27
1 Novel insights into the role of IL-27 and IL-23 in human malignant and normal plasma cells Nicola Giuliani 1 and Irma Airoldi 2 1 Hematology and BMT Center, University of Parma, Parma, Italy 2 AIRC Laboratory of Immunology and Tumors, Department of Experimental and Laboratory Medicine, G. Gaslini Institute, Genova, Italy Corresponding Author Irma Airoldi, Ph.D., A.I.R.C. Laboratory of Immunology and Tumors Department of Experimental and Laboratory Medicine G. Gaslini Institute, 16148 Genova, Italy Phone: +390105636342, Fax: +39010377982 E-mail: [email protected] Research. on December 21, 2018. © 2011 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

1 Novel insights into the role of IL-27 and IL-23 in human malignant

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

1

Novel insights into the role of IL-27 and IL-23 in human malignant and normal plasma cells

Nicola Giuliani1 and Irma Airoldi2

1Hematology and BMT Center, University of Parma, Parma, Italy

2AIRC Laboratory of Immunology and Tumors, Department of Experimental and Laboratory

Medicine, G. Gaslini Institute, Genova, Italy

Corresponding Author

Irma Airoldi, Ph.D.,

A.I.R.C. Laboratory of Immunology and Tumors

Department of Experimental and Laboratory Medicine

G. Gaslini Institute, 16148 Genova, Italy

Phone: +390105636342, Fax: +39010377982

E-mail: [email protected]

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

2

ABSTRACT

Multiple myeloma (MM) is a monoclonal post-germinal center tumor that has phenotypic features

of plasmablasts/plasma cells and usually localizes at multiple sites in the bone marrow.

Pathogenesis of MM is complex and dependent on the interactions between tumor cells and their

microenvironment. Different cytokines, chemokines and pro-angiogenic factors released in the

tumor microenvironment are known to promote MM cell growth. Here we report recent advances

on the role of two stricktly related immunomodulatory cytokines, i.e. IL-27 and IL-23, in human

normal and neoplastic plasma cells highlighting their ability to i) act directly against MM cells, ii)

influence MM microenvironment by targeting osteoclast and osteoblast cells and iii) modulate

normal plasma cell function. Finally, the therapeutic implication of these studies will be discussed.

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

3

BACKGROUND

Interleukin (IL)-27 and IL-23 are immunomodulatory cytokines belonging to the IL-12 superfamily

(1), composed of structurally and functionally related heterodimeric cytokines including IL-12, IL-

23, IL-27 and IL-35 (1). These cytokines are predominantly produced by antigen presenting cells in

response to microbial or host immune stimuli and are involved in the regulation of immune

responses against infections and tumor development (2). Recent pre-clinical studies demonstrated

the direct anti-tumor activities of IL-12 family cytokines in different human hematological

malignancies including pediatric acute leukemias and multiple myeloma, as well as in solid tumors

(3-7). Here, we reported and discuss recent advances on the role of IL-27 and IL-23 in multiple

myeloma (MM) cells and its microenvironment, as well as in human normal plasma cells.

Interleukin-27

IL-27 is composed of the EBI3 and p35 subunits and activates both signal transducer and activator

of transcription (STAT)1 and STAT3 through distinct IL-27 receptor (R) that consists of the unique

receptor subunit WSX-1 paired with the gp130 chain (8). In T lymphocytes, the cytokine induces

STAT1 and STAT3 activation thus resulting in enhancement of naïve cluster of differentiation

(CD)4+ T cell proliferation, promotion of the early T helper (Th)1 differentiation and suppression of

the differentiation of Th2 and Th17 cells. In addition IL-27 plays a key role in generating IL-10

producing regulatory T cells (1, 9).

In human tonsils, the different B cell subsets show variable expression of IL-27R (10). In this study

it has been demonstrated that naïve and memory B lymphocytes express constitutively both chains

of IL-27R whereas germinal center B cells exhibited only barely detectable levels of the WSX-1

chain. However, the WSX-1 was found to be strongly up-regulated during germinal center to

memory B cell transition (10). In terms of signal transduction, IL-27 induces in naïve B cells a

strong phosphorylation of both STAT1 and STAT3, whereas a moderate STAT1 and a low

activation of STAT3 were observed in memory B lymphocytes (10). In addition, human plasma

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

4

cells (PC) express constitutively WSX-1 and gp130 and IL-27 specifically induces STAT-1

phosphorylation while unaffecting STAT-3 or STAT-5 activation (5). Differences in the STAT

activation driven by IL-27 may reflect the different ability of human B cell subsets to respond to IL-

27. For example, IL-27 enhances proliferation in naïve and activated germinal center B cells (10),

induces surface expression of CD54, CD95 and CD86 in stimulated memory B cells (10) and exerts

chemotactic activity on PC (5).

Anti-tumor activity of IL-27 against myeloma cells

IL-27, similarly to IL-12, shows anti-tumor activity in different cancers through indirect

mechanisms such as induction of natural killer (NK) and cytotoxic T lymphocyte (CTL) response or

inhibition of angiogenesis primarily due to induction of CXCL10 and CXCL9 (3, 11-14). In

addition, it has been reported that IL-27 inhibits directly the growth of melanoma cell lines (7) and

B-acute lymphoblastic leukemia cells (12) expressing complete IL-27R. In multiple myeloma (MM)

it has been reported that both chains of IL-27 receptor are expressed in CD138+ MM cells from

patients (5, 11). Although IL-27 does not modulate MM cell proliferation and apoptosis, a strong

reduction of the angiogenic potential of MM cells was clearly documented (5). It is of note that BM

neo-vascularization contributes to the biology of several hematologic malignancies (15) and MM

neo-angiogenesis may be induced by angiogenic cytokines produced by tumor and

microenvironmental cells (16). Cytokines such as basic fibroblast growth factor (bFGF), vascular

endothelial growth factor (VEGF) and angiopoietins (ANGPT) play a leading role in new vessel

formation as well as in tumor angiogenesis (17-19). However, other known and unknown factors

may be also involved and are under investigation. We reported that IL-27 down-regulates in vitro of

a wide panel of pro-angiogenic genes including VEGF-A, -C and –D, angiopoietins, and matrix

metalloproteinases and up-regulates the anti-angiogenic chemokines CXCL9 and CXCL10 (Figure

1) (11). Accordingly, pre-clinical studies using highly immunodeficient SCID/NOD mice injected

with MM cells revealed that IL-27 inhibits the MM cell growth through inhibition of angiogenesis,

that caused ischemic necrosis in the tumors. In this model, the main pro-angiogenic molecules and

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

5

MM growth factors down-regulated by IL-27 were IL-6, VEGF-D and CCL2 that serve also as

autocrine and/or paracrine growth factors (20, 21). Thus, we can envisage that IL-27 may interrupt

or reduce the “growth loop” (Figure 1) in which cytokines and growth factors released by MM cells

induce paracrine stimulation of stromal cells that, in turn, secrete additional growth factors such as

IL-6 and VEGF that induce new vessel formation and support MM cell proliferation, progression

and migration. In addition, MM cells and BM stromal cells produce matrix metalloproteinases

(MMP) that degrade collagens and fibronectin, thus rendering MM cells capable of invading both

stroma and the subendothelial basement membrane (Figure 1) (22-25). The enhanced invasive

ability of MM cells along with their increased angiogenic capacity may explain the frequent

intramedullary and extramedullary dissemination observed in MM pathogenesis. Pre-clinical studies

showed that IL-27 down-regulates MMP9, thus suggesting that IL-27 may also reduced the MM

cells invasion (Figure 1) (11).

Interleukin-23

IL-23 is a heterodimeric cytokine composed of the promiscuous IL-12p40 and the exclusive p19

subunits(26). The IL-23 receptor (R) is composed of the IL-12Rβ1 chain and the unique IL-23R

chain, that associated with Jak2 and STAT3 (26). In T lymphocytes and leukemic T cells, IL-23

induces activation of STAT family members, i.e. STAT1, 3, 4 and STAT5 (26). IL-23 is produced

predominantly by myeloid dendritic cells (DC) activated by Toll like receptor-2, -4 and -8 ligands

and by type 1 macrophages (27, 28). This cytokine is now considered the master switch in several T

cell mediated inflammatory disorders whereas its anti-tumor activity of IL-23 remains still

controversial. In this context, it has been reported that pro-inflammatory cytokines including IL-

17A, IL-6, and IL-23 can impair CD8+ T lymphocytes mediated immune surveillance, and promote

de novo carcinogenesis and tumor neo-vascularization (29-31). In contrast, other groups have shown

that IL-23 exerts anti-tumor activity through stimulation of T and NK cells (32-37).

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

6

Actually any information is not available on IL-23R expression and function in normal human B

cells with the only exception for plasma cells and early B lymphocytes (4, 5). In this context, it has

been demonstrated that human plasma cells express complete IL-23 receptors and that IL-23 may

modulate normal plasma cell functions (5). However the signal transduction activated by IL-23

treatment has never been investigated in these cells. On the contrary, it has been reported that in

human normal early B cells IL-23 induces a specific activation of the STAT3 molecule through

phosphorylation of serine727 and tyrosine 701 residues, while unaffecting the activation of STAT1,

4, 5 and 6 (4).

Anti-tumor activity of IL-23 against myeloma and B-acute lymphoblastic leukemia (ALL).

Although complete IL-23 receptor is expressed in MM cells ((3) and Airoldi I. personal

communication), IL-23 is ineffective in terms of modulation of MM cell proliferation and

angiogenesis, induction of apoptosis and chemotaxis, regulation of chemokine receptor expression

and cytokine production. The mechanisms underlying the lack of function of IL-23R in these cells

remain to be established.

A different scenario has been envisaged using IL-23 as potential anti-tumor agent against (4)

pediatric B-acute lymphoblastic leukemia (ALL). In this context, it has been demonstrated that IL-

23 dampens B-ALL cell growth in vitro and in pre-clinical models by inhibiting proliferation and

inducing apoptosis. Such effects were related to IL-23 induced up-regulation of miR-15a in

leukemic cells and to the consequent down-regulation of the target proteins BCL-2 and Cyclin D1

(38). These two molecules function as oncogenes, play an important role in the regulation of

apoptosis and proliferation respectively and were found to be over-expressed in different tumors.

The proof of evidence that mir-15a was sufficient and necessary for IL-23 anti-tumor activity

against pediatric B-ALL cells has been obtained using two different and complementary

approaches. Thus, it has been investigated whether i) silencing of mir-15a in B-ALL cells abolished

the effects driven by IL-23 and ii) forced expression of miR15a had a similar anti-tumor activity to

the treatment of IL-23. In vitro and in vivo experiments allowed to demonstrate unambiguously that

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

7

the anti-leukemic function of IL-23 was strictly associated to the up-regulation of miR-15a and that

enforced expression of miR-15a had a similar anti-tumor activity to the treatment of IL-23 causing a

clear inhibition of leukemic cell growth in vivo (4). Although the molecular mechanisms underlying

regulation of miR-15a mediated by IL-23 were not elucidated, it was hypothesized that, similarly to

other cytokines, components of IL-23 signaling pathway may interfere in the biogenesis or

transcription of miR-15a. This pre-clinical study provided the first evidence that cytokines-mediated

regulation of miRNA expression pattern play a key role in the control of B-ALL cell growth.

Finally, studies on the mechanisms underlying the pro-apoptotic effect of IL-23 showed that the

cytokine not only down-regulated the anti-apoptotic BCL-2 but also induced PARP cleavage in the

absence of caspase activation (4).

IL-23 and Th17 function in MM

IL-23 is a well known Th17 differentiating cytokine (39). Recent data highlighted that Th17 play an

important role in MM pathobiology and may be an important therapeutic target for anti-MM

activity and to improve immune function (40). In this view, a number of Th17-associated cytokines,

including IL-23, IL-21, IL-22 and IL-17, were found to be significantly elevated in MM compared

with healthy donors and elevated IL-17 serum levels have been correlated with stage II and III MM

(41). In addition, IL-23 was associated with reduced CD8+ T-cell infiltration in the tumor micro-

environment (30) and a combination of Th17-associated pro-inflammatory cytokines may suppress

T-cell responses. This recent evidence suggests a potential role of IL-23 in Th17-mediated

promotion of MM cell growth and inhibition of immune function. Finally, an imbalance between

Treg/Th17 in favor of Th17, that are not adequately suppressed by T regulatory (reg) cells, may be

suggested since has been reported that Treg cells are decreased in number and dysfunctional in MM

patients (42).

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

8

Bone remodeling in MM

Bone destruction is a hallmark of MM (43) and approximately 85-90% of MM patients present lytic

bone lesions frequently associated with severe bone pains and fractures. Osteolytic lesions result

from increased bone resorption due to stimulation of osteoclast formation and a severely

unbalanced bone remodelling decreased or absent bone formation activity that occur close to MM

cells (44, 45).

The critical role of the receptor activator of nuclear factor kappaB ligand (RANKL), in MM-

induced osteoclastogenesis (44) has been demonstrated. RANKL is over-expressed in the BM in

relationship with MM cell infiltration whereas its decoy receptor osteoprotegerin (OPG) is down-

regulated (44). Other cytokines produced by MM cells such as Macrophage Inhibitory Protein

(MIP)-1α (namely CCL3) or induced in the microenvironment by the cell contact are responsible

for enhancement of osteoclast differentiation and activity as well as for inhibition of osteoblast

functions (46). Interestingly, the growth factors released in the course of increased bone resorptive

process as well as activated osteoclasts in turn support MM cell growth and survival, thus inducing

a “vicious cycle”(47-49). Consistently it has been reported that anti-osteoclastic agents may exert

anti-MM effect both in vitro and in vivo (50).

Effects of IL-27 and IL-23 on osteoblastic and osteoclastic cells and bone remodelling

Recent evidences suggest that both IL-27 and IL-23 may affect in a different manner the bone

remodeling process. Osteoclast progenitors from MM patients and controls (5) express functional

IL-27R and IL-27 dampens directly osteoclast differentiation and activity (11). It has been also

reported that IL-27 induced proliferation in osteoblastic cells while unaffecting osteoblast formation

(11, 51), indicating that IL-27 amplifies the mature osteoblastic compartment without modulating

its functionality. Thus, IL-27 may counteract the unbalanced osteoclast and osteoblast activity

induced by MM cells mainly exerting an inhibitory effect on osteoclastic cells.

The effect of IL-23 on osteoclast and bone resorption has been evaluated showing conflicting

results. Yago et al. (52) demonstrated that in human peripheral blood mononuclear cells, IL-23

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

9

induced osteoclastogenesis in the absence of exogenous soluble RANKL. In this study, IL-17

induced by IL-23 from human activated T cells was considered the pivotal cytokine for

osteoclastogenesis through the involvement of the RANK–RANKL system and tumor necrosis

factor (TNF)-α. Thus, IL-17 acted on monocytes and induced osteoclast differentiation and TNF-α

production by cooperating with RANKL. Similarly, it was reported that IL-23 stimulate RANKL

expression on CD4+ T cells and contribute to osteoclastogenesis in an arthritis mouse model (53). In

contrast, Quinn et al. reported that IL-23 inhibited osteoclastogenesis indirectly thought CD4+ T

cells and that IL-23p19-/- had reduced bone mass (54). Kamiya and coworkers (51) showed that IL-

23 was ineffective on RANKL expression and that osteoclastogenesis induced by soluble RANKL

was partially inhibited by IL-23, while unaffecting proliferation of osteoclast progenitors. Any

significant effect of IL-23 was reported on osteoblastic cells and on osteoblast formation (51).

Overall these evidences suggest that under physiological condition IL-23 favors high bone mass by

limiting bone resorption whereas in pathophisiological conditions IL-23 share a stimulatory effect

on osteoclast formation mainly through the induction of RANKL by T cells and the differentiation

of Th17 subset and IL-17 production. The role and the involvement of Th17 and IL-17 in osteoclast

activation in MM has been recently demonstrated (55). Because IL-23 is a well known cytokine

involved in Th17 differentiation and expansion, it is possible that the high BM levels of IL-23

observed in MM patients (40) contribute to the increased osteoclastogenesis through the expansion

of Th17 and consequently the overproduction of IL-17.

Human normal plasma cells

In humans, three major subsets of plasma cells have been isolated and are supposed to represent a

gradual increase in PC maturation: early PC in tonsils, transitional PC in peripheral blood and

mature PC in the BM (56). However, PC are rare cells in vivo representing only 1-2% of tonsil

mononuclear cells and less than 0.5% of BM cells in healthy individuals. Thus, it has been

developed an in vitro reproducible model of peripheral blood B cell differentiation into polyclonal

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

10

plasmablastic cells (PPC) that have morphological and phenotypic features of PC (57). In addition,

this model allowed to obtain a homogenous population of normal plasmablastic cells that provided a

powerful tool to identify genes specifically involved in normal plasma cell differentiation, to further

understand the biology of MM and to improve therapeutic developments..

Expression and role of I-L27 and IL-23 receptor in normal PC

The expression of both chains of the IL-27R and of IL-23R was demonstrated in tonsil and BM PC

as well as in PPC generated in vitro and the functional role of IL-27 and IL-23 was recently

elucidated (5). Since production of immunoglobulin (Ig) represents the main PC function and a key

step that occurs during antigen-specific immune response, we asked whether IL-27 and IL-23 may

modulate Ig secretion in these cells. These experiments showed that stimulation of PPC with IL-2 in

association with IL-27 or IL-23 caused significant up-regulation of IgM secretion paralleled by

inhibition of IgG secretion and unaltered IgA production. However, no effects were reported in

terms of modulation of IgG class switching (5, 10).

Thus, it has been hypothesized that these cytokines are effective on PPC that have not yet

undergone terminal differentiation into antibody-secreting cells since such effects were reverted

when IL-6 was present for the last days of PPC culture. This latter consideration was strongly

supported by the finding that IL-23 and IL-27 had no effect on modulation of Ig secretion in

CD138+ tonsil PC.

Another novel finding was the first demonstration that IL-27 possesses chemotactic properties

versus PC and PPC (5) conceivably related to dependent to STAT-1 activation, as recently

demonstrated for dendritic cells in response to cytokines, such as IFN-β, was (58).

Finally, IL-27 up-regulated in PC and PPC the expression and production of chemokines such as

CXCL9 that induce migration of activated T and NK cells and possess potent anti-angiogenic

activity through interferon (IFN)-γ dependent/independent pathways (11, 59, 60). However, in PPC

and PC, CXCL9 and 10 modulated expressions by IL-27 was not accompanied by induction of IFN-

γ production.

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

11

Taken together, these findings support the concept that IL-27 and more marginally IL-23 plays an

important role in the course of immunological response to pathogens by acting at different levels.

First, IL-23 and IL-27 produced by antigen-presenting cells stimulated by Toll-like receptor signals

may induce short-lived PC to produce low affinity IgM providing a rapid initial response to

pathogens. Thus, IL-27 may attract plasmablast/short-lived PC that up-regulate CXCL9, but not

CCL chemokines, thus creating chemokine gradients that recruit to inflamed tissue activated T,

memory T and NK cells but not granulocytes and monocytes. Finally, CXCL9 and 10 may limit

tissue damage provoked by persistent activation of inflammatory cells in view of their well known

anti-angiogenic activities. These findings may be also relevant in the MM pathogenesis since

normal PC take part in the MM microenvironment. Thus, the anti-angiogenic CXCL9 up-regulated

in normal PC may amplify the inhibition of angiogenesis caused by the direct activity of IL-27 on

MM cells.

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

12

CLINICAL-TRANSLATIONAL ADVANCES

Cytokines of the IL-12 family: potential clinical implications

The potential translational implication of the new evidences on the role of IL-23 and/or IL-27 in the

biology of normal and malignant PC is related to the ability of these cytokines to stimulate immune

responses, target directly tumor cells expressing the corresponding receptors and, at least for IL-27,

to inhibit tumor angiogenesis. Either the modulation of IL-12 family cytokines including IL-23 and

IL-27 or the direct use of these cytokines could be supposed in the future in clinical trials.

In the case of IL-12, pre-clinical studies demonstrated that this cytokine can target directly MM

cells and cause a strong reduction of MM pro-angiogenic potential, with marginal effects on MM

cell proliferation and survival (3). However a particular note of caution regarding the potential

therapeutic implication of IL-12, comes from the side effects that have been reported in clinical

trials addressing the anti-tumor activity of IL-12 against different malignancies.

The initial clinical studies have been performed in phase I trials with patients affected by advanced

malignancies (renal cancer, colon carcinoma and melanoma) to determine the optimal protocol of

treatment; the parameters analyzed were toxicity, maximum tolerated dose (MTD), biological and

anti-tumor activities, and route of administration. Both intravenous (i.v) (61-63) and sub-cutaneous

(s.c) (64, 65) injection schedules have been tested in patients at different times and doses. The IL-12

MTD was different in i.v. and s.c. route of injection, 500 ng/kg and 1000-1500 ng/kg respectively,

and s.c administration resulted in fewer and milder toxic effects than i.v. treatment. Although

preliminary studies suggested that IL-12 might be efficacious, subsequent trials did not confirm the

first encouraging results. Thus, despite an enhancement of immune response, the clinical efficacy

was minimal. Moreover, escalating doses of IL-12 induced serious side effects in patients. Good

therapeutic results have been obtained in patients affected by cutaneous T cell lymphoma, AIDS-

related Kaposi sarcoma and non-Hodgkin’s lymphoma which showed a clear increase in circulating

and tumor infiltrating T cells with objective responses of 56%, 50%, 43%, respectively (66-68). The

favourable results obtained in immunodeficient AIDS patients suffering from Kaposi’s sarcoma was

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

13

not surprising since this tumor is strongly angiogenic and angiogenesis inhibition is one of the most

prominent IL-12 activities. Thus, it was speculated that the anti-neoplastic activity of IL-12 was

largely dependent on its ability to inhibit angiogenesis.

In order to get over or reduce systemic toxicity, novel forms of IL-12 non-viral delivery such as

alum, liposome and polymer-based delivery, have shown in pre-clinical models promising effects

(69). A further refinement of tumor targeted IL-12 therapy would be the use of targeted

formulations of the cytokine such as that developed by Halin and coworkers (70), who have

generated a fusion protein containing an IL-12 moiety fused to a single chain monoclonal antibody

specific for tenascin-C, an isoform of this extacellular matrix protein expressed predominantly in

the tumor stroma. This fusion protein, however, has not yet been tested in clinical trials. More

recently, the effectiveness of electrogene therapy with IL-12 has been evaluated in preclinical

studies (71) and the anti-tumor efficacy was tested in different types of primary tumors, distantly

growing tumors and induced metastases. Thus, it has been shown that intra-tumoral IL-12

electrogene therapy has been proved to be very effective in local tumor control, having also a

systemic effect, whereas intramuscular and peri-tumoral IL-12 electrogene therapy had also a

pronounced systemic effect. The anti-tumor effectiveness of IL-12 electrogene therapy was due to

the induction of adaptive immunity and innate resistance and anti-angiogenic action.

A further promising area of investigation, with potentially relevant clinical applications, stems from

the emerging knowledge on both IL-23 and IL-27 have been used to effectively treat tumors in

murine models. Systemic administration of IL-23 has been demonstrated to be effective in mouse

tumor system including melanoma and fibrosarcoma (32, 72). In addition preclinical evidences

suggest that IL-23 is a promising candidate as a tumor vaccine adjuvant (37). Interestingly adverse

toxic effect of systemic treatment with IL-23 appear to be much less than those observed for IL-12

although some authors reported weight loss and inflammation of the intestinal tract on the basis of

IL-23 schedule treatment (37). On the other hand, mice injected intra-peritoneally with recombinant

IL-23 did not show splenomegaly and liver inflammation as compared to those treated with IL-12

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

14

(72). Therefore in a clinical perspective, the route of administration for IL-23 should be adequately

designed to avoid its adverse effects and to exert efficient anti-tumor effect.

Despite the several evidences describing the anti-tumoral effect of IL-23 in tumoral models, caution

in its clinical application would be justified due to either the role of IL-23 in inflammatory diseases

or the possible role of this cytokine in tumoral progression (30, 36, 73, 74). In fact in mice lacking

IL-23p19 or IL-23 receptor, IL-23 showed a tumoral promoting effect in contrast to its potent anti-

tumor effect. The reasons for this discrepancy are not known, however the potential clinical use of

this cytokines should be carefully evaluated. In addition, the IL-23 seems to be ineffective against

MM cells and its role in the control of MM microenvironment has not been clearly elucidated.

By contrast, we could envisage combinatorial approaches using IL-12 and IL-27 to promote T and

NK cell responses against MM in addition to their direct anti-tumor activity against MM and to the

IL-27 ability to stimulate the osteoblast proliferation and to inhibit osteoclast differentiation and

function. In this view, exciting results have been reported against melanoma and breast cancer using

IL-12 and IL-27 sequential gene therapy through intramuscular electroporation (75). More recently,

it has been reported the efficacy of IL-27 as anti-tumor agent against prostate cancer using a new

mode of gene delivery named 'sonoporation' that involved low-frequency ultrasound irradiation

(76). In this study, three models of immune-competent prostate adenocarcinoma have been used and

detailed characterizations of the tumor growth reduction, gene profile expression, and effector

cellular profiles have been reported. Results from this investigation suggested that IL-27 can be

effective in reducing tumor growth and can help enhance accumulation of effector cells in prostate

tumors in vivo (76).

In conclusion, pre-clinical studies support the concept that IL-27 may represent a novel, promising

therapeutic agent for patients affected by different oncologic diseases including potentially MM,

based upon its documented multifunctional activity. Before this step, the development of future

clinical trials to evaluate the toxicity and efficacy of IL-27 will be mandatory. An additional

argument in favor of this proposal is the low toxicity shown by IL-27 in animal models, likely in

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

15

relation to the low induction of IFN-γ in vivo (32). Clearly, dedicated studies are required to define

the clinical utility of IL-27 as anti-angiogenic and anti-tumoral agent in patients with solid cancer

and MM.

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

16

Acknowledgments

This work was supported by grants from Associazione Italiana Ricerca sul Cancro (A.I.R.C.)

Milano, Italy (grant number 4014 to I.A. and 8530 to N.G.), Italian Ministry of Health (RF, RC,

5/1000, Progetto Strategico Oncologico 2006 rif070701), International Myeloma Foundation (to

N.G.), Italian Ministry of Health-Progetti Regione Emilia Romagna and the and Special Program

Molecular Clinical Oncology 5 per mille (n. 9965).

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

17

References

1. Trinchieri G, Pflanz S and Kastelein RA. The IL-12 family of heterodimeric cytokines: new

players in the regulation of T cell responses. Immunity 2003;19: 641-4

2. Hunter CA. New IL-12-family members: IL-23 and IL-27, cytokines with divergent functions.

Nat Rev Immunol 2005;5: 521-31

3. Airoldi I, Cocco C, Giuliani N, Ferrarini M, Colla S, Ognio E, et al. Constitutive expression of

IL-12R beta 2 on human multiple myeloma cells delineates a novel therapeutic target. Blood

2008;112: 750-9

4. Cocco C, Canale S, Frasson C, Di Carlo E, Ognio E, Ribatti D, et al. Interleukin-23 acts as

antitumor agent on childhood B-acute lymphoblastic leukemia cells. Blood 2010;116: 3887-98

5. Cocco C, Morandi F and Airoldi I. Interleukin-27 and interleukin-23 modulate human plasmacell

functions. J Leukoc Biol 2011; 89:729-34

6. Ferretti E, Di Carlo E, Cocco C, Ribatti D, Sorrentino C, Ognio E, et al. Direct inhibition of

human acute myeloid leukemia cell growth by IL-12. Immunol Lett 2010;133: 99-105

7. Yoshimoto T, Morishima N, Mizoguchi I, Shimizu M, Nagai H, Oniki S, et al. Antiproliferative

activity of IL-27 on melanoma. J Immunol 2008;180: 6527-35

8. Pflanz S, Hibbert L, Mattson J, Rosales R, Vaisberg E, Bazan JF, et al. WSX-1 and glycoprotein

130 constitute a signal-transducing receptor for IL-27. J Immunol 2004;172: 2225-31

9. Xu M, Mizoguchi I, Morishima N, Chiba Y, Mizuguchi J and Yoshimoto T. Regulation of

antitumor immune responses by the IL-12 family cytokines, IL-12, IL-23, and IL-27. Clin Dev

Immunol 2010: 2010. pii: 832454

10. Larousserie F, Charlot P, Bardel E, Froger J, Kastelein RA and Devergne O. Differential effects

of IL-27 on human B cell subsets. J Immunol 2006;176: 5890-7

11. Airoldi I, Di Carlo E, Banelli B, Moserle L, Cocco C, Pezzolo A, et al. The IL-12Rbeta2 gene

functions as a tumor suppressor in human B cell malignancies. J Clin Invest 2004;113: 1651-9

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

18

12. Airoldi I, Di Carlo E, Cocco C, Taverniti G, D'Antuono T, Ognio E, et al. Endogenous IL-12

triggers an antiangiogenic program in melanoma cells. Proc Natl Acad Sci U S A 2007;104: 3996-

4001

13. Hisada M, Kamiya S, Fujita K, Belladonna ML, Aoki T, Koyanagi Y, et al. Potent antitumor

activity of interleukin-27. Cancer Res 2004;64: 1152-6

14. Shimizu M, Shimamura M, Owaki T, Asakawa M, Fujita K, Kudo M, et al. Antiangiogenic and

antitumor activities of IL-27. J Immunol 2006;176: 7317-24

15. Rajkumar SV, Leong T, Roche PC, Fonseca R, Dispenzieri A, Lacy MQ, et al. Prognostic value

of bone marrow angiogenesis in multiple myeloma. Clin Cancer Res 2000;6: 3111-6

16. Vacca A and Ribatti D. Bone marrow angiogenesis in multiple myeloma. Leukemia 2006;20:

193-9

17. Giuliani N, Colla S, Lazzaretti M, Sala R, Roti G, Mancini C, et al. Proangiogenic properties of

human myeloma cells: production of angiopoietin-1 and its potential relationship to myeloma-

induced angiogenesis. Blood 2003;102: 638-45

18. Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, et al. Vessel

cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science

1999;284: 1994-8

19. Uneda S, Matsuno F, Sonoki T, Tniguchi I, Kawano F and Hata H. Expressions of vascular

endothelial growth factor and angiopoietin-2 in myeloma cells. Haematologica 2003;88: 113-5

20. Podar K, Tai YT, Davies FE, Lentzsch S, Sattler M, Hideshima T, et al. Vascular endothelial

growth factor triggers signaling cascades mediating multiple myeloma cell growth and migration.

Blood 2001;98: 428-35

21. Vincent L, Jin DK, Karajannis MA, Shido K, Hooper AT, Rashbaum WK, et al. Fetal stromal-

dependent paracrine and intracrine vascular endothelial growth factor-a/vascular endothelial growth

factor receptor-1 signaling promotes proliferation and motility of human primary myeloma cells.

Cancer Res 2005;65: 3185-92

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

19

22. Barille S, Akhoundi C, Collette M, Mellerin MP, Rapp MJ, Harousseau JL, et al.

Metalloproteinases in multiple myeloma: production of matrix metalloproteinase-9 (MMP-9),

activation of proMMP-2, and induction of MMP-1 by myeloma cells. Blood 1997;90: 1649-55

23. Di Raimondo F, Azzaro MP, Palumbo G, Bagnato S, Giustolisi G, Floridia P, et al. Angiogenic

factors in multiple myeloma: higher levels in bone marrow than in peripheral blood. Haematologica

2000;85: 800-5

24. Vacca A, Ribatti D, Presta M, Minischetti M, Iurlaro M, Ria R, et al. Bone marrow

neovascularization, plasma cell angiogenic potential, and matrix metalloproteinase-2 secretion

parallel progression of human multiple myeloma. Blood 1999;93: 3064-73

25. Vacca A, Ribatti D, Ria R, Pellegrino A, Bruno M, Merchionne F, et al. Proteolytic activity of

human lymphoid tumor cells. Correlation with tumor progression. Dev Immunol 2000;7: 77-88

26. Parham C, Chirica M, Timans J, Vaisberg E, Travis M, Cheung J, et al. A receptor for the

heterodimeric cytokine IL-23 is composed of IL-12Rbeta1 and a novel cytokine receptor subunit,

IL-23R. J Immunol 2002;168: 5699-708

27. Carmody RJ, Ruan Q, Liou HC and Chen YH. Essential roles of c-Rel in TLR-induced IL-23

p19 gene expression in dendritic cells. J Immunol 2007;178: 186-91

28. Verreck FA, de Boer T, Langenberg DM, Hoeve MA, Kramer M, Vaisberg E, et al. Human IL-

23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert

immunity to (myco)bacteria. Proc Natl Acad Sci U S A 2004;101: 4560-5

29. Langowski JL, Kastelein RA and Oft M. Swords into plowshares: IL-23 repurposes tumor

immune surveillance. Trends Immunol 2007;28: 207-12

30. Langowski JL, Zhang X, Wu L, Mattson JD, Chen T, Smith K, et al. IL-23 promotes tumour

incidence and growth. Nature 2006;442: 461-5

31. Tartour E, Fossiez F, Joyeux I, Galinha A, Gey A, Claret E, et al. Interleukin 17, a T-cell-

derived cytokine, promotes tumorigenicity of human cervical tumors in nude mice. Cancer Res

1999;59: 3698-704

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

20

32. Oniki S, Nagai H, Horikawa T, Furukawa J, Belladonna ML, Yoshimoto T, et al. Interleukin-23

and interleukin-27 exert quite different antitumor and vaccine effects on poorly immunogenic

melanoma. Cancer Res 2006;66: 6395-404

33. Ugai S, Shimozato O, Yu L, Wang YQ, Kawamura K, Yamamoto H, et al. Transduction of the

IL-21 and IL-23 genes in human pancreatic carcinoma cells produces natural killer cell-dependent

and -independent antitumor effects. Cancer Gene Ther 2003;10: 771-8

34. Yuan X, Hu J, Belladonna ML, Black KL and Yu JS. Interleukin-23-expressing bone marrow-

derived neural stem-like cells exhibit antitumor activity against intracranial glioma. Cancer Res

2006;66: 2630-8

35. Hu J, Yuan X, Belladonna ML, Ong JM, Wachsmann-Hogiu S, Farkas DL, et al. Induction of

potent antitumor immunity by intratumoral injection of interleukin 23-transduced dendritic cells.

Cancer Res 2006;66: 8887-96

36. Kaiga T, Sato M, Kaneda H, Iwakura Y, Takayama T and Tahara H. Systemic administration of

IL-23 induces potent antitumor immunity primarily mediated through Th1-type response in

association with the endogenously expressed IL-12. J Immunol 2007;178: 7571-80

37. Overwijk WW, de Visser KE, Tirion FH, de Jong LA, Pols TW, van der Velden YU, et al.

Immunological and antitumor effects of IL-23 as a cancer vaccine adjuvant. J Immunol 2006;176:

5213-22

38. Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, et al. miR-15 and miR-16

induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A 2005;102: 13944-9

39. Korn T, Bettelli E, Oukka M and Kuchroo VK. IL-17 and Th17 Cells. Annu Rev Immunol

2009;27: 485-517

40. Prabhala RH, Pelluru D, Fulciniti M, Prabhala HK, Nanjappa P, Song W, et al. Elevated IL-17

produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple

myeloma. Blood 2010;115: 5385-92

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

21

41. Alexandrakis MG, Pappa CA, Miyakis S, Sfiridaki A, Kafousi M, Alegakis A, et al. Serum

interleukin-17 and its relationship to angiogenic factors in multiple myeloma. Eur J Intern Med

2006;17: 412-6

42. Prabhala RH, Neri P, Bae JE, Tassone P, Shammas MA, Allam CK, et al. Dysfunctional T

regulatory cells in multiple myeloma. Blood 2006;107: 301-4

43. Kyle RA and Rajkumar SV. Multiple myeloma. N Engl J Med 2004;351: 1860-73

44. Giuliani N, Colla S and Rizzoli V. New insight in the mechanism of osteoclast activation and

formation in multiple myeloma: focus on the receptor activator of NF-kappaB ligand (RANKL).

Exp Hematol 2004;32: 685-91

45. Roodman GD. Pathogenesis of myeloma bone disease. Blood Cells Mol Dis 2004;32: 290-2

46. Giuliani N and Rizzoli V. Myeloma cells and bone marrow osteoblast interactions: role in the

development of osteolytic lesions in multiple myeloma. Leuk Lymphoma 2007;48: 2323-9

47. Giuliani N, Mangoni M and Rizzoli V. Osteogenic differentiation of mesenchymal stem cells in

multiple myeloma: identification of potential therapeutic targets. Exp Hematol 2009;37: 879-86

48. Giuliani N, Rizzoli V and Roodman GD. Multiple myeloma bone disease: Pathophysiology of

osteoblast inhibition. Blood 2006;108: 3992-6

49. Roodman GD. Pathogenesis of myeloma bone disease. Leukemia 2009;23: 435-41

50. Fowler JA, Edwards CM and Croucher PI. Tumor-host cell interactions in the bone disease of

myeloma. Bone 48: 121-8

51. Kamiya S, Nakamura C, Fukawa T, Ono K, Ohwaki T, Yoshimoto T, et al. Effects of IL-23 and

IL-27 on osteoblasts and osteoclasts: inhibitory effects on osteoclast differentiation. J Bone Miner

Metab 2007;25: 277-85

52. Yago T, Nanke Y, Kawamoto M, Furuya T, Kobashigawa T, Kamatani N, et al. IL-23 induces

human osteoclastogenesis via IL-17 in vitro, and anti-IL-23 antibody attenuates collagen-induced

arthritis in rats. Arthritis Res Ther 2007;9: R96

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

22

53. Ju JH, Cho ML, Moon YM, Oh HJ, Park JS, Jhun JY, et al. IL-23 induces receptor activator of

NF-kappaB ligand expression on CD4+ T cells and promotes osteoclastogenesis in an autoimmune

arthritis model. J Immunol 2008;181: 1507-18

54. Quinn JM, Sims NA, Saleh H, Mirosa D, Thompson K, Bouralexis S, et al. IL-23 inhibits

osteoclastogenesis indirectly through lymphocytes and is required for the maintenance of bone mass

in mice. J Immunol 2008;181: 5720-9

55. Noonan K, Marchionni L, Anderson J, Pardoll D, Roodman GD and Borrello I. A novel role of

IL-17-producing lymphocytes in mediating lytic bone disease in multiple myeloma. Blood 116:

3554-63

56. Medina F, Segundo C, Campos-Caro A, Gonzalez-Garcia I and Brieva JA. The heterogeneity

shown by human plasma cells from tonsil, blood, and bone marrow reveals graded stages of

increasing maturity, but local profiles of adhesion molecule expression. Blood 2002;99: 2154-61

57. Tarte K, De Vos J, Thykjaer T, Zhan F, Fiol G, Costes V, et al. Generation of polyclonal

plasmablasts from peripheral blood B cells: a normal counterpart of malignant plasmablasts. Blood

2002;100: 1113-22

58. Yen JH, Kong W and Ganea D. IFN-beta inhibits dendritic cell migration through STAT-1-

mediated transcriptional suppression of CCR7 and matrix metalloproteinase 9. J Immunol 184:

3478-86

59. Kanegane C, Sgadari C, Kanegane H, Teruya-Feldstein J, Yao L, Gupta G, et al. Contribution of

the CXC chemokines IP-10 and Mig to the antitumor effects of IL-12. J Leukoc Biol 1998;64: 384-

92

60. Mahalingam S, Chaudhri G, Tan CL, John A, Foster PS and Karupiah G. Transcription of the

interferon gamma (IFN-gamma )-inducible chemokine Mig in IFN-gamma-deficient mice. J Biol

Chem 2001;276: 7568-74

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

23

61. Atkins MB, Robertson MJ, Gordon M, Lotze MT, DeCoste M, DuBois JS, et al. Phase I

evaluation of intravenous recombinant human interleukin 12 in patients with advanced

malignancies. Clin Cancer Res 1997;3: 409-17

62. Gollob JA, Mier JW, Veenstra K, McDermott DF, Clancy D, Clancy M, et al. Phase I trial of

twice-weekly intravenous interleukin 12 in patients with metastatic renal cell cancer or malignant

melanoma: ability to maintain IFN-gamma induction is associated with clinical response. Clin

Cancer Res 2000;6: 1678-92

63. Robertson MJ, Cameron C, Atkins MB, Gordon MS, Lotze MT, Sherman ML, et al.

Immunological effects of interleukin 12 administered by bolus intravenous injection to patients with

cancer. Clin Cancer Res 1999;5: 9-16

64. Motzer RJ, Rakhit A, Thompson JA, Nemunaitis J, Murphy BA, Ellerhorst J, et al. Randomized

multicenter phase II trial of subcutaneous recombinant human interleukin-12 versus interferon-

alpha 2a for patients with advanced renal cell carcinoma. J Interferon Cytokine Res 2001;21: 257-

63

65. Portielje JE, Kruit WH, Schuler M, Beck J, Lamers CH, Stoter G, et al. Phase I study of

subcutaneously administered recombinant human interleukin 12 in patients with advanced renal cell

cancer. Clin Cancer Res 1999;5: 3983-9

66. Geller AC, Swetter SM, Brooks K, Demierre MF and Yaroch AL. Screening, early detection,

and trends for melanoma: current status (2000-2006) and future directions. J Am Acad Dermatol

2007;57: 555-72; quiz 573-6

67. Little RF, Pluda JM, Wyvill KM, Rodriguez-Chavez IR, Tosato G, Catanzaro AT, et al. Activity

of subcutaneous interleukin-12 in AIDS-related Kaposi sarcoma. Blood 2006;107: 4650-7

68. Younes A, Pro B, Robertson MJ, Flinn IW, Romaguera JE, Hagemeister F, et al. Phase II

clinical trial of interleukin-12 in patients with relapsed and refractory non-Hodgkin's lymphoma and

Hodgkin's disease. Clin Cancer Res 2004;10: 5432-8

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

24

69. Salem ML, Gillanders WE, Kadima AN, El-Naggar S, Rubinstein MP, Demcheva M, et al.

Review: novel nonviral delivery approaches for interleukin-12 protein and gene systems: curbing

toxicity and enhancing adjuvant activity. J Interferon Cytokine Res 2006;26: 593-608

70. Halin C, Rondini S, Nilsson F, Berndt A, Kosmehl H, Zardi L, et al. Enhancement of the

antitumor activity of interleukin-12 by targeted delivery to neovasculature. Nat Biotechnol 2002;20:

264-9

71. Cemazar M, Jarm T and Sersa G. Cancer electrogene therapy with interleukin-12. Curr Gene

Ther 10: 300-11

72. Nagai H, Oniki S, Fujiwara S, Yoshimoto T and Nishigori C. Antimelanoma immunotherapy:

clinical and preclinical applications of IL-12 family members. Immunotherapy 2010;2: 697-709

73. Hao JS and Shan BE. Immune enhancement and anti-tumour activity of IL-23. Cancer Immunol

Immunother 2006;55: 1426-31

74. Lo CH, Lee SC, Wu PY, Pan WY, Su J, Cheng CW, et al. Antitumor and antimetastatic activity

of IL-23. J Immunol 2003;171: 600-7

75. Zhu S, Lee DA and Li S. IL-12 and IL-27 sequential gene therapy via intramuscular

electroporation delivery for eliminating distal aggressive tumors. J Immunol 2010; 184: 2348-54

76. Zolochevska O, Xia X, Williams J, Ramsay A, Li S and Figueiredo ML. Sonoporation delivery

of Interleukin 27 gene therapy efficiently reduces prostate tumor cell growth in vivo. Hum Gene

Ther 2011 Jul 29. [Epub ahead of print]

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

25

Legend to Figure 1

IL-27 activity against MM cells and the “growth paracrine loop”

IL-27 acts directly on MM cells that express complete IL-27R. IL-27 inhibits the angiogenic

potential of MM cells and down-regulates different angiogenic factors serve also as autocrine

and/or paracrine growth factors. Thus, IL-27 may interrupt or reduce the “growth loop” in which

cytokines and growth factors released by MM cells induce paracrine stimulation of stromal cells

that, in turn, secrete additional growth factors that stimulate new vessel formation and support MM

cell proliferation, progression and migration.

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

© 2011 American Association for Cancer Research

MMP2MMP9IL-1βTNF

CXCL9CXCL10

MMP1MMP2IL-1βTNF

VEGF-AVEGF-CVEGF-DTGF-β

Angiopoietins

VEGF-AVEGF-CVEGF-D

IL-6

Activatedbone marrowstromal cells

Invasiveness

Angiogenesis

IL-27

IL-27

IL-27

IL-27

MM cells

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724

Published OnlineFirst August 31, 2011.Clin Cancer Res   Nicola Giuliani and Irma Airoldi  malignant and normal plasma cellsNovel insights into the role of IL-27 and IL-23 in human

  Updated version

  10.1158/1078-0432.CCR-11-1724doi:

Access the most recent version of this article at:

  Manuscript

Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/early/2011/08/31/1078-0432.CCR-11-1724To request permission to re-use all or part of this article, use this link

Research. on December 21, 2018. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 31, 2011; DOI: 10.1158/1078-0432.CCR-11-1724