throbber
Journal of the Neurological Sciences 217 (2004) 125 – 130
`
`www.elsevier.com/locate/jns
`
`Review article
`
`Angiogenesis in multiple sclerosis: is it good, bad or an epiphenomenon?
`
`Shauna Kirk a, Joseph A. Frank b, Stephen Karlik a,c,d,*
`
`a Department of Pathology, University of Western Ontario, London, Ontario, Canada
`b Experimental Neuroimaging Section, Laboratory of Diagnostic Radiology Research, Clinical Center, National Institutes of Health, Bethesda, MD, USA
`c Department of Diagnostic Radiology, University of Western Ontario, London, Ontario, Canada
`d Department of Physiology, University of Western Ontario, London, Ontario, Canada
`
`Received 23 June 2003; received in revised form 20 October 2003; accepted 24 October 2003
`
`Abstract
`
`Characteristic pathological features of multiple sclerosis (MS) include inflammation, demyelination and axonal and oligodendrocyte loss.
`In addition, lesions can also have a significant vascular component. In this review, morphological, biochemical and radiological evidence is
`presented suggesting angiogenesis as a potential focus for investigation in MS. We hypothesize that angiogenesis plays a significant role in
`the MS lesion, perpetuating disease progression. Thus, treatment strategies that inhibit angiogenesis may decrease clinical and pathological
`signs of disease. Several approaches for testing this hypothesis are outlined.
`D 2003 Elsevier B.V. All rights reserved.
`
`Keywords: Angiogenesis; Multiple sclerosis; Experimental allergic encephalomyelitis; Neuroinflammation; VEGF; CNS pathology
`
`1. Introduction
`
`Multiple sclerosis (MS) is an inflammatory demyelinat-
`ing disease of the central nervous system (CNS) affecting
`approximately 350,000 individuals in North America alone
`[1]. Despite strong research efforts, the cause of MS remains
`elusive, the pathological mechanisms are not fully under-
`stood and the clinical course is highly variable, explaining
`why treatment options are still very limited. Without an
`established etiology or pathophysiology, the selection of
`treatments has concentrated almost exclusively on modify-
`ing the immune response. Research in MS concentrates on
`inflammatory changes, demyelination, axonal and oligoden-
`drocyte loss, and other characteristic pathological features of
`the MS plaque [2]; however, lesions also have a significant
`vascular component
`that warrants further investigation.
`Angiogenesis is the process leading to the development of
`new blood vessels from pre-existing ones [3,4]. An angio-
`genic response has been found to cause or contribute to an
`
`* Corresponding author. Diagnostic Imaging Room, 2MR21, London
`Health Sciences Center-University Campus, 339 Windermere Road,
`London, Ontario, Canada N6A 5A5. Tel.: +1-519-663-3648; fax: +1-519-
`663-3544.
`E-mail address: skarlik@uwo.ca (S. Karlik).
`
`0022-510X/$ - see front matter D 2003 Elsevier B.V. All rights reserved.
`doi:10.1016/j.jns.2003.10.016
`
`increasing number of pathological conditions [5,6]. Al-
`though angiogenesis and other forms of inflammation ex-
`hibit a positive feedback relationship [3],
`the role of
`angiogenesis (rather than altered existing vessels) in the
`pathogenesis of MS and its potential as a therapeutic target
`has not yet been explored.
`
`2. Hypothesis
`
`We propose that the association between MS lesions and
`neovascularization is not merely an epiphenomenon, but
`represents a pathological mechanism contributing directly to
`sustained disease. It is our hypothesis that angiogenesis
`represents a significant component of the MS lesion con-
`tributing to disease progression, thus inhibition of angio-
`genesis may suppress pathological changes, ameliorating
`clinical signs of disease.
`
`3. Current knowledge
`
`Angiogenesis is generally quiescent in adults with the
`exception of certain tightly controlled physiological situa-
`tions such as female reproductive functions and tissue
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2044
`
`

`

`126
`
`S. Kirk et al. / Journal of the Neurological Sciences 217 (2004) 125–130
`
`regeneration and repair [4]. An increase in vascularity acts to
`return the body to normal homeostasis by providing the
`necessary oxygen, nutrients and waste removal [7]. Whether
`or not angiogenesis occurs in a particular tissue depends on
`the balance between the relative amounts of pro- and anti-
`angiogenic factors [8]. Although the exact mechanism con-
`trolling angiogenesis in the CNS is not yet fully understood,
`many of the factors driving angiogenesis in other tissue are
`present during hypoxia/ischemia and tumor induced angio-
`genesis in the CNS [9 – 11]. Angiogenesis has been shown in
`an increasing number of pathological conditions including
`cancer, ischemic disease, diabetic retinopathy, blindness and
`inflammatory diseases (such as rheumatoid arthritis and
`psoriasis) [5,6]. While in some situations, such as cerebral
`and cardiovascular ischemic disease, angiogenesis is bene-
`ficial [9]; in many cases, such as rheumatoid arthritis,
`psoriasis and diabetic retinopathy, this increase in vascularity
`has been shown to be detrimental, leading to further damage
`[12,13].
`A relationship between blood vessels and MS lesions has
`been recognized for over 130 years. Edward Rindfleish [14]
`identified changes in both large and small vessels in the
`spinal cord and commented that all MS lesions were
`associated with abnormal blood vessels. MS lesions are
`typically centered on one or more veins in the white matter
`with particular association with the watershed veins of the
`periventricular white matter. Finger-like projections (Daw-
`son’s fingers) often extend from lesions and directly follow
`the course of the veins or venules [15 – 17]. Dawson’s
`fingers are thought to represent the invasion of the demy-
`
`elinating process into normal white matter [15]. Blood
`vessels are also associated with three of the four recently
`described MS lesion pathology classifications [18]. Pattern
`III, found primarily in acute disease, was not centered on
`veins and venules. This pattern has been shown to be
`associated with hypoxia-inducible factor 1 (HIF-1) [19],
`essential to hypoxia induced, vascular endothelial growth
`factor (VEGF) mediated angiogenesis [20,21], suggesting
`pattern III lesions may progress to a more typical perivas-
`cular pattern. As cells must be within 100 – 200 Am of a
`blood vessel to survive [22,23], MS lesions would require
`sufficient vascularization to permit delivery of additional
`hematogenous cells to maintain the inflammatory state.
`Several key components in the pathophysiology of MS
`(Fig. 1) are also associated with angiogenesis. Matrix metal-
`loproteases (MMP)-1, -2, -3 and -9, intercellular cell adhe-
`sion molecule (ICAM)-1, vascular cell adhesion molecule
`(VCAM)-1 and E-selectin facilitate in the entry of mono-
`nuclear cells through the blood – brain barrier (BBB) in MS
`[16,24] and are involved with the breakdown of the base-
`ment membrane in angiogenesis. This breakdown results in
`the further release of growth factors and angiogenic signal-
`ing molecules [25]. Inflammatory mediators,
`interferon
`(IFN)-g and tumor necrosis factor (TNF)-a/-h, are promi-
`nent in MS [16] and are angiogenic regulators [26]. In MS,
`elevated nitric oxide (NO) levels correlate well with clinical
`and magnetic resonance (MR) markers of disease progres-
`sion [27]. NO has been found to contribute both directly and
`indirectly to angiogenesis in inflammatory, vascular diseases
`and tumor expansion [28]. Endothelin-1 (ET-1), which
`
`Fig. 1. Potential role for angiogenesis in MS. (1) Initially immune cells bind to the BBB through a variety of cell adhesion molecules. (2) Cells infiltrate the
`perivascular space. (3) Cytokine, immune and antibody mediated attack on the myelin in the perivascular space. (4) Infiltration of immune cells into the
`parenchyma. (5) Release of inflammatory and hypoxic mediators including VEGF, HIF-1, NO, ET-1 from various cell types. (6) Angiogenic signals cause the
`initiation of the formation of new blood vessels into the lesion area.
`
`

`

`S. Kirk et al. / Journal of the Neurological Sciences 217 (2004) 125–130
`
`127
`
`stimulates neovascularization in concert with VEGF, corre-
`lates with tumor vascularity and malignancy, and induces
`MMP-2 production [29], has also been reported to be
`significantly elevated in MS patients [30]. VEGF, an un-
`equivocal angiogenic factor that signals the proliferation and
`migration of endothelial cells in angiogenesis [8,31], has
`recently been reported in both MS and its animal model
`experimental allergic encephalomyelitis (EAE) plaques
`[32]. In EAE induced rats an intracerebral infusion of VEGF
`resulted in an inflammatory response in the brain which was
`not found in vehicle-infused animals [32]. VEGF acts both
`directly and indirectly to promote angiogenesis, contributing
`directly to angiogenesis by acting as a specific mitogen and
`potent chemo-attractant
`to endothelial cells, as well as
`enhancing vascular permeability [33]. Indirectly, VEGF
`induces endothelial cells to release other factors, such as
`MMPs and ICAM-1, involved in the angiogenic process
`[33,34]. Several other angiogenic cytokines act at least in
`part by up-regulating VEGF expression [35].
`Further evidence for altered vascularity in MS is the
`existence of ‘‘ring enhancement’’ in contrast enhanced MRI
`at the periphery, but not at the center of chronic lesions [36].
`Early enhancing nodular lesions progressed to ring enhance-
`ment growing in size over time, supporting the belief that
`enhancement and possibly demyelination occurred from the
`center outwards [37]. The radiological evidence of perme-
`ability changes in MS has been assumed to represent the
`increased leakiness of existing vessels produced by inflam-
`matory modulators [38]. However, the evidence is equally
`indicative of new vessels and expansion of the regional blood
`volume, wherein the newly formed vessels are permeable to
`gadolinium-DTPA (Gd). Increased permeability in newly
`formed vessels could also be the cause of the prolonged T1
`and T2 seen in MS lesions or altered magnetization transfer
`ratio and diffusion weighted imaging observed in normal
`appearing white matter (NAWM).
`Rashid et al. [39] showed an increase in cerebral perfu-
`sion in relapsing-remitting (RR-) and secondary-progressive
`(SP-) MS patients compared to controls, which may provide
`evidence that there is an increase in the number of vessels in
`these inflammatory lesions. Recently, a serial MRI study of
`Gd enhancing MS lesions for local perfusion measurement
`found a significant increase in cerebral blood volume and
`cerebral blood flow, not only at the time of initial Gd
`enhancement compared to baseline, but also as early as
`three weeks prior to enhancement [40]. This indicates a
`significant role for the vasculature in NAWM very early on
`in lesion formation.
`In addition to an up-regulation of VEGF expression in
`both acute and chronic plaques, Proescholtd et al. [32] also
`demonstrated that blood vessels within lesions showed an
`irregular morphology consistent with angiogenesis. In acute
`MS lesions Ludwin et al. [41] reported an increase in the
`number and size of blood vessels with increases in endo-
`thelial cell number and mitotic count. Neovascularization
`was marked within, and at the edge of MS lesions, but also
`
`extended into the surrounding peri-plaque, often highlight-
`ing areas of acute inflammation or reactive gliosis. Ring
`enhancing lesions on MRI demonstrated increased vascu-
`larity and angiogenesis around the peripheral aspects of the
`lesion. VEGF and HIF were also found to be elevated in MS
`brains compared to normal tissues. In addition, chronic MS
`lesions appeared to have an increase in the number of
`vessels within a lesion although exact quantification was
`difficult due to scarring and shrinkage of the tissues [41].
`This is the first study in MS to demonstrate an increase in
`the number of blood vessels, however, the specific role of
`angiogenesis in disease progression and in treatment has not
`yet been evaluated.
`
`4. Angiogenesis as a therapeutic target in MS
`
`Many diseases are driven by persistent unregulated
`angiogenesis providing an opportunity for therapeutic in-
`tervention [12]. As new blood vessels are known to play a
`role both in the continued pathogenesis of chronic inflam-
`mation and in wound healing and repair processes, the issue
`of therapeutic intervention in angiogenesis remains contro-
`versial. An important consideration with anti-angiogenic
`therapy is the potential for impaired healing [42]. There is
`evidence to suggest that repair and regeneration can occur
`in the absence of angiogenesis. An anti-angiogenic therapy,
`CM101, was beneficial in the treatment of an experimental
`model of spinal cord injury. In this model, the inhibition of
`angiogenesis minimized both acute and chronic inflamma-
`tion in the CNS leading to the recovery of walking ability
`in the majority of treated mice [43]. In a study of cutaneous
`wound healing, CM101 was found to stimulate wound
`healing. The overall number of blood vessels was decreased
`and the inflammatory process was inhibited, reducing
`infiltration of activated leukocytes thus, preventing further
`tissue damage [44]. A study of dermal wound repair
`showed inhibition of angiogenesis though an alpha-v integ-
`rin blocking antibody, had no detrimental effects on wound
`repair [45]. When an avh3 antagonist was used in the
`treatment of an experimental model of rheumatoid arthritis
`it was shown to be effective not only in acute disease, but
`also resulted in the apoptosis of angiogenic blood vessels
`and effectively diminished clinical and pathological signs of
`disease [46].
`The positive feedback relationship between inflammation
`and angiogenesis can be advantageous under some circum-
`stances, however, as MS is an autoimmune disorder, per-
`sistent
`inflammation against a self antigen results in
`destruction of normal tissue. The potential advantages of
`inhibiting angiogenesis include halting the supply of
`nutrients and further inflammatory cells into the inflamed
`tissue and inhibiting the production of endothelial cell
`derived soluble factors [23]. The benefit of inhibiting
`angiogenesis has been seen in the animal models and early
`clinical studies of other autoimmune disorders such as
`
`

`

`128
`
`S. Kirk et al. / Journal of the Neurological Sciences 217 (2004) 125–130
`
`rheumatoid arthritis [46 – 50], diabetic retinopathy [51 – 54]
`and psoriasis [55]. Although angiogenesis is not likely the
`primary event
`in the pathogenesis of MS,
`its role in
`continued disease progression makes it an important target
`for therapeutic intervention.
`Most, if not all, of the treatments currently used in MS
`display anti-angiogenic properties in other diseases as
`shown by in vivo or in vitro studies. In MS, IFN-h reduces
`the proliferation of T cells, decreases the production of
`TNFa and antigen presentation and alters cytokine produc-
`tion to favor a Th2 pattern [56]. IFN-h also reduces the
`passage of immune cells across the BBB by affecting
`adhesion molecules, chemokines and proteases [16]. IFNs
`in general, are multi-functional cytokines shown to have
`anti-angiogenic properties shown both in vivo and in vitro
`and are used to treat tumor angiogenesis [57,58].
`Corticosteroids have been found beneficial in reducing
`the length of significant clinical relapses in some patients
`with RR-MS [59]. They have potent anti-inflammatory and
`immunosuppressive properties as well as having a stabiliz-
`ing effect on the BBB [60]. Certain corticosteroids have
`been found to be angiostatic, inhibiting the development of
`new blood vessels in tumors [61] and chronic inflammation
`[62,63].
`Immunosuppressive therapies (i.e. mitoxantrone, cyclo-
`phosphamide) are used in approximately 10% of MS
`patients to slow the progression of disease in RR-MS
`patients [64]. Immunosuppressive therapies are commonly
`used in the treatment of cancer and have been found to be
`highly anti-angiogenic [65,66].
`
`5. Hypothesis testing
`
`Clinical, pathological and imaging studies can be uti-
`lized to determine the role of angiogenesis and its potential
`as a therapeutic target
`in MS. EAE can be induced in
`several strains of rodents and non-human primates resulting
`in disease ranging from an acute attack with little or no
`demyelination to a chronic progressive disorder with ex-
`tensive demyelination and some remyelination [67,1].
`Studies of EAE using immunohistochemistry and histology
`can be used to evaluate the temporal relationship of
`angiogenic markers, blood vessel formation and clinical
`and pathological disease progression. Therapeutic studies
`in EAE can be utilized to test both anti-angiogenic and
`angiogenesis inducing agents. Inhibition of angiogenesis
`has the potential to also alleviate chronic inflammation [6].
`By suppressing the development of neovascularization,
`nutrients and new inflammatory cells will not reach the
`site of inflammation. This will prevent endothelial cell
`activation, proliferation and vascular remodeling, inhibiting
`the production of endothelial cell derived factors such as
`MMPs, VEGF receptors and cytokines [23]. Therapy that
`induces angiogenesis can help to determine if an increase
`in vascularity contributes to accelerated disease progres-
`
`sion. Conversely, some evidence suggests revascularization
`is necessary for promoting neural repair and regeneration,
`and prevention of the formation of new blood vessels could
`be detrimental [68]. EAE offers an opportunity to test our
`belief that
`tissue repair cannot be initiated until further
`damage has been halted, potentially requiring the resolu-
`tion of angiogenesis.
`Studies assessing the clinical signs and circulating an-
`giogenic markers (e.g. VEGF, bFGF) of both treated and
`untreated MS patients over time may be used as biomarkers
`to determine if current therapies exhibit an anti-angiogenic
`effect on MS patients and if so, if there is clinical benefit to
`decreased angiogenesis.
`Randomized clinical trials using anti-angiogenic agents
`either currently in use or in clinical trials for cancer and
`arthritis may provide additional therapeutic options for MS
`patients. Therapeutic strategies to inhibit angiogenesis target
`various aspects such as preventing matrix breakdown,
`inhibiting endothelial cell proliferation,
`interfering with
`specific activators of angiogenesis and inhibiting endothelial
`cell survival signaling. There are several ongoing clinical
`trials of anti-angiogenic therapies [69], however, to date, the
`studies have met varying degrees of success [70]. As
`knowledge of the biochemical and molecular mechanisms
`involved in angiogenesis increases, promising new thera-
`peutic strategies will offer more focused treatment leading to
`new therapeutic trials.
`Certain medications used for other indications have been
`shown to have anti-angiogenic properties. Minocycline
`hydrochloride used in the long-term treatment of acne
`patients has been effective in two different models of EAE
`[71,72] and is a potent inhibitor of angiogenesis in tumors
`[73] and in vitro [74,75]. A retrospective study of the
`disease course of individuals with MS concurrently receiv-
`ing long-term therapy with minocyline for acne may provide
`additional information into the benefit of inhibiting angio-
`genesis in MS.
`Clinical and EAE imaging studies could be utilized to
`determine the extent to which neovascularization contrib-
`utes to MRI findings and establish the clinical relevance of
`these findings.
`
`6. Summary and hypothesis
`
`MS is an inflammatory, demyelinating disorder of the
`CNS with vascular features. A firm link has been estab-
`lished between chronic inflammation and angiogenesis and
`evidence in MS is mounting to suggest a significant role
`for neovascularization in the progression of disease. Stud-
`ies of the role of angiogenesis over the course of MS and
`EAE have the potential to provide evidence concerning a
`key mechanism in the progression of disease. The evolu-
`tion and spread of lesions facilitated by angiogenesis may
`present a potential target for therapeutic intervention for
`MS.
`
`

`

`S. Kirk et al. / Journal of the Neurological Sciences 217 (2004) 125–130
`
`129
`
`Acknowledgements
`
`Supported by the MS Society of Canada.
`
`References
`
`[1] Steinman L. Assessment of animal models for MS and demyelinating
`disease in the design of rational therapy. Neuron 1999;24(3):511 – 4.
`[2] Ludwin SK. The neuropathology of multiple sclerosis. Neuroimaging
`Clin N Am 2000;10:625 – 48.
`[3] Folkman J, Brem H. Angiogenesis and inflammation. In: Gallin JI,
`Goldstein IM, Snyderman R, editors. Inflammation: Basic Princi-
`ples and Clinical Correlates. 2nd ed. New York: Raven Press; 1992.
`p. 821 – 39.
`[4] Forget MA, Desrosiers RR, Beliveau R. Physiological roles of matrix
`metalloproteinases: implications for tumor growth and metastasis.
`Can J Physiol Pharmacol 1999;77(7):465 – 80.
`[5] Polverini PJ. The pathophysiology of angiogenesis. Crit Rev Oral
`Biol Med 1995;6(3):230 – 47.
`[6] Winkler JD, Jackson JR. Chronic inflammation and angiogenesis. In:
`Rubanyi GM, editor. Angiogenesis in Health and Disease: Basic
`Mechanisms and Clinical Applications. New York: Marcel Dekker;
`2000. p. 407 – 16.
`[7] Kontos CD, Annex BH. Angiogenesis. Curr Atheroscler Rep 1999;
`1(2):165 – 71.
`[8] Lingen MW. Role of leukocytes and endothelial cells in the develop-
`ment of angiogenesis in inflammation and wound healing. Arch
`Pathol Lab Med 2001;125(1):67 – 71.
`[9] Plate KH. Mechanisms of angiogenesis in the brain. J Neuropathol
`Exp Neurol 1999;58(4):313 – 20.
`[10] Dunn IF, Heese O, Black PM. Growth factors in glioma angiogenesis:
`FGFs, PDGF, EGF and TGFs. J Neurooncol 2000;50(1 – 2):121 – 37.
`[11] Lopes MB. Angiogenesis in brain tumors. Microsc Res Tech 2003;
`1;60(2):225 – 30.
`[12] Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992;267(16):
`10931 – 4.
`[13] Creamer D, Sullivan D, Bicknell R, Barker J. Angiogenesis in psor-
`iasis. Angiogenesis 2002;5(4):231 – 6.
`[14] Rindfleisch E. Pathological Histology: An Introduction to the study of
`Pathological Anatomy. Translated from the German by WC Kloman
`and FT Miles. London: Trun bner & Co.; 1872. p. 652 – 8.
`[15] Adams C. A Colour Atlas of Multiple Sclerosis and Other Myelin
`Disorders. United Kingdom: Wolfe Medical Publications; 1989.
`p. 184 – 201.
`[16] Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG. Mul-
`tiple sclerosis. N Engl J Med 2000;343(13):938 – 52.
`[17] Tan IL, van Schijndel RA, Pouwels PJ, van Walderveen MA,
`Reichenbach JR, Manoliu RA, et al. MR venography of multiple
`sclerosis. AJNR Am J Neuroradiol 2000;21(6):1039 – 42.
`[18] Lucchinetti C, Bruck W, Parisi J, Scheithauer B, Rodriguez M,
`Lassmann H. Heterogeneity of multiple sclerosis lesions: implica-
`tions for the pathogenesis of demyelination. Ann Neurol 2000;47(6):
`707 – 17.
`[19] Lassmann H. Hypoxia-like tissue injury as a component of multiple
`sclerosis lesions. J Neurol Sci 2003;206(2):187 – 91.
`[20] Semenza GL. Surviving ischemia: adaptive responses mediated by
`hypoxia-inducible factor 1. J Clin Invest 2000;106(7):809 – 12.
`[21] Aboul-Enein F, Rauschka H, Kornek B, Stedelmann C, Stefferi A,
`Brucket W, et al. Preferential loss of Myelin-Associated Glycoprotein
`reflects hypoxia-like white matter damage in stroke and inflammatory
`brain diseases. J Neuropathol Exp Neurol 2003;62(1):25 – 33.
`[22] Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases.
`Nature 2000;407(6801):249 – 57.
`[23] Griffioen AW, Molema G. Angiogenesis: potentials for pharmaco-
`
`logic intervention in the treatment of cancer, cardiovascular diseases,
`and chronic I inflammation. Pharmacol Rev 2000;52(2):237 – 68.
`[24] Benveniste EN. Role of macrophages/microglia in multiple sclerosis
`and experimental allergic encephalomyelitis. J Mol Med 1997;75(3):
`165 – 73.
`[25] Dines KC, Powell HC. Mast cell interactions with the nervous system:
`relationship to mechanisms of disease. J Neuropathol Exp Neurol
`1997;56(6):627 – 40.
`[26] Van Meir EG. Cytokines and tumors of the central nervous system.
`Glia 1995;15(3):264 – 88.
`[27] Giovannoni G, Miller DH, Losseff NA, Sailer M, Lewellyn-Smith
`N, Thompson AJ, et al. Serum inflammatory markers and clinical/
`MRI markers of disease progression in multiple sclerosis. J Neurol
`2001;248:487 – 95.
`[28] Ziche M, Morbidelli L. Nitric oxide and angiogenesis. J Neurooncol
`2000;50(1 – 2):139 – 48.
`[29] Salani D, Taraboletti G, Rosano L, Di Castro V, Borsotti P, Giavazzi
`R, et al. Endothelin-1 induces an angiogenic phenotype in cultured
`endothelial cells and stimulates neovascularization in vivo. Am J
`Pathol 2000;157(5):1703 – 11.
`[30] Haufschild T, Shaw SG, Kesselring J, Flammer J. Increased endothe-
`lin-1 plasma levels in patients with multiple sclerosis. J Neurooph-
`thalmol 2001;21(1):37 – 8.
`[31] Jackson JR, Seed MP, Kircher CH, Willoughby DA, Winkler JD. The
`codependence of angiogenesis and chronic inflammation. FASEB J
`1997;11(6):457 – 65.
`[32] Proescholdt MA, Jacobson S, Tresser N, Oldfield EH, Merrill MJ.
`Vascular endothelial growth factor is expressed in multiple sclerosis
`plaques and can induce inflammatory lesions in experimental aller-
`gic encephalomyelitis rats. J Neuropathol Exp Neurol 2002;61(10):
`914 – 25.
`[33] Kolch W, Martiny-Baron A, Kieser A, Marme D. Regulation of the
`expression of the VEGF/VPS and its receptors: role in tumor angio-
`genesis. Breast Cancer Res Treat 1995;36(2):139 – 55.
`[34] Radisavljevic Z, Avraham H, Avraham S. Vascular endothelial
`growth factor up-regulates ICAM-1 expression via phosphatidylino-
`sitol 3 OH-kinase/AKT/nitric oxide pathway and modu migration of
`brain microvascular endothelial cells. J Biol Chem 2000;275(27):
`20770 – 4.
`[35] Dvorak HF. VPF/VEGF and the angiogenic response. Semin Perinatol
`2000;24(1):75 – 8.
`[36] Hiehle Jr JF, Grossman RI, Ramer KN, Gonzalez-Scarano F, Cohen
`JA. Magnetization transfer effects in MR-detected multiple sclerosis
`lesions: comparison with gadolinium-enhanced spin-echo images and
`nonenhanced T1-weighted images. AJNR Am J Neuroradiol 1995;
`16(1):69 – 77.
`[37] He J, Grossman RI, Ge Y, Mannon LJ. Enhancing patterns in multi-
`ple sclerosis: evolution and persistence. Am J Neuroradiol 2001;22:
`649 – 64.
`[38] Plumb J, McQuaid S, Mirakhur M, Kirk J. Abnormal endothelial tight
`junctions in active lesions and normal-appearing white matter in mul-
`tiple sclerosis. Brain Pathol 2002;12(2):154 – 69.
`[39] Rashid W, Parkes LM, Ingle GT, Chard DT, Symms M, Tofts PS, et al.
`Comparative investigation of cerebral perfusion in multiple sclerosis
`using a novel technique. Baltimore, USA: ECTRIMS; 2002.
`[40] Wuerfel J, Bellman-Strobl J, Brunecker P, Aktas O, McFarland H,
`Villringer A, et al. Changes in cerebral perfusion precede plaque
`formation in multiple sclerosis. Mult Scler 2003;9(S1):S19 – 20.
`[41] Ludwin SK, Henry JM, McFarland HF. Vascular proliferation and
`angiogenesis in MS: clinical and pathogenic implications. J Neuro-
`pathol Exp Neurol 2001;60:505.
`[42] Paleolog EM. Angiogenesis in rheumatoid arthritis. Arthritis Res
`2002;4(Suppl. 3):S81 – 90.
`[43] Wamil AW, Wamil BD, Hellerqvist CG. CM101-mediated recovery of
`walking ability in adult mice paralyzed by spinal cord injury. Proc
`Natl Acad Sci U S A 1998;95(22):13188 – 93.
`[44] Nanney LB, Wamil BD, Whitsitt J, Cardwell NL, Davidson JM, Yan
`
`

`

`130
`
`S. Kirk et al. / Journal of the Neurological Sciences 217 (2004) 125–130
`
`HP, et al. CM101 stimulates cutaneous wound healing through an
`anti-angiogenic mechanism. Angiogenesis 2001;4(1):61 – 70.
`[45] Jang YC, Arumugam S, Gibran NS, Isik FF. Role of alpha(v) integ-
`rins and angiogenesis during wound repair. Wound Repair Regen
`1999;7(5):375 – 80.
`[46] Storgard CM, Stupack DG, Joncyzk A, Goodman SL, Fox RI,
`Cheresh DA. Decreased angiogenesis and arthritic disease in rabbits
`treated with an alphavbeta3 antagonist. J Clin Invest 1999;103(1):
`47 – 54.
`[47] Peacock DJ, Banquerigo ML, Brahn E. Angiogenesis inhibition sup-
`presses collagen arthritis. J Exp Med 1992;175(4):1135 – 8.
`[48] Tilley BC, Alarcon GS, Heyse SP, Trentham DE, Neuner R, Kaplan
`DA, et al. Minocycline in rheumatoid arthritis. A 48-week, double-
`blind, placebo-controlled trial. MIRA Trial Group. Ann Intern Med
`1995;122(2):81 – 9.
`[49] Arsenault AL, Lhotak S, Hunter WL, Banquerigo ML, Brahn E.
`Taxol involution of collagen-induced arthritis: ultrastructural correla-
`tion with the inhibition of synovitis and neovascularization. Clin
`Immunol Immunopathol 1998;86(3):280 – 9.
`[50] Sone H, Kawakami Y, Sakauchi M, Nakamura Y, Takahashi A, Shi-
`mano H, et al. Neutralization of vascular endothelial growth factor
`prevents collagen-induced arthritis and ameliorates established dis-
`ease in mice. Biochem Biophys Res Commun 2001;281(2):562 – 8.
`[51] Riecke B, Chavakis E, Bretzel RG, Linn T, Preissner KT, Brownlee
`M, et al. Topical application of integrin antagonists inhibits prolifer-
`ative retinopathy. Horm Metab Res 2001;33(5):307 – 11.
`[52] Auricchio A, Behling KC, Maguire AM, O’Connor EM, Bennett J,
`Wilson JM, et al. Inhibition of retinal neovascularization by intra-
`ocular viral-mediated delivery of anti-angiogenic agents. Mol Ther
`2002;6(4):490 – 4.
`[53] Griggs J, Skepper JN, Smith GA, Brindle KM, Metcalfe JC, Hesketh
`R. Inhibition of proliferative retinopathy by the anti-vascular agent
`combretastatin-A4. Am J Pathol 2002;160(3):1097 – 103.
`[54] Penn JS, Rajaratnam VS, Collier RJ, Clark AF. The effect of an
`angiostatic steroid on neovascularization in a rat model of retinopathy
`of prematurity. Invest Ophthalmol Vis Sci 2001;42(1):283 – 90.
`[55] Sauder DN, Dekoven J, Champagne P, Croteau D, Dupont E. Neo-
`vastat (AE-941), an inhibitor of angiogenesis: randomized phase I/II
`clinical trial results in patients with plaque psoriasis. J Am Acad
`Dermatol 2002;47(4):535 – 41.
`[56] Rohowsky-Kochan C, Molinaro D, Cook SD. Cytokine secretion
`profile of myelin basic protein-specific T cells in multiple sclerosis.
`Mult Scler 2000;6(2):69 – 77.
`[57] Lindner DJ, Borden EC. Synergistic antitumor effects of a combina-
`tion of interferon and tamoxifen on estrogen receptor-positive and
`receptor-negative human tumor cell lines in vivo and in vitro. J In-
`terferon Cytokine Res 1997;17(11):681 – 93.
`[58] Fidler IJ. Angiogenesis and cancer metastasis. Cancer J 2000;
`6(Suppl. 2):S134 – 41.
`[59] Izikson L, Klein RS, Luster AD, Weiner HL. Targeting monocyte re-
`cruitment in CNS autoimmune disease. Clin Immunol 2002;103(2):
`125 – 31.
`
`[60] MacLean HJ, Freedman MS. Immunologic therapy for relapsing –
`remitting multiple sclerosis. Curr Neurol Neurosci Rep 2001;1(3):
`277 – 85.
`[61] Folkman J, Langer R, Linhardt RJ, Haudenschild C, Taylor S. Angio-
`genesis inhibition and tumor regression caused by heparin or a hep-
`arin fragment in the presence of cortisone. Science 1983;221(4612):
`719 – 25.
`[62] Colville-Nash PR, Alam CA, Appleton I, Brown JR, Seed MP,
`Willoughby DA. The pharmacological modulation of angiogenesis
`in chronic granulomatous inflammation. J Pharmacol Exp Ther
`1995;274(3):1463 – 72.
`[63] Nauck M, Karakiulakis G, Perruchoud AP, Papakonstantinou E, Roth
`M. Corticosteroids inhibit the expression of the vascular endothelial
`growth factor gene in human vascular smooth muscle cells. Eur J
`Pharmacol 1998;341(2 – 3):309 – 15.
`[64] Hommes OR, Weiner HL. Results of an international questionnaire on
`immunosuppressive treatment of multiple sclerosis. Mult Scler 2002;
`8(2):139 – 41.
`[65] Polverini PJ, Novak RF. Inhibition of angiogenesis by the antineo-
`plastic agents mitoxantrone and bisantrene. Biochem Biophys Res
`Commun 1986;140(3):901 – 7.
`[66] Billington DC. Angiogenesis and its inhibition: potential new thera-
`pies in oncology and non-neoplastic diseases. Drug Des Discov 1991;
`8(1):3 – 35.
`[67] Lublin FD. Experimental models of autoimmune demyelination. In:
`Cook SD, editor. Handbook of Multiple Sclerosis. 2nd ed. New York:
`Marcel Dekker; 1996. p. 119 – 43.
`[68] Zhang Z, Guth L. Experimental spinal cord injury: Wallerian degener-
`ation in the dorsal column is followed by revascularization, glial pro-
`liferation, and nerve regeneration. Exp Neurol 1997;147(1):159 – 71.
`[69] The Angiogenesis Foundation: online database [ http://www.cancer.
`gov/clinical_trials/doc.aspx?viewid = B0959CBB-3004-4160-A679-
`6DD204BEE68C].
`[70] Longo R, Sarmiento R, Fanelli M, Capaccetti B, Gattuso D, Gasparini
`G. Anti-angiogenic therapy: rationale, challenges and clinical studies.
`Angiogenesis 2002;5(4):237 – 56.
`[71] Brundula V, Rewcastle NB, Metz LM, Bernard CC, Yong VW. Tar-
`geting leukocyte MMPs and transmigration Minocycline as a poten-
`tial therapy for multiple sclerosis. Brain 2002;125(Pt. 6): 1297 – 308.
`[72] Popovic N, Schubart A, Goetz BD, Zhang SC, Linington C, Duncan
`ID. Inhibition of autoimmune encephalomyelitis by a tetracycline.
`Ann Neurol 2002;51(2):215 – 23.
`[73] Weingart JD, Sipos EP, Brem H. The role of minocycline in the
`treatment of intracranial 9

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket