• No results found

University of Groningen Circulating biomarkers in classical Hodgkin lymphoma Plattel, Wouter Johannes

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Circulating biomarkers in classical Hodgkin lymphoma Plattel, Wouter Johannes"

Copied!
173
0
0

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

Hele tekst

(1)

Circulating biomarkers in classical Hodgkin lymphoma

Plattel, Wouter Johannes

DOI:

10.33612/diss.97631424

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:

2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Plattel, W. J. (2019). Circulating biomarkers in classical Hodgkin lymphoma. Rijksuniversiteit Groningen.

https://doi.org/10.33612/diss.97631424

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)
(3)

978-94-034-1959-6 (electronic version)

The studies described in this thesis were financially supported by the Dutch Cancer Society (Grant no. RUG 2010-4860) and the Dutch Organization of Scientific Research (NWO ZonMW Grant no. 92003569).

© Copyright by Wouter Plattel, 2019. All rights reserved. Cover design and layout: © evelienjagtman.com

(4)

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de

rector magnificus prof. dr. C. Wijmenga en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op woensdag 16 oktober 2019 om 16.15 uur

door

Wouter Johannes Plattel

geboren op 26 januari 1985 te

(5)

Prof. dr. J.H.M. van den Berg Prof. dr. J.C. Kluin-Nelemans Copromotores Dr. G.W. van Imhoff Dr. A. Visser Beoordelingscommissie Prof. dr. M.J. Kersten Prof. dr. D. de Jong Prof. dr. G. de Haan

(6)

Chapter 1 Introduction and review of circulating biomarkers in Hodgkin Lymphoma

In preparation

7

PART 1

Chapter 2 Plasma Thymus and Activation-Regulated Chemokine as an early response marker in classical Hodgkin lymphoma

Haematologica. 2012 Mar;97(3):410-5

31

Chapter 3 Biomarkers for evaluation of treatment response in classical Hodgkin lymphoma: comparison of sGalectin-1, sCD163 and sCD30 with TARC

Br J Haematol. 2016 Dec;175(5):868-875

53

Chapter 4 Interim TARC versus interim 18F-FDG-PET in classical Hodgkin

lymphoma response evaluation Submitted

71

PART 2

Chapter 5 The role of microRNAs in Hodgkin lymphoma In MicroRNAs in Medicine, C. H. Lawrie (Ed.). 2013

83 Chapter 6 Pre-analytical, analytical and post-analytical challenges in

circulating miRNA studies

99 Chapter 7 Circulating miRNAs in serum of patients with classical Hodgkin

Lymphoma

117

SUMMARY AND DISCUSSION

Chapter 8 Summary, general discussion and future perspectives 133

Chapter 9 Nederlandse samenvatting 149

APPENDICES Dankwoord List of publications Curriculum Vitae 161 169 171

(7)
(8)

Modified from: Circulating biomarkers in classical Hodgkin Lymphoma

Wouter J. Plattel, Anke van den Berg, Lydia Visser, Hanneke C. Kluin-Nelemans, Gustaaf W. van Imhoff, and Arjan Diepstra

In preparation

CHAPTER 1

Introduction and review of circulating

(9)
(10)

1

Introduction and scope of the thesis

Epidemiology and pathobiology

Hodgkin lymphoma (HL), formally known as Hodgkin’s disease, is a malignant disease named after Thomas Hodgkin who first described this entity in 1832.1 HL has an age-standardized

incidence rate of 3 per 100.000 per year and accounts for 15-25% of all lymphoma patients in the western world.2 Remarkably, about half of the patients are adolescents or young adults.

HL is characterized by the presence of large malignant cells, which contain two nuclei or nuclear lobes. Together with its mononuclear variants, these cells are called Hodgkin Reed-Sternberg (HRS) cells and are typical for classical HL (cHL).2 Classical HL accounts for approximately 95%

of all HL cases and the remaining 5% consist of the non-classical subtype nodular lymphocyte predominant Hodgkin lymphoma (NLPHL). This non-classical variant is regarded as a distinct entity based on its distinct histopathological features, clinical presentation and clinical behavior. In this thesis we will focus on the cHL subtype only.

Classical HL has a unique and fascinating pathobiology. First, the malignant cells are greatly outnumbered by a massive inflammatory component which is unique not only among lymphomas but also among solid cancers (Figure 1). Second, HRS are of B-cell origin, but they have lost their entire B-cell phenotype including surface immunoglobulin expression.3

Last, about 30% of cHL cases contain the Epstein Barr Virus (EBV), which is involved in the pathogenesis of these cases.2

Figure 1. Histopathological pictures of cHL. The scarcity of the HRS cells with its surrounding

microenvironment (A) and an example of the interaction between HRS cells and its microenvironment as demonstrated by HRS cell specific TARC staining (blue) and its receptor CCR4 (brown) on lymphocytes (B). H = Hodgkin cells; RS = Reed Sternberg cells; T = T-cells; H = histiocytes; E = eosinophils; P = plasma cells.

(11)

Normal immature B-cells undergo somatic recombination in germinal centers to transform into plasma cells or memory B-cells containing a B-cell receptor (BCR) with high affinity to a certain antigen. B-cells that lack a high affinity BCR or express a BCR with high affinity to self-antigens are either clonally deleted, undergo receptor editing or end in an anergic state. The lack of a BCR or a functional BCR as characteristic for HRS cells would normally lead to apoptosis. However, HRS cells have found a way to escape from apoptosis during the germinal center reaction. Constitutive activation of the canonical and non-canonical nuclear factor kappa-B (NFkB) pathways are a main survival mechanism by upregulating anti-apoptotic genes like c-FLIP and XIAP.4 The canonical NFkB signaling can be activated via tumor necrosis receptor (TNFR) family

members like CD30, CD40, RANK and TNFR1 that are expressed on the cell membrane of HRS cells. The non-canonical pathway can be activated by both autocrine and exocrine signaling by HRS cells and surrounding immune cells, respectively.5 Latent membrane protein 1 (LMP-1) is

an EBV- derived protein that mimics CD40 receptor stimulation, whereas LMP2 mimics BCR signaling. Together these signals can also induce activation of the NFkB pathway in EBV+ cHL.6,7

The extensive micro-environment of cHL consists of varying numbers of plasma cells, granulocytes, histiocytes, eosinophils, mast cells and macrophages, but the main cell type present in this reactive infiltrate is the T-cell (Figure 2).8 Remarkably, this reactive infiltrate is

unable to induce an effective immune response against the HRS cells. Moreover, the HRS cells need the reactive infiltrate to survive. This protective micro-environment is for a main part self-orchestrated by the HRS cells by various mechanisms. Constitutive activation of the NFkB and Janus kinase–signal transducer and activator of transcription (JAK-STAT) signaling pathways are central players in this aberrant interaction. Genetic alterations including copy number gains and activating mutations have been found in components of both the NFkB and JAK-STAT pathways in 53% and 90% of cases, respectively.9 Moreover, HRS cells directly suppress a Th1 response

by producing IL-10 and TGF-β and by expressing cell surface molecules including Fas ligand and galectin-1 that are associated with decreased amounts of effector T-cells surrounding the HRS cells. On the other hand, HRS cells promote and attract T-helper 2 cells (Th2) and regulatory T-cells (Treg) type immune cells by secreting CCL17 (also known as thymus and activation regulated chemokine, TARC), CCL5, CCL22 chemokines and IL-7, the latter of which enables differentiation of naïve CD4+ T-cells into Treg cells.10 The T-cells in the reactive infiltrate mainly

consist of CD4+ T-cells and they produce Th2 stimulatory cytokines like 4 and 5 and IL-10. The inhibition of an effective Th1 response is illustrated by the scarcity of CD8+ cytotoxic T-lymphocytes (CTLs) and natural killer (NK) cells in the direct vicinity of the tumor cells. In addition, the CD4+ T-cells directly surrounding the HRS cells seem to be in an anergic state as a result of several mechanisms including lack of expression of the T-cell activation marker CD26 and variable expression of immune check-point molecules CTLA-4 and PD-1 in these T-cells, as well as increased PD-1 ligand expression by HRS cells and the abundant production of IL-10 and TGF-β.11

(12)

1

The composition of the reactive infiltrate and stroma is the basis of a further histopathological separation of cHL into four subtypes: nodular sclerosis cHL, mixed cellularity cHL, lymphocyte rich cHL and lymphocyte depleted cHL. Nodular sclerosis cHL is the most common subtype and represents about 60% of the cHL cases in the western world, whereas mixed cellularity is more common in developing countries and is frequently associated with presence of EBV (about 70%).2 Although these subtypes have shown differences in gene-expression, cytokine

production and also clinical behavior, this distinction does not result in different treatment approaches. Galec tin-1 TGFβ , IL-10 CCL28 CCL28, IL -6 TNFα CCL17, C CL22 NK Th2 Th2 T Treg T

Growth Factors Immune Escape

CD40L CD30L,CD40L IL-3 IL-3 IL3R CD30 CD40 RTK IL9R IL6R IL7R IL15R IL13R IL-13 CCL17, C CL22 CD30L,CD40L Eosinophil Mast cell

IL-9, IL-15, IL-6, IL-13, IL-7

IL-7 HLA-G HLA TCR LAG3 PD-1 IL-10 IDO PD-L1, PD-L2 EBI3 IL-12 IL-7 Eosinophil Shaping the environment

Plasma Th1 Exhausted T Macrophage T Treg HRS cell JAK STAT FasL fibroblast fibroblast

Figure 2. Cellular composition of cHL and crosstalk between HRS cells and its micro-environment. HRS

cell = Hodgkin Reed-Sternberg cell; T = T-cell; NK = Natural killer cell; Treg = regulatory T-cell; Th1 = T-helper 1 cell; plasma = plasma cell.

Staging, treatment and prognostic factors

After the introduction of multi-agent chemotherapy regimens in the 1960s and improvements in radiotherapy, cHL can now be cured in more than 80% of all newly diagnosed patients (stage I-IV).12-14 Current treatment algorithms are based on a combination of clinical and laboratory

factors. The Ann Arbor staging system separates early stage from advanced stage patients based on number and location of the involved lymph nodes and presence of extranodal disease. Several large study groups further classify early stage patients for allocation of treatment into

(13)

early favorable and early unfavorable risk groups using slightly different criteria (Table 1). For advanced stage disease, a more widely accepted scoring system, the International Prognostic Score (IPS), has been developed based on six factors with independent significance for freedom of progression of disease in multivariate analysis.15 However, the clinically utility of the IPS to

allocate treatment is limited.16

Table 1. Clinical prognostic risk classification for early stage cHL

EORTC# favorable GHSG$ favorable NCCN& favorable

Age < 50 Not included Not included

ESR and B-symptoms < 50 mm/hr no B-symptoms or < 30 mm/hr + B-symptoms < 50 mm/hr no B-symptoms or < 30 mmh/hr + B-symptoms <50 mm/hr no B-symptoms

Large mediastinal mass Absent Absent (advanced stage if present)

Absent

No. involved sites <4 <3 <4

Extranodal disease Not included Absent Not included

#European Organisation of Research and Treatment of Cancer; $German Hodgkin Study Group; &National

Comprehensive Cancer Network

With current treatment regimens over 90% of early stage patients can be cured with the combination of 2-4 courses of chemotherapy (Adriamycin, Bleomycin, Vinblastine and Dacarbazine (ABVD)) followed by involved node radiotherapy.17,18 The drawbacks of this highly

curative combined modality treatment are the long term cardiovascular toxicities and secondary malignancies that can for an important part be attributed to former extensive radiotherapy fields and anthracycline-based therapies.19,20 Advanced stage patients can be treated with 6 cycles of

ABVD chemotherapy or 4-6 cycles of dose intensified chemotherapy (Bleomycin, Etoposide, Adriamycin, Cyclophosphamide, Oncovin, Procarbazine, Prednisone (escalated BEACOPP)) followed by radiotherapy on residual active disease sites.21 With these treatments about 75 and

90% of advanced stage patients can be cured, respectively. Especially for patients treated with regimens such as escBEACOPP, the risk of death due to short and long-term treatment-related toxicities is approaching the risk of dying of cHL itself.22 EscBEACOPP is also strongly associated

with infertility in both men and women.23

The main challenge in cHL management is therefore to minimize both overtreatment and undertreatment by better tailoring treatment to the prognosis of the individual patient. Recognition of patients at high risk of treatment failure before or early during treatment is therefore of utmost importance.

(14)

1

FDG-PET guided treatment

Functional imaging with Fluorodeoxyglucose Positron Emission Tomography (FDG-PET)-scans combined with computer tomography (FDG-PET/CT) has become the standard tool for staging and response assessment in cHL. FDG-PET/CT is more sensitive than conventional imaging and led to significant upstaging in cHL.24,25 Staging in the era of FDG-PET/CT has also made

routine bone marrow biopsy unnecessary because of its higher sensitivity for bone marrow involvement.26,27

Response evaluation during treatment using FDG-PET scanning (interim FDG-PET (iPET)) enables evaluation of early metabolic changes rather than the morphologic changes visualized by CT occurring later during and after completion of treatment. Several studies using iPET after two or three cycles of ABVD have shown that early metabolic changes are predictive of the final treatment response and PFS.28-30 Based on these studies, which were mainly performed among

advanced stage patients, several cooperative groups have incorporated iPET imaging in their trials to reduce treatment exposure in responding patients to prevent overtreatment and/or to intensify treatment in case of non-responsiveness. Very recently this strategy of treatment escalation or de-escalation based on iPET has shown to improve progression free survival or safely decrease treatment exposure in early- as well as advanced stage patients.17,18,31,32

Nevertheless, iPET does not perfectly predict final outcome. In patients with a negative iPET after two cycles of ABVD, failure rates were observed ranging from 10% up to 25% in early- and advanced stage patients, respectively.17,33 On the other hand, 25% of advanced stage patients

treated with ABVD who are iPET positive become FDG-PET negative after completion of ABVD treatment and experience durable remissions.34 Also, advanced stage patients treated

with escBEACOPP show an excellent prognosis after completion of treatment despite iPET positivity.32 Although current efforts are being made to improve iPET adapted strategies by

investigating different timing and interpretation methods, there is still a need for better early stratification of high and low risk patients.

Biomarkers

A plethora of biomarkers derived from patient tissue or from peripheral blood have shown to correlate with prognosis and survival in cHL. These can be grouped into two categories, one being general factors related to inflammation / the abundant reactive infiltrate; the second being tumor cell-specific biomarkers. In addition, the biomarkers can be grouped based on being tissue-based or blood-tissue-based markers. At this moment, only the blood-tissue-based factors such as elevated erythrocyte sedimentation rate, anemia, leukocytosis, lymphocytopenia and hypoalbuminia are included in clinical prognostic models. Excellent reviews on prognostic factors in cHL have been published during the last years, mainly focusing on tissue markers.35-38 A disadvantage of almost

all published tissue markers is the lack of reproducibility due to differences in tissue fixation, staining, often difficult scoring methods and impractical cut-offs.

(15)

Tumor cell specific biomarkers that correlate with treatment response have the potential to be used in the same way as FDG-PET imaging with additional benefits of low-costs, being more patient-friendly, and potentially more specific. Blood-based biomarkers have the advantage over tissue markers that cell numbers, protein levels or DNA copy numbers can usually be quantified with higher reproducibility using standardized assays. From a clinical view, blood-based biomarkers have the great advantage that they can be sampled not only before but also during and after treatment. During the last 40 years an extensive number of studies have been published on blood-based biomarkers either focusing on prognosis (progression or disease free survival) or on evaluating treatment response. In the remainder of this chapter we summarize both prognostic blood-based biomarkers identified at diagnosis and blood-based biomarkers that can be used as treatment response biomarkers.

Circulating biomarkers

Blood cell counts and systemic inflammatory markers

Leukocytes: leukocytosis, lymphocytopenia, monocytosis

Leukocytosis defined as leukocyte count >15x103/mm3 is a common feature in cHL patients

with an incidence of about 10-20% at diagnosis (see Table 2).15,39 It is included in the IPS as

an independent prognostic factor, but it is not consistently observed as such in recent, albeit smaller studies.40,41 Leukocytosis was shown to have prognostic value when other prognostic

factors including leukocyte subsets were not taken into account.42,43

Lymphocytopenia was already identified in 1971 as an adverse prognostic factor in cHL.44 This

finding was later confirmed in multiple studies. The Hasenclever study incorporated it into the IPS.15,45

Lymphocytopenia, either absolute (<0,6x103/mm3) or relative (<8% of total WBC) is observed in

about one fifth of the patients at diagnosis. It is hypothesized that lymphocytopenia reflects impaired host immune homeostasis or even depletion of lymphocytes by infiltration into the tumor.39,46

Monocytosis was more recently recognized as a negative prognostic factor in cHL.39 It has been

hypothesized that monocyte counts might reflect number of tumor-associated macrophages, which originate from monocytes.39 A monocyte count of >0,9 x103/mm3 was found to correlate

with adverse progression-free, disease-free and overall survival. The lymphocyte/monocyte

ratio further increased the prognostic impact of monocyte counts. A ratio of <1.1 correlated with

very poor prognosis both in early and advanced stage patients and was independent of IPS. The independent prognostic value of this ratio was confirmed for overall survival in a non-Western cohort, for both progression free and overall survival in a very large study involving patients from Italy and Israel and in multivariate analysis with interim FDG-PET result.41,47,48 In conclusion, there

(16)

1

Hemoglobin level and iron metabolism

Low Hemoglobin level or anemia has been identified as a negative prognostic factor in many

malignancies, including HL.15,45 A hemoglobin level below 10.5 g/dl in both males and females is

included in the IPS as a negative prognostic factor, which is present in about 40% of cHL patients at diagnosis.15 Anemia as seen in cHL patients mirrors anemia of patients with chronic diseases

and correlates with interleukin (IL)-6 and hepcidin levels. IL-6 can induce hepcidin, which in turn inhibits release of iron stores from the mononuclear phagocyte system and from the intestine and results in elevated levels of ferritin. Both IL-6 and ferritin correlate with prognosis as has been shown decades ago for ferritin and has been confirmed in multiple recent studies.40,49-53

Clinically, ferritin is mainly used as a reflection of actual iron stores, but ferritin also acts as an acute phase protein. Indeed, ferritin levels highly correlate with inflammatory parameters like erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP).53 In cHL patients, anemia

probably reflects an active immune state and impaired iron metabolism. It is currently not known whether anemia, serum IL-6 or ferritin is the most potent prognostic factor or whether they have independent prognostic value since a direct comparison of those markers is lacking.

Other circulating cells

In a search for peripheral blood biomarkers that reflect tumor micro-environment or host immune response, there are two smaller studies that have found new prognostic markers. The first study showed that higher levels of circulating CD34+ myeloid-derived suppressor

cells (immature MDSCs) correlate with adverse prognosis.43 Multivariate analysis in this iPET

treatment adapted cohort, revealed that elevation of CD34+ MDSCs was the only remaining significant parameter for survival and outperformed iPET. Another study found the CD4/CD19

ratio to be a negative prognostic factor independent of iPET and stage.42

Systemic inflammatory markers

Systemic inflammatory markers are detected in about half of the patients with cHL and correlate with tumor burden. These markers mainly reflect the abundant micro-environment characteristic of cHL. The most well-known non-specific inflammatory biomarker is the ESR. Elevated ESR (>30 or >50 mm) is present in about half of the patients and is included as one of the risk factors for unfavorable early stage disease by major study groups, i.e., the European Organisation for Research and Treatment of Cancer (EORTC), the German Hodgkin Study Group (GHSG) and the National Comprehensive Cancer Network (NCCN) (see Table 1). The initial finding of a correlation of ESR with clinical outcome in both untreated and treated patients published more than four decades ago was confirmed in subsequent papers.54,55 In a more recent paper the ESR remained an

important factor in currently applied early stage risk classification.56 However, the prognostic value

of ESR is only modest or even absent in multivariate analysis with other prognostic biomarkers.57,58

This can probably be explained by the fact that the ESR is influenced by many other prognostic factors like erythrocyte count, fibrinogen levels, presence of acute phase proteins or increased

(17)

gamma globulins. The studies investigating these individual factors as well as C-reactive protein are generally small or show correlation only in univariate analysis.59-61

Low albumin is a negative prognostic factor in cHL. Albumin is the most abundant plasma protein

and accounts for about 15% of the protein producing capacity of the liver. Albumin levels inversely correlate with inflammatory status and inflammatory proteins.62 This can be explained by inhibition

of albumin synthesis by molecules associated with inflammatory states like tumor necrosis factor (TNF), IL-11 and IL-6 that shift the protein production of the liver to production of acute phase proteins.63 Low albumin was the only negative prognostic factor in a large cohort of cHL patients

treated from 1970 until 1980 in a model with ESR, hemoglobulin, alkaline phosphatase and lactate dehydrogenase.57 This international study confirmed the prognostic impact of low albumin levels

with a cut off at 4.0 g/dl and was included in the IPS as a negative prognostic factor.15

Table 2. Blood cell counts and systemic inflammatory markers with adverse prognostic value. % at diagnosis HR for PFS RR (from ref 15)

cut off level Status#

Hemoglobulin 15-30 1.35 <10.5 g/dl Established15, 45

Leukocytosis 15-20 1.41 >15x109/mm3 Established 15,39

Lymphopenia (L) 15-25 1.38 <600/mm3 or <8% Established15,45

Monocytosis (M) 35 1.8 >700-900 cells/ul Potential39

L/M ratio ~35 2.9 - 3.8 <1.1 / <2.1 High potential 41,47,48

ESR 35-50 1.5 -1.6* 30-50 mm/1sthr Established 54-58

Albumin 40-60 1.49 4 g/dl Established 15,57

Ferritin 30-40 4.0 350 ug/l Potential40,49-53

HR = hazard ratio; PFS = progression free survival; RR = relative risk

* Odds ratio for treatment failure within 2.5 years after diagnosis in early stage patients only

# Definitions used for status: established = biomarkers included in currently applied prognostic models;

potential = one or limited number of papers indicating prognostic value; high potential = multiple reports indicating high prognostic impact

Tumor and microenvironment-derived markers

Cytokines, chemokines and soluble receptors

Multiple studies addressed the prognostic value of soluble levels of cytokines, chemokines and their receptors. IL-6 is a pro-inflammatory cytokine produced by HRS cells and by cells from the microenvironment including lymphocytes, macrophages and fibroblasts (see Figure 2). IL-6 levels are elevated in about 20-30% of the patients depending on the applied cut-off.52,62 The

prognostic value of IL-6 was confirmed in a study including 30 cytokines, whereas in another study with a case-control design, IL-6 did not have independent prognostic value, showing that thus far the value of IL-6 is inconclusive.52,64

(18)

1

IL-10 is produced by HRS cells and regulatory T-cells and inhibits a Th1 antitumor response.

IL-10 levels are elevated in about 40-60% of patients and correlate positively with tumor cell EBV status.65 The prognostic value of IL-10 is controversial with multiple studies showing a

prognostic value of IL-10 independent from clinical parameters, whereas other studies did not find any independent prognostic value.52,59,62,64,66-69 An explanation for these findings might be

that in the latter studies multiple cytokines have been included.

The soluble form of the alpha chain of the receptor of IL-2 (sIL-2R, sCD25) is another immunomodulatory molecule that correlates with prognosis in cHL.52,70 sCD25 is derived

by proteolytic cleavage of CD25 by Matrix metallopeptidase 9 (MMP9) produced by tumor-associated macrophages. Levels of sCD25 correlate with the presence of tumor-tumor-associated macrophages in the microenvironment of NHL.71 sCD25 is a marker for activated B- and

T-cells and is considered to be a marker of regulatory T-cells. Upon activation by IL-2, CD25 induces FOXP3 expression in CD4+ T-cells thereby promoting a regulatory T-cell phenotype in the tumor microenvironment.72 A negative prognostic value of high levels of sCD25 was first

reported in 1987.70 These findings were confirmed in several subsequent studies,73-76 but could

not in be confirmed in other studies.59,77-79

Soluble IL-1 receptor antagonist (sIL-1RA) is produced by various types of immune cells and is

elevated in about 35% of cHL patients.80 IL-1RA competes with type I and type II IL-1R and can

partly neutralize the inflammatory effects of IL-1 secreted by HRS cells. Interestingly, secretion of sIL1RA is dependent on IL-6, IL-13 and IL-10 and the prognostic value of these cytokines might be interdependent.62

Soluble CD30 (sCD30) is considered to be derived from HRS cells and thus presents a tumor

cell-derived marker. CD30 belongs to the TNFR family and can be actively shed from the membrane resulting in sCD30. High sCD30 serum levels were first described as a negative prognostic marker in 1990.81 This has been confirmed in multiple studies and was shown to be

independent of clinical features. or other soluble molecules.59,62,68,73,74,78,82-85

A few attempts have been made to include a set of these prognostic markers in a new prognostic model. Casasnovas et al. found that the combination of IL-6, sIL-1RA and sCD30 in a three cytokine/soluble receptor model had better prognostic value than the IPS.62

Unfortunately, no multivariate analysis with individual factors of the IPS was shown. Marri et al. performed a multi-cytokine study in which a two-cytokine model containing IL-6 and sCD25 had the best prognostic value. In this study, sCD30 did not maintain prognostic value in multivariate analysis.52 Unfortunately, sCD25 was not analyzed in the study by

(19)

In the last decade, Thymus and Activation Regulated Chemokine (TARC) or CCL17 has gained interest as a prognostic marker and treatment response marker. TARC is produced by HRS cells at very high levels and is responsible for the attraction of CCR4 positive cells, which are mainly regulatory T-cells and Th2 cells (Figures 1 and 2).86 Thus, TARC is for an important

part responsible for the immunosuppressive direct micro-environment of the HRS cells. In line with the high production, extremely high plasma or serum levels have been observed in cHL patients.87,88 TARC levels are elevated in >85% of patients at diagnosis and high levels correlate

with negative prognostic features and higher disease stage.89 In line with the fact that stage

of disease itself is a potent prognostic factor in cHL, high TARC levels at diagnosis correlated with adverse prognosis.90 This was confirmed in a large study in which a prognostic model that

included both TARC and clinical features showed strong prognostic value.58 However, in a

smaller study no correlation with prognosis was found for TARC.52 Since elevation of TARC is

strikingly high compared to healthy controls, TARC has the ideal features to serve as a biomarker for treatment response.

There are several other protein biomarkers like soluble Galectin-1, sCD163, M-CSF, sCD8, sICAM-1, CA125, B-lymphocyte stimulator and polyclonal free light chains that might serve as prognostic biomarkers since they correlate with adverse clinical or disease characteristics. Future studies are needed to elucidate whether these markers have real prognostic value independent of established markers within current treatment era. Soluble Galectin-1 (sGal-1) and soluble CD163 (sCD163) are of special interest since sGal-1 is thought to be derived from tumor cells itself and sCD163 is the soluble form of the M2 macrophage marker CD163. Tumor-associated macrophages are associated with adverse prognosis in tissue of patients with cHL. 91,92

Table 3. Cytokines, chemokines and soluble receptors related to prognosis % patients

with marker

cut off level Adverse prognosis for high/low levels

Status#

IL6 20-30 30 pg/ml High Controversial52,64

IL-10 40-60 10 pg/ml High Controversial52,59,62,64,66-69

sIL2R 60-75 ~1000 U/ml High Controversial59,73-79

sIL1RA 35 668 pg/ml High Potential62

sCD30 40-80 20-200 U/ml High High potential59,62,68,73,74,78,82-85

TARC 70-90 500-10,000 pg/ml High Controversial52,58,59

# Definitions used for status: controversial = papers showing contradictory results; potential = one or

limited number of papers indicating prognostic value; high potential = multiple reports indicating high prognostic impact

(20)

1

Markers of cell or membrane turnover

Beta-2-microglobulin (B2M) is a component of the HLA class I complex. Serum levels are

thought to reflect membrane turnover and have been correlated with adverse prognosis in several smaller studies with different treatment regimens.93-99 Several reports showed an

independent negative prognostic value for high serum lactate dehydrogenase (LDH).45,97,100-102

However, in the International Consortium on Prognostic factors study, LDH levels did not have a significant prognostic value.15 Also elevated alkaline phosphatase levels have been linked

with adverse prognosis in cHL, but did not remain significant in multivariate analyses in the Hasenclever study.15,40

Molecular markers

High levels of circulating Epstein Barr Virus DNA (EBV-DNA) were found to closely correlate with tumor EBV-status as detected by EBER in situ hybridization (EBER-ISH) on tissue samples.103 Patients with EBV positive disease have detectable EBV DNA levels in their

circulation in about 65% of cases.104-108 An adverse prognostic value of the presence of plasma

EBV-DNA independent of IPS was shown in a relatively large cohort of cHL patients.103 This is in

line with the finding that EBV positivity in tumor tissue has an adverse outcome among elderly cHL patients.109,110 The subtype of cHL in elderly patients is more often of the mixed cellularity

subtype and the HRS cells more often harbor EBV. Unfortunately, the study by Kanakry et al. did not specify the prognostic value of plasma EBV in relation with age. Jones et al. showed that patients with high pre-treatment EBV-DNA copy numbers showed a decrease in copy numbers rapidly after start of treatment, showing its potential to serve as treatment response biomarker among patients with EBV positive disease.92

Circulating microRNAs (miRNAs) have also received increasing attention as non-invasive

biomarkers for diagnosis or prognosis in cancer. MiRNAs are small non-coding RNAs of about 20-25 nucleotides that regulate expression of protein coding genes at the post-transcriptional level. In cHL, two miRNAs were found to correlate with treatment response.111 However,

differences in miRNAs disappeared when levels were normalized to a cellular small RNA (RNA U6), indicating possible blood cell origin of the miRNAs. A more recent paper isolated extracellular vesicles from plasma and identified several miRNAs that were enriched in plasma of cHL patients compared to healthy controls.112 A first analysis showed correlation between a

decrease of these vesicle-derived miRNAs and a metabolic response upon treatment.

Studies investigating genomic aberrations have always been challenging because of the scarcity of tumor cells in cHL tissue (see Figure 1). Analysis of circulating tumor DNA (ctDNA) levels showed to be feasible in cHL and allows non-invasive detection of tumor cell specific genomic aberrations including mutations and copy number alterations (CNAs). Genomic aberrations in cell-free DNA were initially detected in a pregnant woman, who was later diagnosed with

(21)

early stage cHL, indicating the presence of ctDNA.113 Subsequent testing on nine additional

cHL patients revealed the presence of 2p and 9p gains, two commonly observed aberrations in tumor cells of cHL, in ctDNA of seven and five of the nine cHL patients, respectively. A larger scale study that was carried out more recently showed that mutations detected in ctDNA mirror the genetics of cHL tumor cells.114 Due to the small sample size and heterogeneity of the

patient cohort no conclusions could be made on any predictive or prognostic value. Sequential samples collected from a small number of patients showed that persistence of high ctDNA levels correlated with treatment failure or relapse. This indicates its potential to serve as a biomarker for treatment response.

In conclusion, there are multiple circulating biomarkers that have potential to be applied at diagnosis and during treatment to optimize decision making upfront and during treatment. Large multicenter studies are needed to define the optimal set of prognostic biomarkers that can be applied at diagnosis given current treatment regimens and the known prognostic value of iFDG-PET imaging. Research on treatment response biomarkers in cHL is more limited, but several biomarkers have high potential to improve on or to be applied next to FDG-PET imaging.

Scope of the thesis

This thesis aims to address the relevance of selected circulating biomarkers. In the first part of this thesis we study the application of TARC as a biomarker for treatment response and compare TARC with galectin-1, sCD163 and sCD30, and interim FDG-PET imaging. In Chapter 2 we studied the correlation of TARC with clinical response in a cohort of 63 patients from the University Medical Center Groningen (UMCG). In Chapter 3 we added three other potential treatment response biomarkers to compare with TARC, i.e. galactin-1, sCD163 and sCD30 in a larger cohort of 103 patients. In Chapter 4 we compared TARC during treatment (interim TARC) with interim FDG-PET imaging for their ability to predict for modified progression-free survival.

In the second part of this thesis we focus on the applicability of circulating microRNAs as biomarkers in cHL. In Chapter 5 we summarize the current knowledge on mircroRNAs in cHL. In

Chapter 6 we address the pre-analytical, analytical, and post-analytical challenges in circulating

microRNA studies. Finally, in Chapter 7 we investigated the expression profiles of circulating microRNAs in serum of a cHL patient cohort from Vancouver.

In Chapter 8, all results are summarized and discussed, followed by a general perspective on the future application of biomarkers in cHL.

(22)

1

References

1. Hodgkin. On some Morbid Appearances of the Absorbent Glands and Spleen. Med Chir Trans. 1832;17:68-114.

2. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375-2390. 3. Kuppers R, Rajewsky K, Zhao M, et al. Hodgkin

disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development. Proc Natl Acad Sci U S A. 1994;91(23):10962-10966.

4. Kuppers R, Engert A, Hansmann ML. Hodgkin lymphoma. J Clin Invest. 2012;122(10):3439-3447.

5. Liu WR and Shipp MA. Signaling pathways and immune evasion mechanisms in classical Hodgkin lymphoma. Blood. 2017;130(21):2265-2270.

6. Mathas S, Hartmann S, Kuppers R. Hodgkin lymphoma: Pathology and biology. Semin Hematol. 2016;53(3):139-147.

7. Tan GW, Visser L, Tan LP, van den Berg A, Diepstra A. The Microenvironment in Epstein-Barr Virus-Associated Malignancies. P a t h o g e n s . 2 0 1 8 ; 7 ( 2 ) : 1 0 . 3 3 9 0 / pathogens7020040.

8. Liu Y, Sattarzadeh A, Diepstra A, Visser L, van den Berg A. The microenvironment in classical Hodgkin lymphoma: an actively shaped and essential tumor component. Semin Cancer Biol. 2014;24:15-22.

9. Tiacci E, Ladewig E, Schiavoni G, et al. Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma. Blood. 2018;131(22):2454-2465.

10. Maggio E, van den Berg A, Diepstra A, Kluiver J, Visser L, Poppema S. Chemokines, cytokines and their receptors in Hodgkin’s lymphoma cell lines and tissues. Ann Oncol. 2002;13 Suppl 1:52-6.

11. Vardhana S and Younes A. The immune microenvironment in Hodgkin lymphoma: T cells, B cells, and immune checkpoints. Haematologica. 2016;101(7):794-802. 12. Boll B and Gorgen H. The treatment of older

Hodgkin lymphoma patients. Br J Haematol. 2019;184(1):82-92.

13. Depaus J, Delcourt A, Andre M. Therapeutic recommendations for early stage Hodgkin lymphomas. Br J Haematol. 2019;184(1):9-16.

14. Spinner MA and Advani RH. Risk-adapted therapy for advanced-stage Hodgkin lymphoma. Hematology Am Soc Hematol Educ Program. 2018;2018(1):200-206. 15. Hasenclever D and Diehl V. A prognostic

score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease. N Engl J Med. 1998;%19;339(21):1506-1514.

16. Franklin J, Paulus U, Lieberz D, Breuer K, Tesch H, Diehl V. Is the international prognostic score for advanced stage Hodgkin’s disease applicable to early stage patients? German Hodgkin Lymphoma Study Group. Ann Oncol. 2000;11(5):617-623.

17. Andre MPE, Girinsky T, Federico M, et al. Early Positron Emission Tomography Response-Adapted Treatment in Stage I and II Hodgkin Lymphoma: Final Results of the Randomized EORTC/LYSA/FIL H10 Trial. J Clin Oncol. 2017;35(16):1786-1794.

18. Radford J, Illidge T, Counsell N, et al. Results of a trial of PET-directed therapy for early-stage Hodgkin’s lymphoma. N Engl J Med. 2015;372(17):1598-1607.

19. Maraldo MV, Giusti F, Vogelius IR, et al. Cardiovascular disease after treatment for Hodgkin’s lymphoma: an analysis of nine collaborative EORTC-LYSA trials. Lancet Haematol. 2015;2(11):e492-502.

20. Schaapveld M, Aleman BM, van Eggermond AM, et al. Second Cancer Risk Up to 40 Years

(23)

after Treatment for Hodgkin’s Lymphoma. N Engl J Med. 2015;373(26):2499-2511. 21. Skoetz N, Will A, Monsef I, Brillant C, Engert

A, von Tresckow B. Comparison of first-line chemotherapy including escalated BEACOPP versus chemotherapy including ABVD for people with early unfavourable or advanced stage Hodgkin lymphoma. Cochrane Database Syst Rev. 2017;5:CD007941.

22. Viviani S, Zinzani PL, Rambaldi A, et al. ABVD versus BEACOPP for Hodgkin’s lymphoma when high-dose salvage is planned. N Engl J Med. 2011;365(3):203-212.

23. Behringer K, Mueller H, Goergen H, et al. Gonadal function and fertility in survivors after Hodgkin lymphoma treatment within the German Hodgkin Study Group HD13 to HD15 trials. J Clin Oncol. 2013;31(2):231-239.

24. Weihrauch MR, Re D, Bischoff S, et al. Whole-body positron emission tomography using 18F-fluorodeoxyglucose for initial staging of patients with Hodgkin’s disease. Ann Hematol. 2002;81(1):20-25.

25. El-Galaly TC, Hutchings M, Mylam KJ, et al. Impact of 18F-fluorodeoxyglucose positron emission tomography/computed tomography staging in newly diagnosed classical Hodgkin lymphoma: fewer cases with stage I disease and more with skeletal involvement. Leuk Lymphoma. 2014;55(10):2349-2355. 26. El-Galaly TC, d’Amore F, Mylam KJ, et al.

Routine bone marrow biopsy has little or no therapeutic consequence for positron emission tomography/computed tomography-staged treatment-naive patients with Hodgkin lymphoma. J Clin Oncol. 2012;30(36):4508-4514.

27. Voltin CA, Goergen H, Baues C, et al. Value of bone marrow biopsy in Hodgkin lymphoma patients staged by FDG PET: results from the German Hodgkin Study Group trials HD16, HD17, and HD18. Ann Oncol. 2018;29(9):1926-1931.

28. Hutchings M, Loft A, Hansen M, et al. FDG-PET after two cycles of chemotherapy

predicts treatment failure and progression-free survival in Hodgkin lymphoma. Blood. 2006;107(1):52-59.

29. Gallamini A, Hutchings M, Rigacci L, et al. Early interim 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography is prognostically superior to international prognostic score in advanced-stage Hodgkin’s lymphoma: a report from a joint Italian-Danish study. J Clin Oncol. 2007;20;25(24):3746-3752.

30. Gallamini A, Barrington SF, Biggi A, et al. The predictive role of interim positron emission tomography for Hodgkin lymphoma treatment outcome is confirmed using the interpretation criteria of the Deauville five-point scale. Haematologica. 2014;99(6):1107-1113. 31. Johnson P, Federico M, Kirkwood A, et al.

Adapted Treatment Guided by Interim PET-CT Scan in Advanced Hodgkin’s Lymphoma. N Engl J Med. 2016;374(25):2419-2429. 32. Borchmann P, Goergen H, Kobe C, et al.

PET-guided treatment in patients with advanced-stage Hodgkin’s lymphoma (HD18): final results of an open-label, international, randomised phase 3 trial by the German Hodgkin Study Group. Lancet. 2018;390(10114):2790-2802.

33. Stephens DM, Li H, Schoder H, et al. Long-Term Follow-up of SWOG S0816: Response-Adapted Therapy for Stage III/IV Hodgkin Lymphoma Demonstrates Limitations of PET-Adapted Approach. Blood. 2018;132(Suppl 1):929-929 (abstract).

34. Biggi A, Gallamini A, Chauvie S, et al. International validation study for interim PET in ABVD-treated, advanced-stage hodgkin lymphoma: interpretation criteria and concordance rate among reviewers. J Nucl Med. 2013;54(5):683-690.

35. Steidl C, Connors JM, Gascoyne RD. Molecular pathogenesis of Hodgkin’s lymphoma: increasing evidence of the importance of the microenvironment. J Clin Oncol. 2011;29(14):1812-1826.

(24)

1

in Hodgkin lymphoma: better risk profiling and novel treatments. Clin Cancer Res. 2013;19(11):2797-2803.

37. Venkataraman G, Mirza MK, Eichenauer DA, Diehl V. Current status of prognostication in classical Hodgkin lymphoma. Br J Haematol. 2014;165(3):287-299.

38. Connors JM. Risk assessment in the management of newly diagnosed classical Hodgkin lymphoma. Blood. 2015;125(11):1693-1702.

39. Porrata LF, Ristow K, Colgan JP, et al. Peripheral blood lymphocyte/monocyte ratio at diagnosis and survival in classical Hodgkin’s lymphoma. Haematologica. 2012;97(2):262-269.

40. deAndres-Galiana EJ, Fernandez-Martinez JL, Luaces O, et al. On the prediction of Hodgkin lymphoma treatment response. Clin Transl Oncol. 2015;17(8):612-619.

41. Koh YW, Kang HJ, Park C, et al. Prognostic significance of the ratio of absolute neutrophil count to absolute lymphocyte count in classic Hodgkin lymphoma. Am J Clin Pathol. 2012;138(6):846-854.

42. Gaudio F, Perrone T, Mestice A, et al. Peripheral blood CD4/CD19 cell ratio is an independent prognostic factor in classical Hodgkin lymphoma. Leuk Lymphoma. 2014;55(7):1596-1601.

43. Romano A, Parrinello NL, Vetro C, et al. Circulating myeloid-derived suppressor cells correlate with clinical outcome in Hodgkin Lymphoma patients treated up-front with a risk-adapted strategy. Br J Haematol. 2015;168(5):689-700.

44. Swan HT and Knowelden J. Prognosis in Hodgkin’s disease related to the lymphocyte count. Br J Haematol. 1971;21(3):343-349. 45. Ferme C, Bastion Y, Brice P, et al. Prognosis

of patients with advanced Hodgkin’s disease: evaluation of four prognostic models using 344 patients included in the Group d’Etudes des Lymphomes de l’Adulte Study. Cancer. 1997;80(6):1124-1133.

46. Franzke A, Koenecke C, Geffers R, et al.

Classical Hodgkin’s lymphoma: molecular evidence for specific alterations in circulating T lymphocytes. Tumour Biol. 2006;27(6):329-333.

47. Tadmor T, Bari A, Marcheselli L, et al. Absolute Monocyte Count and Lymphocyte-Monocyte Ratio Predict Outcome in Nodular Sclerosis Hodgkin Lymphoma: Evaluation Based on Data From 1450 Patients. Mayo Clin Proc. 2015;90(6):756-764.

48. Romano A, Parrinello NL, Vetro C, et al. Prognostic meaning of neutrophil to lymphocyte ratio (NLR) and lymphocyte to monocyte ration (LMR) in newly diagnosed Hodgkin lymphoma patients treated upfront with a PET-2 based strategy. Ann Hematol. 2018;97(6):1009-1018.

49. Jacobs A, Slater A, Whittaker JA, Canellos G, Wiernik PH. Serum ferritin concentration in untreated Hodgkin’s disease. Br J Cancer. 1976;34(2):162-166.

50. Hann HW, Lange B, Stahlhut MW, McGlynn KA. Prognostic importance of serum transferrin and ferritin in childhood Hodgkin’s disease. Cancer. 1990;66(2):313-316. 51. Hohaus S, Massini G, Giachelia M, et al.

Anemia in Hodgkin’s lymphoma: the role of interleukin-6 and hepcidin. J Clin Oncol. 2010;28(15):2538-2543.

52. Marri PR, Hodge LS, Maurer MJ, et al. Prognostic significance of pretreatment serum cytokines in classical Hodgkin lymphoma. Clin Cancer Res. 2013;19(24):6812-6819. 53. Fernandez-Alvarez R, Gonzalez-Rodriguez

AP, Gonzalez ME, et al. Serum ferritin as prognostic marker in classical Hodgkin lymphoma treated with ABVD-based therapy. Leuk Lymphoma. 2015;56(11):3096-3102. 54. Tubiana M, Attie E, Flamant R,

Gerard-Marchant R, Hayat M. Prognostic factors in 454 cases of Hodgkin’s disease. Cancer Res. 1971;31(11):1801-1810.

55. Gobbi PG, Cavalli C, Federico M, et al. Hodgkin’s disease prognosis: a directly predictive equation. Lancet. 1988;1(8587):675-679.

(25)

56. Klimm B, Goergen H, Fuchs M, et al. Impact of risk factors on outcomes in early-stage Hodgkin’s lymphoma: an analysis of international staging definitions. Ann Oncol. 2013;24(12):3070-3076.

57. Gobbi PG, Comelli M, Grignani GE, Pieresca C, Bertoloni D, Ascari E. Estimate of expected survival at diagnosis in Hodgkin’s disease: a means of weighting prognostic factors and a tool for treatment choice and clinical research. A report from the International Database on Hodgkin’s Disease (IDHD). Haematologica. 1994;79(3):241-255.

58. Sauer M, Plutschow A, Jachimowicz RD, et al. Baseline serum TARC levels predict therapy outcome in patients with Hodgkin lymphoma. Am J Hematol. 2013;88(2):113-115. 59. Axdorph U, Sjoberg J, Grimfors G, Landgren O,

Porwit-MacDonald A, Bjorkholm M. Biological markers may add to prediction of outcome achieved by the International Prognostic Score in Hodgkin’s disease. Ann Oncol. 2000;11(11):1405-1411.

60. Wieland A, Kerbl R, Berghold A, Schwinger W, Mann G, Urban C. C-reactive protein (CRP) as tumor marker in pediatric and adolescent patients with Hodgkin disease. Med Pediatr Oncol. 2003;41(1):21-25.

61. Stanca O, Ciobanu AM, Lupu AR, et al. Onset risk factors and treatment response features of refractory hodgkin lymphoma. Maedica (Buchar). 2013;8(4):343-346.

62. Casasnovas RO, Mounier N, Brice P, et al. Plasma cytokine and soluble receptor signature predicts outcome of patients with classical Hodgkin’s lymphoma: a study from the Groupe d’Etude des Lymphomes de l’Adulte. J Clin Oncol. 2007;25(13):1732-1740.

63. Moshage HJ, Janssen JA, Franssen JH, Hafkenscheid JC, Yap SH. Study of the molecular mechanism of decreased liver synthesis of albumin in inflammation. J Clin Invest. 1987;79(6):1635-1641.

64. Rautert R, Schinkothe T, Franklin J, et al. Elevated pretreatment interleukin-10 serum

level is an International Prognostic Score (IPS)-independent risk factor for early treatment failure in advanced stage Hodgkin lymphoma. Leuk Lymphoma. 2008;49(11):2091-2098. 65. Herling M, Rassidakis GZ, Medeiros LJ,

et al. Expression of Epstein-Barr virus latent membrane protein-1 in Hodgkin and Reed-Sternberg cells of classical Hodgkin’s lymphoma: associations with presenting features, serum interleukin 10 levels, and clinical outcome. Clin Cancer Res. 2003;9(6):2114-2120.

66. Bohlen H, Kessler M, Sextro M, Diehl V, Tesch H. Poor clinical outcome of patients with Hodgkin’s disease and elevated interleukin-10 serum levels. Clinical significance of interleukin-10 serum levels for Hodgkin’s disease. Ann Hematol. 2000;79(3):110-113. 67. Vassilakopoulos TP, Nadali G, Angelopoulou

MK, et al. Serum interleukin-10 levels are an independent prognostic factor for patients with Hodgkin’s lymphoma. Haematologica. 2001;86(3):274-281.

68. Visco C, Vassilakopoulos TP, Kliche KO, et al. Elevated serum levels of IL-10 are associated with inferior progression-free survival in patients with Hodgkin’s disease treated with radiotherapy. Leuk Lymphoma. 2004;45(10):2085-2092.

69. Viviani S, Notti P, Bonfante V, Verderio P, Valagussa P, Bonadonna G. Elevated pretreatment serum levels of Il-10 are associated with a poor prognosis in Hodgkin’s disease, the milan cancer institute experience. Med Oncol. 2000;17(1):59-63.

70. Pizzolo G, Chilosi M, Vinante F, et al. Soluble interleukin-2 receptors in the serum of patients with Hodgkin’s disease. Br J Cancer. 1987;55(4):427-428.

71. Sakai A and Yoshida N. The role of tumor-associated macrophages on serum soluble IL-2R levels in B-cell lymphomas. J Clin Exp Hematop. 2014;54(1):49-57.

72. Yang ZZ, Grote DM, Ziesmer SC, et al. Soluble IL-2Ralpha facilitates IL-2-mediated immune responses and predicts reduced survival

(26)

1

in follicular B-cell non-Hodgkin lymphoma. Blood. 2011;118(10):2809-2820.

73. Gause A, Pohl C, Tschiersch A, et al. Clinical significance of soluble CD30 antigen in the sera of patients with untreated Hodgkin’s disease. Blood. 1991;77(9):1983-1988. 74. Gause A, Jung W, Schmits R, et al. Soluble

CD8, CD25 and CD30 antigens as prognostic markers in patients with untreated Hodgkin’s lymphoma. Ann Oncol. 1992;3 Suppl 4:49-52. 75. Ambrosetti A, Nadali G, Vinante F, et al. Serum levels of soluble interleukin-2 receptor in Hodgkin disease. Relationship with clinical stage, tumor burden, and treatment outcome. Cancer. 1993;72(1):201-206.

76. Enblad G, Sundstrom C, Gronowitz S, Glimelius B. Serum levels of interleukin-2 receptor (CD 25) in patients with Hodgkin’s disease, with special reference to age and prognosis. Ann Oncol. 1995;6(1):65-70. 77. Gorschluter M, Bohlen H, Hasenclever D,

Diehl V, Tesch H. Serum cytokine levels correlate with clinical parameters in Hodgkin’s disease. Ann Oncol. 1995;6(5):477-482. 78. Nadali G, Vinante F, Chilosi M, Pizzolo G.

Soluble molecules as biological markers in Hodgkin’s disease. Leuk Lymphoma. 1997;26 Suppl 1:99-105.

79. Viviani S, Camerini E, Bonfante V, et al. Soluble interleukin-2 receptors (sIL-2R) in Hodgkin’s disease: outcome and clinical implications. Br J Cancer. 1998;77(6):992-997.

80. Gruss HJ, Dolken G, Brach MA, Mertelsmann R, Herrmann F. High concentrations of the interleukin-1 receptor antagonist in serum of patients with Hodgkin’s disease. Lancet. 1992;340(8825):968-72.

81. Pizzolo G, Vinante F, Chilosi M, et al. Serum levels of soluble CD30 molecule (Ki-1 antigen) in Hodgkin’s disease: relationship with disease activity and clinical stage. Br J Haematol. 1990;75(2):282-284.

82. Gause A, Jung W, Keymis S, et al. The clinical significance of cytokines and soluble forms of membrane-derived activation antigens in the serum of patients with Hodgkin’s disease.

Leuk Lymphoma. 1992;7(5-6):439-447. 83. Nadali G, Vinante F, Ambrosetti A, et al. Serum

levels of soluble CD30 are elevated in the majority of untreated patients with Hodgkin’s disease and correlate with clinical features and prognosis. J Clin Oncol. 1994;12(4):793-797.

84. Nadali G, Tavecchia L, Zanolin E, et al. Serum level of the soluble form of the CD30 molecule identifies patients with Hodgkin’s disease at high risk of unfavorable outcome. Blood. 1998;91(8):3011-3016.

85. Zanotti R, Trolese A, Ambrosetti A, et al. Serum levels of soluble CD30 improve International Prognostic Score in predicting the outcome of advanced Hodgkin’s lymphoma. Ann Oncol. 2002;13(12):1908-1914.

86. van den Berg A, Visser L, Poppema S. High expression of the CC chemokine TARC in Reed-Sternberg cells. A possible explanation for the characteristic T-cell infiltratein Hodgkin’s lymphoma. Am J Pathol. 1999;154(6):1685-1691.

87. Ma Y, Visser L, Roelofsen H, et al. Proteomics analysis of Hodgkin lymphoma: identification of new players involved in the cross-talk between HRS cells and infiltrating lymphocytes. Blood. 2008;111(4):2339-2346.

88. Niens M, Visser L, Nolte IM, et al. Serum chemokine levels in Hodgkin lymphoma patients: highly increased levels of CCL17 and CCL22. Br J Haematol. 2008;140(5):527-536.

89. Plattel WJ, van den Berg A, Visser L, et al. Plasma thymus and activation-regulated chemokine as an early response marker in classical Hodgkin’s lymphoma. Haematologica. 2012;97(3):410-415.

90. Weihrauch MR, Manzke O, Beyer M, et al. Elevated serum levels of CC thymus and activation-related chemokine (TARC) in primary Hodgkin’s disease: potential for a prognostic factor. Cancer Res. 2005;65(13):5516-5519.

(27)

Strandmann E, et al. Galectin-1 serum levels reflect tumor burden and adverse clinical features in classical Hodgkin lymphoma. Blood. 2013;121(17):3431-3433.

92. Jones K, Vari F, Keane C, et al. Serum CD163 and TARC as disease response biomarkers in classical Hodgkin lymphoma. Clin Cancer Res. 2013;19(3):731-742.

93. Dimopoulos MA, Cabanillas F, Lee JJ, et al. Prognostic role of serum beta 2-microglobulin in Hodgkin’s disease. J Clin Oncol. 1993;11(6):1108-1111.

94. Chronowski GM, Wilder RB, Tucker SL, et al. An elevated serum beta-2-microglobulin level is an adverse prognostic factor for overall survival in patients with early-stage Hodgkin disease. Cancer. 2002;95(12):2534-2538. 95. Vassilakopoulos TP, Nadali G, Angelopoulou

MK, et al. The prognostic significance of beta(2)-microglobulin in patients with Hodgkin’s lymphoma. Haematologica. 2002;87(7):701-8; discussion 708.

96. Albitar M, Vose JM, Johnson MM, et al. Clinical relevance of soluble HLA-I and beta2-microglobulin levels in non-Hodgkin’s lymphoma and Hodgkin’s disease. Leuk Res. 2007;31(2):139-145.

97. Itoh K, Kinoshita T, Watanabe T, et al. Prognostic analysis and a new risk model for Hodgkin lymphoma in Japan. Int J Hematol. 2010;91(3):446-455.

98. Nakajima Y, Tomita N, Watanabe R, et al. Prognostic significance of serum beta-2 microglobulin level in Hodgkin lymphoma treated with ABVD-based therapy. Med Oncol. 2014;31(9):185-014-0185-3. Epub 2014 Aug 21.

99. Wang Q, Qin Y, Zhou S, et al. Prognostic value of pretreatment serum beta-2 microglobulin level in advanced classical Hodgkin lymphoma treated in the modern era. Oncotarget. 2016;7(44):72219-72228.

100. Schilling RF, McKnight B, Crowley JJ. Prognostic value of serum lactic dehydrogenase level in Hodgkin’s disease. J Lab Clin Med. 1982;99(3):382-387.

101. Straus DJ, Gaynor JJ, Myers J, et al. Prognostic factors among 185 adults with newly diagnosed advanced Hodgkin’s disease treated with alternating potentially noncross-resistant chemotherapy and intermediate-dose radiation therapy. J Clin Oncol. 1990;8(7):1173-1186.

102. Garcia R, Hernandez JM, Caballero MD, et al. Serum lactate dehydrogenase level as a prognostic factor in Hodgkin’s disease. Br J Cancer. 1993;68(6):1227-1231.

103. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013;121(18):3547-3553.

104. Gallagher A, Armstrong AA, MacKenzie J, et al. Detection of Epstein-Barr virus (EBV) genomes in the serum of patients with EBV-associated Hodgkin’s disease. Int J Cancer. 1999;84(4):442-448.

105. Lei KI, Chan LY, Chan WY, Johnson PJ, Lo YM. Quantitative analysis of circulating cell-free Epstein-Barr virus (EBV) DNA levels in patients with EBV-associated lymphoid malignancies. Br J Haematol. 2000;111(1):239-246.

106. Gandhi MK, Lambley E, Burrows J, et al. Plasma Epstein-Barr virus (EBV) DNA is a biomarker for EBV-positive Hodgkin’s lymphoma. Clin Cancer Res. 2006;12(2):460-464.

107. Spacek M, Hubacek P, Markova J, et al. Plasma EBV-DNA monitoring in Epstein-Barr virus-positive Hodgkin lymphoma patients. APMIS. 2011;119(1):10-16.

108. Hohaus S, Santangelo R, Giachelia M, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17(9):2885-2892. 109. Jarrett RF, Stark GL, White J, et al. Impact of

tumor Epstein-Barr virus status on presenting features and outcome in age-defined subgroups of patients with classic Hodgkin lymphoma: a population-based study. Blood.

(28)

1

2005;106(7):2444-2451.

110. Diepstra A, van Imhoff GW, Karim-Kos HE, et al. HLA class II expression by Hodgkin Reed-Sternberg cells is an independent prognostic factor in classical Hodgkin’s lymphoma. J Clin Oncol. 2007;20;25(21):3101-3108. 111. Jones K, Nourse JP, Keane C, Bhatnagar A,

Gandhi MK. Plasma microRNA are disease response biomarkers in classical Hodgkin lymphoma. Clin Cancer Res. 2014;20(1):253-264.

112. van Eijndhoven MA, Zijlstra JM, Groenewegen NJ, et al. Plasma vesicle miRNAs for therapy response monitoring in Hodgkin lymphoma

patients. JCI Insight. 2016;1(19):e89631. 113. Vandenberghe P, Wlodarska I, Tousseyn T,

et al. Non-invasive detection of genomic imbalances in Hodgkin/Reed-Sternberg cells in early and advanced stage Hodgkin’s lymphoma by sequencing of circulating cell-free DNA: a technical proof-of-principle study. Lancet Haematol. 2015;2(2):e55-65. 114. Spina V, Bruscaggin A, Cuccaro A, et al.

Circulating tumor DNA reveals genetics, clonal evolution, and residual disease in classical Hodgkin lymphoma. Blood. 2018;131(22):2413-2425.

(29)
(30)
(31)
(32)

CHAPTER 2

Plasma Thymus and Activation-Regulated

Chemokine as an early response marker

in classical Hodgkin lymphoma

Wouter J. Plattel, Anke van den Berg, Lydia Visser, Anne-Marijn van der Graaf, Jan Pruim, Hans Vos, Bouke Hepkema, Arjan Diepstra, and Gustaaf W. van Imhoff

(33)

Abstract

INTRODUCTION:

Plasma Thymus and Activation-Regulated Chemokine (TARC) is a potential biomarker for classical Hodgkin lymphoma. To define the value of plasma TARC as a marker to monitor treatment response, we correlated serial plasma TARC levels with clinical response in newly diagnosed and relapsed classical Hodgkin lymphoma patients.

DESIGN AND METHODS:

Plasma was collected from 60 (39 early stage and 21 advanced stage) newly diagnosed classical Hodgkin lymphoma patients before, during and after treatment and from 12 relapsed patients before and after treatment. Plasma TARC levels were determined by enzyme-linked immunosorbent assay and were related to pre-treatment metabolic tumor volume as measured by quantification of 2-[18F]fluoro-2-deoxyglucose positron emission tomography images and to treatment response.

RESULTS:

Baseline plasma TARC levels correlated with stage of disease and bulky disease and more closely with metabolic tumor volume. Response to treatment was observed among 38/39 early stage and 19/21 advanced stage patients. Reduction in plasma TARC to normal range levels could be observed as early as after one cycle of chemotherapy in all responsive patients, while plasma TARC remained elevated during and after treatment in the three non-responsive patients. Plasma TARC was elevated in all 12 relapsed patients at time of relapse and remained elevated after salvage treatment in the four non-responsive patients only.

CONCLUSIONS:

Baseline plasma TARC levels correlate with classical Hodgkin lymphoma tumor burden and serial plasma TARC levels correlate with response to treatment in patients with classical Hodgkin lymphoma.

(34)

2

Introduction

In classical Hodgkin Lymphoma (cHL) the neoplastic Hodgkin Reed-Sternberg (HRS) cells are vastly outnumbered by cells in the surrounding reactive infiltrate. This infiltrate is of major importance for the proliferation and survival of HRS cells. Different chemokines and cytokines produced by HRS cells and cells in the reactive infiltrate are responsible for the formation and maintenance of this reactive environment.1-4

The CC chemokine ligand 17 (CCL17) or Thymus and Activation-Regulated Chemokine (TARC) is highly expressed by HRS cells in cHL, but not by the tumor cells of nodular lymphocyte predominant Hodgkin lymphoma or other B-cell derived non-Hodgkin lymphomas.5,6 TARC binds

specifically to the CC chemokine receptor 4 (CCR4). CCR4 is mainly expressed on regulatory T and Th2 cells that are both abundantly present in the reactive infiltrate of cHL.6-8 Approximately

90% of the cHL patients show positive TARC staining in HRS cells by immunohistochemistry (IHC) and about 85% have significantly elevated levels of TARC in their pre-treatment serum or plasma sample compared to healthy controls.9,10 Although patients with active atopic

diseases can also have elevated plasma TARC levels, this is only a modest elevation which is in a significantly lower range than the high plasma TARC levels observed in cHL.11 Pre-treatment

serum TARC levels correlate with stage of disease, erythrocyte sedimentation rate, leukocyte and lymphocyte counts in cHL.9,10 Niens et al.9 reported TARC levels within the normal range

after successful treatment in seven cHL patients and persistent elevated TARC in a single non-responsive patient. Weihrauch et al.10 reported diminished survival rates among patients with

higher TARC levels after treatment. However, nothing is known about TARC dynamics during treatment in relation to clinical treatment response.

We therefore prospectively collected serial plasma samples from newly diagnosed and relapsed cHL patients. The aim of the current study was to correlate plasma TARC levels with tumor burden at time of diagnosis and to correlate serial plasma TARC levels during and after treatment with cHL treatment response.

(35)

Design and Methods

Patient inclusion and treatment

Serial plasma samples were prospectively collected from all newly diagnosed and relapsed cHL patients that were treated in the University Medical Center Groningen (UMCG) from January 2006 until June 2011.

Inclusion criteria for both newly diagnosed and relapsed cHL patients were (1) receipt of standard treatment regimens, (2) availability of a plasma sample before start of treatment and one or more plasma samples during or after treatment as well as (3) confirmation of TARC expression in diagnostic tissue by immunohistochemistry or by elevated baseline plasma TARC if diagnostic tissue was not available. From 78 newly diagnosed patients treated in the UMCG, 60 were included, while 18 were excluded (1 because of patient refusal, 2 because of receiving palliative treatment, 9 because of lack of a pre-treatment plasma sample, 3 with negative tissue TARC staining and 3 with no available tissue and normal pre-treatment plasma TARC levels). From 17 relapsed patients, 12 patients eligible for DHAP salvage treatment followed by autologous stem cell transplantation were included, while 5 patients were excluded (4 because of receiving only palliative treatment and one because of lack of a plasma sample after treatment).

Permission for this study was obtained from the institutional review board of the UMCG and all participating patients and healthy controls signed informed consent. Routine staging of patients at diagnosis or at relapse included diagnostic Computer Tomography (CT) imaging, ‘whole body’ 2-[18F]fluoro-2-deoxyglucose positron emission tomography (FDG-PET) imaging, and bone marrow biopsy. Presence of bulky disease was defined as presence of a mediastinal mass greater than one third of the thoracic diameter on chest X-ray (on level Th5-Th6) and/or a nodal mass of more than 10 cm CT imaging. Response to treatment was evaluated according to the revised International Working Group response criteria.12 Evaluation included FDG-PET/CT scanning

which was interpreted according to the International Harmonization Project criteria (IHP).13

FDG-PET/CT scanning was performed using a Siemens Biograph PET/CT mCT 64 scanner in the vast majority of patients

Patients were treated according to clinical trial (European Organisation for Research and Treatment of Cancer (EORTC)) protocols. Table 1 shows the patient characteristics and data on chemo- and radiotherapy regimens. Briefly, standard treatment for stage I/II (early stage) patients consisted of 3-6 cycles of ABVD chemotherapy with or without 30-36 Gy involved node radiotherapy (IN-RT) according to the EORTC (20051) H10 trial.14 Standard treatment for stage

III/IV (advanced stage) patients consisted of 6-8 cycles of ABVD without radiotherapy or in case of participation in the EORTC 20012 trial, patients randomized between 8 cycles of ABVD and

Referenties

GERELATEERDE DOCUMENTEN

Circulating biomarkers in classical Hodgkin lymphoma Plattel, Wouter

level is an International Prognostic Score (IPS)- independent risk factor for early treatment failure in advanced stage Hodgkin lymphoma. Expression of Epstein-Barr virus latent

Reduction in plasma TARC to normal range levels could be observed as early as after one cycle of chemotherapy in all responsive patients, while plasma TARC remained elevated

Plasma sGal-1, sCD163 and sCD30 levels did not significantly correlate with disease stage or metabolic tumor volume (Figure 2), whereas TARC levels were significantly higher in

The other ten patients with discrepant results had a low iTARC and a positive iPET: seven out of ten became FDG-PET negative at end-treatment, remained TARC negative and did

MiRNA expression studies have been performed on HL cell lines, laser microdissected tumor cells and total tumor tissue sections.. Despite the use of different tissue specimens, a

Om snot te voorkomen worden momenteel toetsen ontwikkeld voor de verschillende veroor- zakers van snot, zodat van partijen snel vastge- steld kan worden of ze besmet zijn en of telers

De natuurlijke vijanden uit de oorsprongsgebieden van japanse vliegen, Japan en Noord-Amerika, kunnen niet zonder meer worden ingevoerd als klassieke biologische bestrijding.. Wel