liquid biopsy in breast cancer - uef electronic...

66
Liquid biopsy in breast cancer: Identifying circulating tumor DNA from breast cancer patient serum samples using PIK3CA-H1047R mutation specific castPCR Emmi Lotta Karoliina Honkala Master’s thesis University of Eastern Finland School of Medicine Biomedicine April 2016

Upload: hadan

Post on 03-Jul-2019

223 views

Category:

Documents


0 download

TRANSCRIPT

Liquid biopsy in breast cancer: Identifying circulating tumor DNA from

breast cancer patient serum samples using

PIK3CA-H1047R mutation specific castPCR

Emmi Lotta Karoliina Honkala

Master’s thesis

University of Eastern Finland

School of Medicine

Biomedicine

April 2016

University of Eastern Finland, Faculty of Health Sciences

Department of biomedicine

Lotta Honkala: Liquid biopsy in breast cancer: Identifying of circulating tumor DNA from

breast cancer patient serum samples using PIK3CA-H1047R mutation specific castPCR

April 2016

Key words: breast cancer, cell-free DNA, PIK3CA, castPCR

ABSTRACT

Circulating nucleic acids have been researched as a new potential biomarker for the diagnosis,

prognosis and treatment of breast cancer. Circulating tumor DNA can be accessed by a liquid

biopsy, which would give a snapshot of the primary tumor with a noninvasive technique. This

could allow early diagnosis and enable monitoring during and after cancer therapies even if

the primary tumor has been surgically removed.

The aim of this Master’s Thesis was to study if circulating DNA could be used as a biomarker

in breast cancer. Serum samples from 82 breast cancer patients were chosen for this study.

The first 38 patient samples had ductal breast carcinoma with a T1N0 status at the time of

diagnosis and thus a good prognosis. However, 19 of these patients relapsed or died during

the follow-up time. The second set of 44 patients had triple negative breast cancer.

DNA was extracted using the CNA-kit (Qiagen) and DNA concentration and quality was

measured with Qubit (Thermo Fisher Scientific). Those serum samples which showed a high-

enough DNA concentration were further analyzed with castPCR PIK3CA-H1047R mutation

analysis. Also 22 primary tumor DNA samples were run with castPCR.

In this study, the concentration of circulating cell free DNA was found to be associated with

tumor grade, hormone receptor status of the tumors and relapse. Patients with higher

concentrations of circulating cell free DNA were more likely to express hormone receptors

and to relapse. Furthermore they had a higher mortality rate than patients with low

concentrations. castPCR PIK3CA mutation analysis did not detect the H1047R mutation in

serum samples although 9 out of 22 primary tumor DNA samples were positive in the analysis.

This study shows the potential of circulating nucleic acids as a tumor biomarker for breast

cancer. Quantification of circulating cell free DNA could serve as a prognostic marker for

relapse and survival. However, according to the castPCR analysis, none of the serum DNA

contained the H1047R mutation, and the method for assessing cfDNA must be considered for

the clinical setting. Further research is required to enable the use of cfDNA as an informative

biomarker in the diagnosis, prognosis and treatment of breast cancer.

Itä-Suomen yliopisto, Terveystieteiden tiedekunta

Biolääketieteen laitos

Lotta Honkala: Liquid biopsy in breast cancer: Identifying of circulating tumor DNA from

breast cancer patient serum samples using PIK3CA-H1047R mutation specific castPCR

Huhtikuu 2016

Avainsanat: rintasyöpä, soluvapaa DNA, PIK3CA, castPCR

TIIVISTELMÄ

Sirkuloivia nukleiinihappoja ja niiden potentiaalista roolia on tutkittu rintasyövän

diagnosoinnissa, ennusteessa ja hoidossa. Tuumorista peräisin olevaan DNA:han voidaan

päästä käsiksi ns. nestemäisen biopsian avulla, joka voi olla esimerkiksi yksinkertainen

verinäyte. Nestemäinen biopsia mahdollistaa diagnosoinnin ja taudin seurannan, vaikka

primäärituumori olisi poistettu.

Tutkimukseen valittiin 82 rintasyöpäpotilasta, joilla 38:lla oli pienikokoinen tuumori ja hyvä

ennuste. Näistä potilaista 19 kuitenkin kuoli rintasyöpään tai se uusiutui. Lopuilla 44 potilaalla

oli triple-negatiivinen rintasyöpä.

DNA eristettiin CNA-kitillä (Qiagen) ja sen pitoisuus ja laatu mitattiin Qubit-fluorometrillä

(Thermo Fisher Scientific). Ne seeruminäytteet, joiden DNA-pitoisuus oli tarpeeksi korkea

tutkittiin castPCR PIK3CA -mutaatioanalyysillä. Näiden lisäksi 22 primäärituumori-DNA-

näytettä ajettiin castPCR:llä.

Tässä tutkimuksessa todettiin soluvapaan DNA:n pitoisuudella olevan assosiaatio tuumorin

koon ja reseptoristatuksen sekä relapsin kanssa. Potilailla, joilla oli korkeammat pitoisuudet

soluvapaata DNA:ta, oli korkeampi taipumus relapsiin sekä korkeampi kuolleisuus. castPCR

PIK3CA -mutaatioanalyysi ei havainnut H1047R-mutaatiota seeruminäytteistä, vaikka 9

primaarituumorinäytettä antoivat analyysissä positiivisen tuloksen.

Tämä tutkimus osoittaa sirkuloivien nukleiinihappojen potentiaalin rintasyövän

biomarkkereina. Soluvapaan DNA:n kvantifiointi voisi toimia rintasyövän ennusteen

markkerina syövän uusiutumiselle ja eloonjäämiselle. Mutaatioanalyysi ei kuitenkaan

löytänyt H1047R-mutaatiota seeruminäytteistä, joten menetelmää, jolla soluvapaata DNA:ta

tutkitaan kliinisessä ympäristössä, täytyy harkita. Jatkotutkimus on tarpeen, jotta soluvapaan

DNA:n käyttöön informatiivisena biomarkkerina rintasyövässä voidaan luottaa.

Abbreviations

ATM = ATM serine/threonine kinase gene

BRCA 1/2 = Breast cancer gene 1/2

BRIP1 = BRCA1 interacting protein C-terminal helicase 1 gene

castPCR = Competitive allele-specific TaqMan polymerase chain reaction

cfDNA = cell-free DNA

CHEK2 = Checkpoint kinase 2 gene

CNA = Circulating nucleic acid

COSMIC = Catalogue of somatic mutations in cancer

ctDNA = circulating tumor DNA

DCIS = Ductal carcinoma in situ

DNA = Deoxyribonucleic acid

IDC = Invasive ductal carcinoma

ER = estrogen receptor

FGFR2 = Fibroblast growth factor receptor 2 gene

HER2 = human epidermal growth factor receptor 2

H1047R = histidine to arginine substitution at position 1047 on PIK3CA

IPC = Internal positive control

MAP3K1 = Mitogen-activated protein kinase kinase kinase 1 gene

OS = Overall survival

PALB2 = Partner and localizer of BRCA2 gene

PIK3 = phosphatidylinositol-3-kinase

PIK3CA = phosphoinositide-3-kinase catalytic alpha polypeptide gene

PIK3CA = phosphatidylinositol-4,5-biphosphate-3-kinase catalytic subunit α

PR = progesterone receptor

PTEN = Phosphatase and tensin homolog gene

RFS = relapse-free survival

RNA = Ribonucleic acid

RT = room temperature

SNP = single nucleotide polymorphism

STK11 = Serine/threonine kinase 11 gene

TNM = Tumor, Node, Metastasis

TP53 = Tumor protein p53 gene

UICC = International Union Against Cancer

Acknowledgements

I had the opportunity to carry out this Master’s Thesis in the Department of Clinical Pathology

and Forensic Medicine at the University of Eastern Finland.

I had the privilege to work with a talented research team who took me under their wing and

who helped and guided me through this process. I would like to thank my supervisors PhD,

Associate Professor Arto Mannermaa and PhD, Hanna Peltonen for their guidance, support

and expertise. This project was very interesting and the results exciting. I am very grateful to

have been able to do this particular thesis work and with these individuals. Other than my

supervisors I would also like to extend my thanks to the whole research group at the

Department of Clinical Pathology and Forensic Medicine, including Helena Kemiläinen for

technical assistance and especially Eija Myöhänen, who was my mentor in the lab.

Also I want to express my thanks to my fellow students and my friends, who kept motivating

me and supporting me throughout the process. I won’t name names but you know who you

are and I am forever grateful.

TABLE OF CONTENTS

ABSTRACT……………………………………………………………………………….

TIIVISTELMÄ…………………………………………………………………………….

Abbreviations………………………………………………………………………………

Acknowledgements………………………………………………………………………..

LITERARY REVIEW ........................................................................................................... 10

1. Introduction ........................................................................................................................ 10

1.1. Breast cancer ............................................................................................................... 10

1.1.1 Classification ......................................................................................................... 11

1.1.2 Risk factors ............................................................................................................ 13

1.1.3 Diagnosis and treatment......................................................................................... 15

1.2 Tumor biomarkers ........................................................................................................ 16

1.3. Circulating cell-free nucleic acids as markers for breast cancer ................................. 17

1.3.1 Origin of cell-free DNA in the blood ..................................................................... 18

1.3.2 Amounts of cfDNA found in healthy individuals and cancer patients .................. 19

1.4. PIK3CA is a frequently mutated gene in sporadic breast cancer................................. 19

1.4.1. The H1047R mutation located on the catalytic domain of PIK3CA .................... 20

2. Aims of the study ............................................................................................................... 21

3. MATERIALS AND METHODS ....................................................................................... 22

3.1 Patients and samples ..................................................................................................... 22

3.1.1 Patients ................................................................................................................... 22

3.1.2 Serum samples ....................................................................................................... 22

3.1.3 Cell lines ................................................................................................................ 24

3.1.5 Primary tumor samples .......................................................................................... 24

3.2 DNA extraction ............................................................................................................ 24

3.2.1 Circulating nucleic acid extraction ........................................................................ 24

3.2.2 Extraction of MDA-MB-453 cell line DNA .......................................................... 24

3.3 Quantification of serum and MDA-MB-453 DNA ...................................................... 25

3.4 Competitive Allele-Specific TaqMan PCR .................................................................. 25

3.4.1 The castPCR protocol ............................................................................................ 26

3.4.2 Workflow ............................................................................................................... 27

3.5 Statistical analysis ........................................................................................................ 29

4. RESULTS .......................................................................................................................... 30

4.1 Quantification of cell-free DNA from patient serum samples ..................................... 30

4.2 Analysis of tumor-related cell-free DNA using PIK3CA mutation specific cast-PCR 32

4.2.1 The reference genome assay has high sensitivity at all tested concentrations ....... 32

4.2.2 MDA-MB-453 is a positive control for the H1047R mutation ............................. 33

4.2.3 The PIK3CA mutation assay has high sensitivity when mutation frequency is 100

% ..................................................................................................................................... 33

4.2.4 Dilution of the mutation frequency lowers sensitivity of the PIK3CA mutation assay

........................................................................................................................................ 34

4.2.5 Serum cell-free DNA samples are mutation negative when using PIK3CA mutation-

specific castPCR ............................................................................................................. 36

4.2.6 Association between patients’ clinicopathological characteristics and cell-free DNA

concentrations obtained from extraction......................................................................... 41

5. DISCUSSION .................................................................................................................... 50

5.1 Low tumor burden and prolonged storage of serum may account for lower cfDNA

concentration ...................................................................................................................... 51

5.2 Detecting tumor-related genetic changes in cell-free DNA ......................................... 52

5.2.1 Competitive Allele-specific TaqMan PCR is sensitive and specific ..................... 53

5.2.2 Quality control of castPCR using specified control samples ................................. 54

5.2.3 Patient serum DNA samples with triple negative or T1N0 breast cancer analyzed

with Competitive Allele-Specific TaqMan PCR PIK3CA mutation assay .................... 55

5.2.4 castPCR run of available primary tumor DNA samples and re-run of serum samples

with mutation positive primary tumor DNA................................................................... 55

5.2.5. No detection of H1047R mutation with high enough frequency from cfDNA using

PIK3CA mutation specific castPCR ............................................................................... 56

5.3 cfDNA concentration may be a viable prognostic marker in breast cancer ................. 57

5.4 Effects of disease progression and treatment must be considered when examining

cfDNA as a prognostic marker ........................................................................................... 58

5.4.1 Sequencing study shows correlation between common samples ........................... 59

5.4.2 Limited mutation detection range of castPCR may hinder its clinical application 60

6. Conclusions ........................................................................................................................ 60

7. References .......................................................................................................................... 61

10

LITERARY REVIEW

1. Introduction

1.1. Breast cancer

Breast cancer is one of the most prevalent cancers worldwide. In 2012 over 1.6 million new

breast cancer cases were diagnosed making it the most common cancer in women and the

second-most common cancer overall. One in four of all cancers in women is breast cancer and

it is ranked the fifth most common cancer-related cause of death worldwide. Breast cancer alone

accounts for approximately half a million deaths per year. (Ferlay et al., 2015)

In Finland 4831 new breast cancer cases were diagnosed in 2013 representing 30.6 % of all

cancers diagnosed in females that year. It has 22.4 % prevalence in the Finnish adult population

and caused 878 deaths in the year 2013. (Ferlay et al., 2015; the Finnish Cancer Registry, 2015)

Breast cancer can occur in men also but cases are very rare with 231 prevalent cases in Finland

in 2014 (the Finnish Cancer Registry, 2015).

Cancer is a heterogeneous disease. Cancerous growth begins when cells start to grow

uncontrollably, with regular cell cycle arrest dysfunctioning. This is usually due to DNA

damage which is not repaired by normal DNA repair systems. In a normal situation if DNA

damage is not repaired, the cell goes into apoptosis and dies, preventing damaged cells from

growing. Most mutations cause programmed cell death, however some remain. In cancer cells

this regulated cell death does not occur. Genes which attain mutations that cause cancerous

growth are called oncogenes. These genes encode proteins and enzymes that take part in normal

cell functions such as cell growth, however when these genes contain mutated DNA the proteins

encoded are faulty and cause changes in these functions. Mutations in DNA can be inherited or

they are caused by mistakes made in normal DNA replication processes or by environmental

factors. Most mutations are spontaneous. (Girish et al., 2014)

11

1.1.1 Classification

Breast cancers are classified according to the tissue that the cancer originates from. Solid tumors

derived from epithelial tissues, like tumors of the breast, are known as carcinomas (Alix-

Panabieres et al., 2012). Invasive ductal carcinoma (IDC) is the most common type of breast

cancer which represents up to 80 % of all cases (www.breastcancer.org;

www.cancerresearchuk.org). The anatomy of the female breast and its most prominent

structures can be visualized in Figure 1.

Tissue origin

Ductal carcinoma signifies that the cancer is of ductal origin, i.e. the cancer has originated from

the normal ductal tissue of the breast. It can be classified as ductal carcinoma in situ (DCIS) or

invasive ductal carcinoma (IDC). In situ indicates that the cancer is growing locally and has not

yet invaded surrounding tissue, in particular the basement membrane. In situ carcinoma’s

priority treatment is dissection of the mass with enough margin to be sure that all cancerous

cells are removed. This type of breast cancer can be of varying size and it is usually benign.

However, ductal carcinoma can become malignant, meaning that it is able to invade

surrounding tissues and spread to other organs of the body via the blood stream and lymph

vessels. It is not known whether breast cancer always starts out as in situ from which it evolves

into a malignant cancer, or if it can be in situ without connection to the malignant or vice versa.

Some studies support the notion of DCIS being a precursor to IDC due to similar non-genetic

risk factors. (Petridis et al., 2016)

Breast cancer can also be lobular, which originates from milk-producing glands (lobules) in the

breast. Lobular breast cancer is much less common; only about 1 in 10 cases are lobular. There

are also very rare forms of breast cancer including tubular, medullar, and papillary carcinoma,

in which the names indicate where the cancer originated from. Other rare forms are Paget

disease and inflammatory breast cancer. (Abdulkareem, 2013)

12

Figure 1. Anatomy of the female breast. (National Cancer Institute, 2011)

Receptor status of breast cancer

Breast cancer cells have protein receptors which are located either on the cell surface or in the

nucleus. Breast cancer can be classified according to which receptors its cells express, with

three major receptors being important markers for classification. These three receptors are the

estrogen receptor (ER), the progesterone receptor (PR) and the human epidermal growth factor

receptor 2 (HER2). The majority of breast cancers are ER-positive, where the estrogen receptors

are amplified. Breast cancers can also be progesterone receptor amplified or HER2-amplified.

Breast cancers can express one, two or three of these receptors. Breast tumors which do not

express estrogen or progesterone receptors and have lacking HER2 amplification are classified

as triple negative. Triple negative breast cancers have the worst prognosis due to there being no

receptor targets for medical treatments and can only be treated with non-specific therapies such

as chemotherapy and surgical excision. (Vuong et al., 2014; Griffiths and Olin, 2012)

The TNM classification system

The TNM classification system is a specific classification system for breast cancer which

represents the status of the cancer according to its size and invasiveness; the T indicates the size

of the solid primary tumor, N the status of the lymph nodes under the arms and M the state of

metastasis (UICC, 2009). A complete table of the classification system can be seen in Table 1.

13

Table 1. The TNM breast tumor classification system (UICC, 2009)

Primary tumor (T)

TX Primary tumor cannot be assessed

T0 No evidence of tumor

Tis Carcinoma in situ

T1

T1a

T1b

T1c

Tumor is < 2 cm in its greatest dimension

Tumor is > 0.1 cm but less than 0.5 cm

Tumor is more than 0.5 cm but less than 1 cm

Tumor is more than 1 cm but not more than 2 cm

T2 Tumor is > 2 cm but no more than 5 cm in its greatest dimension

T3 Tumor is > 5 cm in its greatest dimension

T4 Tumor has attached to chest wall and/or skin

Regional lymph nodes (N)

NX Regional lymph nodes cannot be assessed

N0 No regional lymph node metastasis

N1 Metastasis in ipsilateral level I and II axillary lymph node(s)

N2

N2a

N2b

Metastasis in ipsilateral level I and II axillary OR internal mammary lymph

node(s)

Metastasis in axillary lymph node(s) fixed to one another or to other structures

Metastasis in internal mammary lymph node(s) in the absence of metastasis in

axillary lymph node(s)

N3

N3a

N3b

N3c

Metastasis in level III axillary ipsilateral infraclavicular lymph node(s)

Metastasis in infraclavicular lymph node(s)

Metastasis in internal mammary and axillary lymph node(s)

Metastasis in supraclavicular lymph node(s)

Metastasis (M)

M0 No distant metastasis

M1 Distant metastasis

1.1.2 Risk factors

Genetic factors

Breast cancer is caused by genetic abnormalities such as somatic mutations and epigenetic

changes. These genetic changes can be either sporadic or hereditary. Approximately 10 % of

breast cancers cause a highly increased inherited risk of the disease for first-degree relatives.

Hereditary breast cancers, as any other hereditary diseases, are caused by genetic mutations in

the germ line, which can be passed on from mother to daughter. (Kenemans et al., 2008) These

mutations cause susceptibility to breast cancer. Two of the most known susceptibility factors

14

are breast cancer susceptibility gene 1 and 2, BRCA1 and BRCA2 respectively (Campeau et al.,

2008; Wooster and Weber, 2003). These two genes can cause over a 60 % increase in risk of

breast cancer if mutations occur and are known as high-penetrance genes (Wooster and Weber,

2003). There are also other so called high-risk hereditary breast cancer susceptibility genes

which include tumor suppressors TP53, PTEN and STK11. Tumor suppressors, in normal

conditions, have roles in cell cycle repression and apoptosis. Breast cancer susceptibility genes

give different levels of increased risk. Genes of intermediate risk include ATM, CHEK2, PALB2

and BRIP1. (Shiovitz and Korde, 2015; Campeau et al., 2008) Breast cancer hereditary risk

genes give a predisposition to cancer, meaning and increased risk of cancerous cell growth, but

somatic mutations are necessary for cancer to evolve. It is these mutations which cause the

genes to become oncogenes and cause the predisposition. (Shiovitz and Korde, 2015) New loci

for breast cancer susceptibility are constantly researched and found, with 76 known low

penetrance loci that account for approximately 35 % of hereditary breast cancer risk (Petridis

et al., 2016; Michailidou et al., 2013). However researchers are mainly focusing on those which

have a high frequency in breast cancer patients, such as the ones mentioned above (Shiovitz

and Korde, 2015).

Most breast cancers are sporadic, i.e. the genetic alteration that causes evolvement of the tumor

is not inherited but acquired (Kenemans et al., 2008). These somatic mutations are usually

single point mutations caused by single nucleotide polymorphisms (SNPs) that cause damage

to DNA and are not corrected by the normal DNA correcting system. Sporadic breast cancer

requires multiple “hits”, meaning more than one acquired uncorrected mutation in somatic

genes, to cause the formation of cancer cells. The cell fails to go to apoptosis and so it divides,

giving daughter cells the same damaged DNA. Uncontrolled cell division is the basis of all

cancers. Cancer requires more of these so called “hits” in order to evolve further, for example

to become invasive, metastasize or develop angiogenesis. In sporadic breast cancer only few

genes are mutated frequently with PIK3CA and TP53 among them (Nik-Zainal et al., 2012).

Low penetrance or low risk genes which affect to sporadic breast cancer have been found

through genome wide association studies and the most common ones are FGFR2 and MAP3K1.

(Campeau et al., 2008)

It is important to recognize the genetic alterations each individual patient has as they are specific

to each cancer and oncologists can target these alterations when deciding on treatment for

patients diagnosed with breast cancer. By genotyping a patient cancer oncologists can recognize

the mutations it harbors and find a drug that would reverse the functions of these mutations.

15

They can then decide what treatment to pursue and even more importantly researchers can get

new knowledge about the genetics of breast cancer and develop new drugs and new treatment

methods. Oncologists and researchers work together to provide the best possible treatment for

each patient.

Environmental factors

There are many environmental factors which can increase an individual’s risk of breast cancer.

These factors include lifestyle habits and other factors that cannot be controlled, such as age.

Lifestyle habits such as smoking, exercise and use of alcohol can greatly effect ones risk of

breast cancer. Many risk factors are related to overall female estrogen exposure. After

menopause the risk is greatly increased when female estrogen hormone production changes.

The risk of getting breast cancer in Finland before the age of 75 is 10 %, meaning one in ten

females will be diagnosed with breast cancer before their 75th birthday (The Finnish Cancer

Registry, 2015). One controllable factor is the age that one chooses to reproduce; childlessness

can increase the risk of breast cancer. Menstruation and pregnancy have an effect on overall

estrogen production, which in turn effects development of breast cancer. (Abdulkareem, 2013)

1.1.3 Diagnosis and treatment

As for any cancer, early diagnosis is key to survival. Breast cancer is often not diagnosed until

symptoms occur. Breast cancer diagnosis is done by a physician by performing a breast

examination and mammography which is essentially an X-ray of the breast. An ultrasonography

may also be performed, which might give more information of the nature of the finding. If

something is found, a sample or biopsy can be taken to determine whether the finding is benign

or malignant and how to proceed. (Girish et al., 2014)

Breast cancer can be treated with surgery, which is primary, and hormone therapy targeting

ER/PR/HER2, chemotherapy or radiation therapy. For early stage breast cancers the standard

treatment is excision of the tumor with or without additional treatment. For in situ cancers the

tumor mass is always excised by surgery, usually without a combination of other treatment.

Malignant cancers are treated with a combination of treatments depending on the type of breast

cancer. (Girish et al., 2014; Le Du et al., 2013) Triple negative breast cancer can only be treated

by chemotherapy, radiation therapy and/or surgery as it expresses no receptors that can be

specifically targeted (Griffiths and Olin, 2012).

16

As important as finding the right treatment is, it is equally important to monitor the patient’s

response to the treatment. Proper monitoring helps oncologists further optimize the treatment

to be patient specific. Personalized treatment is one the main goals in cancer research today.

Also monitoring shows which treatments are ineffective so these therapies can be avoided.

The main current monitoring method is by a primary tumor biopsy. A tissue biopsy is highly

invasive, and as monitoring of the tumor must be done frequently it is not ideal. Also the

primary treatment after diagnosis is excision of the primary tumor and so a tissue biopsy cannot

be done. A new method of taking a biopsy of tumors is being researched, the so called liquid

biopsy. A liquid biopsy is for example a simple blood sample, which contains tumor DNA and

so gives a snap-shot of the primary tumor. This type of biopsy is non-invasive and is easy for

both the patient and physician. It can also be taken even though the primary tumor has been

surgically removed.

1.2 Tumor biomarkers

Biomarkers are different types of molecules, genes, proteins etc. something biological which

gives an indication of something happening in the body. Tumor biomarkers can indicate the

presence of a tumor and its characteristics and they are classified as something produced either

by the tumor or in response to the tumor. (Paoletti and Hayes, 2014; Vuong et al., 2014)

One of the most important factors in overall survival for a cancer patient is how early the disease

is detected; the earlier a tumor is detected, the better. Biomarkers make early diagnosis possible.

Overall survival is greatly affected when the disease is diagnosed while still asymptomatic. Not

only early diagnosis, but at a later stage early identification of possible relapse is essential to

conquering the disease (Reid et al., 2015; Crowley et al., 2013). Another important aspect of

success in treating breast cancer is the ability to monitor disease progression and the effect of

treatment to decrease use of toxic non-effective drugs (Dawson et al., 2013; Elshimali et al.,

2013). Personalized cancers require personalized therapies.

Tumor biomarkers are needed to measure tumor burden (Diaz and Bardelli, 2014). They can

give a more accurate prognosis and assessment of overall survival. Biomarkers also give us an

assessment of what stage the cancer is in and can in this way help evaluate how to proceed with

treatment.

The most used biomarkers currently are the BRCA1 and 2 genes along with the hormone

receptor status (ER/PR/HER2) of the tumor. New biomarkers are continually studied and

17

verified including markers such as DNA and microRNAs. Tumor DNA contains tumor specific

mutations and these mutations could be potentially used as biomarkers. (Paoletti and Hayes,

2014; Vuong et al., 2014)

In the next section one of the most recently discovered candidates for a cancer biomarker will

be discussed.

1.3. Circulating cell-free nucleic acids as markers for breast cancer

Circulating or cell-free nucleic acids (CNAs) are fragments of DNA or RNA that circulate in

the bloodstream free from confinement within cells. CNAs can form complexes with

proteolipids, be contained in apoptotic vessels or in a complex with the p53 protein or “naked”

(Schwarzenbach, 2013; Fleischhacker and Schmidt, 2007). Small amounts of extracellular

DNA can be found in the blood of all humans, including healthy individuals. The amounts differ

between individuals, with different aspects affecting to this such as disease status.

Concentrations in healthy individuals is very low due to removal by phagocytes (Elshimali et

al., 2013) but increasing amounts of CNAs have been found in cancer patients (Dawson et al.,

2013; Fleischhacker and Schmidt, 2007; Anker et al., 1999). The variability of results from

studies investigating cfDNA concentrations between cancer patients and healthy individuals

must be noted. Some studies state that they were able to discriminate between healthy

individuals and cancer patients, but could not indicate whether the patients had benign or

malignant breast lesions relying only to cfDNA concentration (Schwarzenbach, 2013). Others

on the other hand show increasing amounts in patients with advanced disease and patients with

low disease burden presenting a CNA amount of a healthy individual (Anker et al., 1999).

Diseases other than cancer have also been shown to increase CNA levels, including

inflammatory bowel disease, hepatitis and rheumatoid arthritis (Anker et al., 1999). CNA levels

have been shown to change also in states of inflammation and for example after periods of

extensive exercise (Fleischhacker and Schmidt, 2007).

All knowledge of CNAs is still considered to be hypothetical with more research needed to

know exactly how and why CNAs are released into the bloodstream in normal and pathological

conditions.

Recent discoveries have found cancer-related genetic changes also in the serum and plasma of

cancer patients which further indicates the presence of tumor-derived DNA in the blood. These

discoveries open up a new prospect in cancer diagnostics, monitoring and treatment. With a

18

liquid biopsy characteristics of the cancer could be seen by merely taking a blood sample

compared to a highly invasive tissue biopsy. A biopsy of the cancer is vital in order for correct

treatment methods to be chosen as well as for monitoring the effect of treatment. A liquid biopsy

would be ideal for monitoring treatment, as it is much less invasive and the procedure is much

simpler for the physician as well as for the patient. It is also less time consuming and

inexpensive. Another important implication is the biomarker potential of circulating cell-free

DNA, which could be accessed via liquid biopsy. (Schwarzenbach, 2013)

1.3.1 Origin of cell-free DNA in the blood

The origin of cell-free DNA (cfDNA) in the blood is not yet fully understood. It was first

discovered in 1948 but it was not until the 1970’s when it started to peak interest and was

investigated in cancer patients (Leon et al., 1977). Leon et al. (1977) showed the presence of

cell free DNA in the blood of healthy individuals and of cancer patients. The origin of the DNA

was thought to be due to normal tissue and cell injury which would cause release of DNA into

the bloodstream. This release would be increased in the pathologic condition of cancer.

Apoptosis has been confirmed as a key source of cell-free DNA in the bloodstream (Elshimali

et al., 2013). In normal conditions cell injury leads to apoptosis and the release of DNA into the

circulation whereas in pathologic conditions the release is due to either apoptosis or necrosis of

cells (Schwarzenbach, 2013; Fleischhacker and Schmidt, 2007; Jahr et al., 2001). Other sources

of cfDNA are cell lysis by necrosis, spontaneous release, blood cell lysis and break down of

pathogens (Elshimali et al., 2013). It has also been speculated whether cancer actively releases

DNA into the circulation as preparation for metastasis, but the method for this is not yet known

(Schwarzenbach, 2013; Jahr et al., 2001).

Circulating DNAs are double-stranded in various fragment sizes ranging from under 180 base

pairs to over 30 kilobases (Elshimali et al., 2013; Fleischhacker and Schmidt, 2007). This

varying size can hinder its detection. Cell-free DNA has been mainly extracted from plasma or

serum using commercial DNA extraction kits. Whether plasma or serum is the better material

for extraction has been unclear, with studies stating pros and cons for both (Elshimali et al.,

2013). cfDNA levels are shown to be generally higher in serum than in plasma, but this is agreed

to be due to possible release of DNA from hemapoietic cells during the clotting process

(Benesova et al., 2013; Schwarzenbach, 2013).

19

1.3.2 Amounts of cfDNA found in healthy individuals and cancer patients

In healthy controls the amount of cfDNA is minimal and even at non-detectable levels as was

the case with 42 out of 55 tested healthy individuals in the study by Leon et al. (1977). The

range for these 55 individuals was from 0 to 50 ng/ml. The range for cfDNA of the 173

cancerous patients serum tested was from 0 to 5000 ng/ml with a mean of 180 ng/ml. This study

demonstrates the broad range of cell free DNA in pathologic conditions and Leon et al. (1977)

point out that this may hinder the diagnostic value of a liquid biopsy with a relatively large

proportion of cancer patients showing normal amounts of cfDNA. It must be kept in mind that

the study is quite dated and methods for quantifying cfDNA are not as sensitive as today.

However, more recent studies do concur with these findings showing mean concentrations from

less than 10 ng/ml to over 1500 ng/ml (Elshimali et al., 2013), with one study showing as low

as 1 ng/ml in healthy individuals (Board et al., 2010). Nearly all of the patients in Board and

colleagues’ (2010) study had received some form of treatment and it has been shown that there

are drops in the amount of cfDNA in cancer patients after treatment such as surgery (Elshimali

et al., 2013). Leon et al. (1977) suggest that the biopsy should be taken before initiation of

treatment. The concentrations of cfDNA varies between individuals whether they are healthy

or have cancer.

As concentration of cfDNA is believed to be increased in the disease state of cancer, it must be

concluded that the increase is due to tumor DNA in the blood. The proportion of cfDNA of

tumor origin is dependent on factors such as tumor size and state of metastasis, however each

patient is an individual and not all factors are known (Schwarzenbach, 2013). Now the tide is

turning from just quantitating the cfDNA found in cancer patients’ plasma/serum to qualifying

how much of the free DNA is actually of tumor origin and characterizing the DNA in order to

identify characteristics in the primary tumor.

1.4. PIK3CA is a frequently mutated gene in sporadic breast cancer

The phosphoinositide-3-kinase catalytic alpha polypeptide gene or PIK3CA is a 34 kb sized

gene located on chromosome 3 and is composed of 20 coding exons. It encodes the

phosphatidylinositol-4.5-biphosphate-3-kinase catalytic subunit α (p110α or better known as

PIK3CA) of a regulatory kinase PI3K (phosphatidylinositol 3-kinase). PI3K is part of an

essential signaling pathway which regulates cell proliferation among other important functions

such as cell motility and morphology. (Oshiro et al., 2015; Bader et al., 2005)

20

The PIK3CA gene is known to have frequently occurring alterations that have been linked to

different types of solid cancers in humans, including breast cancer. Somatic mutations in this

gene are present in 15 % of human cancers and it is the most commonly mutated gene in patients

with breast cancer, with a frequency of up to 40 % (Oshiro et al., 2015; Schwarzenbach, 2013;

Higgins et al., 2012; Karakas et al., 2006). PIK3CA harbors three so called ‘hotspot’ mutations

which frequently occur in the oncogene in cancer patients. These mutations, H1047R, E545K

and E542K, account for up to 80 % of all PIK3CA mutations in breast cancer (Oshiro et al.,

2015; Board et al., 2010; Karakas et al., 2006; Bader et al., 2005). As it has a major role in

regulation of cell growth, PIK3CA is thought to give rise to cancerous growth when its activity

is enhanced (Board et al., 2010; Karakas et al., 2006).

It has been speculated whether PIK3CA mutations could be used as prognostic markers in

addition to monitoring tumor status. In a recent study patients with PIK3CA mutated circulating

tumor DNA (ctDNA) had worse prognosis with lower relapse-free survival (RFS) and overall

survival (OS) rates than did patients without these mutations. (Oshiro et al., 2015)

1.4.1. The H1047R mutation located on the catalytic domain of PIK3CA

One of the most common oncogenic alterations in the PIK3CA gene is a somatic missense

mutation located on exon 20 called H1047R, where the amino acid histidine has been

substituted by arginine at position 1047 (Figure 2). Exon 20 encodes the catalytic domain of

the protein. Somatic mutations in exon 20 have been reported in multiple studies before the

year 2006 with a combined frequency of 26 % in breast cancer patients (Karakas et al., 2006).

According to the COSMIC database (Catalogue of somatic mutations in cancer, Wellcome

Trust Sanger Institute, Cambridge, UK), H1047R is the most frequent mutation in sporadic

breast cancer. It is a substitution mutation where adenine is substituted with guanine causing a

missense read frame.

21

Figure. 2 The p110α subunit of PIK3CA which harbors the H1047R mutation in its catalytic domain. (Adapted

from (Kalinsky et al., 2009)

2. Aims of the study

This study constitutes to a larger biomarker study which strives to find tools for oncologists for

earlier diagnosis and to aid in planning more efficient personalized treatments. Ultimately the

purpose was to find a plausible reason for why patients with the same prognosis have such

differing outcomes with their disease and to possibly discover new knowledge about the reasons

behind the poor prognosis of triple negative breast cancer. The goal was to investigate whether

cell-free tumor DNA could be used as a biomarker in breast cancer. This was done two ways:

firstly studying whether a single point mutation could be found from cfDNA, in effect detecting

DNA which is of tumor origin and secondly examining the relationship between cfDNA

concentration and clinical variables and survival. The main aim of this study was to identify the

H1047R mutation of the PIK3CA gene from blood serum DNA of patients with early-stage

breast cancer and to find connections between cfDNA concentration and outcome of the

disease.

22

3. MATERIALS AND METHODS

3.1 Patients and samples

3.1.1 Patients

Patient samples were acquired through the Kuopio Breast Cancer Project (Mannisto et al.,

1999). Samples were taken from women with all types of breast cancer at the time of diagnosis

during the years 1990-1995 in Kuopio University Hospital. These women are still followed-up

on intensely with the most recent follow-up session done in 2015 and they have had additional

samples taken over the years. Altogether samples from 82 patients were studied in this project.

3.1.2 Serum samples

Serum is a component of blood which does not contain blood cells or coagulation factors. Serum

is obtained by centrifugation, to remove cellular components after which the remaining plasma

is allowed to clot.

Two sets of serum samples were chosen for this study. The first set of 38 samples were chosen

according to the criteria of initial diagnosis of ductal breast carcinoma with the status T1, N0.

In this case T1 means that the primary tumor is less than 2 cm in diameter and N0 that the tumor

has not spread to the lymph nodes under the arms (UICC, 2009). A complete table of the

classification system can be seen in Table 1. Out of the 38 samples, 19 had ductal carcinoma

that lead to relapse and/or death while the remaining 19 had ductal carcinoma but no relapse.

The first 19 study samples are compared to the other 19 breast cancer samples with same

clinicopathological characteristics, excluding relapse and/or death. These samples were

matched up to each other as well as possible, including criteria such as age, treatment and breast

cancer type. These samples were chosen because of the abnormality in their cancer

development; the T1, N0 status indicates a form of breast cancer with a good prognosis,

unfortunately these 19 patients had poor outcomes. From the patients with good prognosis, but

bad outcome, 9 have died of causes related to breast cancer and 8 have gone through relapse.

From the 19 patients with good prognosis and good outcome 15 are still alive today with no

relapse and 4 have died of causes not directly related to breast cancer.

The second set of 44 patient samples were chosen with the sole criteria of having triple negative

breast cancer. The triple negative form is known to be the most deadly form of breast cancer as

it has limited treatment possibilities (Griffiths and Olin, 2012). From the 44 patients with triple

23

negative breast cancer 12 have died of causes related to breast cancer and 4 have gone through

relapse. Clinicopathological data of all the patients are summarized in table 2.

A blood serum sample from a healthy individual was used as the negative control in this study.

Table 2. Clinicopathological data of the patients

All patients (N=82) T1N0 patients (N=38) TN patients (N=44)

Clinical variable N (%) N (%) N (%)

Age

≤ 52 43 (52.4) 20 (52.6) 23 (52.3)

> 52 39 (47.6) 18 (47.4) 21 (47.7)

Status

alive 32 (39.0) 15 (39.4) 17 (38.6)

alive, relapse 12 (14.6) 8 (21.1) 4 (9.1)

dead 17 (20.8) 6 (15.8) 11 (25.0)

dead, breast cancer 21 (25.6) 9 (23.7) 12 (27.3)

Histology

Ductal 69 (84.1) 38 (100) 31 (70.5)

Lobular 3 (3.8) 0 3 (6.8)

Other 10 (12.1) 0 10 (22.7)

Stage

in situ 2 (2.4) 0 2 (4.6)

I 56 (68.3) 38 (100) 18 (40.9)

II 18 (22.0) 0 18 (40.9)

III 6 (7.3) 0 6 (13.6)

Grade

1 11 (13.4) 9 (23.7) 2 (4.5)

2 29 (35.4) 22 (57.9) 7 (16.0)

3 40 (48.8) 7 (18.4) 33 (75.0)

Unknown 2 (2.4) 0 2 (4.5)

Estrogen receptor

positive 33 (40.2) 33 (86.9) 0

negative 48 (58.6) 4 (10.5) 44 (100)

Unknown 1 (1.2) 1 (2.6) 0

Progesterone receptor

positive 28 (34.2) 28 (73.7) 0

negative 53 (64.6) 9 (23.7) 44 (100)

Unknown 1 (1.2) 1 (2.6) 0

24

3.1.3 Cell lines

The cell lines used in this study were MDA-MB-453 (MDA) and SKOV-3 (SKOV). These cell

lines were previously known to carry the H1047R mutation of the PIK3CA gene (ATCC, 2014)

and were available as cell pellets stored at -80 °C. The MDA cell line DNA was used as a

positive control in this study.

3.1.5 Primary tumor samples

DNA extracted from primary tumor tissue was available from 22 samples in common with the

serum samples. The DNA was stored at – 80 °C in TE-buffer.

3.2 DNA extraction

3.2.1 Circulating nucleic acid extraction

CNAs were isolated from serum using the QIAamp Circulating Nucleic Acid kit (QIAGEN,

Hilden, Germany) according to manufacturer’s instructions.

Before extraction the CNA-kit buffers were prepared according to the manufacturer’s

instructions. All buffers were provided by the manufacturer. Also the equipment required for

extraction was set up. The equipment included a vacuum pump (600 mbar), a vacuum manifold

compatible with QIAamp Mini columns provided by the manufacturer and a connecting waste

system. All other equipment used can be viewed from the manufacturer’s manual.

Prior to extraction serum samples were thawed at room temperature and 1 ml was taken for

extraction. Each 1 ml of sample was first centrifuged at 2000 g for 10 min at room temperature

(RT). After this the extraction was done from the supernatant. This was done to rid the sample

of possible white cell debris which is associated with DNA extraction from serum

(Schwarzenbach, 2013).

Extracted DNA was eluted with 50 µl of AVE buffer and stored at -20 °C.

3.2.2 Extraction of MDA-MB-453 cell line DNA

A batch of MDA-MB-453 cells was stored at -80 °C. The amount of cells was not known and

so the amounts of reagents used were approximated.

25

On the first day 0.5 ml of lysis buffer, 50 µl of 10 % sodium dodecyl sulfate and 10 µl of

proteinase K were added to the cell pellet and mixed gently by turning the tube. The tube was

incubated overnight in 37 °C.

On the next day the DNA was extracted and precipitated. First 250 µl of phenol and 250 µl of

chloroform was added to the Eppendorf tube. The suspension was mixed by turning the tube

upside-down for approximately 15 min on a mechanical mixer. The tube was then centrifuged

for 15 min at RT to separate the phases. The topmost phase was collected and the same steps

starting the extraction protocol were carried out. Again the topmost phase was collected and the

extraction repeated, however this time with only chloroform 500 µl.

Next the DNA was precipitated by adding 1/10 of the volume (50 µl) of 3M sodium acetate and

2 volumes (1 ml) of - 20 °C absolute ethanol and then mixed by turning. The now visible DNA

strand was moved to a clean tube and washed with 500 µl of 70 % ethanol by turning the tube.

To dry the DNA the tube was centrifuged twice for approximately 2 min at RT with removing

the supernatant after the first centrifugation and then incubated with the lid open for 15 min in

a fume hood. Then the DNA precipitate was dissolved into TE-buffer. TE-buffer was added

several times and incubated at RT until the DNA had dissolved. All in all 450 µl of TE-buffer

was added with 24 h of incubation including 4 h on a shaker for the DNA to be dissolved.

3.3 Quantification of serum and MDA-MB-453 DNA

Serum DNA samples and MDA DNA were quantified using Qubit fluorometer (Thermo Fisher

Scientific Inc., Waltham, MA, USA). The samples were prepared according to the

manufacturer’s instructions. Serum DNA samples were diluted into Qubit mastermix with the

ratio of 190 µl of mastermix and 10 µl of sample DNA and MDA DNA samples with the ratio

of 199 µl of mastermix and 1 µl of sample DNA. Qubit double stranded DNA BR assay reagent

containing a fluorescent marker was added. The Qubit fluorometer determines DNA

concentration by measuring fluorescence emitted by the double stranded DNA. The Qubit

fluorometer is very sensitive minimizing the chance to detect contaminants, as it does not detect

single stranded nucleic acids, giving a high quality DNA concentration.

3.4 Competitive Allele-Specific TaqMan PCR

The TaqMan Mutation Assay uses Competitive Allele-Specific TaqMan PCR (castPCR)

technology as a tool to detect rare mutations in a pool of normal DNA. This is an assay which

26

can be applied to the detection of tumor-originated DNA from normal DNA in a liquid biopsy

sample by screening for and amplifying cancer related mutations. It makes it possible to identify

1 mutant allele in a pool of 1000 wild-type alleles, meaning a sensitivity of 0.1 %. CastPCR is

a mutation detection assay in which an allele-specific primer detects a mutant allele and an

allele-specific blocker suppresses the wild-type allele thus ensuring amplification of the target

and blocking non-specific amplification. A locus-specific TaqMan probe, containing a

fluorescent marker, binds to both mutant and wild-type DNA of the target gene. The mutation

is amplified by a mutation specific primer and a fluorescent signal is detected during a PCR

run. The blocker stops the primer from attaching to wild-type DNA ensuring amplification of

the mutant allele and no amplification of the wild-type allele. In this study the TaqMan mutation

detection assay for the H1047R mutation of the PIK3CA gene was used (ID: Hs00000831_mu).

(Thermo Fisher Scientific Inc. Waltham, MA, USA, 2015)

A reference mutation assay can be used alongside the castPCR mutation assay. The reference

genome assay detects the gene of interest by using a locus-specific primer which attaches itself

to both mutant and wild-type allele containing genes. This primer amplifies a “normal” part of

the gene and can be used to ensure the presence of the gene. The reference genome assay used

in this study was the PIK3CA_rf kit (ID: Hs00001025_rf).

An internal positive control (IPC) is used in the assays to differentiate a target negative result

from a PCR failure.

3.4.1 The castPCR protocol

After sample serum DNA was extracted, samples were run with castPCR. Samples were diluted

in nuclease-free water for each castPCR run so that all samples on the same plate in the same

PCR run were of equal concentration, with the exception of one run in which DNA was not

diluted. The sample amounts per well varied between 1 ng and 20 ng for different runs. The

manufacturer recommends 20 ng, but because of low DNA concentrations in our samples we

used less. The manufacturer states that 1 ng of sample may be used, but the sensitivity is much

lower as they guarantee a sensitivity of only 1.0 % when 2 ng of sample is used. The castPCR

protocol included preparation of a supermix to which sample and assay would be added (Table

3). The reaction volume used for one sample was 20 µl, containing 2 or 4 µl of sample and 2 µl

of either mutation assay or reference genome assay, run on a 96-well plate. Positive and

negative controls were used in all runs as well as a non-template control. Every sample was run

twice on the same plate, once with the mutation assay and once with reference genome assay.

27

Both sensitivity tests and runs with study samples were done. The manufacturer recommends

using 40 cycles in the thermo cycling program. In some runs 50 cycles were used due to low

DNA concentrations in our samples. The thermocycling program and conditions used can be

viewed in Table 4.

Table 3. Contents of the PCR supermix used in castPCR analysis

castPCRsupermix 1x (reaction volume for one sample 20 µl)

TaqMan Genotyping Master Mix, 2x 10 µl

50 X Exogenous IPC Template DNA 0,4 µl

10X Exogenous IPC Mix 2,0 µl

Nuclease free water 1,6 µl

Prepared gDNA (added separately to each well) 2-4,0 µl

Total 18 µl

Table 4.Thermocycling PCR program

Stage Temperature Time

(mm:ss)

Cycles

1 95 10:00 1

2 92 00:15 5

58 01:00

3 92 00:15 40-50

60 01:00

3.4.2 Workflow

The workflow of this project can be visualized in three separate charts, with DNA extraction

and reference genome assays in Figure 3 and castPCR assays in Figure 4. DNA was extracted

from all 82 samples and quantified. The reference genome assay was tested for its sensitivity

using a sample of wild-type DNA, which was then used as a negative control in following

mutation detection castPCR runs.

28

Figure 3. Workflow of serum DNA extraction and castPCR PIK3CA reference genome assay.

Figure 4. Workflow of castPCR, containing details of each run. Sensitivity tests with diluted frequency have

been done with mutation frequencies of 10 %, 5 %, 2.5 %, 1 %, 0.1 % and 0.01 %

29

3.5 Statistical analysis

Statistical analysis was performed with the SPSS program version 21.0 (IBM Co., Armonk,

NY, USA). Samples were distributed into groups based on cfDNA concentration including five

different types of distribution: median, two types of percentiles, extremities and high

concentration against the rest. These distributions have been clarified in Figure 5. The groups

were then compared to see whether they had significant difference according to important

clinical variables including cancer histology, stage, grade, receptor status and relapse. Chi-

square analysis and Mann-Whitney two-sample rank-test were used. The clinical variables

found significantly different (relapse and receptor status) between sample groups in chi-square

analysis were used as covariates in the Mann-Whitney analysis. The nodal status and the tumor

size classification of patients has been omitted due to patients being chosen with certain criteria

according to these variables and the distribution is therefore not random. Survival rates were

analyzed using Kaplan-Meier and Cox regression survival analyses. Multiple test correction

was not done.

Figure 5. Clarification of samples

distributed into groups according to

cfDNA concentration used in statistical

analysis.

30

4. RESULTS

4.1 Quantification of cell-free DNA from patient serum samples

Cell-free DNA was extracted from patient serum samples with the QIAamp CNA-kit

(QIAGEN, Hilden, Germany). Detectable levels of cfDNA were obtained from 56 out of 82

patient serum samples (68 %). The castPCR analysis requires a reaction volume of 20 µl which

includes a maximum of 4 µl of sample. Only those samples from which the manufacturers

recommended minimum of 1 ng of sample could be used in the following PCR assay were

further analyzed. This means that a cfDNA sample could be used only if its concentration was

higher than or equal to 0.25 ng/µl. This dropped out three additional samples making the total

amount of serum cfDNA samples that went into castPCR PIK3CA-mutation analysis 53. In

Table 5 measured DNA concentrations from all samples can be seen. The 26 samples that are

shown to have a concentration of 0 ng/µl do not necessarily have no DNA, the instrument used

to measure the concentration merely was not sensitive enough to detect such small amounts of

DNA. These samples would not have been able to be used in further analysis even if the

instrument would have been sensitive enough.

31

Table 5. A table showing the measured cfDNA concentrations from each sample in ng/µl. cfDNA

levels were measure with Qubit. PNRO = patient number, cfDNA= cell-free DNA

PNRO cfDNA ng/µl PNRO cfDNA ng/µl PNRO cfDNA ng/µl

75 0.354 720 0.238 1618 0.928

78 1.11 732 0 1629 0

106 0 735 0.536 1634 0.27

124 0 762 0.25 1653 0.286

130 1.29 839 0 1670 0

134 1.03 867 0.412 1672 0

205 0.68 910 0.48 1677 0

213 1.99 916 0 1679 0

313 0.6 938 0.574 1680 0.508

316 0.606 940 0 1685 0.426

317 0.786 945 0.594 1707 0.532

372 0 975 0 1735 0.206

389 0.97 985 0.256 1746 0

395 0.254 997 1.52 1750 0.61

416 0.244 1004 0.586 1751 1.91

435 0.774 1021 0.594 1763 0

458 1.4 1029 0 1788 0

462 0.896 1280 0.54 1798 0.276

467 1.67 1390 0 1801 0.914

558 0 1414 0 1810 0

565 0.77 1472 0.466 1814 1.41

566 0.254 1496 1.36 1822 0.426

613 0 1497 0 1857 0.464

616 2.12 1505 0.75 1863 0.266

622 0.292 1513 0.812 1868 0.928

670 0 1575 0 1904 0.594

699 1.97 1596 0

706 0.23 1602 0.388

32

4.2 Analysis of tumor-related cell-free DNA using PIK3CA mutation specific cast-

PCR

4.2.1 The reference genome assay has high sensitivity at all tested concentrations

The castPCR assay comes in two parts: the reference genome assay which amplifies a portion

of the gene in question which does not include the mutation and the PIK3CA mutation assay

which amplifies only the mutation. The first thing that was done was to test the sensitivity of

the reference genome assay with wild-type serum by making a dilution series of the sample.

The sample was diluted to concentrations of 10 ng/µl, 5 ng/µl, 2.5 ng/µl, 1.25 ng/µl and 0.625

ng/µl. After addition of 2 µl of each dilution per well the sample amounts in the assay were 20

ng, 10 ng, 5 ng, 2.5 ng and 1.25 ng respectively. In Figure 6 the amplification curves of this

castPCR run are visualized. Ct values increase as the DNA concentration decreases. One well

of 20 ng of wild-type serum sample was also run with the mutation assay and as expected this

sample was not amplified (Figure 6).

Figure 6. Amplification curves of castPCR run testing the sensitivity of the reference genome

assay. Different amounts (20 ng, 10 ng, 5 ng, 2.5 ng and 1.25 ng) of cell-free DNA extracted from

healthy patient serum were added to each well. Each curve is marked according to the amount of

sample in that well. One well was with 20 ng of healthy cfDNA was run with the mutation assay,

marked here as 775mu. The unmarked curves below the threshold constitute the IPC from samples.

33

4.2.2 MDA-MB-453 is a positive control for the H1047R mutation

The next thing that was needed was a positive control. Two available cell lines, MDA-MB-453

(MDA) and SKOV-3 (SKOV), were known to harbor the H1047R mutation that was used in

the PIK3CA mutation assay (ATCC, 2014) and a PCR run was performed with 20 ng of DNA

extracted from these two cell lines. In Figure 7 the results from the castPCR run can be

visualized. Both MDA and SKOV were amplified by the mutation assay, MDA with a Ct value

of 27.97 and SKOV 28.64. The cell line MDA was chosen to be used as a positive control for

following PCR analyses.

Figure 7. Amplification curves of castPCR run using the PIK3CA mutation assay

with 20 ng of DNA extracted from the cell lines MDA-MB-453 (MDA) and SKOV-

3 (SKOV). In the figure each amplification curve is marked according to the cell line.

4.2.3 The PIK3CA mutation assay has high sensitivity when mutation frequency is 100 %

The sensitivity of the castPCR mutation analysis was examined by making a similar dilution

series as with the wild-type serum sample. DNA extracted from MDA was diluted to 10 ng/µl,

5 ng/µl, 2.5 ng/µl, 1.25 ng/µl and 0.625 ng/µl. After addition of 2 µl of each dilution per well,

34

the sample amounts in the assay were 20 ng, 10 ng, 5 ng, 2.5 ng and 1.25 ng respectively. Figure

8 shows the results of the amplification. The amplification curve of the 5 ng sample is not seen

and is thought to be due to a possible pipetting error.

Figure 8. Amplification curves of castPCR run testing the sensitivity of the

PIK3CA mutation assay. Different amounts (20 ng, 10 ng, 5 ng, 2.5 ng and 1.25

ng) of DNA extracted from the MDA-MB-453 (MDA) cell line were added to

each well. Each curve is marked according to the amount of sample in that well.

The curves below the threshold constitute the IPC from samples as well as the

NTC sample.

4.2.4 Dilution of the mutation frequency lowers sensitivity of the PIK3CA mutation assay

The H1047R mutation frequency in the MDA cell line is assumed to be 100 %. This does not

represent the situation in real life patient cases where tumor DNA is mixed with normal DNA

and the frequency can in fact be very low. Thus the mutation frequency of DNA extracted from

the MDA cell line was diluted using wild-type DNA. This was done twice, with the

35

manufacturers recommended 20 ng of sample and with our minimum of 1 ng of sample per

assay. According to the manufacturer the PIK3CA mutation assay should be able to detect a

mutation frequency as low as 0.1 % when 20 ng of sample DNA is used. The created frequencies

were 10 %, 5 %, 1 %, 0.5 %, 0.1 % and 0.01 % for the assays with both 20 ng and 1 ng. The

results of these PCR runs can be seen in Figure 9. In both figures (9A and 9B) the dilutions with

the frequencies 0.5 % - 10 % can be seen to have amplification curves above the threshold,

meaning that the PIK3CA mutation with these frequencies have been amplified in the PCR

reaction. However, the Ct values are quite high. The manufacturer recommends using 40 cycles

in the castPCR reaction, which we have changed to 50 due to low DNA concentrations. In

Figure 9A the Ct value for the sample with a mutation frequency of 0.5 % is 40, which is

equivalent to the amount of cycles recommended by the manufacturer. If the cycle amount

would not have been increased to 50, this amplification curve would not be seen at all. When

using the sample minimum of 1 ng, all samples have Ct values that are higher than 40, meaning

that if 40 cycles would have been used in the PCR the whole amplification plot would show

negative (Figure 9B). These amplifications can however be taken to be positive due to no

amplification in the negative control. In Figure 9A the frequencies 0.1 % and 0.01 % fall

beneath the negative control curve and so cannot be taken to be valid. These respective

frequencies from the assay using 1 ng have not been amplified at all (Figure 9B). The castPCR

PIK3CA mutation assay was not found to be sensitive enough to locate and amplify the H1047R

mutation with the frequency of 0.1 % or lower when either 20 ng or 1 ng of sample was used.

36

Figure 9. Amplification curves of castPCR run to test the sensitivity of the PIK3CA mutation assay when the

mutation frequency is low. DNA extracted from the MDA-MB-453 cell line was diluted into DNA extracted

from healthy serum to frequencies of 10 %, 5 %, 1 %, 0.5 %, 0.1 % and 0.01 %. In A) 20 ng of DNA and in B)

1 ng of DNA was used. Each curve is marked by its according frequency. The unmarked curves below the

threshold constitute the IPC from samples. Positive control = POS.CTRL, negative control = NEG.CTRL.

4.2.5 Serum cell-free DNA samples are mutation negative when using PIK3CA mutation-

specific castPCR

After sensitivity testing the actual study samples were run with the castPCR PIK3CA-mutation

assay.

Triple negative serum DNA samples are negative for the H1047R mutation

Triple negative breast cancer was one criteria for which certain patient samples were chosen.

The results from castPCR analysis of these samples can be visualized in Figure 10. The amount

of sample used per well was 1 ng. Only the positive control has been amplified when all samples

show no amplification and so a negative result. In Figure 10A the amplification plot shows

curves of samples with the positive and negative controls and Figure 10B the same plot

excluding the controls. The controls have been excluded in order for sample curves to be

37

visualized more clearly and to see that there is no amplification. Figure 10B clearly visualizes

no amplification in the study samples. Triple negative serum cfDNA samples from which 5 ng

of sample could be obtained for the assay were re-run to see whether increasing the amount of

sample would have any effect on the results. This meant that when adding the maximum of 4

µl of sample per well samples which had 1.25 ng/µl or more DNA could be re-run with 5 ng.

The amplification plot of the re-run shows no amplification of samples, only the positive control

has been amplified (Figure 11). The only other curve which has just been starting to rise at the

end of the PCR program with a Ct value of 42.08 is the negative control.

Figure 10. Amplification curves of castPCR run analyzing triple negative breast cancer samples with

PIK3CAmutation assay. 1 ng of sample was used in each analysis and 40 cycles was used. A) Amplification curves

including the positive and negative controls and B) zooming in on the red circle with controls excluded (NB.

fluorescence scale differs between A and B). Positive control = POS.CTRL

38

Figure 11. Amplification curves of castPCR run analyzing triple negative breast

cancer samples with PIK3CA mutation assay. 5 ng of sample was used per assay.

Positive control = POS.CTRL, Negative control = NEG.CTRL

T1N0 serum DNA samples are negative for the H1047R mutation

In Figure 12 the amplification plots from a castPCR run with 1 ng of study samples that had the

criteria of being T1N0 can be visualized. The plots indicate one possible positive result; sample

number 1857 has begun to amplify with a Ct value of 47.86. Figure 12A shows the result

including the positive and negative controls and Figure 12B the result excluding them. The

controls have been excluded from Figure 12B to make it easier to visualize the result. All other

samples except 1857 remain unamplified beneath the threshold along with the negative control

and NTC (Figure 12B). This possible positive result must be taken under consideration due to

the fact that it has such a high Ct value. The manufacturer recommends using 40 cycles in the

castPCR assay and 50 cycles have been used here. If only 40 cycles would have been used this

result would not show in the amplification plot. The sample was re-run at a later time.

39

Figure 12. Amplification curves of castPCR run analyzing T1N0 study samples with PIK3CA mutation assay.

1 ng of sample was used for each analysis. A) Amplification curves with positive and negative controls included

and B) zooming in on red circle in A with controls excluded. Positive control = POS.CTRL, 1857 = sample number.

Nine patients found mutation positive when assessing mutation status from primary tumor

DNA

Next the mutation status of the patients’ primary tumors were evaluated. Out of the 53 serum

cfDNA samples that we were able to run with castPCR, 22 had available primary tumor tissue

DNA samples. The primary tumor samples were run with castPCR PIK3CA mutation assay

with the manufacturers recommended 20 ng of sample per assay. The amplification curves from

this castPCR run can be visualized in Figure 13. In the figure the positive control MDA can be

seen to have the lowest Ct value, after which nine, possibly 10 samples are amplified before the

negative control. This gives us nine patients who harbor the H1047R mutation of the PIK3CA

gene. When examining the 9 patients who had PIK3CA positive primary tumors, 5 have either

died of causes related to breast cancer or gone through relapse. 7 out of the 9 have triple negative

breast cancer and 3 belong to the high cfDNA concentration group (75th percentile).

40

Figure 13. Amplification curves of castPCR run analyzing primary tumor tissue samples with

PIK3CA mutation assay. 20 ng of primary tumor DNA was used per sample. Samples are marked

according to their sample number. Positive control = POS.CTRL, negative control = NEG.CTRL.

Serum DNA samples are negative for the H1047R mutation when re-run with maximum

sample concentration of cell-free DNA in castPCR analysis

After the castPCR run with primary tumor DNA samples, those patient serum cfDNA samples

that had a positive result in the primary tumor sample were re-run with the PIK3CA mutation

assay. Eight samples out of the nine positive results had serum cfDNA concentrations over 0.25

ng/µl and could be run again with highest sample amounts possible. The samples that were re-

run were 467, 1618, 1801, 75, 910, 1685, 1868 and 1904. Also the possible positive, sample

1857 from a previous castPCR PIK3CA mutation assay run was re-run on the same plate with

maximum sample amount. Maximum sample amount means addition of 4 µl to each well of the

original concentration obtained from cfDNA extraction. Samples contained amounts from 1.42-

41

6.68 ng in this run. The original cfDNA concentrations can be viewed in Table 5. As the results

show in Figure 14, none of the samples run showed a positive amplification. Even the sample

1857 which previously showed a possible positive result (Figure 12) was clearly negative here.

Figure 14A shows the amplification curves including the controls whereas Figure 14B excludes

the controls and distinctly shows no amplification of study samples.

Figure 14. Amplification curves of castPCR run analysing serum cfDNA samples of patients, who had mutation

positive primary tumor DNA, with PIK3CA mutation assay. Also includes sample 1857 from a previous run. All

samples were run with their maximum sample concentration that could be utilized. A) Amplification curves

including positive and negative controls and B) zooming in on the red circle with controls excluded (NB.

fluorescence scale differs between A and B). POS.CTRL = positive control, NEG.CTRL = negative control.

4.2.6 Association between patients’ clinicopathological characteristics and cell-free DNA

concentrations obtained from extraction

Statistical analysis was performed using cfDNA concentrations to analyze distribution

significance and survival. Distributions used in statistical analysis can be viewed in Figure 5.

cfDNA concentrations were distributed into groups by several types of distributions. The data

was then analyzed by chi-square analysis under certain clinical variables such as histology

status, tumor stage, tumor grade, receptor status and relapse. In Table 6 the cfDNA

42

concentrations have been divided by median distribution, where the median divides the data set

into low or high expression (Figure 5A). With median distribution a significant difference can

be seen only in estrogen/progesterone receptor status. The results of this analysis show that it

is more likely to belong to the high cfDNA expression group when estrogen receptor (p = 0.003)

or progesterone receptor (p = 0.004) positive (Table 6). The triple negative status also supports

this result. Being triple negative makes it more likely to express lower concentrations of cfDNA,

and not having triple negative breast cancer makes it more likely to express higher cfDNA

concentrations (p = 0.023).

Table 6. The median distribution (figure 5A) of the patients (n = 81-82) in regards to clinical variables relevant

to this study. A chi-square test shows the statistical significance of these variables according to serum DNA

concentration. High expression is cfDNA concentration above the median and low expression below the

median. The significance value is p ≤ 0.05*. ns = not significant

Distribution Median distribution of DNA

Clinical variable Low expression High expression p-value

Histology ns.

Ductal 31 35

Lobular 2 1

Other 7 3

Stage ns.

in situ 1 1

I 29 27

II 9 9

III 2 4

Grade ns.

1 5 6

2 11 18

3 24 16

Estrogen receptor 0,033*

positive 12 21

negative 29 19

Progesterone receptor 0,004*

positive 8 20

negative 33 20

Triple negative 0,023*

yes 27 17

no 14 24

Relapse ns.

no 26 21

yes 15 20

43

In Table 7 the cfDNA concentration data is divided into three percentiles: the 25th percentile,

the 50th percentile and the 75th percentile (Figure 5B). When changing the distribution this way

the tumor grade becomes significant, showing a majority of patients having a less differentiated

tumor with cfDNA concentrations in the 50th and 75th percentiles. The progesterone receptor

status is also significant, again indicating a higher cfDNA concentration when the patient is

progesterone receptor positive (p = 0.012).

Table 7. Distribution of the patients (n = 81-82) within three percentile classes (figure 5B) in regards to clinical

variables relevant to this study A Chi-square test shows the statistical significance of these variables under this

distribution according to serum cfDNA concentration. The significance value is p ≤ 0.05*. ns = not significant

Distribution Distribution of DNA in three percentiles

Clinical variable 25th 50th 75th p-value

Histology ns.

Ductal 21 27 18

Lobular 1 2 0

Other 3 6 1

Stage ns.

in situ 1 1 0

I 16 26 14

II 7 7 4

III 2 2 2

Grade 0.048*

1 2 8 1

2 7 10 12

3 16 17 7

Estrogen receptor ns. (0.078)

positive 6 17 10

negative 20 19 9

Progesterone receptor 0.012*

positive 4 13 11

negative 22 23 8

Triple negative ns.

yes 18 19 7

no 8 17 13

Relapse ns.

no 18 21 8

yes 8 15 12

Table 8 shows the results from the chi-square analysis when cfDNA concentrations are divided

into four percentile groups where each group represents 25 % of the whole dataset. Each group

is marked according to the percentage values between which its data falls when values are in

ascending order (Figure 5C). With this distribution, only the progesterone receptor status is

44

significantly different between groups, showing that with higher cfDNA concentrations it is

more likely to be progesterone receptor positive (p = 0.02; Table 8).

Table 8. Distribution of the patients (n = 81-82) within four percentile classes (figure 5C) in regards to clinical

variables relevant to this study. A chi-square test shows the statistical significance of these variables according

to serum cfDNA concentration. The significance value is p ≤ 0.05*. ns = not significant

Distribution Distribution of DNA in four percentiles

Clinical variable 0-25 25-50 50-75 75-100 p-value

Histology ns.

Ductal 21 10 17 18

Lobular 1 1 1 0

Other 3 4 2 1

Stage ns.

in situ 1 0 1 0

I 16 13 13 14

II 7 2 5 4

III 2 0 2 2

Grade ns.

1 2 3 5 1

2 7 4 6 12

3 16 8 9 7

Estrogen receptor ns.

positive 6 6 11 10

negative 20 9 10 9

Progesterone receptor 0.02*

positive 4 4 9 11

negative 22 11 12 8

Triple negative ns.

yes 18 9 10 7

no 8 6 11 13

Relapse ns.

no 18 8 13 8

yes 8 7 8 12

The next distribution that is shown here is distribution of the patient cfDNA concentrations into

extremities, omitting the middle set of values. Here the values are distributed into the 25th

percentile and the 75th percentile and excluding all middle values (Figure 5D). This distribution

was done to try to analyze cfDNA concentrations which are clearly high and low without middle

values. This distribution shows that patients with high expression of cfDNA tend to have more

relapse than patients with low cfDNA concentrations (p = 0.047). Also the receptor status is

significant. Positive estrogen receptor status (p = 0.041) and progesterone receptor status (p =

0.003) and not having triple negative cancer (p = 0.021) indicates higher cfDNA concentrations.

45

Table 9. Distribution of the patients (n = 45-46) with taking into account only those patients in the extremities

(figure 5D) in regards to clinical variables relevant to this study. A chi-square test shows the statistical

importance of these variables according to serum DNA concentration. The significance value is p ≤ 0.05*. ns

= not significant

Distribution Distribution of DNA of patients in extremities

Clinical variable 25th 75th p-value

Histology ns.

Ductal 21 18

Lobular 1 0

Other 3 1

Stage ns.

in situ 1 0

I 16 14

II 7 4

III 2 2

Grade ns. (0.097)

1 2 1

2 7 12

3 16 7

Estrogen receptor 0.041*

positive 6 10

negative 20 9

Progesterone receptor 0.003*

positive 4 11

negative 22 8

Triple negative 0.022*

yes 18 7

no 8 13

Relapse 0.047*

no 18 8

yes 8 12

In the final distribution the patients have been divided into a high-rest distribution so that one

group is formed from the 75th percentile and the other group includes all the rest of the values

(Figure 5E). Here we see a significance in tumor grade (p = 0.033), progesterone receptor (p =

0.016) and triple negative status (p = 0.048). A greater tumor grade indicates higher cfDNA

concentrations as does a progesterone receptor positive status and not being triple negative.

46

Table 10. High-rest distribution (figure 5E) of the patients (n = 81-82) in regards to clinical variables relevant

to this study. A chi-square test shows the statistical significance of these variables according to serum DNA

concentration. The significance value is p ≤ 0.05*. ns = not significant

Distribution High - rest distribution of DNA

Clinical variable < 75th 75th p-value

Histology ns.

Ductal 48 18

Lobular 3 0

Other 9 1

Stage ns.

in situ 2 0

I 42 14

II 14 4

III 4 2

Grade 0.033*

1 10 1

2 17 12

3 33 7

Estrogen receptor ns.

positive 23 10

negative 39 9

Progesterone receptor 0.016*

positive 17 11

negative 45 8

Triple negative 0.048*

yes 37 7

no 25 13

Relapse ns.

no 26 6

yes 9 3

The Mann-Whitney two-sample rank-sum test was performed between patient samples divided

according to five different distributions used in Chi-Square test above and tested against clinical

variables such as relapse and receptor status. Table 11 shows the significant results obtained

from the analysis. All distributions except high-rest distribution show significance when taking

into account the clinical variables estrogen receptor, progesterone receptor and triple negative

status. Patients are less likely to be triple negative and more likely to express at least one

hormone receptor when cfDNA concentrations are higher. When patients are distributed to

extremities also relapse is significant (p = 0.05) showing a greater tendency to relapse when

having high cfDNA concentrations.

47

Table 11. The p-values of the Mann-Whitney U statistical test according to the serum DNA levels showing only

those clinical variables that showed a significant result. The significance level p ≤ 0.05*. ns = not significant.

Confidence interval 0.95.

p-value

Distribution Median Three percentiles Four percentiles Extremities

Clinical variable low high 25th 50th 75th 25th 25-50 50-75 75th 25th 75th

Estrogen

receptor 0.034* 0.035* 0.026* 0.043*

positive 12 21 6 17 10 6 6 11 10 6 10

negative 29 19 20 19 9 20 9 10 9 20 9

Progesterone

receptor 0.004* 0.003* 0.002* 0.003*

positive 8 20 4 13 11 4 4 9 11 4 11

negative 33 20 22 23 8 22 11 12 8 22 8

Triple negative

status 0.016* 0.02* 0.012* 0.022*

yes 27 17 18 19 7 18 9 10 7 18 7

no 14 24 8 17 13 8 6 11 13 8 13

Relapse ns. ns. ns. 0.05*

no 26 21 18 21 8 18 8 13 8 18 8

yes 15 20 8 15 12 8 7 8 12 8 12

Patients with high cell-free DNA concentrations have a decline in survival rate and are more

likely to relapse than patients with low cell-free DNA concentrations

Survival was evaluated using the Kaplan-Meier analysis. Survival rates were analyzed in all

distribution groups of which two showed significance: when data was distributed to extremities

and when data was distributed into a high-rest distribution (distributions Figure 5D-E) Analyses

according to three distributions are shown in Figure 15, including the two mentioned above as

well as distribution into three percentile groups (Figure 5B). When distributing into extremities

the results show a significant decline of survival rate in patients with high serum DNA

concentrations compared to patients with low serum DNA concentration when taking either

relapse as an end point (p = 0.033; figure 15B) or death by breast cancer as an end-point (p =

0.05; Figure 15A). The same is seen with high-rest distribution with either relapse (p = 0.023,

Figure 15D) or death by breast cancer as an end-point (p = 0.024; Figure 15E). A similar trend

is evident with cfDNA concentration divided into three percentiles and having relapse as an

end-point, but the result is not significant (Figure 15 C). Patients with higher concentrations of

cfDNA have clear decline of overall survival to breast cancer and are more likely to suffer

relapse.

48

Figure 15. Kaplan-Meier survival analysis according to three different distributions of

cfDNA concentrations with A) follow-up survival in extremities (p = 0.05), B) relapse free

survival in extremities (p = 0.033), C) relapse-free survival in three percentile groups (ns),

D) relapse-free survival in high-rest distribution (p = 0.023) and E) follow-up survival in

high-rest distribution (p = 0.024) Significance level p ≤ 0.05.

49

Patients with high cell-free DNA concentrations have a decline in survival rate compared to

those with low cell-free DNA concentrations in multivariate survival analysis

Logistic regression analysis was done to compare survival of patients under criteria of the

covariates age, nodal status and estrogen/progesterone receptor status with death by breast

cancer used as an end-point. Regression analysis was done under three distributions, with

distribution into extremities and high-rest distribution. Survival was also evaluated with

distribution into TN and T1N0 patients. The results of the cox regression analysis can be

visualized in Figure 16. In Figure 16A the cfDNA data has been distributed into extremities,

with DNA low indicating the 25th percentile and DNA high the 75th percentile. The analysis

showed a decline in survival rate of those patients with higher concentrations of cfDNA, but

the result was not significant. In Figure 16B the analysis has been performed under high-rest

distribution. When changing the distribution this way the survival analysis becomes significant

(p = 0.041) showing a clear decline of survival of patients with high concentrations of cfDNA.

There was no significance found in survival between TN and T1N0 groups (Figure 16C).

50

Figure 16. Cox regression analysis from cfDNA

concentration including the covariates age, nodal

status, PR status and ER status where death by breast

cancer is used as an end-point. A) According to

distribution to extremities, ns. = not significant, B)

according to high-rest distribution (p = 0.041) and C)

according to distribution into TN and T1N0 groups

(ns.). Significance level p ≤ 0.05.

5. DISCUSSION

This study demonstrates the importance of exploring circulating cell-free nucleic acids and their

role in cancer. The results indicate patients with high concentrations of cfDNA to be more prone

to relapse. Also a multivariate survival analysis shows a trend of higher mortality in patients

with higher cfDNA concentrations. By examining the cfDNA status of cancer patients it could

be possible to specify which patients are more susceptible to recurrence. The examination of

51

cfDNA needs to go beyond concentration and focus on specific characteristics of possible tumor

DNA in the blood. We attempted to find ctDNA by a castPCR assay which works by amplifying

single point mutations from DNA extracted from blood serum or plasma. The method is very

sensitive in finding mutations, the problem is the ability of studying only one mutation per assay

which may not be practical in a clinical setting. Here we were not able to find the H1047R

mutation from cfDNA samples of 82 breast cancer patients.

5.1 Low tumor burden and prolonged storage of serum may account for lower

cfDNA concentration

The patient material available for use contained samples from nearly all breast cancer patients

in Kuopio university hospital during the years 1990-1995 and have been very thoroughly

followed up on. This gave us the ability to basically take our pick. The patient samples used

were all over 15 years old with some as old as 25 years, which was an advantage as well as a

disadvantage. The advantage was that the outcome of these patient cases were known and this

knowledge could be used when choosing patient samples for the study as well as giving an

initial reason for this study. Samples that were of interest were patients who had a good

prognosis at the time of their diagnosis, but the disease progressed in an opposite way from that

which was predicted. The rest of the samples were chosen for their sole characteristic of being

triple negative samples, which are interesting due to the limited treatment possibilities with this

particular type of cancer, as they have no receptors that can be targeted and are thus usually

more lethal.

The most prominent disadvantage was the storage issue. DNA integrity is greatly decreased

with prolonged storage time (Xue et al., 2009; Sozzi et al., 2005). The samples were stored in -

80 °C. One study showed that already storage over 3 years degrades DNA in plasma (Sozzi et

al., 2005). This may be one reason for the low DNA concentration obtained from extraction

from the serum samples. Also due to the age of the samples it is not necessarily known whether

they have been thawed and used previously. Thawing and refreezing has been shown to also

degrade DNA (Xue et al., 2009).

Previous studies have stated that detectable amounts of cfDNA can be reliably found only from

patients with progressive disease and here our patient samples were from the time of diagnosis

when disease burden is likely to be much smaller (Dawson et al., 2013; Schwarzenbach et al.,

2011; Board et al., 2010; Anker et al., 1999). Schwarzenbach and colleagues (2011) did find

that they were able to distinguish between healthy patients and patients with breast cancer based

52

on cfDNA concentration, but could not show what stage the disease was in. However, Hashad

and colleagues (2012) found a significant difference in cfDNA levels between patients with

benign disease and those with malignant breast lesions (p < 0.001). Other researchers were able

to detect tumor derived cfDNA also in patients with early-stage breast cancer, not only late

malignant disease (Beaver et al., 2014; Bettegowda et al., 2014; Shaw and Stebbing, 2014).

Many researchers argue that serum is less suitable than plasma as a starting material for DNA

extraction due to contamination of wild-type DNA (Schwarzenbach, 2013; Benesova et al.,

2013) and the majority of studies have used plasma. Board and colleagues (2010) observed a

higher mutation detection rate of PIK3CA mutations in plasma than in serum and hypothesized

this to be due to lysis of white blood cells resulting in a higher background of healthy DNA.

Cell-free DNA is found in varying sizes, especially when tumor derived (Schwarzenbach et al.,

2011; Fleischhacker et al., 2011; Jahr et al., 2001), Qiagen columns used for elution of DNA

might lose the very smallest fragments (Xue et al. 2009).

In this study, detectable amounts of cfDNA were obtained from 56 out of 82 patients, with

concentrations ranging from 0.23 ng/µl to 2.12 ng/µl. Qubit was not able to detect DNA from

26 patient serum samples, which most likely indicates very low concentrations instead of no

concentration.

5.2 Detecting tumor-related genetic changes in cell-free DNA

Liquid biopsies and their possible clinical applications are a constantly rising topic with the

possibilities of cfDNA and its use in cancer diagnostics at its core. There are multiple techniques

to study the genetic make-up of cfDNA and these methods are continually developed, with

many researchers comparing several methods within studies. The most common approaches are

PCR techniques manufactured to find SNPs and mutations that would identify cfDNA

originated from a tumor. Digital PCR is one of the most popular techniques researchers today

use to analyze the genetics of cfDNA mainly for its high sensitivity. Digital PCR is more

sensitive and precise than quantitative real-time PCR, but also much more expensive (Reid et

al., 2015; Beaver et al., 2014). Reid and coworkers (2014) found droplet digital PCR to be more

robust and up to 200 times more sensitive than castPCR which manufacturers give a sensitivity

of 0.1 %. In this study we used castPCR to establish which patients were mutation positive by

amplifying a target point mutation and blocking unspecific amplification. castPCR is a

relatively less used method with only 11 publications using the technique found in Pubmed

(http://www.ncbi.nlm.nih.gov/pubmed, accessed: 11.04.2016). However qPCR has much more

range and can analyze hundreds of sequences per run. Other PCR techniques used are

53

BEAMing (beads, emulsion, amplification and magnetics) and Amplication-Refractory

Mutation System (ARMS-PCR). A problem with PCR methods may be low mutation frequency

with a high background of normal DNA that could hinder detection and amplification of

specific mutations.

Next-generation sequencing (NGS) tools have been developed to find every SNP in a gene

(Dempsey, 2013). Whole exome sequencing gives every exon of a gene, with all SNPs that it

harbors, and though the method is time consuming and requires some form of manual labor or

a written program to identify wanted mutations, it is quite efficient. Developments are being

made to continuously enhance the method. (Dawson et al., 2013; Banerji et al., 2012)

Sequencing methods are also yet not as sensitive as PCR methods and require larger amounts

of starting material (Diaz and Bardelli, 2014). However, strides have already been taken to

overcome these issues and it is more likely that NGS methods will be more valuable in the

clinical setting than methods for identifying single mutations. The challenge for both may be

determining which findings are important and what their significance is. Most of the studies

examined have used multiple methods together to achieve the most accurate result, most using

some form of sequencing with a PCR technique. After initial findings using sequencing, a

mutation considered significant may then be studied using PCR techniques.

The aim for researchers is to find a practical way to use cfDNA in a clinical setting to diagnose

and monitor cancer and advancing its usage into widespread practice (Bettegowda et al., 2014;

Sausen et al., 2014).

5.2.1 Competitive Allele-specific TaqMan PCR is sensitive and specific

The castPCR method was tested for its sensitivity in many different ways, starting with the

sensitivity of the reference genome assay. The results of a standard series dilution showed that

with smaller DNA concentrations there is also less amplification (Figure 6). Also the healthy

serum sample was not amplified when using the mutation assay, as it should not have. We were

able to use this healthy serum sample as a negative control.

Next the sensitivity of the PIK3CA mutation assay was tested with the positive control for

which the assumption was held that its DNA carries a mutation frequency of 100 %. The

mutation assay was able to amplify the positive control with as low as 1.5 ng of sample (Figure

8).

The positive control was then diluted into wild-type DNA, thus diluting the mutation frequency.

The manufacturer gives a guarantee that the assay can detect a mutation frequency as low as

54

0.1 %. When 20 ng of sample was used, the curves rose nicely up to 0.5 % after which the

negative control could be seen and below this the amplification curves of samples with mutation

frequencies of 0.1 % and 0.01 % (Figure 9A). The fact that the negative control has been

amplified indicates possible inaccuracies in all samples in this run and for further use this whole

run should be redone. The current results show that the assay was not able to amplify the

PIK3CA mutation with a frequency of 0.1 % or lower. The same result was seen when 1 ng of

sample was used (Figure 9B). This shows that the castPCR PIK3CA mutation assay can detect

this particular mutation to a mutation frequency as low as 0.5 %. This further implies that in

order for results from this study to be obtained, the DNA extracted from the patient serum

samples must first contain the mutation in question and at the frequency of no less than 0.5 %.

Sample amount of 1 ng, however, should not be an issue. Probably the most crucial steps to be

considered are the preparation of the plate and reactions, where when having such small

amounts of starting material it is essential to perform these steps accurately.

5.2.2 Quality control of castPCR using specified control samples

The positive control is used as a measure of a positive result and also to determine that the PCR

reaction has worked. The negative control is used to ensure that no contamination has happened

and to recognize false positives. The negative control is also used as a boundary control,

meaning that in the event that the negative control is amplified all samples amplified after it are

to be considered as negative and should be run again. For example when testing the sensitivity

of the method, the samples with the two lowest frequencies of 0.1 % and 0. 01 % had to be

negative as the negative control has been amplified before these with a lower Ct value (Figure

11A). The negative control may be amplified for several different reasons. Most likely it is due

to unspecific amplification, which is why it is usually amplified at the very end of the run, when

the cycles start to rise and so the negative control has a very high Ct value. The fault may also

be mechanical. The negative control is very important to be able to register false positives.

Whenever there is amplification in the negative control the whole run should be redone. Also

an internal positive control (IPC) was used, which has the main role of identifying target

negatives and distinguishing them from PCR failure. The IPC in each run was positive,

indicating that negative results were not the result of PCR failure.

55

5.2.3 Patient serum DNA samples with triple negative or T1N0 breast cancer analyzed

with Competitive Allele-Specific TaqMan PCR PIK3CA mutation assay

The study samples were chosen on certain criteria which made them interesting to this study.

The first set of samples were chosen with good prognostic markers, T1N0 ductal carcinoma,

from patients who either had relapse or eventually died of the disease. The second set of samples

was chosen with the same criteria, but from patients who overcame the disease. The third set of

samples were chosen with the sole criteria of being triple negative, as it is the most aggressive

type of breast cancer with no hormonal treatment available (Griffiths and Olin, 2012).

Unfortunately due to poor results when extracting DNA from serum samples, 35 % of the

samples could not be run with castPCR mutation assay.

Study samples were run on castPCR with PIK3CA mutation assay using 1 ng of sample. No

amplification of triple negative samples could be seen when using 1 ng of sample (Figure 10).

There were only 5 triple negative samples with high enough concentration to be able to be run

again with 5 ng of sample. These samples were not amplified either (Figure 11). The results

from the triple negative castPCR runs indicate that there is no observable mutation with high

enough frequency to be amplified (Figure 10, 11).

Next the T1N0 samples were run in the same way as the triple negative samples using 1 ng of

sample in the assay. One sample (sample number 1857) had a Ct value, meaning a possible

positive result (Figure 12). None of the other samples were amplified. This one sample was re-

run later using a maximum amount of sample, but showed a negative result (Figure 14). As with

the triple negative samples, the results from the T1N0 castPCR runs show no observable

mutation in these samples.

5.2.4 castPCR run of available primary tumor DNA samples and re-run of serum samples

with mutation positive primary tumor DNA

The next idea was that possibly the patient could have the H1047R mutation, but the tumor has

not yet spread its cells and DNA into the bloodstream. We obtained primary tumor DNA from

22 of our 82 patients and ran them with the castPCR PIK3CA mutation assay with the

recommended 20 ng of sample. This analysis showed a positive result in 9 samples (Figure 14).

More of the samples were somewhat amplified, but the curve of the negative control can be

seen to be above these and so these must be taken as false positives.

Eight of the 9 samples that showed positive results in the primary tumor DNA analysis were re-

run with castPCR PIK3CA mutation assay from their serum DNA samples with the maximum

56

sample amount that could be obtained per assay. All samples showed negative results (Figure

14).

The results show that even though the primary tumor DNA sample was found to be mutation

positive the corresponding serum DNA sample was negative. This conflict in the result may

indicate that the mutation is present, but the tumor has not yet spread its DNA into the

bloodstream or that tumor DNA is present but at very small amounts. Tumor DNA could be

ample in the bloodstream, but due to low tumor DNA to normal DNA ratio the mutation is too

diluted to be observed. The sensitivity test done previously shows that even a mutation with a

frequency of 10 % is amplified very little when using 1 ng of sample (Figure 9B). All analyses

indicate a need for higher serum DNA concentrations to be used in castPCR, though according

to the sensitivity test done previously it should not be a problem. The need for higher serum

concentrations comes from the need of higher mutation frequency, which may be the real

problem here.

5.2.5. No detection of H1047R mutation with high enough frequency from cfDNA using

PIK3CA mutation specific castPCR

The blood serum samples that were taken from the patients were taken at the time of diagnosis,

which can indicate low tumor burden. Tumors are however individual for each case and the

situation at time of diagnosis could vary greatly between individuals. Breast cancer is a type of

cancer that can be detected early e.g. by mammogram but in many cases it is detected later on

when the tumor has grown so that it is palpable (www.breastcancer.org). The initial cfDNA

concentrations obtained from extraction from the serum samples indicate that the amount of

tumor DNA in the serum has been minimal (Table 5).

The small concentration of cfDNA obtained from the serum samples was the first pitfall in this

study. This does not however mean that this was the reason for negative results. The patients

with negative results may not have the H1047R mutation at all. However 9 patients were

positive for the PIK3CA H1047R mutation in their primary tumor DNA. Perhaps the tumor

DNA in the serum has a different tumor profile than the primary tumor and does not contain

the H1047R mutation. Sporadic breast cancer comes in so many different forms meaning

multiple different somatic mutations accounting to cancerous growth in patients. The current

approximated frequency for the PIK3CA H1047R mutation in patients with breast cancer is 26

%, which means that approximately 74 % of sporadic breast cancer patients do not carry this

particular mutation (Catalogue of somatic mutations in cancer, Wellcome Trust Sanger

57

Institute, Cambridge, UK). This might not have been such a large problem if there would have

been more patient samples used in this study.

Due to there being no positive results from the serum DNA PIK3CA mutation PCR analysis

the ratio of tumor to healthy DNA in the blood could not be determined.

The reason for no positive results may be a too small of a sample group. The criteria for the

patient samples chosen to be in this study limited the amount of patient samples. Also as the

samples were so old, some which could have been used have been lost in storage. 36 % of the

serum samples could not be run with castPCR as no detectable amounts of cfDNA were

observed after extraction. With a larger pool of patient samples the probability of positive

results rises. Bettegowda and colleagues (2014) found a similar fraction of patients without

detectable amounts of cfDNA (45 %) in patients with localized cancers, but they had 223

patients all together as the present study had only 82.

Based on the results from the primary tumor DNA analysis nine patients (11 %) did in fact carry

the H1047R mutation, but it could not be determined from serum DNA. There could be more

patients with the mutation in question, which could possibly be seen from primary tumor DNA,

but not all of the samples had primary tumor DNA samples available that could have been

analyzed. Also the fact that only one hot-spot mutation was analyzed limits this study and in

the future multiple mutations should be considered.

Some of the results could also be false negatives. The amount of DNA could be too small for

the PCR program to detect. On the other hand the proportion of mutation may be so minimal

that the PCR program does not detect this even though it would be present. Board and

colleagues (2010) detected PIK3CA mutations from plasma cfDNA samples from 8 patients

with known PIK3CA mutations in their primary tumor who had advanced cancer. They did not

however detect any PIK3CA mutations from 14 patient cfDNA samples also with known

PIK3CA mutations in their primary tumor with localized cancer. It must be concluded that

either there is no mutation yet present in the blood or it is not detectable in patients with early-

stage cancer. This is why the primary tumor DNA samples were studied. If the patient harbored

the H1047R mutation, it would have to show in the primary tumor sample as a positive PCR

result.

5.3 cfDNA concentration may be a viable prognostic marker in breast cancer

Even though results were not obtained from PCR analysis, we were able to move further with

cfDNA obtained from extraction and examine associations between cfDNA concentration and

58

clinical variables. Out of the 82 patients 53 had detectable amounts of cfDNA in the serum, but

all 82 samples were taken into account when doing statistical analysis.

Many significant associations can be observed especially when patient samples have been

distributed to extremities and when patients with high cfDNA concentrations have been taken

as a separate group. Patients with high serum DNA concentration have had more relapse than

patients with low serum DNA concentration (p = 0.047, Table 9). This indicates that patients

with higher concentrations of DNA in the serum have higher possibility of relapse and so a

poorer prognosis.

In both survival analyses the results clearly indicated a higher mortality rate in patients with

high concentrations of cfDNA (Figure 15, 16). Patients with higher concentrations of cfDNA

are more likely to relapse and more often succumb to breast cancer than patients with lower

concentrations. These results are supported by multiple previous studies (Takeshita et al., 2015;

Newman et al., 2014; Dawson et al., 2013). Out of the 20 patients that belong to the high -group

of the high-rest distribution, 9 have died of causes related to breast cancer and 3 have gone

through relapse. Six of the patients who died of causes related to breast cancer and all 3 patients

who have gone through relapse belong to the T1N0 group. The results demonstrate a clear

association between the severity of breast cancer and cell-free DNA concentration. Cell-free

DNA has been a prominent research target mainly for its relationship with cancer and these

results show that its use as a biomarker is justified. The challenge now is going beyond cfDNA.

5.4 Effects of disease progression and treatment must be considered when

examining cfDNA as a prognostic marker

A great advantage of this study is that the samples used were old, so the outcome of disease

was known. This study could be repeated with fresh samples from patients with breast cancer,

only then the outcome would not be known and the criteria for samples would have to be

changed. This would give many more samples to be studied. It would also be of interest to see

what the difference in extracted DNA concentration would be when using stored and fresh

samples. These patients’ diseases would have to be monitored and their disease progression

followed up on.

As noted in the experimental section, the patient serum samples were taken at the time of

diagnosis. Serum samples from these same patients taken at later time points are also in storage

and would be available for use. It would be interesting to see whether the DNA amount in the

blood has increased at all over time. Studies have shown a correlation between blood DNA

59

levels and tumor evolution (Anker et al., 1999) with increasing amounts of cfDNA

concentrations as tumor burden increases. Also from those patients whose primary tumor

samples gave positive results, it would be interesting to see if from a later time point the

mutation could also be identified from the serum sample. However studies also showed that

cfDNA concentrations decrease after cancer treatment (Elshimali et al., 2013; Leon et al., 1977)

and as patients have most likely gone through some form of treatment these later samples may

have lower cfDNA concentrations. Previous studies show that the DNA levels in blood decrease

after radiotherapy treatment by up to 90 % (Anker et al., 1999). Also patients may undergo

different types of treatment, such as hormone therapy, chemotherapy and/or surgery, all of

which could have different and unknown effects. For example excision of the primary tumor

has been shown to drop cfDNA levels to as low as that of a healthy individual (Catarino et al.,

2008).

It could also be possible to try to concentrate the serum DNA samples that are now available to

see whether a greater concentration could be obtained and then used in the castPCR PIK3CA

mutation assay for a higher sample amount. The inability to identify cfDNA in some early-stage

patients is one of the obstacles of its usage in a clinical setting (Bettegowda et al., 2014).

5.4.1 Sequencing study shows correlation between common samples

Another study done simultaneously used 22 samples that were also used in this study. In that

study tumor DNA was sequenced from DNA extracted from formalin-fixed paraffin-embedded

(FFPE) tissue samples. Out of the 22 samples only three were shown to harbor the H1047R

mutation. These three samples were also shown to be mutation positive in this study when run

using primary tumor DNA with the PIK3CA mutation assay. One sample which showed a

positive result from the primary tumor DNA run in this study did not show the H1047R

mutation when sequenced. Unfortunately more samples were not in common between these two

studies, for example it would have been very interesting to see the sequencing of sample number

1857. The objective was to first have all the T1N0 samples in common between these two

studies, but samples were dropped out of this study for various reason, for example some

samples were lost in storage. Samples were also dropped from the other study, mainly due to

the paraffin-embedded tumor not being a good representation of the tumor and so did not suffice

for extraction and sequencing. Also only tumor tissue was sequenced, giving mutation status of

the tumor.

Sequencing is however a very robust method in finding mutations from sample DNA and might

be one route to follow when further analyzing cfDNA.

60

5.4.2 Limited mutation detection range of castPCR may hinder its clinical application

The application of using castPCR which identifies one specific mutation is one thing that needs

to be considered. The method used in this study is very specific. It is able to analyze the presence

of one specific mutation from a sample in one analysis. The mutation used here is one that is

found frequently in patients with sporadic breast cancer. Breast cancer types vary widely and

each case of breast cancer is unique. The use of only one specific mutation may not be optimal

when samples are analyzed. To be able to expand the use of this method and to gain maximum

benefit, multiple mutations should be studied from each sample as it is not known which

mutations the patients harbor. Even though the H1047R mutation is the most frequent in cases

of sporadic breast cancer, it is harbored in only 9 patients in this study that we know. This means

that nearly 17 % of patients (9/53) who had their serum DNA sample run with castPCR mutation

assay were mutation positive, and when taking into account all 82 patients 11 % were mutation

positive. It would be possibly wise to take a few more mutations, others also with high

frequencies (e.g. mutations of TP53) and test them alongside the H1047R mutation. Which

mutation is found would be indifferent to the goals of this study, where identification of tumor

DNA was primary.

Evaluating total circulating DNA concentrations from patients may have clinical value.

Extracting DNA or RNA is quite a simple procedure which is not very costly. If more

knowledge could be obtained from the relationship of cancer and cfDNA, quantifying it from a

blood sample could become an easy first step for diagnosis and prognosis. However this

requires more work for there are many unanswered questions to what the connection ultimately

is and whether it is the same for all patients.

6. Conclusions

Increasing levels of circulating cell-free DNA account to a higher tendency to relapse and

higher mortality rate in patients with breast cancer. Patients with higher concentrations of

cfDNA have poorer outcomes and therefore worse initial prognosis. Outcome could be

evaluated by studying cfDNA concentration and identifying the ratio of tumor to healthy DNA.

cfDNA concentration could be used as a prognostic marker among other clinicopathological

characteristics. Identifying tumor characteristics from cfDNA is possible, but the method must

be considered for clinical use. Studying single point mutations might not be efficient in the

clinical setting. More research is required to fully be able to consider cell-free DNA as a tumor

biomarker in diagnostics, prognostics and treatment of breast cancer.

61

7. References

Internet references

www.breastcancer.org, accessed on 05.09.2015

www.cancerresearchuk.org, accessed on 05.09.2015

Catalogue of somatic mutations in cancer, http://cancer.sanger.ac.uk/cosmic, Wellcome Trust

Sanger Institute, Cambridge, UK. Accessed on 20.02.2016

The Finnish Cancer Registry, www.syoparekisteri.fi, updated 08.10.2015, accessed on

16.02.2016

National Cancer Institute, http://www.cancer.gov/publications/dictionaries/cancer-terms,

accessed on 29.02.2016

http://www.ncbi.nlm.nih.gov/pubmed, “Competitive allele-specific TaqMan PCR”, accessed:

16.10.2015

Literature references

Abdulkareem, I.H. 2013. Aetio-pathogenesis of breast cancer. Niger.Med.J. 54:371-375.

Alix-Panabieres, C., H. Schwarzenbach, and K. Pantel. 2012. Circulating tumor cells and

circulating tumor DNA. Annu.Rev.Med. 63:199-215.

Anker, P., H. Mulcahy, X.Q. Chen, and M. Stroun. 1999. Detection of circulating tumour DNA

in the blood (plasma/serum) of cancer patients. Cancer Metastasis Rev. 18:65-73.

Bader, A.G., S. Kang, L. Zhao, and P.K. Vogt. 2005. Oncogenic PI3K deregulates transcription

and translation. Nat.Rev.Cancer. 5:921-929.

Banerji, S., K. Cibulskis, C. Rangel-Escareno, K.K. Brown, S.L. Carter, A.M. Frederick, M.S.

Lawrence, A.Y. Sivachenko, C. Sougnez, L. Zou, M.L. Cortes, J.C. Fernandez-Lopez, S. Peng,

K.G. Ardlie, D. Auclair, V. Bautista-Pina, F. Duke, J. Francis, J. Jung, A. Maffuz-Aziz, R.C.

Onofrio, M. Parkin, N.H. Pho, V. Quintanar-Jurado, A.H. Ramos, R. Rebollar-Vega, S.

Rodriguez-Cuevas, S.L. Romero-Cordoba, S.E. Schumacher, N. Stransky, K.M. Thompson, L.

Uribe-Figueroa, J. Baselga, R. Beroukhim, K. Polyak, D.C. Sgroi, A.L. Richardson, G.

Jimenez-Sanchez, E.S. Lander, S.B. Gabriel, L.A. Garraway, T.R. Golub, J. Melendez-Zajgla,

A. Toker, G. Getz, A. Hidalgo-Miranda, and M. Meyerson. 2012. Sequence analysis of

mutations and translocations across breast cancer subtypes. Nature. 486:405-409.

Beaver, J.A., D. Jelovac, S. Balukrishna, R.L. Cochran, S. Croessmann, D.J. Zabransky, H.Y.

Wong, P. Valda Toro, J. Cidado, B.G. Blair, D. Chu, T. Burns, M.J. Higgins, V. Stearns, L.

Jacobs, M. Habibi, J. Lange, P.J. Hurley, J. Lauring, D.A. VanDenBerg, J. Kessler, S. Jeter,

M.L. Samuels, D. Maar, L. Cope, A. Cimino-Mathews, P. Argani, A.C. Wolff, and B.H. Park.

2014. Detection of cancer DNA in plasma of patients with early-stage breast cancer.

Clin.Cancer Res. 20:2643-2650.

62

Benesova, L., B. Belsanova, S. Suchanek, M. Kopeckova, P. Minarikova, L. Lipska, M. Levy,

V. Visokai, M. Zavoral, and M. Minarik. 2013. Mutation-based detection and monitoring of

cell-free tumor DNA in peripheral blood of cancer patients. Anal.Biochem. 433:227-234.

Bettegowda, C., M. Sausen, R.J. Leary, I. Kinde, Y. Wang, N. Agrawal, B.R. Bartlett, H. Wang,

B. Luber, R.M. Alani, E.S. Antonarakis, N.S. Azad, A. Bardelli, H. Brem, J.L. Cameron, C.C.

Lee, L.A. Fecher, G.L. Gallia, P. Gibbs, D. Le, R.L. Giuntoli, M. Goggins, M.D. Hogarty, M.

Holdhoff, S.M. Hong, Y. Jiao, H.H. Juhl, J.J. Kim, G. Siravegna, D.A. Laheru, C. Lauricella,

M. Lim, E.J. Lipson, S.K. Marie, G.J. Netto, K.S. Oliner, A. Olivi, L. Olsson, G.J. Riggins, A.

Sartore-Bianchi, K. Schmidt, l. Shih, S.M. Oba-Shinjo, S. Siena, D. Theodorescu, J. Tie, T.T.

Harkins, S. Veronese, T.L. Wang, J.D. Weingart, C.L. Wolfgang, L.D. Wood, D. Xing, R.H.

Hruban, J. Wu, P.J. Allen, C.M. Schmidt, M.A. Choti, V.E. Velculescu, K.W. Kinzler, B.

Vogelstein, N. Papadopoulos, and L.A. Diaz Jr. 2014. Detection of circulating tumor DNA in

early- and late-stage human malignancies. Sci.Transl.Med. 6:224ra24.

Board, R.E., A.M. Wardley, J.M. Dixon, A.C. Armstrong, S. Howell, L. Renshaw, E. Donald,

A. Greystoke, M. Ranson, A. Hughes, and C. Dive. 2010. Detection of PIK3CA mutations in

circulating free DNA in patients with breast cancer. Breast Cancer Res.Treat. 120:461-467.

Campeau, P.M., W.D. Foulkes, and M.D. Tischkowitz. 2008. Hereditary breast cancer: new

genetic developments, new therapeutic avenues. Hum.Genet. 124:31-42.

Catarino, R., M.M. Ferreira, H. Rodrigues, A. Coelho, A. Nogal, A. Sousa, and R. Medeiros.

2008. Quantification of free circulating tumor DNA as a diagnostic marker for breast cancer.

DNA Cell Biol. 27:415-421.

Crowley, E., F. Di Nicolantonio, F. Loupakis, and A. Bardelli. 2013. Liquid biopsy: monitoring

cancer-genetics in the blood. Nat.Rev.Clin.Oncol. 10:472-484.

Dawson, S.J., D.W. Tsui, M. Murtaza, H. Biggs, O.M. Rueda, S.F. Chin, M.J. Dunning, D.

Gale, T. Forshew, B. Mahler-Araujo, S. Rajan, S. Humphray, J. Becq, D. Halsall, M. Wallis,

D. Bentley, C. Caldas, and N. Rosenfeld. 2013. Analysis of circulating tumor DNA to monitor

metastatic breast cancer. N.Engl.J.Med. 368:1199-1209.

Dempsey, P.W. 2013. All in a blood draw: breast cancer monitoring in the age of genetic

medicine. MLO Med.Lab.Obs. 45:14-15.

Diaz, L.A.,Jr, and A. Bardelli. 2014. Liquid biopsies: genotyping circulating tumor DNA.

J.Clin.Oncol. 32:579-586.

Elshimali, Y.I., H. Khaddour, M. Sarkissyan, Y. Wu, and J.V. Vadgama. 2013. The clinical

utilization of circulating cell free DNA (CCFDNA) in blood of cancer patients. Int.J.Mol.Sci.

14:18925-18958.

Ferlay, J., I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers, M. Rebelo, D.M. Parkin, D.

Forman, and F. Bray. 2015. Cancer incidence and mortality worldwide: sources, methods and

major patterns in GLOBOCAN 2012. Int.J.Cancer. 136:E359-86.

Fleischhacker, M., and B. Schmidt. 2007. Circulating nucleic acids (CNAs) and cancer--a

survey. Biochim.Biophys.Acta. 1775:181-232.

63

Fleischhacker, M., B. Schmidt, S. Weickmann, D.M. Fersching, G.S. Leszinski, B. Siegele,

O.J. Stotzer, D. Nagel, and S. Holdenrieder. 2011. Methods for isolation of cell-free plasma

DNA strongly affect DNA yield. Clin.Chim.Acta. 412:2085-2088.

Girish, C., P. Vijayalakshmi, R. Mentham, C. Babu Rao, and S. Nama. 2014. A REVIEW ON

BREAST CANCER. International Journal of Pharmacy and Biological Sciences. 4:47-52.

Griffiths, C.L., and J.L. Olin. 2012. Triple negative breast cancer: a brief review of its

characteristics and treatment options. J.Pharm.Pract. 25:319-323.

Higgins, M.J., D. Jelovac, E. Barnathan, B. Blair, S. Slater, P. Powers, J. Zorzi, S.C. Jeter, G.R.

Oliver, J. Fetting, L. Emens, C. Riley, V. Stearns, F. Diehl, P. Angenendt, P. Huang, L. Cope,

P. Argani, K.M. Murphy, K.E. Bachman, J. Greshock, A.C. Wolff, and B.H. Park. 2012.

Detection of tumor PIK3CA status in metastatic breast cancer using peripheral blood.

Clin.Cancer Res. 18:3462-3469.

Jahr, S., H. Hentze, S. Englisch, D. Hardt, F.O. Fackelmayer, R.D. Hesch, and R. Knippers.

2001. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for

their origin from apoptotic and necrotic cells. Cancer Res. 61:1659-1665.

Kalinsky, K., L.M. Jacks, A. Heguy, S. Patil, M. Drobnjak, U.K. Bhanot, C.V. Hedvat, T.A.

Traina, D. Solit, W. Gerald, and M.E. Moynahan. 2009. PIK3CA mutation associates with

improved outcome in breast cancer. Clin.Cancer Res. 15:5049-5059.

Karakas, B., K.E. Bachman, and B.H. Park. 2006. Mutation of the PIK3CA oncogene in human

cancers. Br.J.Cancer. 94:455-459.

Kenemans, P., R.A. Verstraeten, and R.H. Verheijen. 2008. Oncogenic pathways in hereditary

and sporadic breast cancer. Maturitas. 61:141-150.

Le Du, F., N.T. Ueno, and A.M. Gonzalez-Angulo. 2013. Breast Cancer Biomarkers: Utility in

Clinical Practice. Curr.Breast Cancer.Rep. 5:10.1007/s12609-013-0125-9.

Leon, S.A., B. Shapiro, D.M. Sklaroff, and M.J. Yaros. 1977. Free DNA in the serum of cancer

patients and the effect of therapy. Cancer Res. 37:646-650.

Mannisto, S., P. Pietinen, M. Virtanen, V. Kataja, and M. Uusitupa. 1999. Diet and the risk of

breast cancer in a case-control study: does the threat of disease have an influence on recall bias?

J.Clin.Epidemiol. 52:429-439.

Michailidou, K., P. Hall, A. Gonzalez-Neira, M. Ghoussaini, J. Dennis, R.L. Milne, M.K.

Schmidt, J. Chang-Claude, S.E. Bojesen, M.K. Bolla, Q. Wang, E. Dicks, A. Lee, C. Turnbull,

N. Rahman, Breast and Ovarian Cancer Susceptibility Collaboration, O. Fletcher, J. Peto, L.

Gibson, I. Dos Santos Silva, H. Nevanlinna, T.A. Muranen, K. Aittomaki, C. Blomqvist, K.

Czene, A. Irwanto, J. Liu, Q. Waisfisz, H. Meijers-Heijboer, M. Adank, Hereditary Breast and

Ovarian Cancer Research Group Netherlands (HEBON), R.B. van der Luijt, R. Hein, N.

Dahmen, L. Beckman, A. Meindl, R.K. Schmutzler, B. Muller-Myhsok, P. Lichtner, J.L.

Hopper, M.C. Southey, E. Makalic, D.F. Schmidt, A.G. Uitterlinden, A. Hofman, D.J. Hunter,

S.J. Chanock, D. Vincent, F. Bacot, D.C. Tessier, S. Canisius, L.F. Wessels, C.A. Haiman, M.

Shah, R. Luben, J. Brown, C. Luccarini, N. Schoof, K. Humphreys, J. Li, B.G. Nordestgaard,

64

S.F. Nielsen, H. Flyger, F.J. Couch, X. Wang, C. Vachon, K.N. Stevens, D. Lambrechts, M.

Moisse, R. Paridaens, M.R. Christiaens, A. Rudolph, S. Nickels, D. Flesch-Janys, N. Johnson,

Z. Aitken, K. Aaltonen, T. Heikkinen, A. Broeks, L.J. Veer, C.E. van der Schoot, P. Guenel, T.

Truong, P. Laurent-Puig, F. Menegaux, F. Marme, A. Schneeweiss, C. Sohn, B. Burwinkel,

M.P. Zamora, J.I. Perez, G. Pita, M.R. Alonso, A. Cox, I.W. Brock, S.S. Cross, M.W. Reed,

E.J. Sawyer, I. Tomlinson, M.J. Kerin, N. Miller, B.E. Henderson, F. Schumacher, L. Le

Marchand, I.L. Andrulis, J.A. Knight, G. Glendon, A.M. Mulligan, kConFab Investigators,

Australian Ovarian Cancer Study Group, A. Lindblom, S. Margolin, M.J. Hooning, A.

Hollestelle, A.M. van den Ouweland, A. Jager, Q.M. Bui, J. Stone, G.S. Dite, C. Apicella, H.

Tsimiklis, G.G. Giles, G. Severi, L. Baglietto, P.A. Fasching, L. Haeberle, A.B. Ekici, M.W.

Beckmann, H. Brenner, H. Muller, V. Arndt, C. Stegmaier, A. Swerdlow, A. Ashworth, N. Orr,

M. Jones, J. Figueroa, J. Lissowska, L. Brinton, M.S. Goldberg, F. Labreche, M. Dumont, R.

Winqvist, K. Pylkas, A. Jukkola-Vuorinen, M. Grip, H. Brauch, U. Hamann, T. Bruning,

GENICA (Gene Environment Interaction and Breast Cancer in Germany) Network, P. Radice,

P. Peterlongo, S. Manoukian, B. Bonanni, P. Devilee, R.A. Tollenaar, C. Seynaeve, C.J. van

Asperen, A. Jakubowska, J. Lubinski, K. Jaworska, K. Durda, A. Mannermaa, V. Kataja, V.M.

Kosma, J.M. Hartikainen, N.V. Bogdanova, N.N. Antonenkova, T. Dork, V.N. Kristensen, H.

Anton-Culver, S. Slager, A.E. Toland, S. Edge, F. Fostira, D. Kang, K.Y. Yoo, D.Y. Noh, K.

Matsuo, H. Ito, H. Iwata, A. Sueta, A.H. Wu, C.C. Tseng, D. Van Den Berg, D.O. Stram, X.O.

Shu, W. Lu, Y.T. Gao, H. Cai, S.H. Teo, C.H. Yip, S.Y. Phuah, B.K. Cornes, M. Hartman, H.

Miao, W.Y. Lim, J.H. Sng, K. Muir, A. Lophatananon, S. Stewart-Brown, P. Siriwanarangsan,

C.Y. Shen, C.N. Hsiung, P.E. Wu, S.L. Ding, S. Sangrajrang, V. Gaborieau, P. Brennan, J.

McKay, W.J. Blot, L.B. Signorello, Q. Cai, W. Zheng, S. Deming-Halverson, M. Shrubsole, J.

Long, J. Simard, M. Garcia-Closas, P.D. Pharoah, G. Chenevix-Trench, A.M. Dunning, J.

Benitez, and D.F. Easton. 2013. Large-scale genotyping identifies 41 new loci associated with

breast cancer risk. Nat.Genet. 45:353-61, 361e1-2.

Newman, A.M., S.V. Bratman, J. To, J.F. Wynne, N.C. Eclov, L.A. Modlin, C.L. Liu, J.W.

Neal, H.A. Wakelee, R.E. Merritt, J.B. Shrager, B.W. Loo Jr, A.A. Alizadeh, and M. Diehn.

2014. An ultrasensitive method for quantitating circulating tumor DNA with broad patient

coverage. Nat.Med. 20:548-554.

Nik-Zainal, S., P. Van Loo, D.C. Wedge, L.B. Alexandrov, C.D. Greenman, K.W. Lau, K.

Raine, D. Jones, J. Marshall, M. Ramakrishna, A. Shlien, S.L. Cooke, J. Hinton, A. Menzies,

L.A. Stebbings, C. Leroy, M. Jia, R. Rance, L.J. Mudie, S.J. Gamble, P.J. Stephens, S.

McLaren, P.S. Tarpey, E. Papaemmanuil, H.R. Davies, I. Varela, D.J. McBride, G.R. Bignell,

K. Leung, A.P. Butler, J.W. Teague, S. Martin, G. Jonsson, O. Mariani, S. Boyault, P. Miron,

A. Fatima, A. Langerod, S.A. Aparicio, A. Tutt, A.M. Sieuwerts, A. Borg, G. Thomas, A.V.

Salomon, A.L. Richardson, A.L. Borresen-Dale, P.A. Futreal, M.R. Stratton, P.J. Campbell,

and Breast Cancer Working Group of the International Cancer Genome Consortium. 2012. The

life history of 21 breast cancers. Cell. 149:994-1007.

Oshiro, C., N. Kagara, Y. Naoi, M. Shimoda, A. Shimomura, N. Maruyama, K. Shimazu, S.J.

Kim, and S. Noguchi. 2015. PIK3CA mutations in serum DNA are predictive of recurrence in

primary breast cancer patients. Breast Cancer Res.Treat. 150:299-307.

Paoletti, C., and D.F. Hayes. 2014. Molecular testing in breast cancer. Annu.Rev.Med. 65:95-

110.

65

Petridis, C., M.N. Brook, V. Shah, K. Kohut, P. Gorman, M. Caneppele, D. Levi, E. Papouli,

N. Orr, A. Cox, S.S. Cross, I. Dos-Santos-Silva, J. Peto, A. Swerdlow, M.J. Schoemaker, M.K.

Bolla, Q. Wang, J. Dennis, K. Michailidou, J. Benitez, A. Gonzalez-Neira, D.C. Tessier, D.

Vincent, J. Li, J. Figueroa, V. Kristensen, A.L. Borresen-Dale, P. Soucy, J. Simard, R.L. Milne,

G.G. Giles, S. Margolin, A. Lindblom, T. Bruning, H. Brauch, M.C. Southey, J.L. Hopper, T.

Dork, N.V. Bogdanova, M. Kabisch, U. Hamann, R.K. Schmutzler, A. Meindl, H. Brenner, V.

Arndt, R. Winqvist, K. Pylkas, P.A. Fasching, M.W. Beckmann, J. Lubinski, A. Jakubowska,

A.M. Mulligan, I.L. Andrulis, R.A. Tollenaar, P. Devilee, L. Le Marchand, C.A. Haiman, A.

Mannermaa, V.M. Kosma, P. Radice, P. Peterlongo, F. Marme, B. Burwinkel, C.H. van

Deurzen, A. Hollestelle, N. Miller, M.J. Kerin, D. Lambrechts, G. Floris, J. Wesseling, H.

Flyger, S.E. Bojesen, S. Yao, C.B. Ambrosone, G. Chenevix-Trench, T. Truong, P. Guenel, A.

Rudolph, J. Chang-Claude, H. Nevanlinna, C. Blomqvist, K. Czene, J.S. Brand, J.E. Olson, F.J.

Couch, A.M. Dunning, P. Hall, D.F. Easton, P.D. Pharoah, S.E. Pinder, M.K. Schmidt, I.

Tomlinson, R. Roylance, M. Garcia-Closas, and E.J. Sawyer. 2016. Genetic predisposition to

ductal carcinoma in situ of the breast. Breast Cancer Res. 18:22-016-0675-7.

Reid, A.L., J.B. Freeman, M. Millward, M. Ziman, and E.S. Gray. 2015. Detection of BRAF-

V600E and V600K in melanoma circulating tumour cells by droplet digital PCR. Clin.Biochem.

48:999-1002.

Sausen, M., S. Parpart, and L.A. Diaz Jr. 2014. Circulating tumor DNA moves further into the

spotlight. Genome Med. 6:35.

Schwarzenbach, H. 2013. Circulating nucleic acids as biomarkers in breast cancer. Breast

Cancer Res. 15:211.

Schwarzenbach, H., D.S. Hoon, and K. Pantel. 2011. Cell-free nucleic acids as biomarkers in

cancer patients. Nat.Rev.Cancer. 11:426-437.

Shaw, J.A., and J. Stebbing. 2014. Circulating free DNA in the management of breast cancer.

Ann.Transl.Med. 2:3-5839.2013.06.06.

Shiovitz, S., and L.A. Korde. 2015. Genetics of breast cancer: a topic in evolution. Ann.Oncol.

Sozzi, G., L. Roz, D. Conte, L. Mariani, F. Andriani, P. Verderio, and U. Pastorino. 2005.

Effects of prolonged storage of whole plasma or isolated plasma DNA on the results of

circulating DNA quantification assays. J.Natl.Cancer Inst. 97:1848-1850.

Takeshita, T., Y. Yamamoto, M. Yamamoto-Ibusuki, T. Inao, A. Sueta, S. Fujiwara, Y. Omoto,

and H. Iwase. 2015. Prognostic role of PIK3CA mutations of cell-free DNA in early-stage triple

negative breast cancer. Cancer.Sci. 106:1582-1589.

UICC, International Union Against Cancer, editors: Sobin, L.H., Gospodarowicz, M.K. and

Wittekind, C. 2009. TNM Classification of Malignant Tumours. Seventh Edition

Vuong, D., P.T. Simpson, B. Green, M.C. Cummings, and S.R. Lakhani. 2014. Molecular

classification of breast cancer. Virchows Arch. 465:1-14.

Wooster, R., and B.L. Weber. 2003. Breast and ovarian cancer. N.Engl.J.Med. 348:2339-2347.

66

Xue, X., M.D. Teare, I. Holen, Y.M. Zhu, and P.J. Woll. 2009. Optimizing the yield and utility

of circulating cell-free DNA from plasma and serum. Clin.Chim.Acta. 404:100-104.