You are on page 1of 9

Clinical

Cancer
Research

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

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 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 interferon
Interferons (IFN) were discovered in 1957 by Isaacs and
Lindenmann as soluble proteins able to inhibit virus replication 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 sigAuthors' Affiliation: 1Division 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 3Division of Oncology, CIMA, University of
 Paris Descartes,
Navarra, Pamplona, Spain; 4Institut Cochin, Universite
CNRS (UMR 8104), and 5INSERM, 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: imelero@unav.es or mshervas@unav.es
doi: 10.1158/1078-0432.CCR-10-1114
2011 American Association for Cancer Research.

www.aacrjournals.org

nals through a receptor complex consisting of interleukin10 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 regulated 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 immunostimulatory 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
(68).
Receptor occupancy rapidly triggers several signaling
cascades (Fig. 1), which culminate in the transcriptional
regulation of hundreds of IFN-stimulated genes (ISG). The

2619

Hervas-Stubbs et al.

Type I IFNs signaling pathways


(A) JAK-STAT
IFN-/

(B) CRK
IFN-/

(C) PI3K and NFB


IFN-/

(D) MAPK

IFN-/

TCR complex

C3G

PI3K

AKT

mTOR
Inhibition of
GSK-3/CDKN1A/
CDKN1B

IKK

IKK

Ras

PKC

Rac1
Raf1

NFB

Proliferation
Differentiation
Survival
....

IRF9

p38

Jun
MEF2
MAPKAP Elk-1
NFAT
Rsk
ATF2

Fos
Elk-1
Sap-1

STAT STAT
IRF9

Heterodimers

STAT STAT
1
2

STAT STAT

STAT CRKL
5

ISRE

GAS

GAS

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

NFB Rel

Regulated
genes or
functions

JNK

MEK1/2
ERK1/2

Homodimers

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

VAV

Rap1

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

Lck
Zap70

C3G

Heterodimers

STAT STAT
1
2

SLP76
VAV

RS1/2

Homodimers
STAT STAT STAT1, STAT3
STAT4, STAT5
STAT CRKL
STAT6
5

JAK1

VAV

STAT
3

LAT

STAT CRKL
5

Tyk2

JAK1

IFNAR2
IFNAR1

Tyk2

TRAF STAT
3
NIK

IFNAR2
IFNAR1

STAT

JAK1

IFNAR2
IFNAR1

STAT

Tyk2

JAK1

IFNAR2
IFNAR1

IFNAR2
IFNAR1

Tyk2

CD45 IFN-/

CD4/8

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

...

...

ISG15
Apoptosis
Histone acetylation
Antiviral effect
Growth inhibitory effects
Hematopoietic suppressive signals

NFAT
Ets
Elk-1
MEF2
Stat1/3

...

c-Myc
SAP-1
P53
SP1
SMADs

Cellular growth
Differentiation
Ser phosphorylation
of STAT1

2011 American Association for Cancer Research

Figure 1. Signaling pathways activated by the IFN-I receptor. Schematic of signaling pathways downstream of the IFNAR, emphasizing cross-talk with
different signaling cascades, which depends on the identity and activation status of the cell responding to IFN-I. A, JAK-STAT signal pathway activated by
IFNs-I. B, CRKL pathway. Upon JAK activation, CRKL associates with TYK2 and undergoes tyrosine phosphorylation. The activated form of CRKL
forms a complex with STAT5, which also undergoes TYK2-dependent tyrosine phosphorylation. The CRKLSTAT5 complex translocates to the nucleus
and binds specific GAS elements. C, PI3K and NF-kB pathways. Phosphorylation of TYK2 and JAK1 results in activation of PI3K and AKT, which
in turn mediates downstream activation of mTOR, inactivation of GSK-3, CDKN1A, and CDKN1B, and activation of IKKb, which results in activation of
NF-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 also
activate NF-kB through an activation loop via PKCq. D, MAPK pathway. Activation of JAK results in tyrosine phosphorylation of Vav, which leads to the
activation 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); and NFAT, ETS, ELK1, MEF2, STAT1/2, c-Myc, SAP-1, p53, SP1,
and SMADs (activated by ERK1/2).

first signaling pathway shown to be activated by IFNs-I was


the Janus activated kinasesignal transducer and activation
of transcription (JAK-STAT) pathway (Fig. 1A; ref. 9), which
is active in almost all cell types. IFNAR1 is constitutively
associated with tyrosine kinase 2 (TYK2), whereas IFNAR2
is associated with JAK1. Receptor binding results in transphosphorylation of JAK1 and TYK2. The activated JAKs
then phosphorylate conserved tyrosine residues in the
cytoplasmic tails of the IFNAR. These phosphotyrosyl residues subsequently serve as docking sites for the recruitment
of src-homology 2 (SH2)containing signaling molecules,
such as STAT1 and STAT2. Once at the receptor, the STAT
proteins themselves become JAK substrates. STAT1 and

2620

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

STAT2 are both phosphorylated on a single tyrosine


(Y701 for STAT1 and Y690 for STAT2). Once phosphorylated, STATs dimerize and move to the nucleus, where they
associate with the IFN-regulatory factor 9 (IRF9) to form
the 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, namely
STAT3 and STAT5, have been found to be phosphorylated
(activated) by IFNs-I (Fig. 1A; refs. 1013). STAT4
and STAT6 can also be activated by IFN-a; however, this
activation seems to be restricted to certain cell types, such
as endothelial or lymphoid cells (1417). Following

Clinical Cancer Research

IFN-a on Immune Cells

phosphorylation, activated STATs form homo- (STAT1,


STAT3, STAT4, STAT5, and STAT6) or heterodimers
(STAT1/2, STAT1/3, STAT1/4, STAT1/5, STAT2/3, and
STAT5/6; refs. 10, 11, 18, 19), which translocate to the
nucleus and bind other regulatory sequences known as
IFN-gactivated sites (GAS).
Interestingly, distinct STATs have opposing biological
effects. Thus, STAT3 stimulates growth, whereas on the
contrary, STAT1 is a growth inhibitor. On the other hand,
IFNs-Imediated activation of STAT4 is required for IFN-g
production, whereas surprisingly, STAT1 negatively regulates the IFNs-Idependent induction of IFN-g (20). The
relative abundance of these dimeric transcription factors,
which may vary substantially depending on cell type and
activation and/or differentiation state, is likely to have a
major 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, which
accumulates at the cell surface where it physically associates
with the IFNAR and usurps the IFNAR-STAT1 pathway by
stimulating the transcription of suppressors of cytokine
signaling (SOCS; refs. 24, 25). The SOCS proteins extinguish IFN-I signaling in a negative feedback loop. For
instance, SOCS1 directly interacts with JAKs blocking their
catalytic activity, whereas SOCS3 seems to inhibit JAKs after
gaining access by receptor binding. In addition, SOCS
proteins interact with the cellular ubiquitination machinery through the SOCS-box region and may redirect associated proteins, such as JAKs or IFNAR chains, to
proteasomal degradation. It has been shown that SOCS1
knockout mice develop lupus-like disease symptoms (26)
and that SOCS1 silencing enhances the antitumor activity
of IFNs-I (27), suggesting that SOCS are key elements in
keeping IFNs-I effects under control.
Alternative signaling pathways and cross-talk with
other receptors
Evidence is accumulating that several other signaling
elements and cascades are required for the generation of
many of the responses to IFNs-I, such as the v-crk sarcoma
virus CT10 oncogene homolog (avian)-like (CRKL) pathway, the mitogen activated protein kinase (MAPK) pathway, the phosphoinositide 3-kinase (PI3K) pathway, and
either the classical or the alternative NF-kB cascade
(Fig. 1B-D; refs. 9, 28). Some of these pathways operate
independently of the JAK-STAT, whereas others cooperate
with STATs to tune the response to IFNs-I. Interestingly,
MAPK, PI3K, and NF-kB pathways involve immunoreceptor tyrosine-based activation motives used by classical
immunoreceptors, suggesting a cross-talk among IFNAR
and 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 IFNAR1
subunit. PI3K subsequently activates the serine-threonine
kinase AKT, which in turn mediates downstream the activation of mTOR and the inactivation of several proteins,

www.aacrjournals.org

such as glycogen synthase kinase 3 (GSK-3) and cyclin


dependent kinase inhibitors 1A and 1B (CDKN1A and
CDKN1B), all of them involved in the control of cell
division and proliferation. The AKT pathway also results
in the activation of IkB kinase beta (IKKb), which gives
rise to NF-kB activation. IFNs-I also activate NF-kB by an
alternative pathway, which involves the linkage of TNF
receptorassociated factors (TRAF) to the activation of
the NF-kBinducing kinase (NIK), which in turn results
in activation of NF-kB2. This alternative pathway is
strictly dependent on the activity of IKKa. In addition,
PI3K/AKT can also activate NF-kB through an activation
loop via protein kinase C zeta (PKCq). Overall, NF-kB
activated by IFNs-I regulates prosurvival signals and
enhances the expression of several GTP-binding proteins
as well as molecules involved in antigen processing and/
or presentation.
The MAPK pathway. Activation of JAK results in tyrosine phosphorylation of the guanine nucleotide exchange
factor Vav, which leads to downstream activation of Rat
sarcoma protein (Ras) and ras-related C3 botulinum toxin
substrate 1 (Rac1), which subsequently leads to the activation of several MAPKs: p38, c-Jun N-terminal kinases
(JNK), and extracellular signal regulated kinases (ERK) 1
and 2 (9). Vav can also activate PKCq, leading to the
activation of the NF-kB cascade. Several studies have shown
that IFN-amediated activation of ERK1/2 in T lymphocytes requires proteins involved in the T-cell receptor (TCR)
early signaling complex, such as CD45, lymphocyte-specific protein tyrosine kinase (Lck), Zeta-chain-associated
protein kinase 70 (Zap70), SH2 domain-containing leukocyte protein of 76 kDa (SLP76), and Vav1, and possibly
linker for activation of T cells (LAT), indicating cross-talk
between pathways (2931). Moreover, it has been shown
recently that TCR deletion in Jurkat cells abrogated IFNARstimulated MAPK activity, whereas the canonical JAK-STAT
pathway remained unaffected (31). A hypothetical pathway that involves all these molecules and results in Vav
activation is depicted in Fig. 1D. The different MAPKs
activate different sets of transcription factors. Importantly,
p38 functions are essential for the antiviral and antileukemic properties of IFNs-I, it plays a role in the hematopoietic-suppressive signals and is required for the growthinhibitory effects of IFNs-I observed in certain lymphocyte
cultures. IFNs-Iactivated ERK cascades regulate cellular
growth, differentiation, and serine phosphorylation of
STAT1. The search for biochemical signals to explain the
effects of IFN-I on immune system cells is far from over.
Effects of IFNs-I on cells of the immune system
It has long been established that the main mechanism
accounting for the efficacy of the IFN-Ibased therapies was
due to their direct effect on malignant or virus-infected
cells. In fact, IFNs-I directly inhibit the proliferation of
tumor and virus-infected cells and increase MHC class I
expression, enhancing antigen recognition. Moreover,
IFNs-I repress oncogene expression and induce that of
tumor suppressor genes, which may contribute to the

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

2621

Hervas-Stubbs et al.

inhibitory effects of IFN-I on malignant cells in conjunction with the antiangiogenic effects.
IFNs-I have also proven to be involved in immune
system regulation (7, 8, 32). IFNs-I exert their effects on
immune cells either directly, through IFNAR triggering, or
indirectly (i) by the induction of chemokines, which allow
the recruitment of immune cells to the site of infection; (ii)
by the secretion of a second wave of cytokines, which could
further regulate cell numbers and activities (as for example
IL-15, which plays a critical role in proliferation and
maintenance of NK cells and memory CD8 T cells; refs.
33, 34); or (iii) by the stimulation of other cell types critical
for the activation of certain immune cells, such as DC for
the activation of nave T cells.
One of the earliest described immunoregulatory functions of IFNs-I was their ability to regulate NK functions
(32). IFNs-I enhance the ability of NK cells to kill target
cells and to produce IFN-g by indirect and direct mechanisms (33, 35, 36). Furthermore, IFNs-I promote the accumulation and/or survival of proliferating NK cells by the
IFN-I/STAT1dependent induction of IL-15 (33). IFNs-I
also enhance the production or secretion of other cytokines
by 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 the
differentiation of monocytes into DC with high capacity
for Ag presentation, stimulate macrophage antibodydependent cytotoxicity, and positively or negatively regulate the production of various cytokines (e.g., TNF, IL-1, IL6, IL-8, IL-12, and IL-18) by macrophages (38). In addition,
autocrine IFN-I is required for the enhancement of macrophage phagocytosis by macrophage colony-stimulating
factor and IL-4 (39) and for the lipopolysaccharide-,
virus-, and IFN-ginduced oxidative burst through the
generation 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 cell
type able to prime nave T cells and, therefore, are critical
antigen presenting cells (APC). IFNs-I have multiple effects
on DCs, affecting their differentiation, maturation, and
migration. Thus, human monocytes cultured in granulocyte-macrophage colony-stimulating factor (GM-CSF)
IFN-a/b, rather than the more commonly used combination of GM-CSF IL-4, differentiate more rapidly into DCs,
exhibiting the phenotype of partially mature DCs, while
showing a strong capability to induce a primary human
antibody response and CTL expansion when pulsed with
antigen and injected into humanized severe combined
immunodeficiency (SCID) mice (4042).
In vitro treatment of immature conventional DCs with
IFN-a/b has been shown to upregulate surface expression
of MHC class I, class II, CD40, CD80, CD86, and CD83
molecules in the human system (4345), associated with a
heightened capacity to induce CD8 T-cell responses.
Accordingly, it has been observed in mice that IFN-a acting
on DC plays a key role in achieving efficient cross-priming
of antigen-specific CD8 T lymphocytes (7, 8, 32). IFNs-I
also affect the ability of DC to secrete IL-12p70. Low

2622

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

concentrations of IFNs-I are essential for the optimal production of IL-12p70 (46), whereas higher levels of IFN-I
suppress IL-12p40 expression, thus dampening IL-12p70
production (47).
A key requisite to initiate the adaptive immune response
is the arrival of professional APC from infected tissue into
lymph nodes. It has been shown that human DCs differentiated from monocytes in the presence of IFN-a exhibit
upregulation of CC chemokine receptor type 7 (CCR7),
correlating with an enhanced chemotactic response in vitro
to CCR7 natural ligand (CCL19) and with strong migratory
behavior in SCID mice (48). Interestingly, experiments in
mice have also shown that IFN-a/b is required for plasmacytoid DCs (pDC) to migrate from the marginal zone into
the T-cell area, where they form clusters (49). We have also
recently shown that IFNs-I enhance the adhesion and
extravasations of DCs across inflamed lymphatic
endothelium in a lymphocyte function-associated antigen 1 (LFA-1) and very late antigen-4 (VLA4)dependent fashion (50). IFNs-I may also favor the encounter
between DCs and specific lymphocytes in lymph nodes
by promoting lymphocyte trapping in lymph nodes upon
downmodulation of sphingosine 1-phosphate (51).
IFNs-I have also been shown to potently enhance the
primary antibody response to soluble antigen, stimulating
the production of all subtypes of immunoglobulin G (IgG),
and inducing long-lived antibody production and immunologic memory (8). Direct effects of IFN-a on DCs (8), B
cells (52, 53), and CD4 T cells (53) all contribute to its
adjuvant activities on humoral immune responses. Interestingly, a direct effect of IFNs-I on B cells seems to be
crucial for the development of local humoral responses
against viruses (54).
As mentioned above, CD4 T cells are direct targets of IFNI in the enhancement of antibody responses (53). In
humans, it has also been described that direct effects of
IFN-I on naive CD4 T cells favor their differentiation into
IFN-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-I
promote IFN-g production by CD8 T cells in a STAT4dependent manner (56) and promote survival of CD8 T
cells from wild-type (WT) but not IFNAR-deficient
(IFNAR/) mice (57). The definitive report showing that
CD8 T cells represent direct targets of IFN-Imediated
stimulation in vivo came from Kolumam and colleagues
(58). By adoptively transferring virus-specific naive CD8 T
cells from IFNAR/ or IFNAR-sufficient mice into normal
IFNAR WT hosts, IFNs-I were shown to act directly on
murine CD8 T cells, allowing their clonal expansion and
memory differentiation. Subsequent studies confirmed this
work (5961). Elegant experiments in mice by Curtsinger
and colleagues (62) have shown that, in addition to signals
via TCR (signal-1) and CD28 (signal-2), nave CD8 T cells
required a third signal to differentiate into effector cells.
cDNA microarray analyses showed that IFN-a could
regulate critical genes involved in CTL functions (63),
providing evidence that IFNa promoted activation and

Clinical Cancer Research

IFN-a on Immune Cells

differentiation of CD8 T cells by sustaining the expression


of T-bet and Eomes through chromatin remodeling.
Recently, we have shown that IFN-a provides a strong
and direct signal to human CD8 T cells, thereby resulting
in upregulation of critical genes for cytotoxic T-cell activity
(cytolysis and IFN-g secretion) and for the production of
chemokines that would coattract other effector lymphocytes. IFN-a was absolutely critical in the case of human
nave CD8 T cells for effector function acquisition (64). In
many instances, T cells may sense IFN-I before the cognate
antigen is actually presented to them. As a result of a
preexposure to IFN-I, CD8 T cells become preactivated
and acquire much faster effector functions upon antigen
recognition (65). This preactivation may reflect the upregulation by IFN-I of several mRNAs in antigen-naive CD8 T
lymphocyte, albeit without changes in the expression of the
corresponding protein, unless antigen-specific activation
ensues (64).

Clinical-Translational Advances
The contracting current indications of IFN-a as an
anticancer agent
IFN-a has received approval for treatment of several
neoplastic diseases (66). The most frequent indications
for IFN-a are chronic viral hepatitis. For the treatment of
these infectious conditions, stabilized forms of IFN-a have
been created by directed conjugation of polyethylene glycol, which, by increasing molecular weight, retards renal
clearance and, thereby, extends plasma concentrations with
sustained receptor occupancy (67). However, PEG-conjugated IFN is not widely used in oncology in the absence of
comparative 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-a
prolongs disease-free survival and shows a trend toward
increased overall survival (68). However, the advent of
CTLA-4 antagonist antibodies will very likely displace
the use of IFN-a for melanoma therapy (69).
Table 1 shows the spectrum of neoplastic diseases in which
IFN-a has been used. As shown, its role in cancer therapy is
steadily decreasing because of its limited efficacy and the
advent of new, more effective, and/or safer treatments.
Future perspectives
IFNs-I are powerful tools to directly and indirectly
modulate the functions of the immune system. Evolution
has shaped these cytokines as a key sign of alarm in case
of viral infection. The type of immune response that we
would like to induce and sustain against cancer antigens
is identical to the one we commonly observe upon acute
viral infections. Obviously, the effects of IFN-I are integrated with many other key molecules that need to act in a
concerted fashion.
As described in Table 1, side effects of systemic longterm treatments and lack of sufficiently high efficacy have
dampened the interest of IFN-a for clinical use in oncology. However, we believe that IFN-a is likely to be more

www.aacrjournals.org

efficacious when acting locally and intermittently at the


malignant tissue and tumor-draining lymph node than
when administered to achieve systemic bioavailability.
Local delivery can be achieved with pharmaceutical formulations and gene therapy approaches (70). In our
opinion, the clinical use of recombinant IFN-a as a vaccine
adjuvant should be reconsidered and new investigations
carried out with an eye to clinical applications (71).
The rationale for using intermittent delivery arises from
observations indicating that IFN-I turns on signaling desensitizing mechanisms (i.e., because of SOCS1 induction).
Intermittency at an optimized pace may help to avoid these
negative feedback mechanisms in the responding immune
cells. These ideas contrast with stabilized formulations,
such as the widely used polyethylene glycol-conjugated
IFN-a and fusion proteins with albumin that extend the
half-life of the cytokine.
It is of much interest that a gene expression signature
induced by IFN-I in melanoma lesions predicts benefit
from vaccines and may have a key role in recruiting
effector T cells to tumors by inducing chemokine genes
(72). IFN-a has been recently used in promising combinatorial immunotherapy approaches alongside DC vaccination for glioblastoma, precisely to promote T-cell
infiltration and DC activation (73). In this regard, using
endogenous IFN-I inducers instead of recombinant cytokines may have advantages, because substances such as
the viral RNA analog poly I:C will also elicit other cytokines that may act in concert with IFNs-I to enhance the
antitumor immune response.
Creative combination strategies are building on the
knowledge that IFN-a effects on the immune system
are likely to improve its therapeutic profile against malignant diseases. For instance, combinations with immunostimulatory monoclonal antibodies, IL-15, IL-12, and
IL-21, may hold much promise. Indeed, intratumoral
release of mouse IFN-a along with systemic treatment
with anti-CD137 mAb results in synergistic therapeutic
effects (74). Macrophages homing to tumor, transduced
to express IFN-a, exert therapeutic effects without the
systemic side effects (75). These studies suggest that local
delivery as opposed to systemic administration should be
tested. Of great importance is the notion that the chemokines induced by IFN-I (i.e., CXCL9, CXCL10) attract
activated lymphocytes, thereby calling more immune
cells to infiltrate the malignant focus.
When reflecting on the use of IFN-I for cancer, we must
think about reprogramming the tumor microenvironment, frequently devoid of any stimulatory signals. Local
IFN-I release with concomitant irradiation and chemotherapy may help to turn some of the macroscopic tumor
lesions into immunogenic vaccines. Factors such as CpG
oligonucleotides or poly I:C that induce the release of
endogenous IFNs-I at a given tumor lesion (76) are
promising in this regard. These agents may constitute
an advantageous alternative because these agents also
induce an additional array of proinflammatory mediators
acting in synergy with IFNs-I.

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

2623

Hervas-Stubbs et al.

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


Indication

Stage

Comment

Standard Current Therapy

Reference

Melanoma

II/III

Prolongs disease-free
survival and shows a trend
toward increased overall
survival.
Combination with
IL2: Increased response
rates but no survival
improvement.
Two phase III trials showed
negative results.

IFN-a is the standard option.

68

Chemotherapy. The advent


of CTLA-4 antagonist antibody
will displace the use of IFN-a
for melanoma inmunotherapy.
Novel targeted agents such
as sunitinib, temsirolimus,
sorafenib, or everolimus
have decreased the use of
IFN-a to a minimum in both
resectable and advanced RCC.

77, 78

IV

RCC

II/III

IV

Hairy cell leukemia

Multiple myeloma

CMS

PV

ET

PM
CML

Widely studied as a single


agent or in combination with
IL-2, chemotherapy, or both:
modest or negative results.
First treatment that showed
clinical benefit in this disease.

Recommended, in low
dose, as a therapeutic
option for maintenance
therapy and as an
alternative for frail
patients.
Extensively studied as
maintenance treatment,
as single treatment, or in
combination with
chemotherapy: slight
efficacy.
Optional treatment for
cytoreduction (94.6%
complete responses).
Treatment of choice
in women with
childbearing potential
and optional treatment
in young patients
resistant to
hydroxyurea.
Cytoreduction therapy.
First-line therapy in
combination with
cytarabine: 71% clinical
response rate and 39% major
cytogenetic response rate.

79, 80

81

Purine analogs are now


considered standard
treatment for this
disease.

82, 83

Largely replaced by newer


and more effective agents
such as thalidomide,
lenalidomide, or bortezomib.

84

The mainstay of therapy


remains repeated phlebotomy.

8587

Hydroxyurea. Anagrelide.

86

Hydroxyurea. Thalidomide.
Imatinib, in a phase III trial,
has been superior in
tolerability, complete
hematologic and cytogenetic
response rates, and
progression-free survival.

77
8892

(Continued on the following page)

2624

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

Clinical Cancer Research

IFN-a on Immune Cells

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


Indication

Hemangioma

AIDS-related Kaposi
sarcoma

Stage

Comment

Standard Current Therapy

Reference

In comparison with
chemotherapy IFNa
has been shown to
be superior.
Complicate hemangiomas that
do not respond to steroids.
Rate regression in
58% of the patients.
Evidence for clinical activity.

In imatinib resistance, IFNa


has been precluded by the
appearance
of dasatinib.
Steroids.

93

Liposomal anthracyclines are


preferred as the first
treatment option.

94

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

In conclusion, the notion of key direct effects of IFNs-I


on immune system cells and detailed knowledge on the
elicited signaling pathways should help biomarker discovery to identify the small number of patients who could
actually benefit from current treatments. More importantly,
these ideas might change the way we use IFN-a for cancer
therapy, suggesting new strategies and combinations with
other agents.
Disclosure of Potential Conflicts of Interest
I. Melero, A. Rouzaut, and S. Hervas-Stubbs receive laboratory reagents
and research funding from Digna Biotech. I. Melero has acted as a consultant
for Bristol-Myers Squibb and Pfizer Inc.

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

Grant Support
MEC/MCI (SAF200503131, SAF200803294, and TRA20090030), Departamento de Educacion y Departamento de Salud (Beca Ortiz de Landazuri) del
Gobierno de Navarra. Redes tematicas de investigaci
on cooperativa RETIC
(RD06/0020/0065), Fondo de investigaci
on sanitaria (FIS PI060932), European
commission VII framework program (ENCITE), Immunonet SUDOE. Fundaci
on
Mutua Madrilea, and "UTE for project FIMA". S. Hervas-Stubbs was supported
by AECC and by MCI (RYC-200700928).
Received October 22, 2010; revised January 17, 2011; accepted January
17, 2011; published OnlineFirst March 3, 2011.

References
1.
2.
3.
4.
5.

6.

7.

8.

9.

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


Soc Lond B Biol Sci 1957;147:25867.
Pestka S, Krause CD, Walter MR. Interferons, interferon-like cytokines, and their receptors. Immunol Rev 2004;202:832.
Vilcek J. Novel interferons. Nat Immunol 2003;4:89.
Miller CH, Maher SG, Young HA. Clinical Use of Interferon-gamma.
Ann N Y Acad Sci 2009;1182:6979.
de Weerd NA, Samarajiwa SA, Hertzog PJ. Type I interferon receptors:
biochemistry and biological functions. J Biol Chem 2007;282:20053
7.
Hwang SY, Hertzog PJ, Holland KA, Sumarsono SH, Tymms MJ,
Hamilton JA, et al. A null mutation in the gene encoding a type I
interferon receptor component eliminates antiproliferative and antiviral responses to interferons alpha and beta and alters macrophage
responses. Proc Natl Acad Sci U S A 1995;92:112848.
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 I
interferon. Nat Immunol 2003;4:100915.
Le Bon A, Schiavoni G, DAgostino G, Gresser I, Belardelli F, Tough
DF. Type i interferons potently enhance humoral immunity and can
promote isotype switching by stimulating dendritic cells in vivo.
Immunity 2001;14:46170.
Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated
signalling. Nat Rev Immunol 2005;5:37586.

www.aacrjournals.org

10. Aaronson DS, Horvath CM. A road map for those who don't know
JAK-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 transcriptional activation in response to IFNs and other extracellular signaling
proteins. Science 1994;264:141521.
hrl S, Stoiber D, Decker T. Activa13. Meinke A, Barahmand-Pour F, Wo
tion of different Stat5 isoforms contributes to cell-type-restricted
signaling in response to interferons. Mol Cell Biol 1996;16:693744.
14. Farrar JD, Smith JD, Murphy TL, Murphy KM. Recruitment of Stat4 to
the human interferon-alpha/beta receptor requires activated Stat2. J
Biol Chem 2000;275:26937.
15. Fasler-Kan E, Pansky A, Wiederkehr M, Battegay M, Heim MH.
Interferon-alpha activates signal transducers and activators of transcription 5 and 6 in Daudi cells. Eur J Biochem 1998;254:5149.
16. Matikainen S, Sareneva T, Ronni T, Lehtonen A, Koskinen PJ, Julkunen I. Interferon-alpha activates multiple STAT proteins and upregulates proliferation-associated IL-2Ralpha, c-myc, and pim-1 genes
in human T cells. Blood 1999;93:198091.
17. Torpey N, Maher SE, Bothwell AL, Pober JS. Interferon alpha but not
interleukin 12 activates STAT4 signaling in human vascular endothelial
cells. J Biol Chem 2004;279:2678996.
18. Parmar S, Platanias LC. Interferons: mechanisms of action and clinical
applications. Curr Opin Oncol 2003;15:4319.

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

2625

Hervas-Stubbs et al.

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 interferon
gamma: 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 of
Stat3 in mouse embryonic fibroblasts. Biochem Biophys Res Commun 2005;327:72733.
24. Alexander WS. Suppressors of cytokine signalling (SOCS) in the
immune system. Nat Rev Immunol 2002;2:4106.
25. Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and
immune 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 dendritic cell activation and systemic autoimmunity. Immunity 2003;
19:43750.
ke B, Meinecke J, et al.
27. Zitzmann K, Brand S, De Toni EN, Baehs S, Go
SOCS1 silencing enhances antitumor activity of type I IFNs by
regulating apoptosis in neuroendocrine tumor cells. Cancer Res
2007;67:502532.
28. Du Z, Wei L, Murti A, Pfeffer SR, Fan M, Yang CH, et al. Nonconventional signal transduction by type 1 interferons: the NF-kappaB
pathway. J Cell Biochem 2007;102:108794.
29. Ahmed Z, Beeton CA, Williams MA, Clements D, Baldari CT, Ladbury
JE. Distinct spatial and temporal distribution of ZAP70 and Lck
following stimulation of interferon and T-cell receptors. J Mol Biol
2005;353:100110.
30. Petricoin EF 3rd, Ito S, Williams BL, Audet S, Stancato LF, Gamero A,
et al. Antiproliferative action of interferon-alpha requires components
of T-cell-receptor signalling. Nature 1997;390:62932.
31. Stevens CN, Simeone AM, John S, Ahmed Z, Lucherini OM, Baldari
CT, et al. T-cell receptor early signalling complex activation in
response to interferon-alpha receptor stimulation. Biochem J
2010;428:42937.
32. Trinchieri G, Santoli D. Anti-viral activity induced by culturing lymphocytes with tumor-derived or virus-transformed cells. Enhancement of human natural killer cell activity by interferon and antagonistic
inhibition 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 Immunol
2002;169:427987.
34. Zhang X, Sun S, Hwang I, Tough DF, Sprent J. Potent and selective
stimulation 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. Distinct
requirements for IFNs and STAT1 in NK cell function. J Immunol
2000;165:35717.
36. Trinchieri G, Santoli D, Granato D, Perussia B. Antagonistic effects of
interferons on the cytotoxicity mediated by natural killer cells. Fed
Proc 1981;40:270510.
37. Ortaldo JR, Phillips W, Wasserman K, Herberman RB. Effects of
metabolic inhibitors on spontaneous and interferon-boosted human
natural killer cell activity. J Immunol 1980;125:183944.
38. Bogdan C, Mattner J, Schleicher U. The role of type I interferons in
non-viral infections. Immunol Rev 2004;202:3348.
39. Sampson LL, Heuser J, Brown EJ. Cytokine regulation of complement
receptor-mediated ingestion by mouse peritoneal macrophages. MCSF and IL-4 activate phagocytosis by a common mechanism requiring autostimulation by IFN-beta. J Immunol 1991;146:100513.
40. Lapenta C, Santini SM, Logozzi M, Spada M, Andreotti M, Di Pucchio
T, et al. Potent immune response against HIV-1 and protection from
virus challenge in hu-PBL-SCID mice immunized with inactivated
virus-pulsed dendritic cells generated in the presence of IFN-alpha.
J Exp Med 2003;198:3617.

2626

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

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-derived
dendritic cell development and activity in vitro and in Hu-PBL-SCID
mice. 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 a
short-term culture with IFN-alpha and granulocyte-macrophage colony-stimulating factor stimulate a potent Epstein-Barr virus-specific
CD8 T cell response. J Immunol 2003;170:5195202.
43. Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous
activators of dendritic cells. Nat Med 1999;5:124955.
44. Ito T, Amakawa R, Inaba M, Ikehara S, Inaba K, Fukuhara S. Differential regulation of human blood dendritic cell subsets by IFNs. J
Immunol 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 their
phenotypic 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-like
receptor-induced interleukin-12p70 secretion by dendritic cells. J Exp
Med 2005;201:143546.
47. Byrnes AA, Ma X, Cuomo P, Park K, Wahl L, Wolf SF, et al. Type I
interferons and IL-12: convergence and cross-regulation among
mediators 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 I
IFN-induced monocyte-derived dendritic cells: importance for the
rapid acquisition of potent migratory and functional activities. Blood
2001;98:30229.
49. Asselin-Paturel C, Boonstra A, Dalod M, Durand I, Yessaad N, Dezutter-Dambuyant C, et al. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nat Immunol 2001;2:1144
50.
50. Rouzaut A, Garasa S, Teijeira A, Gonzalez I, Martinez-Forero I, Larrea
E, et al. Dendritic cells adhere to and transmigrate across lymphatic
endothelium in response to IFN-a. Eur J Immunol 2010;40:305463.
 N, Xu Y, An J, Lanier LL, et al. CD69
51. Shiow LR, Rosen DB, Brdickova
acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte 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 specifically
enhance antiviral humoral responses. Eur J Immunol 2006;36:2094
105.
53. Le Bon A, Thompson C, Kamphuis E, Durand V, Rossmann C, Kalinke
U, et al. Cutting edge: enhancement of antibody responses through
direct stimulation of B and T cells by type I IFN. J Immunol 2006;176:
20748.
54. Coro ES, Chang WL, Baumgarth N. Type I IFN receptor signals directly
stimulate local B cells early following influenza virus infection. J
Immunol 2006;176:434351.
55. Brinkmann V, Geiger T, Alkan S, Heusser CH. Interferon alpha
increases the frequency of interferon gamma-producing human
CD4 T cells. J Exp Med 1993;178:165563.
56. Nguyen KB, Watford WT, Salomon R, Hofmann SR, Pien GC, Morinobu A, et al. Critical role for STAT4 activation by type 1 interferons in
the interferon-gamma response to viral infection. Science 2002;297:
20636.
57. Marrack P, Kappler J, Mitchell T. Type I interferons keep activated T
cells 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 expansion
and memory formation in response to viral infection. J Exp Med
2005;202:63750.
59. Aichele P, Unsoeld H, Koschella M, Schweier O, Kalinke U, Vucikuja S.
CD8 T cells specific for lymphocytic choriomeningitis virus require
type 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 enhances
the CD8 T cell response during cross-priming. J Immunol 2006;176:
46829.

Clinical Cancer Research

IFN-a on Immune Cells

61. Thompson LJ, Kolumam GA, Thomas S, Murali-Krishna K. Innate


inflammatory signals induced by various pathogens differentially
dictate the IFN-I dependence of CD8 T cells for clonal expansion
and 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 clonal
expansion 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-12
and type I IFN in programming for CD8 T cell effector function and
memory. 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 cytokine on human nave
and antigen-experienced CD8 T-cells. Eur J Immunol 2010;40:3389402.
65. Marshall HD, Prince AL, Berg LJ, Welsh RM. IFN-alpha beta and selfMHC divert CD8 T cells into a distinct differentiation pathway characterized by rapid acquisition of effector functions. J Immunol
2010;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. Polyethylene
glycol-interferon: current status in hepatitis C virus therapy. J Gastroenterol Hepatol 2002;17[Suppl 3]:S34450.
68. Garbe C, Eigentler TK. Diagnosis and treatment of cutaneous melanoma: state of the art 2006. Melanoma Res 2007;17:11727.
69. Hanley PJ, Cruz CR, Savoldo B, Leen AM, Stanojevic M, 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 blood
and 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 interferonbeta 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 as
vaccine adjuvants against infectious diseases and cancer. Expert Rev
Vaccines 2008;7:37381.
72. Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, et al.
Chemokine expression in melanoma metastases associated with
CD8 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-associated
antigen peptides and clinical activity by vaccinations with alpha-type
1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent
malignant glioma. J Clin Oncol 2011;29:3306.
 n A, Alfaro C, Azpilikueta A, Ochoa MC, Rouzaut A,
74. Dubrot J, Palazo
et al. Intratumoral injection of interferon-a and systemic delivery of
agonist anti-CD137 monoclonal antibodies synergize for immunotherapy. Int J Cancer 2011;128:10518.
75. De Palma M, Mazzieri R, Politi LS, Pucci F, Zonari E, Sitia G, et al. Tumortargeted interferon-alpha delivery by Tie2-expressing monocytes inhibits tumor growth and metastasis. Cancer Cell 2008;14:299311.
76. Brody JD, Ai WZ, Czerwinski DK, Torchia JA, Levy M, Advani RH, et al.
In situ vaccination with a TLR9 agonist induces systemic lymphoma
regression: 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-based
interdisciplinary guideline. Eur J Cancer 2010;46:27083.
78. Hodi FS, ODay SJ, McDermott DF, Weber RW, Sosman JA, Haanen
JB, et al. Improved survival with ipilimumab in patients with metastatic
melanoma. N Engl J Med 2010;363:71123.

www.aacrjournals.org

79. Messing EM, Manola J, Wilding G, Propert K, Fleischmann J, Crawford ED, et al. Eastern Cooperative Oncology Group/Intergroup trial.
Phase III study of interferon alfa-NL as adjuvant treatment for resectable 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 III
renal cell carcinoma: a multicentric randomized study. J Clin Oncol
2001;19:42531.
81. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM,
Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell
carcinoma. N Engl J Med 2007;356:11524.
82. Quesada JR, Reuben J, Manning JT, Hersh EM, Gutterman JU. Alpha
interferon for induction of remission in hairy-cell leukemia. N Engl J
Med 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 or
in combination on the inhibitory effects of VEGF on human dendritic
cell 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 polycythemia
vera, essential thrombocythemia, and primary myelofibrosis in the
JAK2 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 myelofibrosis. 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 patients
treated with pegylated interferon alpha-2a. Blood 2006;108:2037
40.
88. Talpaz M, Kantarjian HM, McCredie KB, Keating MJ, Trujillo J, Gutterman J. Clinical investigation of human alpha interferon in chronic
myelogenous leukemia. Blood 1987;69:12808.
89. Baccarani M, Rosti G, de Vivo A, Bonifazi F, Russo D, Martinelli G, et
al; Italian Cooperative Study Group on Myeloid Leukemia. A randomized study of interferon-alpha versus interferon-alpha and low-dose
arabinosyl cytosine in chronic myeloid leukemia. Blood 2002;99:
152735.
90. Guilhot F, Chastang C, Michallet M, Guerci A, Harousseau JL, Maloisel F, et al; French Chronic Myeloid Leukemia Study Group. Interferon alfa-2b combined with cytarabine versus interferon alone in
chronic 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 transplantation
in treating the chronic phase of chronic myeloid leukemia: developed
for the American Society of Hematology. Blood 1999;94:1517
36.
92. OBrien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F, et al. IRIS Investigators. Imatinib compared with interferon
and low-dose cytarabine for newly diagnosed chronic-phase chronic
myeloid leukemia. N Engl J Med 2003;348:9941004.
93. Greinwald JH Jr, Burke DK, Bonthius DJ, Bauman NM, Smith RJ. An
update on the treatment of hemangiomas in children with interferon
alfa-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 sarcoma: current status and perspectives. Cancer Treat Rev 2006;32:
44555.

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

2627

You might also like