Targeting synovial neoangiogenesis in rheumatoid arthritis

Targeting synovial neoangiogenesis in rheumatoid arthritis

    Targeting synovial neoangiogenesis in rheumatoid arthritis Agathe Leblond, Yannick Allanore, J´erˆome Avouac PII: DOI: Reference: S1...

734KB Sizes 1 Downloads 113 Views

    Targeting synovial neoangiogenesis in rheumatoid arthritis Agathe Leblond, Yannick Allanore, J´erˆome Avouac PII: DOI: Reference:

S1568-9972(17)30100-3 doi:10.1016/j.autrev.2017.04.005 AUTREV 1997

To appear in:

Autoimmunity Reviews

Received date: Accepted date:

27 February 2017 3 March 2017

Please cite this article as: Leblond Agathe, Allanore Yannick, Avouac J´erˆome, Targeting synovial neoangiogenesis in rheumatoid arthritis, Autoimmunity Reviews (2017), doi:10.1016/j.autrev.2017.04.005

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Targeting synovial neoangiogenesis in Rheumatoid arthritis. Agathe Leblond1, Yannick Allanore (MD, PhD)1,2, Jérôme Avouac (MD, PhD)1,2 1

Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016 and CNRS UMR8104,

Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital

IP

2

T

Institut Cochin, Paris, France

Corresponding author: Dr. Jérôme Avouac Service de Rhumatologie A, Hôpital Cochin

NU

Université Paris Descartes 27 rue du Faubourg Saint-Jacques

MA

75014 Paris, France Telephone: + 33 1 58.41.25.63

SC R

Cochin, Paris, France

Fax: + 33 1 58.41.26.24

D

e-mail: [email protected]

TE

Total word count: 4389

CE P

Acknowledgement: “Arthritis foundation” and “Société Française de Rhumatologie”

AC

The authors have no competing interest to declare

1

ACCEPTED MANUSCRIPT Abstract: In Rheumatoid arthritis (RA), neoangiogenesis is an early and crucial event to promote the development of the hyperplasic proliferative pathologic synovium. Endothelial cells are critical for the formation of new blood vessels since they highly contribute to

T

angiogenesis and vasculogenesis. Current therapies in RA target the inflammatory

IP

consequences of autoimmune activation and despite major improvements these last years still refractory patients or incomplete responders may be seen raising the point of the need to

SC R

identify complementary additive and innovative therapies. This review resumes the mechanisms of synovial neoangiogenesis in RA, including recent insights on the implication of vasculogenesis, and the regulation of synovial neoangiogenesis by angiogenic and

NU

inflammatory mediators. In line with the recent development of vascular-targeted therapies used in cancer and beyond, we also discuss possible therapeutic implications in RA, in

MA

particular the combination of targeted immunotherapies with anti-angiogenic molecules.

D

Keywords: Rheumatoid arthritis, synovial angiogenesis, vasculogenesis, endothelial cells.

TE

Highlights

Neoangiogenesis is a key process in the development of the synovial pannus.

-

The formation of new synovial vessels depends on angiogenesis and vasculogenesis

-

A positive feedback loop exists between synovial inflammation and neoangiogenesis

-

Hypoxia and VEGF are the most potent proangiogenic growth factors in RA synovium

-

Modulation of angiogenesis in experimental arthritis has shown promising results

AC

CE P

-

2

ACCEPTED MANUSCRIPT Introduction: Rheumatoid arthritis (RA) is the most common cause of chronic inflammatory arthritis with a

T

prevalence ranging from 0.5% to 1% of the adult population worldwide (1, 2).

IP

The synovium is the primary site of the inflammatory process in RA. The synovium becomes

SC R

inflamed, with infiltration of blood-derived inflammatory cells at the interface between cartilage and bone. This invasive and destructive front (termed ‘pannus’) promotes the development of the erosions observed in RA. Progressive destruction of the articular cartilage,

NU

subchondral bone and periarticular soft tissues results in deformities that characterize long-

MA

standing RA. These deformities lead to functional deterioration and long term profound irreversible disability.

An important feature of RA is the role of vascular structures in these invasive and destructive

TE

D

processes. Indeed, there are increased number and density of synovial blood vessels in RA, which are required to supply the expansion of synovial tissue and to develop the hyperplasic

CE P

and invasive nature of RA synovium. Endothelial cells (ECs) lining the blood vessels also appear to be an active target for the action of cytokines, growth factors, permeability factors,

AC

and matrix-degrading enzymes. EC response to these factors both maintain and promotes RA. Thus, neoangiogenesis appears to be central to maintaining and promoting RA. It is also possible that a potential method of attenuating the development of the pannus is to interfere with its blood supply. This possibility is supported by several studies in animal models of arthritis which have suggested that blocking angiogenesis during the course of RA might actually be of therapeutic benefit. The aim of this review is to summarize the current understanding of synovial neoangiogenesis in RA and its regulation. We are also giving a focus on the application of angiogenesis inhibitors in animal models of arthritis, and on the potential for development of new vasculartargeted therapies for treatment of RA.

3

ACCEPTED MANUSCRIPT 1/ Increased vascular density in RA synovium and the contrast with tissue hypoxia The synovium is normally a physiologically relatively paucicellular structure with a delicate

T

intimal lining between macrophage-like and fibroblast-like synoviocytes. This lining region is

IP

one or two cells deep and is highly vascularized. Synovial blood flow provides oxygen and

SC R

nutrients to the synoviocytes and to the avascular articular cartilage.

RA is first characterized by a transitory pre-vascular highly inflammatory stage, followed by a prominent vascular stage with a strong increase in vessel growth.

NU

The pre-vascular stage is characterized by a marked hyperplasia of macrophage-like and

MA

fibroblast-like synoviocytes in the lining layer. In parallel, the sublining layer is infiltrated by CD4+ T cells, B cells and macrophages, leading to the formation of an invasive and destructive front, called the synovial pannus. This pannus acts like a local tumor that invades

TE

D

and damages cartilage and bone (2) (Figure 1). Then, the vascular stage rapidly arises and is usually set at the time of RA clinical diagnosis.

CE P

In this stage, capillary density is increased in the RA synovium, with a more deeply distribution as compared with normal tissue (Figure 2) (3, 4). Increased density of sublining

AC

blood vessels perpetuates synovitis by increasing the delivery of nutrients and oxygen to the proliferating pannus, and allows immune cells to emigrate from the blood into inflamed synovium where they highly produce a network of pro-inflammatory cytokines and chemokines (5). The increased number of blood vessels correlates with synovial hyperplesia, mononuclear cell infiltration and tender/swollen joint counts (6). Vascular proliferation is usually primarily detected in inflamed joints, whereas mononuclear cell infiltration and increased thickness of the synovial lining layer are observed both in inflamed and noninflamed joints (3). Despite increased vessel density related to active endothelial proliferation and increased EC survival, synovium in RA is chronically hypoxic, particulary in the lining layer (3) (Figure 1).

4

ACCEPTED MANUSCRIPT The presence of reduced oxygen levels in the RA synovium was demonstrated by direct measurements of the oxygen tension and more indirectly by an increase of hypoxic

T

metabolites in the synovium (7). This observation is not unexpected given the raise in

IP

synovial cell proliferation and the consequent increase in the distance between the

SC R

proliferating cells and the nearest blood vessels. This leads to a growing metabolic demand for oxygen and nutrients resulting in local hypoxia and relative hypoperfusion. Moreover the vasculature of RA synovium is compromised by movement and accumulation of synovial

NU

fluid, thus exacerbating hypoxia in an already ischemic environment. Such a combination of

MA

increased metabolic demand and hypoxia is a potent signal for new vessel formation.

2/ Source of new synovial blood vessels

TE

D

The recruitement of ECs is required to form new vascular structures. ECs can be recruited locally through angiogenesis, defined by the capillary sprouting of resident ECs. Circulating

CE P

bone marrow-derived endothelial progenitor cells (EPCs or hemangioblasts) are a second potential source of ECs.

AC

Angiogenesis

Angiogenesis is the growth of new blood vessels from existing ones and is an important aspect of new tissue development, growth and repair. The numerous proangiogenic signals that target ECs derive from cells primed in an abnormal environment, where the proliferation rate exceeds the supply of nutrients and oxygen (8). Angiogenic stimulation is triggered in RA synovium by the proinflammatory and hypoxic miceoenvironment, with production of a large array of growth factors, cytokines, and chemokines. These factors induce the sprouting of ECs from preexisting vessels, their proliferation and migration into inflamed sites, launching the vascular stage of the disease (Figure 1). This sequence is not specific to RA and is observed in other diseases with angiogenic compentent including cancer, diabetes, and other chronic

5

ACCEPTED MANUSCRIPT inflammatory conditions. However, specific cells from the synovium are also able to early drive angiogenenis together with ECs. Indeed, local synovial inflammation drives resident

T

stromal synovial cells to acquire a pro-angiogenic profile. Synovial fibroblast from RA

IP

synovium are sufficient under hypoxic conditions to induce angiogenesis, when used in a

SC R

matrigel plug system engrafted in immunodeficient mouse. (9, 10). In addition, these cells express growth factors (VEGF, b FGF, TGFβ), cytokines (IL-6, IL-8), chemokines (CXCL12), adhesion molecules (ICAM-1, VCAM-1) and matrix remodeling enzymes

NU

(MMP1, 2, 3 and 9) that regulate angiogenesis.

MA

Infiltrating macrophages are also a major source of pro-angiogenic molecules producing a broad range of mediators including growth factors, chemokines and matrix-remodeling enzymes (11).

TE

D

In RA, increased angiogenesis is associated with morphological alterations of new formed vessels. This fraction of neoangiogenic immature, dilated and leak vessels lacks -smooth

CE P

muscle actin (-SMA) positive mural cells. Chronic VEGF overexpression is implicated in this imbalance between EC proliferation and the lack of concomitant development of pericyte

AC

coverage. These small size vessels are preferentially located in sublining layer and are surrounded by inflamatory infiltrates (12) (Figure 1). Interestingly, disease progression and activity are related to the density of immature vessels, which is the sole vessel fraction to regress in response to anti-TNFα therapy (12). Vasculogenesis EPCs, first described by Asahara and al (13), are a population of bone marrow-derived cells characterized by the presence of surface markers such as CD34, VEGF receptor-2 (VEGFR-2 or kinase-insert domain receptor, KDR) and CD133, able to differentiate into mature endothelial cells and to participate in the formation of new blood vessels (14) (Figure 1).

6

ACCEPTED MANUSCRIPT Two studies have reported the presence of endothelial precursor cells in the synovial tissue of RA patients. In a first study (15), a population of cells expressing CD34 on their surface was

T

found in the synovial tissue of 18 RA patients. These cells were detected in close proximity to

IP

CD133+ cells, forming cell clusters in the area under the synovial membrane. CD34+

SC R

precursor cells produced high levels of the chemokine receptor CXCR4, and VEGFR-2 was expressed on CD34+ and CD133+ cells. In the second study (16), CD34+ cells, purified from the bone marrow of 13 patients with active RA and 9 control subjects, were cultured in the

NU

presence of stem cell factor and GM-CSF. Significantly more von Willebrand factor-positive

MA

cells (vWF+) and CD31+/vWF+ cells were generated from RA bone marrow-derived CD34+ cells for RA than control samples. The generation of vWF+ cells from bone marrow CD34+ cells was related to the microvessel densities in the synovial tissues. Thus, bone marrow

TE

D

CD34+ cells may contribute to synovial neovascularization by supplying endothelial precursor cells and may be important in the pathogenesis of RA.

CE P

In the peripheral blood of RA patients, contradictory results have been reported (17-21), which could be related to several methodological issues or the characteristics of the RA popu-

AC

lation enrolled. EPC depletion in the peripheral blood has been reported, which could result from their enhanced recruitment from peripheral blood to synovium. On the other hand, our group has reported increased circulating EPC levels in RA patients, with a correlation between EPC counts and RA disease activity (22). These results were obtained in a relatively large number of patients by using a well-characterized definition of late-outgrowth EPCs and fulfilling recent recommendations for the detection of EPCs. These results emphasize a key role for vasculogenesis in RA, and the association with disease activity supports the implication of vasculogenesis in the perpetuation of synovitis.

7

ACCEPTED MANUSCRIPT 3/ Regulation of synovial neoangiogenesis in RA Neovascularization in RA is dependent on the balance of proangiogenic and antiangiogenic

T

mediators, including growth factors, cytokines, chemokines, cell adhesion molecules and

IP

matrix metalloproteinases (23).

SC R

Hypoxia and Hypoxia inducible factor 1-alpha (HIF-1α)

RA synovium is characterised by chronic tissue hypoxia, which is partly related to synovial cell hyperplasia and the disorganization of vascular system within RA synovium. Hypoxia

NU

may be responsible for rendering RA synovial lining pro-angiogenic and pro-invasive, thus

MA

contributing to synovial and cartilage damage. Indeed, upregulation of metalloproteinases and RA fibroblast migration across collagen have been showed under hypoxic conditions (24). Furthermore, expression of angiogenic stimuli, including vascular endothelial growth factor

TE

D

(VEGF), and VEGF/placental growth factor heterodimer, was also increased. And crucially, it has been demonstrated that hypoxia increases the angiogenic drive of RA cells, as

CE P

demonstrated by enhanced blood vessel formation in an in vitro angiogenesis assay (24). A key regulator of angiogenesis is the transcription factor HIF (Hypoxia-Inducible Factor)

AC

which consists of an inducible oxygen-regulated subunit (HIF-1α) and a  subunit constitutively expressed in the nucleus (HIF-1β). Regulation of HIF-dependent gene expression requires -subunit accumulation in the cytoplasm and translocation into the nucleus, which enables it to dimerize with -subunits of HIF. HIF heterodimers are then recognized by co-activators and bind to the hypoxia response elements in the target gene to initiate transcription In RA, two, closely related HIF- isoforms (HIF-1, HIF-2) are expressed in human RA synovium and also in experimental rat arthritis, suggesting that synovial hypoxia leads to upregulation of HIF in the synovial joint, accumulation of VEGF and induction of synovial angiogenesis (25-27) (Figure 1). The number of HIF1-α positive cells correlated strongly

8

ACCEPTED MANUSCRIPT with the number of blood vessels in RA synovial tissue and with inflammatory EC infiltration, cell proliferation and the synovitis score. Some studies showed the beneficial

T

effect of compound targeting HIF-1α on the neovascularization in the RA. Benzophenones are

IP

potent anti-inflammatory compounds and are tested for anti-arthritic and anti-angiogenic

SC R

agents in animals models (28). A benzophenone analogue, 2-benzoyl-phenoxy acetamide BP1, was known to inhibit HIF-1α mediated VEGF expression and angiogenesis (29). It was shown that BP-1 inhibited HIF-1α nuclear accumulation in HUVECS and in the synovium

NU

tissue of adjuvant induced arthritis (AIA) rats (30). Moreover BP-1 inhibited the hypoxia

MA

induced increase of VEGF gene expression in HUVECS and also in AIA rats. The treatment with BP-1 in synovium section of AIA rats were characterized by a pronounced decrease in vascular density causing in a reduction of arthritis severity in AIA rats. More recently, a high-

TE

D

mobility group box protein 1 (HGMB1), a nonhistone nuclear protein and a cytokine mediator, have been shown to be implicated in the pathogenesis of RA (31). Stimulation with

CE P

HGMB1 increased both HIF-1α mRNA and protein levels in synovial fibroblasts obtained from RA patients. Moreover, HGMB1 induced HIF-1α transcription in RA fibroblasts occurs

AC

via NF-ΚB activation. The use of anti-HGMB1 neutralizing antibody prominently ameliorated the inflammatory features in collagen-induce arthritis (CIA) mice and also showed a significant reduction in the number of vessels in CIA mice in association with diminished HIF-1α and VEGF levels in the knee joints of CIA mice. VEGF signaling VEGF is the main signaling protein involved in angiogenesis ; it is detected in RA in synovial tissue and fluid, as well as in serum (32). During RA, VEGF165 is the mainly involved isoform (8) and it exerts its effects through tyrosine kinase receptors Flt-1 (fms-like tyrosine kinase receptor; also known as VEGF-R1) and Flk-1/KDR (fetal liver kinase receptor/kinaseinsert-domain-containing receptor; also known as VEGF-R2). The dual activities of VEGF as

9

ACCEPTED MANUSCRIPT an endothelial-cell mitogen and a modulator of changes in vascular permeability are of relevance in the pathogenesis of RA (Figure 1). Immunohistochemical and in situ

T

hybridization studies of synovial tissues have shown that VEGF is strongly expressed by

IP

subsynovial macrophages, fibroblast surrounding microvessels, vascular smooth muscle cells,

SC R

and synovial lining cells (33). The VEGF expression level in synovial fluid and tissues correlates with the clinical severity of human RA and with the degree of joint destruction (34, 35). Interestingly serum VEGF concentrations have been found higher in patients with newly

NU

diagnosed RA than in those with long-standing, treated disease. This observation may

MA

represent a response to drug treatment, supporting several observations demonstrating reduction in serum VEGF concentrations after therapeutic intervention. In fact patients with early RA responding to disease-modifying antirheumatic drugs showed significant reductions

TE

D

in serum VEGF concentrations compared to non esponders to the same treatment, who showed no significant change in serum VEGF (36). Furthermore, blockade of TNF- in RA

CE P

results in early marked reduction, but not normalization, of serum VEGF concentrations in a dose-dependent manner (37). The synthesis of VEGF by inflammatory and synovial cells is

AC

induced by numerous cytokines (such as TNF-, IL1-, TGF- and platelet derived growth factor, PDGF), by oxidative stress and by hypoxia (34, 35) (Figure 1). Increasing evidence suggests a complex interaction between VEGF and the angiopoietin (Ang)/Tie-2 system. It is demonstrated that Ang/Tie-2 is overexpressed in human RA synovium (38). Tie-2 signaling regulates arthritis-related angiogenesis in vivo and Ang-1 stabilize new blood vessels (Figure 1). Ang-1 is secreted by pericytes or synoviocytes and activation of Tie2 signalling through Akt pathway is responsible for endothelial cell proliferation and survival. Indeed, TNF-α upregulates Tie-2 in ECs through NFκB and increases the expression of Ang-1 in synoviocytes (39, 40). Gene therapy with the used of a soluble Tie-2 receptor inhibits neovascularization and arthritis development and protects

10

ACCEPTED MANUSCRIPT against bone destruction in a mouse model of CIA (41). Interestingly, the effect of a bispecific antibody targeting TNF- and Ang-2 in a TNF- transgenic mouse model of arthritis has

T

been recently described (42). The authors has showed a dose-dependent reduction in both

IP

clinical symptoms and histological scores with the use of this antibody. Moreover the

SC R

magnitude of results observed with this bispecific antibody were more important than those observed with adalimumab. Cytokines

NU

It is known that many of cytokines such as TNF-α, IL6, IL1-β or IL17 are important

MA

contributors in RA and may even work in part by regulating neovascularization in the arthritic joint.

TNF-α is a potent inducer of new blood vessel growth (43). Exposure of endothelial cells to

TE

D

TNF-α has been reported to induce the release of VEGF and FGF-2 (44) (Figure 1). Production by synovial-joint cells of angiogenic cytokines such as VEGF is at least in part

CE P

induced by TNF-α. Moreover, anti-TNF-α therapy was found to reduce the levels of circulating VEGF, the synovial blood flow assessed by power Doppler ultrasound (45), and

(46).

AC

the synovial vascularity assessed by immunostaining for CD31 and von Willebrand factor

IL-17 is found in RA synovial tissue and fluid, and the percentage of TH-17 cells is significantly higher in RA synovial fluid compared to RA or normal peripheral blood (47, 48). It was shown that the local expression of IL-17 increases vascularity in mouse ankle joints (49). Moreover, IL-17 markedly promotes blood vessels growth using matrigel plugs in C57BL/6 mice. In vitro, IL-17 induces endothelial Human microvascular endohtelial cells (HMVEC) migration and the used of neutralizing antibody to IL-17 suppressed HMVEC migration (49). IL-17 induced HMVEC chemotaxis and tube formation are mediated

11

ACCEPTED MANUSCRIPT primarily through IL-17 receptor (R)-C. Neutralization of either IL-17 in RA fluids or IL17R-C on HMVECS significantly reduces the induction of HMVEC migration.

T

Chemokines

IP

In RA, synoviocytes and inflammatory cells produces chemokines which plays a central role

SC R

in atttracting additional leucocytes. Among these chemokines, one has been mainly studied in RA. CXCL12 or stromal cell derived factor-1 (SDF-1) and it’s receptor, CXCR4, are present in joints of RA patients (50, 51). The injection of a CXCR4 antagonist (52), the bicyclam

NU

derivative AMD3100, in IFN-γR-deficient DBA/1 mice showed a reduction of incidence of

MA

arthritis, disease onset and clinical scores. Moreover, the histological examination demonstrated the protective effect of AMD3100 in IFN-γR-deficient DBA/1 mice. Indeed, infiltration of mono-and-polymorphonuclear cells, hyperplesia of the synovium and pannus

TE

D

formation were dramatically reduced in IFN-γR-deficient DBA/1 mice. More recently, CXCR7 was identified as an alternative receptor for CXCL12. CXCR7 is

CE P

expressed on RA synovial endothelial cells (53). The use of CXCR7 inhibitor, CCX733, inhibited CXCL12-induced angiogenesis, reduced clinical arthrtitis scores and prevented joint

AC

destruction in mice with CIA (53). Treatment with a humanized antibody against CXCL12, ch30D8, ameliorated arthritis in CIA mice (54). Indeed, this antibody reduced disease progression, clinical score and bone-erosive changes in CIA mice. The chemokine CXCL16 is also overexpressed in RA SF (55). The depletion of CXCL16 in CIA mice reduced clinical arthritis score, infiltration of inflammatory cells and bone destruction in the synovium of mice with CIA (56). The presence of CXCR6 in HMVECs and EPCs may be upregulated by IL-1β (57) and in vitro CXCL16 can induce HMVEC migration and tube formation. In vivo, intragraft with RA SF immunodepleted of CXCL16 in SCID mice showed a significant diminution in EPC recruitment.

12

ACCEPTED MANUSCRIPT 4/ Targeting angiogenesis: Therapeutic application in RA Effects of usual RA trearment on angiogenesis

T

Disease-modifying antirheumatic drugs (DMARDs) might affect synovial angiogenesis.

IP

Methotrexate (MTX) has been shown to inhibit vascular endothelial cell proliferation in vitro

SC R

(58). However, MTX failed to decrease VEGF levels in patients with RA and did not significantly reduce vessel growth in a murine in vivo matrigel model for angiogenesis.(59, 60).

NU

The use of TNF- inhibitors in RA results in early marked reduction, but not normalization,

MA

of serum VEGF concentrations in a dose-dependent manner (37, 59, 61). Moreover, antiTNF-α therapy was found to reduce the synovial blood flow assessed by power Doppler

D

ultrasound (45), and the synovial vascularity assessed by immunostaining for CD31 and von

TE

Willebrand factor (36).

Regarding other biologics, treatment with abatacept was associated in RA patients with

CE P

reduced serum levels of A disintegrin and metalloprotease 17 (ADAM17), which is recognized as an important player in exacerbation of inflammation related with increased

AC

activities of angiogenesis (62). A single study has observed reduced microvessel density in synovial tissues of RA patients treated with tocilizumab, as well as degeneration of lining layers (63).

Available preclinical evidence of anti-angiogenic treatments Direct Targeting of the VEGF pathway may be beneficial in treating RA. In the model of transgenic K/BxN mice, treatment with anti-VEGF-RI strongly attenuated the disease clinically and on histological analyses throughout the study period, while anti-VEGF only transiently delayed disease onset (64). Treatment with anti-VEGF-RII had no effect. Treatment with a VEGF-RI tyrosine kinase inhibitor almost abolished the disease. These results show that VEGF is a key factor in pannus development. This has been emphasised by

13

ACCEPTED MANUSCRIPT data showing that enhanced expression of PlGF and VEGF-R1 may contribute to rheumatoid inflammation by triggering production of proinflammatory cytokines (65).

T

Of the most interest, treatment with Avastin (Bevacizumab), a VEGF humanized monoclonal

IP

antibody, on collagen induced arthritis in rat model reduces significantly the arthritis index,

SC R

synovial pathological injury index and serum levels of VEGF and TNFα (66). Avastatin exhibits similar effects to Etanercept to relieve RA in rat model Moreover, angiostatin is known to be an endogenous inhibitor of angiogenesis. It has been described that angiostatin

NU

inhibits primary tumor growth and metastasis in vivo (67) and endothelial cells migration and

MA

induces endothelial cell-specific apoptosis in vitro (68). The use of a HIV vector containing the murine angiostatin expression unit significantly reduced synovial cell hyperplesia, pannus formation and progression of CIA (69).

TE

D

Other anti-angiogenic treatements have been used, which indirectly target the VEGF pathway. TNP-470, an angiogenesis inhibitor derived from fumagillin, has proven its efficacy in

CE P

inhibiting tumor growth and metastasis in various animals models (70-72). TNP-470 has been shown to inhibit angiogenesis and, in particular, VEGF synthesis (71, 72). The preventive

AC

treatment with TNP-470 (73), at a dosage of 60 mg/kg of body weight, delayed the onset of arthritis and its clinical intensity was rather mild. At a dosage of 90 mg/kg of TNP-470 the appearance of clinical signs was delayed for a longer period of time and disease was almost abolished. Moreover, the treatment with TNP-470 reduced cartilage and bone destruction, as well as circulating VEGF blood levels. Norisoboldine (NOR), an alkaloid coumpound, can significantly suppress synovial angiogenesis by selectively inhibiting endothelial cell migration (74). Indeed, the use of NOR in the adjuvant-induced arthritis rat model caused a significant reduction in the number of blood vessels and inhibited VEGF-induced endothelial cell migration via the CAMP-PKANF-ΚB/Notch 1 signaling.

14

ACCEPTED MANUSCRIPT Treatment with a recombinant human (rh) endostatin in the adjuvant-induced arthritis rat model led to decreased VEGF expression in both cartilage and synovial tissue, which was

T

accompanied by an attenuation of paw swelling and a reduction in new blood vessels

IP

formation (75). Moreover, administration of rh endostatin in rat adjuvant arthritis led to the

SC R

downregulation of VEGF, TNF-α and IL-1β production, wich was associated with decreased arthritis severity and bone destruction (76). In vitro, treatment with rh endostatin showed a reduction of osteoclast formation and bone resorbing function (76).

NU

Levels of resistin, a small cysteine-rich adipose-derived peptide hormone were overexpressed

MA

in the synovial fluid and tissue from patients with RA and mice with CIA. Resistin promoted the homing of EPCs into the synovium, thereby inducing angiogenesis (77). Resistin also

TE

and neoangiogenesis in vivo.

D

directly induced a marked VEGF expression in EPCs and its inhibition reduced EPC homing

CE P

Conclusion and research agenda

Neoangiogenesis is a dynamic and a key process in the development and the perpetuation of

AC

RA. New vessel formation depends on two main processes that are angiogenesis and vasculogenesis, which lead to the recruitment of resident and systemic ECs, respectively. A positive feedback loop exists between chronic inflammation and neoangiogenesis. Inflammatory state promotes enhanced synovial neovasvularization and new blood vessel formation facilitates chronic inflammation, leading to bone and cartilage damages. However, the question whether angiogenesis drives RA or whether the enhanced synovial proliferation promotes angiogenesis is still unsolved since angiogenesis, hypoxia and synovial hyperplasia are intricately linked and are regulated by common mediators. Multiple molecules contribute to the tight regulation of the angiogenic processes, especially hypoxia and VEGF, which is one of the most potent pro-angiogenic growth factors identified.

15

ACCEPTED MANUSCRIPT VEGF is implicated in orchestrating the generation of synovial neovessels, and persistent VEGF overexpression leads to the development of an altered vasculature characterized by

T

serious structural and functional defects.

IP

The modulation of many pro- and anti-angiogenic molecules in mouse models of

SC R

experimental arthritis has shown promising results. Consequently, targeting angiogenesis might be considered as a new alternative in the treatment of RA. This is supported by the fact that about 30% of patients have a primary or secondary lack of response, regardless the

NU

inflammatory cytokine or the immune cell targeted by the different available drugs. However,

MA

several questions need to be addressed prior positionning anti-angiogenic therapies in RA. The first question is the selection of the most accurate angiogenic pathway to target. Based on the data presented in this review, VEGF pathway seems the most appropriate given the

TE

D

prominent synovial and systemic overexpression of this growth factor in RA. Moreover, promising results both on efficacy and safety of anti-VEGF therapies in cancer and other

CE P

chronic diseases are available. Combination of anti-VEGF, bevacizumab and chemotherapy in many cancers (neuroblastoma, melanoma, malignant glioma, metastatic colerectal cancer,

AC

lung cancer and cervical cancer) showed a significant survival benefit in comparaison with chemotherapy alone (78-87). Moreover, the effect of bevacizumab combined with chemotherapy was better when bevacizumab was administered few days (3 days) before chemotherapy. In addition, inhibition of angiogenesis by anti-VEGF therapy (bevacizumab) has now been used in other diseases than e.g age-related macular degeneration (88) or diabetic retinopathy (89). Research agenda proposed to further address this question: further research is still required to definitely validate the choice of VEGF or identify another candidate molecules or pathways that could be targeted. Several proangiogenic or angiostatic factors have not been studied in RA (Table 1). Moreover, unbiaised genomic approaches on ECs isolated from

16

ACCEPTED MANUSCRIPT patients with RA are ongoing and could help to identify pertinent candidates to be further validated.

T

The second question is the selection of the most appropriate patients who could receive anti-

IP

angiogenic therapies. It makes sense to primarily focus on patients in the vascular phase of the

SC R

disease. High circulating levels of angiogenic biomarkers might be helpful for this selection, but the precise nature of the biomarkers should be assessed, as well as a routine dosage of these markers. Power Doppler ultrasound might also be useful to select patient with high

NU

synovial blood flow. Several data support to focus on patiens with newly diagnosed patients

MA

given the detection of high VEGF serum and synovial fluid levels, compared to patients with long lasting disease. Another important point to consider is the high cardiovascular risk of RA

TE

upon antiangiogenic therapies.

D

patients, given the demonstration of reduced collateral vessel formation following ischaemia

Research agenda proposed to further address this question: Further studies assessing the

CE P

link between angiogenic markers, clinical/biological signs of disease activity and synovial blood flow on doppler are required to better define the population that could benefit of anti-

AC

angiogenic therapies.

The third question is the administration of angiogenic therapies alone or in combination. The combination of an anti-angiogenic theray with a drug targeting the inflammation process seems more appropriate, by analogy with cancer therapy, in which of cytotoxic treatments are combined with anti-angiogenic molecules (90). Treatment targeting inflammation has already a variable anti-angiogenic effect, but which is in all cases incomplete. The addition of an antiangiogenic treatment could help to reach a complete effect on the pathological angiogenic process.

17

ACCEPTED MANUSCRIPT Research agenda proposed to further address this question: New preclinical studies testing this combination therapy is required before positionning anti-angiogenic drugs with

AC

CE P

TE

D

MA

NU

SC R

IP

T

human proof of concept studies.

18

ACCEPTED MANUSCRIPT References: 1.

Criscione LG, E.W. SC. Tumor necrosis factor-alpha for the treatment of rheumatic

Firestein GS. Starving the synovium: angiogenesis and inflammation in rheumatoid

IP

2.

T

diseases. . Curr Opin Rheumatol. 2002;14:204-11.

3.

SC R

arthritis. J Clin Invest. 1999;103:3-4.

FitzGerald O, Soden M, Yanni G, Robinson R, Bresnihan B. Morphometric analysis

of blood vessels in synovial membranes obtained from clinical affected and unaffected knee

Stevens CR, Blake DR, Merry P, Revell PA, Levick JR. A comparative study by

MA

4.

NU

joints of patients with rheumatoid arthritis. Ann Rheum Dis. 1991;50:792-6.

morphometry of the microvasculature in normal and rheumatoid synovium. Arthritis Rheum. 1991;34:1508-13.

Paleolog EM. Angiogenesis in rheumatoid arthritis. Arthritis Res. 2002;4.

6.

Rooney M, Condell D, Quinlan W, Daly L, Whelan A, Feighery C, et al. Analysis of

TE

D

5.

7.

CE P

the histologic variation of synovitis in rheumatoid arthritis. Arthritis Rheum. 1988;31:956-63. Bodamyali T, Stevens CR, Billingham ME, Ohta S, Blake DR. Influence of hypoxia in

8.

AC

inflammatory synovitis. Ann Rheum Dis. 1998;57:703-10. Marrelli A, Cipriani P, Liakouli V, Carubbi F, Perricone C, Perricone R, et al.

Angiogenesis in rheumatoid arthritis: a disease specific process or a common response to chronic inflammation? Autoimmun Rev. 2011;10:595-8. 9.

del Rey MJ, Izquierdo E, Caja S, Usategui A, Santiago B, Galindo M, et al. Human

inflammatory synovial fibroblasts induce enhanced myeloid cell recruitment and angiogenesis through a hypoxia-inducible transcription factor 1alpha/vascular endothelial growth factormediated pathway in immunodeficient mice. Arthritis Rheum. 2009;60:2926-34.

19

ACCEPTED MANUSCRIPT 10.

Del Rey MJ, Izquierdo E, Usategui A, Gonzalo E, Blanco FJ, Acquadro F, et al. The

transcriptional response of normal and rheumatoid arthritis synovial fibroblasts to hypoxia.

Maracle CX, Tas SW. Inhibitors of angiogenesis: ready for prime time? Best Pract Res

IP

11.

T

Arthritis Rheum. 2010;62:3584-94.

12.

SC R

Clin Rheumatol. 2014;28:637-49.

Izquierdo E, Canete JD, Celis R, Santiago B, Usategui A, Sanmarti R, et al. Immature

blood vessels in rheumatoid synovium are selectively depleted in response to anti-TNF

Asahara T, Murohara T, Sullivan A, Silver M, van der zee R, Li T, et al. Isolation of

MA

13.

NU

therapy. PLoS One. 2009;4:e8131.

putative progenitor endothelial cells for angiogenesis. SCIENCE. 1997;275:964-7. 14.

Avouac J, Wipff J, Goldman O, Ruiz B, Couraud PO, Chiocchia G, et al.

TE

D

Angiogenesis in systemic sclerosis: impaired expression of vascular endothelial growth factor receptor 1 in endothelial progenitor-derived cells under hypoxic conditions. Arthritis Rheum.

15.

CE P

2008;58:3550-61.

Ruger B, Giurea A, Wanivenhaus AH, Zehetgruber H, Hollemann D, Yanagida G, et

AC

al. Endothelial precursor cells in the synovial tissue of patients with rheumatoid arthritis and osteoarthritis. Arthritis Rheum. 2004;50:2157-66. 16.

Hirohata S, Yanagida T, Nampei A, Kunugiza Y, Hashimoto H, Tomita T, et al.

Enhanced generation of endothelial cells from CD34+ cells of the bone marrow in rheumatoid arthritis: possible role in synovial neovascularization. Arthritis Rheum. 2004;50:3888-96. 17.

Grisar J, Aletaha D, Steiner CW, Kapral T, Steiner S, Seidinger D, et al. Depletion of

endothelial progenitor cells in the peripheral blood of patients with rheumatoid arthritis. Circulation. 2005;111:204-11.

20

ACCEPTED MANUSCRIPT 18.

Herbrig K, Haensel S, Oelschlaegel U, Pistrosch F, Foerster S, Passauer J. Endothelial

dysfunction in patients with rheumatoid arthritis is associated with a reduced number and

Allanore Y, Batteux F, Avouac J, Assous N, Weill B, Kahan A. Levels of circulating

IP

19.

T

impaired function of endothelial progenitor cells. Ann Rheum Dis. 2006;65:157-63.

20.

SC R

endothelial progenitor cells in systemic sclerosis. Clin Exp Rheumatol 2007;25:60-6. Egan CG, Caporali F, Garcia-Gonzalez E, Galeazzi M, Sorrentino V. Endothelial

progenitor cells and colony-forming units in rheumatoid arthritis: association with clinical

Kuwana M, Okazaki Y, Yasuoka H, Kawakami Y, Ikeda Y. Defective vasculogenesis

MA

21.

NU

characteristics. Rheumatology (Oxford). 2008;47:1484-8.

in systemic sclerosis. The Lancet. 2004;364:603-10. 22.

Jodon de Villeroche V, Avouac J, Ponceau A, Ruiz B, Kahan A, Boileau C, et al.

TE

D

Enhanced late-outgrowth circulating endothelial progenitor cell levels in rheumatoid arthritis and correlation with disease activity. Arthritis Res Ther. 2010;12:R27. Tas SW, Maracle CX, Balogh E, Szekanecz Z. Targeting of proangiogenic signalling

CE P

23.

pathways in chronic inflammation. Nat Rev Rheumatol. 2016;12:111-22. Akhavani MA, Madden L, Buysschaert I, Sivakumar B, Kang N, Paleolog EM.

AC

24.

Hypoxia upregulates angiogenesis and synovial cell migration in rheumatoid arthritis. Arthritis Res Ther. 2009;11:R64. 25.

Gaber T, Dziurla R, Tripmacher R, Burmester GR, Buttgereit F. Hypoxia inducible

factor (HIF) in rheumatology: low O2! See what HIF can do! Ann Rheum Dis. 2005;64:97180. 26.

Peters CL, Morris CJ, Mapp PI, Blake DR, Lewis CE, Winrow VR. The transcription

factors hypoxia-inducible factor 1alpha and Ets-1 colocalize in the hypoxic synovium of inflamed joints in adjuvant-induced arthritis. Arthritis Rheum. 2004;50:291-6.

21

ACCEPTED MANUSCRIPT 27.

Brouwer E, Gouw ASH, Posthumus MD, van Leeuwen MA, Boerboom AL, Bijzet J,

et al. Hypoxia inducible factor-1-alpha is related to both angiogenesis and inflammation in

Khanum SA, Shashikanth S, Deepak AV. Synthesis and anti-inflammatory activity of

29.

SC R

benzophenone analogues. Bioorg Chem. 2004;32:211-22.

IP

28.

T

rheumatoid arthritis. Clin Exp Rheumatol. 2009;27:945-51.

Prabhakar BT, Khanum SA, Shashikanth S, Salimath BP. Antiangiogenic effect of 2-

benzoyl-phenoxy acetamide in EAT cell is mediated by HIF-1alpha and down regulation of

Shankar J, Thippegowda PB, Kanum SA. Inhibition of HIF-1alpha activity by BP-1

MA

30.

NU

VEGF of in-vivo. Invest New Drugs. 2006;24:471-8.

ameliorates adjuvant induced arthritis in rats. Biochem Biophys Res Commun. 2009;387:2238.

D

Park SY, Lee SW, Kim HY, Lee WS, Hong KW, Kim CD. HMGB1 induces

TE

31.

angiogenesis in rheumatoid arthritis via HIF-1alpha activation. Eur J Immunol. 2015;45:1216-

32.

CE P

27.

Ballara S, Taylor PC, Reusch P, Marmé D, Feldmann M, Maini RN, et al. Raised

AC

Serum Vascular Endothelial Growth Factor Levels Are Associated With Destructive Change in Inflammatory Arthritis. Arthritis Rheum. 2001;44:2055-64. 33.

Scola MP, Imagawa T, Boivin GP, Giannini EH, Glass DN, Hirsch R, et al.

Expression of angiogenic factors in juvenile rheumatoid arthritis: Correlation with revascularization of human synovium engrafted into SCID mice. Arthritis Rheum 2001;44:794-801. 34.

Berse B, Hunt RJ, Diegel P, Morganelli K-T, Yeo F, Fava BRA. Hypoxia augments

cytokine (transforming growth factor-beta (TGF-B) and IL-1)- induced vascular endothelial growth factor secretion by human synovial fibroblasts. Clin Exp Immunol. 1991;115:176-82.

22

ACCEPTED MANUSCRIPT 35.

Cho CS, Cho ML, Min SY, Kim WU, Min DJ, Lee SS, et al. CD40 Engagement on

Synovial Fibroblast Up-Regulates Production of Vascular Endothelial Growth Factor. J

Paleolog EM, Young S, Stark AC, McCloskey RV, Feldmann M, Maini RN.

IP

36.

T

Immunol. 2000;164:5055-61.

SC R

Modulation of angiogenic vascular endothelial growrh factor by tumor necrosis factor alpha and interleukin-1 in rheumatoid arthritis. Arthritis Rheum. 1998;41:1258-65. 37.

Macias I, Garcia-Perez S, Ruiz-Tudela M, Medina F, Chozas N, Giron-Gonzales J.

NU

Modification of pro-and anti inflammatory cytokines and vascular-related molecules by tumor

MA

necrosis factor alpha blockade in patients with rheumatoid arthritis. J Rheumatol. 2005;32:2102-8. 38.

Shahrara S, Volin MV, Connors MA, Haine GK, Koch AE. Differential expression of

TE

D

the angiogenic Tie receptor family in arthritic and normal synovial tissue. Arthritis Res. 2002;4:201-8.

DeBusk LM, Chen Y, Nishishita T, Chen J, Thomas JW, Lin PC. Tie2 receptor

CE P

39.

tyrosine kinase, a major mediator of tumor necrosis factor alpha-induced angiogenesis in

40.

AC

rheumatoid arthritis. Arthritis Rheum. 2003;48:2461-71. Scott BB, Zaratin PF, Gilmartin AG, Hansbury MJ, Colombo A, Belpasso C, et al.

TNF-alpha modulates angiopoietin-1 expression in rheumatoid synovial fibroblasts via the NF-kappa B signalling pathway. Biochem Biophys Res Commun. 2005;328:409-14. 41.

Chen Y, Donnelly E, Kobayashi H, Debusk LM, Lin PC. Gene therapy targeting the

Tie2 function ameliorates collagen-induced arthritis and protects against bone destruction. Arthritis Rheum. 2005;52:1585-94. 42.

Kanakaraj P, Puffer BA, Yao XT, Kankanala S, Boyd E, Shah RR, et al. Simultaneous

targeting of TNF and Ang2 with a novel bispecific antibody enhances efficacy in an in vivo model of arthritis. MAbs. 2012;4:600-13.

23

ACCEPTED MANUSCRIPT 43.

Leibovich SJ, Polverini PJ, Shepard HM, Wiseman DM, Shively V, Nuseir N.

Macrophage-induced angiogenesis is mediated by tumour necrosis factor-alpha. Nature.

Yoshida S, Ono M, Shono T, Izumi H, Ishibashi T, Suzuki H, et al. Involvement of

IP

44.

T

1987;329:630-2.

SC R

interleukin-8, vascular endothelial growth factor, and basic fibroblast factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol. 1997;17:4015-23. 45.

Fisher B, Donatien P, Filer A, Winlove C, McInnes I, Buckley CD, et al. Decrease in

NU

articular hypoxia and synovial blood flow at early time points following infliximab and

46.

MA

etanercept treatment in rheumatoid arthritis. Clin Exp Rheumatol. 2016;34:1072-1076. Taylor P, Patel S, Paleolog E, McCloskey RV, Feldmann M, Maini RN. Reduced

synovial vascularity following TNF alpha blockade in rheumatoid arthritis. Arthritis Rheum.

TE

47.

D

1998;41.

Stamp LK, James MJ, Cleland LG. Interleukin-17: the missing link between T-cell

CE P

accumulation and effector cell actions in rheumatoid arthritis ? Immunology and Cell Biology. 2004;82:1-9.

Shahrara S, Huang Q, Mardelin AMI, Pope RM. TH-17 cells in Rheumatoid arthritis.

AC

48.

Arthritis Res Ther. 2008;10. 49.

Pickens SR, Volin MV, Mandelin AM, 2nd, Kolls JK, Pope RM, Shahrara S. IL-17

contributes to angiogenesis in rheumatoid arthritis. J Immunol. 2010;184:3233-41. 50.

Buckley CD, Amft N, Bradfield PF, Pilling D, Ross E, Arenzana-Seisdedos F, et al.

Persistent Induction of the Chemokine Receptor CXCR4 by TGF- 1 on Synovial T Cells Contributes to Their Accumulation Within the Rheumatoid Synovium. J Immunol. 2000;165:3423-9.

24

ACCEPTED MANUSCRIPT 51.

Nanki T, Hayashida K, El-Gabalawy HS, Suson S, Shi K, Girschick HJ, et al. Stromal

Cell-Derived Factor-1-CXC Chemokine Receptor 4 Interactions Play a Central Role in CD4+

Matthys P, Hatse S, Vermeire K, Wuyts A, Bridger G, Henson GW, et al. AMD3100,

IP

52.

T

T Cell Accumulation in Rheumatoid Arthritis Synovium. Journal Immunol. 2000;165:6590-8.

SC R

a Potent and Specific Antagonist of the Stromal Cell-Derived Factor-1 Chemokine Receptor CXCR4, Inhibits Autoimmune Joint Inflammation in IFN- Receptor-Deficient Mice. Immunol. 2001;167:4686-92.

Watanabe K, Penfold ME, Matsuda A, Ohyanagi N, Kaneko K, Miyabe Y, et al.

NU

53.

J

54.

MA

Pathogenic role of CXCR7 in rheumatoid arthritis. Arthritis Rheum. 2010;62:3211-20. Zhong C, Wang J, Li B, Xiang H, Ultsch M, Coons M, et al. Development and

preclinical characterization of a humanized antibody targeting CXCL12. Clin Cancer Res.

TE

55.

D

2013;19:4433-45.

Ruth JH, Haas CS, Park CC, Amin MA, Martinez RJ, Haines GK, 3rd, et al. CXCL16-

CE P

mediated cell recruitment to rheumatoid arthritis synovial tissue and murine lymph nodes is dependent upon the MAPK pathway. Arthritis Rheum. 2006;54:765-78. Nanki T, Shimaoka T, Hayashida K, Taniguchi K, Yonehara S, Miyasaka N.

AC

56.

Pathogenic role of the CXCL16-CXCR6 pathway in rheumatoid arthritis. Arthritis Rheum. 2005;52:3004-14. 57.

Isozaki T, Arbab AS, Haas CS, Amin MA, Arendt MD, Koch AE, et al. Evidence that

CXCL16 is a potent mediator of angiogenesis and is involved in endothelial progenitor cell chemotaxis : studies in mice with K/BxN serum-induced arthritis. Arthritis Rheum. 2013;65:1736-46. 58.

Hirata S, Mastubara T, Saura R, Tateishi H, Hirohata K. Inhibition of in vitro vascular

endothelial cell proliferation and in vivo neovascularization by low-dose methotrexate. Arthritis Rheum. 1989;32:1065-73.

25

ACCEPTED MANUSCRIPT 59.

Nagashima M, Yoshino S, Aono H, Takai M, Sasano M. Inhibitory effects of anti-

rheumatic drugs on vascular endothelial growth factor in cultured rheumatoid synovial cells.

Fiehn C, Wunder A, Krienke S, Max R, Ho AD, Moehler T. Lack of evidence for

IP

60.

T

Clin Exp Immunol. 1999;116:360-5.

SC R

inhibition of angiogenesis as a central mechanism of the antiarthritic effect of methotrexate. Rheumatol Int. 2005;25:108-13. 61.

Ballara S, Taylor PC, Reusch P, Marme D, Feldmann M, Maini RN, et al. Raised

NU

serum vascular endothelial growth factor levels are associated with destructive change in

62.

MA

inflammatory arthritis. Arthritis Rheum. 2001;44:2055-64. Umemura M, Isozaki T, Ishii S, Seki S, Oguro N, Miura Y, et al. Reduction of Serum

ADAM17 Level Accompanied with Decreased Cytokines after Abatacept Therapy in Patients

63.

TE

D

with Rheumatoid Arthritis. Int J Biomed Sci. 2014;10:229-35. Hirohata S, Abe A, Murasawa A, Kanamono T, Tomita T, Yoshikawa H. Differential

CE P

effects of IL-6 blockade tocilizumab and TNF inhibitors on angiogenesis in synovial tissues from patients with rheumatoid arthritis. Mod Rheumatol. 2016:1-22. De Bandt M, Ben Mahdi MH, Ollivier V, Grossin M, Dupuis M, Gaudry M, et al.

AC

64.

Blockade of vascular endothelial growth factor receptor I (VEGF-RI), but not VEGF-RII, suppresses joint destruction in the K/BxN model of rheumatoid arthritis. J Immunol. 2003;171:4853-9. 65.

Yoo SA, Yoon HJ, Kim HS, Chae CB, De Falco S, Cho CS, et al. Role of placenta

growth factor and its receptor flt-1 in rheumatoid inflammation: a link between angiogenesis and inflammation. Arthritis Rheum. 2009;60:345-54. 66.

Wang Y, Da G, Li H, Zheng Y. Avastin exhibits therapeutic effects on collagen-

induced arthritis in rat model. Inflammation. 2013;36:1460-7.

26

ACCEPTED MANUSCRIPT 67.

O'Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, et al.

Angiostatin: A novel angiogenesis inhibitor that mediates the suppression of metastases by a

Holmgren L, O'Reilly MS, Folkman J. Dormancy of micrometastases: balanced

IP

68.

T

Lewis Lung Carcinoma. Cell 1994;79:315-28.

SC R

proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med. 1995;1:149-53. 69.

Kato K, Miyake K, Igarashi T, Yoshino S, Shimada T. Human immunodeficiency

NU

virus vector-mediated intra-articular expression of angiostatin inhibits progression of

70.

MA

collagen-induced arthritis in mice. Rheumatol Int. 2005;25:522-9. Antoine N, Greimers R, De Roanne C, Kusaka M, Heinen E, Simar LJ, et al. AGM-

1470, a potent angiogenesis inhibitor, prevents the entry of normal but not transformed

71.

TE

D

endothelial cells into the G1 phase of the cell cycle. Cancer Res. 1994;54:2073-6. Maniwa Y, Okada M, Ishii N, Kiyooka K. Vascular endothelial growth factor

CE P

increased by pulmonary surgery accelerates the growth of micrometastases in metastatic lung cancer. Chest. 1998;114:1668-75. Shishido T, Yashoshima T, Denno R, Mukaiya M, Sato N, Hirata K. Inhibition of liver

AC

72.

metastasis of human pancreatic carcinoma by angiogenesis inhibitor TNP-470 in combination with cysplatin. J Cancer Res. 1998;89:963-9. 73.

De Bandt M, Grossin M, Weber A-J, Chopin M, Elbim C, Pla M, et al. Suppression of

arthritis and protection from bone destruction by treatment with TNP-470/AGM-170 in a transgenic mouse model of rheumatoid arthritis. Arthritis Rheum. 2000;43:2056-63. 74.

Lu Q, Tong B, Luo Y, Sha L, Chou G, Wang Z, et al. Norisoboldine suppresses

VEGF-induced endothelial cell migration via the cAMP-PKA-NF-kappaB/Notch1 pathway. PLoS One. 2013;8:e81220.

27

ACCEPTED MANUSCRIPT 75.

Yue L, Shen YX, Feng LJ, Chen FH, Yao HW, Liu LH, et al. Blockage of the

formation of new blood vessels by recombinant human endostatin contributes to the

Chen N, Gao RF, Yuan FL, Zhao MD. Recombinant Human Endostatin Suppresses

IP

76.

T

regression of rat adjuvant arthritis. Eur J Pharmacol. 2007;567:166-70.

SC R

Mouse Osteoclast Formation by Inhibiting the NF-kappaB and MAPKs Signaling Pathways. Front Pharmacol. 2016;7:145. 77.

Su CM, Hsu CJ, Tsai CH, Huang CY, Wang SW, Tang CH. Resistin Promotes

NU

Angiogenesis in Endothelial Progenitor Cells Through Inhibition of MicroRNA206: Potential

78.

MA

Implications for Rheumatoid Arthritis. Stem Cells. 2015;33:2243-55. Dickson PV, Hamner JB, Sims TL, Fraga CH, Ng CY, Rajasekeran S, et al.

Bevacizumab-induced transient remodeling of the vasculature in neuroblastoma xenografts

TE

D

results in improved delivery and efficacy of systemically administered chemotherapy. Clin Cancer Res. 2007;13:3942-50.

Turley RS, Fontanella AN, Padussis JC, Toshimitsu H, Tokuhisa Y, Cho EH, et al.

CE P

79.

Bevacizumab-induced alterations in vascular permeability and drug delivery: a novel

AC

approach to augment regional chemotherapy for in-transit melanoma. Clin Cancer Res. 2012;18:3328-39. 80.

Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, Dowell JM, Reardon DA, Quinn JA,

et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13:1253-9. 81.

Giantonio BJ, Catalano PJ, Meropol NJ, O'Dwyer PJ, Mitchell EP, Alberts SR, et al.

Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J Clin Oncol. 2007;25:1539-44.

28

ACCEPTED MANUSCRIPT 82.

Saltz LB, Clarke S, Diaz-Rubio E, Scheithauer W, Figer A, Wong R, et al.

Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in

Reck M, von Pawel J, Zatloukal P, Ramlau R, Gorbounova V, Hirsh V, et al. Phase III

IP

83.

T

metastatic colorectal cancer: a randomized phase III study. J Clin Oncol. 2008;26:2013-9.

SC R

trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil. J Clin Oncol. 2009;27:1227-34. 84.

Tebbutt NC, Wilson K, Gebski VJ, Cummins MM, Zannino D, van Hazel GA, et al.

NU

Capecitabine, bevacizumab, and mitomycin in first-line treatment of metastatic colorectal

MA

cancer: results of the Australasian Gastrointestinal Trials Group Randomized Phase III MAX Study. J Clin Oncol. 2010;28(19):3191-8. 85.

Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al.

TE

D

Bevacizumab plus Irinotecan, Fluorouracil, and Leucovorin for metastatic colorectal cancer. New Engl J Med. 2004;350:2335-43.

Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al. Paclitaxel-

CE P

86.

Carboplatin alone or with Bevacizumab for non-small-cell lung cancer. N Engl J Med.

87.

AC

2006;355:2542-50.

Tewari KS, Sill MW, Long HJ, 3rd, Penson RT, Huang H, Ramondetta LM, et al.

Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med. 2014;370:734-43. 88.

Yonekawa Y, Kim IK. Clinical characteristics and current treatment of age-related

macular degeneration. Cold Spring Harb Perspect Med. 2014;5:a017178. 89.

Agarwal A, Afridi R, Hassan M, Sadiq MA, Sepah YJ, Do DV, et al. Novel Therapies

in Development for Diabetic Macular Edema. Curr Diab Rep. 2015;15:75. 90.

Yadav L, Puri N, Rastogi V, Satpute P, Sharma V. Tumour Angiogenesis and

Angiogenic Inhibitors: A Review. J Clin Diagn Res. 2015;9:XE01-XE5.

29

ACCEPTED MANUSCRIPT Table 1: Pro and anti-angiogenic molecules that are know or might contribute to synovial neoangiogenesis

Chemokines

TE CE P

Angiogenin ANG-1

IP

NU

IL-8 (CXCL8)

AC

Other mediators

Anti-angiogenic molecules

TGF-

TNF- IL-1

Cryptic proteins

Proangiogenic molecules

IL-12

MA

Cytokines

VEGF FGF-1, FGF-2, HGF TGF- PDGF PlGF G-CSF

D

Growth factors

Anti-angiogenic molecules

SC R

Proangiogenic molecules

Molecules suspected to contribute to synovial neoangiogenesis

T

Molecules known to contribute to synovial neoangiogenesis

Endostatin Angiostatin

Fibronectin fragment Kringle-5 Vasostatin Midkine, Pleiotrophin Ephrin-B2 Ephrin B4

Cartilage-derived inhibitor Interferon-inducible protein (IP-10) TIMPs Plasminogen activator inhibitor Platelet factor-4 Thrombospondin-1

VEGF: Vascular Endothelial Growth Factor, FGF: Fibroblast Growth Factor, HGF: Hepatocyte Growth Factor, TGF-β: Transforming Growth Factor beta, PDGF: Plateletderived Growth Factor, PIGF: Phosphatidylinositol-glycan biosynthesis class F protein, GCSF: Granulocyte colony-stimulating factor, TNF-α: Tumor Necrosis Factor α, IL-1β: Interleukin-1β, IL-12: Interleukin-12, IL-8: Interleukin-8, ANG-1: Angiopoietin-1, TIMPs: Tissue Inhibitor of metalloproteinase.

30

ACCEPTED MANUSCRIPT Figure legends

CE P

TE

D

MA

NU

SC R

IP

T

Figure 1: Mechanisms of synovial neovasculariation in rheumatoid arthritis A, Rheumatoid Arthritis (RA) is characterized by inflammatory polyarthritis involving particularly metacarpophalangeal and proximal interphalangeal joints. B, The synovium (in red) is the primary site of inflammation in RA leading to bone erosions and cartilage destruction. C-F: Neoangiogenesis is a crucial factor for the development of the synovial pannus. The lining layer becomes hypertrophic due to the marked hyperplasia of fibroblast-like synoviocytes. This cell hyperplasia promotes chronic tissue hypoxia with activation of the transcription factor HIF (Hypoxia inducible factor), leading to VEGF overexpression in the sublining layer of the synovium, the synovial fluid and the serum. The synovial sublining layer is characterized by increased vessel density, which perpetuates synovitis by allowing inflammatory cells to emigrate from the blood to inflamed synovium. These cells secrete matrix metalloproteinases and activate the RANK/RANKL pathway to induce bone and cartilage damages. Increased vessel density contrasts with hypoxic areas favored by synovial cell proliferation and formation of morphologically abnormal, immature and non-functional vessels secondary to chronic VEGF overexpression. VEGF is the main signaling protein involved in synovial angiogenesis. Hypoxia of the lining and sublining layers and inflammatory cytokines are the major source of VEGF production. VEGF induces the proliferation and the migration of endothelial cells (ECs) to form new blood vessels (D). The proinflammatory cytokine TNF- stimulates the production of VEGF through the NFKB pathway in ECs (E) and fibroblast-like synoviocytes (F). Angiopoietin-1 synthetized by pericytes stimulates vessel stabilization (D) through the activation of the tie2/AKT pathway (E). The overexpression of joint and systemic VEGF stimulates vasculogenesis with the recruitment of endothelial progenitor cells from the bone marrow (EPCs) (C). Circulating EPCs are recruited in the inflamed synovial to stimulate synovial neovascularization (D).

AC

Figure 2: Sections of synovial tissue stained for CD31 from a patient with osteoarthritis (OA) (A) and rheumatoid arthritis (RA). Increased vessel density is observed in the syovial tissue of the patient with RA compared to the patient with OA (black arrows). Perivascular inflammatory infiltrates are also visible in the patient with RA.

31

TE

D

MA

NU

SC R

IP

T

ACCEPTED MANUSCRIPT

AC

CE P

Figure 1

32

TE

D

MA

NU

SC R

IP

T

ACCEPTED MANUSCRIPT

AC

CE P

Figure 2

33