direct effects of type i interferons on cells of immune system (stubbs et al. 2011)

9
Molecular Pathways Direct Effects of Type I Interferons on Cells of the Immune System Sandra Hervas-Stubbs 1 , Jose Luis Perez-Gracia 2 , Ana Rouzaut 3 , Miguel F. Sanmamed 1,2 , Agnes Le Bon 4,5 , and Ignacio Melero 1,2 Abstract Type I interferons (IFN-I) are well-known inducers of tumor cell apoptosis and antiangiogenesis via signaling through a common receptor interferon alpha receptor (IFNAR). IFNAR induces the Janus activated kinase–signal transducer and activation of transcription (JAK-STAT) pathway in most cells, along with other biochemical pathways that may differentially operate, depending on the responding cell subset, and jointly control a large collection of genes. IFNs-I were found to systemically activate natural killer (NK) cell activity. Recently, mouse experiments have shown that IFNs-I directly activate other cells of the immune system, such as antigen-presenting dendritic cells (DC) and CD4 and CD8 T cells. Signaling through the IFNAR in T cells is critical for the acquisition of effector functions. Cross-talk between IFNAR and the pathways turned on by other surface lymphocyte receptors has been described. Importantly, IFNs-I also increase antigen presentation of the tumor cells to be recognized by T lymphocytes. These IFN-driven immunostimulatory pathways offer opportunities to devise combinatorial immunotherapy strategies. Clin Cancer Res; 17(9); 2619–27. Ó2011 AACR. Background Types of interferon Interferons (IFN) were discovered in 1957 by Isaacs and Lindenmann as soluble proteins able to inhibit virus repli- cation in cell cultures (1). There are 3 types of IFNs: type I (IFN-I), type II (IFN-II), and type III (IFN-III; refs. 2, 3). IFNs belonging to all IFN classes are very important for fighting viral infection. In humans, IFNs-I include IFN-a subtypes, IFN-b, IFN-e, IFN-k, and IFN-w (2). From an immunologic perspective, IFN-a and IFN-b are the main IFN-I subtypes of interest. Both are produced by almost every cell of the body in response to viral infection and share important antiviral properties. The IFN-a family is a multigenic group of highly homologous polypeptides encoded by more than 13 intronless genes in humans. IFN-b, in contrast, exists as a single gene in most species. IFN-g is the only IFN-II. This cytokine does not have marked structural homology with IFNs-I and binds a different cell-surface receptor (IFNGR), also composed of 2 different subunits (IFNGR1 and IFNGR2; ref. 2). The recently classified IFN-III consists of 3 IFN-l molecules (IFN-l1, IFN-l2, and IFN-l3) and sig- nals through a receptor complex consisting of interleukin- 10 receptor 2 subunit (IL-10R2) and IL-28 receptor alpha subunit (IL-28Ra; ref. 3). It is well established that IFNs induce the expression of hundreds of genes, which mediate various biological responses. Some of these genes are regu- lated by the 3 classes of IFNs, whereas others are selectively regulated by distinct IFNs. Although the role of IFN-II and IFN-III is very important in immune responses, IFN-II has not yet shown any clinical activity for human cancer treatment (4), and IFN-III is not currently developed for any indication (3). However, since the discovery of IFNs, the use of IFN-I in therapy has been very widespread. In fact, systemic injections of IFN-I are approved for the treatment of a variety of diseases, which include solid and hematologic malignancies, multiple sclerosis, and, above all, chronic viral hepatitis. In this review, we focus on the role of IFN-I as a direct immunos- timulatory agent directly modifying functions of immune system cells and how these functions can be applied to better therapeutic strategies. IFN-I signaling pathways All types of IFN-I signal through a unique heterodimeric receptor, interferon alpha receptor (IFNAR), composed of 2 subunits, IFNAR1 and IFNAR2, which are expressed in most tissues (5). IFNAR1 knockout mice have not only confirmed the role this subunit plays in mediating the biological response to IFNs-I, but have also established the pleiotropic role that these IFNs play in regulating the host response to viral infections and in adaptive immunity (6–8). Receptor occupancy rapidly triggers several signaling cascades (Fig. 1), which culminate in the transcriptional regulation of hundreds of IFN-stimulated genes (ISG). The Authors' Affiliation: 1 Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, 2 Medical Oncology Department and Cell Therapy Unit, University Clinic, University of Navarra, and 3 Division of Oncology, CIMA, University of Navarra, Pamplona, Spain; 4 Institut Cochin, Universit e Paris Descartes, CNRS (UMR 8104), and 5 INSERM, U1016, Paris, France Corresponding Author: Ignacio Melero, CIMA and Facultad de Medicina Universidad de Navarra, Av. Pio XII, 57, 31008 Pamplona, Spain. Phone: 34-948194700; Fax: 34-948194717; E-mail: [email protected] or msher- [email protected] doi: 10.1158/1078-0432.CCR-10-1114 Ó2011 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 2619

Upload: gdhbar

Post on 07-Nov-2015

214 views

Category:

Documents


0 download

DESCRIPTION

Effects of Interferons of Type I on cells of the immune system.

TRANSCRIPT

  • Molecular Pathways

    Direct Effects of Type I Interferons on Cells of the Immune System

    Sandra Hervas-Stubbs1, Jose Luis Perez-Gracia2, Ana Rouzaut3, Miguel F. Sanmamed1,2,Agnes Le Bon4,5, and Ignacio Melero1,2

    AbstractType I interferons (IFN-I) are well-known inducers of tumor cell apoptosis and antiangiogenesis via

    signaling through a common receptor interferon alpha receptor (IFNAR). IFNAR induces the Janus

    activated kinasesignal transducer and activation of transcription (JAK-STAT) pathway in most cells,

    along with other biochemical pathways that may differentially operate, depending on the responding cell

    subset, and jointly control a large collection of genes. IFNs-I were found to systemically activate natural

    killer (NK) cell activity. Recently, mouse experiments have shown that IFNs-I directly activate other cells of

    the immune system, such as antigen-presenting dendritic cells (DC) and CD4 and CD8 T cells. Signaling

    through the IFNAR in T cells is critical for the acquisition of effector functions. Cross-talk between IFNAR

    and the pathways turned on by other surface lymphocyte receptors has been described. Importantly, IFNs-I

    also increase antigen presentation of the tumor cells to be recognized by T lymphocytes. These IFN-driven

    immunostimulatory pathways offer opportunities to devise combinatorial immunotherapy strategies. Clin

    Cancer Res; 17(9); 261927. 2011 AACR.

    Background

    Types of interferonInterferons (IFN) were discovered in 1957 by Isaacs and

    Lindenmann as soluble proteins able to inhibit virus repli-cation in cell cultures (1). There are 3 types of IFNs: type I(IFN-I), type II (IFN-II), and type III (IFN-III; refs. 2, 3). IFNsbelonging to all IFN classes are very important for fightingviral infection. In humans, IFNs-I include IFN-a subtypes,IFN-b, IFN-e, IFN-k, and IFN-w (2). From an immunologicperspective, IFN-a and IFN-b are themain IFN-I subtypes ofinterest. Both are produced by almost every cell of the bodyin response to viral infection and share important antiviralproperties. The IFN-a family is amultigenic group of highlyhomologous polypeptides encoded by more than 13intronless genes in humans. IFN-b, in contrast, exists as asingle gene in most species. IFN-g is the only IFN-II. Thiscytokine does not have marked structural homology withIFNs-I and binds a different cell-surface receptor (IFNGR),also composed of 2 different subunits (IFNGR1 andIFNGR2; ref. 2). The recently classified IFN-III consists of

    3 IFN-l molecules (IFN-l1, IFN-l2, and IFN-l3) and sig-

    nals through a receptor complex consisting of interleukin-10 receptor 2 subunit (IL-10R2) and IL-28 receptor alphasubunit (IL-28Ra; ref. 3). It is well established that IFNsinduce the expression of hundreds of genes, which mediatevarious biological responses. Some of these genes are regu-lated by the 3 classes of IFNs, whereas others are selectivelyregulated by distinct IFNs.Although the role of IFN-II and IFN-III is very important

    in immune responses, IFN-II has not yet shown any clinicalactivity for human cancer treatment (4), and IFN-III is notcurrently developed for any indication (3). However, sincethe discovery of IFNs, the use of IFN-I in therapy has beenvery widespread. In fact, systemic injections of IFN-I areapproved for the treatment of a variety of diseases, whichinclude solid and hematologic malignancies, multiplesclerosis, and, above all, chronic viral hepatitis. In thisreview, we focus on the role of IFN-I as a direct immunos-timulatory agent directly modifying functions of immunesystem cells and how these functions can be applied tobetter therapeutic strategies.

    IFN-I signaling pathwaysAll types of IFN-I signal through a unique heterodimeric

    receptor, interferon alpha receptor (IFNAR), composed of 2subunits, IFNAR1 and IFNAR2, which are expressed inmost tissues (5). IFNAR1 knockout mice have not onlyconfirmed the role this subunit plays in mediating thebiological response to IFNs-I, but have also establishedthe pleiotropic role that these IFNs play in regulating thehost response to viral infections and in adaptive immunity(68).Receptor occupancy rapidly triggers several signaling

    cascades (Fig. 1), which culminate in the transcriptionalregulation of hundreds of IFN-stimulated genes (ISG). The

    Authors' Affiliation: 1Division of Gene Therapy andHepatology, Centre forApplied Medical Research (CIMA), University of Navarra, Pamplona,2Medical Oncology Department and Cell Therapy Unit, University Clinic,University of Navarra, and 3Division of Oncology, CIMA, University ofNavarra, Pamplona, Spain; 4Institut Cochin, Universite Paris Descartes,CNRS (UMR 8104), and 5INSERM, U1016, Paris, France

    Corresponding Author: Ignacio Melero, CIMA and Facultad de MedicinaUniversidad de Navarra, Av. Pio XII, 57, 31008 Pamplona, Spain. Phone:34-948194700; Fax: 34-948194717; E-mail: [email protected] or [email protected]

    doi: 10.1158/1078-0432.CCR-10-1114

    2011 American Association for Cancer Research.

    ClinicalCancer

    Research

    www.aacrjournals.org 2619

  • first signaling pathway shown to be activated by IFNs-I wasthe Janus activated kinasesignal transducer and activationof transcription (JAK-STAT) pathway (Fig. 1A; ref. 9), whichis active in almost all cell types. IFNAR1 is constitutivelyassociated with tyrosine kinase 2 (TYK2), whereas IFNAR2is associated with JAK1. Receptor binding results in trans-phosphorylation of JAK1 and TYK2. The activated JAKsthen phosphorylate conserved tyrosine residues in thecytoplasmic tails of the IFNAR. These phosphotyrosyl resi-dues subsequently serve as docking sites for the recruitmentof src-homology 2 (SH2)containing signaling molecules,such as STAT1 and STAT2. Once at the receptor, the STATproteins themselves become JAK substrates. STAT1 and

    STAT2 are both phosphorylated on a single tyrosine(Y701 for STAT1 and Y690 for STAT2). Once phosphory-lated, STATs dimerize and move to the nucleus, where theyassociate with the IFN-regulatory factor 9 (IRF9) to formthe ISG factor 3 (ISGF3) transcription factor complex,which binds to IFN-stimulated response elements.Although STAT1 and STAT2 are the most important

    mediators of the response to IFNs-I, other STATs, namelySTAT3 and STAT5, have been found to be phosphorylated(activated) by IFNs-I (Fig. 1A; refs. 1013). STAT4and STAT6 can also be activated by IFN-a; however, thisactivation seems to be restricted to certain cell types, suchas endothelial or lymphoid cells (1417). Following

    2011 American Association for Cancer Research

    Tyk2

    IFN-/(A) JAK-STAT

    STAT

    STAT STAT

    STAT

    STAT3

    STAT3

    C3G

    IRF9

    IRF9

    ISG15, IP-10,IRF-7, PKR,2-5-OAS,etc

    IRF-1, IRF-2,IRF-8, IRF-9,etc

    Regulation of expression of GTP-binding protein Ag processing/presentation proteinSurvival signals

    Apoptosis Cellular growthDifferentiationSer phosphorylationof STAT1

    c-MycSAP-1P53SP1SMADs

    NFATEtsElk-1MEF2Stat1/3

    JunElk-1NFATATF2

    Ras

    Raf1

    MEK1/2

    ERK1/2JNKp38

    Rac1Rap1

    ProliferationDifferentiationSurvival....

    mTORInhibition ofGSK-3/CDKN1A/CDKN1B

    PKCIKKIKK

    NFB

    MEF2MAPKAPRsk

    FosElk-1Sap-1

    ISG15Histone acetylationAntiviral effectGrowth inhibitory effectsHematopoietic suppressive signals

    STAT1

    STAT5

    STAT5

    CRKL

    CRKL

    CRKL

    STAT2

    STAT1

    STAT2

    VAV

    JAK1 JAK1 JAK1 JAK1

    VAV

    VAV

    STAT1, STAT3STAT4, STAT5STAT6

    STAT1/2,3,4 or 5STAT2/3STAT5/6

    STATSTAT

    IFNAR

    1IFN

    AR2

    IFN-/(B) CRK

    IFN-/ IFN-/ IFN-/

    Type I IFNs signaling pathways(C) PI3K and NFB

    TCR complexCD45

    CD4/8

    (D) MAPK

    IFNAR

    1IFN

    AR2

    Homodimers

    STAT STAT

    STAT STAT

    Homodimers

    Heterodimers

    Heterodimers

    ISRE GAS GAS

    Regulatedgenes orfunctions

    NFB Rel ... ... ...

    Tyk2

    IFNAR

    1IFN

    AR2

    IFNAR

    1IFN

    AR2

    IFNAR

    1IFN

    AR2

    Tyk2

    NIK

    Tyk2

    RS1/2PI3K

    AKT

    Zap70

    Lck

    LA

    TSLP76TRAFSTAT

    5C3G

    Figure 1. Signaling pathways activated by the IFN-I receptor. Schematic of signaling pathways downstream of the IFNAR, emphasizing cross-talk withdifferent signaling cascades, which depends on the identity and activation status of the cell responding to IFN-I. A, JAK-STAT signal pathway activated byIFNs-I. B, CRKL pathway. Upon JAK activation, CRKL associates with TYK2 and undergoes tyrosine phosphorylation. The activated form of CRKLforms a complex with STAT5, which also undergoes TYK2-dependent tyrosine phosphorylation. The CRKLSTAT5 complex translocates to the nucleusand binds specific GAS elements. C, PI3K and NF-kB pathways. Phosphorylation of TYK2 and JAK1 results in activation of PI3K and AKT, whichin turn mediates downstream activation of mTOR, inactivation of GSK-3, CDKN1A, and CDKN1B, and activation of IKKb, which results in activation ofNF-kB. IFNs-I also activate NF-kB by an alternative pathway that involves the linkage of TRAFs, which results in activation of NF-kB2. PI3K/AKT can alsoactivate NF-kB through an activation loop via PKCq. D, MAPK pathway. Activation of JAK results in tyrosine phosphorylation of Vav, which leads to theactivation of several MAPKs such as p38, JNK, and ERK1/2. The different MAPKs activate different sets of transcription factors, such as Fos, ELK1, Sap-1,MEF2, MAPKAP, and Rsk (activated by p38); Jun, ELK1, NFAT, and ATF2 (activated by JNK); andNFAT, ETS, ELK1,MEF2, STAT1/2, c-Myc, SAP-1, p53, SP1,and SMADs (activated by ERK1/2).

    Hervas-Stubbs et al.

    Clin Cancer Res; 17(9) May 1, 2011 Clinical Cancer Research2620

  • phosphorylation, activated STATs form homo- (STAT1,STAT3, STAT4, STAT5, and STAT6) or heterodimers(STAT1/2, STAT1/3, STAT1/4, STAT1/5, STAT2/3, andSTAT5/6; refs. 10, 11, 18, 19), which translocate to thenucleus and bind other regulatory sequences known asIFN-gactivated sites (GAS).Interestingly, distinct STATs have opposing biological

    effects. Thus, STAT3 stimulates growth, whereas on thecontrary, STAT1 is a growth inhibitor. On the other hand,IFNs-Imediated activation of STAT4 is required for IFN-gproduction, whereas surprisingly, STAT1 negatively regu-lates the IFNs-Idependent induction of IFN-g (20). Therelative abundance of these dimeric transcription factors,which may vary substantially depending on cell type andactivation and/or differentiation state, is likely to have amajor impact on the overall response to IFNs-I (2123).Stimulation of the IFNAR/JAK/STAT1 pathway can also

    lead to the upregulation of the TAM receptor Axl, whichaccumulates at the cell surface where it physically associateswith the IFNAR and usurps the IFNAR-STAT1 pathway bystimulating the transcription of suppressors of cytokinesignaling (SOCS; refs. 24, 25). The SOCS proteins extin-guish IFN-I signaling in a negative feedback loop. Forinstance, SOCS1 directly interacts with JAKs blocking theircatalytic activity, whereas SOCS3 seems to inhibit JAKs aftergaining access by receptor binding. In addition, SOCSproteins interact with the cellular ubiquitination machin-ery through the SOCS-box region and may redirect asso-ciated proteins, such as JAKs or IFNAR chains, toproteasomal degradation. It has been shown that SOCS1knockout mice develop lupus-like disease symptoms (26)and that SOCS1 silencing enhances the antitumor activityof IFNs-I (27), suggesting that SOCS are key elements inkeeping IFNs-I effects under control.

    Alternative signaling pathways and cross-talk withother receptorsEvidence is accumulating that several other signaling

    elements and cascades are required for the generation ofmany of the responses to IFNs-I, such as the v-crk sarcomavirus CT10 oncogene homolog (avian)-like (CRKL) path-way, the mitogen activated protein kinase (MAPK) path-way, the phosphoinositide 3-kinase (PI3K) pathway, andeither the classical or the alternative NF-kB cascade(Fig. 1B-D; refs. 9, 28). Some of these pathways operateindependently of the JAK-STAT, whereas others cooperatewith STATs to tune the response to IFNs-I. Interestingly,MAPK, PI3K, and NF-kB pathways involve immunorecep-tor tyrosine-based activation motives used by classicalimmunoreceptors, suggesting a cross-talk among IFNARand multiple receptors on immune system cells.The PI3K and NF-kB pathways. Upon activation, TYK2

    and JAK1 phosphorylate insulin receptor substrate 1 (IRS1)and 2 (IRS2), which provide docking sites for the PI3K (9).STAT3 acts as an adapter to couple PI3K to the IFNAR1subunit. PI3K subsequently activates the serine-threoninekinase AKT, which in turn mediates downstream the acti-vation of mTOR and the inactivation of several proteins,

    such as glycogen synthase kinase 3 (GSK-3) and cyclindependent kinase inhibitors 1A and 1B (CDKN1A andCDKN1B), all of them involved in the control of celldivision and proliferation. The AKT pathway also resultsin the activation of IkB kinase beta (IKKb), which givesrise to NF-kB activation. IFNs-I also activate NF-kB by analternative pathway, which involves the linkage of TNFreceptorassociated factors (TRAF) to the activation ofthe NF-kBinducing kinase (NIK), which in turn resultsin activation of NF-kB2. This alternative pathway isstrictly dependent on the activity of IKKa. In addition,PI3K/AKT can also activate NF-kB through an activationloop via protein kinase C zeta (PKCq). Overall, NF-kBactivated by IFNs-I regulates prosurvival signals andenhances the expression of several GTP-binding proteinsas well as molecules involved in antigen processing and/or presentation.The MAPK pathway. Activation of JAK results in tyro-

    sine phosphorylation of the guanine nucleotide exchangefactor Vav, which leads to downstream activation of Ratsarcoma protein (Ras) and ras-related C3 botulinum toxinsubstrate 1 (Rac1), which subsequently leads to the activa-tion of several MAPKs: p38, c-Jun N-terminal kinases(JNK), and extracellular signal regulated kinases (ERK) 1and 2 (9). Vav can also activate PKCq, leading to theactivation of theNF-kB cascade. Several studies have shownthat IFN-amediated activation of ERK1/2 in T lympho-cytes requires proteins involved in the T-cell receptor (TCR)early signaling complex, such as CD45, lymphocyte-speci-fic protein tyrosine kinase (Lck), Zeta-chain-associatedprotein kinase 70 (Zap70), SH2 domain-containing leu-kocyte protein of 76 kDa (SLP76), and Vav1, and possiblylinker for activation of T cells (LAT), indicating cross-talkbetween pathways (2931). Moreover, it has been shownrecently that TCR deletion in Jurkat cells abrogated IFNAR-stimulated MAPK activity, whereas the canonical JAK-STATpathway remained unaffected (31). A hypothetical path-way that involves all these molecules and results in Vavactivation is depicted in Fig. 1D. The different MAPKsactivate different sets of transcription factors. Importantly,p38 functions are essential for the antiviral and antileuke-mic properties of IFNs-I, it plays a role in the hematopoie-tic-suppressive signals and is required for the growth-inhibitory effects of IFNs-I observed in certain lymphocytecultures. IFNs-Iactivated ERK cascades regulate cellulargrowth, differentiation, and serine phosphorylation ofSTAT1. The search for biochemical signals to explain theeffects of IFN-I on immune system cells is far from over.

    Effects of IFNs-I on cells of the immune systemIt has long been established that the main mechanism

    accounting for the efficacy of the IFN-Ibased therapies wasdue to their direct effect on malignant or virus-infectedcells. In fact, IFNs-I directly inhibit the proliferation oftumor and virus-infected cells and increase MHC class Iexpression, enhancing antigen recognition. Moreover,IFNs-I repress oncogene expression and induce that oftumor suppressor genes, which may contribute to the

    IFN-a on Immune Cells

    www.aacrjournals.org Clin Cancer Res; 17(9) May 1, 2011 2621

  • inhibitory effects of IFN-I on malignant cells in conjunc-tion with the antiangiogenic effects.IFNs-I have also proven to be involved in immune

    system regulation (7, 8, 32). IFNs-I exert their effects onimmune cells either directly, through IFNAR triggering, orindirectly (i) by the induction of chemokines, which allowthe recruitment of immune cells to the site of infection; (ii)by the secretion of a second wave of cytokines, which couldfurther regulate cell numbers and activities (as for exampleIL-15, which plays a critical role in proliferation andmaintenance of NK cells and memory CD8 T cells; refs.33, 34); or (iii) by the stimulation of other cell types criticalfor the activation of certain immune cells, such as DC forthe activation of nave T cells.One of the earliest described immunoregulatory func-

    tions of IFNs-I was their ability to regulate NK functions(32). IFNs-I enhance the ability of NK cells to kill targetcells and to produce IFN-g by indirect and direct mechan-isms (33, 35, 36). Furthermore, IFNs-I promote the accu-mulation and/or survival of proliferating NK cells by theIFN-I/STAT1dependent induction of IL-15 (33). IFNs-Ialso enhance the production or secretion of other cytokinesby the NK cell through the autocrine IFN-g loop (37).IFNs-I also affect monocyte and/or macrophage function

    and differentiation. Thus, IFNs-I markedly support thedifferentiation of monocytes into DC with high capacityfor Ag presentation, stimulate macrophage antibody-dependent cytotoxicity, and positively or negatively regu-late the production of various cytokines (e.g., TNF, IL-1, IL-6, IL-8, IL-12, and IL-18) bymacrophages (38). In addition,autocrine IFN-I is required for the enhancement of macro-phage phagocytosis by macrophage colony-stimulatingfactor and IL-4 (39) and for the lipopolysaccharide-,virus-, and IFN-ginduced oxidative burst through thegeneration of nitric oxide synthase 2.The main function of DCs is to uptake and process

    antigens for presentation to T cells. DCs are a unique celltype able to prime nave T cells and, therefore, are criticalantigen presenting cells (APC). IFNs-I have multiple effectson DCs, affecting their differentiation, maturation, andmigration. Thus, human monocytes cultured in granulo-cyte-macrophage colony-stimulating factor (GM-CSF) IFN-a/b, rather than the more commonly used combina-tion of GM-CSF IL-4, differentiatemore rapidly into DCs,exhibiting the phenotype of partially mature DCs, whileshowing a strong capability to induce a primary humanantibody response and CTL expansion when pulsed withantigen and injected into humanized severe combinedimmunodeficiency (SCID) mice (4042).

    In vitro treatment of immature conventional DCs withIFN-a/b has been shown to upregulate surface expressionof MHC class I, class II, CD40, CD80, CD86, and CD83molecules in the human system (4345), associated with aheightened capacity to induce CD8 T-cell responses.Accordingly, it has been observed in mice that IFN-a actingon DC plays a key role in achieving efficient cross-primingof antigen-specific CD8 T lymphocytes (7, 8, 32). IFNs-Ialso affect the ability of DC to secrete IL-12p70. Low

    concentrations of IFNs-I are essential for the optimal pro-duction of IL-12p70 (46), whereas higher levels of IFN-Isuppress IL-12p40 expression, thus dampening IL-12p70production (47).A key requisite to initiate the adaptive immune response

    is the arrival of professional APC from infected tissue intolymph nodes. It has been shown that human DCs differ-entiated from monocytes in the presence of IFN-a exhibitupregulation of CC chemokine receptor type 7 (CCR7),correlating with an enhanced chemotactic response in vitroto CCR7 natural ligand (CCL19) and with strongmigratorybehavior in SCID mice (48). Interestingly, experiments inmice have also shown that IFN-a/b is required for plasma-cytoid DCs (pDC) to migrate from the marginal zone intothe T-cell area, where they form clusters (49). We have alsorecently shown that IFNs-I enhance the adhesion andextravasations of DCs across inflamed lymphaticendothelium in a lymphocyte function-associated anti-gen 1 (LFA-1) and very late antigen-4 (VLA4)depen-dent fashion (50). IFNs-I may also favor the encounterbetween DCs and specific lymphocytes in lymph nodesby promoting lymphocyte trapping in lymph nodes upondownmodulation of sphingosine 1-phosphate (51).IFNs-I have also been shown to potently enhance the

    primary antibody response to soluble antigen, stimulatingthe production of all subtypes of immunoglobulin G (IgG),and inducing long-lived antibody production and immu-nologic memory (8). Direct effects of IFN-a on DCs (8), Bcells (52, 53), and CD4 T cells (53) all contribute to itsadjuvant activities on humoral immune responses. Inter-estingly, a direct effect of IFNs-I on B cells seems to becrucial for the development of local humoral responsesagainst viruses (54).Asmentioned above, CD4 T cells are direct targets of IFN-

    I in the enhancement of antibody responses (53). Inhumans, it has also been described that direct effects ofIFN-I on naive CD4 T cells favor their differentiation intoIFN-gsecreting Th1-like T cells (55).Several studies suggested that IFNs-I might exert a direct

    effect on CD8 T cells. Thus, it was reported that IFNs-Ipromote IFN-g production by CD8 T cells in a STAT4-dependent manner (56) and promote survival of CD8 Tcells from wild-type (WT) but not IFNAR-deficient(IFNAR/) mice (57). The definitive report showing thatCD8 T cells represent direct targets of IFN-Imediatedstimulation in vivo came from Kolumam and colleagues(58). By adoptively transferring virus-specific naive CD8 Tcells from IFNAR/ or IFNAR-sufficient mice into normalIFNAR WT hosts, IFNs-I were shown to act directly onmurine CD8 T cells, allowing their clonal expansion andmemory differentiation. Subsequent studies confirmed thiswork (5961). Elegant experiments in mice by Curtsingerand colleagues (62) have shown that, in addition to signalsvia TCR (signal-1) and CD28 (signal-2), nave CD8 T cellsrequired a third signal to differentiate into effector cells.cDNA microarray analyses showed that IFN-a couldregulate critical genes involved in CTL functions (63),providing evidence that IFNa promoted activation and

    Hervas-Stubbs et al.

    Clin Cancer Res; 17(9) May 1, 2011 Clinical Cancer Research2622

  • differentiation of CD8 T cells by sustaining the expressionof T-bet and Eomes through chromatin remodeling.Recently, we have shown that IFN-a provides a strongand direct signal to human CD8 T cells, thereby resultingin upregulation of critical genes for cytotoxic T-cell activity(cytolysis and IFN-g secretion) and for the production ofchemokines that would coattract other effector lympho-cytes. IFN-a was absolutely critical in the case of humannave CD8 T cells for effector function acquisition (64). Inmany instances, T cells may sense IFN-I before the cognateantigen is actually presented to them. As a result of apreexposure to IFN-I, CD8 T cells become preactivatedand acquire much faster effector functions upon antigenrecognition (65). This preactivation may reflect the upre-gulation by IFN-I of several mRNAs in antigen-naive CD8 Tlymphocyte, albeit without changes in the expression of thecorresponding protein, unless antigen-specific activationensues (64).

    Clinical-Translational Advances

    The contracting current indications of IFN-a as ananticancer agentIFN-a has received approval for treatment of several

    neoplastic diseases (66). The most frequent indicationsfor IFN-a are chronic viral hepatitis. For the treatment ofthese infectious conditions, stabilized forms of IFN-a havebeen created by directed conjugation of polyethylene gly-col, which, by increasing molecular weight, retards renalclearance and, thereby, extends plasma concentrations withsustained receptor occupancy (67). However, PEG-conju-gated IFN is not widely used in oncology in the absence ofcomparative clinical studies with the unconjugated form.In oncology, the main indication of IFN-a is for patients

    with resected stage II and III melanoma, in whom IFN-aprolongs disease-free survival and shows a trend towardincreased overall survival (68). However, the advent ofCTLA-4 antagonist antibodies will very likely displacethe use of IFN-a for melanoma therapy (69).Table 1 shows the spectrumofneoplastic diseases inwhich

    IFN-a has been used. As shown, its role in cancer therapy issteadily decreasing because of its limited efficacy and theadvent of new, more effective, and/or safer treatments.

    Future perspectivesIFNs-I are powerful tools to directly and indirectly

    modulate the functions of the immune system. Evolutionhas shaped these cytokines as a key sign of alarm in caseof viral infection. The type of immune response that wewould like to induce and sustain against cancer antigensis identical to the one we commonly observe upon acuteviral infections. Obviously, the effects of IFN-I are inte-grated with many other key molecules that need to act in aconcerted fashion.As described in Table 1, side effects of systemic long-

    term treatments and lack of sufficiently high efficacy havedampened the interest of IFN-a for clinical use in oncol-ogy. However, we believe that IFN-a is likely to be more

    efficacious when acting locally and intermittently at themalignant tissue and tumor-draining lymph node thanwhen administered to achieve systemic bioavailability.Local delivery can be achieved with pharmaceutical for-mulations and gene therapy approaches (70). In ouropinion, the clinical use of recombinant IFN-a as a vaccineadjuvant should be reconsidered and new investigationscarried out with an eye to clinical applications (71).The rationale for using intermittent delivery arises from

    observations indicating that IFN-I turns on signaling desen-sitizing mechanisms (i.e., because of SOCS1 induction).Intermittency at an optimized pace may help to avoid thesenegative feedback mechanisms in the responding immunecells. These ideas contrast with stabilized formulations,such as the widely used polyethylene glycol-conjugatedIFN-a and fusion proteins with albumin that extend thehalf-life of the cytokine.It is of much interest that a gene expression signature

    induced by IFN-I in melanoma lesions predicts benefitfrom vaccines and may have a key role in recruitingeffector T cells to tumors by inducing chemokine genes(72). IFN-a has been recently used in promising combi-natorial immunotherapy approaches alongside DC vac-cination for glioblastoma, precisely to promote T-cellinfiltration and DC activation (73). In this regard, usingendogenous IFN-I inducers instead of recombinant cyto-kines may have advantages, because substances such asthe viral RNA analog poly I:C will also elicit other cyto-kines that may act in concert with IFNs-I to enhance theantitumor immune response.Creative combination strategies are building on the

    knowledge that IFN-a effects on the immune systemare likely to improve its therapeutic profile against malig-nant diseases. For instance, combinations with immu-nostimulatory monoclonal antibodies, IL-15, IL-12, andIL-21, may hold much promise. Indeed, intratumoralrelease of mouse IFN-a along with systemic treatmentwith anti-CD137 mAb results in synergistic therapeuticeffects (74). Macrophages homing to tumor, transducedto express IFN-a, exert therapeutic effects without thesystemic side effects (75). These studies suggest that localdelivery as opposed to systemic administration should betested. Of great importance is the notion that the che-mokines induced by IFN-I (i.e., CXCL9, CXCL10) attractactivated lymphocytes, thereby calling more immunecells to infiltrate the malignant focus.When reflecting on the use of IFN-I for cancer, we must

    think about reprogramming the tumor microenviron-ment, frequently devoid of any stimulatory signals. LocalIFN-I release with concomitant irradiation and chemo-therapy may help to turn some of the macroscopic tumorlesions into immunogenic vaccines. Factors such as CpGoligonucleotides or poly I:C that induce the release ofendogenous IFNs-I at a given tumor lesion (76) arepromising in this regard. These agents may constitutean advantageous alternative because these agents alsoinduce an additional array of proinflammatory mediatorsacting in synergy with IFNs-I.

    IFN-a on Immune Cells

    www.aacrjournals.org Clin Cancer Res; 17(9) May 1, 2011 2623

  • Table 1. Contracting Indications of IFN-a for Malignant Conditions

    Indication Stage Comment Standard Current Therapy Reference

    Melanoma II/III Prolongs disease-freesurvival and shows a trendtoward increased overallsurvival.

    IFN-a is the standard option. 68

    IV Combination withIL2: Increased responserates but no survivalimprovement.

    Chemotherapy. The adventof CTLA-4 antagonist antibodywill displace the use of IFN-afor melanoma inmunotherapy.

    77, 78

    RCC II/III Two phase III trials showednegative results.

    Novel targeted agents suchas sunitinib, temsirolimus,sorafenib, or everolimushave decreased the use ofIFN-a to a minimum in bothresectable and advanced RCC.

    79, 80

    IV Widely studied as a singleagent or in combination withIL-2, chemotherapy, or both:modest or negative results.

    81

    Hairy cell leukemia First treatment that showedclinical benefit in this disease.

    Purine analogs are nowconsidered standardtreatment for thisdisease.

    82, 83

    Recommended, in lowdose, as a therapeuticoption for maintenancetherapy and as analternative for frailpatients.

    Multiple myeloma Extensively studied asmaintenance treatment,as single treatment, or incombination withchemotherapy: slightefficacy.

    Largely replaced by newerand more effective agentssuch as thalidomide,lenalidomide, or bortezomib.

    84

    CMS PV Optional treatment forcytoreduction (94.6%complete responses).

    The mainstay of therapyremains repeated phlebotomy.

    8587

    ET Treatment of choicein women withchildbearing potentialand optional treatmentin young patientsresistant tohydroxyurea.

    Hydroxyurea. Anagrelide. 86

    PM Cytoreduction therapy. Hydroxyurea. Thalidomide. 77CML First-line therapy in

    combination withcytarabine: 71% clinicalresponse rate and 39% majorcytogenetic response rate.

    Imatinib, in a phase III trial,has been superior intolerability, completehematologic and cytogeneticresponse rates, andprogression-free survival.

    8892

    (Continued on the following page)

    Hervas-Stubbs et al.

    Clin Cancer Res; 17(9) May 1, 2011 Clinical Cancer Research2624

  • In conclusion, the notion of key direct effects of IFNs-Ion immune system cells and detailed knowledge on theelicited signaling pathways should help biomarker discov-ery to identify the small number of patients who couldactually benefit from current treatments. More importantly,these ideas might change the way we use IFN-a for cancertherapy, suggesting new strategies and combinations withother agents.

    Disclosure of Potential Conflicts of Interest

    I. Melero, A. Rouzaut, and S. Hervas-Stubbs receive laboratory reagentsand research funding fromDigna Biotech. I. Melero has acted as a consultantfor Bristol-Myers Squibb and Pfizer Inc.

    Acknowledgments

    We are grateful for continuous scientific collaboration on IFNs-I withJesus Prieto, Esther Larrea, Jose I. Riezu-Boj, Iranzu Gonzalez, and Juan Ruiz.

    Grant Support

    MEC/MCI (SAF200503131, SAF200803294, and TRA20090030), Departa-mento de Educacion y Departamento de Salud (Beca Ortiz de Landazuri) delGobierno de Navarra. Redes tematicas de investigacion cooperativa RETIC(RD06/0020/0065), Fondo de investigacion sanitaria (FIS PI060932), Europeancommission VII framework program (ENCITE), Immunonet SUDOE. FundacionMutua Madrilea, and "UTE for project FIMA". S. Hervas-Stubbs was supportedby AECC and by MCI (RYC-200700928).

    Received October 22, 2010; revised January 17, 2011; accepted January17, 2011; published OnlineFirst March 3, 2011.

    References1. Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R

    Soc Lond B Biol Sci 1957;147:25867.2. Pestka S, Krause CD, Walter MR. Interferons, interferon-like cyto-

    kines, and their receptors. Immunol Rev 2004;202:832.3. Vilcek J. Novel interferons. Nat Immunol 2003;4:89.4. Miller CH, Maher SG, Young HA. Clinical Use of Interferon-gamma.

    Ann N Y Acad Sci 2009;1182:6979.5. deWeerd NA, Samarajiwa SA, Hertzog PJ. Type I interferon receptors:

    biochemistry and biological functions. J Biol Chem 2007;282:200537.

    6. Hwang SY, Hertzog PJ, Holland KA, Sumarsono SH, Tymms MJ,Hamilton JA, et al. A null mutation in the gene encoding a type Iinterferon receptor component eliminates antiproliferative and anti-viral responses to interferons alpha and beta and alters macrophageresponses. Proc Natl Acad Sci U S A 1995;92:112848.

    7. Le Bon A, Etchart N, Rossmann C, Ashton M, Hou S, Gewert D, et al.Cross-priming of CD8 T cells stimulated by virus-induced type Iinterferon. Nat Immunol 2003;4:100915.

    8. Le Bon A, Schiavoni G, DAgostino G, Gresser I, Belardelli F, ToughDF. Type i interferons potently enhance humoral immunity and canpromote isotype switching by stimulating dendritic cells in vivo.Immunity 2001;14:46170.

    9. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediatedsignalling. Nat Rev Immunol 2005;5:37586.

    10. Aaronson DS, Horvath CM. A road map for those who don't knowJAK-STAT. Science 2002;296:16535.

    11. Darnell JE Jr. STATs and gene regulation. Science 1997;277:16305.12. Darnell JE Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcrip-

    tional activation in response to IFNs and other extracellular signalingproteins. Science 1994;264:141521.

    13. Meinke A, Barahmand-Pour F, Wohrl S, Stoiber D, Decker T. Activa-tion of different Stat5 isoforms contributes to cell-type-restrictedsignaling in response to interferons. Mol Cell Biol 1996;16:693744.

    14. Farrar JD, Smith JD, Murphy TL, Murphy KM. Recruitment of Stat4 tothe human interferon-alpha/beta receptor requires activated Stat2. JBiol Chem 2000;275:26937.

    15. Fasler-Kan E, Pansky A, Wiederkehr M, Battegay M, Heim MH.Interferon-alpha activates signal transducers and activators of tran-scription 5 and 6 in Daudi cells. Eur J Biochem 1998;254:5149.

    16. Matikainen S, Sareneva T, Ronni T, Lehtonen A, Koskinen PJ, Julk-unen I. Interferon-alpha activates multiple STAT proteins and upre-gulates proliferation-associated IL-2Ralpha, c-myc, and pim-1 genesin human T cells. Blood 1999;93:198091.

    17. Torpey N, Maher SE, Bothwell AL, Pober JS. Interferon alpha but notinterleukin 12 activates STAT4 signaling in human vascular endothelialcells. J Biol Chem 2004;279:2678996.

    18. Parmar S, Platanias LC. Interferons: mechanisms of action and clinicalapplications. Curr Opin Oncol 2003;15:4319.

    Table 1. Contracting Indications of IFN- for Malignant Conditions (cont'd )

    Indication Stage Comment Standard Current Therapy Reference

    In comparison withchemotherapy IFNahas been shown tobe superior.

    In imatinib resistance, IFNahas been precluded by theappearanceof dasatinib.

    Hemangioma Complicate hemangiomas thatdo not respond to steroids.

    Steroids. 93

    Rate regression in58% of the patients.

    AIDS-related Kaposisarcoma

    Evidence for clinical activity. Liposomal anthracyclines arepreferred as the firsttreatment option.

    94

    PV, polycythemia vera; ET, essential thrombocythemia; PM, primary myelofibrosis; RCC, renal cell cancer; CMS, chronic myelo-proliferative syndrome; CML, chronic myeloid leukemia

    IFN-a on Immune Cells

    www.aacrjournals.org Clin Cancer Res; 17(9) May 1, 2011 2625

  • 19. Platanias LC, Fish EN. Signaling pathways activated by interferons.Exp Hematol 1999;27:158392.

    20. Nguyen KB, Cousens LP, Doughty LA, Pien GC, Durbin JE, Biron CA.Interferon alpha/beta-mediated inhibition and promotion of interferongamma: STAT1 resolves a paradox. Nat Immunol 2000;1:706.

    21. Levy DE, Lee CK. What does Stat3 do?J Clin Invest 2002;109:11438.22. Qing Y, Stark GR. Alternative activation of STAT1 and STAT3 in

    response to interferon-gamma. J Biol Chem 2004;279:4167985.23. Ramana CV, Kumar A, Enelow R. Stat1-independent induction of

    SOCS-3 by interferon-gamma is mediated by sustained activation ofStat3 in mouse embryonic fibroblasts. Biochem Biophys Res Com-mun 2005;327:72733.

    24. Alexander WS. Suppressors of cytokine signalling (SOCS) in theimmune system. Nat Rev Immunol 2002;2:4106.

    25. Yoshimura A, Naka T, KuboM. SOCS proteins, cytokine signalling andimmune regulation. Nat Rev Immunol 2007;7:45465.

    26. Hanada T, Yoshida H, Kato S, Tanaka K, Masutani K, Tsukada J, et al.Suppressor of cytokine signaling-1 is essential for suppressing den-dritic cell activation and systemic autoimmunity. Immunity 2003;19:43750.

    27. Zitzmann K, Brand S, De Toni EN, Baehs S, Goke B, Meinecke J, et al.SOCS1 silencing enhances antitumor activity of type I IFNs byregulating apoptosis in neuroendocrine tumor cells. Cancer Res2007;67:502532.

    28. Du Z, Wei L, Murti A, Pfeffer SR, Fan M, Yang CH, et al. Non-conventional signal transduction by type 1 interferons: the NF-kappaBpathway. J Cell Biochem 2007;102:108794.

    29. Ahmed Z, Beeton CA, Williams MA, Clements D, Baldari CT, LadburyJE. Distinct spatial and temporal distribution of ZAP70 and Lckfollowing stimulation of interferon and T-cell receptors. J Mol Biol2005;353:100110.

    30. Petricoin EF 3rd, Ito S, Williams BL, Audet S, Stancato LF, Gamero A,et al. Antiproliferative action of interferon-alpha requires componentsof T-cell-receptor signalling. Nature 1997;390:62932.

    31. Stevens CN, Simeone AM, John S, Ahmed Z, Lucherini OM, BaldariCT, et al. T-cell receptor early signalling complex activation inresponse to interferon-alpha receptor stimulation. Biochem J2010;428:42937.

    32. Trinchieri G, Santoli D. Anti-viral activity induced by culturing lym-phocytes with tumor-derived or virus-transformed cells. Enhance-ment of human natural killer cell activity by interferon and antagonisticinhibition of susceptibility of target cells to lysis. J Exp Med 1978;147:131433.

    33. Nguyen KB, Salazar-Mather TP, Dalod MY, Van Deusen JB, Wei XQ,Liew FY, et al. Coordinated and distinct roles for IFN-alpha beta, IL-12,and IL-15 regulation of NK cell responses to viral infection. J Immunol2002;169:427987.

    34. Zhang X, Sun S, Hwang I, Tough DF, Sprent J. Potent and selectivestimulation of memory-phenotype CD8 T cells in vivo by IL-15.Immunity 1998;8:5919.

    35. Lee CK, Rao DT, Gertner R, Gimeno R, Frey AB, Levy DE. Distinctrequirements for IFNs and STAT1 in NK cell function. J Immunol2000;165:35717.

    36. Trinchieri G, Santoli D, Granato D, Perussia B. Antagonistic effects ofinterferons on the cytotoxicity mediated by natural killer cells. FedProc 1981;40:270510.

    37. Ortaldo JR, Phillips W, Wasserman K, Herberman RB. Effects ofmetabolic inhibitors on spontaneous and interferon-boosted humannatural killer cell activity. J Immunol 1980;125:183944.

    38. Bogdan C, Mattner J, Schleicher U. The role of type I interferons innon-viral infections. Immunol Rev 2004;202:3348.

    39. Sampson LL, Heuser J, Brown EJ. Cytokine regulation of complementreceptor-mediated ingestion by mouse peritoneal macrophages. M-CSF and IL-4 activate phagocytosis by a common mechanism requir-ing autostimulation by IFN-beta. J Immunol 1991;146:100513.

    40. Lapenta C, Santini SM, Logozzi M, Spada M, Andreotti M, Di PucchioT, et al. Potent immune response against HIV-1 and protection fromvirus challenge in hu-PBL-SCID mice immunized with inactivatedvirus-pulsed dendritic cells generated in the presence of IFN-alpha.J Exp Med 2003;198:3617.

    41. Santini SM, Lapenta C, Logozzi M, Parlato S, Spada M, Di Pucchio T,et al. Type I interferon as a powerful adjuvant for monocyte-deriveddendritic cell development and activity in vitro and in Hu-PBL-SCIDmice. J Exp Med 2000;191:177788.

    42. Santodonato L, DAgostino G, Nisini R, Mariotti S, Monque DM,Spada M, et al. Monocyte-derived dendritic cells generated after ashort-term culture with IFN-alpha and granulocyte-macrophage col-ony-stimulating factor stimulate a potent Epstein-Barr virus-specificCD8 T cell response. J Immunol 2003;170:5195202.

    43. Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenousactivators of dendritic cells. Nat Med 1999;5:124955.

    44. Ito T, Amakawa R, Inaba M, Ikehara S, Inaba K, Fukuhara S. Differ-ential regulation of human blood dendritic cell subsets by IFNs. JImmunol 2001;166:29619.

    45. Montoya M, Schiavoni G, Mattei F, Gresser I, Belardelli F, Borrow P,et al. Type I interferons produced by dendritic cells promote theirphenotypic and functional activation. Blood 2002;99:326371.

    46. Gautier G, Humbert M, Deauvieau F, Scuiller M, Hiscott J, Bates EE,et al. A type I interferon autocrine-paracrine loop is involved in Toll-likereceptor-induced interleukin-12p70 secretion by dendritic cells. J ExpMed 2005;201:143546.

    47. Byrnes AA, Ma X, Cuomo P, Park K, Wahl L, Wolf SF, et al. Type Iinterferons and IL-12: convergence and cross-regulation amongmediators of cellular immunity. Eur J Immunol 2001;31:202634.

    48. Parlato S, Santini SM, Lapenta C, Di Pucchio T, Logozzi M, Spada M,et al. Expression of CCR-7, MIP-3beta, and Th-1 chemokines in type IIFN-induced monocyte-derived dendritic cells: importance for therapid acquisition of potent migratory and functional activities. Blood2001;98:30229.

    49. Asselin-Paturel C, Boonstra A, Dalod M, Durand I, Yessaad N, Dezut-ter-Dambuyant C, et al. Mouse type I IFN-producing cells are imma-ture APCswith plasmacytoid morphology. Nat Immunol 2001;2:114450.

    50. Rouzaut A, Garasa S, Teijeira A, Gonzalez I, Martinez-Forero I, LarreaE, et al. Dendritic cells adhere to and transmigrate across lymphaticendothelium in response to IFN-a. Eur J Immunol 2010;40:305463.

    51. Shiow LR, Rosen DB, Brdickova N, Xu Y, An J, Lanier LL, et al. CD69acts downstream of interferon-alpha/beta to inhibit S1P1 and lym-phocyte egress from lymphoid organs. Nature 2006;440:5404.

    52. Fink K, Lang KS, Manjarrez-Orduno N, Junt T, Senn BM, Holdener M,et al. Early type I interferon-mediated signals on B cells specificallyenhance antiviral humoral responses. Eur J Immunol 2006;36:2094105.

    53. Le Bon A, Thompson C, Kamphuis E, Durand V, Rossmann C, KalinkeU, et al. Cutting edge: enhancement of antibody responses throughdirect stimulation of B and T cells by type I IFN. J Immunol 2006;176:20748.

    54. Coro ES, ChangWL, Baumgarth N. Type I IFN receptor signals directlystimulate local B cells early following influenza virus infection. JImmunol 2006;176:434351.

    55. Brinkmann V, Geiger T, Alkan S, Heusser CH. Interferon alphaincreases the frequency of interferon gamma-producing humanCD4 T cells. J Exp Med 1993;178:165563.

    56. Nguyen KB, Watford WT, Salomon R, Hofmann SR, Pien GC, Mor-inobu A, et al. Critical role for STAT4 activation by type 1 interferons inthe interferon-gamma response to viral infection. Science 2002;297:20636.

    57. Marrack P, Kappler J, Mitchell T. Type I interferons keep activated Tcells alive. J Exp Med 1999;189:52130.

    58. Kolumam GA, Thomas S, Thompson LJ, Sprent J, Murali-Krishna K.Type I interferons act directly on CD8 T cells to allow clonal expansionand memory formation in response to viral infection. J Exp Med2005;202:63750.

    59. Aichele P, Unsoeld H, KoschellaM, Schweier O, Kalinke U, Vucikuja S.CD8 T cells specific for lymphocytic choriomeningitis virus requiretype I IFN receptor for clonal expansion. J Immunol 2006;176:45259.

    60. Le Bon A, Durand V, Kamphuis E, Thompson C, Bulfone-Paus S,Rossmann C, et al. Direct stimulation of T cells by type I IFN enhancesthe CD8 T cell response during cross-priming. J Immunol 2006;176:46829.

    Hervas-Stubbs et al.

    Clin Cancer Res; 17(9) May 1, 2011 Clinical Cancer Research2626

  • 61. Thompson LJ, Kolumam GA, Thomas S, Murali-Krishna K. Innateinflammatory signals induced by various pathogens differentiallydictate the IFN-I dependence of CD8 T cells for clonal expansionand memory formation. J Immunol 2006;177:174654.

    62. Curtsinger JM, Valenzuela JO, Agarwal P, Lins D, Mescher MF, Type I.Type I IFNs provide a third signal to CD8 T cells to stimulate clonalexpansion and differentiation. J Immunol 2005;174:44659.

    63. Agarwal P, Raghavan A, Nandiwada SL, Curtsinger JM, Bohjanen PR,Mueller DL, et al. Gene regulation and chromatin remodeling by IL-12and type I IFN in programming for CD8 T cell effector function andmemory. J Immunol 2009;183:1695704.

    64. Hervas-Stubbs S, Riezu-Boj J-I, Gonzalez I, Mancheno U, Dubrot J,Azpilicueta A, et al. Effects of IFNa as a signal-3 cytokineonhuman naveandantigen-experiencedCD8T-cells. EurJ Immunol 2010;40:3389402.

    65. Marshall HD, Prince AL, Berg LJ, Welsh RM. IFN-alpha beta and self-MHC divert CD8 T cells into a distinct differentiation pathway char-acterized by rapid acquisition of effector functions. J Immunol2010;185:141928.

    66. Kirkwood J. Cancer immunotherapy: the interferon-alpha experience.Semin Oncol 2002;29:1826.

    67. Wedemeyer H, Wiegand J, Cornberg M, Manns MP. Polyethyleneglycol-interferon: current status in hepatitis C virus therapy. J Gastro-enterol Hepatol 2002;17[Suppl 3]:S34450.

    68. Garbe C, Eigentler TK. Diagnosis and treatment of cutaneous mela-noma: state of the art 2006. Melanoma Res 2007;17:11727.

    69. Hanley PJ, Cruz CR, Savoldo B, Leen AM, StanojevicM, Khalil M, et al.Functionally active virus-specific T cells that target CMV, adenovirus,and EBV can be expanded from naive T-cell populations in cord bloodand will target a range of viral epitopes. Blood 2009;114:195867.

    70. Sterman DH, Recio A, Haas AR, Vachani A, Katz SI, Gillespie CT, et al.A phase I trial of repeated intrapleural adenoviral-mediated interferon-beta gene transfer for mesothelioma and metastatic pleural effusions.Mol Ther 2010;18:85260.

    71. Bracci L, La Sorsa V, Belardelli F, Proietti E. Type I interferons asvaccine adjuvants against infectious diseases and cancer. Expert RevVaccines 2008;7:37381.

    72. Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, et al.Chemokine expression in melanoma metastases associated withCD8 T-cell recruitment. Cancer Res 2009;69:307785.

    73. Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, et al.Induction of CD8 T-cell responses against novel glioma-associatedantigen peptides and clinical activity by vaccinations with alpha-type1 polarized dendritic cells and polyinosinic-polycytidylic acid stabi-lized by lysine and carboxymethylcellulose in patients with recurrentmalignant glioma. J Clin Oncol 2011;29:3306.

    74. Dubrot J, Palazon A, Alfaro C, Azpilikueta A, Ochoa MC, Rouzaut A,et al. Intratumoral injection of interferon-a and systemic delivery ofagonist anti-CD137monoclonal antibodies synergize for immunother-apy. Int J Cancer 2011;128:10518.

    75. DePalmaM,MazzieriR,Politi LS,PucciF, Zonari E,SitiaG,et al. Tumor-targeted interferon-alpha delivery by Tie2-expressing monocytes inhi-bits tumor growth and metastasis. Cancer Cell 2008;14:299311.

    76. Brody JD, Ai WZ, Czerwinski DK, Torchia JA, LevyM, Advani RH, et al.In situ vaccination with a TLR9 agonist induces systemic lymphomaregression: a phase I/II study. J Clin Oncol 2010;28:432432.

    77. Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Spatz A, et al.Diagnosis and treatment of melanoma: European consensus-basedinterdisciplinary guideline. Eur J Cancer 2010;46:27083.

    78. Hodi FS, ODay SJ, McDermott DF, Weber RW, Sosman JA, HaanenJB, et al. Improved survival with ipilimumab in patients with metastaticmelanoma. N Engl J Med 2010;363:71123.

    79. Messing EM, Manola J, Wilding G, Propert K, Fleischmann J, Craw-ford ED, et al. Eastern Cooperative Oncology Group/Intergroup trial.Phase III study of interferon alfa-NL as adjuvant treatment for resect-able renal cell carcinoma: an Eastern Cooperative Oncology Group/Intergroup trial. J Clin Oncol 2003;21:121422.

    80. Pizzocaro G, Piva L, Colavita M, Ferri S, Artusi R, Boracchi P, et al.Interferon adjuvant to radical nephrectomy in Robson stages II and IIIrenal cell carcinoma: a multicentric randomized study. J Clin Oncol2001;19:42531.

    81. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM,Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cellcarcinoma. N Engl J Med 2007;356:11524.

    82. Quesada JR, Reuben J, Manning JT, Hersh EM, Gutterman JU. Alphainterferon for induction of remission in hairy-cell leukemia. N Engl JMed 1984;310:158.

    83. Alfaro C, Suarez N, Gonzalez A, Solano S, Erro L, Dubrot J, et al.Influence of bevacizumab, sunitinib and sorafenib as single agents orin combination on the inhibitory effects of VEGF on human dendriticcell differentiation from monocytes. Br J Cancer 2009;100:11119.

    84. Kumar S. Multiple myeloma current issues and controversies.Cancer Treat Rev 2010;36[Suppl 2]:S311.

    85. Zhan H, Spivak JL. The diagnosis and management of polycythemiavera, essential thrombocythemia, and primary myelofibrosis in theJAK2 V617F era. Clin Adv Hematol Oncol 2009;7:33442.

    86. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al.Activating mutation in the tyrosine kinase JAK2 in polycythemia vera,essential thrombocythemia, and myeloid metaplasia with myelofibro-sis. Cancer Cell 2005;7:38797.

    87. Kiladjian JJ, Cassinat B, Turlure P, Cambier N, Roussel M, Bellucci S,et al. High molecular response rate of polycythemia vera patientstreated with pegylated interferon alpha-2a. Blood 2006;108:203740.

    88. Talpaz M, Kantarjian HM, McCredie KB, Keating MJ, Trujillo J, Gutter-man J. Clinical investigation of human alpha interferon in chronicmyelogenous leukemia. Blood 1987;69:12808.

    89. Baccarani M, Rosti G, de Vivo A, Bonifazi F, Russo D, Martinelli G, etal; Italian Cooperative Study Group on Myeloid Leukemia. A rando-mized study of interferon-alpha versus interferon-alpha and low-dosearabinosyl cytosine in chronic myeloid leukemia. Blood 2002;99:152735.

    90. Guilhot F, Chastang C, Michallet M, Guerci A, Harousseau JL, Mal-oisel F, et al; French Chronic Myeloid Leukemia Study Group. Inter-feron alfa-2b combined with cytarabine versus interferon alone inchronic myelogenous leukemia. N Engl J Med 1997;337:2239.

    91. Silver RT, Woolf SH, Hehlmann R, Appelbaum FR, Anderson J,Bennett C, et al. An evidence-based analysis of the effect of busulfan,hydroxyurea, interferon, and allogeneic bone marrow transplantationin treating the chronic phase of chronic myeloid leukemia: developedfor the American Society of Hematology. Blood 1999;94:151736.

    92. OBrien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cer-vantes F, et al. IRIS Investigators. Imatinib compared with interferonand low-dose cytarabine for newly diagnosed chronic-phase chronicmyeloid leukemia. N Engl J Med 2003;348:9941004.

    93. Greinwald JH Jr, Burke DK, Bonthius DJ, Bauman NM, Smith RJ. Anupdate on the treatment of hemangiomas in children with interferonalfa-2a. Arch Otolaryngol Head Neck Surg 1999;125:217.

    94. Vanni T, Sprinz E, Machado MW, Santana RdeC, Fonseca BA,Schwartsmann G. Systemic treatment of AIDS-related Kaposi sar-coma: current status and perspectives. Cancer Treat Rev 2006;32:44555.

    IFN-a on Immune Cells

    www.aacrjournals.org Clin Cancer Res; 17(9) May 1, 2011 2627