625

B1

Line 1 79 RGB(244,165,230) STANDARD 0.0039063

B2

Line 1 79 RGB(244,165,230) STANDARD 0.0039063

B3

Line 1 79 RGB(244,165,230) STANDARD 0.0039063

B4

Line 1 79 RGB(244,165,230) STANDARD 0.0009766

B5

Line 1 79 RGB(244,165,230) STANDARD 0.0009766

B6

Line 1 79 RGB(244,165,230) STANDARD 0.0009766

B7 Sample 1 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

B8 Sample 1 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

B9 Sample 1 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

B10 Sample 2 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

B11 Sample 2 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

B12 Sample 2 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

Page 135: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-135

C1 Sample 3 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

C2 Sample 3 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

C3 Sample 3 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

C4 Sample 4 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

C5 Sample 4 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

C6 Sample 4 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

C7 Sample 5 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

C8 Sample 5 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

C9 Sample 5 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

C10 Sample 6 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

C11 Sample 6 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

C12 Sample 6 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

D1 Sample 7 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

Page 136: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-136

D2 Sample 7 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

D3 Sample 7 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

D4 Sample 8 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

D5 Sample 8 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

D6 Sample 8 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

D7 Sample 9 RGB(96,255,160) Line 1 79 RGB(244,165,230) UNKNOWN

D8 Sample 9 RGB(96,255,160) Line 1 79 RGB(244,165,230) UNKNOWN

D9 Sample 9 RGB(96,255,160) Line 1 79 RGB(244,165,230) UNKNOWN

D10 Sample 10 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

D11 Sample 10 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

D12 Sample 10 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

E1 Sample 11 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

E2 Sample 11 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

Page 137: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-137

E3 Sample 11 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

E4 Sample 12 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

E5 Sample 12 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

E6 Sample 12 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

E7 Sample 13 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

E8 Sample 13 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

E9 Sample 13 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

E10 Sample 14 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

E11 Sample 14 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

E12 Sample 14 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

F1 Sample 15 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

F2 Sample 15 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

F3 Sample 15 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

Page 138: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-138

F4 Sample 16 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

F5 Sample 16 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

F6 Sample 16 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

F7 Sample 17 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

F8 Sample 17 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

F9 Sample 17 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

F10 Sample 18 RGB(96,255,160) Line 1 79 RGB(244,165,230) UNKNOWN

F11 Sample 18 RGB(96,255,160) Line 1 79 RGB(244,165,230) UNKNOWN

F12 Sample 18 RGB(96,255,160) Line 1 79 RGB(244,165,230) UNKNOWN

G1 Sample 19 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

G2 Sample 19 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

G3 Sample 19 RGB(132,193,241) Line 1 79 RGB(244,165,230) UNKNOWN

G4 Sample 20 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

Page 139: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-139

G5 Sample 20 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

G6 Sample 20 RGB(168,255,222) Line 1 79 RGB(244,165,230) UNKNOWN

G7 Sample 21 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

G8 Sample 21 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

G9 Sample 21 RGB(223,221,142) Line 1 79 RGB(244,165,230) UNKNOWN

G10 Sample 22 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

G11 Sample 22 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

G12 Sample 22 RGB(247,255,168) Line 1 79 RGB(244,165,230) UNKNOWN

H1 Sample 23 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

H2 Sample 23 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

H3 Sample 23 RGB(180,255,0) Line 1 79 RGB(244,165,230) UNKNOWN

H4 Sample 24 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

H5 Sample 24 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

Page 140: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-140

H6 Sample 24 RGB(255,204,153) Line 1 79 RGB(244,165,230) UNKNOWN

H7 Sample 25 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

H8 Sample 25 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

H9 Sample 25 RGB(253,138,88) Line 1 79 RGB(244,165,230) UNKNOWN

H10 Sample 26 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

H11 Sample 26 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

H12 Sample 26 RGB(213,244,165) Line 1 79 RGB(244,165,230) UNKNOWN

Page 141: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-141

Raw data - results Line 1 79bp

Block Type

96fast

Chemistry

SYBR_GREEN

Experiment File Name

C:\Applied Biosystems\7500\experiments\experiments\Line 1 79.eds

Experiment Run End Time

2010-03-08 04:58:43 AM GMT

Instrument Type

sds7500fast

Passive

Reference

ROX

Page 142: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-142

Well

Sample

Name

Target

Name Task Reporter Cт Cт Mean Cт SD Quantity

Quantity

Mean Quantity SD

Automatic

Ct

Threshold

Ct

Threshold

Automatic

Baseline

Baseline

Start

Baseline

End Tm1

A1

Line 1

79 NTC SYBR Undetermined

TRUE 0.035783 TRUE 3 29 71.80394

A2

Line 1

79 NTC SYBR Undetermined

TRUE 0.035783 TRUE 3 29 71.6244

A3

Line 1

79 NTC SYBR Undetermined

TRUE 0.035783 TRUE 3 29 71.6244

A4

Line 1

79 STANDARD SYBR 16.56781197 16.62403 0.13666 0.25

TRUE 0.035783 TRUE 3 14 70.18819

A5

Line 1

79 STANDARD SYBR 16.77983665 16.62403 0.13666 0.25

TRUE 0.035783 TRUE 3 14 70.18819

A6

Line 1

79 STANDARD SYBR 16.52445221 16.62403 0.13666 0.25

TRUE 0.035783 TRUE 3 14 70.36771

A7

Line 1

79 STANDARD SYBR 18.64361763 18.71006 0.057687 0.0625

TRUE 0.035783 TRUE 3 16 70.36771

A8

Line 1

79 STANDARD SYBR 18.73918533 18.71006 0.057687 0.0625

TRUE 0.035783 TRUE 3 16 70.36771

Page 143: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-143

A9

Line 1

79 STANDARD SYBR 18.7473793 18.71006 0.057687 0.0625

TRUE 0.035783 TRUE 3 16 70.18819

A10

Line 1

79 STANDARD SYBR 20.71775818 20.80297 0.077126 0.015625

TRUE 0.035783 TRUE 3 18 70.00866

A11

Line 1

79 STANDARD SYBR 20.86799622 20.80297 0.077126 0.015625

TRUE 0.035783 TRUE 3 18 70.00866

A12

Line 1

79 STANDARD SYBR 20.82315445 20.80297 0.077126 0.015625

TRUE 0.035783 TRUE 3 18 70.00866

B1

Line 1

79 STANDARD SYBR 22.94385529 22.85642 0.078897 0.003906

TRUE 0.035783 TRUE 3 21 69.82913

B2

Line 1

79 STANDARD SYBR 22.79053688 22.85642 0.078897 0.003906

TRUE 0.035783 TRUE 3 20 70.00866

B3

Line 1

79 STANDARD SYBR 22.83488083 22.85642 0.078897 0.003906

TRUE 0.035783 TRUE 3 20 70.18819

B4

Line 1

79 STANDARD SYBR 24.94140053 24.93026 0.009655 0.000977

TRUE 0.035783 TRUE 3 22 70.18819

B5

Line 1

79 STANDARD SYBR 24.92507553 24.93026 0.009655 0.000977

TRUE 0.035783 TRUE 3 22 70.18819

Page 144: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-144

B6

Line 1

79 STANDARD SYBR 24.92430687 24.93026 0.009655 0.000977

TRUE 0.035783 TRUE 3 23 70.36771

B7

Sample

1

Line 1

79 UNKNOWN SYBR 21.09013176 21.09132 0.007957 0.012741 0.012731317 6.76101E-05 TRUE 0.035783 TRUE 3 19 70.36771

B8

Sample

1

Line 1

79 UNKNOWN SYBR 21.09980583 21.09132 0.007957 0.012659 0.012731317 6.76101E-05 TRUE 0.035783 TRUE 3 18 70.18819

B9

Sample

1

Line 1

79 UNKNOWN SYBR 21.08402634 21.09132 0.007957 0.012793 0.012731317 6.76101E-05 TRUE 0.035783 TRUE 3 19 70.18819

B10

Sample

2

Line 1

79 UNKNOWN SYBR 21.11586952 21.23754 0.197495 0.012524 0.011612135 0.001469633 TRUE 0.035783 TRUE 3 19 70.18819

B11

Sample

2

Line 1

79 UNKNOWN SYBR 21.13134193 21.23754 0.197495 0.012395 0.011612135 0.001469633 TRUE 0.035783 TRUE 3 18 70.00866

B12

Sample

2

Line 1

79 UNKNOWN SYBR 21.46541405 21.23754 0.197495 0.009917 0.011612135 0.001469633 TRUE 0.035783 TRUE 3 19 69.82913

C1

Sample

3

Line 1

79 UNKNOWN SYBR 21.10075378 21.07293 0.128714 0.012651 0.012920561 0.001124214 TRUE 0.035783 TRUE 3 19 70.00866

C2

Sample

3

Line 1

79 UNKNOWN SYBR 21.18545914 21.07293 0.128714 0.011955 0.012920561 0.001124214 TRUE 0.035783 TRUE 3 19 70.00866

Page 145: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-145

C3

Sample

3

Line 1

79 UNKNOWN SYBR 20.93258095 21.07293 0.128714 0.014155 0.012920561 0.001124214 TRUE 0.035783 TRUE 3 19 70.18819

C4

Sample

4

Line 1

79 UNKNOWN SYBR 22.65699768 22.74297 0.093927 0.004475 0.00423065 0.000263387 TRUE 0.035783 TRUE 3 20 70.36771

C5

Sample

4

Line 1

79 UNKNOWN SYBR 22.72868156 22.74297 0.093927 0.004266 0.00423065 0.000263387 TRUE 0.035783 TRUE 3 20 70.18819

C6

Sample

4

Line 1

79 UNKNOWN SYBR 22.84321594 22.74297 0.093927 0.003952 0.00423065 0.000263387 TRUE 0.035783 TRUE 3 20 70.36771

C7

Sample

5

Line 1

79 UNKNOWN SYBR 22.53641319 22.53805 0.036776 0.00485 0.00484575 0.000118905 TRUE 0.035783 TRUE 3 20 70.18819

C8

Sample

5

Line 1

79 UNKNOWN SYBR 22.50211143 22.53805 0.036776 0.004962 0.00484575 0.000118905 TRUE 0.035783 TRUE 3 20 70.18819

C9

Sample

5

Line 1

79 UNKNOWN SYBR 22.57560921 22.53805 0.036776 0.004725 0.00484575 0.000118905 TRUE 0.035783 TRUE 3 20 70.18819

C10

Sample

6

Line 1

79 UNKNOWN SYBR 21.93924141 22.03871 0.08617 0.007227 0.006769878 0.00039581 TRUE 0.035783 TRUE 3 19 70.18819

C11

Sample

6

Line 1

79 UNKNOWN SYBR 22.09057999 22.03871 0.08617 0.006532 0.006769878 0.00039581 TRUE 0.035783 TRUE 3 20 70.00866

Page 146: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-146

C12

Sample

6

Line 1

79 UNKNOWN SYBR 22.08631134 22.03871 0.08617 0.006551 0.006769878 0.00039581 TRUE 0.035783 TRUE 3 20 70.00866

D1

Sample

7

Line 1

79 UNKNOWN SYBR 22.06984711 22.14585 0.065967 0.006623 0.006299497 0.000280888 TRUE 0.035783 TRUE 3 19 70.00866

D2

Sample

7

Line 1

79 UNKNOWN SYBR 22.17944908 22.14585 0.065967 0.006156 0.006299497 0.000280888 TRUE 0.035783 TRUE 3 20 70.18819

D3

Sample

7

Line 1

79 UNKNOWN SYBR 22.1882534 22.14585 0.065967 0.00612 0.006299497 0.000280888 TRUE 0.035783 TRUE 3 20 70.18819

D4

Sample

8

Line 1

79 UNKNOWN SYBR 22.7189579 22.88522 0.177087 0.004293 0.003860029 0.000451203 TRUE 0.035783 TRUE 3 20 70.18819

D5

Sample

8

Line 1

79 UNKNOWN SYBR 23.07144165 22.88522 0.177087 0.003393 0.003860029 0.000451203 TRUE 0.035783 TRUE 3 21 70.18819

D6

Sample

8

Line 1

79 UNKNOWN SYBR 22.86526489 22.88522 0.177087 0.003894 0.003860029 0.000451203 TRUE 0.035783 TRUE 3 21 70.36771

D7

Sample

9

Line 1

79 UNKNOWN SYBR 21.90338516 21.92776 0.06958 0.007402 0.007287627 0.000334898 TRUE 0.035783 TRUE 3 19 70.36771

D8

Sample

9

Line 1

79 UNKNOWN SYBR 22.00624657 21.92776 0.06958 0.006911 0.007287627 0.000334898 TRUE 0.035783 TRUE 3 20 70.36771

Page 147: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-147

D9

Sample

9

Line 1

79 UNKNOWN SYBR 21.87364578 21.92776 0.06958 0.00755 0.007287627 0.000334898 TRUE 0.035783 TRUE 3 19 70.18819

D10

Sample

10

Line 1

79 UNKNOWN SYBR 20.88681412 20.96103 0.088881 0.014594 0.013904788 0.000815655 TRUE 0.035783 TRUE 3 18 70.18819

D11

Sample

10

Line 1

79 UNKNOWN SYBR 20.93674278 20.96103 0.088881 0.014116 0.013904788 0.000815655 TRUE 0.035783 TRUE 3 18 70.18819

D12

Sample

10

Line 1

79 UNKNOWN SYBR 21.05952835 20.96103 0.088881 0.013004 0.013904788 0.000815655 TRUE 0.035783 TRUE 3 19 70.00866

E1

Sample

11

Line 1

79 UNKNOWN SYBR 22.44207191 22.4341 0.007133 0.005165 0.005193074 2.47115E-05 TRUE 0.035783 TRUE 3 20 70.00866

E2

Sample

11

Line 1

79 UNKNOWN SYBR 22.42832375 22.4341 0.007133 0.005213 0.005193074 2.47115E-05 TRUE 0.035783 TRUE 3 20 70.18819

E3

Sample

11

Line 1

79 UNKNOWN SYBR 22.43190193 22.4341 0.007133 0.005201 0.005193074 2.47115E-05 TRUE 0.035783 TRUE 3 20 70.18819

E4

Sample

12

Line 1

79 UNKNOWN SYBR 22.91010857 22.9426 0.044894 0.003779 0.003698872 0.000110023 TRUE 0.035783 TRUE 3 20 70.18819

E5

Sample

12

Line 1

79 UNKNOWN SYBR 22.99382782 22.9426 0.044894 0.003573 0.003698872 0.000110023 TRUE 0.035783 TRUE 3 21 70.18819

Page 148: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-148

E6

Sample

12

Line 1

79 UNKNOWN SYBR 22.92386436 22.9426 0.044894 0.003744 0.003698872 0.000110023 TRUE 0.035783 TRUE 3 20 70.36771

E7

Sample

13

Line 1

79 UNKNOWN SYBR 21.27283478 21.15307 0.191925 0.011278 0.01228549 0.001627657 TRUE 0.035783 TRUE 3 19 70.36771

E8

Sample

13

Line 1

79 UNKNOWN SYBR 21.25467682 21.15307 0.191925 0.011415 0.01228549 0.001627657 TRUE 0.035783 TRUE 3 19 70.36771

E9

Sample

13

Line 1

79 UNKNOWN SYBR 20.93170357 21.15307 0.191925 0.014163 0.01228549 0.001627657 TRUE 0.035783 TRUE 3 18 70.36771

E10

Sample

14

Line 1

79 UNKNOWN SYBR 22.49219704 22.53241 0.082388 0.004995 0.004867907 0.000263599 TRUE 0.035783 TRUE 3 20 70.18819

E11

Sample

14

Line 1

79 UNKNOWN SYBR 22.47784615 22.53241 0.082388 0.005044 0.004867907 0.000263599 TRUE 0.035783 TRUE 3 20 70.18819

E12

Sample

14

Line 1

79 UNKNOWN SYBR 22.6271801 22.53241 0.082388 0.004565 0.004867907 0.000263599 TRUE 0.035783 TRUE 3 20 70.00866

F1

Sample

15

Line 1

79 UNKNOWN SYBR 21.77561569 21.74323 0.061909 0.008061 0.008242189 0.000344685 TRUE 0.035783 TRUE 3 19 69.82913

F2

Sample

15

Line 1

79 UNKNOWN SYBR 21.78224182 21.74323 0.061909 0.008026 0.008242189 0.000344685 TRUE 0.035783 TRUE 3 19 70.00866

Page 149: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-149

F3

Sample

15

Line 1

79 UNKNOWN SYBR 21.67185211 21.74323 0.061909 0.00864 0.008242189 0.000344685 TRUE 0.035783 TRUE 3 19 70.18819

F4

Sample

16

Line 1

79 UNKNOWN SYBR 21.57563591 21.65517 0.08086 0.009213 0.008744922 0.000471443 TRUE 0.035783 TRUE 3 19 70.18819

F5

Sample

16

Line 1

79 UNKNOWN SYBR 21.65259171 21.65517 0.08086 0.008752 0.008744922 0.000471443 TRUE 0.035783 TRUE 3 19 70.18819

F6

Sample

16

Line 1

79 UNKNOWN SYBR 21.73729324 21.65517 0.08086 0.00827 0.008744922 0.000471443 TRUE 0.035783 TRUE 3 19 70.18819

F7

Sample

17

Line 1

79 UNKNOWN SYBR 22.34988594 22.39887 0.059147 0.005493 0.005319388 0.00020845 TRUE 0.035783 TRUE 3 20 70.18819

F8

Sample

17

Line 1

79 UNKNOWN SYBR 22.38216019 22.39887 0.059147 0.005376 0.005319388 0.00020845 TRUE 0.035783 TRUE 3 20 70.18819

F9

Sample

17

Line 1

79 UNKNOWN SYBR 22.46458244 22.39887 0.059147 0.005088 0.005319388 0.00020845 TRUE 0.035783 TRUE 3 20 70.18819

F10

Sample

18

Line 1

79 UNKNOWN SYBR 22.10458755 22.25439 0.130002 0.006471 0.005870172 0.000521499 TRUE 0.035783 TRUE 3 19 70.18819

F11

Sample

18

Line 1

79 UNKNOWN SYBR 22.3209362 22.25439 0.130002 0.005601 0.005870172 0.000521499 TRUE 0.035783 TRUE 3 20 70.18819

Page 150: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-150

F12

Sample

18

Line 1

79 UNKNOWN SYBR 22.33764648 22.25439 0.130002 0.005539 0.005870172 0.000521499 TRUE 0.035783 TRUE 3 20 70.00866

G1

Sample

19

Line 1

79 UNKNOWN SYBR 22.1571579 22.14281 0.027467 0.006248 0.006308919 0.000116324 TRUE 0.035783 TRUE 3 20 69.82913

G2

Sample

19

Line 1

79 UNKNOWN SYBR 22.11113358 22.14281 0.027467 0.006443 0.006308919 0.000116324 TRUE 0.035783 TRUE 3 20 70.00866

G3

Sample

19

Line 1

79 UNKNOWN SYBR 22.16012001 22.14281 0.027467 0.006236 0.006308919 0.000116324 TRUE 0.035783 TRUE 3 20 70.00866

G4

Sample

20

Line 1

79 UNKNOWN SYBR 20.70654106 20.77372 0.058243 0.016461 0.01574721 0.000619154 TRUE 0.035783 TRUE 3 18 70.00866

G5

Sample

20

Line 1

79 UNKNOWN SYBR 20.80999947 20.77372 0.058243 0.015362 0.01574721 0.000619154 TRUE 0.035783 TRUE 3 18 70.00866

G6

Sample

20

Line 1

79 UNKNOWN SYBR 20.80462646 20.77372 0.058243 0.015418 0.01574721 0.000619154 TRUE 0.035783 TRUE 3 18 70.18819

G7

Sample

21

Line 1

79 UNKNOWN SYBR 22.96528816 23.01903 0.05269 0.003642 0.003515227 0.000123808 TRUE 0.035783 TRUE 3 20 70.18819

G8

Sample

21

Line 1

79 UNKNOWN SYBR 23.02118683 23.01903 0.05269 0.003509 0.003515227 0.000123808 TRUE 0.035783 TRUE 3 20 70.18819

Page 151: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-151

G9

Sample

21

Line 1

79 UNKNOWN SYBR 23.07060242 23.01903 0.05269 0.003395 0.003515227 0.000123808 TRUE 0.035783 TRUE 3 21 70.00866

G10

Sample

22

Line 1

79 UNKNOWN SYBR 22.87696838 22.88319 0.023635 0.003863 0.00384775 6.05555E-05 TRUE 0.035783 TRUE 3 20 70.18819

G11

Sample

22

Line 1

79 UNKNOWN SYBR 22.86328697 22.88319 0.023635 0.003899 0.00384775 6.05555E-05 TRUE 0.035783 TRUE 3 20 70.00866

G12

Sample

22

Line 1

79 UNKNOWN SYBR 22.9093132 22.88319 0.023635 0.003781 0.00384775 6.05555E-05 TRUE 0.035783 TRUE 3 20 70.00866

H1

Sample

23

Line 1

79 UNKNOWN SYBR 19.9601059 20.09171 0.134209 0.027099 0.02488552 0.002222565 TRUE 0.035783 TRUE 3 17 69.82913

H2

Sample

23

Line 1

79 UNKNOWN SYBR 20.08666039 20.09171 0.134209 0.024903 0.02488552 0.002222565 TRUE 0.035783 TRUE 3 17 69.82913

H3

Sample

23

Line 1

79 UNKNOWN SYBR 20.2283802 20.09171 0.134209 0.022654 0.02488552 0.002222565 TRUE 0.035783 TRUE 3 17 69.82913

H4

Sample

24

Line 1

79 UNKNOWN SYBR 21.16566277 21.32312 0.165309 0.012115 0.010949518 0.001198132 TRUE 0.035783 TRUE 3 19 69.82913

H5

Sample

24

Line 1

79 UNKNOWN SYBR 21.30839348 21.32312 0.165309 0.011013 0.010949518 0.001198132 TRUE 0.035783 TRUE 3 19 70.00866

Page 152: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

7-152

H6

Sample

24

Line 1

79 UNKNOWN SYBR 21.49529648 21.32312 0.165309 0.009721 0.010949518 0.001198132 TRUE 0.035783 TRUE 3 19 70.00866

H7

Sample

25

Line 1

79 UNKNOWN SYBR 22.63315392 22.65711 0.024832 0.004547 0.004474884 7.41448E-05 TRUE 0.035783 TRUE 3 20 70.00866

H8

Sample

25

Line 1

79 UNKNOWN SYBR 22.65543175 22.65711 0.024832 0.004479 0.004474884 7.41448E-05 TRUE 0.035783 TRUE 3 20 70.00866

H9

Sample

25

Line 1

79 UNKNOWN SYBR 22.68273354 22.65711 0.024832 0.004399 0.004474884 7.41448E-05 TRUE 0.035783 TRUE 3 20 70.00866

H10

Sample

26

Line 1

79 UNKNOWN SYBR 23.22262764 23.24241 0.15147 0.003067 0.003037081 0.000303832 TRUE 0.035783 TRUE 3 20 70.00866

H11

Sample

26

Line 1

79 UNKNOWN SYBR 23.4027977 23.24241 0.15147 0.002719 0.003037081 0.000303832 TRUE 0.035783 TRUE 3 21 69.82913

H12

Sample

26

Line 1

79 UNKNOWN SYBR 23.10180092 23.24241 0.15147 0.003325 0.003037081 0.000303832 TRUE 0.035783 TRUE 3 20 69.82913

Page 153: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

153

References

1. Pohl H, and Welch HG. The role of overdiagnosis and reclassification in the marked increase of esophageal adenocarcinoma incidence. J Natl Cancer Inst 2005;97:142-6.

2. UK CR. Cancer stats. http://wwwcancerresearchukorg/ 2010. 3. Steevens J, Botterweck AA, Dirx MJ, et al. Trends in incidence of oesophageal and stomach

cancer subtypes in Europe. Eur J Gastroenterol Hepatol 2010;22:669-78. 4. Qu GZ, Grundy PE, Narayan A, et al. Frequent hypomethylation in Wilms tumors of

pericentromeric DNA in chromosomes 1 and 16. Cancer Genet Cytogenet 1999;109:34-9. 5. Fraga MF, Ballestar E, Villar-Garea A, et al. Loss of acetylation at Lys16 and trimethylation at

Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet 2005;37:391-400. 6. Skobe M, and Fusenig NE. Tumorigenic conversion of immortal human keratinocytes through

stromal cell activation. Proc Natl Acad Sci U S A 1998;95:1050-5. 7. Kinzler KW, and Vogelstein B. Landscaping the cancer terrain. Science 1998;280:1036-7. 8. Hudson JD, Shoaibi MA, Maestro R, et al. A proinflammatory cytokine inhibits p53 tumor

suppressor activity. J Exp Med 1999;190:1375-82. 9. Vogelstein B, Fearon ER, Hamilton SR, et al. Genetic alterations during colorectal-tumor

development. N Engl J Med 1988;319:525-32. 10. Cho KR, and Vogelstein B. Genetic alterations in the adenoma--carcinoma sequence. Cancer

1992;70:1727-31. 11. Fearon ER, and Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990;61:759-

67. 12. Hermsen M, Postma C, Baak J, et al. Colorectal adenoma to carcinoma progression follows

multiple pathways of chromosomal instability. Gastroenterology 2002;123:1109-19. 13. Kudo S, Lambert R, Allen JI, et al. Nonpolypoid neoplastic lesions of the colorectal mucosa.

Gastrointest Endosc 2008;68:S3-47. 14. Gille JJP, Hogervorst FBL, Pals G, et al. Genomic deletions of MSH2 and MLH1 in colorectal

cancer families detected by a novel mutation detection approach. Br J Cancer 2002;87:892-7. 15. Gill S, Lindor NM, Burgart LJ, et al. Isolated Loss of PMS2 Expression in Colorectal Cancers:

Frequency, Patient Age, and Familial Aggregation. Clinical Cancer Research 2005;11:6466-71. 16. Kurzawski G, Suchy J, Debniak T, et al. Importance of microsatellite instability (MSI) in

colorectal cancer: MSI as a diagnostic tool. Ann Oncol 2004;15 Suppl 4:iv283-4. 17. Kinzler KW, and Vogelstein B. Lessons from hereditary colorectal cancer. Cell 1996;87:159-70. 18. Kambara T, Simms LA, Whitehall VL, et al. BRAF mutation is associated with DNA methylation

in serrated polyps and cancers of the colorectum. Gut 2004;53:1137-44. 19. Barrett NR. Chronic peptic ulcer of the oesophagus and 'oesophagitis'. Br J Surg 1950;38:175-

82. 20. Kazumori H, Ishihara S, Rumi MA, et al. Bile acids directly augment caudal related homeobox

gene Cdx2 expression in oesophageal keratinocytes in Barrett's epithelium. Gut 2006;55:16-25.

21. Wong NA, Wilding J, Bartlett S, et al. CDX1 is an important molecular mediator of Barrett's metaplasia. Proc Natl Acad Sci U S A 2005;102:7565-70.

22. Eda A, Osawa H, Satoh K, et al. Aberrant expression of CDX2 in Barrett's epithelium and inflammatory esophageal mucosa. J Gastroenterol 2003;38:14-22.

23. Armstrong D, Rytina ER, Murphy GM, et al. Gastric mucosal toxicity of duodenal juice constituents in the rat. Acute studies using ex vivo rat gastric chamber model. Dig Dis Sci 1994;39:327-39.

24. Vaezi MF, and Richter JE. Synergism of acid and duodenogastroesophageal reflux in complicated Barrett's esophagus. Surgery 1995;117:699-704.

Page 154: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

154

25. Caldwell MT, Lawlor P, Byrne PJ, et al. Ambulatory oesophageal bile reflux monitoring in Barrett's oesophagus. Br J Surg 1995;82:657-60.

26. Nehra D, Howell P, Williams CP, et al. Toxic bile acids in gastro-oesophageal reflux disease: influence of gastric acidity. Gut 1999;44:598-602.

27. Lillemoe KD, Johnson LF, and Harmon JW. Alkaline esophagitis: a comparison of the ability of components of gastroduodenal contents to injure the rabbit esophagus. Gastroenterology 1983;85:621-8.

28. Niemantsverdriet EC, Timmer R, Breumelhof R, et al. The roles of excessive gastro-oesophageal reflux, disordered oesophageal motility and decreased mucosal sensitivity in the pathogenesis of Barrett's oesophagus. Eur J Gastroenterol Hepatol 1997;9:515-9.

29. Wild CP, and Hardie LJ. Reflux, Barrett's oesophagus and adenocarcinoma: burning questions. Nat Rev Cancer 2003;3:676-84.

30. Qiao D, Gaitonde SV, Qi W, et al. Deoxycholic acid suppresses p53 by stimulating proteasome-mediated p53 protein degradation. Carcinogenesis 2001;22:957-64.

31. Wilson KT, Fu S, Ramanujam KS, et al. Increased expression of inducible nitric oxide synthase and cyclooxygenase-2 in Barrett's esophagus and associated adenocarcinomas. Cancer Res 1998;58:2929-34.

32. Buttar NS, Wang KK, Anderson MA, et al. The effect of selective cyclooxygenase-2 inhibition in Barrett's esophagus epithelium: an in vitro study. J Natl Cancer Inst 2002;94:422-9.

33. Zhang F, Altorki NK, Wu YC, et al. Duodenal reflux induces cyclooxygenase-2 in the esophageal mucosa of rats: evidence for involvement of bile acids. Gastroenterology 2001;121:1391-9.

34. Souza RF, Shewmake K, Terada LS, et al. Acid exposure activates the mitogen-activated protein kinase pathways in Barrett's esophagus. Gastroenterology 2002;122:299-307.

35. Tselepis C, Morris CD, Wakelin D, et al. Upregulation of the oncogene c-myc in Barrett's adenocarcinoma: induction of c-myc by acidified bile acid in vitro. Gut 2003;52:174-80.

36. Theisen J, Nehra D, Citron D, et al. Suppression of gastric acid secretion in patients with gastroesophageal reflux disease results in gastric bacterial overgrowth and deconjugation of bile acids. J Gastrointest Surg 2000;4:50-4.

37. Haigh CR, Attwood SE, Thompson DG, et al. Gastrin induces proliferation in Barrett's metaplasia through activation of the CCK2 receptor. Gastroenterology 2003;124:615-25.

38. Katzka DA, and Castell DO. Successful elimination of reflux symptoms does not insure adequate control of acid reflux in patients with Barrett's esophagus. Am J Gastroenterol 1994;89:989-91.

39. Sontag SJ. The medical management of reflux esophagitis. Role of antacids and acid inhibition. Gastroenterol Clin North Am 1990;19:683-712.

40. Ouatu-Lascar R, and Triadafilopoulos G. Complete elimination of reflux symptoms does not guarantee normalization of intraesophageal acid reflux in patients with Barrett's esophagus. Am J Gastroenterol 1998;93:711-6.

41. Goldstein SR, Yang GY, Curtis SK, et al. Development of esophageal metaplasia and adenocarcinoma in a rat surgical model without the use of a carcinogen. Carcinogenesis 1997;18:2265-70.

42. Kawaura Y, Tatsuzawa Y, Wakabayashi T, et al. Immunohistochemical study of p53, c-erbB-2, and PCNA in barrett's esophagus with dysplasia and adenocarcinoma arising from experimental acid or alkaline reflux model. J Gastroenterol 2001;36:595-600.

43. Jankowski J, Hopwood D, Pringle R, et al. Increased expression of epidermal growth factor receptors in Barrett's esophagus associated with alkaline reflux: a putative model for carcinogenesis. Am J Gastroenterol 1993;88:402-8.

44. Hanahan D, and Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70. 45. Hanahan D, and Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-

74.

Page 155: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

155

46. Gormally E, Vineis P, Matullo G, 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.

47. Cairns SR, Scholefield JH, Steele RJ, et al. Guidelines for colorectal cancer screening and surveillance in moderate and high risk groups (update from 2002). Gut 2010;59:666-89.

48. Playford RJ. New British Society of Gastroenterology (BSG) guidelines for the diagnosis and management of Barrett's oesophagus. Gut 2006;55:442.

49. Allum WH, Griffin SM, Watson A, et al. Guidelines for the management of oesophageal and gastric cancer. Gut 2002;50 Suppl 5:v1-23.

50. Goddard AF, McIntyre AS, and Scott BB. Guidelines for the management of iron deficiency anaemia. British Society of Gastroenterology. Gut 2000;46 Suppl 3-4:IV1-IV5.

51. Locker GY, Hamilton S, Harris J, et al. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol 2006;24:5313-27.

52. guidelines N. Guidance on Cancer Services Improving Outcomes in Colorectal Cancers Manual Update. http://guidanceniceorguk/CSGCC/Guidance/pdf/English 2004.

53. Weller D. Diagnosing colorectal cancer in primary care. BMJ 2010;340:c1714. 54. Richards MA. The size of the prize for earlier diagnosis of cancer in England. Br J Cancer

2009;101 Suppl 2:S125-9. 55. http://www.cancerscreening.nhs.uk/bowel/. Report to the English Bowel Cancer Screening

Working Group September 2004. On Line 2010. 56. van Dam L, Hol L, de Bekker-Grob EW, et al. What determines individuals' preferences for

colorectal cancer screening programmes? A discrete choice experiment. Eur J Cancer 2010;46:150-9.

57. Vernon SW. Participation in colorectal cancer screening: a review. J Natl Cancer Inst 1997;89:1406-22.

58. Parente F, Marino B, DeVecchi N, et al. Faecal occult blood test-based screening programme with high compliance for colonoscopy has a strong clinical impact on colorectal cancer. Br J Surg 2009;96:533-40.

59. Ore L, Hagoel L, Lavi I, et al. Screening with faecal occult blood test (FOBT) for colorectal cancer: assessment of two methods that attempt to improve compliance. Eur J Cancer Prev 2001;10:251-6.

60. Federici A, Giorgi Rossi P, Borgia P, et al. The immunochemical faecal occult blood test leads to higher compliance than the guaiac for colorectal cancer screening programmes: a cluster randomized controlled trial. J Med Screen 2005;12:83-8.

61. Fenocchi E, Martinez L, Tolve J, et al. Screening for colorectal cancer in Uruguay with an immunochemical faecal occult blood test. Eur J Cancer Prev 2006;15:384-90.

62. Yang H, Ge Z, Dai J, et al. Effectiveness of the Immunofecal Occult Blood Test for Colorectal Cancer Screening in a Large Population. Dig Dis Sci 2010.

63. Chen LS, Liao CS, Chang SH, et al. Cost-effectiveness analysis for determining optimal cut-off of immunochemical faecal occult blood test for population-based colorectal cancer screening (KCIS 16). J Med Screen 2007;14:191-9.

64. Li S, Wang H, Hu J, et al. New immunochemical fecal occult blood test with two-consecutive stool sample testing is a cost-effective approach for colon cancer screening: results of a prospective multicenter study in Chinese patients. Int J Cancer 2006;118:3078-83.

65. van Rossum LG, van Rijn AF, Laheij RJ, et al. Cutoff value determines the performance of a semi-quantitative immunochemical faecal occult blood test in a colorectal cancer screening programme. Br J Cancer 2009;101:1274-81.

66. Atkin WS, Edwards R, Kralj-Hans I, et al. Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: a multicentre randomised controlled trial. Lancet 2010;375:1624-33.

67. Atkin WS, Hart A, Edwards R, et al. Uptake, yield of neoplasia, and adverse effects of flexible sigmoidoscopy screening. Gut 1998;42:560-5.

Page 156: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

156

68. Cameron AJ, Zinsmeister AR, Ballard DJ, et al. Prevalence of columnar-lined (Barrett's) esophagus. Comparison of population-based clinical and autopsy findings. Gastroenterology 1990;99:918-22.

69. Kadri SR, Lao-Sirieix P, O'Donovan M, et al. Acceptability and accuracy of a non-endoscopic screening test for Barrett's oesophagus in primary care: cohort study. BMJ 2010;341:c4372.

70. Grimm I, Shaheen N, and Bozymski EM. Surveillance for Barrett's esophagus: Are we saving lives? Gastroenterology 1997;112:661-2.

71. Spechler SJ. Barrett's esophagus: an overrated cancer risk factor. Gastroenterology 2000;119:587-9.

72. Spechler SJ, Robbins AH, Rubins HB, et al. Adenocarcinoma and Barrett's esophagus. An overrated risk? Gastroenterology 1984;87:927-33.

73. Sharma P, and Sidorenko EI. Are screening and surveillance for Barrett's oesophagus really worthwhile? Gut 2005;54 Suppl 1:i27-32.

74. Corley DA, Levin TR, Habel LA, et al. Surveillance and survival in Barrett's adenocarcinomas: a population-based study. Gastroenterology 2002;122:633-40.

75. Shaheen NJ, Crosby MA, Bozymski EM, et al. Is there publication bias in the reporting of cancer risk in Barrett's esophagus? Gastroenterology 2000;119:333-8.

76. Bhat S, Coleman HG, Yousef F, et al. Risk of Malignant Progression in Barrett's Esophagus Patients: Results from a Large Population-Based Study. J Natl Cancer Inst 2011;103:1049-57.

77. Wright TA, Gray MR, Morris AI, et al. Cost effectiveness of detecting Barrett's cancer. Gut 1996;39:574-9.

78. Inadomi JM. Surveillance in Barrett's esophagus: a failed premise. Keio J Med 2009;58:12-8. 79. Longcroft-Wheaton G, Duku M, Mead R, et al. Acetic acid spray is an effective tool for the

endoscopic detection of neoplasia in patients with Barrett's esophagus. Clin Gastroenterol Hepatol 2010;8:843-7.

80. Lieberman R. Evidence-based medical perspectives: the evolving role of PSA for early detection, monitoring of treatment response, and as a surrogate end point of efficacy for interventions in men with different clinical risk states for the prevention and progression of prostate cancer. Am J Ther 2004;11:501-6.

81. Levenson VV. Biomarkers for early detection of breast cancer: what, when, and where? Biochim Biophys Acta 2007;1770:847-56.

82. Ahlquist DA. Molecular detection of colorectal neoplasia. Gastroenterology 2010;138:2127-39.

83. http://www.cancerscreening.nhs.uk/bowel/. NHS Bowel Cancer Screening Programme. On Line 2010.

84. Van Duijnhoven HL, Van De Warenburg FJ, Willems RJ, et al. A comparison of ELISA assays as routine diagnostic test for detection of autoantibodies against extractable nuclear antigens. Clin Biochem 1999;32:179-83.

85. Boom R, Sol C, Weel J, et al. A highly sensitive assay for detection and quantitation of human cytomegalovirus DNA in serum and plasma by PCR and electrochemiluminescence. J Clin Microbiol 1999;37:1489-97.

86. Drew WL. Laboratory diagnosis of cytomegalovirus infection and disease in immunocompromised patients. Curr Opin Infect Dis 2007;20:408-11.

87. Arkel YS, Ku DH, Le P, et al. Comparison of a test for soluble fibrin polymer (TpP) with a standard quantitative ELISA for D-dimer in patients, without current thrombosis, who have cancer or renal disease. Thromb Haemost 2001;86:1127-8.

88. Mandel P MP. Les acides nucléiques du plasma sanquin chez l’homme. C R Acad Sci Paris 1948;142:241-3.

89. Ellinger J, Wittkamp V, Albers P, et al. Cell-free circulating DNA: diagnostic value in patients with testicular germ cell cancer. J Urol 2009;181:363-71.

Page 157: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

157

90. Liggett T, Melnikov A, Yi QL, et al. Differential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitis. Cancer 2010;116:1674-80.

91. Salbe C, Trevisiol C, Ferruzzi E, et al. Molecular detection of codon 12 K-RAS mutations in circulating DNA from serum of colorectal cancer patients. Int J Biol Markers 2000;15:300-7.

92. Bianchi DW. Circulating fetal DNA: its origin and diagnostic potential-a review. Placenta 2004;25 Suppl A:S93-S101.

93. Guan T, Guo XY, Ye CL, et al. [Analysis of circulating DNA level in the plasma of cervical cancer patients]. Nan Fang Yi Ke Da Xue Xue Bao 2008;28:1663-4, 7.

94. Tomita H, Ichikawa D, Sai S, et al. [Quantification of circulating plasma DNA fragments as tumor markers in patients with esophageal and gastric cancer]. Gan To Kagaku Ryoho 2007;34:1908-10.

95. Kolesnikova EV, Tamkovich SN, Bryzgunova OE, et al. Circulating DNA in the blood of gastric cancer patients. Ann N Y Acad Sci 2008;1137:226-31.

96. Ellinger J, Muller SC, Stadler TC, et al. The role of cell-free circulating DNA in the diagnosis and prognosis of prostate cancer. Urol Oncol 2009.

97. Ellinger J, Albers P, Perabo FG, et al. CpG island hypermethylation of cell-free circulating serum DNA in patients with testicular cancer. J Urol 2009;182:324-9.

98. An Q, Liu Y, Gao Y, et al. Detection of p16 hypermethylation in circulating plasma DNA of non-small cell lung cancer patients. Cancer Lett 2002;188:109-14.

99. Board RE, Wardley AM, Dixon JM, et al. Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer. Breast Cancer Res Treat 2010;120:461-7.

100. Board RE, Williams VS, Knight L, et al. Isolation and extraction of circulating tumor DNA from patients with small cell lung cancer. Ann N Y Acad Sci 2008;1137:98-107.

101. Boni L, Cassinotti E, Canziani M, et al. Free circulating DNA as possible tumour marker in colorectal cancer. Surg Oncol 2007;16 Suppl 1:S29-31.

102. Ma L, Liu FR, and Zhang SL. [Detection of circulating hypermethylated tumor-specific RASSF1A DNA in ovarian cancer patients]. Zhonghua Bing Li Xue Za Zhi 2005;34:785-7.

103. Moreira VG, Prieto B, Rodriguez JS, et al. Usefulness of cell-free plasma DNA, procalcitonin and C-reactive protein as markers of infection in febrile patients. Ann Clin Biochem 2010;47:253-8.

104. Bengtsson A, Nezlin R, Shoenfeld Y, et al. DNA levels in circulating immune complexes decrease at severe SLE flares-correlation with complement component C1q. J Autoimmun 1999;13:111-9.

105. Gahan PB. Circulating nucleic acids in plasma and serum: roles in diagnosis and prognosis in diabetes and cancer. Infect Disord Drug Targets 2008;8:100-8.

106. Andreadis C, Touloupidis S, Galaktidou G, et al. Serum CYFRA 21-1 in patients with invasive bladder cancer and its relevance as a tumor marker during chemotherapy. J Urol 2005;174:1771-5; discussion 5-6.

107. Lam NY, Rainer TH, Chan LY, et al. Time course of early and late changes in plasma DNA in trauma patients. Clin Chem 2003;49:1286-91.

108. Jahr S, Hentze H, Englisch S, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001;61:1659-65.

109. Giacona MB, Ruben GC, Iczkowski KA, et al. Cell-free DNA in human blood plasma: length measurements in patients with pancreatic cancer and healthy controls. Pancreas 1998;17:89-97.

110. Vogelstein B, and Kinzler KW. Cancer genes and the pathways they control. Nat Med 2004;10:789-99.

111. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007;35:495-516. 112. Chan KC, Zhang J, Hui AB, et al. Size distributions of maternal and fetal DNA in maternal

plasma. Clin Chem 2004;50:88-92.

Page 158: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

158

113. Lui YY, and Dennis YM. Circulating DNA in plasma and serum: biology, preanalytical issues and diagnostic applications. Clin Chem Lab Med 2002;40:962-8.

114. Bremnes RM, Sirera R, and Camps C. Circulating tumour-derived DNA and RNA markers in blood: a tool for early detection, diagnostics, and follow-up? Lung Cancer 2005;49:1-12.

115. Umetani N, Giuliano AE, Hiramatsu SH, et al. Prediction of breast tumor progression by integrity of free circulating DNA in serum. J Clin Oncol 2006;24:4270-6.

116. Isaacs P. Non-specificity of elevated serum ribonuclease as a pancreatic tumour marker. Digestion 1981;22:101-7.

117. Odaka C, and Mizuochi T. Role of macrophage lysosomal enzymes in the degradation of nucleosomes of apoptotic cells. J Immunol 1999;163:5346-52.

118. Stroun M, Maurice P, Vasioukhin V, et al. The origin and mechanism of circulating DNA. Ann N Y Acad Sci 2000;906:161-8.

119. Stroun M, Lyautey J, Lederrey C, et al. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta 2001;313:139-42.

120. Waterhouse M, Themeli M, Metaxas Y, et al. Horizontal DNA and mRNA transfer between donor and recipient cells after allogeneic hematopoietic cell transplantation? Front Biosci 2009;14:2704-13.

121. Garcia-Olmo D, Garcia-Olmo DC, Ontanon J, et al. Horizontal transfer of DNA and the "genometastasis hypothesis". Blood 2000;95:724-5.

122. Ehnfors J, Kost-Alimova M, Persson NL, et al. Horizontal transfer of tumor DNA to endothelial cells in vivo. Cell Death Differ 2009;16:749-57.

123. Holmgren L, Szeles A, Rajnavolgyi E, et al. Horizontal transfer of DNA by the uptake of apoptotic bodies. Blood 1999;93:3956-63.

124. Tamkovich SN, Bryzgunova OE, Rykova EY, et al. Circulating nucleic acids in blood of healthy male and female donors. Clin Chem 2005;51:1317-9.

125. Rykova E, Skvortsova TE, Hoffmann AL, et al. [Breast cancer diagnostics based on extracellular DNA and RNA circulating in blood]. Biomed Khim 2008;54:94-103.

126. Tamkovich SN, Litviakov NV, Bryzgunova OE, et al. Cell-surface-bound circulating DNA as a prognostic factor in lung cancer. Ann N Y Acad Sci 2008;1137:214-7.

127. Rachmilewitz D, Karmeli F, Takabayashi K, et al. Immunostimulatory DNA ameliorates experimental and spontaneous murine colitis. Gastroenterology 2002;122:1428-41.

128. Chandler VL. Paramutation: from maize to mice. Cell 2007;128:641-5. 129. Hoque MO, Begum S, Topaloglu O, et al. Quantitation of promoter methylation of multiple

genes in urine DNA and bladder cancer detection. J Natl Cancer Inst 2006;98:996-1004. 130. Attallah AM, el-Didi M, Seif F, et al. Comparative study between cytology and dot-ELISA for

early detection of bladder cancer. Am J Clin Pathol 1996;105:109-14. 131. Zancan M, Franceschini R, Mimmo C, et al. Free DNA in urine: a new marker for bladder

cancer? Preliminary data. Int J Biol Markers 2005;20:134-6. 132. Zancan M, Galdi F, Di Tonno F, et al. Evaluation of cell-free DNA in urine as a marker for

bladder cancer diagnosis. Int J Biol Markers 2009;24:147-55. 133. Serdyuk OI, Botezatu IV, Shelepov VP, et al. Detection of mutant k-ras sequences in the urine

of cancer patients. Bull Exp Biol Med 2001;131:283-4. 134. Hou JQ, He J, Wen DG, et al. Survivin mRNA expression in urine as a biomarker for patients

with transitional cell carcinoma of bladder. Chin Med J (Engl) 2006;119:1118-20. 135. Su YH, Song J, Wang Z, et al. Removal of high-molecular-weight DNA by carboxylated

magnetic beads enhances the detection of mutated K-ras DNA in urine. Ann N Y Acad Sci 2008;1137:82-91.

136. Su YH, Wang M, Brenner DE, et al. Detection of mutated K-ras DNA in urine, plasma, and serum of patients with colorectal carcinoma or adenomatous polyps. Ann N Y Acad Sci 2008;1137:197-206.

Page 159: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

159

137. Diehl F, Li M, Dressman D, 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.

138. Muller I, Beeger C, Alix-Panabieres C, et al. Identification of loss of heterozygosity on circulating free DNA in peripheral blood of prostate cancer patients: potential and technical improvements. Clin Chem 2008;54:688-96.

139. Thierry AR, Mouliere F, Gongora C, et al. Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res 2010.

140. Su YH, Wang M, Aiamkitsumrit B, et al. Detection of a K-ras mutation in urine of patients with colorectal cancer. Cancer Biomark 2005;1:177-82.

141. Bryzgunova OE, Morozkin ES, Yarmoschuk SV, et al. Methylation-specific sequencing of GSTP1 gene promoter in circulating/extracellular DNA from blood and urine of healthy donors and prostate cancer patients. Ann N Y Acad Sci 2008;1137:222-5.

142. Utting M, Werner W, Dahse R, et al. Microsatellite analysis of free tumor DNA in urine, serum, and plasma of patients: a minimally invasive method for the detection of bladder cancer. Clin Cancer Res 2002;8:35-40.

143. Hoque MO, Begum S, Topaloglu O, et al. Quantitative detection of promoter hypermethylation of multiple genes in the tumor, urine, and serum DNA of patients with renal cancer. Cancer Res 2004;64:5511-7.

144. Yazici H, Terry MB, Cho YH, et al. Aberrant methylation of RASSF1A in plasma DNA before breast cancer diagnosis in the Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev 2009;18:2723-5.

145. Yoon KA, Park S, Lee SH, et al. Comparison of circulating plasma DNA levels between lung cancer patients and healthy controls. J Mol Diagn 2009;11:182-5.

146. Toth K, Galamb O, Spisak S, et al. [Free circulating DNA based colorectal cancer screening from peripheral blood: the possibility of the methylated septin 9 gene marker]. Orv Hetil 2009;150:969-77.

147. Sunami E, Vu AT, Nguyen SL, et al. Quantification of LINE1 in circulating DNA as a molecular biomarker of breast cancer. Ann N Y Acad Sci 2008;1137:171-4.

148. Umetani N, Hiramatsu S, and Hoon DS. Higher amount of free circulating DNA in serum than in plasma is not mainly caused by contaminated extraneous DNA during separation. Ann N Y Acad Sci 2006;1075:299-307.

149. Umetani N, Kim J, Hiramatsu S, et al. Increased integrity of free circulating DNA in sera of patients with colorectal or periampullary cancer: direct quantitative PCR for ALU repeats. Clin Chem 2006;52:1062-9.

150. Jung M, Klotzek S, Lewandowski M, et al. Changes in concentration of DNA in serum and plasma during storage of blood samples. Clin Chem 2003;49:1028-9.

151. Lee TH, Montalvo L, Chrebtow V, et al. Quantitation of genomic DNA in plasma and serum samples: higher concentrations of genomic DNA found in serum than in plasma. Transfusion 2001;41:276-82.

152. Taback B, O'Day SJ, and Hoon DS. Quantification of circulating DNA in the plasma and serum of cancer patients. Ann N Y Acad Sci 2004;1022:17-24.

153. Xue X, Teare MD, Holen I, et al. Optimizing the yield and utility of circulating cell-free DNA from plasma and serum. Clin Chim Acta 2009;404:100-4.

154. van der Vaart M, and Pretorius PJ. Is the role of circulating DNA as a biomarker of cancer being prematurely overrated? Clin Biochem 2010;43:26-36.

155. Behera D, Gumbhir K, Malik SK, et al. Lung cancer and circulating DNA. Indian J Chest Dis Allied Sci 1986;28:54-5.

156. Altimari A, Grigioni AD, Benedettini E, et al. Diagnostic role of circulating free plasma DNA detection in patients with localized prostate cancer. Am J Clin Pathol 2008;129:756-62.

Page 160: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

160

157. Agostini M, Pucciarelli S, Enzo MV, et al. Circulating Cell-Free DNA: A Promising Marker of Pathologic Tumor Response in Rectal Cancer Patients Receiving Preoperative Chemoradiotherapy. Ann Surg Oncol 2011.

158. Catarino R, Ferreira MM, Rodrigues H, et al. Quantification of free circulating tumor DNA as a diagnostic marker for breast cancer. DNA Cell Biol 2008;27:415-21.

159. Bastian PJ, Palapattu GS, Yegnasubramanian S, et al. Prognostic value of preoperative serum cell-free circulating DNA in men with prostate cancer undergoing radical prostatectomy. Clin Cancer Res 2007;13:5361-7.

160. Dobrzycka B, Terlikowski SJ, Mazurek A, et al. Circulating free DNA, p53 antibody and mutations of KRAS gene in endometrial cancer. Int J Cancer 2010;127:612-21.

161. Gal S, Fidler C, Lo YM, et al. Quantitation of circulating DNA in the serum of breast cancer patients by real-time PCR. Br J Cancer 2004;90:1211-5.

162. Takeshita H, Ichikawa D, Komatsu S, et al. Prediction of CCND1 amplification using plasma DNA as a prognostic marker in oesophageal squamous cell carcinoma. Br J Cancer 2010;102:1378-83.

163. Rago C, Huso DL, Diehl F, et al. Serial assessment of human tumor burdens in mice by the analysis of circulating DNA. Cancer Res 2007;67:9364-70.

164. Lam NY, Rainer TH, Chiu RW, et al. Plasma mitochondrial DNA concentrations after trauma. Clin Chem 2004;50:213-6.

165. Wang BG, Huang HY, Chen YC, et al. Increased plasma DNA integrity in cancer patients. Cancer Res 2003;63:3966-8.

166. Holdenrieder S, Burges A, Reich O, et al. DNA integrity in plasma and serum of patients with malignant and benign diseases. Ann N Y Acad Sci 2008;1137:162-70.

167. Kumar S, Guleria R, Singh V, et al. Efficacy of circulating plasma DNA as a diagnostic tool for advanced non-small cell lung cancer and its predictive utility for survival and response to chemotherapy. Lung Cancer 2010.

168. Sozzi G, Conte D, Leon M, et al. Quantification of free circulating DNA as a diagnostic marker in lung cancer. J Clin Oncol 2003;21:3902-8.

169. Ellinger J, Bastian PJ, Haan KI, et al. Noncancerous PTGS2 DNA fragments of apoptotic origin in sera of prostate cancer patients qualify as diagnostic and prognostic indicators. Int J Cancer 2008;122:138-43.

170. Chang HW, Lee SM, Goodman SN, et al. Assessment of plasma DNA levels, allelic imbalance, and CA 125 as diagnostic tests for cancer. J Natl Cancer Inst 2002;94:1697-703.

171. Huang ZH, Li LH, and Hua D. Quantitative analysis of plasma circulating DNA at diagnosis and during follow-up of breast cancer patients. Cancer Lett 2006;243:64-70.

172. Deligezer U, Eralp Y, Akisik EE, et al. Size distribution of circulating cell-free DNA in sera of breast cancer patients in the course of adjuvant chemotherapy. Clin Chem Lab Med 2008;46:311-7.

173. Kohler C, Radpour R, Barekati Z, et al. Levels of plasma circulating cell free nuclear and mitochondrial DNA as potential biomarkers for breast tumors. Mol Cancer 2009;8:105.

174. Holdenrieder S, Stieber P, Bodenmuller H, et al. Nucleosomes in serum of patients with benign and malignant diseases. Int J Cancer 2001;95:114-20.

175. Alvarado MD, Jensen EH, and Yeatman TJ. The potential role of gene expression in the management of primary and metastatic colorectal cancer. Cancer Control 2006;13:27-31.

176. Perego RA, Corizzato M, Brambilla P, et al. Concentration and microsatellite status of plasma DNA for monitoring patients with renal carcinoma. Eur J Cancer 2008;44:1039-47.

177. deVos T, Tetzner R, Model F, et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 2009;55:1337-46.

178. Chan KC, Lai PB, Mok TS, et al. Quantitative analysis of circulating methylated DNA as a biomarker for hepatocellular carcinoma. Clin Chem 2008;54:1528-36.

Page 161: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

161

179. Grutzmann R, Molnar B, Pilarsky C, et al. Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS One 2008;3:e3759.

180. Rashid A, Shen L, Morris JS, et al. CpG island methylation in colorectal adenomas. Am J Pathol 2001;159:1129-35.

181. Zou H, Harrington JJ, Shire AM, et al. Highly methylated genes in colorectal neoplasia: implications for screening. Cancer Epidemiol Biomarkers Prev 2007;16:2686-96.

182. Xu XL, Yu J, Zhang HY, et al. Methylation profile of the promoter CpG islands of 31 genes that may contribute to colorectal carcinogenesis. World J Gastroenterol 2004;10:3441-54.

183. Johnson PJ, and Lo YM. Plasma nucleic acids in the diagnosis and management of malignant disease. Clin Chem 2002;48:1186-93.

184. Lavon I, Refael M, Zelikovitch B, et al. Serum DNA can define tumor-specific genetic and epigenetic markers in gliomas of various grades. Neuro Oncol 2010;12:173-80.

185. Leung WK, To KF, Man EP, et al. Quantitative detection of promoter hypermethylation in multiple genes in the serum of patients with colorectal cancer. Am J Gastroenterol 2005;100:2274-9.

186. Sabbioni S, Miotto E, Veronese A, et al. Multigene methylation analysis of gastrointestinal tumors: TPEF emerges as a frequent tumor-specific aberrantly methylated marker that can be detected in peripheral blood. Mol Diagn 2003;7:201-7.

187. Timothy RC, Michael W, Catherine LD, et al. Prospective Clinical Validation of an Assay for Methylated SEPT9 DNA in Human Plasma as a Colorectal Cancer Screening Tool in Average Risk Men and Women ≥50 Years. Gastroenterology 2010;139:e18.

188. Eads CA, Lord RV, Kurumboor SK, et al. Fields of aberrant CpG island hypermethylation in Barrett's esophagus and associated adenocarcinoma. Cancer Res 2000;60:5021-6.

189. McCabe DC, and Caudill MA. DNA methylation, genomic silencing, and links to nutrition and cancer. Nutr Rev 2005;63:183-95.

190. Lim U, Flood A, Choi SW, et al. Genomic methylation of leukocyte DNA in relation to colorectal adenoma among asymptomatic women. Gastroenterology 2008;134:47-55.

191. Hsiung DT, Marsit CJ, Houseman EA, et al. Global DNA methylation level in whole blood as a biomarker in head and neck squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev 2007;16:108-14.

192. Chalitchagorn K, Shuangshoti S, Hourpai N, et al. Distinctive pattern of LINE-1 methylation level in normal tissues and the association with carcinogenesis. Oncogene 2004;23:8841-6.

193. Hibi K, Robinson CR, Booker S, et al. Molecular detection of genetic alterations in the serum of colorectal cancer patients. Cancer Res 1998;58:1405-7.

194. Anker P, Lefort F, Vasioukhin V, et al. K-ras mutations are found in DNA extracted from the plasma of patients with colorectal cancer. Gastroenterology 1997;112:1114-20.

195. Diehl F, Schmidt K, Durkee KH, et al. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology 2008;135:489-98.

196. Holdhoff M, Schmidt K, Donehower R, et al. Analysis of circulating tumor DNA to confirm somatic KRAS mutations. J Natl Cancer Inst 2009;101:1284-5.

197. Rawnaq T, Schwarzenbach H, Schurr PG, et al. Monitoring of Loss of Heterozygosity in Serum Microsatellite DNA Among Patients with Gastrointestinal Stromal Tumors Indicates Tumor Recurrence. J Surg Res 2010.

198. Chen X, Bonnefoi H, Diebold-Berger S, et al. Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin Cancer Res 1999;5:2297-303.

199. Wachowiak R, Kaifi JT, Schurr PG, et al. Similar patterns of loss of heterozygosity in serum of adenocarcinoma of the distal oesophagus and the cardia in early diagnosis. Anticancer Res 2007;27:477-81.

200. Nakamoto D, Yamamoto N, Takagi R, et al. Detection of microsatellite alterations in plasma DNA of malignant mucosal melanoma using whole genome amplification. Bull Tokyo Dent Coll 2008;49:77-87.

Page 162: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

162

201. Kolble K, Ullrich OM, Pidde H, et al. Microsatellite alterations in serum DNA of patients with colorectal cancer. Lab Invest 1999;79:1145-50.

202. Eisenberger CF, Stoecklein NH, Jazra S, et al. The detection of oesophageal adenocarcinoma by serum microsatellite analysis. Eur J Surg Oncol 2006;32:954-60.

203. Ulvik A, and Ueland PM. Single nucleotide polymorphism (SNP) genotyping in unprocessed whole blood and serum by real-time PCR: application to SNPs affecting homocysteine and folate metabolism. Clin Chem 2001;47:2050-3.

204. Silva JM, and Bonilla F. Correspondence re: F. Coulet et al., Detection of plasma tumor DNA in head and neck squamous cell carcinoma by microsatellite typing and p53 mutation analysis. Cancer Res., 60: 707-709, 2000. Cancer Res 2001;61:8595-6.

205. Brankley SM, Wang KK, Harwood AR, et al. The development of a fluorescence in situ hybridization assay for the detection of dysplasia and adenocarcinoma in Barrett's esophagus. J Mol Diagn 2006;8:260-7.

206. Prasad GA, Bansal A, Sharma P, et al. Predictors of progression in Barrett's esophagus: current knowledge and future directions. Am J Gastroenterol 2010;105:1490-502.

207. Reid BJ, Levine DS, Longton G, et al. Predictors of progression to cancer in Barrett's esophagus: baseline histology and flow cytometry identify low- and high-risk patient subsets. Am J Gastroenterol 2000;95:1669-76.

208. Beck J, Urnovitz HB, Mitchell WM, et al. Next generation sequencing of serum circulating nucleic acids from patients with invasive ductal breast cancer reveals differences to healthy and nonmalignant controls. Mol Cancer Res 2010;8:335-42.

209. Baggerly KA, Edmonson SR, Morris JS, et al. High-resolution serum proteomic patterns for ovarian cancer detection. Endocr Relat Cancer 2004;11:583-4; author reply 5-7.

210. Croft DT, Jr., Jordan RM, Patney HL, et al. Performance of whole-genome amplified DNA isolated from serum and plasma on high-density single nucleotide polymorphism arrays. J Mol Diagn 2008;10:249-57.

211. Lin CS, Wang LS, Tsai CM, et al. Low copy number and low oxidative damage of mitochondrial DNA are associated with tumor progression in lung cancer tissues after neoadjuvant chemotherapy. Interact Cardiovasc Thorac Surg 2008;7:954-8.

212. Ellinger J, Albers P, Muller SC, et al. Circulating mitochondrial DNA in the serum of patients with testicular germ cell cancer as a novel noninvasive diagnostic biomarker. BJU Int 2009;104:48-52.

213. Chan SK, Griffith OL, Tai IT, et al. Meta-analysis of colorectal cancer gene expression profiling studies identifies consistently reported candidate biomarkers. Cancer Epidemiol Biomarkers Prev 2008;17:543-52.

214. Han M, Liew CT, Zhang HW, et al. Novel blood-based, five-gene biomarker set for the detection of colorectal cancer. Clin Cancer Res 2008;14:455-60.

215. Marshall KW, Mohr S, Khettabi FE, et al. A blood-based biomarker panel for stratifying current risk for colorectal cancer. Int J Cancer 2010;126:1177-86.

216. Lledo SM, Garcia-Granero E, Dasi F, et al. Real time quantification in plasma of human telomerase reverse transcriptase (hTERT) mRNA in patients with colorectal cancer. Colorectal Dis 2004;6:236-42.

217. Dasi F, Lledo S, Garcia-Granero E, et al. Real-time quantification in plasma of human telomerase reverse transcriptase (hTERT) mRNA: a simple blood test to monitor disease in cancer patients. Lab Invest 2001;81:767-9.

218. Gehan Kamal El-Saeed MAE-B, Hala Hany El-Saeed ,, and Iman A. Gaweesh MAS, Hatim Attiya. Molecular Detection of Human Telomerase mRNA (hTERT-mRNA) in Egyptian Patients with Hepatocellular Carcinoma. Hepatitis Monthly 2009;9:114-21.

219. Silva JM, Rodriguez R, Garcia JM, et al. Detection of epithelial tumour RNA in the plasma of colon cancer patients is associated with advanced stages and circulating tumour cells. Gut 2002;50:530-4.

Page 163: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

163

220. Schuster R, Max N, Mann B, et al. Quantitative real-time RT-PCR for detection of disseminated tumor cells in peripheral blood of patients with colorectal cancer using different mRNA markers. Int J Cancer 2004;108:219-27.

221. Molnar B. Colorectal Cancer (CRC), Adenoma and IBD Detection in Blood Cells by Array PCR - Role of Circulating Free DNA as Mononuclear Cell Activator. Gastroenterology 2010;Supplement:s 67.

222. Grimminger P, Vallbohmer D, Hoffmann A, et al. Quantitative analysis of survivin RNA expression in blood as a non-invasive predictor of response to neoadjuvant radiochemotherapy in esophageal cancer. J Surg Oncol 2009;100:447-51.

223. Shariat SF, Casella R, Khoddami SM, et al. Urine detection of survivin is a sensitive marker for the noninvasive diagnosis of bladder cancer. J Urol 2004;171:626-30.

224. Tanaka K, Yano M, Motoori M, et al. CEA-antigen and SCC-antigen mRNA expression in peripheral blood predict hematogenous recurrence after resection in patients with esophageal cancer. Ann Surg Oncol 2010;17:2779-86.

225. Setoyama T, Natsugoe S, Okumura H, et al. Carcinoembryonic antigen messenger RNA expression in blood predicts recurrence in esophageal cancer. Clin Cancer Res 2006;12:5972-7.

226. Anderson NL, and Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics 2002;1:845-67.

227. Hung KE, and Yu KH. Proteomic approaches to cancer biomarkers. Gastroenterology 2010;138:46-51 e1.

228. de Noo ME, Mertens BJ, Ozalp A, et al. Detection of colorectal cancer using MALDI-TOF serum protein profiling. Eur J Cancer 2006;42:1068-76.

229. Sorace JM, and Zhan M. A data review and re-assessment of ovarian cancer serum proteomic profiling. BMC Bioinformatics 2003;4:24.

230. Zhang Z, Bast RC, Jr., Yu Y, et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res 2004;64:5882-90.

231. Petricoin EF, Ardekani AM, Hitt BA, et al. Use of proteomic patterns in serum to identify ovarian cancer. Lancet 2002;359:572-7.

232. Whistler T, Rollin D, and Vernon SD. A method for improving SELDI-TOF mass spectrometry data quality. Proteome Sci 2007;5:14.

233. De Bock M, de Seny D, Meuwis MA, et al. Challenges for biomarker discovery in body fluids using SELDI-TOF-MS. J Biomed Biotechnol 2010;2010:906082.

234. Engwegen JY, Depla AC, Smits ME, et al. Detection of Colorectal Cancer by Serum and Tissue Protein Profiling: A Prospective Study in a Population at Risk. Biomark Insights 2008;3:375-85.

235. Xue A, Scarlett CJ, Chung L, et al. Discovery of serum biomarkers for pancreatic adenocarcinoma using proteomic analysis. Br J Cancer 2010;103:391-400.

236. Habermann JK, Roblick UJ, Luke BT, et al. Increased serum levels of complement C3a anaphylatoxin indicate the presence of colorectal tumors. Gastroenterology 2006;131:1020-9; quiz 284.

237. Roessler M, Rollinger W, Palme S, et al. Identification of nicotinamide N-methyltransferase as a novel serum tumor marker for colorectal cancer. Clin Cancer Res 2005;11:6550-7.

238. Lee H, Rhee H, Kang HJ, et al. Macrophage migration inhibitory factor may be used as an early diagnostic marker in colorectal carcinomas. Am J Clin Pathol 2008;129:772-9.

239. Gold P, and Freedman SO. Demonstration of Tumor-Specific Antigens in Human Colonic Carcinomata by Immunological Tolerance and Absorption Techniques. J Exp Med 1965;121:439-62.

240. Metser U, You J, McSweeney S, et al. Assessment of tumor recurrence in patients with colorectal cancer and elevated carcinoembryonic antigen level: FDG PET/CT versus contrast-enhanced 64-MDCT of the chest and abdomen. AJR Am J Roentgenol 2010;194:766-71.

Page 164: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

164

241. Zheng CX, Zhan WH, Zhao JZ, et al. The prognostic value of preoperative serum levels of CEA, CA19-9 and CA72-4 in patients with colorectal cancer. World J Gastroenterol 2001;7:431-4.

242. Flamini E, Mercatali L, Nanni O, et al. Free DNA and carcinoembryonic antigen serum levels: an important combination for diagnosis of colorectal cancer. Clin Cancer Res 2006;12:6985-8.

243. D'Armento G, Daniele L, Mariani S, et al. Added value of combined gene and protein expression of CK20 and CEA in non-macroscopically involved lymph nodes of colorectal cancer. Int J Surg Pathol 2009;17:93-8.

244. Joergensen MT, Brunner N, and De Muckadell OB. Comparison of circulating MMP-9, TIMP-1 and CA19-9 in the detection of pancreatic cancer. Anticancer Res 2010;30:587-92.

245. Haglund C, Roberts PJ, Jalanko H, et al. Tumour markers CA 19-9 and CA 50 in digestive tract malignancies. Scand J Gastroenterol 1992;27:169-74.

246. Rothlin MA, Joller H, and Largiader F. CA 242 is a new tumor marker for pancreatic cancer. Cancer 1993;71:701-7.

247. Bast RC, Jr., Xu FJ, Yu YH, et al. CA 125: the past and the future. Int J Biol Markers 1998;13:179-87.

248. Joypaul B, Browning M, Newman E, et al. Comparison of serum CA 72-4 and CA 19-9 levels in gastric cancer patients and correlation with recurrence. Am J Surg 1995;169:595-9.

249. Given M, Scott M, Mc Grath JP, et al. The predictive of tumour markers CA 15-3, TPS and CEA in breast cancer recurrence. Breast 2000;9:277-80.

250. Bon GG, von Mensdorff-Pouilly S, Kenemans P, et al. Clinical and technical evaluation of ACS BR serum assay of MUC1 gene-derived glycoprotein in breast cancer, and comparison with CA 15-3 assays. Clin Chem 1997;43:585-93.

251. Chan DW, Beveridge RA, Muss H, et al. Use of Truquant BR radioimmunoassay for early detection of breast cancer recurrence in patients with stage II and stage III disease. J Clin Oncol 1997;15:2322-8.

252. Li M, Yuan YH, Han Y, et al. Expression profile of cancer-testis genes in 121 human colorectal cancer tissue and adjacent normal tissue. Clin Cancer Res 2005;11:1809-14.

253. Akcakanat A, Kanda T, Tanabe T, et al. Heterogeneous expression of GAGE, NY-ESO-1, MAGE-A and SSX proteins in esophageal cancer: Implications for immunotherapy. Int J Cancer 2006;118:123-8.

254. Jager E, Stockert E, Zidianakis Z, et al. Humoral immune responses of cancer patients against "Cancer-Testis" antigen NY-ESO-1: correlation with clinical events. Int J Cancer 1999;84:506-10.

255. Kilic A, Schuchert MJ, Luketich JD, et al. Use of novel autoantibody and cancer-related protein arrays for the detection of esophageal adenocarcinoma in serum. J Thorac Cardiovasc Surg 2008;136:199-204.

256. Sahin U, Tureci O, Schmitt H, et al. Human neoplasms elicit multiple specific immune responses in the autologous host. Proc Natl Acad Sci U S A 1995;92:11810-3.

257. Chan CC, Fan CW, Kuo YB, et al. Multiple serological biomarkers for colorectal cancer detection. Int J Cancer 2010;126:1683-90.

258. Zhong L, Ge K, Zu JC, et al. Autoantibodies as potential biomarkers for breast cancer. Breast Cancer Res 2008;10:R40.

259. Zhong L, Hidalgo GE, Stromberg AJ, et al. Using protein microarray as a diagnostic assay for non-small cell lung cancer. Am J Respir Crit Care Med 2005;172:1308-14.

260. Cawley HM, Meltzer SJ, De Benedetti VM, et al. Anti-p53 antibodies in patients with Barrett's esophagus or esophageal carcinoma can predate cancer diagnosis. Gastroenterology 1998;115:19-27.

261. Hurst NG, Stocken DD, Wilson S, et al. Elevated serum matrix metalloproteinase 9 (MMP-9) concentration predicts the presence of colorectal neoplasia in symptomatic patients. Br J Cancer 2007;97:971-7.

Page 165: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

165

262. Maurel J, Nadal C, Garcia-Albeniz X, et al. Serum matrix metalloproteinase 7 levels identifies poor prognosis advanced colorectal cancer patients. Int J Cancer 2007;121:1066-71.

263. Ebrahimzadeh MEa MM, Fattahi Ec. The prognostic value of preoperative serum levels of CEA and CA 242 in patients with colorectal cancer. The International Medical Journal 2005.

Dec Vol. 4 8-13. 264. Kim SB, Fernandes LC, Saad SS, et al. Assessment of the value of preoperative serum levels of

CA 242 and CEA in the staging and postoperative survival of colorectal adenocarcinoma patients. Int J Biol Markers 2003;18:182-7.

265. Waas ET, Hendriks T, Lomme RM, et al. Plasma levels of matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-1 correlate with disease stage and survival in colorectal cancer patients. Dis Colon Rectum 2005;48:700-10.

266. Nilsson O, Johansson C, Glimelius B, et al. Sensitivity and specificity of CA242 in gastro-intestinal cancer. A comparison with CEA, CA50 and CA 19-9. Br J Cancer 1992;65:215-21.

267. Birgisson H, Nielsen HJ, Christensen IJ, et al. Preoperative plasma TIMP-1 is an independent prognostic indicator in patients with primary colorectal cancer: A prospective validation study. Eur J Cancer 2010.

268. Holten-Andersen MN, Christensen IJ, Nielsen HJ, et al. Total levels of tissue inhibitor of metalloproteinases 1 in plasma yield high diagnostic sensitivity and specificity in patients with colon cancer. Clin Cancer Res 2002;8:156-64.

269. Komosinska-Vassev K, Olczyk P, Winsz-Szczotka K, et al. Age- and Gender-Dependent Changes in Connective Tissue Remodeling: Physiological Differences in Circulating MMP-3, MMP-10, TIMP-1 and TIMP-2 Level. Gerontology 2010.

270. Fernandes LC, Kim SB, and Matos D. Cytokeratins and carcinoembryonic antigen in diagnosis, staging and prognosis of colorectal adenocarcinoma. World J Gastroenterol 2005;11:645-8.

271. Carpelan-Holmstrom M, Haglund C, Lundin J, et al. Independent prognostic value of preoperative serum markers CA 242, specific tissue polypeptide antigen and human chorionic gonadotrophin beta, but not of carcinoembryonic antigen or tissue polypeptide antigen in colorectal cancer. Br J Cancer 1996;74:925-9.

272. Pujol J, Grenier J, Parrat E, et al. Cytokeratins as serum markers in lung cancer: a comparison of CYFRA 21- 1 and TPS. Am J Respir Crit Care Med 1996;154:725-33.

273. Brockmann JG, St Nottberg H, Glodny B, et al. CYFRA 21-1 serum analysis in patients with esophageal cancer. Clin Cancer Res 2000;6:4249-52.

274. Miller TE, Beausang LA, Winchell LF, et al. Detection of nuclear matrix proteins in serum from cancer patients. Cancer Res 1992;52:422-7.

275. Keesee SK, Meneghini MD, Szaro RP, et al. Nuclear matrix proteins in human colon cancer. Proc Natl Acad Sci U S A 1994;91:1913-6.

276. Keesee SK, Briggman JV, Thill G, et al. Utilization of nuclear matrix proteins for cancer diagnosis. Crit Rev Eukaryot Gene Expr 1996;6:189-214.

277. Brunagel G, Vietmeier BN, Bauer AJ, et al. Identification of nuclear matrix protein alterations associated with human colon cancer. Cancer Res 2002;62:2437-42.

278. Leman ES, Schoen RE, Magheli A, et al. Evaluation of colon cancer-specific antigen 2 as a potential serum marker for colorectal cancer. Clin Cancer Res 2008;14:1349-54.

279. Leman ES, Schoen RE, Weissfeld JL, et al. Initial analyses of colon cancer-specific antigen (CCSA)-3 and CCSA-4 as colorectal cancer-associated serum markers. Cancer Res 2007;67:5600-5.

280. Seefeld M, El Tarhouny S, Fan AX, et al. Parallel assessment of circulatory cell-free DNA by PCR and nucleosomes by ELISA in breast tumors. Int J Biol Markers 2008;23:69-73.

281. Kolomecki K, Stepien H, Bartos M, et al. Usefulness of VEGF, MMP-2, MMP-3 and TIMP-2 serum level evaluation in patients with adrenal tumours. Endocr Regul 2001;35:9-16.

282. Kozlowski M, Kowalczuk O, Milewski R, et al. Serum vascular endothelial growth factors C and D in patients with oesophageal cancer. Eur J Cardiothorac Surg 2010;38:260-7.

Page 166: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

166

283. El Tarhouny S, Seefeld M, Fan AX, et al. Comparison of serum VEGF and its soluble receptor sVEGFR1 with serum cell-free DNA in patients with breast tumor. Cytokine 2008;44:65-9.

284. Lein M, Weiss S, Jung K, et al. Soluble CD44 variants in serum of patients with prostate cancer and other urological malignancies. Prostate 1996;29:334-5.

285. Martin S, Jansen F, Bokelmann J, et al. Soluble CD44 splice variants in metastasizing human breast cancer. Int J Cancer 1997;74:443-5.

286. Masson D, Denis MG, Denis M, et al. Soluble CD44: quantification and molecular repartition in plasma of patients with colorectal cancer. Br J Cancer 1999;80:1995-2000.

287. Guo YJ, Liu G, Wang X, et al. Potential use of soluble CD44 in serum as indicator of tumor burden and metastasis in patients with gastric or colon cancer. Cancer Res 1994;54:422-6.

288. Inoue Y, Yokobori T, Yokoe T, et al. Clinical Significance of Human Kallikrein7 Gene Expression in Colorectal Cancer. Ann Surg Oncol 2010.

289. Diamandis EP, and Yousef GM. Human tissue kallikreins: a family of new cancer biomarkers. Clin Chem 2002;48:1198-205.

290. Shan SJ, Scorilas A, Katsaros D, et al. Unfavorable prognostic value of human kallikrein 7 quantified by ELISA in ovarian cancer cytosols. Clin Chem 2006;52:1879-86.

291. Ewan King L, Li X, Cheikh Saad Bouh K, et al. Human kallikrein 10 ELISA development and validation in breast cancer sera. Clin Biochem 2007;40:1057-62.

292. Feng B, Xu WB, Zheng MH, et al. Clinical significance of human kallikrein 10 gene expression in colorectal cancer and gastric cancer. J Gastroenterol Hepatol 2006;21:1596-603.

293. Saito N, and Kameoka S. Serum laminin is an independent prognostic factor in colorectal cancer. Int J Colorectal Dis 2005;20:238-44.

294. Cohen SJ, Punt CJ, Iannotti N, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol 2008;26:3213-21.

295. Maheswaran S, Sequist LV, Nagrath S, et al. Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med 2008;359:366-77.

296. Tol J, Koopman M, Miller MC, et al. Circulating tumour cells early predict progression-free and overall survival in advanced colorectal cancer patients treated with chemotherapy and targeted agents. Ann Oncol 2010;21:1006-12.

297. Jeng KS, Sheen IS, and Tsai YC. Does the presence of circulating hepatocellular carcinoma cells indicate a risk of recurrence after resection? Am J Gastroenterol 2004;99:1503-9.

298. Rahbari NN, Aigner M, Thorlund K, et al. Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer. Gastroenterology 2010;138:1714-26.

299. Bessa X, Elizalde JI, Boix L, et al. Lack of prognostic influence of circulating tumor cells in peripheral blood of patients with colorectal cancer. Gastroenterology 2001;120:1084-92.

300. Peach G, Kim C, Zacharakis E, et al. Prognostic significance of circulating tumour cells following surgical resection of colorectal cancers: a systematic review. Br J Cancer 2010;102:1327-34.

301. Bressler B, Paszat LF, Vinden C, et al. Colonoscopic miss rates for right-sided colon cancer: a population-based analysis. Gastroenterology 2004;127:452-6.

302. Sachelarie I, Kerr K, Ghesani M, et al. Integrated PET-CT: evidence-based review of oncology indications. Oncology (Williston Park) 2005;19:481-90; discussion 90-2, 95-6.

303. Denis MG, Lipart C, Leborgne J, et al. Detection of disseminated tumor cells in peripheral blood of colorectal cancer patients. Int J Cancer 1997;74:540-4.

304. Ally MS, Al-Ghnaniem R, and Pufulete M. The relationship between gene-specific DNA methylation in leukocytes and normal colorectal mucosa in subjects with and without colorectal tumors. Cancer Epidemiol Biomarkers Prev 2009;18:922-8.

305. Hibi K, and Nakao A. [Molecular detection of tumor DNA in the serum of colorectal cancer patients]. Nippon Rinsho 2003;61 Suppl 7:197-200.

Page 167: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

167

306. Thijssen MA, Swinkels DW, Ruers TJ, et al. Difference between free circulating plasma and serum DNA in patients with colorectal liver metastases. Anticancer Res 2002;22:421-5.

307. Gahan PB, and Swaminathan R. Circulating nucleic acids in plasma and serum. Recent developments. Ann N Y Acad Sci 2008;1137:1-6.

308. Chan KC, Yeung SW, Lui WB, et al. Effects of preanalytical factors on the molecular size of cell-free DNA in blood. Clin Chem 2005;51:781-4.

309. Chiu RW, Poon LL, Lau TK, et al. Effects of blood-processing protocols on fetal and total DNA quantification in maternal plasma. Clin Chem 2001;47:1607-13.

310. Kirsch C, Weickmann S, Schmidt B, et al. An improved method for the isolation of free-circulating plasma DNA and cell-free DNA from other body fluids. Ann N Y Acad Sci 2008;1137:135-9.

311. Loffler J, Hebart H, Schumacher U, et al. Comparison of different methods for extraction of DNA of fungal pathogens from cultures and blood. J Clin Microbiol 1997;35:3311-2.

312. Adams DS. Lab math : a handbook of measurements, calculations, and other quantitative skills for use at the bench. Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory Press; 2003.

313. Griffiths LJ, Anyim M, Doffman SR, et al. Comparison of DNA extraction methods for Aspergillus fumigatus using real-time PCR. J Med Microbiol 2006;55:1187-91.

314. Schwarzenbach H, Stoehlmacher J, Pantel K, et al. Detection and monitoring of cell-free DNA in blood of patients with colorectal cancer. Ann N Y Acad Sci 2008;1137:190-6.

315. Avent ND, Madgett TE, Maddocks DG, et al. Cell-free fetal DNA in the maternal serum and plasma: current and evolving applications. Curr Opin Obstet Gynecol 2009;21:175-9.

316. laboratories I. <http://tools.invitrogen.com/content/sfs/brochures/F-065432%20quantit%20htp_FLR.pdf>. 2011.

317. Kline M. Testing Candidate DNA Quantitation Standards with Several Real-Time Quantitative PCR Methods. talk at the 16th International Symposium on Human Identification (Grapevine, TX), 2005.

318. Benzel I, Bansal A, Browning BL, et al. Interactions among genes in the ErbB-Neuregulin signalling network are associated with increased susceptibility to schizophrenia. Behav Brain Funct 2007;3:31.

319. Goldblum JR. Barrett's esophagus and Barrett's-related dysplasia. Mod Pathol 2003;16:316-24.

320. Goldblum JR. Controversies in the diagnosis of Barrett esophagus and Barrett-related dysplasia: one pathologist's perspective. Arch Pathol Lab Med 2010;134:1479-84.

321. Allin KH, Bojesen SE, and Nordestgaard BG. Baseline C-reactive protein is associated with incident cancer and survival in patients with cancer. J Clin Oncol 2009;27:2217-24.

322. Lo YM, Tein MS, Lau TK, et al. Quantitative analysis of fetal DNA in maternal plasma and serum: implications for noninvasive prenatal diagnosis. Am J Hum Genet 1998;62:768-75.

323. Pucciarelli S, Del Bianco P, Efficace F, et al. Patient-reported outcomes after neoadjuvant chemoradiotherapy for rectal cancer: a multicenter prospective observational study. Ann Surg 2011;253:71-7.

324. Schmidt B, Weickmann S, Witt C, et al. Integrity of cell-free plasma DNA in patients with lung cancer and nonmalignant lung disease. Ann N Y Acad Sci 2008;1137:207-13.

325. Huang P, Zhang YX, and Zhao T. [The relationship between copy number and microsatellite instability of mitochondrial DNA in colorectal cancer]. Zhonghua Nei Ke Za Zhi 2009;48:837-40.

326. Xia P, An HX, Dang CX, et al. Decreased mitochondrial DNA content in blood samples of patients with stage I breast cancer. BMC Cancer 2009;9:454.

327. Hosgood HD, 3rd, Liu CS, Rothman N, et al. Mitochondrial DNA copy number and lung cancer risk in a prospective cohort study. Carcinogenesis 2010;31:847-9.

Page 168: December 2012 Circulating DNA markers of Gastro-intestinal … · 2017. 1. 19. · gastro-intestinal cancer biomarker literature reporting the strengths, weaknesses and successes

References

168

328. Dorssers LC, Grebenchtchikov N, Brinkman A, et al. Application of a newly developed ELISA for BCAR1 protein for prediction of clinical benefit of tamoxifen therapy in patients with advanced breast cancer. Clin Chem 2004;50:1445-7.

329. Taback B, Saha S, and Hoon DS. Comparative analysis of mesenteric and peripheral blood circulating tumor DNA in colorectal cancer patients. Ann N Y Acad Sci 2006;1075:197-203.

330. Gauthier VJ, Tyler LN, and Mannik M. Blood clearance kinetics and liver uptake of mononucleosomes in mice. J Immunol 1996;156:1151-6.

331. Sozzi G, Roz L, Conte D, et al. Effects of prolonged storage of whole plasma or isolated plasma DNA on the results of circulating DNA quantification assays. J Natl Cancer Inst 2005;97:1848-50.

332. Kawakami K, Brabender J, Lord RV, et al. Hypermethylated APC DNA in plasma and prognosis of patients with esophageal adenocarcinoma. J Natl Cancer Inst 2000;92:1805-11.

333. Banki F, Yacoub WN, Hagen JA, et al. Plasma DNA is more reliable than carcinoembryonic antigen for diagnosis of recurrent esophageal cancer. J Am Coll Surg 2008;207:30-5.

334. Wang M, Block TM, Steel L, et al. Preferential isolation of fragmented DNA enhances the detection of circulating mutated k-ras DNA. Clin Chem 2004;50:211-3.

335. Ross KS, Haites NE, and Kelly KF. Repeated freezing and thawing of peripheral blood and DNA in suspension: effects on DNA yield and integrity. J Med Genet 1990;27:569-70.

336. Stroun M, Lyautey J, Lederrey C, et al. Alu repeat sequences are present in increased proportions compared to a unique gene in plasma/serum DNA: evidence for a preferential release from viable cells? Ann N Y Acad Sci 2001;945:258-64.

337. Rogers JC. Characterization of DNA excreted from phytohemagglutinin-stimulated lymphocytes. J Exp Med 1976;143:1249-64.

338. Stroun M, Anker P, Beljanski M, et al. Presence of RNA in the nucleoprotein complex spontaneously released by human lymphocytes and frog auricles in culture. Cancer Res 1978;38:3546-54.

339. Tomita H, Ichikawa D, Ikoma D, et al. Quantification of circulating plasma DNA fragments as tumor markers in patients with esophageal cancer. Anticancer Res 2007;27:2737-41.

340. Shi X, Seluanov A, and Gorbunova V. Cell divisions are required for L1 retrotransposition. Mol Cell Biol 2007;27:1264-70.

341. Nystrom-Lahti M, Kristo P, Nicolaides NC, et al. Founding mutations and Alu-mediated recombination in hereditary colon cancer. Nat Med 1995;1:1203-6.

342. Boldt DH, MacDermott RP, Speckart SF, et al. Excretion of DNA by purified human lymphocyte subpopulations. J Immunol 1977;118:1495-8.

343. Bazan V, Bruno L, Augello C, et al. Molecular detection of TP53, Ki-Ras and p16INK4A promoter methylation in plasma of patients with colorectal cancer and its association with prognosis. Results of a 3-year GOIM (Gruppo Oncologico dell'Italia Meridionale) prospective study. Ann Oncol 2006;17 Suppl 7:vii84-90.

344. Boher JM, Pujol JL, Grenier J, et al. Markov model and markers of small cell lung cancer: assessing the influence of reversible serum NSE, CYFRA 21-1 and TPS levels on prognosis. Br J Cancer 1999;79:1419-27.

345. Bonfrer JM, Gaarenstroom KN, Kenter GG, et al. Prognostic significance of serum fragments of cytokeratin 19 measured by Cyfra 21-1 in cervical cancer. Gynecol Oncol 1994;55:371-5.

346. Chen F, Luo X, Zhang J, et al. Elevated serum levels of TPS and CYFRA 21-1 predict poor prognosis in advanced non-small-cell lung cancer patients treated with gefitinib. Med Oncol 2009.