Tyrosine kinases in inflammatory dermatologic disease.
Journal: 2011/September - Journal of the American Academy of Dermatology
ISSN: 1097-6787
Abstract:
Tyrosine kinases (TKs) are enzymes that catalyze the phosphorylation of tyrosine residues on protein substrates. They are key components of signaling pathways that drive an array of cellular responses including proliferation, differentiation, migration, and survival. Specific TKs have recently been identified as critical to the pathogenesis of several autoimmune and inflammatory diseases. Small-molecule inhibitors of TKs are emerging as a novel class of therapy that may provide benefit in certain patient subsets. In this review, we highlight TK signaling implicated in inflammatory dermatologic diseases, evaluate strategies aimed at inhibiting these aberrant signaling pathways, and discuss prospects for future drug development.
Relations:
Content
Citations
(2)
References
(153)
Diseases
(3)
Drugs
(1)
Chemicals
(3)
Organisms
(1)
Processes
(2)
Affiliates
(2)
Similar articles
Articles by the same authors
Discussion board
J Am Acad Dermatol 65(2): 389-403

Tyrosine kinases in inflammatory dermatologic disease

Introduction

In recent years, the number of studies on tyrosine kinases (TKs) in autoimmune or inflammatory disease has exponentially increased. The use of TK inhibitors have been proposed as potential therapies for rheumatoid arthritis, pulmonary arterial hypertension, Crohn's disease, and type-1 diabetes17. Here, we present experimental evidence that casts TKs as key players in the etiology and pathogenesis of inflammatory dermatologic diseases, and discuss the potential of TK inhibition for the treatment of these diseases.

Tyrosine kinases

Reversible phosphorylation is a post-translational mechanism that controls an array of fundamental cellular events. TKs contribute to phosphorylation-mediated regulation by catalyzing the transfer of a phosphate group from ATP or GTP to tyrosine residues on protein substrates. The human genome encodes 90 TKs, which can be divided into two main classes: receptor and non-receptor TKs8. Receptor TKs are transmembrane proteins composed of an extracellular ligand-binding domain, a transmembrane domain, and an intracellular domain containing the catalytic components. The 58 receptor TKs are grouped into 20 families that include the platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR), fibroblast growth factor receptor (FGFR), and RET9. In the absence of ligand binding, all receptor TKs (with the exception of the insulin receptor family) exist in the cell membrane in a monomeric and non-phosphorylated form. Ligand binding to the extracellular domain of receptor TKs induces oligomerization through conformational changes, in addition to stabilizing the active oligomeric form of receptor TKs10 (Figure 1). Oligomerization of receptor TKs classically leads to their activation via autophosphorylation of tyrosine residues in the activation loop of the intracellular domain, which leads to an increase in intrinsic catalytic activity and the formation of additional binding sites for substrate proteins11. Active receptor TKs can then catalyze the transfer of phosphate groups to tyrosine residues on substrate proteins, thus propagating the signal from the cell surface to the cell cytoplasm and nucleus.

An external file that holds a picture, illustration, etc.
Object name is nihms-200971-f0001.jpg

Receptor tyrosine kinase activation. A. In the absence of ligand binding, receptor tyrosine kinases (TKs) usually exist in the cell membrane in a monomeric and non-phosphorylated form. B. Ligand binding to the extracellular domain causes conformational changes that induce and stabilize oligomerization of the receptor TKs, leading to autophosphorylation of their cytoplasmic domains. The active kinase catalyzes the transfer of phosphate groups (P) to substrate molecules, thereby promoting signal transduction, including through MAPKs, Akt, and STATs, and downstream effector functions. The conformational changes involved in receptor TK activation may also promote signal transduction by releasing inhibitory constraints on substrate molecules. C. In the presence of a TK inhibitor (TKI), the cytosolic components of the receptor TK fail to effectively oligomerize and autophosphorylate, which prevents signal transduction and effector function.

Non-receptor, or cytosolic, TKs lack extracellular and transmembrane domains and are activated by signals that cause either their dissociation from inhibitors or the phosphorylation of tyrosine residues within the TK complex12. The 32 non-receptor TKs can be grouped into 10 families including Abl, Src, and janus kinase (JAK)9. As with receptor TKs, non-receptor TKs exist in an inactive conformation under basal conditions, and phosphorylation stabilizes the active kinase conformation, enabling the catalytic transfer of phosphate groups to the bound substrate protein (Figure 2). Excellent insights into TK structure-function relationships are reviewed elsewhere111315.

An external file that holds a picture, illustration, etc.
Object name is nihms-200971-f0002.jpg

Activation of tyrosine kinases. A. Tyrosine kinases (TKs) contain a substrate-binding domain, an ATP-binding site, and a catalytic site where the phosphate group (P) will be transferred. The substrate is the molecule to which the phosphate will be transferred. Under basal conditions, TKs exist in an inactive (“closed”) conformation (not shown), and phosphorylation of TKs stabilizes the active (“open”) kinase conformation that permits catalytic transfer of phosphate groups to substrate molecules. B. An activated TK transfers a phosphate group from ATP (or GTP) to a tyrosine residue on a substrate molecule. C. Phosphorylation of substrates by TKs is an important cellular mechanism by which a signal is propagated from one part of the cell to another and leads to various effector functions. D. TK inhibitors usually binds the kinase at the ATP-binding site, thus preventing ATP from binding and transferring a phosphate group to the substrate, and consequently preventing the active substrate from signaling to other parts of the cell. Selectivity of TK inhibitors is made possible by generating inhibitors that bind to specific chemical pockets adjacent to the ATP-binding site.

Systemic Sclerosis

Systemic sclerosis (SSc), a chronic connective tissue disease of unknown etiology, is characterized by extensive fibrosis of the skin and internal organs, production of autoantibodies, and widespread vasculopathy16. SSc remains an incurable disease with a median of 11 years of survival from the time of diagnosis17. Although the pathogenesis of SSc is unclear, current evidence implicates profibrotic pathways initiated by the cytokines PDGF and transforming growth factor β (TGFβ).

Members of the PDGFR family are PDGFRα, PDGFRβ, c-Fms (CSF1R), c-Kit, and c-Fms-like tyrosine kinase 3 (FLT3). PDGF ligands bind and activate PDGFRα and PDGFRβ, macrophage colony-stimulating factor (M-CSF; also known as CSF1) bind c-Fms, stem-cell factor (SCF) binds c-Kit, and FLT3 ligand binds FLT318. The PDGF isoforms, PDGF-A, PDGF-B, PDGF-C and PDGF-D, combine to form either homo- (PDGF-AA, -BB, -CC, -DD) or heterodimers (PDGF-AB only) that are biologically active19. PDGFs are produced by discrete cell populations including macrophages and endothelial cells. They bind to either PDGFRα or PDGFRβ found primarily on mesenchymal cells such as fibroblasts, smooth muscle cells, and glial cells, thereby driving local cellular responses including proliferation, migration, and survival.

Upregulation of the PDGF ligands PDGF-AA, PDGF-AB, and PDGF-BB are detected in bronchoalveolar lavage fluid, blood, endothelial cells, and infiltrating macrophages in SSc patients2022. Locally-captured platelets, endothelial cells, monocyte lineage cells and fibroblasts are all potential sources of PDGF ligands, however the exact cellular origin of these PDGF ligands in SSc patients remains to be elucidated. Expression of PDGFRs is high in skin biopsies and cultured fibroblasts from SSc patients; in contrast, PDGFR expression is low or absent in tissue or fibroblasts from healthy controls2326. In a recent study, autoantibodies against the inactive monomers of PDGFR were detected exclusively in SSc patients, and these PDGFR-specific autoantibodies could induce mitogen-activated protein kinase (MAPK) signaling and type I collagen gene expression in fibroblasts27. Although follow-up studies by other research groups have been unable to confirm the presence of stimulatory PDGFR-specific autoantibodies in SSc patients2830, the upregulation of both PDGFRs and their ligands in SSc patients suggests that PDGFR signaling is involved in SSc fibrosis. In theory, autoantibodies against receptor tyrosine kinases, such as PDGFR, have the potential to either act as agonists (stimulating signaling through the receptor) or antagonists (by preventing the binding of ligand).

In addition to PDGF, TGFβ is a critical cytokine in SSc, promoting fibroblast growth, differentiation, and extracellular matrix synthesis31. In animal models, constitutive TGFβ signaling in fibroblasts induces fibrotic features that are similar to those of human SSc32. TGFβ is secreted by several cell types including monocytes, macrophages, fibroblasts, and T cells, in a latent form; activation of TGFβ occurs via several distinct mechanisms including catalysis by thrombospondin, plasmin, and cell-surface integrins. Binding of TGFβ to oligomeric TGFβ receptor complexes, which are serine/threonine receptor kinases and not tyrosine kinases, activates both Smad-dependent and -independent signaling pathways33. TGFβ induces Smad2-and Smad3-independent activation of the TK c-Abl in fibroblasts3435; thus, TGFβ signaling could potentially be dampened with a TK inhibitor that targets c-Abl.

Interestingly, c-Abl and Arg, both members of the Abl TK family, are also activated, as part of a feedforward loop, by the PDGFR and EGFR signaling cascades3638. Furthermore, TGFβ stimulation upregulates PDGFRα expression in SSc fibroblasts25, suggesting that synergy exists between TGFβ and PDGFR signaling. Indeed, we showed that costimulation of SSc fibroblasts with PDGF and TGFβ, by activation of c-Abl, induced a greater and synergistic proliferative response than would be expected compared to stimulation with PDGF or TGFβ alone39. This evidence suggests that there are a multitude of opportunities to negate synergistic stimuli to fibroplasia, such as c-Abl and PDGFR, and further suggests that blocking multiple tyrosine kinase pathways may produce better efficacy outcomes compared to inhibition of a single tyrosine kinase.

In preclinical models of SSc, small-molecule inhibitors of PDGFR and c-Abl have shown efficacy in the treatment of skin fibrosis. For instance, the small-molecule inhibitor imatinib mesylate (imatinib), which inhibits PDGFR, Abl, c-Fms and c-Kit at clinically relevant concentrations, has been shown to provide significant efficacy in several animal models of fibrosis343540. Imatinib reduced dermal thickening in both the bleomycin-induced acute model of dermal fibrosis and the tight-skin-1 mouse late-fibrotic model of SSc4142. Nilotinib, which inhibits PDGFR, Abl, and c-Kit, and dasatinib, which inhibits PDGFR, Abl, c-Kit, and Src, have also been shown to decrease TGFβ- and PDGF-driven production of extracellular matrix proteins by dermal fibroblasts and to block bleomycin-induced dermal fibrosis43.

Case reports on the effects of imatinib on SSc-related disease are conflicting. Recent case reports describe patients with refractory SSc whose symptoms, particularly cutaneous sclerosis, improved following treatment with imatinib394445. Case reports have also described improvement in cardiopulmonary hemodynamics in patients with refractory SSc-associated PAH treated with imatinib46. However, in another case series that explored the addition of imatinib to cyclophosphamide therapy in patients with SSc-related interstitial lung disease, only one patient exhibited improvement in pulmonary status after 12 months of treatment with imatinib47. Similarly, there was no statistical improvement in skin score (6 month delta MRSS change from baseline = −2.0), inflammatory markers, or global assessments in four patients who completed 6 months of imatinib in patients with diffuse cutaneous SSc48. Five patients in this open-label study withdrew due to intolerable side effects48. In sharp contrast, a larger Phase II study which followed patients with diffuse cutaneous SSc for 12 months identified a benefit with imatinib therapy49. In this latter study, improvements did not start to emerge until the imatinib treatment period reached 6 months (3 month delta MRSS change from baseline = −0.4, 6 month delta MRSS change = −4.9), and improvements continued to increase through the 12 month endpoint (12 month delta MRSS change −7.3)49. This study also found mean forced vital capacity and diffusion capacity statistical improvements from 84% and 80% at baseline to 90% and 88% at 12 months, respectively49.

The reasons for the discrepancies in these case reports and open label trials may be multifold. The current data suggests that at least 6–9 months of consistent imatinib therapy are required until benefits are first observed. Perhaps at least part of the conflicting reports stems from insufficient time on imatinib therapy. Also, clearly imatinib tolerability is an issue in these reports. Our unpublished observations on the use of imatinib in SSc suggest that lower doses of imatinib (e.g., 100–200mg per day) is equally effective and better tolerated than the higher doses used in the aforementioned studies (400+mg per day). Unlike cancers, the fibroplasia seen in SSc is driven by wild-type kinases (e.g., PDGFR and c-Abl) in which low doses of TK inhibitors may be more appropriate. Lower doses of imatinib, with a corresponding reduced side effect profile, would increase the likelihood that SSc patients would safely reach the 6–9 month mark that appears to be required for benefits. Further, we found that a fibroblast gene-expression signature was markedly changed in SSc patients following treatment with imatinib39. We speculate that there are several patient population subsets that currently fall under the umbrella category of SSc, and that at least one subset may be responsive to imatinib therapy while other subsets may not be responsive. Molecular biomarkers could prove useful in guiding therapeutic decision-making, an approach that must be validated by rigorous clinical trials, which are currently underway.

Graft-versus-host-disease

Graft-versus-host-disease (GVHD) is a potentially lethal complication of allogeneic hematopoietic cell transplantation (HCT)50. HCT is of significant value for patients with leukemia or other diseases that stem from aberrations in hematopoiesis51. Acute GVHD generally occurs 20–40 days after HCT and is caused by activation of host antigen-presenting cells (APCs), activation of donor T cells in response to the activated host APCs, and finally secondary activation of host effector cells, resulting in further inflammation, local tissue injury, and organ attack52. Skin manifestations of acute GVHD include lichenoid macules and papules that may blister or ulcerate in severe cases52. Chronic GVHD is less well understood but recent insights suggest that B cells may be involved53. Skin lesions in chronic GVHD are classically divided into lichenoid and sclerodermoid categories, and recently an eczematoid form has been described5455.

Patients with extensive sclerodermoid chronic GVHD frequently have cutaneous manifestations similar to those of patients with SSc. Similar to SSc, GVHD may be associated with the development of autoantibodies against PDGFR. In a study on a small number of HCT patients, those patients with extensive chronic GVHD possessed stimulatory autoantibodies against PDGFR, whose levels correlated with fibrosis severity; in contrast, HCT patients without chronic GVHD and healthy individuals did not possess anti-PDGFR antibodies56. In a recent study on the use of low-dose imatinib in the treatment of chronic GVHD, imatinib was well tolerated overall, and the rate of response to treatment with imatinib was 86% after 8 months, which included a reduction in the sclerodermal disease as well as in chronic bronchiolitis and osteomyalgia57. Targeting B-cell activity and PDGFR autoantibodies holds promise for the treatment of chronic GVHD, and studies assessing the efficacy of small-molecule TK inhibitors of PDGFR, such as imatinib, in the treatment of chronic GVHD are ongoing.

Nephrogenic Systemic Fibrosis

Less than ten years have elapsed since the first published description of nephrogenic systemic fibrosis (NSF; formerly nephrogenic fibrosing dermopathy) in patients with severe renal disease58. The number of cases has been increasing, and over 200 patients are currently described in the International Center for Nephrogenic Fibrosing Dermopathy Research59. The major risk factor for developing NSF is the use of gadolinium-containing magnetic resonance contrast agents by patients with impaired renal function59. Cutaneous changes are the most prominent sign, occurring in all patients, and include skin tightening and thickening, induration, contractures, and hyperpigmentation60. Systemic involvement may also be observed and includes fibrosis of internal organs such as lungs, myocardium, and pericardium61. NSF skin biopsies show an increase in collagen levels and in proliferation of fibrocytes thought to be derived from the bone marrow6263.

Levels of the profibrotic cytokine TGFβ are increased in affected skin, fascia, and muscle of NSF patients64, suggesting that TGFβ plays a role in the development of fibrosis in NSF. Since TGFβ stimulates extracellular matrix production in fibroblasts by Smad-independent activation of the c-Abl TK, inhibitors of c-Abl could potentially provide therapeutic benefit in NSF. In addition, PDGFR may also be involved in the pathogenesis of NSF, as in other fibrotic disorders such as SSc and GVHD. Two recent case reports describe the use of imatinib—which inhibits both c-Abl and PDGFR— in the treatment of three patients with NSF6566. Administration of imatinib at 400–600 mg per day significantly improved NSF in two men (a 65-year-old and a 75-year-old) with stage-5 kidney disease65. In another study, imatinib at 400 mg per day or 300 mg twice a day provided significant therapeutic benefit in a 57-year-old woman with end-stage renal disease and biopsy-proven NSF66. In a recent open-label clinical trial, the therapeutic efficacy of imatinib was assessed in 6 patients with biopsy-proven NSF after 4 months of treatment67. The patients' cutaneous symptoms improved by 24% within 2 months of therapy, but worsened in 4 out of 6 patients after the discontinuation of imatinib treatment67. Additional studies using larger numbers of NSF patients to determine the clinical efficacy of imatinib or other c-Abl/PDGFR inhibitors are warranted.

Psoriasis

Psoriasis is a common inflammatory autoimmune disease with a strong genetic component. It is characterized by epidermal hyperproliferation and a diverse infiltration of leukocytes, which classically lead to the development of well-demarcated erythematous plaques with white scales6869. Evidence to date underscores the importance of angiogenic factors, keratinocytes, and T cells in the pathophysiology of psoriasis7072.

VEGF ligands are growth factors important in angiogenesis and signal through the TK VEGFR family members VEGFR1 (aka Flt-1/Flk-2), VEGFR2 (aka KDR/Flk-1), and VEGFR3 (aka Flt-4). Induction of inflammation with oxazolone in transgenic mice that heterozygously overexpress VEGF-A in the epidermis induces features that resemble those of human psoriasis, including epidermal hyperplasia, hyperkeratosis, and T-cell infiltration73. Homozygous overexpression of VEGF-A in transgenic mice resulted in the spontaneous development of a psoriasiform phenotype that mirrored human psoriasis74. Although VEGFRs are expressed by vascular endothelial cells and epidermal keratinocytes7576, aberrations in VEGFR signaling in psoriasis appear to occur primarily in epidermal keratinocytes. Compared to skin sections from normal patients, skin sections from psoriasis patients exhibit markedly higher expression of VEGF, VEGFR1, VEGFR2, and VEGFR-3, all of which localize to epidermal keratinocytes7779.

Compared to keratinocytes from normal skin, VEGFR1 and VEGFR2 on keratinocytes from both involved psoriatic and normal-appearing uninvolved psoriatic skin are significantly upregulated78. In contrast, tissue levels of VEGF ligands are significantly elevated only in involved psoriatic skin and are low in both uninvolved psoriatic skin and normal skin80. This raises the possibility that VEGFR1 and VEGFR2 signaling may be aberrantly upregulated in both involved and uninvolved psoriatic skin independent of the presence of VEGF ligands.

Thus VEGF/VEGFR signaling may be important in psoriasis, and blocking this pathway may have potential as a therapeutic strategy in psoriasis. NVP-BAW2881, a TK inhibitor that potently inhibits VEGFR1–3 at 1.0–4.3 nanomolar (nM) concentrations and inhibits PDGFRβ, c-Kit, and RET at 45–72 nM concentrations reduced psoriasis-like symptoms in heterozygous VEGF-A transgenic mice challenged with oxazolone72. Interestingly, topical administration of NVP-BAW2881 was almost as efficacious as oral administration. Since topical administration will reduce systemic toxicities associated with TK inhibition, these studies may represent an important step toward the use of topical TK inhibitors for the treatment of skin lesions.

Like VEGF/VEGFR signaling, EGF/EGFR signaling has been implicated in keratinocyte dysregulation in psoriasis. The EGFR family is composed of four members: EGFR (also known as ErbB1 or Her1), ErbB2 (Her2), ErbB3 (Her3), and ErbB4 (Her4). EGFR, ErbB2, and ErbB3 are expressed in epithelial tissues and promote cell survival, migration, and proliferation81. The EGF ligands are produced largely by keratinocytes and include TGFα, amphiregulin, EGF, and epiregulin82. Levels of TGFα8384, amphiregulin8586, heparin-binding EGF87, and epiregulin88 are increased in psoriatic keratinocytes and psoriatic skin biopsies. Furthermore, transgenic mice that overexpress EGF ligands in basal keratinocytes develop psoriasis-like skin lesions89. EGFR expression is also significantly increased in psoriatic skin, and even in normal-appearing skin adjacent to psoriatic lesions, compared to normal skin8384. Psoriatic skin biopsies were shown to maintain histological features of psoriasis while in tissue culture, a phenotype that partially reverted to normal in the presence of neutralizing EGFR antibodies90 or the EGFR inhibitor PD16954091.

T cells play a major role in the autoimmune response in psoriasis, and both Th1 and Th17 cells have been proposed as the dominant T-cell type that secretes cytokines and interacts with the local cell population, thereby promoting the formation of a psoriatic plaque697192. The cytoplasmic TK JAK3 plays critical roles in T-cell development, activation, and proliferation93, Whereas other members of the JAK family are involved in a broad array of cytokine signaling, JAK3 is expressed predominantly by lymphocytes. The JAK1/3-specific TK inhibitor R333 was recently shown to provide therapeutic efficacy in the CD18-deficient PL/J mouse model of psoriasis94. CP-690,550, a more specific JAK3 inhibitor95, also showed significant benefit in a psoriasis phase 1 clinical trial96. Thus, inhibiting T-cell signaling by targeting JAK3 may have potential as an alternative treatment strategy for psoriasis.

Case reports have documented both beneficial and detrimental effects of currently available oral small-molecule TK inhibitors in human psoriasis. Psoriasis improved in a 64-year-old man receiving sunitinib for the treatment of metastatic renal cell carcinoma (RCC)97. Sunitinib inhibits several TKs including VEGFR, PDGFR, c-Kit, c-Fms, FLT3, and RET; this broad inhibitory spectrum makes it difficult to determine whether or not angiogenesis was the primary process targeted by sunitinib. Case reports on the use of imatinib for the treatment of psoriasis are conflicting. An early case report detailed the prompt disappearance of long-standing psoriasis in a 64-year-old man whose metastatic gastrointestinal stromal tumor (GIST) was treated with imatinib98; the psoriasis partially reappeared when the dose of imatinib was lowered98. In contrast, more recent cases have been described in which treatment of chronic myelogenous leukemia or GIST with imatinib exacerbated underlying psoriasis99101.

EGFR inhibitors have proven to be an effective therapy for several types of cancers. The majority of cancer patients treated with EGFR inhibitors exhibit skin toxicities due to the effects of EGFR blockade in normal keratinocytes102. However, in the correct clinical setting, EGFR inhibition could potentially provide benefit against skin pathology. A recent report described the effects of lapatinib, an EGFR/ErbB2 TK inhibitor, used to treat a 53-year-old man with renal cell carcinoma who simultaneously had long-standing severe psoriasis103. Although one month of lapatinib treatment completely resolved his psoriasis, the patient developed skin toxicities—including acneiform skin rash and facial seborrheic dermatitis—that typically occur secondary to EGFR inhibitor treatment103. Cetuximab, a monoclonal antibody against EGFR, reduced psoriasis in a patient treated for metastatic colorectal cancer104. In sharp contrast, one month of treatment with gefitinib, an EGFR inhibitor that exhibits minimal activity against ErbB2105, exacerbated psoriasis in a 69-year-old man with non-small cell lung cancer106. Clearly, furtherclinical data are needed to determine whether inhibition EGFR family members may provide benefit in psoriatic patients.

Pemphigous vulgaris

Pemphigus vulgaris (PV) is a potentially fatal autoimmune blistering disease caused by antibodies against desmoglein (Dsg) 1 and Dsg 3—transmembrane adhesion proteins located on the cell surface of keratinocytes107—as well as non-Dsg antigenic targets108. More than 25 years have elapsed since the landmark study by Diaz and colleagues demonstrating that passive transfer of the IgG fraction of PV sera (PV-IgG) induces pemphigus in mice109. Although the molecular mechanisms by which PV-IgG leads to loss of adhesion between adjacent endothelial cells (acantholysis) and to skin blistering remain uncertain, recent studies indicate that keratinocyte TK signaling pathways are critical in the pathogenesis of PV-IgG-mediated acantholysis.

In a study on the role of EGFR in PV-IgG-mediated acantholysis, PV-IgG was shown to induce EGFR phosphorylation in cultured keratinocytes and the consequent activation of downstream signaling molecules including the MAPK extracellular signal-regulated kinase (ERK)110. Moreover, AG1478, a specific EGFR inhibitor, reversed PV-IgG-mediated apoptosis in cultured keratinocytes110. The mechanisms by which PV-IgG induced EGFR activation were not elucidated. In a separate study, injection of human PV-IgG was shown to induce the phosphorylation of EGFR, ErbB2, and ErbB3 in mice111. Clinical and molecular findings could be reversed with erlotinib, which inhibits EGFR, ErbB2, and ErbB3. Furthermore, TGFα, EGF, and betacellulin were upregulated in PV epidermal lesions following injection of human PV-IgG compared to injection of normal human IgG111. Whether the upregulation of EGFR family ligands is a direct effect of PV-IgG binding to keratinocytes, or an indirect effect resulting from the induction of acantholysis, remains unclear. However, PV-IgG may activate keratinocytes to produce these EGFR family ligands, which in turn drive the EGFR/ErbB2/ErbB3 signaling cascade.

Activation of Src has been observed in the epidermis following injection of PV-IgG in mice, and in vivo administration of the Src inhibitor PP1 successfully prevented acantholysis111. Recent analyses suggest that PV-IgG induction of Src signaling occurs upstream of EGFR activation. PV-IgG stimulation of keratinocytes elicited peak phosphorylation activities of Src, EGFR, and p38 MAPK at 30 min, 60 min, and 240 min, respectively112. Thus, Src signaling preceded EGFR activation. However, Src inhibition with PP2 following PV-IgG stimulation only partially abrogated EGFR and p38 MAPK phosphorylation112. Together, these studies suggest that either (i) the concentration of PP2 did not completely inhibit Src activation, or (ii) PP1 may inhibit kinases other than Src (as has been previously suggested113) and that non-Src pathways also trigger EGFR activation.

Whether or not it is regulated primarily by Src, EGFR appears to play a critical role in acantholysis, making it an attractive therapeutic target in PV. Genistein, a pan-TK inhibitor114, has been shown to block PV-IgG-induced acantholysis in vivo115116. TK inhibition with genistein has also been shown to block Dsg3 internalization in PV-IgG-stimulated keratinocytes117. Further insight into the functional significance of Dsg3 internalization will be important in attempts to identify therapeutic targets in keratinocyte signaling pathways. In addition, blocking p38, which is activated downstream of EGFR, with specific inhibitors prevented acantholysis in the IgG passive-transfer model of PV118. Despite the availability of FDA-approved inhibitors—such as gefitinib, erlotinib, cetuximab, lapatinib, and panitumumab—that target members of the EGFR family, there have been no case reports describing the use of EGFR inhibitors in human PV. Therapies that target key signaling molecules in PV acantholysis, such as EGFR, may represent a novel strategy to treat this life-threatening autoimmune disease. On the basis of the availability and increasingly extensive use of EGFR inhibitors, we expect that there will soon be case reports that document the use of EGFR inhibitors in patients with a primary oncologic disease who concomitantly have PV.

Bullous Pemphigoid

Bullous pemphigoid, a common autoimmune blistering disease119, exhibits increased VEGFR1 and VEGFR2 expression in affected and surrounding skin lesions compared to normal skin, and increased VEGF levels in serum and in blister fluid120121. Further evidence implicating TKs in the pathogenesis of bullous pemphigoid stems from a case report that describes a patient with severe hypereosinophilic syndrome and bullous pemphigoid whose skin lesions and eosinophilia both dramatically regressed when treated with imatinib at 400–600 mg per day122. Additional research is necessary to define the role of TK inhibitors in the treatment of bullous pemphigoid.

Dermatomyositis and Systemic Lupus Erythematosus

Dermatomyositis (DM) is a chronic inflammatory disorder that exhibits progressive proximal symmetric muscle weakness and cutaneous disease. It is an autoimmune disorder involving deposition of autoantibodies in the microvasculature, which leads to complement activation and subsequently to capillary necrosis and a mixed leukocytic infiltration123. Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibody formation, chronic inflammation, and immune complex deposition leading to irreversible end-organ failure. Cutaneous manifestations of SLE occur in the majority of patients and the three forms are acute cutaneous lupus, subacute cutaneous lupus, and chronic cutaneous (discoid) lupus124. Recent studies have implicated the type I interferons (IFNα and IFNβ) in the pathogeneses of DM and SLE. Tyk2, a non-receptor TK, is essential for type I IFN signaling and may represent a novel target for DM and SLE therapy.Large-scale analysis of transcriptional profiles in muscle biopsies from patients with DM, other myopathies, or neuromuscular disorders identified a gene-expression profile characteristic of DM, in which 12 of the 14 most highly upregulated genes were type I IFN-inducible genes125. MxA, a type I IFN-inducible protein that is not induced by other cytokines including IFNγ126, was highly expressed in DM muscle fibers and capillaries125. Type I IFN-inducible genes were also found to be the most highly upregulated genes in peripheral blood mononuclear cells from patients with DM, in which the level of type I IFN-inducible transcripts correlated with DM disease activity127. Levels of MxA protein are also significantly increased in DM skin lesions compared to healthy skin128 and our unpublished data based on transcriptional profiling demonstrate a clear IFN signature in the skin (D Fiorentino, unpublished). Plasmacytoid dendritic cells (DCs), whose numbers were shown to be increased in DM skin lesions compared to normal skin, are proposed to be the primary source of type I IFN in DM skin lesions128; however, keratinocytes and other cell types are also known to produce type I IFNs. Expression of type I IFNs and type I IFN-inducible genes is also increased in SLE patients, and has been shown to correlate with disease activity129. In SLE, immune complexes containing nucleic acids are thought to be endocytosed by plasmacytoid DCs, resulting in activation of Toll-like receptor-7 (TLR-7) and TLR-9, and the consequent upregulation of type I IFN gene expression129.

Tyk2, a member of the JAK family, is a crucial TK in signal transduction through type I IFN receptors130. Splenocytes isolated from Tyk2-deficient mice were unable to respond to a low concentration of IFNα131. DCs from a separate mouse strain that also exhibit defective Tyk2 signaling were significantly less responsive to stimulation through TLR-9 or TLR-4, and mice were protected against collagen-induced arthritis132. These mice were also resistant to experimental allergic encephalomyelitis133. In addition, polymorphisms in the Tyk2 gene are associated with certain subtypes of SLE134135. These studies suggest that Tyk2 signaling may contribute to the etiology and pathogenesis of certain autoimmune diseases; given the role of type I IFN in DM and SLE, we speculate that targeting Tyk2 may provide therapeutic benefit in these diseases. Although Tyk2 inhibitors are being developed, there are currently no Tyk2 inhibitors in clinical trials. We await the availability of Tyk2 inhibitors and suggest that there is rationale to targeting Tyk2 in DM and SLE.

Other TKs have also been implicated in SLE and DM. The nonreceptor TK spleen tyrosine kinase (Syk), which is critical for T-cell receptor, B-cell receptor, and activating Fc receptor signaling, is upregulated in lymphocytes from SLE patients, and Syk inhibition ameliorated disease in an animal model of SLE136137. Imatinib has also been found to treat systemic manifestations of SLE and enhance survival in mice, possibly through interruption of the PDGFR pathway138139. A recent case report found that sorafenib, which inhibits PDGFR and VEGFR in addition to the serine-threonine kinases Raf-1 and B-Raf, administered to a patient with DM and hepatocellular carcinoma led to improvement of DM severity, although therapy was discontinued after only 6 weeks140. These studies suggest the potential utility of TK inhibitors in the treatment of DM and SLE.

Concluding Remarks

TKs play pivotal roles in cellular responses that may contribute to the pathogenesis of several autoimmune and inflammatory dermatologic diseases (Figure 3). Targeting implicated TKs with small-molecule inhibitors may provide a powerful therapeutic approach for these difficult-to-treat disorders. It will be important to continue to define the relative contributions of specific TKs to the disease processes in order to determine which inhibitors have potential as therapeutic agents.

An external file that holds a picture, illustration, etc.
Object name is nihms-200971-f0003.jpg

Cell types and responses mediated by exemplary kinases that may contribute to the pathogenesis of inflammatory dermatologic disorders. Fibroblast proliferation, differentiation, migration, and survival are promoted by signaling through PDGFRs, as well as through TGFβ receptors via c-Abl. Signaling through VEGF or EGF receptors induces proliferation, migration, survival, and adhesion of keratinocytes and endothelial cells. Syk contributes to B-cell receptor signaling, inducing B-cell proliferation and activation. JAK3 propagates T-cell signals through Type I cytokine receptors that contain the common gamma chain, thereby promoting T-cell proliferation, activation, and development. Tyk2 is important in Type I IFN signaling, which leads to cellular responses including IFN signature gene upregulation, DC differentiation, and Th responses. Abbreviations: DC, dendritic cell; EGFR, epidermal growth factor receptor; IFN-R, interferon receptor; PDGFR, platelet-derived growth factor receptor; TGF, transforming growth factor; VEGFR, vascular endothelial cell growth factor receptor; Syk, spleen tyrosine kinase; JAK3, janus kinase 3.

Findings from oncology studies have taught us that the presence of a target, for examplean upregulated TK, does not guarantee clinical response to targeted therapy, for example with a TK inhibitor141. Nor do animal models always mirror human disease to a degree that would allow for confident translation of therapies from mouse to human. Nevertheless, the studies described herein suggest that TKs have central roles in the pathogenesis of several important dermatologic diseases. Effective targeting of the appropriate molecules with specific TK inhibitors may provide significant benefit in these diseases (Table 1).

Table 1

TKs implicated in the pathogenesis of select dermatologic diseases and TK inhibitors with at least one case report describing potential therapeutic benefit. Note that the TK inhibitors are not necessarily directly linked to the TKs in the middle column.

DiseaseImplicated TKsTK inhibitors reported to provide potential benefit
Systemic sclerosisPDGFR2027, 39, Abl32, 3439, EGFR144Imatinib34, 35, 3942, 4446, 49, Nilotinib43, Dasatinib43
GVHDPDGFR56Imatinib57
NSFc-Abl64Imatinib6567
PsoriasisVEGFR73, 74, 7780, JAK1/394, EGFR8389, 145147, Src148, 149NVP-BAW288172, R33394, PD16954091, Sunitinib97, Imatinib98, Lapatinib103, Cetuximab104
Pemphigus vulgarisEGFR110112, Src112AG1478110
Bullous pemphigoidVEGFR120, 121Imatinib122
DermatomyositisTyk2125, 127, 128, 130Sorafenib140
SLETyk2129, 134, 135, 150, 151, Syk136, 137Imatinib138, 139

Taking another lesson from oncology research, we may find that specific inhibition of a single TK is not sufficient to provide substantial benefit in certain inflammatory dermatologic diseases. A landmark study demonstrated that, in cells from patients with glioblastoma multiforme, multiple receptor TKs are activated simultaneously and cooperate in the maintenance of pathogenic signaling142. Inhibition of multiple receptor TKs, rather than inhibition of a single receptor TK, was required to disrupt pathogenic signaling142. PDGFR and FGFR signaling pathways have been shown to synergize in the neovascularization of tumors. Likewise, our group has shown that PDGF and TGFβ, by activating c-Abl, synergize in the induction of SSc fibroblast proliferation, suggesting that inhibiting multiple TKs may be more efficacious than inhibiting a single TK in autoimmune diseases as well143. Thus, treatment with TK inhibitors that target multiple pathways should not be neglected as an approach to the treatment of complex inflammatory dermatologic diseases. However, inhibition of more TKs increases the likelihood of detrimental side effects, such that the optimal TK inhibitor will represent a trade-off between efficacy and toxicity.

Cancer is frequently driven by mutations in kinases, and thus successful treatment of cancer requires high doses of TK inhibitors. In contrast, autoimmune diseases are mediated by aberrant activation of wild-type kinases, against which low doses of inhibitor may be effective. The use of low doses of TK inhibitors in autoimmune disease would result in enhanced tolerability and safety.

We expect that the next generation of TK inhibitors will include topical formulations, which may specifically treat the cutaneous manifestations of inflammatory dermatologic diseases. Topical TK inhibitors should theoretically provide local benefit while avoiding the systemic toxicity observed with orally administered TK inhibitors.

Inhibition of TKs by molecules that are already FDA-approved or in earlier stages of clinical development represents a promising treatment strategy for inflammatory dermatologic diseases. Significant rationale exists for prospective clinical trials to determine whether TK inhibitors may provide therapeutic efficacy, and it is essential that data from rigorous clinical trials be used to guide treatment decisions in this important clinical area.

Acknowledgements

This work was supported by the NIH National Institute of Arthritis and Musculoskeletal and Skin Diseases grant R01-AR-054822 and by the Department of Veterans Affairs funding to W.H.R. Dr. Chung receives support from the Department of Veterans Affairs. Dr. Fiorentino receives support from the Scleroderma Research Foundation. The authors appreciate the editorial input of Dr. Tamsin M. Lindstrom.

Funding sources: This work was supported by the NIH National Institute of Arthritis and Musculoskeletal and Skin Diseases grant R01-AR-054822 and by the Department of Veterans Affairs funding to W.H.R. Dr. Chung receives support from the Department of Veterans Affairs. Dr. Fiorentino receives support from the Scleroderma Research Foundation.

Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
GRECC, Palo Alto VA Health Care System, 3801 Miranda Ave., Palo Alto, CA 94304, USA
Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
Correspondence and reprint requests should be addressed to W.H.R.: ude.drofnats@sniborw; 650.849.1207 phone, 650.849.1208 fax.
Publisher's Disclaimer

Abstract

Tyrosine kinases are enzymes that catalyze the phosphorylation of tyrosine residues on protein substrates. They are key components of signaling pathways that drive an array of cellular responses including proliferation, differentiation, migration, and survival. Specific tyrosine kinases have recently been identified as critical to the pathogenesis of several autoimmune and inflammatory diseases. Small-molecule inhibitors of tyrosine kinases are emerging as a novel class of therapy that may provide benefit in certain patient subsets. In this review, we highlight tyrosine kinase signaling implicated in inflammatory dermatologic diseases, evaluate strategies aimed at inhibiting these aberrant signaling pathways, and discuss prospects for future drug development.

Keywords: Tyrosine kinase, phosphorylation, inflammatory, autoimmune, dermatology, fibrosis, psoriasis, pemphigus, dermatomyositis
Abstract

Abbreviations and Acronyms

TKtyrosine kinase
PDGFRplatelet-derived growth factor receptor
VEGFRvascular endothelial growth factor receptor
EGFRepidermal growth factor receptor
FGFRfibroblast growth factor receptor
JAKjanus kinase
AblAbelson
ARGAbelson-related gene
Srcsarcoma
SScsystemic sclerosis
TGFtransforming growth factor
PVpemphigus vulgaris
Dsgdesmoglein
GISTgastrointestinal stromal tumor
DMdermatomyositis
IFNinterferon
DCdendritic cell
TLRToll-like receptor
GVHDgraft-versus-host-disease
HCThematopoietic cell transplantation
APCantigen-presenting cell
NSFnephrogenic systemic fibrosis
Sykspleen tyrosine kinase
FLT3c-Fms-like tyrosine kinase 3
Abbreviations and Acronyms

Footnotes

Publisher's Disclaimer: 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 citable 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.

Statement on prior presentation: This manuscript has not been published or submitted for publication elsewhere. The authors have no conflicts of interest to declare.

Footnotes

References

  • 1. Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S, Savai R, Roth M, et al Reversal of experimental pulmonary hypertension by PDGF inhibition. J Clin Invest. 2005;115:2811–21.[Google Scholar]
  • 2. Ghofrani HA, Seeger W, Grimminger FImatinib for the treatment of pulmonary arterial hypertension. N Engl J Med. 2005;353:1412–3.[PubMed][Google Scholar]
  • 3. Wong BR, Grossbard EB, Payan DG, Masuda ESTargeting Syk as a treatment for allergic and autoimmune disorders. Expert Opin Investig Drugs. 2004;13:743–62.[PubMed][Google Scholar]
  • 4. Louvet C, Szot GL, Lang J, Lee MR, Martinier N, Bollag G, et al Tyrosine kinase inhibitors reverse type 1 diabetes in nonobese diabetic mice. Proc Natl Acad Sci U S A. 2008;105:18895–900.[Google Scholar]
  • 5. D'Aura Swanson C, Paniagua RT, Lindstrom TM, Robinson WHTyrosine kinases as targets for the treatment of rheumatoid arthritis. Nat Rev Rheumatol. 2009;5:317–24.[Google Scholar]
  • 6. Eklund KK, Lindstedt K, Sandler C, Kovanen PT, Laasonen L, Juurikivi A, et al Maintained efficacy of the tyrosine kinase inhibitor imatinib mesylate in a patient with rheumatoid arthritis. J Clin Rheumatol. 2008;14:294–6.[PubMed][Google Scholar]
  • 7. Paniagua RT, Sharpe O, Ho PP, Chan SM, Chang A, Higgins JP, et al Selective tyrosine kinase inhibition by imatinib mesylate for the treatment of autoimmune arthritis. J Clin Invest. 2006;116:2633–42.[Google Scholar]
  • 8. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam SThe protein kinase complement of the human genome. Science. 2002;298:1912–34.[PubMed][Google Scholar]
  • 9. Robinson DR, Wu YM, Lin SFThe protein tyrosine kinase family of the human genome. Oncogene. 2000;19:5548–57.[PubMed][Google Scholar]
  • 10. Schlessinger JCell signaling by receptor tyrosine kinases. Cell. 2000;103:211–25.[PubMed][Google Scholar]
  • 11. Hubbard SR, Till JHProtein tyrosine kinase structure and function. Annu Rev Biochem. 2000;69:373–98.[PubMed][Google Scholar]
  • 12. Krause DS, Van Etten RATyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353:172–87.[PubMed][Google Scholar]
  • 13. Ghoreschi K, Laurence A, O'Shea JJSelectivity and therapeutic inhibition of kinases: to be or not to be? Nat Immunol. 2009;10:356–60.[Google Scholar]
  • 14. Johnson LNProtein kinase inhibitors: contributions from structure to clinical compounds. Q Rev Biophys. 2009;42:1–40.[PubMed][Google Scholar]
  • 15. Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B, Kuriyan JStructural mechanism for STI-571 inhibition of abelson tyrosine kinase. Science. 2000;289:1938–42.[PubMed][Google Scholar]
  • 16. Gabrielli A, Avvedimento EV, Krieg TScleroderma. N Engl J Med. 2009;360:1989–2003.[PubMed][Google Scholar]
  • 17. Mayes MD, Lacey JV, Jr., Beebe-Dimmer J, Gillespie BW, Cooper B, Laing TJ, et al Prevalence, incidence, survival, and disease characteristics of systemic sclerosis in a large US population. Arthritis Rheum. 2003;48:2246–55.[PubMed][Google Scholar]
  • 18. Dibb NJ, Dilworth SM, Mol CDSwitching on kinases: oncogenic activation of BRAF and the PDGFR family. Nat Rev Cancer. 2004;4:718–27.[PubMed][Google Scholar]
  • 19. Andrae J, Gallini R, Betsholtz CRole of platelet-derived growth factors in physiology and medicine. Genes Dev. 2008;22:1276–312.[Google Scholar]
  • 20. Gay S, Jones RE, Jr., Huang GQ, Gay REImmunohistologic demonstration of platelet-derived growth factor (PDGF) and sis-oncogene expression in scleroderma. J Invest Dermatol. 1989;92:301–3.[PubMed][Google Scholar]
  • 21. Ludwicka A, Ohba T, Trojanowska M, Yamakage A, Strange C, Smith EA, et al Elevated levels of platelet derived growth factor and transforming growth factor-beta 1 in bronchoalveolar lavage fluid from patients with scleroderma. J Rheumatol. 1995;22:1876–83.[PubMed][Google Scholar]
  • 22. Pandolfi A, Florita M, Altomare G, Pigatto P, Donati MB, Poggi AIncreased plasma levels of platelet-derived growth factor activity in patients with progressive systemic sclerosis. Proc Soc Exp Biol Med. 1989;191:1–4.[PubMed][Google Scholar]
  • 23. Klareskog L, Gustafsson R, Scheynius A, Hallgren RIncreased expression of platelet-derived growth factor type B receptors in the skin of patients with systemic sclerosis. Arthritis Rheum. 1990;33:1534–41.[PubMed][Google Scholar]
  • 24. Zheng XY, Zhang JZ, Tu P, Ma SQExpression of platelet-derived growth factor B-chain and platelet-derived growth factor beta-receptor in fibroblasts of scleroderma. J Dermatol Sci. 1998;18:90–7.[PubMed][Google Scholar]
  • 25. Yamakage A, Kikuchi K, Smith EA, LeRoy EC, Trojanowska MSelective upregulation of platelet-derived growth factor alpha receptors by transforming growth factor beta in scleroderma fibroblasts. J Exp Med. 1992;175:1227–34.[Google Scholar]
  • 26. Soria A, Cario-Andre M, Lepreux S, Rezvani HR, Pasquet JM, Pain C, et al The effect of imatinib (Glivec) on scleroderma and normal dermal fibroblasts: a preclinical study. Dermatology. 2008;216:109–17.[PubMed][Google Scholar]
  • 27. Baroni SS, Santillo M, Bevilacqua F, Luchetti M, Spadoni T, Mancini M, et al Stimulatory autoantibodies to the PDGF receptor in systemic sclerosis. N Engl J Med. 2006;354:2667–76.[PubMed][Google Scholar]
  • 28. Loizos N, Lariccia L, Weiner J, Griffith H, Boin F, Hummers L, et al Lack of detection of agonist activity by antibodies to platelet-derived growth factor receptor alpha in a subset of normal and systemic sclerosis patient sera. Arthritis Rheum. 2009;60:1145–51.[PubMed][Google Scholar]
  • 29. Balada E, Simeon-Aznar CP, Ordi-Ros J, Rosa-Leyva M, Selva-O'Callaghan A, Pardos-Gea J, et al Anti-PDGFR-alpha antibodies measured by non-bioactivity assays are not specific for systemic sclerosis. Ann Rheum Dis. 2008;67:1027–9.[PubMed][Google Scholar]
  • 30. Classen JF, Henrohn D, Rorsman F, Lennartsson J, Lauwerys BR, Wikstrom G, et al Lack of evidence of stimulatory autoantibodies to platelet-derived growth factor receptor in patients with systemic sclerosis. Arthritis Rheum. 2009;60:1137–44.[PubMed][Google Scholar]
  • 31. Varga J, Pasche BTransforming growth factor beta as a therapeutic target in systemic sclerosis. Nat Rev Rheumatol. 2009;5:200–6.[Google Scholar]
  • 32. Sonnylal S, Denton CP, Zheng B, Keene DR, He R, Adams HP, et al Postnatal induction of transforming growth factor beta signaling in fibroblasts of mice recapitulates clinical, histologic, and biochemical features of scleroderma. Arthritis Rheum. 2007;56:334–44.[PubMed][Google Scholar]
  • 33. Derynck R, Zhang YESmad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature. 2003;425:577–84.[PubMed][Google Scholar]
  • 34. Daniels CE, Wilkes MC, Edens M, Kottom TJ, Murphy SJ, Limper AH, et al Imatinib mesylate inhibits the profibrogenic activity of TGF-beta and prevents bleomycin-mediated lung fibrosis. J Clin Invest. 2004;114:1308–16.[Google Scholar]
  • 35. Wang S, Wilkes MC, Leof EB, Hirschberg RImatinib mesylate blocks a non-Smad TGF-beta pathway and reduces renal fibrogenesis in vivo. FASEB J. 2005;19:1–11.[PubMed][Google Scholar]
  • 36. Boureux A, Furstoss O, Simon V, Roche SAbl tyrosine kinase regulates a Rac/JNK and a Rac/Nox pathway for DNA synthesis and Myc expression induced by growth factors. J Cell Sci. 2005;118:3717–26.[PubMed][Google Scholar]
  • 37. Plattner R, Koleske AJ, Kazlauskas A, Pendergast AMBidirectional signaling links the Abelson kinases to the platelet-derived growth factor receptor. Mol Cell Biol. 2004;24:2573–83.[Google Scholar]
  • 38. Sini P, Cannas A, Koleske AJ, Di Fiore PP, Scita GAbl-dependent tyrosine phosphorylation of Sos-1 mediates growth-factor-induced Rac activation. Nat Cell Biol. 2004;6:268–74.[PubMed][Google Scholar]
  • 39. Chung L, Fiorentino DF, Benbarak MJ, Adler AS, Mariano MM, Paniagua RT, et al Molecular framework for response to imatinib mesylate in systemic sclerosis. Arthritis Rheum. 2009;60:584–91.[Google Scholar]
  • 40. Yoshiji H, Noguchi R, Kuriyama S, Ikenaka Y, Yoshii J, Yanase K, et al Imatinib mesylate (STI-571) attenuates liver fibrosis development in rats. Am J Physiol Gastrointest Liver Physiol. 2005;288:G907–13.[PubMed][Google Scholar]
  • 41. Akhmetshina A, Venalis P, Dees C, Busch N, Zwerina J, Schett G, et al Treatment with imatinib prevents fibrosis in different preclinical models of systemic sclerosis and induces regression of established fibrosis. Arthritis Rheum. 2009;60:219–24.[PubMed][Google Scholar]
  • 42. Distler JH, Jungel A, Huber LC, Schulze-Horsel U, Zwerina J, Gay RE, et al Imatinib mesylate reduces production of extracellular matrix and prevents development of experimental dermal fibrosis. Arthritis Rheum. 2007;56:311–22.[PubMed][Google Scholar]
  • 43. Akhmetshina A, Dees C, Pileckyte M, Maurer B, Axmann R, Jungel A, et al Dual inhibition of c-abl and PDGF receptor signaling by dasatinib and nilotinib for the treatment of dermal fibrosis. FASEB J. 2008;22:2214–22.[PubMed][Google Scholar]
  • 44. van Daele PL, Dik WA, Thio HB, van Hal PT, van Laar JA, Hooijkaas H, et al Is imatinib mesylate a promising drug in systemic sclerosis? Arthritis Rheum. 2008;58:2549–52.[PubMed][Google Scholar]
  • 45. Sfikakis PP, Gorgoulis VG, Katsiari CG, Evangelou K, Kostopoulos C, Black CMImatinib for the treatment of refractory, diffuse systemic sclerosis. Rheumatology (Oxford) 2008;47:735–7.[PubMed][Google Scholar]
  • 46. ten Freyhaus H, Dumitrescu D, Bovenschulte H, Erdmann E, Rosenkranz SSignificant improvement of right ventricular function by imatinib mesylate in scleroderma-associated pulmonary arterial hypertension. Clin Res Cardiol. 2009;98:265–7.[PubMed][Google Scholar]
  • 47. Sabnani I, Zucker MJ, Rosenstein ED, Baran DA, Arroyo LH, Tsang P, et al A novel therapeutic approach to the treatment of scleroderma-associated pulmonary complications: safety and efficacy of combination therapy with imatinib and cyclophosphamide. Rheumatology (Oxford) 2009;48:49–52.[PubMed][Google Scholar]
  • 48. Pope J, McBain DL, Petrlich L, Watson S, Bender L, F. D, et al. A proof of concept trial of Gleevec (imatinib) in active diffuse scleroderma (DSSc). American College of Rheumatology (ACR) / Association of Rheumatology Health Professionals (ARHP) Scientific Meeting; Philadelphia, PA. 2009. [PubMed]
  • 49. Gordon J, Mersten J, Lyman S, Kloiber SA, Wildman HF, Crow MK, et al. Imatinib mesylate (Gleevec) in the treatment of systemic sclerosis: Interim results of a Phase IIa, one year, open label clinical trial. American College of Rheumatology (ACR) / Association of Rheumatology Health Professionals (ARHP) Scientific Meeting; Philadelphia, PA. 2009. [PubMed]
  • 50. Welniak LA, Blazar BR, Murphy WJImmunobiology of allogeneic hematopoietic stem cell transplantation. Annu Rev Immunol. 2007;25:139–70.[PubMed][Google Scholar]
  • 51. Copelan EAHematopoietic stem-cell transplantation. N Engl J Med. 2006;354:1813–26.[PubMed][Google Scholar]
  • 52. Ferrara JL, Levine JE, Reddy P, Holler EGraft-versus-host disease. Lancet. 2009;373:1550–61.[Google Scholar]
  • 53. Small TN, Robinson WH, Miklos DBB cells and transplantation: an educational resource. Biol Blood Marrow Transplant. 2008;15:104–13.[Google Scholar]
  • 54. Schaffer JV, McNiff JM, Seropian S, Cooper DL, Bolognia JLLichen sclerosus and eosinophilic fasciitis as manifestations of chronic graft-versus-host disease: expanding the sclerodermoid spectrum. J Am Acad Dermatol. 2005;53:591–601.[PubMed][Google Scholar]
  • 55. Creamer D, Martyn-Simmons CL, Osborne G, Kenyon M, Salisbury JR, Devereux S, et al Eczematoid graft-vs-host disease: a novel form of chronic cutaneous graft-vs-host disease and its response to psoralen UV-A therapy. Arch Dermatol. 2007;143:1157–62.[PubMed][Google Scholar]
  • 56. Svegliati S, Olivieri A, Campelli N, Luchetti M, Poloni A, Trappolini S, et al Stimulatory autoantibodies to PDGF receptor in patients with extensive chronic graft-versus-host disease. Blood. 2007;110:237–41.[PubMed][Google Scholar]
  • 57. Olivieri ALF, Sanna R, Raimondi R, Bacigalupo A. Imatinib for the treatment of advanced refractory chronic GVHD. European Group for Blood and Marrow Transplantation Meeting; Florence, Italy. 2008. [PubMed]
  • 58. Cowper SE, Robin HS, Steinberg SM, Su LD, Gupta S, LeBoit PEScleromyxoedema-like cutaneous diseases in renal-dialysis patients. Lancet. 2000;356:1000–1.[PubMed][Google Scholar]
  • 59. Kribben A, Witzke O, Hillen U, Barkhausen J, Daul AE, Erbel RNephrogenic systemic fibrosis: pathogenesis, diagnosis, and therapy. J Am Coll Cardiol. 2009;53:1621–8.[PubMed][Google Scholar]
  • 60. Bangsgaard N, Marckmann P, Rossen K, Skov LNephrogenic systemic fibrosis: late skin manifestations. Arch Dermatol. 2009;145:183–7.[PubMed][Google Scholar]
  • 61. Daram SR, Cortese CM, Bastani BNephrogenic fibrosing dermopathy/nephrogenic systemic fibrosis: report of a new case with literature review. Am J Kidney Dis. 2005;46:754–9.[PubMed][Google Scholar]
  • 62. Galan A, Cowper SE, Bucala RNephrogenic systemic fibrosis (nephrogenic fibrosing dermopathy) Curr Opin Rheumatol. 2006;18:614–7.[PubMed][Google Scholar]
  • 63. Quan TE, Cowper S, Wu SP, Bockenstedt LK, Bucala RCirculating fibrocytes: collagen-secreting cells of the peripheral blood. Int J Biochem Cell Biol. 2004;36:598–606.[PubMed][Google Scholar]
  • 64. Jimenez SA, Artlett CM, Sandorfi N, Derk C, Latinis K, Sawaya H, et al Dialysis-associated systemic fibrosis (nephrogenic fibrosing dermopathy): study of inflammatory cells and transforming growth factor beta1 expression in affected skin. Arthritis Rheum. 2004;50:2660–6.[PubMed][Google Scholar]
  • 65. Kay J, High WAImatinib mesylate treatment of nephrogenic systemic fibrosis. Arthritis Rheum. 2008;58:2543–8.[PubMed][Google Scholar]
  • 66. Chandran S, Petersen J, Jacobs C, Fiorentino D, Doeden K, Lafayette RAImatinib in the treatment of nephrogenic systemic fibrosis. Am J Kidney Dis. 2009;53:129–32.[PubMed][Google Scholar]
  • 67. Kay J, Sullivan ME, Patel TV. Efficacy and safety of oral imatinib mesylate in patients with nephrogenic systemic fibrosis. American College of Rheumatology (ACR) / Association of Rheumatology Health Professionals (ARHP) Scientific Meeting; Philadelphia, PA. 2009. [PubMed]
  • 68. Schon MP, Boehncke WHPsoriasis. N Engl J Med. 2005;352:1899–912.[PubMed][Google Scholar]
  • 69. Bowcock AM, Krueger JGGetting under the skin: the immunogenetics of psoriasis. Nat Rev Immunol. 2005;5:699–711.[PubMed][Google Scholar]
  • 70. Zenz R, Eferl R, Kenner L, Florin L, Hummerich L, Mehic D, et al Psoriasis-like skin disease and arthritis caused by inducible epidermal deletion of Jun proteins. Nature. 2005;437:369–75.[PubMed][Google Scholar]
  • 71. Boniface K, Diveu C, Morel F, Pedretti N, Froger J, Ravon E, et al Oncostatin M secreted by skin infiltrating T lymphocytes is a potent keratinocyte activator involved in skin inflammation. J Immunol. 2007;178:4615–22.[PubMed][Google Scholar]
  • 72. Halin C, Fahrngruber H, Meingassner JG, Bold G, Littlewood-Evans A, Stuetz A, et al Inhibition of chronic and acute skin inflammation by treatment with a vascular endothelial growth factor receptor tyrosine kinase inhibitor. Am J Pathol. 2008;173:265–77.[Google Scholar]
  • 73. Kunstfeld R, Hirakawa S, Hong YK, Schacht V, Lange-Asschenfeldt B, Velasco P, et al Induction of cutaneous delayed-type hypersensitivity reactions in VEGF-A transgenic mice results in chronic skin inflammation associated with persistent lymphatic hyperplasia. Blood. 2004;104:1048–57.[PubMed][Google Scholar]
  • 74. Xia YP, Li B, Hylton D, Detmar M, Yancopoulos GD, Rudge JSTransgenic delivery of VEGF to mouse skin leads to an inflammatory condition resembling human psoriasis. Blood. 2003;102:161–8.[PubMed][Google Scholar]
  • 75. Roy H, Bhardwaj S, Yla-Herttuala SBiology of vascular endothelial growth factors. FEBS Lett. 2006;580:2879–87.[PubMed][Google Scholar]
  • 76. Wilgus TA, Matthies AM, Radek KA, Dovi JV, Burns AL, Shankar R, et al Novel function for vascular endothelial growth factor receptor-1 on epidermal keratinocytes. Am J Pathol. 2005;167:1257–66.[Google Scholar]
  • 77. Detmar M, Brown LF, Claffey KP, Yeo KT, Kocher O, Jackman RW, et al Overexpression of vascular permeability factor/vascular endothelial growth factor and its receptors in psoriasis. J Exp Med. 1994;180:1141–6.[Google Scholar]
  • 78. Man XY, Yang XH, Cai SQ, Bu ZY, Zheng MOverexpression of vascular endothelial growth factor (VEGF) receptors on keratinocytes in psoriasis: regulated by calcium independent of VEGF. J Cell Mol Med. 2008;12:649–60.[Google Scholar]
  • 79. Zhang Y, Matsuo H, Morita EVascular endothelial growth factor 121 is the predominant isoform in psoriatic scales. Exp Dermatol. 2005;14:758–64.[PubMed][Google Scholar]
  • 80. Bhushan M, McLaughlin B, Weiss JB, Griffiths CELevels of endothelial cell stimulating angiogenesis factor and vascular endothelial growth factor are elevated in psoriasis. Br J Dermatol. 1999;141:1054–60.[PubMed][Google Scholar]
  • 81. Pastore S, Mascia F, Mariani V, Girolomoni GThe epidermal growth factor receptor system in skin repair and inflammation. J Invest Dermatol. 2008;128:1365–74.[PubMed][Google Scholar]
  • 82. Schneider MR, Antsiferova M, Feldmeyer L, Dahlhoff M, Bugnon P, Hasse S, et al Betacellulin regulates hair follicle development and hair cycle induction and enhances angiogenesis in wounded skin. J Invest Dermatol. 2008;128:1256–65.[PubMed][Google Scholar]
  • 83. Gottlieb AB, Chang CK, Posnett DN, Fanelli B, Tam JPDetection of transforming growth factor alpha in normal, malignant, and hyperproliferative human keratinocytes. J Exp Med. 1988;167:670–5.[Google Scholar]
  • 84. Elder JT, Fisher GJ, Lindquist PB, Bennett GL, Pittelkow MR, Coffey RJ, Jr., et al Overexpression of transforming growth factor alpha in psoriatic epidermis. Science. 1989;243:811–4.[PubMed][Google Scholar]
  • 85. Cook PW, Pittelkow MR, Keeble WW, Graves-Deal R, Coffey RJ, Jr., Shipley GDAmphiregulin messenger RNA is elevated in psoriatic epidermis and gastrointestinal carcinomas. Cancer Res. 1992;52:3224–7.[PubMed][Google Scholar]
  • 86. Piepkorn MOverexpression of amphiregulin, a major autocrine growth factor for cultured human keratinocytes, in hyperproliferative skin diseases. Am J Dermatopathol. 1996;18:165–71.[PubMed][Google Scholar]
  • 87. Zheng Y, Peng Z, Wang Y, Tan S, Xi Y, Wang GAlteration and significance of heparin-binding epidermal-growth-factor-like growth factor in psoriatic epidermis. Dermatology. 2003;207:22–7.[PubMed][Google Scholar]
  • 88. Shirakata Y, Kishimoto J, Tokumaru S, Yamasaki K, Hanakawa Y, Tohyama M, et al Epiregulin, a member of the EGF family, is over-expressed in psoriatic epidermis. J Dermatol Sci. 2007;45:69–72.[PubMed][Google Scholar]
  • 89. Cook PW, Piepkorn M, Clegg CH, Plowman GD, DeMay JM, Brown JR, et al Transgenic expression of the human amphiregulin gene induces a psoriasis-like phenotype. J Clin Invest. 1997;100:2286–94.[Google Scholar]
  • 90. Varani J, Kang S, Stoll S, Elder JTHuman psoriatic skin in organ culture: comparison with normal skin exposed to exogenous growth factors and effects of an antibody to the EGF receptor. Pathobiology. 1998;66:253–9.[PubMed][Google Scholar]
  • 91. Varani J, Lateef H, Fay K, Elder JTAntagonism of epidermal growth factor receptor tyrosine kinase ameliorates the psoriatic phenotype in organ-cultured skin. Skin Pharmacol Physiol. 2005;18:123–31.[PubMed][Google Scholar]
  • 92. Di Cesare A, Di Meglio P, Nestle FOThe IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 2009;129:1339–50.[PubMed][Google Scholar]
  • 93. Pesu M, Laurence A, Kishore N, Zwillich S, Chan G, O'Shea JJTherapeutic targeting of janus kinases. Immunol Rev. 2008;223:132–42.[Google Scholar]
  • 94. Chang BY, Zhao F, He X, Ren H, Braselmann S, Taylor V, et al JAK3 inhibition significantly attenuates psoriasiform skin inflammation in CD18 mutant PL/J mice. J Immunol. 2009;183:2183–92.[PubMed][Google Scholar]
  • 95. Changelian PS, Flanagan ME, Ball DJ, Kent CR, Magnuson KS, Martin WH, et al Prevention of organ allograft rejection by a specific Janus kinase 3 inhibitor. Science. 2003;302:875–8.[PubMed][Google Scholar]
  • 96. Boy MG, Wang C, Wilkinson BE, Chow VF, Clucas AT, Krueger JG, et al Double-blind, placebo-controlled, dose-escalation study to evaluate the pharmacologic effect of CP-690,550 in patients with psoriasis. J Invest Dermatol. 2009;129:2299–302.[PubMed][Google Scholar]
  • 97. Keshtgarpour M, Dudek AZSU-011248, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, controls chronic psoriasis. Transl Res. 2007;149:103–6.[PubMed][Google Scholar]
  • 98. Miyagawa S, Fujimoto H, Ko S, Hirota S, Kitamura YImprovement of psoriasis during imatinib therapy in a patient with a metastatic gastrointestinal stromal tumour. Br J Dermatol. 2002;147:406–7.[PubMed][Google Scholar]
  • 99. Woo SM, Huh CH, Park KC, Youn SWExacerbation of psoriasis in a chronic myelogenous leukemia patient treated with imatinib. J Dermatol. 2007;34:724–6.[PubMed][Google Scholar]
  • 100. Cheng H, Geist DE, Piperdi M, Virk R, Piperdi BManagement of imatinib-related exacerbation of psoriasis in a patient with a gastrointestinal stromal tumour. Australas J Dermatol. 2009;50:41–3.[PubMed][Google Scholar]
  • 101. Dickens E, Lewis F, Bienz NImatinib: a designer drug, another cutaneous complication. Clin Exp Dermatol. 2009;34:603–4.[PubMed][Google Scholar]
  • 102. Lacouture MEMechanisms of cutaneous toxicities to EGFR inhibitors. Nat Rev Cancer. 2006;6:803–12.[PubMed][Google Scholar]
  • 103. Wierzbicka E, Tourani JM, Guillet G. Improvement of psoriasis and cutaneous side-effects during tyrosine kinase inhibitor therapy for renal metastatic adenocarcinoma. A role for epidermal growth factor receptor (EGFR) inhibitors in psoriasis? Br J Dermatol. 2006;155:213–4.[PubMed]
  • 104. Neyns B, Meert V, Vandenbroucke FCetuximab treatment in a patient with metastatic colorectal cancer and psoriasis. Curr Oncol. 2008;15:196–7.[Google Scholar]
  • 105. Herbst RS, Fukuoka M, Baselga JGefitinib--a novel targeted approach to treating cancer. Nat Rev Cancer. 2004;4:956–65.[PubMed][Google Scholar]
  • 106. Zorzou MP, Stratigos A, Efstathiou E, Bamias AExacerbation of psoriasis after treatment with an EGFR tyrosine kinase inhibitor. Acta Derm Venereol. 2004;84:308–9.[PubMed][Google Scholar]
  • 107. Stanley JR, Amagai MPemphigus, bullous impetigo, and the staphylococcal scalded-skin syndrome. N Engl J Med. 2006;355:1800–10.[PubMed][Google Scholar]
  • 108. Nguyen VT, Ndoye A, Shultz LD, Pittelkow MR, Grando SAAntibodies against keratinocyte antigens other than desmogleins 1 and 3 can induce pemphigus vulgaris-like lesions. J Clin Invest. 2000;106:1467–79.[Google Scholar]
  • 109. Anhalt GJ, Labib RS, Voorhees JJ, Beals TF, Diaz LAInduction of pemphigus in neonatal mice by passive transfer of IgG from patients with the disease. N Engl J Med. 1982;306:1189–96.[PubMed][Google Scholar]
  • 110. Frusic-Zlotkin M, Raichenberg D, Wang X, David M, Michel B, Milner YApoptotic mechanism in pemphigus autoimmunoglobulins-induced acantholysis--possible involvement of the EGF receptor. Autoimmunity. 2006;39:563–75.[PubMed][Google Scholar]
  • 111. Pretel M, Espana A, Marquina M, Pelacho B, Lopez-Picazo JM, Lopez-Zabalza MJAn imbalance in Akt/mTOR is involved in the apoptotic and acantholytic processes in a mouse model of pemphigus vulgaris. Exp Dermatol. 2009;18:771–80.[PubMed][Google Scholar]
  • 112. Chernyavsky AI, Arredondo J, Kitajima Y, Sato-Nagai M, Grando SADesmoglein versus non-desmoglein signaling in pemphigus acantholysis: characterization of novel signaling pathways downstream of pemphigus vulgaris antigens. J Biol Chem. 2007;282:13804–12.[PubMed][Google Scholar]
  • 113. Tatton L, Morley GM, Chopra R, Khwaja AThe Src-selective kinase inhibitor PP1 also inhibits Kit and Bcr-Abl tyrosine kinases. J Biol Chem. 2003;278:4847–53.[PubMed][Google Scholar]
  • 114. Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, Itoh N, et al Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem. 1987;262:5592–5.[PubMed][Google Scholar]
  • 115. Sanchez-Carpintero I, Espana A, Pelacho B, Lopez Moratalla N, Rubenstein DS, Diaz LA, et al In vivo blockade of pemphigus vulgaris acantholysis by inhibition of intracellular signal transduction cascades. Br J Dermatol. 2004;151:565–70.[PubMed][Google Scholar]
  • 116. Marquina M, Espana A, Fernandez-Galar M, Lopez-Zabalza MJThe role of nitric oxide synthases in pemphigus vulgaris in a mouse model. Br J Dermatol. 2008;159:68–76.[PubMed][Google Scholar]
  • 117. Delva E, Jennings JM, Calkins CC, Kottke MD, Faundez V, Kowalczyk APPemphigus vulgaris IgG-induced desmoglein-3 endocytosis and desmosomal disassembly are mediated by a clathrin- and dynamin-independent mechanism. J Biol Chem. 2008;283:18303–13.[Google Scholar]
  • 118. Berkowitz P, Hu P, Warren S, Liu Z, Diaz LA, Rubenstein DSp38MAPK inhibition prevents disease in pemphigus vulgaris mice. Proc Natl Acad Sci U S A. 2006;103:12855–60.[Google Scholar]
  • 119. Joly P, Roujeau JC, Benichou J, Picard C, Dreno B, Delaporte E, et al A comparison of oral and topical corticosteroids in patients with bullous pemphigoid. N Engl J Med. 2002;346:321–7.[PubMed][Google Scholar]
  • 120. Ameglio F, D'Auria L, Cordiali-Fei P, Mussi A, Valenzano L, D'Agosto G, et al Bullous pemphigoid and pemphigus vulgaris: correlated behaviour of serum VEGF, sE-selectin and TNF-alpha levels. J Biol Regul Homeost Agents. 1997;11:148–53.[PubMed][Google Scholar]
  • 121. Brown LF, Harrist TJ, Yeo KT, Stahle-Backdahl M, Jackman RW, Berse B, et al Increased expression of vascular permeability factor (vascular endothelial growth factor) in bullous pemphigoid, dermatitis herpetiformis, and erythema multiforme. J Invest Dermatol. 1995;104:744–9.[PubMed][Google Scholar]
  • 122. Hofmann SC, Technau K, Muller AM, Lubbert M, Bruckner-Tuderman LBullous pemphigoid associated with hypereosinophilic syndrome: simultaneous response to imatinib. J Am Acad Dermatol. 2007;56:S68–72.[PubMed][Google Scholar]
  • 123. Krathen MS, Fiorentino D, Werth VPDermatomyositis. Curr Dir Autoimmun. 2008;10:313–32.[Google Scholar]
  • 124. Rothfield N, Sontheimer RD, Bernstein MLupus erythematosus: systemic and cutaneous manifestations. Clin Dermatol. 2006;24:348–62.[PubMed][Google Scholar]
  • 125. Greenberg SA, Pinkus JL, Pinkus GS, Burleson T, Sanoudou D, Tawil R, et al Interferon-alpha/beta-mediated innate immune mechanisms in dermatomyositis. Ann Neurol. 2005;57:664–78.[PubMed][Google Scholar]
  • 126. Wenzel J, Worenkamper E, Freutel S, Henze S, Haller O, Bieber T, et al Enhanced type I interferon signalling promotes Th1-biased inflammation in cutaneous lupus erythematosus. J Pathol. 2005;205:435–42.[PubMed][Google Scholar]
  • 127. Walsh RJ, Kong SW, Yao Y, Jallal B, Kiener PA, Pinkus JL, et al Type I interferon inducible gene expression in blood is present and reflects disease activity in dermatomyositis and polymyositis. Arthritis Rheum. 2007;56:3784–92.[Google Scholar]
  • 128. Wenzel J, Schmidt R, Proelss J, Zahn S, Bieber T, Tuting TType I interferon-associated skin recruitment of CXCR3+ lymphocytes in dermatomyositis. Clin Exp Dermatol. 2006;31:576–82.[PubMed][Google Scholar]
  • 129. Ronnblom L, Alm GV, Eloranta MLType I interferon and lupus. Curr Opin Rheumatol. 2009;21:471–7.[PubMed][Google Scholar]
  • 130. Ghoreschi K, Laurence A, O'Shea JJJanus kinases in immune cell signaling. Immunol Rev. 2009;228:273–87.[Google Scholar]
  • 131. Shimoda K, Kato K, Aoki K, Matsuda T, Miyamoto A, Shibamori M, et al Tyk2 plays a restricted role in IFN alpha signaling, although it is required for IL-12-mediated T cell function. Immunity. 2000;13:561–71.[PubMed][Google Scholar]
  • 132. Ortmann R, Smeltz R, Yap G, Sher A, Shevach EM. A heritable defect in IL-12 signaling in B10.Q/J mice. I. In vitro analysis. J Immunol. 2001;166:5712–9.[PubMed]
  • 133. Spach KM, Noubade R, McElvany B, Hickey WF, Blankenhorn EP, Teuscher CA single nucleotide polymorphism in Tyk2 controls susceptibility to experimental allergic encephalomyelitis. J Immunol. 2009;182:7776–83.[Google Scholar]
  • 134. Hellquist A, Jarvinen TM, Koskenmies S, Zucchelli M, Orsmark-Pietras C, Berglind L, et al Evidence for genetic association and interaction between the TYK2 and IRF5 genes in systemic lupus erythematosus. J Rheumatol. 2009;36:1631–8.[PubMed][Google Scholar]
  • 135. Jarvinen TM, Hellquist A, Koskenmies S, Einarsdottir E, Koskinen LL, Jeskanen L, et al Tyrosine kinase 2 and interferon regulatory factor 5 polymorphisms are associated with discoid and subacute cutaneous lupus erythematosus. Exp Dermatol. 2009[PubMed][Google Scholar]
  • 136. Krishnan S, Juang YT, Chowdhury B, Magilavy A, Fisher CU, Nguyen H, et al Differential expression and molecular associations of Syk in systemic lupus erythematosus T cells. J Immunol. 2008;181:8145–52.[Google Scholar]
  • 137. Bahjat FR, Pine PR, Reitsma A, Cassafer G, Baluom M, Grillo S, et al An orally bioavailable spleen tyrosine kinase inhibitor delays disease progression and prolongs survival in murine lupus. Arthritis Rheum. 2008;58:1433–44.[PubMed][Google Scholar]
  • 138. Zoja C, Corna D, Rottoli D, Zanchi C, Abbate M, Remuzzi GImatinib ameliorates renal disease and survival in murine lupus autoimmune disease. Kidney Int. 2006;70:97–103.[PubMed][Google Scholar]
  • 139. Sadanaga A, Nakashima H, Masutani K, Miyake K, Shimizu S, Igawa T, et al Amelioration of autoimmune nephritis by imatinib in MRL/lpr mice. Arthritis Rheum. 2005;52:3987–96.[PubMed][Google Scholar]
  • 140. Apostolidis L, Kahlert C, Siegmund A, Thom R, Horstmann S, Jager D, et al Remission of paraneoplastic dermatomyositis associated with hepatocellular carcinoma under prednisolone and azathiopin, and concommittant sorafenib. Onkologie. 2009;32:50–3.[PubMed][Google Scholar]
  • 141. Bergsland EKWhen does the presence of the target predict response to the targeted agent? J Clin Oncol. 2006;24:213–6.[PubMed][Google Scholar]
  • 142. Stommel JM, Kimmelman AC, Ying H, Nabioullin R, Ponugoti AH, Wiedemeyer R, et al Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science. 2007;318:287–90.[PubMed][Google Scholar]
  • 143. Nissen LJ, Cao R, Hedlund EM, Wang Z, Zhao X, Wetterskog D, et al Angiogenic factors FGF2 and PDGF-BB synergistically promote murine tumor neovascularization and metastasis. J Clin Invest. 2007;117:2766–77.[Google Scholar]
  • 144. Planque S, Zhou YX, Nishiyama Y, Sinha M, O'Connor-Mccourt M, Arnett FC, et al Autoantibodies to the epidermal growth factor receptor in systemic sclerosis, lupus, and autoimmune mice. FASEB J. 2003;17:136–43.[PubMed][Google Scholar]
  • 145. Amagai M, Ozawa S, Ueda M, Nishikawa T, Abe O, Shimizu NDistribution of EGF receptor expressing and DNA replicating epidermal cells in psoriasis vulgaris and Bowen's disease. Br J Dermatol. 1988;119:661–8.[PubMed][Google Scholar]
  • 146. Nanney LB, Stoscheck CM, Magid M, King LE., JrAltered [125I]epidermal growth factor binding and receptor distribution in psoriasis. J Invest Dermatol. 1986;86:260–5.[PubMed][Google Scholar]
  • 147. Higashiyama M, Hashimoto K, Matsumoto K, Yoshikawa KDifferential expression of transforming growth factor-alpha (TGF-alpha) and EGF receptor in transitional area of psoriatic epidermis. J Dermatol Sci. 1994;7:45–53.[PubMed][Google Scholar]
  • 148. Ayli EE, Li W, Brown TT, Witkiewicz A, Elenitsas R, Seykora JTActivation of Src-family tyrosine kinases in hyperproliferative epidermal disorders. J Cutan Pathol. 2008;35:273–7.[Google Scholar]
  • 149. Pulitzer M, Li W, Hanson M, Singh F, Elenitsas R, Gelfand JM, et al Srcasm overexpression in psoriasis-insights into pathogenesis. J Cutan Pathol. 2007;34:160–5.[PubMed][Google Scholar]
  • 150. Cunninghame Graham DS, Akil M, Vyse TJAssociation of polymorphisms across the tyrosine kinase gene, TYK2 in UK SLE families. Rheumatology (Oxford) 2007;46:927–30.[PubMed][Google Scholar]
  • 151. Sigurdsson S, Nordmark G, Goring HH, Lindroos K, Wiman AC, Sturfelt G, et al Polymorphisms in the tyrosine kinase 2 and interferon regulatory factor 5 genes are associated with systemic lupus erythematosus. Am J Hum Genet. 2005;76:528–37.[Google Scholar]
  • 152. Blake S, Lyons A, Hughes TNilotinib inhibits the Src-family kinase Lck and T-cell function in vivo. J Cell Mol Med. 2008;13:599–601.[Google Scholar]
  • 153. Hantschel O, Rix U, Schmidt U, Burckstummer T, Kneidinger M, Schutze G, et al The Btk tyrosine kinase is a major target of the Bcr-Abl inhibitor dasatinib. Proc Natl Acad Sci U S A. 2007;104:13283–8.[Google Scholar]
  • 154. Blake S, Hughes T, Mayrhofer G, Lyons AThe Src/Abl inhibitor dasatinib (BMS-354825) inhibits function of normal human T-lymphocytes in vitro. Clin Immunol. 2008;127:330–9.[PubMed][Google Scholar]
  • 155. Faivre S, Demetri G, Sargent W, Raymond EMolecular basis for sunitinib efficacy and future clinical development. Nat Rev Drug Discov. 2007;6:734–45.[PubMed][Google Scholar]
  • 156. Cameron D, Stein SDrug insight: intracellular inhibitors of HER2 - clinical development of lapatinib in breast cancer. Nat Clin Pract Oncol. 2008;5:512–20.[PubMed][Google Scholar]
  • 157. Wilhelm S, Adnane L, Newell P, Villanueva A, Llovet J, Lynch MPreclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling. Mol Cancer Ther. 2008;7:3129–40.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.