• No results found

Novel preclinical models, therapies and biomarkers for testicular cancer Rosas Plaza, Fernanda

N/A
N/A
Protected

Academic year: 2021

Share "Novel preclinical models, therapies and biomarkers for testicular cancer Rosas Plaza, Fernanda"

Copied!
141
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Novel preclinical models, therapies and biomarkers for testicular cancer Rosas Plaza, Fernanda

DOI:

10.33612/diss.119056452

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Rosas Plaza, F. (2020). Novel preclinical models, therapies and biomarkers for testicular cancer.

Rijksuniversiteit Groningen. https://doi.org/10.33612/diss.119056452

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

NOVEL PRECLINICAL MODELS, THERAPIES AND BIOMARKERS FOR TESTICULAR CANCER

PhD thesis

(3)

Cover and layer-out design: Merit González Printing of this thesis was supported by:

- Stichting Werkgroep Interne Oncologie - The Graduate School of Medical Sciences - University Library

© Copyright 2020, F. X. Rosas Plaza

All rights reserved. No part of this thesis may be reproduced, stored or

transmitted in any form without permission by the author.

(4)

to obtain the degree of PhD at the University of Groningen

on the authority of the

Rector Magnificus Prof. C. Wijmenga and in accordance with

the decision by the College of Deans.

This thesis will be defended in public on Monday 9 March 2020 at 14:30

by

born on 31 May 1987 in Mexico City, Mexico

PhD thesis

Fernanda Ximena Rosas Plaza

therapies and biomarkers

for testicular cancer

(5)

Prof. S. de Jong Prof. J. A. Gietema

Prof. M. A. T. M. van Vugt Assessment committee Prof. W.N. Keith

Prof. A. van den Berg

Prof. F.A.E. Kruyt

(6)

Table of contents

Chapter 1 Chapter 2

Chapter 3

Chapter 4

Chapter 5

Chapter 6 Chapter 7

General Introduction and outline of thesis 7 miR-371a-3p, miR-373-3p and miR-367-3p as serum biomarkers in metastatic testicular germ cell cancers before, during and after chemotherapy 29 Cells, 2019, 8, 1221

Dual mTORC1/2 inhibition sensitizes testicular cancer

models to cisplatin treatment 55

Molecular Cancer Therapeutics, 2020, 19: 590-601 Establishment and characterization of testicular cancer patient-derived xenograft models for preclinical

evaluation of novel therapeutic strategies 81 In preparation

AFP levels do not predict response to cisplatin in

testicular cancer patient-derived xenograft models 105 Summary, general discussion and future perspectives 115 Nederlandse samenvatting (Summary in Dutch) 129

Acknowledgements 135

List of publications 138

(7)
(8)

Chapter 1

General introduction and outline of the thesis

(9)
(10)

9

Clinical and pathological characteristics of testicular cancer

Testicular cancer (TC) accounts for approximately 1% of all cancers in men worldwide, while it is also the most common solid tumor among young men (15-40 years)

1

. Incidence of TC varies widely across different regions, with an age- standardized rate of 8.7% in Western Europe compared to 0.2% in Central Africa

1

. Incidence rates of TC have increased since the 1960s in Northern European countries, affecting age groups older than 15 years. No causative mechanism for this increase had been identified so far

2

.

Well known risk factors associated with the development of TC include cryptorchidism, a family history of TC and a previous diagnosis of TC

3

. Other recently described risk factors are low birth weight, low gestational age at birth, inguinal hernia, high intake of dairy products and ‘heavy’ cannabis use

4–6

. Germ cell tumor (GCT) is the predominant histology of TC patients (95%), divided into seminomas and non-seminomas, of which seminomas are slightly more common than non-seminomas

7

. GCTs arise from gonocytes that failed to differentiate into spermatogonias, and the pluripotency of these cells allow them to generate highly diverse histological tumors. Seminomas are blocked in the earliest differentiation state whereas non-seminomas consist of diverse histological subtypes depending on the degree of differentiation: embryonal carcinoma (undifferentiated), choriocarcinoma, yolk sac tumor or teratoma (well-differentiated)

8

. Mixed GCTs are common and may appear in any form.

Diagnosis of TC is performed with clinical evaluation, ultrasound examination of the testis and determination of serum tumor markers. These tumor biomarkers include LDH (lactate dehydrogenase), AFP (alpha-fetoprotein) and ß-HCG (ß-human chorionic gonadotropin), and assist in the diagnosis, risk stratification and follow-up of TC patients

9

. Current risk stratification was established in 1997 by the International Germ Cell Consensus Classification (IGCCC) and constitutes a prognostic staging system for patients with disseminated disease (Table 1)

10

.

Overall cure rate of TC is good, mostly resulting from the high sensitivity of TCs

to the chemotherapeutic cisplatin in a metastatic setting. Relative 5-year overall

survival of seminoma and non-seminoma patients is 98% and 93%, respectively

2

.

Patients with stage I disease have more than 99% overall survival regardless of

tumor type. However, patients with metastatic disease have different survival

outcomes depending on the tumor type and prognosis group. The IGCCC

classification stratifies patients into good, intermediate and poor prognosis groups

(11)

Table1. Risk stratification for metastatic patients

12

Non-seminoma Seminoma

Risk group RMH

Stage Spread and tumor markers Survival Spread and tumor markers Survival Good

prognosis

II-IV • Testis or retroperitoneal primary

• No metastasis to organs other than the lung and/

or lymph nodes

• Tumor markers level normal or at least 1 tumor marker above normal:

• LFD < 1.5x ULN

• beta-hCG < 5000 mIU/mL

• AFP < 1000 ng/mL

56% of patients 5-year PFS: 89%

5-year OS: 92%

• Any primary site

• No metastasis to organs other than lungs and/or lymph nodes

• Normal AFP

• Any beta-hCG and LDH

90% of patients 5-year PFS: 82%

5-year OS: 86%

Intermediate prognosis

II-IV • Testis or retroperitoneal primary

• No metastasis to organs other than the lungs and/

or lymph nodes

• At least 1 tumor marker substantially above normal:

• LDH 1.5 - 1.0x ULN beta-hCG ≥ 5000 ≤ 50000 mIU/mL

• AFP ≥ 1000 ≤ 10000 ng/mL

28% of patients 5-year PFS: 75%

5-year OS: 80%

Any primary site Metastasis to organs other than the lungs and/or lymph nodes Normal AFP Any beta-hCG and LDH

10% of patients 5-year PFS: 67%

5-year OS: 72%

Poor prognosis

II-IV • Mediastinal primary with or without metastases

• Metastasis to organs other than the lungs and/or lymph nodes

• At least 1 or more tumor maker levels highly elevated:

• LDH > 10x ULN

• beta-hCG > 50000 mIU/mL

• AFP > 10000 ng/mL

16% of patients 5-year PFS: 41%

5-year OS: 48%

No poor prognosis patients

No poor prognosis patients

RMH: Royal Marsden Hospital, LDH: lactate dehydrogenase, hCG: human chorionic gonadotropin, AFP: alpha- fetoprotein, ULN: upper limit of normal, PFS: progression free survival, OS: overall survival

which are determined based on clinical features: the location of the metastasis

and levels of different tumor biomarkers. Even though recent studies have

shown improvement of the initially reported survival rates

11

, general consensus

states that the good prognosis group has an 86% and 92% 5-year overall survival

for seminoma and non-seminoma patients respectively. Intermediate prognosis

TC patients have 72% and 80% 5-year overall survival for seminoma and non-

seminoma tumors, respectively, and the poor prognosis group including only

non- seminoma patients have a 48% 5-year overall survival

10

.

(12)

11

Standard treatment of TC

Stage I or localized disease is treated with orchiectomy, while metastatic patients are given cisplatin based chemotherapy after orchiectomy is performed.

Patients with disseminated disease are treated with three or four courses of BEP regimen including bleomycin, etoposide and cisplatin. Approximately 20% of patients with disseminated disease will need second-line treatment as a consequence of relapse or refractory disease

13

. Different salvage treatments have been reported with long term remissions ranging from 23% using VeIP/

VIP (cisplatin, ifosfamide and etoposide/vinblastine)

14, 15

to 63% and 73% using TIP (paclitaxel, ifosfamide and cisplatin)

13

or GIP (gemcitabine, ifosfamide and cisplatin)

16

respectively. High-dose chemotherapy initially showed promising results, however more recent clinical trials have not demonstrated superior efficacy over standard salvage treatments

17, 18

.

Genetic alterations in TC

Few susceptibility loci associated with the development of TC have been identified. Independent genome wide association studies found that genetic predisposition of TC was associated to variants influencing the KITLG/KIT pathway. Variants within the 12q22 region, comprising the KITLG gene, were associated with an elevated risk of TC

19, 20

. KITLG gene encodes the ligand for the receptor tyrosine kinase c-KIT. Chromosome 5 was also found to have a TC- associated variant affecting the SPRY4 gene. SPRY4 functions as an inhibitor of mitogen-activated protein kinase (MAPK) pathway, which is downstream of the KIT pathway. Both susceptibility loci were associated with seminomas and non-seminomas, suggesting that despite their diverse histology they share a common biological origin, further supported by the frequent observation of mixed GCTs.

TCs are highly aneuploid and frequently show large scale copy number

gains and losses. Copy number gains on chromosomes 7, 8, 21, 22 and X are

commonly observed as well as losses on chromosomes 4, 5, 11, 13 and 18

21

.

However the most common anomaly is the presence of a 12p isochromosome

(i(12p)) affecting more than 80% of TCs

22, 23

. This region of the small arm of

chromosome 12 contains the oncogene KRAS, as well as stem cell related genes

including NANOG and STELLAR. Of note, pre-malignant lesions do not contain

i(12p)

24

suggesting that it is not involved in the development of the precursor

lesion. Importantly, amplifications of the KIT gene were identified in 21% of

seminomas and 9% of non-seminomas. Copy number gains of the KIT gene,

located on chromosome 4, were associated with an increased expression of the

protein

25

.

(13)

Despite the high aneuploidy of TCs, the somatic mutation rate is low. Whole exome sequencing revealed that TC tumors have a mutation rate of 0.51 somatic mutations per Mb, while lung cancers carry 8.0 mutations/Mb, and melanomas 11.0 mutations/Mb

23

. KIT was the most significantly mutated gene in TC, observed predominantly in seminomas

23, 26

. The proportion of KIT hotspot mutations in seminomas ranges from 18%

27

to 31.3%

23

. Studies investigating mutations in K-RAS and N-RAS specifically, have found that incidence of RAS mutations is on average 20%-30% in both seminomas and non-seminomas

27, 28

. Available data on which tumor type has a higher incidence of RAS mutations is non-conclusive. Mutations in cell division cycle 27 gene (CDC27) were recently described in 11.9% of TC patients and few PIK3CA and AKT1 mutations have been reported as well

23, 29

. Accordingly, pathway analysis of somatic mutations identified that genes involved in metabolism were the most frequently mutated (93%), with the PI3K/AKT pathway as one of the most affected pathways (54%) in TC

23

.

Relevance of cisplatin and apoptosis induction in TC

Since the introduction of cisplatin-based chemotherapy in the 1970s, curing rates of metastatic TC improved drastically

30

. Even though cisplatin has been used in the clinic for a long time and is a curative drug in some types of pediatric cancer and young adult cancers in addition to TC, the mechanisms behind the excellent sensitivity in these tumor types are still elusive. TP53 is one of the most commonly mutated genes in cancer, however it is rarely altered in TC before treatment

31

. The presence of wild type (WT) TP53 is thought to be one of the key factors involved in response to chemotherapy in these patients. The TP53 gene encodes the p53 protein, a transcription factor involved in many cellular processes including cell cycle arrest, DNA repair, apoptosis, autophagy and metabolism

32

.

Cellular stress in response to cisplatin treatment has been widely studied. Once

cisplatin enters the cell, it interacts with DNA. Cisplatin generates different forms

of DNA adducts, which are predominantly intrastrand cross-links and secondly

interstrand cross-links, protein-DNA cross-links and DNA monoadducts

33, 34

.

The cisplatin-DNA cross-links disrupt the structure of the DNA which can be

recognized by DNA repair proteins. Cisplatin-induced intrastrand crosslinks are

mostly repaired by the nucleotide excision repair (NER) pathway

35

. However,

human cells deal poorly with this type of DNA damage due to the binding of high

mobility groups (HMG) proteins to the DNA adducts, blocking their repair

36,37

.

In addition, some essential proteins involved in NER including ERCC1, XPF

and XPA have low expression levels in TC, contributing to the high levels of

sensitivity to cisplatin treatment

38

. Furthermore, the cytotoxicity of cisplatin can

partially be explained by to the formation of DNA double strand breaks (DSBs),

(14)

13

during the processing of DNA interstrand cross-links in replicating cells

39

. The DNA damage response (DDR) machinery is activated in response to DSBs and as a consequence leads to activation of the ataxia telangiectasia mutated (ATM) kinase. ATM subsequently phosphorylates p53 as well as the main p53 negative regulator, mouse double minute homolog (MDM2). Both replication stress and DNA damage can stabilize p53, for example, by promoting p53 phosphorylation and blocking the degradation regulated by the p53 inhibitor MDM2. The phosphorylated form of MDM2 is unable to ubiquitinate p53, allowing its full activation

40

. As a result of DNA damage, p53 induces cell cycle arrest and apoptosis. The upregulation of p21/CDKN1A, encoded by the CDKN1A gene, is crucial for the induction of cell cycle arrest in G1 phase. Apoptosis, on the other hand, can be triggered by the p53-mediated transcription of pro-apoptotic BCL-2 family members including PUMA and PMAIP1 (NOXA)

41

. Activation of the DDR induced by cisplatin in TC, leads to a rapid induction of apoptosis with a major role for p53. Contributing to the hypersensitivity of TCs to apoptosis is the low mutation rate of TP53, as mentioned before

31

. Of note, no correlation was found between p53 expression levels and cisplatin sensitivity.

Accumulation of cisplatin and the amount of cisplatin bound to DNA in TC cells are not clearly correlated with cisplatin sensitivity, which is more related to the lack of repair of cisplatin-damaged DNA in TC cells

42, 43

. For TC it has been described that cisplatin-induced p53 activation can induce apoptosis via both the extrinsic apoptotic pathway mediated by the FAS death receptor

44, 45

and the intrinsic apoptotic pathway via the BH3-only family members: p53 upregulated modulator of apoptosis (PUMA) and Noxa

46

. FAS receptor activation results in cleavage of procaspase-8, leading to cleavage of the pro- apoptotic BH3 interacting-domain death agonist (BID). The cleaved BID fragment (tBID) targets the mitochondria and induces the oligomerization of BCL-2- associated X protein (BAX) and BCL-2-antagonist/killer 1 (BAK) in the outer mitochondrial membrane. Mitochondrial leakage leads to the release of cytochrome C and cleavage of procaspase-9 into fully active caspase-9

47

. Activation of the intrinsic apoptotic pathway is regulated by molecular interactions between members of the B cell lymphoma 2 (BCL-2) family of apoptosis regulating proteins, which can be either pro- or anti-apoptotic. The BH3-only proteins promote BAX and BAK activation, by inhibiting the anti-apoptotic Bcl-2 proteins and, in some instances, by directly engaging with BAX and BAK

48

. The anti-apoptotic Bcl-2 members prevent apoptosis initiation by sequestering pro-apoptotic proteins.

Caspase 8 and 9 activation will lead to cleavage of executioner caspases (-3, -6 and -7) that will orchestrate the degradation of the cell

49

.

Even though most TC tumors retain WT TP53, some posttranscriptional

modifications are able to repress p53 pro-apoptotic activity. For example,

carboxyl- terminal lysine methylation on p53 was shown to inhibit PUMA and

CDKN1A expression in teratocarcinoma cells

50

. Methyltransferase knockdown

(15)

and expression of p53 mutants that cannot be methylated restored PUMA and CDKN1A expression. Although effects on apoptosis by PUMA or CDKN1A downregulation and restoration were not evaluated, their involvement in response to cisplatin has been reported in TC

51

and other cancer types

52

. However, whether cells enter cycle arrest or undergoes apoptosis as a consequence of CDKN1A or PUMA upregulation, respectively, remains unclear and might be context dependent

41

.

Cisplatin resistance in TC

Genomic alterations affecting the MDM2/p53 axis have been described in chemo- resistant TC. A large study of 180 TC tumors using whole exome sequencing data (WES) described that most of the samples with MDM2/p53 alterations were found in resistant tumors and the majority were post-treatment specimens

53

. Moreover, chemo-resistant tumors exhibited a trend towards higher positivity for both p53 and MDM2 compared to sensitive specimens, suggesting increased importance of MDM2 mediated inhibition of p53 activity

54

. This hypothesis is supported by functional studies using cisplatin-sensitive and -resistant TC cell lines indicating that the interaction between p53 and MDM2 requires higher doses of cisplatin to be disrupted in resistant cell lines

45

. More genomic alterations in PIK3CA, AKT1 and MTOR are found in resistant tumors, although not statistically significant. KRAS mutations are also overrepresented in resistant tumors. These data are in line with similar reports that compared sensitive versus resistant tumors and found a small number of PIK3CA, AKT1 and KRAS mutations in resistant tumors only

29

.

The PI3K/Akt/mTOR pathway has been described to be involved in response to cisplatin using mutational analyses, as mentioned earlier, but also in several functional studies. Akt kinase hyperactivation was seen in resistant TC cells by putative PDGFRß overexpression

55

(Figure 1). PDGFRß inhibition with Pazopanib resulted in sensitization to cisplatin treatment in vitro and was an effective treatment in combination with the HER2 inhibitor lapatinib to treat resistant TC models in vivo

56

. PI3K and Akt inhibition were tested in combination with cisplatin in resistant TC cell lines showing apoptosis induction compared to cisplatin treatment alone

51

.

As described above excellent sensitivity of TC tumors to cisplatin treatment is partly caused by a diminished capacity to repair cisplatin-DNA adducts.

Consequently, cisplatin resistance may arise from an enhanced ability to resolve

DNA damage, or by increased coping with unrepaired lesions. Overexpression

of HMGB1, which can inhibit NER by binding to the DNA adducts, has been

reported in several tumor types and was associated with poor prognosis

57–61

.

In addition, increased expression of ERCC1 in ovarian cancer cell lines was

(16)

15

associated with cisplatin resistance

62

. Currently there is no data available suggesting that changes to ERCC1 or HMG proteins such as posttranslational modifications are involved in cisplatin resistance of TC. Reduced HR activity may contribute to cisplatin sensitivity in TC. One report has compared cisplatin sensitive and resistant TC cell lines, showing that cisplatin sensitivity correlated with the level of proficiency of TC cells to repair interstrand cross-links. Despite differences in HR proficiency between cisplatin sensitive and resistant cell lines, all TC cell lines proved more HR deficient compared to a non-TC cisplatin resistant cell line

63

. More research should be conducted to determine if and how DNA damage repair pathways are involved in cisplatin resistance of TC.

Figure 1. Pathways involved in cisplatin sensitivity

Once cisplatin enters TC cells it induces different types of DNA damage. This leads to the initiation of DNA damage response (DDR), which is mainly orchestrated by p53. DNA damage induced stabilization of p53 results in p53-mediated transcription of different proteins, i.e. MDM2, its main negative regulator, and the pro-apoptotic proteins PUMA and NOXA. High expression of these pro-apoptotic proteins activates the intrinsic apoptotic pathway. After oligomerization of BAX and BAK in the outer mitochondrial membrane cytochrome C is released from the mitochondria of as a result caspase 9 is activated. The extrinsic apoptotic pathway can be initiated by p53-mediated increase and activation of the FAS receptor, which induces procaspase-8 cleavage. Growth factor stimuli provide pro-survival signals to the cell through the PI3k/

AKT/mTOR pathway by inducing protein synthesis and inhibiting pro-survival proteins. The Ras/Raf/ERK pathway also provides pro-survival signals to the cell. Red line indicates inhibition, black line indicates stimulation.

FAS ligand

FASR

Proliferation Survival

Translational control Protein synthesis

Polyubiquitination and proteasomal degradation Cell cycle arrest DNA repair Metabolism

P P

Ras

PRaf

PI3K

PTEN

p53

BAD

PUMA mTORC2

mTORC1

4e-BP1

PDK1 PIP2

MEK 1/2 P

ERK 1/2 P

P P

PAKTP PPIP3

P P

UbUb

p53P MDM2P

DDR

Cisplatin Hypoxia

Retinoic acid Noxa

Oct4 Cytochrome C

Cytochrome C

BAX BAK

BAX BAK

Apoptosis BCL2,

BCLxL MCL1 BID tBID

Caspase 8Pro

Active Caspase 8

Caspase 9

Caspase 3/7 FADD

P S6P

(17)

Novel targeted therapies in TC

Clinical trials using targeted drugs are scarce. The susceptibility of TC towards inhibition of the PI3K/AKT/mTORC pathway in preclinical studies pushed forward two clinical trials in patients with refractory disease or relapse using everolimus

64

and pazopanib

56

as single agents. Efficacy of the mTORC1 inhibitor everolimus was minimal, while treatment with the multitargeted tyrosine kinase (TK) inhibitor pazopanib achieved limited tumor activity. Two other small clinical trials tested the efficacy of TK inhibition alone in heavily treated TC patients.

Imatinib was used to treat 6 KIT-positive patients with imatinib showing poor activity with 5 out of 6 patients developing progressive disease

65

. Five patients refractory to first-line therapy were included in a phase II study in which the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib was tested

66

. Only one of these patients responded to treatment. Finally, a recent article studied the response of refractory TC to immune checkpoint inhibition.

Twelve patients were treated with the antibody against PD-1, pembrolizumab, with no partial or complete responses observed

67

.

Another interesting pathway as a clinical target for treatment of TC is the DDR pathway. TC tumors have been shown to be deficient for interstrand DNA cross- link repair, leading to formation of DSBs

68

. Normally, DSBs are repaired by HR, a DNA repair mechanism that is impaired in TC cells

63

. Unresolved DSBs, however, provide a therapeutic opportunity as HR-deficient tumors are highly sensitive to PARP1 inhibitors

69–71

. Expression of PARP1 was high in TC tumors and not in normal testis tissue, but did not show a correlation with clinical variables

72

. Treatment of TC cells with the PARP inhibitor olaparib sensitized cells to cisplatin treatment, in particular cisplatin-resistant cell lines

63

. Currently, two phase II trials are evaluating the potential of PARP inhibition in relapsed or refractory TC, either as single agent (NCT02533765, active, not recruiting) or combined with gemcitabine and carboplatin (NCT02860819, active and recruiting).

Novel circulating biomarkers in TC

Serum tumor markers LDH, AFP and ß-HCG are used to follow clinical response

to chemotherapy in patients with metastatic disease. However, only ~60% of TC

patients show elevated tumor markers at start of treatment

73

. 90% of patients

with non-seminomatous tumors are positive for AFP or ß-HCG, while 30% of

seminoma patients are positive for ß-HCG

9

. Of note, AFP can be produced by

other malignancies as well, including hepatocellular carcinoma and chronic

liver diseases, e.g. cirrhosis. Other tumor types can also secrete ß-HCG, like

pancreatic, biliary and gastric cancers

73

. Independent of tumor type, LDH is

elevated in 40- 60% of cases; although LDH is the least specific marker of all

(18)

17

three

74

. The IGCCC stratification helps identifying patients that are at higher risk of dying of the disease. However, a large subset of patients classified as intermediate and poor prognosis will be cured with chemotherapy. Tumor markers that more accurately predict disease outcome are therefore needed.

Only a few novel circulating biomarkers have been studied in TC: the circulating cytokeratin 18

75

, the demethylated promoter regions of the long non-coding RNA X inactive transcript (XIST) gene

76

and the non-coding microRNA cluster miR- 371

77

. Circulating full-length and caspase-cleaved cytokeratin 18 (CK18) are regarded as markers of cell death in patients treated with chemotherapy. CK18 levels were evaluated in 34 patients during the course of chemotherapy to study their correlation with treatment response. Even though CK18 levels rose after start of each chemotherapy course, the study lacked enough patients with poor clinical outcome to confirm the utility of this marker to predict clinical response.

Demethylated promotor regions of XIST have been evaluated in a small study using plasma DNA of TC patients and healthy donors. 55% of non-seminoma and 71% of seminoma patients’ plasma was positive for unmethylated XIST, while 0% of the healthy donors resulted positive. Despite the high specificity of this tumor maker, sensitivity was relatively low. In addition, no studies confirming the applicability of demethylated XIST have been published. Finally, several studies have shown the potential utility of members of the miR-371 cluster (miR-371a-3p, miR-372-3p and miR-373-3p) and the pluripotency associated miR-367-3p

78

in diagnosis

79–83

and follow-up of TC patients. Levels of these miRs in blood have been shown to predict the presence of viable disease

84

or detect relapses after chemotherapy

85, 86

. As such, these miRs are recognized as putative tumor markers for diagnosis and follow-up of TC patients.

Novel preclinical models in TC

Despite the fact that TC is a complex and heterogeneous disease, TC models including cell lines and xenografts are scarce. Only 20 human and mouse cell lines

87, 88

and less than 10 xenograft models

89–98

have been reported. Available cell lines and xenografts almost exclusively cover the embryonal carcinoma subtype highlighting the obvious need for more models representing the other histological subtypes.

Patient derived xenografts (PDX) are used more frequently in current cancer

research due to several advantages over cell line-based xenografts. Some highly

valuable features of PDX models are maintenance of tumor heterogeneity, high

similarity to human tumors

99,100

and prediction of drug response to treatment

101,102

.

Use of TC PDX models in cancer research has been described

55, 56, 103, 104

. So far, only

14 orthotopically established TC PDX models

105

, and 12 subcutaneous TC PDX

models

106

have been reported. However, only one systematic report is available

of their establishment

107

. The aforementioned study described the establishment

(19)

of 14 non-seminomatous PDX models, specifically from the choriocarcinoma, embryonal carcinoma and yolk sac subtypes, as well as mixed tumors with yolk sac, teratoma and embryonal carcinoma components. Orthotopic implantation of the tumor pieces was more successful than subcutaneous implantation.

These PDX models were used to study cisplatin resistance and to test novel combinatorial strategies using a glucosylceramide synthase (GCS) inhibitor, DL- threo-PDMP, and a multi-targeted receptor tyrosine kinase inhibitor, sunitinib, in combination with cisplatin

107, 108

.

Other promising 3D models in cancer research are organoids. Testicular organoids have been developed mostly from non-cancerous human adult tissue with the limitation of testicular histological organization being hardly recognized

109, 110

. One study that used seminoma tumors to develop hanging-drop cultures reported that tumor morphology was preserved, however, it was conserved for only 7 days, along with proliferative capacity

111

. Sensitivity to chemotherapeutic drugs like cisplatin was tested in human organoids derived from non-cancerous tissue showing decreased viability compared to untreated organoids

112

. However, toxicity studies using other anticancer drugs and combinatorial strategies on cancerous TC organoids are lacking.

Thus, even though TC is a highly curable disease there are some therapeutic challenges left to tackle. Tumor marker evaluation before and during treatment is a highly valuable tool, yet, the available tumor markers, AFP, ß-HCG and LDH have several limitations in terms of sensitivity and specificity. This is mostly detrimental for follow-up of patients. Another issue affecting TC patient’s survival is cisplatin resistance, since there are no other therapeutic options for these men that relapse and do not respond to second line treatment or those that develop refractory disease. Finally, one of the most reliable cancer models that researches have are PDX models, but few TC models are available. By increasing the amount of models and the biological knowledge of these models, we could test novel therapies and study cisplatin resistance in depth.

Therefore, the aim of this thesis is to tackle these therapeutic challenges. Clinical

use of miR levels as tumor markers was evaluated in TC patients in order to

improve diagnosis, stratification and follow-up of TC patients. Additionally, we

explored cisplatin resistance in TC and developed in vivo models to test novel

combinatorial therapies identified in the laboratory.

(20)

19

In chapter 2 we explored the behavior of three microRNAs, miR-371a-3p, miR-373- 3p and miR-367-3p, in TC patients with metastatic disease receiving chemotherapy. Recent studies have shown the above mentioned miRs are upregulated specifically in TC patients compared to other cancer patients. Relative levels of miR-371a-3p, miR-373-3p and miR-367-3p were evaluated in serum of metastatic TC patients using the ampTSmiR test. Two cohorts were included:

a prospectively included and a retrospectively selected cohort were studied.

In total, 109 patients were included in the study. Blood samples were drawn at start of treatment and during follow-up at week 1, 3, 6, 8 and after 3-6 months.

The last evaluated sample was taken after 1 year of completing chemotherapy.

We studied the behavior of miR levels during and after chemotherapy and it’s correlation with classical tumor markers, treatment and poor outcome.

In chapter 3 we aimed to find novel combinatorial therapies to sensitize TC cells lines towards cisplatin in order to provide new treatment options for TC patients that do not respond to cisplatin-based chemotherapy. It has been shown that PI3K/Akt downregulation sensitizes resistant TC cell lines and PDX models to cisplatin

51

. Additionally, hyperactivation of the PI3K/AKT/mTORC pathway was linked to cisplatin resistance showing resistant sublines had higher levels of p-AKT473 compared to their sensitive parental cells

56

. In this chapter we studied the activation of kinases, including receptor tyrosine kinases, and downstream substrates in five cisplatin-sensitive or resistant TC cell lines using phospho-kinase arrays and western blotting. We tested inhibitors of most active kinases in the PI3K/AKT/mTORC pathway using apoptosis assays and survival assays to evaluate the most effective treatment in TC cell lines. Additionally, two TC patient-derived xenografts (PDX) from a chemo-sensitive and a -resistant patient, were treated with cisplatin in the absence or presence of kinase inhibitor.

In chapter 4 we evaluated the feasibility to use tumor samples obtained via orchiectomy from TC patients operated at the University Medical Center Groningen to develop subcutaneous TC PDX models between March 2016 and June 2018, we obtained 10 tumor samples from which one tumor sample was diagnosed as a precancerous lesion, eight were non seminomas and one was pure seminoma. We could establish 3 TC PDX models by subcutaneous implantation of solid tumor pieces in NOD scid gamma (NSG) mice. PDX models and matched patient tumors were characterized using immunohistochemistry to assess proliferation (Ki-67), human vs mouse stroma (Cyclophilin A) and caspase activity (cleaved caspase-3) as well as copy number variation and RNA- sequencing analysis to evaluate tumor evolution over several passages. PDX models were treated with cisplatin and other targeted drugs to assess their sensitivity towards them.

OUTLINE OF THE THESIS

(21)

In chapter 5, we investigated whether PDX models can be used as suitable models to study two well-known TC biomarkers: AFP and ß-HCG. These markers are used in diagnosis, and follow-up of TC patients. However, not all tumors from TC patients secrete these proteins. In addition, chemotherapy treatment results in normalized marker levels in some initially marker-positive TC patients, although viable tumor cells are still present. Since PDX tumors consist of human tumor cells and mouse normal cells, these models allow studying tumor cell specific secretion of these markers. We selected three PDX models, derived from three patients that were AFP positive and two of them having detectable ß-HCG levels as well. We related the expression of these markers in the PDX models before and after treatment with cisplatin with tumor volume using immunohistochemistry and ELISA.

In chapter 6, we summarized and analyzed the findings of this thesis in light of

current knowledge, and finally provide future perspectives.

(22)

21

References

1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.

2. Ylönen O, Jyrkkiö S, Pukkala E, Syvänen K, Boström PJ. Time trends and occupational variation in the incidence of testicular cancer in the Nordic countries. BJU Int.

Wiley/Blackwell (10.1111); 2018;122:384–93.

3. Gatta G, Trama A. Epidemiology of Testicular Cancer. Pathol Testic Penile Neoplasms. Cham: Springer International Publishing; 2016. page 3–18.

4. Garner MJ, Birkett NJ, Johnson KC, Shatenstein B, Ghadirian P, Krewski D, et al.

Dietary risk factors for testicular carcinoma. Int J Cancer. 2003;106:934–41.

5. Cook MB, Akre O, Forman D, Madigan MP, Richiardi L, McGlynn KA. A systematic review and meta-analysis of perinatal variables in relation to the risk of testicular cancer—

experiences of the son. Int J Epidemiol. Oxford University Press; 2010;39:1605–18.

6. Callaghan RC, Allebeck P, Akre O, McGlynn KA, Sidorchuk A. Cannabis Use and Incidence of Testicular Cancer: A 42-Year Follow-up of Swedish Men between 1970 and 2011. Cancer Epidemiol Biomarkers Prev. American Association for Cancer Research; 2017;26:1644–52.

7. Richiardi L, Bellocco R, Adami H-O, Torrång A, Barlow L, Hakulinen T, et al.

Testicular Cancer Incidence in Eight Northern European Countries: Secular and Recent Trends. Cancer Epidemiol Biomarkers Prev. 2004.

8. Hanna NH, Einhorn LH. Testicular Cancer — Discoveries and Updates. Longo DL, editor. N Engl J Med. 2014;371:2005–16.

9. Albers P, Albrecht W, Algaba F, Bokemeyer C, Cohn-Cedermark G, Fizazi K, et al.

Guidelines on Testicular Cancer: 2015 Update. Eur Urol. 2015;68:1054– 68.

10. Mead GM, Stenning SP. The International Germ Cell Consensus Classification: a new prognostic factor-based staging classification for metastatic germ cell tumours.

Clin Oncol (R Coll Radiol). 1997;9:207–9.

11. van Dijk MR, Steyerberg EW, Habbema JDF. Survival of non-seminomatous germ cell cancer patients according to the IGCC classification: An update based on meta- analysis. Eur J Cancer. 2006;42:820–6.

12. International Germ Cell Consensus Classification: a prognostic factor-based staging system for metastatic germ cell cancers. International Germ Cell Cancer Collaborative Group. J Clin Oncol. 1997;15:594–603.

13. Kondagunta GV, Bacik J, Donadio A, Bajorin D, Marion S, Sheinfeld J, et al. Combination of Paclitaxel, Ifosfamide, and Cisplatin Is an Effective Second- Line Therapy for Patients With Relapsed Testicular Germ Cell Tumors. J Clin Oncol. 2005;23:6549–55.

14. Miller KD, Loehrer PJ, Gonin R, Einhorn LH. Salvage chemotherapy with vinblastine, ifosfamide, and cisplatin in recurrent seminoma. J Clin Oncol. 1997;15:1427–31.

15. McCaffrey JA, Mazumdar M, Bajorin DF, Bosl GJ, Vlamis V, Motzer RJ. Ifosfamide-

and cisplatin-containing chemotherapy as first-line salvage therapy in germ cell

tumors: response and survival. J Clin Oncol. 1997;15:2559–63.

(23)

16. Fizazi K, Gravis G, Flechon A, Geoffrois L, Chevreau C, Laguerre B, et al. Combining gemcitabine, cisplatin, and ifosfamide (GIP) is active in patients with relapsed metastatic germ-cell tumors (GCT): a prospective multicenter GETUG phase II trial. Ann Oncol Off J Eur Soc Med Oncol. 2014;25:987–91.

17. Pico J-L, Rosti G, Kramar A, Wandt H, Koza V, Salvioni R, et al. A randomised trial of high-dose chemotherapy in the salvage treatment of patients failing first-line platinum chemotherapy for advanced germ cell tumours. Ann Oncol Off J Eur Soc Med Oncol. 2005;16:1152–9.

18. Necchi A, Mariani L, Di Nicola M, Lo Vullo S, Nicolai N, Giannatempo P, et al.

High-dose sequential chemotherapy (HDS) versus PEB chemotherapy as first- line treatment of patients with poor prognosis germ-cell tumors: mature results of an Italian randomized phase II study. Ann Oncol. Oxford University Press;

2015;26:167–72.

19. Kanetsky PA, Mitra N, Vardhanabhuti S, Li M, Vaughn DJ, Letrero R, et al. Common variation in KITLG and at 5q31.3 predisposes to testicular germ cell cancer. Nat Genet. Nature Publishing Group; 2009;41:811–5.

20. Rapley EA, Turnbull C, Al Olama AA, Dermitzakis ET, Linger R, Huddart RA, et al.

A genome-wide association study of testicular germ cell tumor. Nat Genet. Nature Publishing Group; 2009;41:807–10.

21. Summersgill1 B, Goker’ H, Weber-Halll S, Huddart3 R, Horwich3 A, Shipley1 J. Molecular cytogenetic analysis of adult testicular germ cell tumours and identification of regions of consensus copy number change. Br Joumal Cancer.

1998.

22. Sandberg AA, Meloni AM, Suijkerbuijk RF. REVIEWS OF CHROMOSOME STUDIES IN UROLOGICAL TUMORS. 111. CYTOGENETICS AND GENES IN TESTICULAR TUMORS. J UROlOGY. 1996.

23. Litchfield K, Summersgill B, Yost S, Sultana R, Labreche K, Dudakia D, et al. Whole- exome sequencing reveals the mutational spectrum of testicular germ cell tumours.

Nat Commun. Nature Publishing Group; 2015;6:5973.

24. Ottesen AM, Skakkebaek NE, Lundsteen C, Leffers H, Larsen J, Rajpert-De Meyts E. High-resolution comparative genomic hybridization detects extra chromosome arm 12p material in most cases of carcinoma in situ adjacent to overt germ cell tumors, but not before the invasive tumor development. Genes, Chromosom Cancer. 2003;38:117–25.

25. McIntyre A, Summersgill B, Grygalewicz B, Gillis AJM, Stoop J, van Gurp RJHLM, et al.

Amplification and Overexpression of the KIT Gene Is Associated with Progression in the Seminoma Subtype of Testicular Germ Cell Tumors of Adolescents and Adults.

Cancer Res. 2005;65:8085–9.

26. Kemmer K, Corless CL, Fletcher JA, Mcgreevey L, Haley A, Griffith D, et al. KIT Mutations Are Common in Testicular Seminomas. Am J Pathol. 2004.

27. Shen H, Shih J, Hollern DP, Wang L, Bowlby R, Tickoo SK, et al. Integrated Molecular Characterization of Testicular Germ Cell Tumors. Cell Rep. Elsevier; 2018;23:3392–

406.

(24)

23

28. Hacioglu BM, Kodaz H, Erdogan B, Cinkaya A, Tastekin E, Hacibekiroglu I, et al.

K-RAS and N-RAS mutations in testicular germ cell tumors. Bosn J basic Med Sci.

Association of Basic Medical Sciences of Federation of Bosnia and Herzegovina;

2017;17:159–63.

29. Feldman DR, Iyer G, Van Alstine L, Patil S, Al-Ahmadie H, Reuter VE, et al. Presence of Somatic Mutations within PIK3CA, AKT, RAS, and FGFR3 but not BRAF in Cisplatin-Resistant Germ Cell Tumors. Clin Cancer Res.2014;20:3712–20.

30. Einhorn LH. Treatment of testicular cancer: a new and improved model. J Clin Oncol. 1990;8:1777–81.

31. Peng H-Q, Hogg D, Malkin D, Bailey D, Gallic BL, Bulbul M, et al. Mutations of the p53 Gene Do Not Occur in Testis Cancer1. CANCER Res. 1993.

32. Vousden KH, Ryan KM. p53 and metabolism. Nat Rev Cancer. 2009;9:691– 700.

33. Eastman A. Characterization of the adducts produced in DNA by cis- diamminedichloroplatinum(II) and cis-dichloro(ethylenediamine)platinum(II).

Biochemistry. American Chemical Society; 1983;22:3927–33.

34. Yang D, Wang AH. Structural studies of interactions between anticancer platinum drugs and DNA. Prog Biophys Mol Biol. 1996;66:81–111.

35. Jordan P, Carmo-Fonseca M. Review Molecular mechanisms involved in cisplatin cytotoxicity. C Cell Mol Life Sci. 2000.

36. Sancar J. Repair of Cisplatin-DNA Adducts by the Mammalian Excision Nuclease †.

Proc Natl Acad Sci USA. 1994.

37. Masters JRW, Köberle B. Curing metastatic cancer: lessons from testicular germ- cell tumours. Nat Rev Cancer. Nature Publishing Group; 2003;3:517–25.

38. Köberle B, Masters JR, Hartley JA, Wood RD. Defective repair of cisplatin- induced DNA damage caused by reduced XPA protein in testicular germ cell tumours. Curr Biol. 1999;9:273–6.

39. Frankenberg-Schwager M, Kirchermeier D, Greif G, Baer K, Becker M, Frankenberg D.

Cisplatin-mediated DNA double-strand breaks in replicating but not in quiescent cells of the yeast Saccharomyces cerevisiae. Toxicology. Elsevier; 2005;212:175–84.

40. Cheng Q, Chen J. Mechanism of p53 stabilization by ATM after DNA damage. Cell Cycle. Taylor & Francis; 2010;9:472–8.

41. Kastenhuber ER, Lowe SW. Putting p53 in Context. Cell. Elsevier; 2017;170:1062–78.

42. Sark MW, Timmer-Bosscha H, Meijer C, Uges DR, Sluiter WJ, Peters WH, et al.

Cellular basis for differential sensitivity to cisplatin in human germ cell tumour and colon carcinoma cell lines. Br J Cancer. Nature Publishing Group; 1995;71:684–90.

43. Köberle B, Grimaldi KA, Sunters A, Hartley JA, Kelland LR, Masters JR. DNA repair capacity and cisplatin sensitivity of human testis tumour cells. Int J cancer.

1997;70:551–5.

44. Spierings DCJ, de Vries EGE, Stel AJ, te Rietstap N, Vellenga E, de Jong S. Low p21Waf1/Cip1 protein level sensitizes testicular germ cell tumor cells to Fas- mediated apoptosis. Oncogene. 2004;23:4862–72.

45. Koster R, Timmer-Bosscha H, Bischoff R, Gietema JA, De Jong S. Disruption of the

MDM2–p53 interaction strongly potentiates p53-dependent apoptosis in cisplatin-

(25)

resistant human testicular carcinoma cells via the Fas/FasL pathway. Cell Death Dis. 2011;2.

46. Gutekunst M, Oren M, Weilbacher A, Dengler MA, Markwardt C, Thomale J, et al. p53 Hypersensitivity Is the Predominant Mechanism of the Unique Responsiveness of Testicular Germ Cell Tumor (TGCT) Cells to Cisplatin. Gartel AL, editor. PLoS One. Public Library of Science; 2011;6:e19198.

47. Spierings DC, de Vries EG, Vellenga E, de Jong S. The attractive Achilles heel of germ cell tumours: an inherent sensitivity to apoptosis-inducing stimuli. J Pathol.

John Wiley & Sons, Ltd; 2003;200:137–Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR. The BCL-2 Family Reunion. Mol Cell. Cell Press; 2010;37:299–310.

48. Slee EA, Adrain C, Martin SJ. Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J Biol Chem.

American Society for Biochemistry and Molecular Biology; 2001;276:7320–6.

49. Zhu J, Dou Z, Sammons MA, Levine AJ, Berger SL. Lysine methylation represses p53 activity in teratocarcinoma cancer cells. Proc Natl Acad Sci U S

50. A. National Academy of Sciences; 2016;113:9822–7.

51. Koster R, di Pietro A, Timmer-Bosscha H, Gibcus JH, van den Berg A, Suurmeijer AJ, et al. Cytoplasmic p21 expression levels determine cisplatin resistance in human testicular cancer. J Clin Invest. 2010;120:3594–605.

52. Jiang M, Wei Q, Wang J, Du Q, Yu J, Zhang L, et al. Regulation of PUMA-α by p53 in cisplatin-induced renal cell apoptosis. Oncogene. Nature Publishing Group;

2006;25:4056–66.

53. Bagrodia A, Lee BH, Lee W, Cha EK, Sfakianos JP, Iyer G, et al. Genetic Determinants of Cisplatin Resistance in Patients With Advanced Germ Cell Tumors. J Clin Oncol.

American Society of Clinical Oncology; 2016;34:4000–7.

54. Kersemaekers A-MF, Mayer F, Molier M, van Weeren PC, Oosterhuis JW, Bokemeyer C, et al. Role of P53 and MDM2 in treatment response of human germ cell tumors.

J Clin Oncol. American Society of Clinical Oncology; 2002;20:1551–61.

55. Juliachs M, Muñoz C, Moutinho CA, Vidal A, Condom E, Esteller M, et al. The PDGFRß- AKT pathway contributes to CDDP-acquired resistance in testicular germ cell tumors.

Clin Cancer Res. American Association for Cancer Research; 2014;20:658–67.

56. Juliachs M, Vidal A, Del Muro XG, Piulats JM, Condom E, Casanovas O, et al.

Effectivity of pazopanib treatment in orthotopic models of human testicular germ cell tumors. BMC Cancer. 2013;13:382.

57. Zhao C-B, Bao J-M, Lu Y-J, Zhao T, Zhou X-H, Zheng D-Y, et al. Co- expression of RAGE and HMGB1 is associated with cancer progression and poor patient outcome of prostate cancer. Am J Cancer Res. 2014;4:369–77.

58. Zhang L, Han J, Wu H, Liang X, Zhang J, Li J, et al. The association of HMGB1 expression with clinicopathological significance and prognosis in hepatocellular carcinoma: A meta-analysis and literature review. PLoS One. Public Library of Science; 2014.

59. Yang GL, Zhang LH, Bo JJ, Hou XJ, Chen HG, Cao M, et al. Increased expression of HMGB1 is associated with poor prognosis in human bladder cancer. J Surg Oncol.

2012;106:57–61.

(26)

25

60. Xu Y, Chen Z, Zhang G, Xi Y, Sun R, Chai F, et al. HMGB1 overexpression correlates

with poor prognosis in early-stage squamous cervical cancer. Tumor Biol. Springer Netherlands; 2015;36:9039–47.

61. Wu D, Ding Y, Wang S, Zhang Q, Liu L. Increased expression of high mobility group box I (HMGBI) is associated with progression and poor prognosis in human nasopharyngeal carcinoma. J Pathol. 2008;216:167–75.

62. Ferry K V., Hamilton TC, Johnson SW. Increased nucleotide excision repair in cisplatin- resistant ovarian cancer cells: Role of ERCC1-XPF. Biochem Pharmacol. 2000;60:1305–13.

63. Cavallo F, Graziani G, Antinozzi C, Feldman DR, Houldsworth J, Bosl GJ, et al. Reduced Proficiency in Homologous Recombination Underlies the High Sensitivity of Embryonal Carcinoma Testicular Germ Cell Tumors to Cisplatin and Poly (ADP-Ribose) Polymerase Inhibition. Lichten M, editor. PLoS One. Public Library of Science; 2012;7:e51563.

64. Mego M, Svetlovska D, Miskovska V, Obertova J, Palacka P, Rajec J, et al. Phase II study of everolimus in refractory testicular germ cell tumors. Urol Oncol Semin Orig Investig. 2016;34:122.e17-122.e22.

65. Einhorn LH, Brames MJ, Heinrich MC, Corless CL, Madani A. Phase II Study of Imatinib Mesylate in Chemotherapy Refractory Germ Cell Tumors Expressing KIT.

Am J Clin Oncol •. 2006;29.

66. Subbiah V, Meric-Bernstam F, Mills GB, Shaw KRM, Bailey AM, Rao P, et al. Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent

®

) a VEGFR2/PDGFRß/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. J Hematol Oncol. BioMed Central; 2014;7:52.

67. Adra N, Einhorn LH, Althouse SK, Ammakkanavar NR, Musapatika D, Albany C, et al. Phase II trial of pembrolizumab in patients with platinum refractory germ- cell tumors: a Hoosier Cancer Research Network Study GU14-206. Ann Oncol.

2018;29:209–14.

68. Usanova S, Piée-Staffa A, Sied U, Thomale J, Schneider A, Kaina B, et al. Cisplatin sensitivity of testis tumour cells is due to deficiency in interstrand- crosslink repair and low ERCC1-XPF expression. Mol Cancer. BioMed Central; 2010;9:248.

69. Farmer H, McCabe N, Lord CJ, Tutt ANJ, Johnson DA, Richardson TB, et al.

Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy.

Nature. Nature Publishing Group; 2005;434:917–21.

70. Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al.

Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. Nature Publishing Group; 2005;434:913–7.

71. McCabe N, Turner NC, Lord CJ, Kluzek K, Białkowska A, Swift S, et al. Deficiency in the Repair of DNA Damage by Homologous Recombination and Sensitivity to Poly(ADP-Ribose) Polymerase Inhibition. Cancer Res. American Association for Cancer Research; 2006;66:8109–15.

72. Mego M, Cierna Z, Svetlovska D, Macak D, Machalekova K, Miskovska V, et al. PARP expression in germ cell tumours. J Clin Pathol. BMJ Publishing Group;

2013;66:607–12.

(27)

73. Murray MJ, Huddart RA, Coleman N. The present and future of serum diagnostic tests for testicular germ cell tumours. Nat Rev Urol. 2016;13:715– 25.

74. Gilligan TD, Hayes DF, Seidenfeld J, Temin S. ASCO Clinical Practice Guideline on Uses of Serum Tumor Markers in Adult Males With Germ Cell Tumors. J Oncol Pract. American Society of Clinical Oncology; 2010;6:199– 202.

75. de Haas EC, di Pietro A, Simpson KL, Meijer C, Suurmeijer AJH, Lancashire LJ, et al.

Clinical evaluation of M30 and M65 ELISA cell death assays as circulating biomarkers in a drug-sensitive tumor, testicular cancer. Neoplasia. 2008;10:1041–8.

76. Kawakami T, Okamoto K, Ogawa O, Okada Y. XIST unmethylated DNA fragments in male-derived plasma as a tumour marker for testicular cancer. Lancet. 2004;363:40–2.

77. Voorhoeve PM, le Sage C, Schrier M, Gillis AJM, Stoop H, Nagel R, et al. A Genetic Screen Implicates miRNA-372 and miRNA-373 As Oncogenes in Testicular Germ Cell Tumors. Cell. 2006;124:1169–81.

78. Chen L, Heikkinen L, Emily Knott K, Liang Y, Wong G. Evolutionary conservation and function of the human embryonic stem cell specific miR- 302/367 cluster.

Comp Biochem Physiol Part D Genomics Proteomics. Elsevier; 2015;16:83–98.

79. van Agthoven T, Looijenga LHJ. Accurate primary germ cell cancer diagnosis using serum based microRNA detection (ampTSmiR test). Oncotarget. 2017;8:58037–49.

80. Murray MJ, Bell E, Raby KL, Rijlaarsdam MA, Gillis AJM, Looijenga LHJ, et al. A pipeline to quantify serum and cerebrospinal fluid microRNAs for diagnosis and detection of relapse in paediatric malignant germ-cell tumours. Br J Cancer. 2016;114:151–62.

81. Dieckmann K-P, Radtke A, Spiekermann M, Balks T, Matthies C, Becker P, et al.

Serum Levels of MicroRNA miR-371a-3p: A Sensitive and Specific New Biomarker for Germ Cell Tumours. Eur Urol. 2017;71:213–20.

82. Gillis A, Stoop H, Hersmus R, Oosterhuis J, Sun Y, Chen C, et al. High- throughput microRNAome analysis in human germ cell tumours. J Pathol. 2007;213:319–28.

83. Palmer RD, Murray MJ, Saini HK, van Dongen S, Abreu-Goodger C, Muralidhar B, et al.

Malignant Germ Cell Tumors Display Common MicroRNA Profiles Resulting in Global Changes in Expression of Messenger RNA Targets. Cancer Res. 2010;70:2911–23.

84. Leão R, van Agthoven T, Figueiredo A, Jewett MAS, Fadaak K, Sweet J, et al. Serum miRNA Predicts Viable Disease after Chemotherapy in Patients with Testicular Nonseminoma Germ Cell Tumor. J Urol. 2018;

85. Mego M, Agthoven T, Gronesova P, Chovanec M, Miskovska V, Mardiak J, et al.

Clinical utility of plasma miR-371a-3p in germ cell tumors. J Cell Mol Med. John Wiley & Sons, Ltd (10.1111); 2018;jcmm.14013.

86. van Agthoven T, Eijkenboom WMH, Looijenga LHJ. microRNA-371a-3p as informative biomarker for the follow-up of testicular germ cell cancer patients. Cell Oncol. 2017;40:379–88.

87. Rahman NA, Huhtaniemi IT. Testicular cell lines. Mol Cell Endocrinol. Elsevier;

2004;228:53–65.

88. Andrews PW, Bronson DL, Benham F, Strickland S, Knowles BB. A comparative study

of eight cell lines derived from human testicular teratocarcinoma. Int J Cancer. John

Wiley & Sons, Ltd; 1980;26:269–80.

(28)

27

89. Harstrick A, Casper J, Guba R, Wilke H, Poliwoda H, Schmoll HJ. Comparison of

the antitumor activity of cisplatin, carboplatin, and iproplatin against established human testicular cancer cell lines in vivo and in vitro. Cancer. 1989;63:1079–83.

90. Albany C, Hever-Jardine MP, von Herrmann KM, Yim CY, Tam J, Warzecha JM, et al. Refractory testicular germ cell tumors are highly sensitive to the second generation DNA methylation inhibitor guadecitabine. Oncotarget. Impact Journals; 2017;8:2949–59.

91. Nakagawa H, Ueda T, Ito S, Shiraishi T, Taniguchi H, Kayukawa N, et al. Androgen suppresses testicular cancer cell growth &lt;i&gt;in vitro&lt;/i&gt; and &lt;i&gt;in vivo&lt;/i&gt; Oncotarget. Impact Journals; 2016;7:35224–32.

92. Rossi M, Colecchia D, Ilardi G, Acunzo M, Nigita G, Sasdelli F, et al. MAPK15 upregulation promotes cell proliferation and prevents DNA damage in male germ cell tumors. Oncotarget. Impact Journals; 2016;7:20981–98.

93. Gan Y, Wang Y, Tan Z, Zhou J, Kitazawa R, Jiang X, et al. TDRG1 regulates chemosensitivity of seminoma TCam-2 cells to cisplatin via PI3K/Akt/mTOR signaling pathway and mitochondria-mediated apoptotic pathway. Cancer Biol Ther. Taylor & Francis; 2016;17:741–50.

94. Aide N, Poulain L, Briand M, Dutoit S, Allouche S, Labiche A, et al. Early evaluation of the effects of chemotherapy with longitudinal FDG small-animal PET in human testicular cancer xenografts: early flare response does not reflect refractory disease. Eur J Nucl Med Mol Imaging. Springer-Verlag; 2009;36:396–405.

95. Douglas ML, Richardson MM, Nicol DL. Testicular germ cell tumors exhibit evidence of hormone dependence. Int J Cancer. John Wiley & Sons, Ltd; 2006;118:98–102.

96. Shirakawa T, Gotoh A, Zhang Z, Kao C, Chung LW., Gardner TA. Development of human chorionic gonadotropin subunit-beta promoter-based toxic gene therapy for testicular cancer. Urology. Elsevier; 2004;63:613–8.

97. Abraham D, Abri S, Hofmann M, Höltl W, Aharinejad S. Low Dose Carboplatin Combined With Angiostatic Agents Prevents Metastasis in Human Testicular Germ Cell Tumor Xenografts. J Urol. 2003;170:1388–93.

98. Dunn TA, Grünwald V, Bokemeyer C, Casper J. Pre-clinical activity of taxol in non- seminomatous germ cell tumor cell lines and nude mouse xenografts. Invest New Drugs. 1997;15:91–8.

99. Cassidy JW, Caldas C, Bruna A. Maintaining Tumor Heterogeneity in Patient- Derived Tumor Xenografts. 2015;

100. Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, et al. High- throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. Nature Publishing Group; 2015;21:1318–25.

101. Pompili L, Porru M, Caruso C, Biroccio A, Leonetti C. Patient-derived xenografts:

a relevant preclinical model for drug development. J Exp Clin Cancer Res. BioMed Central; 2016;35:189.

102. Ricci F, Bizzaro F, Cesca M, Guffanti F, Ganzinelli M, Decio A, et al. Patient- derived ovarian tumor xenografts recapitulate human clinicopathology and genetic alterations.

Cancer Res. American Association for Cancer Research; 2014;74:6980–90.

(29)

103. Hidalgo M, Amant F, Biankin A V, Budinská E, Byrne AT, Caldas C, et al. Patient- derived xenograft models: an emerging platform for translational cancer research.

Cancer Discov. American Association for Cancer Research; 2014;4:998–1013.

104. Berger DP, Fiebig HH, Winterhalter BR, Wallbrecher E, Henss H. Preclinical phase II study of ifosfamide in human tumour xenografts in vivo. Cancer Chemother Pharmacol. 1990;26 Suppl:S7-11.

105. Byrne AT, Alférez DG, Amant F, Annibali D, Arribas J, Biankin A V., et al. Interrogating open issues in cancer precision medicine with patient-derived xenografts. Nat Rev Cancer. 2017;17:254–68.

106. Burger AM, Fiebig H-H. SCREENING USING ANIMAL SYSTEMS. Anticancer Drug Dev. 2002. page 285–99.

107. Piulats JM, Vidal A, García-Rodríguez FJ, Muñoz C, Nadal M, Moutinho C, et al.

Orthoxenografts of Testicular Germ Cell Tumors Demonstrate Genomic Changes Associated with Cisplatin Resistance and Identify PDMP as a Resensitizing Agent.

Clin Cancer Res. American Association for Cancer Research; 2018;24:3755–66.

108. Castillo-Avila W, Piulats JM, Garcia del Muro X, Vidal A, Condom E, Casanovas O, et al. Sunitinib Inhibits Tumor Growth and Synergizes with Cisplatin in Orthotopic Models of Cisplatin-Sensitive and Cisplatin-Resistant Human Testicular Germ Cell Tumors. Clin Cancer Res. 2009;15:3384–95.

109. Baert Y, De Kock J, Alves-Lopes JP, Söder O, Stukenborg J-B, Goossens E. Primary Human Testicular Cells Self-Organize into Organoids with Testicular Properties.

Stem cell reports. Elsevier; 2017;8:30–8.

110. Alves-Lopes JP, Stukenborg J-B. Testicular organoids: a new model to study the testicular microenvironment in vitro? Hum Reprod Update. 2018;24:176– 91.

111. Jørgensen A, Young J, Nielsen JE, Joensen UN, Toft BG, Rajpert-De Meyts E, et al.

Hanging drop cultures of human testis and testis cancer samples: a model used to investigate activin treatment effects in a preserved niche. Br J Cancer. Nature Publishing Group; 2014;110:2604–14.

112. Pendergraft SS, Sadri-Ardekani H, Atala A, Bishop CE. Three-dimensional

testicular organoid: a novel tool for the study of human spermatogenesis and

gonadotoxicity in vitro†. Biol Reprod. Oxford University Press; 2017;96:720– 32.

(30)

Chapter 2

MiR-371a-3p, miR-373-3p and miR-367-3p as serum biomarkers in metastatic testicular germ cell cancers before, during and after chemotherapy

Ximena Rosas Plaza, Ton van Agthoven, Coby Meijer, Marcel A. T. M. van Vugt, Steven de Jong, Jourik A. Gietema and Leendert H. J. Looijenga.

Cells, 2019, 8, 1221

(31)

Background: LDH (lactate dehydrogenase), AFP (alpha-fetoprotein) and ß-HCG (human chorionic gonadotropin) are used in diagnosis, and follow-up of Testicular Germ Cell Cancer (TGCC) patients. Our aim was to investigate the association between levels of miR-371a-3p, miR-373-3p and miR-367-3p and clinical features in metastatic TGCC.

Methods: Relative levels of miR-371a-3p, miR-373-3p and miR-367-3p were evaluated in serum of metastatic TGCC patients. A prospectively included and a retrospectively selected cohort were studied (total patient number=109). Blood samples were drawn at start of chemotherapy and during follow-up. Serum microRNA (miR) levels were determined using the ampTSmiR test.

Results: At start of chemotherapy, miR-371a-3p, miR-373-3p and miR-367-3p levels were positively correlated to LDH. Median level of these miRs was higher in patients who developed a relapse after complete biochemical remission (n=34) than in those who had complete durable remission (n=60). Higher levels of miR-367-3p were found in patients with refractory disease (n=15) compared to those who had complete response. miR levels decreased during the first week of chemotherapy in patients with complete response and stayed below threshold after one year of treatment.

Conclusion: High miR levels at start of chemotherapy are associated with worse clinical outcome and can assist in early diagnosing of relapses.

Keywords: testicular germ cell cancer, microRNA, serum biomarkers, ß-HCG, AFP, LDH.

Abstract

Referenties

GERELATEERDE DOCUMENTEN

Nu de oplossing van het geschil afhangt van de uitleg van de Richtlijn, stelde de Deense rechter in deze zaak de volgende prejudiciële vraag aan het Hof van Justitie:

Novel preclinical models, therapies and biomarkers for testicular cancer Rosas Plaza,

One report has compared cisplatin sensitive and resistant TC cell lines, showing that cisplatin sensitivity correlated with the level of proficiency of TC cells to repair

Before start of chemotherapy, patients who developed a relapse after a complete biochemical response had higher miR-371a-3p, miR-373-3p and miR-367-3p median levels than

Sunitinib Inhibits Tumor Growth and Synergizes with Cisplatin in Orthotopic Models of Cisplatin-Sensitive and Cisplatin-Resistant Human Testicular Germ Cell Tumors. ErbBs

In the present study, we therefore evaluated the possibility of using TC PDX models to study AFP and ß-HCG serum levels in relation to PDX growth and response to cisplatin

In Chapter 2, we investigated three microRNAs: miR-371a-3p, miR-373-3p and miR-367-3p as putative tumor biomarkers during and after treatment with cisplatin combination

We analyzed the genomic profiles of carbapenem- nonsusceptible Pseudomonas aeruginosa (CNPA) strains collected from two intensive care units (ICUs) in the national referral hospital