You are on page 1of 15

Published Ahead of Print on April 11, 2011 as 10.1200/JCO.2010.32.8401 The latest version is at http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2010.32.

8401

JOURNAL OF CLINICAL ONCOLOGY

R E V I E W

A R T I C L E

Molecular Pathogenesis of Hodgkins Lymphoma: Increasing Evidence of the Importance of the Microenvironment
Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne
From the British Columbia Cancer Agency, University of British Columbia, Vancouver, British Columbia, Canada. Submitted September 19, 2010; accepted January 4, 2011; published online ahead of print at www.jco.org on April 11, 2011. Supported in part by the Michael Smith Foundation for Health Research, the Lymphoma Research Foundation, and a postdoctoral fellowship from the Cancer Research Society (Steven E. Drabin fund; C.S.); Project Grant No. 019001 from the Terry Fox Foundation New Frontiers Program (J.M.C., R.D.G.); and Grant No. 178536 from the Canadian Institutes for Health Research (R.D.G.). Authors disclosures of potential conicts of interest and author contributions are found at the end of this article. Corresponding author: Randy Gascoyne, MD, Department of Pathology and Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Ave, Room 5-113, Vancouver, BC V5Z 1L3, Canada; e-mail: rgascoyn@bccancer.bc.ca. 2011 by American Society of Clinical Oncology 0732-183X/11/2999-1/$20.00 DOI: 10.1200/JCO.2010.32.8401

Hodgkins lymphoma (HL) represents the most common subtype of malignant lymphoma in young people in the Western world. Most patients can be cured with modern treatment strategies, although approximately 20% will die after relapse or progressive disease. The histologic hallmark of the disease is the presence of the characteristic Hodgkin Reed-Sternberg (HRS) cells in classical HL and so-called lymphocyte-predominant (LP) cells in nodular lymphocyte-predominant HL. HL is unique among all cancers because malignant cells are greatly outnumbered by reactive cells in the tumor microenvironment and make up only approximately 1% of the tumor. Expression of a variety of cytokines and chemokines by the HRS and LP cells is believed to be the driving force for an abnormal immune response, perpetuated by additional factors secreted by reactive cells in the microenvironment that help maintain the inammatory milieu. The malignant HRS and LP cells manipulate the microenvironment, permitting them to develop their malignant phenotype fully and evade host immune attack. Gene expression signatures derived from non-neoplastic cells correlate well with response to initial and subsequent therapies, reecting their functional relevance. Recent biomarker studies have added texture to clinical outcome predictors, and their incorporation into prognostic models may improve our understanding of the biologic correlates of treatment failure. Moreover, recent preclinical and clinical studies have demonstrated that the tumor microenvironment represents a promising therapeutic target, raising hope that novel treatment strategies focused on the interface between malignant and reactive cells will soon emerge. J Clin Oncol 29. 2011 by American Society of Clinical Oncology

INTRODUCTION

Hodgkins lymphoma (HL) accounts for approximately 11% of all malignant lymphomas.1 The morphologic hallmarks of this unique lymphoma were initially described more than 100 years ago; it is characterized by the presence of Hodgkin ReedSternberg (HRS) cells in classical HL (cHL) and socalled lymphocyte-predominant (LP) cells in nodular lymphocyte-predominant HL (NLPHL).2 Typically, malignant cells are greatly outnumbered by reactive cells in a microenvironment that includes lymphocytes, macrophages, eosinophils, mast cells, plasma cells, stromal cells, broblasts, and other cells.3 Specically in cHL, the frequencies of all these cellular components, including the HRS cells, vary considerably between cHL subtypes; lymphocyte rich, lymphocyte depleted, mixed cellularity (MC), and nodular sclerosis (NS). Independent of advances in understanding the etiology and molecular biology of HL, the introduction of polychemotherapy has reduced the number of patients who succumb to the disease by more than

60% since the early 1970s.4 Consequently, most patients are cured by modern treatment strategies. Important milestones in our understanding of HL include rst, separation of HL into cHL versus NLPHL; second, development of distinct treatment strategies for limited versus advanced disease; and third, validation of the International Prognostic Score for advanced disease.5,6 Since its introduction in 1998, the International Prognostic Score has become the gold standard for risk assessment and stratication and been used to guide treatment decisions in advanced cHL. However, it can be anticipated that progress, especially among those 20% of patients who are still destined to die after relapse or progressive disease, will be tightly linked to novel biomarker discovery and targeted therapies for relapsed disease.7 In this review, we will rst focus on the crosstalk between malignant and reactive cells in the microenvironment mediated by cytokines and chemokines expressed in HL tissue. These soluble factors mediate the specic cellular composition of
2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Copyright 2011 by American Society of Clinical Oncology

Steidl, Connors, and Gascoyne

the reactive inltrate, promote receptor-mediated signaling, and cultivate immune privilege for malignant cells. We will then review the prognostic implications of the tumor microenvironment and nally discuss novel therapeutic approaches targeting the microenvironment or microenvironment-dependent receptor signaling.
CROSSTALK BETWEEN CELLS: THE IMPORTANCE OF CYTOKINES AND CHEMOKINES IN HL

Cytokines and chemokines are lowmolecular weight proteins with a wide variety of functions that work either in a paracrine manner to

modulate the activity of surrounding cells or in an autocrine fashion to directly affect the cells that produce them. Specically, these autocrine and paracrine interactions lead to the particular cellular composition found in HL and contribute to the proliferation and antiapoptotic phenotype of HRS cells (Fig 1). Although the cytokine and chemokine milieu in HL tissues is complex and interactive, a clear understanding of this biology will be clinically relevant in the future as therapies are developed that disrupt the crosstalk between malignant cells and the microenvironment. Hence, improved insight into the biology will be critical to introducing these novel biologic treatments into clinical practice. This section focuses on the involvement of cytokines in the

Fibroblast Fibroblast Mast cell CCL11/ Eotaxin Eosinophil


CCR3

IL8
CXCR1/2

B cell

FGF1/2 IL-13
CD30L IL-13R

CD30L

BLC CCL5 IL-4

TNF- LT- TGF- APRIL BAFF


CD95

Neutrophil

CD30

FASL
TNFR family

CCR3

HRS cell IL-5 MEC


CD30

HLA-G
CCR10 CD40

T cell (Treg /TH2)


CCR4

Cytotoxic T cell

CXCL16
CCR3/ CCR10

CD74

TARC MDC CD40L CCL20 IFN


CD7

Gal-1

MIF

PDL1 CX3CL1 CSF1

IL-10 TGF

CX3CR1 CSF1R

Macrophage

IFN

PD-1

IL-2R

IL-2 Plasma cell IL8 T cell (TH1)

NK cell

Fig 1. Schematic of the crosstalk between malignant Hodgkin Reed-Sternberg (HRS) cells and the tumor microenvironment in classical Hodgkins lymphoma. In the center, a bi-nucleated HRS cell is shown expressing characteristic surface molecules as well as secreted cytokines and chemokines. Surrounding the HRS cells are cell types representative of the non-neoplastic cells attracted and activated by these molecules. The cells in the microenvironment themselves express a variety of chemokines and cytokines that further shape the reactive inltrate and provide signaling for HRS cells. Only the main activating and inhibitory functions (arrows) of predominantly membrane-bound (purple triangles) and secreted molecules mediated by surface receptors (pink) are illustrated; other interactions exist. IL, interleukin; FGF, broblast growth factor; TNF-, tumor necrosis factor ; LT-, lymphotoxin ; TGF-, transforming growth factor ; BAFF, B-cell activating factor; APRIL, a proliferation-inducing ligand; BLC, B lymphocyte chemoattractant; TNFR, tumor necrosis factor receptor; FASL, Fas ligand; MEC, mucosae-associated epithelial chemokine; Treg, T regulatory cell; TH, T helper cell; TARC, thymus and activation-related chemokine; MDC, macrophage-derived chemokine; Gal-1, galectin-1; PDL, programmed cell death 1 ligand; IFN-, interferon gamma; NK, natural killer. 2
2011 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Microenvironment in Hodgkins Lymphoma

formation of the microenvironment and highlights how, in return, HRS cells receive signals from the microenvironment through surface receptors. The clinical and pathologic features of HL reect an abnormal immune response that is thought to be the result of expression of a variety of cytokines and chemokines by the HRS cells, altering the composition and function of the cells in the surrounding microenvironment and shaping the specic histopathologic appearance of the lymph node.8,9 Moreover, the reactive cells in the microenvironment produce specic cytokines and chemokines that help maintain and even amplify the intense inammatory reaction.10 Table 1 summarizes the most commonly described cytokines and chemokines in HL and their main functions. Of note, most of the studies in HL have focused on NSHL and MCHL, because NLPHL, lymphocyte-rich and lymphocyte-depleted cHL account for only a minority of cases.2 In addition to the variability of cytokine expression in the different HL subtypes, Epstein-Barr virus (EBV) positivity also inuences the relative expression of certain cytokines (eg, CXCL10, CXCL9, CCL5, and CCL3).9

B cells of various maturational stages are part of the normal cellular composition of lymph nodes, but in HL, this architecture is disturbed to varying degrees. Therefore, it remains an open question if and how reactive B cells are specically attracted into the microenvironment of HL, or whether they represent remnants not displaced by HL. However, as part of the general inammatory reaction, various cytokines, including TNF- and lymphotoxin , with effects on B-cell differentiation, proliferation, and chemotaxis have been shown to be expressed, which play critical roles in the initiation of germinal center reactions.21,33,66 In NLPHL, expression of B cellattracting chemokine 1 (CXCL13) has been demonstrated in a subset of cases.67

AUTOCRINE AND PARACRINE RECEPTOR SIGNALING BY HRS CELLS

COMPOSITION OF THE MICROENVIRONMENT

The most abundant cells found in HL are inltrating CD4 T cells.58 Several studies have revealed that the main subset of these cells displays a phenotype of T helper 2 (TH2) and T regulatory (Treg) cells, particularly those in the direct vicinity of HRS cells.13,59 HRS cells secrete regulated upon activation, normal T cell expressed and secreted (CCL5), thymus and activation-related chemokine (CCL17), and macrophage-derived chemokine (CCL22), potent chemokines that have all been shown to attract CCR4-expressing TH2 and Treg cells in HL.32,46,47,49-52 CCL5 has also been shown to attract mast cells commonly found in HL.46 Together with T helper 1 (TH1) cells, HRS cells themselves produce interferon gamma,11,15,16 an important enhancer of macrophage function. HRS cells also produce granulocyte colonystimulating factor (CSF1) and fractalkine (CX3CL1), chemokines and differentiation factors of monocytes and their progenitors explaining variable inltration by macrophages.40,43,60,61 In general, macrophages have been shown to be critical enhancers of tumor progression, to promote cell migration, and to suppress antitumor immunity in many cancers, including lymphoma.62 Particularly in HL, secretion of macrophage migration inhibitory factor (MIF) may contribute to the proliferation of HRS cells, which frequently express the CD74 receptor on their surface20,63; CD74 has been previously implicated in signaling and accessory functions for immune cell activation.64 Interleukin (IL) -8 expression by macrophages might also contribute to neutrophil inltration, which has been frequently described, most commonly in NSHL.53,54 Expression of IL-13, tumor necrosis factor (TNF) , and broblast growth factors by HRS cells has been well described,18,27,41 and this leads to the activation and proliferation of broblasts, most prominently seen in NSHL.48,65 Fibroblasts themselves can attract eosinophils and TH2 cells through secretion of eotaxin (CCL11).48 Furthermore, IL-5 and mucosae-associated epithelial chemokine (CCL28) expression of HRS might contribute to tissue eosinophilia.19,39 CCL28 and CXCL16 have been described as correlated with plasma cell inltration, a variable feature in cHL.39
www.jco.org

Although the malignant HRS cells gain a survival advantage by deregulated transcription factor networks and genetic lesions altering intrinsic signaling, the fully developed proliferation and antiapoptotic phenotype of HRS cells are critically dependent on their interaction with the microenvironment.68 In HL, the cooperation of direct genetic hits in signaling pathways with microenvironment-dependent signaling is most evident in the activation of nuclear factor kappa B (NFB) and Janus kinasesignal transducer and activator of transcription signaling (JAK-STAT; Fig 2). Many mutations and structural alterations in the genes involved in these pathways leading to constitutive activity, including chromosomal gains, overexpression of pathway genes,69-74 and inactivating mutations of tumor suppressor genes have been described.75-82 Moreover, the microenvironment provides signaling via surface receptors, namely IL receptors,83 members of the TNF receptor (TNFR) family,84 and receptor tyrosine kinases.85 Prominent receptor family members involved in activation of the canonical NFB pathway, including CD30 (TNFR superfamily [SF], member 8), CD40 (TNFRSF5), receptor activator of NFB (TNFRSF11A), and TNFR1 (TNFRSF1A), have been shown to be expressed on the surface of HRS cells.29,45,86,87 These receptors can be activated by membrane-bound or secreted ligands such as receptor activator of NFB ligand (TNFSF11, autocrine29), CD30L (TNFSF8, mast cells,30 eosinophils31), CD40L (TNFSF5, TH2/Treg86), TNF- (autocrine, macrophages and lymphocytes33,34), and lymphotoxin (autocrine21). Furthermore, in EBV-positive HL, the expression of viral oncogene LMP1 leads to protein aggregates in the cellular membrane mimicking an active CD40 receptor that similarly activates NFB signaling.88 Alternative activation (so-called noncanonical activation) of the NFB pathway can be achieved via the receptors transmembrane activator and calcium modulator and cyclophilin ligand interactor (TNFRSF13B) and B-cell maturation antigen (TNFRSF17) after engagement by their ligands a proliferation-inducing ligand (TNFSF13) and B-cell activating factor (TNFSF13B).28,89 Notably, these ligands are expressed not only by HRS cells but also by a variety of cells in the microenvironment, including macrophages, neutrophils, mast cells, and plasma cells, suggesting a prominent paracrine network.28 The constitutive activity of the Janus kinaseSTAT pathway is exemplied by autocrine and paracrine signaling of IL-13, which engages the IL-13 receptor and leads to HRS cell proliferation and survival by phosphorylated STAT6.18,90 Additionally, STAT3 activation through IL-21 and STAT5 activation have been described in HL.91,92 Overall, expression of a large number of ILs that display
2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Steidl, Connors, and Gascoyne

Table 1. Cytokines and Chemokines Involved in the Pathogenesis of Hodgkins Lymphoma Name TH1 cytokines IL-2 IL-12 IFN- TH2 cytokines IL-4 IL-5 IL-6 IL-9 IL-10 IL-13 TNF receptor ligands TNFSF13B TNFSF11 TNFSF13 TNFSF8 CD40LG LTA TNF- Other cytokines IL-1A IL-1B TGF- IL-3 IL-7 MIF VEGF CSF1 CSF2 Chemokines CC chemokines (-chemokines) CCL1 CCL2 CCL5 CCL11 CCL13 CCL17 CCL19 CCL20 CCL22 CCL28 CXC chemokines (-chemokines) CXCL8 CXCL9 CXCL10 CXCL12 CXCL13 CXCL16 CX3C chemokines CX3CL1 Alternative Name Lymphokine BCGF1 EDF IFNB2 P40 CSIF Biological Function/Function in Hodgkins Lymphoma T-cell growth factor Growth factor for activated T and NK cells Macrophage activation, CXCL10 induction B-cell activation Eosinophil differentiation B- and plasma cell activation and maturation Mast cell activation Negative regulation of TH1 cytokines B-cell maturation and activation, broblast stimulation, CCL17 and CCL22 induction B- and HRS cell proliferation and differentiation Augmentation of T-cell activation by dendritic cells, HRS cell stimulation B- and HRS cell proliferation and differentiation T-cell stimulation B- and HRS cell proliferation B-cell growth factor, collagen synthesis CCL11 production enhancement through broblasts, collagen synthesis Proinammatory cytokine, brosis and sclerosis induction Fibrosis and sclerosis induction B- and T-cell suppression, broblast proliferation, associated with nodular sclerosis subtype Growth factor for HRS cells Growth factor for B and T cells Blocks cytotoxic T lymphocyte response Endothelial cell growth, angiogenesis Monocyte growth and differentiation Granulocyte, macrophage, and eosinophil differentiation Reference No. 11, 12 13, 14 11, 15-17 12, 16 12, 18, 19 12, 20-22 23, 24 16, 25, 26 18, 27

BAFF RANKL APRIL CD30L TNFSF5 TNFSF1 TNFSF2 IL-1 IL-1 Multi-CSF M-CSF GM-CSF

28 29 19 30, 31 32 21 33, 34 33, 35 33, 35 26, 36 37, 38 39 40 41, 42 20, 43, 44 12, 22, 45

I-309 MCP-1 RANTES Eotaxin MCP-4 TARC MIP-3 MIP-3 MDC MEC IL-8 MIG IP-10 SDF1 BLC SRPSOX Fractalkine

Monocyte chemotaxis Monocyte and basophil chemotaxis T-cell, eosinophil, and mast cell recruitment Eosinophil accumulation, produced by broblasts (TNF- induced) Monocyte chemotactic protein Induction of TH2 response and regulatory T cells Promotes B- and T-cell migration Lymphocyte recruitment TH2 response and regulatory T cell induction Plasma cell and eosinophil recruitment Neutrophil recruitment Reactive inltrate in EBV-positive disease Reactive inltrate in EBV-positive disease Monocyte migration stimulation B lymphocyte migration Recruitment of plasma cells Chemotactic for T cells, NK cells and monocytes

32, 39 33 16, 46, 47 16, 39, 48 14 32 49-51 32 32 16, 32, 39, 52 39 53-56 9, 16 16, 32, 40, 50 57 32, 57 39 40

Abbreviations: TH, T helper cell; IL, interleukin; NK, natural killer; IFN-, interferon gamma; TNF, tumor necrosis factor; TNFSF, TNF superfamily; BAFF, B-cell activating factor; HRS, Hodgkin Reed-Sternberg; RANKL, receptor activator of nuclear factor kappa B ligand; APRIL, a proliferation-inducing ligand; LTA, lymphotoxin alpha; GM-CSF, granulocyte macrophage colony-stimulating factor; MIF, macrophage migration inhibitory factor; VEGF, vascular endothelial growth factor; MCP, monocyte chemotactic protein; TARC, thymus and activation-related chemokine; MDC, macrophage-derived chemokine; MEC, mucosae-associated epithelial chemokine; MIG, monokine induced by gamma interferon; EBV, Epstein-Barr virus; IP-10, 10KDa interferon gamma-induced protein; SDF, stromal cell derived factor; BLC, B lymphocyte chemoattractant; SRPSOX, scavenger receptor for phospatidylserine and oxidised low-density lipoprotein.

2011 by American Society of Clinical Oncology

JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Microenvironment in Hodgkins Lymphoma

JAK-STAT signaling
IL-4, IL13 IL-6, IL-10 IL-2, IL-7, IL-9, IL-15 IL-3, IL-5, GM-CSF

Canonical NFB
RANKL CD30L CD40L TNF- LT-

Alternative NFB

Tyrosine receptor kinases

APRIL BAFF

HGF NGF collagen type I

IL-2R IL-3R IL-6R IL-7R I IL-13R

CD40 TNFR1/2 RANK CD30

TACI BCMA

c M c-MET TRKA DDR2

STAT JAK1 JAK2 JAK3 TYK2


P P

STAT
P

RIP TRAF
P

TRAF
MAP3K14

Janus kinase

SHP2 GAB1 GRB2 SOS PI3K

STAT

STAT

NEMO IKK IKK

IKK complex

IKK IKK
p100 RELB

RAS

PDK

STAT-3 STAT-5 STAT-6

STAT STAT

STAT dimers/ oligomers

IB complex IB IB p50 RELA NFB complex Proteasomal degradation

RAF

AKT

p52 RELB

MEK
BAD MDM2 mTOR p21

ERK

Transcriptional regulation: - Inflammation - Survival - Cell proliferation - Differentiation

Apoptosis, cell cycle control, protein synthesis, DNA damage repair

Fig 2. Pathway activation in Hodgkin Reed-Sternberg cells through signaling from the microenvironment. Soluble and membrane-bound signaling molecules produced by reactive cells (paracrine activation) activate the Janus kinasesignal transducer and activator of transcription signaling (JAK-STAT) and canonical and alternative nuclear factor kappa B (NFB) pathways and receptor tyrosine kinases. For the JAK-STAT signaling pathway, the most commonly expressed interleukins (ILs), IL receptors, and activated STATs are shown. For the NFB pathway, only the principal activation cascades are shown, and inhibitor of kinase (IK) complex activation by other kinases are described. Downstream signaling of receptor tyrosine kinases is shown using the example of tyrosine kinase receptor A (TRKA) and illustrating the Ras and Akt pathways; other signal transduction pathways exist. GM-CSF, granulocyte macrophage colony-stimulating factor; RANKL, receptor activator of NFB ligand; TNF-, tumor necrosis factor ; LT-, lymphotoxin ; APRIL, a proliferation-inducing ligand; BAFF, B-cell activating factor; HGF, hepatocyte growth factor; NGF, nerve growth factor; TACI, transmembrane activator and CAML interactor; BCMA, B-cell maturation antigen; c-MET, mesenchymal-epithelial transition factor; DDR, discoidin domain receptor tyrosine kinase; TYK2, tyrosine kinase 2; RIP, receptor-interacting protein; TRAF, TNF receptorassociated factor 1; MAP3K, mitogenactivated protein 3 kinase; SHP2, Src homology 2 domain containing tyrosine phosphatase; SOS, son of sevenless homolog 1 (Drosophila); GRB2, growth factor receptor bound protein 2; PI3K, phosphoinositide 3-kinase; ERK, extracellular signal-regulated kinase; MEK, MAP-ERK kinase; PDK, 3-phosphoinositide dependant protein kinase; BAD, BCL2-associated agonist of cell death; MDM, mouse double minute; mTOR, mechanistic target of rapamycin.

receptor-specic binding similar to that seen with IL-13 has been reported in HL8,93 (Table 1), including ligands binding to IL-2 receptor complexes94,95 and the IL-3,37,38 IL-6,12,96 and IL-7 receptors.97 Recently, receptor tyrosine kinase signaling has been reported to play an important role in the pathogenesis of HL. In particular, the
www.jco.org

surface receptors mesenchymal-epithelial transition factor (c-MET), tyrosine receptor kinase A (TRKA), and discoidin domain receptor tyrosine kinase 2 (DDR2) are expressed and mediate survival signaling in HRS cells through paracrine signaling.98-102 The respective ligands of these receptors are thought to be produced by cells of the
2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Steidl, Connors, and Gascoyne

microenvironment. Hepatocyte growth factor (HGF), a specic ligand of c-MET, is expressed by CD21 follicular dendritic cells.99 Collagen type I, the high-afnity ligand of TRKA, is expressed by broblasts, and nerve growth factor (NGF) producing granulocytes were found abundantly in the direct vicinity of HRS cells.100 Furthermore, autocrine activation loops have been suggested for the plateletderived growth factor receptor (PDGFRA) and EPH receptor B1 (EPHB1). In summary, cytokine and chemokine expression have been extensively studied in HL, and specic ligand-receptor interactions help explain the composition of the tumor environment and downstream receptor signaling critically inuencing the phenotype of malignant cells.
IMMUNE ESCAPE BY HRS CELLS

As discussed, HRS cells are dependent on the extensive inammatory microenvironment and use receptor signaling pathways to maintain their high proliferative capacity and antiapoptotic phenotype. However, HRS cells also appear to orchestrate their microenvironment in many ways to evade host immune attack. As evidenced in HIVassociated HL, malignant cells can thrive in an environment in which host immunity is globally impaired.103 Moreover, in recent years, specic mechanisms have been uncovered detailing how HRS cells induce a favorable microenvironment with suppressed antitumor immune activity. Major advances have been made that characterize the phenotypes and functions of specic T-cell subsets,104 including immunosuppressive Treg cells, which are abundant in tissue biopsies of HL.105,106 Specically, it has been shown that HRS cells produce chemokines such as CCL5, CCL17, and CCL22, which attract TH2 and Treg cells49,52,107 and secrete IL-7, an IL capable of inducing differentiation of CD4 naive T cells toward FOXP3 Treg cells.97,108 Treg and HRS cells themselves produce IL-10 and transforming growth factor (TGF-), which exert inhibitory effects on T-cell effector functions, especially on cytotoxic T lymphocytes (CTLs).8,25,105,109 In addition, prostaglandin E2 (PGE2) has been implicated as a contributor to immunosuppression and pathogenesis of HL110,111 similar to cancer progression of solid tumors.112 PGE2 leads to decreased CD4 T-cell activation through inhibition of leukocytespecic protein tyrosine kinase (LCK),113 suggesting that PGE2 contributes to the CD4 T-cell impairment observed in HL. The cells producing PGE2 have yet to be determined. Another important mechanism by which HRS cells evade host immune response is by overexpression of surface molecules maintaining peripheral tolerance. For example, the overexpression of Fas ligand (CD95L) that has been described in virtually all HRS cells of NS and MC subtypes114-116 induces apoptosis in tumor-specic CTLs.117 Similarly, galectin-1 (LGLAS1) was found to be overexpressed on the surface of HRS cells in HL, correlating with a reduced CD8 inltration at the tumor site.118 In cocultures of activated T cells with HL cell lines, galectin-1 induced secretion of TH2 cytokines and expansion of Treg cells, whereas knockdown of galectin-1 increased overall T-cell viability and restored the TH1/TH2 balance.119 Furthermore, overexpression of ligands of the receptor molecule programmed cell death 1 (PD1), PD1 ligand 1 (PDL1; CD274) itself, and PDL2 (CD273) has been described in HL.120,121 PD1 belongs to the CD28 costimulatory receptor superfamily expressed on CTLs and can modify T-cell activ6
2011 by American Society of Clinical Oncology

ity by providing a second signal to T cells via the PD1 receptor, in conjunction with signaling through the T-cell receptor.122 In the setting of HL, PD1 ligand overexpression has been shown to contribute to the T-cell exhaustion observed in inltrating T cells. A recent study established a link between copy number gains of chromosome 9p where PDL1 and PDL2 reside and overexpression of these molecules in NSHL.121 Loss of HLA class I and II molecules on the surface of HRS cells occurs in a substantial proportion of cases and is correlated with latent EBV infection. HRS cell escape from immune surveillance has been suggested as a functional consequence of this nding,123-125 because the reduced immunogenicity of HRS cells could impair activation of CTLs (major histocompatibility complex [MHC] class I recognition) and CD4 T cells (MHC class II recognition). However, the underlying causes for this downregulation of HLA molecules and the exact functional consequences are largely unexplored in HL. In particular, no correlation between chromosomal loss and underexpression of HLA molecules has been found in two HL cell lines.126 Interestingly, certain HLA types are correlated with genetic susceptibility to HL EBV-positive HL cases in particularsuggesting that certain polymorphisms might effect antigen presentation to CTLs.127-129 Furthermore, expression of the nonclassical MHC class I gene HLA-G, the ligand for inhibitory receptors found on natural killer (NK) cells, subsets of T cells, and other immune cells may lead to evasion of NK-cell and CTL-mediated immune responses and has been implicated in the pathogenesis of HL.130 Such surface expression has been identied by immunohistochemistry (IHC) in more than half of cHL cases, and a strong correlation with absence of classical MHC class I molecule expression and EBV-negative status has been seen. In summary, it is now well established that the phenotypic changes that characterize HRS cells allow for efcient escape from the host immune attack through various mechanisms, including release of chemokines shaping an immunosuppressive microenvironment as well as the expression of surface molecules gaining immune privilege through the reduction of HRS cell immunogenicity. However, most of the underlying genetic mechanisms remain to be discovered.
BIOMARKERS IN HL: CORRELATIONS TO TREATMENT OUTCOME

As the impact of the microenvironment on the specic pathobiology of HL becomes increasingly clear, more and more work is focused on correlation of biomarkers with treatment outcome. Although treatment regimens have been remarkably stable in the last two decades, specic aspects of patient selection, varying histologies, and treatments might inuence cross-comparability and in part explain differing conclusions from these studies. In general, although a plethora of biomarkers associated with clinical outcome have been described, none of these factors has inuenced clinical practice to date. The main reason for this unsuccessful clinical translation lies in the lack of reproducibility between independent patient cohorts and prognostic implications that are not of signicant magnitude to justify a change in clinical management. For individual biomarkers, reproducibility of IHC scoring has been suggested as a reason for inconclusive results, and thus more robust multigene predictors have been reported based on expression proling.131,132 Table 2 summarizes some of the biomarkers for which outcome correlations have been described, separated into studies using IHC and gene expression proling. The focus
JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Microenvironment in Hodgkins Lymphoma

Table 2. Biomarkers With Outcome Correlations Described in the Literature Immunohistochemistry Marker/Signature Granzyme B TIA-1 FOXP3 CD20 BCL11A HLA-DR CD68, PNA ALDH1A1 STAT1 EBV-encoded small RNAs Expressed on/Staining Pattern Cytotoxic T cells Cytotoxic T cells Regulatory T cells Background B cells Background B cells, plasmacytoid dendritic cells HRS cells Macrophages Macrophages Macrophages HRS cells Function Target cell lysis Target cell lysis Transcriptional regulation B-cell differentiation Transcriptional regulation Antigen presentation Scavenger receptor Oxidative pathway/metabolism Transcriptional activation Activation of NFB Outcome Correlation Adverse (PFS, OS) Adverse (EFS, OS) Favorable (EFS) Favorable (OS, EFS) Favorable (OS, EFS) Reference No. 133-137 136,138 137-139 132,140 140

Favorable (FFS) Adverse (PFS, DSS) Adverse (DSS) Adverse in elderly patients, favorable in young patients (FFS, OS) Adverse (PFS)

123 132,141 142 143-147

MMP11

HRS cells, macrophages, endothelial cells, extracellular

Tissue remodeling Gene Expression Proling

132

Main Gene Components Angiogenic signature Adipocyte signature Fibroblast function/extracellular matrix remodeling ADH1B, CD93, SRPX, PLA2G2A, GPR126 GLUL, MGST1, COL1A2, FABP4 Adverse: MMP2, MMP3, TIMP1, COL1A1, COL4A1, COL4A2, COL5A1, COL18A1, COL16A1, MFAP2, THBS1/2, FN1, EDNRA, ITGB5, LAMA4; favorable: TIMP4, SPON1, LAMB1, TACR1, CCL26 BCL11A, BANK1, STAP1, BLNK, FCER2, CD24, CCL21 CD3D, CD8B1, CTSL, CD26, SH2D1A, IFI16, RGS13, CR2, ELL3, CCDC23, PPM1L, TRA@, PIK3CA ITM2A, SRPX, CTSB, APP ALDH1A1, LYZ, STAT1, ITGA4, CCL13, MS4A4A, CCL23, VCAN, HSP90AB3P, HSP90AB1, CTSB, CFL1, JMJD6, MAPK7, IKBKG, RAB7A, RXRA, MAPK13

Outcome Correlation Adverse (primary treatment failure) Adverse (primary treatment failure) Discordant: adverse/favorable (primary treatment outcome)

Reference No. 132 132 142,148

B-cell signature Cytotoxic T-cell signature

Favorable (primary treatment outcome) Adverse (primary treatment outcome) Adverse (primary treatment outcome) Adverse (primary treatment outcome)

132,140 131,132,142

Plasmacytoid dendritic cells Macrophage signature

132 131,132,142

Abbreviations: PFS, progression-free survival; OS, overall survival; TIA-1, T-cellrestricted intracellular antigen 1; EFS, event-free survival; FOXP3, forkhead box protein 3; BCL11A, B-cell lymphoma/leukemia 11A; HRS, Hodgkin Reed-Sternberg; FFS, failure-free survival; PNA, peanut agglutinin; DSS, disease-specic survival; ALDH1A1, aldehyde dehydrogenase 1A1; STAT, signal transducer and activator of transcription signaling; EBV, Epstein-Barr virus; NFB, nuclear factor kappa B; MMP11, matrix metalloproteinase 11.

of these studies has been cHL, because there are no biologic markers yet identied that can predict treatment outcome or transformation to an aggressive lymphoma in patients with NLPHL.
HISTOPATHOLOGY

The rst biomarker studies in cHL used classical histopathologic approaches to describe the specic cellular composition of subtypes and investigate correlations with treatment outcome. Although in initial reports, the rare lymphocyte-depleted subtype and grade 2 nodular sclerosis were reported to be associated with poor response to initial therapy and decreased survival,149 there is now overwhelming evidence that none of the cHL subtypes are correlated with survival using modern treatment modalities.133,150-152 Similarly, the number of HRS cells is not prognostically relevant. Tissue eosinophilia is still a subject of debate; a large study investigating diagnostic biopsies found a strong
www.jco.org

correlation between the presence of eosinophils and freedom from treatment failure and overall survival (OS),31 in agreement with a report showing prolonged disease-free survival in patients with heavy eosinophilic inltration in NSHL.153 However, several other studies could not conrm this nding.150,154,155 Mast cell inltration has also been linked to poor prognosis in cHL,156 and a recent study using c-kit/CD117 IHC was able to link the number of mast cells to unfavorable outcome after primary therapy157; however, these data have yet to be validated. These factors affecting HRS cells and the microenvironment as assessed by routine histopathology are of interest. However, none has yet had an impact on clinical practice.
MACROPHAGES

Some of the rst biomarker studies in the modern treatment era were conducted in the 1980s and suggested that increased numbers of
2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Steidl, Connors, and Gascoyne

macrophages, as measured using peanut agglutinin binding cells, were associated with more frequent constitutional symptoms and relapse.141,158 The association of macrophages with adverse treatment outcome has now been conrmed in an increasing number of patient cohorts. In a gene expression proling study investigating the microenvironment, the authors found several genes expressed by macrophages, including STAT1 and ALDH1A1, to be signicantly correlated with disease-specic survival when investigated by IHC.142 The importance of these markers was also conrmed in an independent patient cohort using quantitative reverse-transcriptase polymerase chain reaction from archival parafn material.131 However, it was not conrmed in a second study by the same group, in which the authors found a correlation of the macrophage activation genes LYZ and STAT1 with favorable outcome.159 Most recently, tumor tissue signatures of monocytes and tumor-associated macrophages were found by gene expression proling in patients with therapy-refractory disease or relapse when compared with patients who sustained longterm remissions.132 Most importantly, in independent validation experiments, the authors demonstrated that the enumeration of CD68 macrophages in lymph node biopsies was a strong and independent predictor of disease-specic survival. These data are encouraging; in this study, it could be demonstrated that the number of CD68 macrophages was also signicantly associated with the outcome of secondary treatments such as autologous stem-cell transplantation. Overall, the adverse prognostic impact of tumor-associated macrophages in cHL biopsies can now be considered as rmly established in patients treated with doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) or ABVD-like chemotherapy regimens. The correlation of tumor-associated macrophages with adverse clinical course and tumor progression has also been reported in follicular lymphoma160 and many other solid tumors in which macrophages stimulate tumor growth, angiogenesis, tumor cell migration, and invasion.62
T-CELL SUBSETS

nosis. Accordingly, high ratios of FOXP3/TIA-1 T cells138 were associated with favorable event- and disease-free survival and OS, whereas high ratios of FOXP3/GrB cells137 were shown to be associated with favorable failure-free survival and OS. The correlation of high numbers of intratumoral FOXP3 regulatory cells with OS and failure-free survival was validated in a subsequent study.139 In contrast, high numbers of regulatory T cells have mostly been reported to be associated with unfavorable outcome in various solid tumors.162 The reason for these unexpected ndings remains unclear. In an attempt to link T-cell inltration to the specic biology of HRS cells, the prognostically favorable low number of cytotoxic cells (CD8 T lymphocytes, CD57 NK cells, and GrB cells) was linked to overexpression of antiapoptotic proteins in HRS cells.163 Interestingly, recent work has shown that lack of HLA class II cell-surface expression on HRS cells was frequently observed, and this nding was associated with EBV-negative disease status, HLA class I loss, and adverse outcome in multivariate analysis.123 However, because loss of HLA class II likely leads to diminished activation of TH1 cells and thereby a reduced cytotoxic antitumor response, the exact functional mechanisms remain to be claried linking increased numbers of intratumoral CTLs with unfavorable outcome. Furthermore, signatures of cytotoxic T cells have been identied using gene expression proling, suggesting that overexpression of certain T-cell activation genes such as CD8B1, CD3D, CTSL, CD26, and SH2D1A might be correlated with inferior outcome.131,132,142 Additional biomarker studies investigating the prognostic impact of T-cell subsets together with tumor-associated macrophages are anticipated to address the question of whether combinations of IHC markers with well-established clinical risk factors might further improve the predictive value of a single biomarker alone.
BACKGROUND B CELLS

Another biomarker in the microenvironment of HL was described by Oudejans et al,134 who found that an increased number of activated CTLs in tissue biopsies of patients was associated with unfavorable clinical outcome. The authors used granzyme B (GrB) as an IHC marker and found GrB-expressing cells in the vast majority of patients, but they showed that patients with more than 15% GrB cells had signicantly shorter progression-free survival and OS. In a subsequent study of 83 patients with cHL using GrB IHC, this same factor was found to correlate with OS and to be independent of both age and stage in multivariate analysis.135 Several subsequent studies by independent groups validated these initial studies of CTLs as useful predictors of prognosis in cHL but also identied other T-cell subsets associated with treatment outcome.133,135-138,157,161 Despite this earlier work, the most useful T-cell marker combination for determining prognosis using IHC is still a subject of debate. In a large series of 257 patients with cHL represented on a tissue microarray, the number of FOXP3 Treg cells correlated with prolonged event- and disease-free survival, and the authors were able to validate the negative prognostic impact of CTLs.138 However, in this study, TIA-1another cytotoxic granule markerwas used to enumerate CTLs, and the number of GrB-activated CTLs was not signicantly associated with poor prog8
2011 by American Society of Clinical Oncology

Recently, the number of CD20 background B cells was reported to be signicantly associated with treatment outcome in two independent studies.132,140 Both studies found an overrepresentation of signatures of benign B cells in diagnostic biopsies of patients whose primary treatments were successful and were able to demonstrate using CD20 IHC signicant correlations with OS and disease-specic survival, respectively. Although in one study the prognostic impact of the number of CD20 background B cells on OS was independent of clinical parameters,140 the second study found a strong correlation of CD20 cells with clinical stage.72 Interestingly, another marker expressed on B cellsBCL11Awas an even better predictor for OS, possibly because of an association of BCL11A plasmacytoid dendritic cells with favorable outcome.
FOLLICULAR DENDRITIC CELLS

The presence and distribution of follicular dendritic cells (FDCs) have been studied in both cHL and NLPHL using IHC.164,165 These studies reveal the close spatial relationship of HRS and LP cells with the FDC network. In one study of cHL, the authors reported a correlation between the absence of FDCs and unfavorable outcome.166 In a follow-up study, the association with outcome was validated, but only in the context of certain patterns of CD21 FDC staining.167 The
JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Microenvironment in Hodgkins Lymphoma

longest OS was observed in patients with well-dened follicle-like structures compared with visible but largely destroyed FDC meshworks (intermediate survival) and absence of FDCs (poor survival). These data suggest that not only the number of FDCs but also the level of destruction of normal lymph node architecture might be an important variable for outcome prediction.
EBV ASSOCIATION

EBV infection of HRS cells occurs in up to 60% of patients in some studies but varies with geographic location, age, sex, clinical stage, and histologic type.168 Recent gene expression proling data have shed more light on immune-related cells in EBV-positive versus EBV-negative disease, specically identifying a TH1/antiviral response in EBV-positive patients.140 Furthermore, an increased number of cytotoxic T cells has been linked to EBV positivity.134 The impact of EBV infection on outcome is controversial, with some studies showing no impact150,169 and others favorable143,170 or unfavorable outcomes.144,145 These discrepancies might result in part from patient selection and confounding differences in demographic factors, because there is an increasing body of work supporting an association between EBV positivity and adverse outcome in older adult patients144-146 and a favorable effect in younger, specically pediatric, patients.143,144,147 Additional studies might clarify the clinical value of these ndings; the apparent age dependence has hampered the translation of EBV status into a widely used biomarker.
SERUM MARKERS

IHC, including tissue-associated macrophages, background reactive B cells, and CTLs132,140,142; however, conrmation is still needed for others (Table 2). Of note, the prognostic implications of angiogenesis, adipoctyes, and dendritic cells have been studied in other lymphomas.179-181 Proteomic studies have recently been performed in HL. Two approaches have been chosen thus far, one using total cell lysates, the other studying the secretome of HRS cells.40,182,183 These studies have demonstrated the potential value for developing novel diagnostic tools by identifying specic proles linked to certain lymphoma entities and have helped to characterize secreted proteins that are critically involved in the crosstalk of HRS cells with their microenvironment.40 However, no outcome correlations have been described to date. Only one study has investigated correlations of a micro-RNA prole, which was previously found in HL,184 with outcome. The authors found that miR-135a expression was associated with a higher probability of relapse and shorter disease-free survival.185

THERAPEUTIC APPROACHES TARGETING THE TUMOR MICROENVIRONMENT

It has been shown that certain cytokines, chemokines, and other soluble factors are detectable in the sera of patients with HL, and multiple studies have focused on the negative prognostic impact of these molecules.17,171,172 In general, increased levels of these serum markers can be interpreted as surrogates for active crosstalk between HRS cells and their microenvironment. Characteristically, many of those markers, including sCD30, serum IL-2 receptor, soluble intercellular adhesion molecule (sCD54), and vascular cell adhesion molecule 1 (sCD106), have shown correlations to one another, and elevated levels have been reported to be associated with advanced tumor stage and unfavorable prognosis.173-176 Although all these markers have shown prognostic signicance, their independent value in comparison with the established clinical surrogate markers for tumor burden and disease activity has not been convincingly demonstrated.
GENOME-WIDE STUDIES

Gene expression proling studies have contributed substantially to improved understanding of the disease with respect to the inherent phenotypic features of malignant HRS cells177,178 and the specic composition of the microenvironment.132,140,142,148 Using the unbiased approach of transcriptome-wide gene expression proling, gene signatures have been identied that correlate with failure or success after primary treatments. Many of these signatures can be related to cellular compartments represented in whole-tissue biopsies. Some of these signatures have been subsequently validated by
www.jco.org

Although HL is overall a successfully treated disease, treatment failure in a substantial proportion of patients and treatment-related early and late sequelae of chemotherapy and radiotherapy underscore the need for novel therapeutic approaches, especially for relapsed HL. No new drugs have been approved for HL in the past two decades186; however, many agents are being tested in preclinical studies and currently enrolling active trials. Table 3 lists a selection of novel therapeutic agents in HL that are thought to target reactive cells in the microenvironment or surface receptors on the malignant cells or to interfere with downstream receptor signaling. The concept that the tumor microenvironment might be a promising therapeutic target has been reinforced by reports in relapsed HL about the efcacy of the pyrimidine analog gemcitabine,210 a cytotoxic agent that has been reported to specically target Treg cells.211 Furthermore, the results of a study combining the anti-CD20 monoclonal antibody rituximab with gemcitabine in relapsed HL are encouraging and suggest that a combined treatment targeting reactive B and T cells in the microenvironment might be an effective approach.188 Trials targeting CD20 B cells using radioimmunoconjugates have been initiated.191 However, it remains controversial if the efcacy of anti-CD20 immunotherapy results from the direct killing of HRS cells that do occasionally express CD20212 or from the depletion of HRS cellsupporting reactive B cells in the microenvironment.187 Of note, two gene expression proling studies have observed a correlation between increased numbers of background B cells and favorable prognosis, a nding that needs further clarication in the context of rituximab treatment.132,140 Support for a direct effect on HRS cells also comes from a recent study suggesting that HRS progenitor cells might express CD20 on their surface and could be eradicated by rituximab.213 Unequivocally, treatment effects in NLPHL have been attributed to a direct effect on the CD20-expressing LP cells.189,190 Other antibodies with a predominant effect on the microenvironment that are currently being tested in clinical trials including patients with HL are represented by alemtuzumab (anti CD52)193 and galiximab (antiCD80).192 Furthermore, immunotherapy with EBV-specic cytotoxic
2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Steidl, Connors, and Gascoyne

Table 3. Selected Therapeutic Agents in Hodgkins Lymphoma and Their Presumed Targets Drug Cellular targets in microenvironment Rituximab
131

Main Target CD20 CD20 CD20 CD20 CD20 CD20 CD80 B cells HRS cells (LP cells) B cells HRS cells (LP cells) B cells HRS cells (LP cells) B cells, follicular dendritic cells, HRS cells

Reference No./Clinical Trial No. 187-190 NCT00484874 191 192 NCT00516217 193 NCT01030900 194 NCT00284804 195,196 NCT00947856, NCT01100502, NCT01060904 197 NCT00791011 198 NCT00670592 199 NCT00967044 200-202 NCT00439361 203,204 205 NCT00742027 206,207 NCT00540007 208,209

Itositumomab

90

Yibritumomab tiuxetan

Galiximab Alemtuzumab Receptor signaling on HRS cells MDX-060 SGN-35 AMG655 HCD122 Downstream signaling Everolimus Bortezomib Vorinostat Panobinostat Other Lenalidomide Cytotoxic T lymphocytes

CD52 cells in microenvironment

CD30 HRS cells CD30 HRS cells TNFRSF10B (TRAIL-R2) on HRS cells CD40 HRS cells, TH2/Treg signaling

TNFR signaling: PI3K, mTOR NFB signaling: inhibition of degradation of IB Histone modication, JAK-STAT signaling (pSTAT6) Histone modication

Immunomodulation, anti-angiogenic EBV-positive disease: viral LMP2

NOTE. US Food and Drug Administration clinical trail numbers according to http://clinicaltrials.gov. Abbreviations: HRS, Hodgkin Reed-Sternberg; LP, lymphocyte predominant; TNFRSF, tumor necrosis factor receptor superfamily; TH, T helper cell; Treg, T regulatory cell; PI3K, phosphoinositide 3-kinase; mTOR, mechanistic target of rapamycin; NFB, nuclear factor kappa B; IB, inhibitory kappa B; JAK-STAT, Janus kinasesignal transducer and activator of transcription signaling; EBV, Epstein-Barr virus; LMP2, latent membrane protein 2.

effector cells has shown promising results, providing proof of principle for adoptive immunotherapy with ex vivo expanded viral antigenspecic T cells.208 Remarkably, in a subsequent study, ve of six patients with relapsed HL had a tumor response; of these ve, four achieved complete remissions that were sustained for more than 9 months.209 Because of the increased expression of members of the TNF receptor family and the dependence of malignant cells on TNF receptor downstream signaling, these molecules are considered ideal targets for specic agents in HL. TNFRSF8 (CD30) in particular has been the target of a number of preclinical and clinical studies, of which the anti-CD30 compounds SGN-30 and MDX060 can be considered representative but disappointing in phase I/II clinical trials194,214 because of limited efcacy in relapsed disease. However, using an alternative strategy of conjugating an anti-CD30 antibody with the cytotoxic antimicrotubule agent monomethyl auristatin E (ie, SGN-35 or brentuximab vedotin),195 much higher partial and complete remission rates were achieved.196 It is noteworthy that in a heavily pretreated patient cohort, tumor reductions were documented in more than 80% of patients. As a result, additional clinical trials with brentuximab vedotin as a
10
2011 by American Society of Clinical Oncology

single agent and in combination with ABVD are currently recruiting. An anti-CD40 (TNFRSF5) antibody (HCD122198) trial and a study of the combination bortezomib with agonistic antiTNFrelated apoptosis-inducing ligand receptor 2 (TNFRSF10B) compound AMG655197 are enrolling patients with HL; however, results are still pending. A number of novel drugs have downstream receptor signaling as their primary targets, of which everolimus (mammalian target of rapamycin, phosphoinositide 3-kinase pathway),199 bortezomib (TNFR signaling, NFB pathway),200 and vorinostat (STAT6, IL signaling)204 have shown promising preclinical results. Of these, however, bortezomib as a single agent in relapsed HL had low clinical efcacy.202 Combination therapies are now being tested. In summary, therapies targeting cells in the microenvironment or disrupting microenvironment-dependent signaling in the malignant cells seem to be effective and show promise in patients with relapsed HL. Current clinical trials are also focusing on combination therapies with classical chemotherapy agents. Randomized trials comparing these novel agents with the current standard of care for rstand second-line therapies will ultimately determine their signicance in the overall landscape of HL treatment.
JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Microenvironment in Hodgkins Lymphoma

AUTHORS DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST


The author(s) indicated no potential conicts of interest.

AUTHOR CONTRIBUTIONS
Manuscript writing: All authors Final approval of manuscript: All authors
33. Xerri L, Birg F, Guigou V, et al: In situ expression of the IL-1-alpha and TNF-alpha genes by Reed-Sternberg cells in Hodgkins disease. Int J Cancer 50:689-693, 1992 34. Kretschmer C, Jones DB, Morrison K, et al: Tumor necrosis factor alpha and lymphotoxin production in Hodgkins disease. Am J Pathol 137:341351, 1990 35. Ree HJ, Crowley JP, Dinarello CA: Antiinterleukin-1 reactive cells in Hodgkins disease. Cancer 59:1717-1720, 1987 36. Hsu SM, Lin J, Xie SS, et al: Abundant expression of transforming growth factor-beta 1 and -beta 2 by Hodgkins Reed-Sternberg cells and by reactive T lymphocytes in Hodgkins disease. Hum Pathol 24:249-255, 1993 37. Aldinucci D, Olivo K, Lorenzon D, et al: The role of interleukin-3 in classical Hodgkins disease. Leuk Lymphoma 46:303-311, 2005 38. Aldinucci D, Poletto D, Gloghini A, et al: Expression of functional interleukin-3 receptors on Hodgkin and Reed-Sternberg cells. Am J Pathol 160:585-596, 2002 39. Hanamoto H, Nakayama T, Miyazato H, et al: Expression of CCL28 by Reed-Sternberg cells denes a major subtype of classical Hodgkins disease with frequent inltration of eosinophils and/or plasma cells. Am J Pathol 164:997-1006, 2004 40. Ma Y, Visser L, Roelofsen H, et al: Proteomics analysis of Hodgkin lymphoma: Identication of new players involved in the cross-talk between HRS cells and inltrating lymphocytes. Blood 111:23392346, 2008 41. Khnykin D, Troen G, Berner JM, et al: The expression of broblast growth factors and their receptors in Hodgkins lymphoma. J Pathol 208:431438, 2006 42. Giles FJ, Vose JM, Do KA, et al: Clinical relevance of circulating angiogenic factors in patients with non-Hodgkins lymphoma or Hodgkins lymphoma. Leuk Res 28:595-604, 2004 43. Hsu PL, Lin YC, Hsu SM: Expression of macrophage colony-stimulating factor (M-CSF) in two Hodgkins Reed-Sternberg (H-RS) cell lines, HDLM-1 and KM-H2, and in H-RS cells in tissues. Int J Hematol 54:315-326, 1991 44. Lamprecht B, Walter K, Kreher S, et al: Derepression of an endogenous long terminal repeat activates the CSF1R proto-oncogene in human lymphoma. Nat Med 16:571-579, 2010 45. Pinto A, Aldinucci D, Gloghini A, et al: Human eosinophils express functional CD30 ligand and stimulate proliferation of a Hodgkins disease cell line. Blood 88:3299-3305, 1996 46. Fischer M, Juremalm M, Olsson N, et al: Expression of CCL5/RANTES by Hodgkin and ReedSternberg cells and its possible role in the recruitment of mast cells into lymphomatous tissue. Int J Cancer 107:197-201, 2003 47. Buri C, Korner M, Scharli P, et al: CC chemokines and the receptors CCR3 and CCR5 are differentially expressed in the nonneoplastic leukocytic inltrates of Hodgkin disease. Blood 97:1543-1548, 2001 48. Jundt F, Anagnostopoulos I, Bommert K, et al: Hodgkin/Reed-Sternberg cells induce broblasts 11

REFERENCES
1. Horner MJ, Ries LAG, Krapcho M, et al (eds): SEER Cancer Statistics Review, 1975-2006, National Cancer Institute. http://seer.cancer.gov/csr/ 1975_2006/ 2. Swerdlow SH, Campo E, Harris NL, et al (eds): WHO Classication of Tumours of Haematopoietic and Lymphoid Tissues. Lyon, France, International Agency for Research on Cancer, 2008 3. Pileri SA, Ascani S, Leoncini L, et al: Hodgkins lymphoma: The pathologists viewpoint. J Clin Pathol 55:162-176, 2002 4. Diehl V, Stein H, Hummel M, et al: Hodgkins lymphoma: Biology and treatment strategies for primary, refractory, and relapsed disease. Hematology Am Soc Hematol Educ Program 225-247, 2003 5. Hasenclever D, Diehl V: A prognostic score for advanced Hodgkins disease: International Prognostic Factors Project on Advanced Hodgkins Disease. N Engl J Med 339:1506-1514, 1998 6. Engert A, Plutschow A, Eich HT, et al: Reduced treatment intensity in patients with earlystage Hodgkins lymphoma. N Engl J Med 363:640652, 2010 7. DeVita VT Jr, Costa J: Toward a personalized treatment of Hodgkins disease. N Engl J Med 362:942-943, 2010 8. Skinnider BF, Mak TW: The role of cytokines in classical Hodgkin lymphoma. Blood 99:42834297, 2002 9. Teruya-Feldstein J, Tosato G, Jaffe ES: The role of chemokines in Hodgkins disease. Leuk Lymphoma 38:363-371, 2000 10. Khan G: Epstein-Barr virus, cytokines, and inammation: A cocktail for the pathogenesis of Hodgkins lymphoma? Exp Hematol 34:399-406, 2006 11. Dukers DF, Jaspars LH, Vos W, et al: Quantitative immunohistochemical analysis of cytokine proles in Epstein-Barr virus-positive and -negative cases of Hodgkins disease. J Pathol 190:143-149, 2000 12. Klein S, Jucker M, Diehl V, et al: Production of multiple cytokines by Hodgkins disease derived cell lines. Hematol Oncol 10:319-329, 1992 13. Ma Y, Visser L, Blokzijl T, et al: The CD4CD26 T-cell population in classical Hodgkins lymphoma displays a distinctive regulatory T-cell prole. Lab Invest 88:482-490, 2008 14. Schwaller J, Tobler A, Niklaus G, et al: Interleukin-12 expression in human lymphomas and nonneoplastic lymphoid disorders. Blood 85:21822188, 1995 15. Gerdes J, Kretschmer C, Zahn G, et al: Immunoenzymatic assessment of interferon-gamma in Hodgkin and Sternberg-Reed cells. Cytokine 2:307310, 1990 16. Teruya-Feldstein J, Jaffe ES, Burd PR, et al: Differential chemokine expression in tissues involved by Hodgkins disease: Direct correlation of eotaxin expression and tissue eosinophilia. Blood 93:2463-2470, 1999 17. Kurzrock R, Redman J, Cabanillas F, et al: Serum interleukin 6 levels are elevated in lymphoma patients and correlate with survival in advanced www.jco.org

Hodgkins disease and with B symptoms. Cancer Res 53:2118-2122, 1993 18. Kapp U, Yeh WC, Patterson B, et al: Interleukin 13 is secreted by and stimulates the growth of Hodgkin and Reed-Sternberg cells. J Exp Med 189: 1939-1946, 1999 19. Samoszuk M, Nansen L: Detection of interleukin-5 messenger RNA in Reed-Sternberg cells of Hodgkins disease with eosinophilia. Blood 75:13-16, 1990 20. Hsu SM, Hsu PL: The nature of ReedSternberg cells: Phenotype, genotype, and other properties. Crit Rev Oncog 5:213-245, 1994 21. Foss HD, Herbst H, Oelmann E, et al: Lymphotoxin, tumour necrosis factor and interleukin-6 gene transcripts are present in Hodgkin and ReedSternberg cells of most Hodgkins disease cases. Br J Haematol 84:627-635, 1993 22. Merz H, Fliedner A, Orscheschek K, et al: Cytokine expression in T-cell lymphomas and Hodgkins disease: Its possible implication in autocrine or paracrine production as a potential basis for neoplastic growth. Am J Pathol 139:1173-1180, 1991 23. Merz H, Houssiau FA, Orscheschek K, et al: Interleukin-9 expression in human malignant lymphomas: Unique association with Hodgkins disease and large cell anaplastic lymphoma. Blood 78:13111317, 1991 24. Glimelius I, Edstrom A, Amini RM, et al: IL-9 expression contributes to the cellular composition in Hodgkin lymphoma. Eur J Haematol 76:278-283, 2006 25. Herbst H, Foss HD, Samol J, et al: Frequent expression of interleukin-10 by Epstein-Barr virusharboring tumor cells of Hodgkins disease. Blood 87:2918-2929, 1996 26. Atayar C, Poppema S, Blokzijl T, et al: Expression of the T-cell transcription factors, GATA-3 and T-bet, in the neoplastic cells of Hodgkin lymphomas. Am J Pathol 166:127-134, 2005 27. Skinnider BF, Elia AJ, Gascoyne RD, et al: Interleukin 13 and interleukin 13 receptor are frequently expressed by Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood 97:250-255, 2001 28. Chiu A, Xu W, He B, et al: Hodgkin lymphoma cells express TACI and BCMA receptors and generate survival and proliferation signals in response to BAFF and APRIL. Blood 109:729-739, 2007 29. Fiumara P, Snell V, Li Y, et al: Functional expression of receptor activator of nuclear factor kappaB in Hodgkin disease cell lines. Blood 98:27842790, 2001 30. Molin D, Fischer M, Xiang Z, et al: Mast cells express functional CD30 ligand and are the predominant CD30L-positive cells in Hodgkins disease. Br J Haematol 114:616-623, 2001 31. von Wasielewski R, Seth S, Franklin J, et al: Tissue eosinophilia correlates strongly with poor prognosis in nodular sclerosing Hodgkins disease, allowing for known prognostic factors. Blood 95: 1207-1213, 2000 32. Maggio EM, Van Den Berg A, Visser L, et al: Common and differential chemokine expression patterns in rs cells of NLP, EBV positive and negative classical Hodgkin lymphomas. Int J Cancer 99:665672, 2002

2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Steidl, Connors, and Gascoyne

to secrete eotaxin, a potent chemoattractant for T cells and eosinophils. Blood 94:2065-2071, 1999 49. van den Berg A, Visser L, Poppema S: High expression of the CC chemokine TARC in ReedSternberg cells: A possible explanation for the characteristic T-cell inltratein Hodgkins lymphoma. Am J Pathol 154:1685-1691, 1999 50. Ohshima K, Tutiya T, Yamaguchi T, et al: Inltration of Th1 and Th2 lymphocytes around Hodgkin and Reed-Sternberg (H&RS) cells in Hodgkin disease: Relation with expression of CXC and CC chemokines on H&RS cells. Int J Cancer 98:567-572, 2002 51. Peh SC, Kim LH, Poppema S: TARC, a CC chemokine, is frequently expressed in classic Hodgkins lymphoma but not in NLP Hodgkins lymphoma, T-cell-rich B-cell lymphoma, and most cases of anaplastic large cell lymphoma. Am J Surg Pathol 25:925-929, 2001 52. Hedvat CV, Jaffe ES, Qin J, et al: Macrophage-derived chemokine expression in classical Hodgkins lymphoma: Application of tissue microarrays. Mod Pathol 14:1270-1276, 2001 53. Foss HD, Herbst H, Gottstein S, et al: Interleukin-8 in Hodgkins disease: Preferential expression by reactive cells and association with neutrophil density. Am J Pathol 148:1229-1236, 1996 54. Luciani MG, Stoppacciaro A, Peri G, et al: The monocyte chemotactic protein a (MCP-1) and interleukin 8 (IL-8) in Hodgkins disease and in solid tumours. Mol Pathol 51:273-276, 1998 55. Trumper L, Jung W, Dahl G, et al: Interleukin-7, interleukin-8, soluble TNF receptor, and p53 protein levels are elevated in the serum of patients with Hodgkins disease. Ann Oncol 5:93-96, 1994 (suppl 1) 56. Gorschlu ter M, Bohlen H, Hasenclever D, et al: Serum cytokine levels correlate with clinical parameters in Hodgkins disease. Ann Oncol 6:477482, 1995 57. Hopken UE, Foss HD, Meyer D, et al: Upregulation of the chemokine receptor CCR7 in classical but not in lymphocyte-predominant Hodgkin disease correlates with distinct dissemination of neoplastic cells in lymphoid organs. Blood 99:11091116, 2002 58. Poppema S, Bhan AK, Reinherz EL, et al: In situ immunologic characterization of cellular constituents in lymph nodes and spleens involved by Hodgkins disease. Blood 59:226-232, 1982 59. Poppema S, Lai R, Visser L, et al: CD45 (leucocyte common antigen) expression in T and B lymphocyte subsets. Leuk Lymphoma 20:217-222, 1996 60. Alexander WS: Cytokines in hematopoiesis. Int Rev Immunol 16:651-682, 1998 61. Truman LA, Ford CA, Pasikowska M, et al: CX3CL1/fractalkine is released from apoptotic lymphocytes to stimulate macrophage chemotaxis. Blood 112:5026-5036, 2008 62. Qian BZ, Pollard JW: Macrophage diversity enhances tumor progression and metastasis. Cell 141:39-51, 2010 63. Stein R, Qu Z, Cardillo TM, et al: Antiproliferative activity of a humanized anti-CD74 monoclonal antibody, hLL1, on B-cell malignancies. Blood 104:3705-3711, 2004 64. Leng L, Metz CN, Fang Y, et al: MIF signal transduction initiated by binding to CD74. J Exp Med 197:1467-1476, 2003 65. Ohshima K, Akaiwa M, Umeshita R, et al: Interleukin-13 and interleukin-13 receptor in Hodgkins disease: Possible autocrine mechanism and involvement in brosis. Histopathology 38: 368-375, 2001 12

66. Vu F, Dianzani U, Ware CF, et al: ICOS, CD40, and lymphotoxin beta receptors signal sequentially and interdependently to initiate a germinal center reaction. J Immunol 180:2284-2293, 2008 67. Nam-Cha SH, Roncador G, Sanchez-Verde L, et al: PD-1, a follicular T-cell marker useful for recognizing nodular lymphocyte-predominant Hodgkin lymphoma. Am J Surg Pathol 32:1252-1257, 2008 68. Ku ppers R: The biology of Hodgkins lymphoma. Nat Rev Cancer 9:15-27, 2009 69. Joos S, Menz CK, Wrobel G, et al: Classical Hodgkin lymphoma is characterized by recurrent copy number gains of the short arm of chromosome 2. Blood 99:1381-1387, 2002 70. Mart n-Subero JI, Gesk S, Harder L, et al: Recurrent involvement of the REL and BCL11A loci in classical Hodgkin lymphoma. Blood 99:14741477, 2002 71. Chui DT, Hammond D, Baird M, et al: Classical Hodgkin lymphoma is associated with frequent gains of 17q. Genes Chromosomes Cancer 38:126136, 2003 72. Steidl C, Telenius A, Shah SP, et al: Genomewide copy number analysis of Hodgkin ReedSternberg cells identies recurrent imbalances with correlations to treatment outcome. Blood 116:418427, 2010 73. Mathas S, Jo hrens K, Joos S, et al: Elevated NF-kappaB p50 complex formation and Bcl-3 expression in classical Hodgkin, anaplastic large-cell, and other peripheral T-cell lymphomas. Blood 106: 4287-4293, 2005 74. Martin-Subero JI, Wlodarska I, Bastard C, et al: Chromosomal rearrangements involving the BCL3 locus are recurrent in classical Hodgkin and peripheral T-cell lymphoma. Blood 108:401-402, 2006; author reply 402-403 75. Cabannes E, Khan G, Aillet F, et al: Mutations in the IkBa gene in Hodgkins disease suggest a tumour suppressor role for IkappaBalpha. Oncogene 18:3063-3070, 1999 76. Emmerich F, Meiser M, Hummel M, et al: Overexpression of I kappa B alpha without inhibition of NF-kappaB activity and mutations in the I kappa B alpha gene in Reed-Sternberg cells. Blood 94:31293134, 1999 77. Jungnickel B, Staratschek-Jox A, Bra uninger A, et al: Clonal deleterious mutations in the IkappaBalpha gene in the malignant cells in Hodgkins lymphoma. J Exp Med 191:395-402, 2000 78. Emmerich F, Theurich S, Hummel M, et al: Inactivating I kappa B epsilon mutations in Hodgkin/ Reed-Sternberg cells. J Pathol 201:413-420, 2003 79. Schmitz R, Hansmann ML, Bohle V, et al: TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. J Exp Med 206:981-989, 2009 80. Weniger MA, Melzner I, Menz CK, et al: Mutations of the tumor suppressor gene SOCS-1 in classical Hodgkin lymphoma are frequent and associated with nuclear phospho-STAT5 accumulation. Oncogene 25:2679-2684, 2006 81. Mottok A, Renne C, Willenbrock K, et al: Somatic hypermutation of SOCS1 in lymphocytepredominant Hodgkin lymphoma is accompanied by high JAK2 expression and activation of STAT6. Blood 110:3387-3390, 2007 82. Maggio EM, Stekelenburg E, Van den Berg A, et al: TP53 gene mutations in Hodgkin lymphoma are infrequent and not associated with absence of Epstein-Barr virus. Int J Cancer 94:60-66, 2001 83. Leonard WJ: Role of Jak kinases and STATs in cytokine signal transduction. Int J Hematol 73: 271-277, 2001

84. Karin M, Gallagher E: TNFR signaling: Ubiquitin-conjugated TRAFc signals control stopand-go for MAPK signaling complexes. Immunol Rev 228:225-240, 2009 85. Manning G, Whyte DB, Martinez R, et al: The protein kinase complement of the human genome. Science 298:1912-1934, 2002 86. Carbone A, Gloghini A, Gruss HJ, et al: CD40 ligand is constitutively expressed in a subset of T cell lymphomas and on the microenvironmental reactive T cells of follicular lymphomas and Hodgkins disease. Am J Pathol 147:912-922, 1995 87. Messineo C, Jamerson MH, Hunter E, et al: Gene expression by single Reed-Sternberg cells: Pathways of apoptosis and activation. Blood 91: 2443-2451, 1998 88. Kilger E, Kieser A, Baumann M, et al: Epstein-Barr virus-mediated B-cell proliferation is dependent upon latent membrane protein 1, which simulates an activated CD40 receptor. Embo J 17: 1700-1709, 1998 89. Schwaller J, Went P, Matthes T, et al: Paracrine promotion of tumor development by the TNF ligand APRIL in Hodgkins disease. Leukemia 21: 1324-1327, 2007 90. Skinnider BF, Elia AJ, Gascoyne RD, et al: Signal transducer and activator of transcription 6 is frequently activated in Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood 99:618-626, 2002 91. Scheeren FA, Diehl SA, Smit LA, et al: IL-21 is expressed in Hodgkin lymphoma and activates STAT5: evidence that activated STAT5 is required for Hodgkin lymphomagenesis. Blood 111:47064715, 2008 92. Kube D, Holtick U, Vockerodt M, et al: STAT3 is constitutively activated in Hodgkin cell lines. Blood 98:762-770, 2001 93. Aldinucci D, Gloghini A, Pinto A, et al: The classical Hodgkins lymphoma microenvironment and its role in promoting tumour growth and immune escape. J Pathol 221:248-263, 2010 94. Tesch H, Gunther A, Abts H, et al: Expression of interleukin-2R alpha and interleukin-2R beta in Hodgkins disease. Am J Pathol 142:1714-1720, 1993 95. Trumper LH, Brady G, Bagg A, et al: Singlecell analysis of Hodgkin and Reed-Sternberg cells: Molecular heterogeneity of gene expression and p53 mutations. Blood 81:3097-3115, 1993 96. Jucker M, Abts H, Li W, et al: Expression of interleukin-6 and interleukin-6 receptor in Hodgkins disease. Blood 77:2413-2418, 1991 97. Cattaruzza L, Gloghini A, Olivo K, et al: Functional coexpression of Interleukin (IL)-7 and its receptor (IL-7R) on Hodgkin and Reed-Sternberg cells: Involvement of IL-7 in tumor cell growth and microenvironmental interactions of Hodgkins lymphoma. Int J Cancer 125:1092-1101, 2009 98. Pons E, Uphoff CC, Drexler HG: Expression of hepatocyte growth factor and its receptor c-met in human leukemia-lymphoma cell lines. Leuk Res 22:797-804, 1998 99. Teoli L, Di Febo AL, Pierconti F, et al: Expression of the c-met proto-oncogene and its ligand, hepatocyte growth factor, in Hodgkin disease. Blood 97:1063-1069, 2001 100. Renne C, Willenbrock K, Ku ppers R, et al: Autocrine- and paracrine-activated receptor tyrosine kinases in classic Hodgkin lymphoma. Blood 105: 4051-4059, 2005 101. Willenbrock K, Ku ppers R, Renne C, et al: Common features and differences in the transcriptome of large cell anaplastic lymphoma and classical
JOURNAL OF CLINICAL ONCOLOGY

2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Microenvironment in Hodgkins Lymphoma

Hodgkins lymphoma. Haematologica 91:596-604, 2006 102. Renne C, Minner S, Ku ppers R, et al: Autocrine NGFbeta/TRKA signalling is an important survival factor for Hodgkin lymphoma derived cell lines. Leuk Res 32:163-167, 2008 103. Carbone A, Gloghini A, Serraino D, et al: HIV-associated Hodgkin lymphoma. Curr Opin HIV AIDS 4:3-10, 2009 104. Sakaguchi S, Miyara M, Costantino CM, et al: FOXP3 regulatory T cells in the human immune system. Nat Rev Immunol 10:490-500 105. Marshall NA, Christie LE, Munro LR, et al: Immunosuppressive regulatory T cells are abundant in the reactive lymphocytes of Hodgkin lymphoma. Blood 103:1755-1762, 2004 106. Gandhi MK, Lambley E, Duraiswamy J, et al: Expression of LAG-3 by tumor-inltrating lymphocytes is coincident with the suppression of latent membrane antigen-specic CD8 T-cell function in Hodgkin lymphoma patients. Blood 108:2280-2289, 2006 107. Aldinucci D, Lorenzon D, Cattaruzza L, et al: Expression of CCR5 receptors on Reed-Sternberg cells and Hodgkin lymphoma cell lines: Involvement of CCL5/Rantes in tumor cell growth and microenvironmental interactions. Int J Cancer 122:769-776, 2008 108. Tanijiri T, Shimizu T, Uehira K, et al: Hodgkins Reed-Sternberg cell line (KM-H2) promotes a bidirectional differentiation of CD4CD25Foxp3 T cells and CD4 cytotoxic T lymphocytes from CD4 naive T cells. J Leukoc Biol 82:576-584, 2007 109. Kadin ME, Agnarsson BA, Ellingsworth LR, et al: Immunohistochemical evidence of a role for transforming growth factor beta in the pathogenesis of nodular sclerosing Hodgkins disease. Am J Pathol 136:1209-1214, 1990 110. Passwell J, Levanon M, Davidsohn J, et al: Monocyte PGE2 secretion in Hodgkins disease and its relation to decreased cellular immunity. Clin Exp Immunol 51:61-68, 1983 111. Cayeux SJ, Beverley PC, Schulz R, et al: Elevated plasma prostaglandin E2 levels found in 14 patients undergoing autologous bone marrow or stem cell transplantation. Bone Marrow Transplant 12:603-608, 1993 112. Harris SG, Padilla J, Koumas L, et al: Prostaglandins as modulators of immunity. Trends Immunol 23:144-150, 2002 113. Chemnitz JM, Driesen J, Classen S, et al: Prostaglandin E2 impairs CD4 T cell activation by inhibition of lck: Implications in Hodgkins lymphoma. Cancer Res 66:1114-1122, 2006 114. Kim LH, Eow GI, Peh SC, et al: The role of CD30, CD40 and CD95 in the regulation of proliferation and apoptosis in classical Hodgkins lymphoma. Pathology 35:428-435, 2003 115. Walker PR, Saas P, Dietrich PY: Role of Fas ligand (CD95L) in immune escape: The tumor cell strikes back. J Immunol 158:4521-4524, 1997 116. Verbeke CS, Wenthe U, Grobholz R, et al: Fas ligand expression in Hodgkin lymphoma. Am J Surg Pathol 25:388-394, 2001 117. Dotti G, Savoldo B, Pule M, et al: Human cytotoxic T lymphocytes with reduced sensitivity to Fas-induced apoptosis. Blood 105:4677-4684, 2005 118. Gandhi MK, Moll G, Smith C, et al: Galectin-1 mediated suppression of Epstein-Barr virus specic T-cell immunity in classic Hodgkin lymphoma. Blood 110:1326-1329, 2007 119. Juszczynski P, Ouyang J, Monti S, et al: The AP1-dependent secretion of galectin-1 by Reed Sternberg cells fosters immune privilege in classical

Hodgkin lymphoma. Proc Natl Acad Sci U S A 104:13134-13139, 2007 120. Yamamoto R, Nishikori M, Kitawaki T, et al: PD-1-PD-1 ligand interaction contributes to immunosuppressive microenvironment of Hodgkin lymphoma. Blood 111:3220-3224, 2008 121. Green MR, Monti S, Rodig SJ, et al: Integrative analysis reveals selective 9p24.1 amplication, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 116: 3268-3277, 2010 122. Keir ME, Butte MJ, Freeman GJ, et al: PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 26:677-704, 2008 123. Diepstra A, van Imhoff GW, Karim-Kos HE, et al: HLA class II expression by Hodgkin ReedSternberg cells is an independent prognostic factor in classical Hodgkins lymphoma. J Clin Oncol 25: 3101-3108, 2007 124. Huang X, van den Berg A, Gao Z, et al: Expression of HLA class I and HLA class II by tumor cells in Chinese classical Hodgkin lymphoma patients. PLoS One 5:e10865, 2010 125. Poppema S: Immunobiology and pathophysiology of Hodgkin lymphomas. Hematology Am Soc Hematol Educ Program 231-238, 2005 126. Kluiver J, Kok K, Pfeil I, et al: Global correlation of genome and transcriptome changes in classical Hodgkin lymphoma. Hematol Oncol 25:21-29, 2007 127. Diepstra A, Niens M, Vellenga E, et al: Association with HLA class I in Epstein-Barr-virus-positive and with HLA class III in Epstein-Barr-virus-negative Hodgkins lymphoma. Lancet 365:2216-2224, 2005 128. Hjalgrim H, Rostgaard K, Johnson PC, et al: HLA-A alleles and infectious mononucleosis suggest a critical role for cytotoxic T-cell response in EBV-related Hodgkin lymphoma. Proc Natl Acad Sci U S A 107:6400-6405, 2010 129. Niens M, Jarrett RF, Hepkema B, et al: HLA-A*02 is associated with a reduced risk and HLA-A*01 with an increased risk of developing EBV Hodgkin lymphoma. Blood 110:3310-3315, 2007 130. Diepstra A, Poppema S, Boot M, et al: HLA-G protein expression as a potential immune escape mechanism in classical Hodgkins lymphoma. Tissue Antigens 71:219-226, 2008 131. Sa nchez-Espiridio n B, Sa nchez-Aguilera A, Montalba n C, et al: A TaqMan low-density array to predict outcome in advanced Hodgkins lymphoma using parafn-embedded samples. Clin Cancer Res 15:1367-1375, 2009 132. Steidl C, Lee T, Shah SP, et al: Tumorassociated macrophages and survival in classic Hodgkins lymphoma. N Engl J Med 362:875-885, 2010 133. Asano N, Oshiro A, Matsuo K, et al: Prognostic signicance of T-cell or cytotoxic molecules phenotype in classical Hodgkins lymphoma: A clinicopathologic study. J Clin Oncol 24:4626-4633, 2006 134. Oudejans JJ, Jiwa NM, Kummer JA, et al: Activated cytotoxic T cells as prognostic marker in Hodgkins disease. Blood 89:1376-1382, 1997 135. ten Berge RL, Oudejans JJ, Dukers DF, et al: Percentage of activated cytotoxic T-lymphocytes in anaplastic large cell lymphoma and Hodgkins disease: An independent biological prognostic marker. Leukemia 15:458-464, 2001 136. Alvaro-Naranjo T, Lejeune M, Salvado -Usach MT, et al: Tumor-inltrating cells as a prognostic factor in Hodgkins lymphoma: A quantitative tissue

microarray study in a large retrospective cohort of 267 patients. Leuk Lymphoma 46:1581-1591, 2005 137. Kelley TW, Pohlman B, Elson P, et al: The ratio of FOXP3 regulatory T cells to granzyme B cytotoxic T/NK cells predicts prognosis in classical Hodgkin lymphoma and is independent of bcl-2 and MAL expression. Am J Clin Pathol 128:958-965, 2007 138. Alvaro T, Lejeune M, Salvado MT, et al: Outcome in Hodgkins lymphoma can be predicted from the presence of accompanying cytotoxic and regulatory T cells. Clin Cancer Res 11:1467-1473, 2005 139. Tzankov A, Meier C, Hirschmann P, et al: Correlation of high numbers of intratumoral FOXP3 regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkins lymphoma. Haematologica 93:193-200, 2008 140. Chetaille B, Bertucci F, Finetti P, et al: Molecular proling of classical Hodgkin lymphoma tissues uncovers variations in the tumor microenvironment and correlations with EBV infection and outcome. Blood 113:2765-3775, 2009 141. Ree HJ, Kadin ME: Macrophage-histiocytes in Hodgkins disease: The relation of peanutagglutinin-binding macrophage-histiocytes to clinicopathologic presentation and course of disease. Cancer 56:333-338, 1985 142. Sa nchez-Aguilera A, Montalba n C, de la Cueva P, et al: Tumor microenvironment and mitotic checkpoint are key factors in the outcome of classic Hodgkin lymphoma. Blood 108:662-668, 2006 143. Engel M, Essop MF, Close P, et al: Improved prognosis of Epstein-Barr virus associated childhood Hodgkins lymphoma: Study of 47 South African cases. J Clin Pathol 53:182-186, 2000 144. Keegan TH, Glaser SL, Clarke CA, et al: Epstein-Barr virus as a marker of survival after Hodgkins lymphoma: A population-based study. J Clin Oncol 23:7604-7613, 2005 145. Diepstra A, van Imhoff GW, Schaapveld M, et al: Latent Epstein-Barr virus infection of tumor cells in classical Hodgkins lymphoma predicts adverse outcome in older adult patients. J Clin Oncol 27:3815-3821, 2009 146. Jarrett RF, Stark GL, White J, et al: Impact of tumor Epstein-Barr virus status on presenting features and outcome in age-dened subgroups of patients with classic Hodgkin lymphoma: A populationbased study. Blood 106:2444-2451, 2005 147. Kwon JM, Park YH, Kang JH, et al: The effect of Epstein-Barr virus status on clinical outcome in Hodgkins lymphoma. Ann Hematol 85:463-468, 2006 148. Devilard E, Bertucci F, Trempat P, et al: Gene expression proling denes molecular subtypes of classical Hodgkins disease. Oncogene 21:30953102, 2002 149. MacLennan KA, Bennett MH, Tu A, et al: Relationship of histopathologic features to survival and relapse in nodular sclerosing Hodgkins disease: A study of 1659 patients. Cancer 64:1686-1693, 1989 150. Hess JL, Bodis S, Pinkus G, et al: Histopathologic grading of nodular sclerosis Hodgkins disease: Lack of prognostic signicance in 254 surgically staged patients. Cancer 74:708-714, 1994 151. van Spronsen DJ, Vrints LW, Hofstra G, et al: Disappearance of prognostic signicance of histopathological grading of nodular sclerosing Hodgkins disease for unselected patients, 1972-92. Br J Haematol 96:322-327, 1997
2011 by American Society of Clinical Oncology

www.jco.org

13

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Steidl, Connors, and Gascoyne

152. Gobbi PG, Cavalli C, Federico M, et al: Increasing interdependency of prognosis- and therapy-related factors in Hodgkins disease. Acta Haematol 81:34-40, 1989 153. Enblad G, Sundstrom C, Glimelius B: Inltration of eosinophils in Hodgkins disease involved lymph nodes predicts prognosis. Hematol Oncol 11:187-193, 1993 154. Mir R, Anderson J, Strauchen J, et al: Hodgkin disease in patients 60 years of age or older: Histologic and clinical features of advanced-stage diseaseThe Cancer and Leukemia Group B. Cancer 71:1857-1866, 1993 155. Axdorph U, Porwit-MacDonald A, Grimfors G, et al: Tissue eosinophilia in relation to immunopathological and clinical characteristics in Hodgkins disease. Leuk Lymphoma 42:1055-1065, 2001 156. Molin D, Edstrom A, Glimelius I, et al: Mast cell inltration correlates with poor prognosis in Hodgkins lymphoma. Br J Haematol 119:122-124, 2002 157. Canioni D, Deau-Fischer B, Taupin P, et al: Prognostic signicance of new immunohistochemical markers in refractory classical Hodgkin lymphoma: A study of 59 cases. PLoS One 4:e6341, 2009 158. Colby TV, Hoppe RT, Warnke RA: Hodgkins disease: A clinicopathologic study of 659 cases. Cancer 49:1848-1858, 1982 159. Sa nchez-Espiridio n B, Montalba n C, Lo pez A, et al: A molecular risk score based on four functional pathways for advanced classical Hodgkin lymphoma. Blood 116:e12-e17, 2010 160. Farinha P, Masoudi H, Skinnider BF, et al: Analysis of multiple biomarkers shows that lymphomaassociated macrophage (LAM) content is an independent predictor of survival in follicular lymphoma (FL). Blood 106:2169-2174, 2005 161. Schreck S, Friebel D, Buettner M, et al: Prognostic impact of tumour-inltrating Th2 and regulatory T cells in classical Hodgkin lymphoma. Hematol Oncol 27:31-39, 2009 162. Me ne trier-Caux C, Gobert M, Caux C: Differences in tumor regulatory T-cell localization and activation status impact patient outcome. Cancer Res 69:7895-7898, 2009 163. Alvaro T, Lejeune M, Garc a JF, et al: Tumorinltrated immune response correlates with alterations in the apoptotic and cell cycle pathways in Hodgkin and Reed-Sternberg cells. Clin Cancer Res 14:685-691, 2008 164. Alavaikko MJ, Hansmann ML, Nebendahl C, et al: Follicular dendritic cells in Hodgkins disease. Am J Clin Pathol 95:194-200, 1991 165. Delsol G, Meggetto F, Brousset P, et al: Relation of follicular dendritic reticulum cells to Reed-Sternberg cells of Hodgkins disease with emphasis on the expression of CD21 antigen. Am J Pathol 142:1729-1738, 1993 166. Alavaikko MJ, Blanco G, Aine R, et al: Follicular dendritic cells have prognostic relevance in Hodgkins disease. Am J Clin Pathol 101:761-767, 1994 167. Baur AS, Meuge -Moraw C, Michel G, et al: Prognostic value of follicular dendritic cells in nodular sclerosing Hodgkins disease. Histopathology 32:512-520, 1998 168. Glaser SL, Lin RJ, Stewart SL, et al: EpsteinBarr virus-associated Hodgkins disease: Epidemiologic characteristics in international data. Int J Cancer 70:375-382, 1997 169. Enblad G, Sandvej K, Lennette E, et al: Lack of correlation between EBV serology and presence of EBV in the Hodgkin and Reed-Sternberg cells of 14

patients with Hodgkins disease. Int J Cancer 72: 394-397, 1997 170. Morente MM, Piris MA, Abraira V, et al: Adverse clinical outcome in Hodgkins disease is associated with loss of retinoblastoma protein expression, high Ki67 proliferation index, and absence of Epstein-Barr virus-latent membrane protein 1 expression. Blood 90:2429-2436, 1997 171. Warzocha K, Bienvenu J, Ribeiro P, et al: Plasma levels of tumour necrosis factor and its soluble receptors correlate with clinical features and outcome of Hodgkins disease patients. Br J Cancer 77:2357-2362, 1998 172. Weihrauch MR, Manzke O, Beyer M, et al: Elevated serum levels of CC thymus and activationrelated chemokine (TARC) in primary Hodgkins disease: Potential for a prognostic factor. Cancer Res 65:5516-5519, 2005 173. Gause A, Pohl C, Tschiersch A, et al: Clinical signicance of soluble CD30 antigen in the sera of patients with untreated Hodgkins disease. Blood 77:1983-1988, 1991 174. 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 72: 201-206, 1993 175. Christiansen I, Enblad G, Kalkner KM, et al: Soluble ICAM-1 in Hodgkins disease: A promising independent predictive marker for survival. Leuk Lymphoma 19:243-251, 1995 176. Christiansen I, Sundstro m C, Enblad G, et al: Soluble vascular cell adhesion molecule-1 (sVCAM-1) is an independent prognostic marker in Hodgkins disease. Br J Haematol 102:701-709, 1998 177. Ku ppers R, Klein U, Schwering I, et al: Identication of Hodgkin and Reed-Sternberg cellspecic genes by gene expression proling. J Clin Invest 111:529-537, 2003 178. Brune V, Tiacci E, Pfeil I, et al: Origin and pathogenesis of nodular lymphocyte-predominant Hodgkin lymphoma as revealed by global gene expression analysis. J Exp Med 205:2251-2268, 2008 179. Lenz G, Wright G, Dave SS, et al: Stromal gene signatures in large B-cell lymphomas. N Engl J Med 359:2313-2323, 2008 180. Farinha P, Kyle A, Minchinton A, et al: Vascularization predicts overall survival and risk of transformation in follicular lymphoma. Haematologica 95: 2157-2160, 2010 181. Iqbal J, Weisenburger DD, Greiner TC, et al: Molecular signatures to improve diagnosis in peripheral T-cell lymphoma and prognostication in angioimmunoblastic T-cell lymphoma. Blood 115: 1026-1036 182. Fujii K, Kondo T, Yokoo H, et al: Protein expression pattern distinguishes different lymphoid neoplasms. Proteomics 5:4274-4286, 2005 183. Carvalho PC, Carvalho Mda G, Degrave W, et al: Differential protein expression patterns obtained by mass spectrometry can aid in the diagnosis of Hodgkins disease. J Exp Ther Oncol 6:137-145, 2007 184. Navarro A, Gaya A, Martinez A, et al: MicroRNA expression proling in classic Hodgkin lymphoma. Blood 111:2825-2832, 2008 185. Navarro A, Diaz T, Martinez A, et al: Regulation of JAK2 by miR-135a: Prognostic impact in classic Hodgkin lymphoma. Blood 114:2945-2951, 2009 186. Younes A: Novel treatment strategies for patients with relapsed classical Hodgkin lymphoma. Hematology Am Soc Hematol Educ Program 507519, 2009

187. Younes A, Romaguera J, Hagemeister F, et al: A pilot study of rituximab in patients with recurrent, classic Hodgkin disease. Cancer 98:310-314, 2003 188. Oki Y, Pro B, Fayad LE, et al: Phase 2 study of gemcitabine in combination with rituximab in patients with recurrent or refractory Hodgkin lymphoma. Cancer 112:831-836, 2008 189. Schulz H, Rehwald U, Morschhauser F, et al: Rituximab in relapsed lymphocyte-predominant Hodgkin lymphoma: Long-term results of a phase 2 trial by the German Hodgkin Lymphoma Study Group (GHSG). Blood 111:109-111, 2008 190. Rehwald U, Schulz H, Reiser M, et al: Treatment of relapsed CD20 Hodgkin lymphoma with the monoclonal antibody rituximab is effective and well tolerated: Results of a phase 2 trial of the German Hodgkin Lymphoma Study Group. Blood 101:420-424, 2003 191. Younes A, Wong F: Experience with 90Yibritumomab tiuxetan for relapsed classical Hodgkin lymphoma. Ann Oncol 20:1147-1148, 2009 192. Leonard JP, Friedberg JW, Younes A, et al: A phase I/II study of galiximab (an anti-CD80 monoclonal antibody) in combination with rituximab for relapsed or refractory, follicular lymphoma. Ann Oncol 18:1216-1223, 2007 193. Rodig SJ, Abramson JS, Pinkus GS, et al: Heterogeneous CD52 expression among hematologic neoplasms: Implications for the use of alemtuzumab (CAMPATH-1H). Clin Cancer Res 12: 7174-7179, 2006 194. Ansell SM, Horwitz SM, Engert A, et al: Phase I/II study of an anti-CD30 monoclonal antibody (MDX-060) in Hodgkins lymphoma and anaplastic large-cell lymphoma. J Clin Oncol 25:2764-2769, 2007 195. Okeley NM, Miyamoto JB, Zhang X, et al: Intracellular activation of SGN-35, a potent antiCD30 antibody-drug conjugate. Clin Cancer Res 16:888-897, 2010 196. Younes A, Forero-Torres A, Bartlett NL, et al: Multiple complete responses in a phase 1 doseescalation study of the antibody-drug conjugate SGN-35 in patients with relapsed or refractory CD30-positive lymphomas. Presented at the Annual Meeting of the American Society of Hematology, San Francisco, CA, December 6-9, 2008 (abstr 1006) 197. Georgakis GV, Li Y, Humphreys R, et al: Activity of selective fully human agonistic antibodies to the TRAIL death receptors TRAIL-R1 and TRAIL-R2 in primary and cultured lymphoma cells: Induction of apoptosis and enhancement of doxorubicin- and bortezomib-induced cell death. Br J Haematol 130: 501-510, 2005 198. Luqman M, Klabunde S, Lin K, et al: The antileukemia activity of a human anti-CD40 antagonist antibody, HCD122, on human chronic lymphocytic leukemia cells. Blood 112:711-720, 2008 199. Johnston PB, Ansell SM, Colgan JP, et al: MTOR inhibition for relapsed or refractory Hodgkin lymphoma: Promising single agent activity with everolimus (RAD001). Presented at the Annual Meeting of the American Society of Hematology, Atlanta, GA, December 8-11, 2007 (abstr 2555) 200. Zheng B, Georgakis GV, Li Y, et al: Induction of cell cycle arrest and apoptosis by the proteasome inhibitor PS-341 in Hodgkin disease cell lines is independent of inhibitor of nuclear factor-kappaB mutations or activation of the CD30, CD40, and RANK receptors. Clin Cancer Res 10:3207-3215, 2004
JOURNAL OF CLINICAL ONCOLOGY

2011 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

Microenvironment in Hodgkins Lymphoma

201. Younes A, Pro B, Fayad L: Experience with bortezomib for the treatment of patients with relapsed classical Hodgkin lymphoma. Blood 107: 1731-1732, 2006 202. Blum KA, Johnson JL, Niedzwiecki D, et al: Single agent bortezomib in the treatment of relapsed and refractory Hodgkin lymphoma: Cancer and leukemia Group B protocol 50206. Leuk Lymphoma 48:1313-1319, 2007 203. Kirschbaum MH, Goldman BH, Zain JM, et al: Vorinostat (suberoylanilide hydroxamic acid) in relapsed or refractory Hodgkin lymphoma: SWOG 0517. Presented at the Annual Meeting of the American Society of Hematology, Atlanta, GA, December 8-11, 2007 (abstr 2574) 204. Buglio D, Georgakis GV, Hanabuchi S, et al: Vorinostat inhibits STAT6-mediated TH2 cytokine and TARC production and induces cell death in Hodgkin lymphoma cell lines. Blood 112:1424-1433, 2008 205. Younes A, Sureda A, Ben-Yehuda D, et al: Phase II study of oral panobinostat in patients with relapsed/refractory Hodgkin lymphoma (HL) after

high-dose chemotherapy with autologous stem cell transplant. Haematologica 94, 2009 (suppl 2; abstr 34) 206. Fehniger TA, Larson S, Trinkaus K, et al: A Phase II Multicenter Study of Lenalidomide in Patients with Relapsed or Refractory Classical Hodgkin Lymphoma (cHL): Preliminary Results. Presented at the Annual Meeting of the American Society of Hematology, San Francisco, CA, December 6-9, 2008 (abstr 2595) 207. Kuruvilla J, Taylor D, Wang L, et al: Phase II trial of lenalidomide in patients with relapsed or refractory Hodgkin lymphoma. Presented at the Annual Meeting of the American Society of Hematology, San Francisco, CA, December 6-9, 2008 (abstr 3052) 208. Bollard CM, Aguilar L, Straathof KC, et al: Cytotoxic T lymphocyte therapy for Epstein-Barr virus Hodgkins disease. J Exp Med 200:16231633, 2004 209. Bollard CM, Gottschalk S, Leen AM, et al: Complete responses of relapsed lymphoma following genetic modication of tumor-antigen present

ing cells and T-lymphocyte transfer. Blood 110: 2838-2845, 2007 210. Oki Y, Younes A: Current role of gemcitabine in the treatment of Hodgkin lymphoma. Leuk Lymphoma 49:883-889, 2008 211. Correale P, Del Vecchio MT, La Placa M, et al: Chemotherapeutic drugs may be used to enhance the killing efcacy of human tumor antigen peptide-specic CTLs. J Immunother 31:132-147, 2008 212. Rassidakis GZ, Medeiros LJ, Viviani S, et al: CD20 expression in Hodgkin and Reed-Sternberg cells of classical Hodgkins disease: Associations with presenting features and clinical outcome. J Clin Oncol 20:1278-1287, 2002 213. Jones RJ, Gocke CD, Kasamon YL, et al: Circulating clonotypic B cells in classic Hodgkin lymphoma. Blood 113:5920-5926, 2009 214. Bartlett NL, Younes A, Carabasi MH, et al: A phase 1 multidose study of SGN-30 immunotherapy in patients with refractory or recurrent CD30 hematologic malignancies. Blood 111: 1848-1854, 2008

Acknowledgment We thank Sibrand Poppema, MD, PhD, for his many contributions to the eld and for reviewing this manuscript prior to submission.

www.jco.org

2011 by American Society of Clinical Oncology

15

Downloaded from jco.ascopubs.org on November 19, 2013. For personal use only. No other uses without permission. Copyright 2011 American Society of Clinical Oncology. All rights reserved.

You might also like