1
|
Bohdziewicz A, Pawlik KK, Maciejewska M, Sikora M, Alda-Malicka R, Czuwara J, Rudnicka L. Future Treatment Options in Systemic Sclerosis-Potential Targets and Ongoing Clinical Trials. J Clin Med 2022; 11:1310. [PMID: 35268401 PMCID: PMC8911443 DOI: 10.3390/jcm11051310] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/14/2022] [Accepted: 02/24/2022] [Indexed: 02/03/2023] Open
Abstract
Systemic sclerosis is an autoimmune connective tissue disease characterized by vasculopathy and fibrosis of the skin and internal organs. The pathogenesis of systemic sclerosis is very complex. Mediators produced by immune cells are involved in the inflammatory processes occurring in the tissues. The currently available therapeutic options are often insufficient to halt disease progress. This article presents an overview of potential therapeutic targets and the pipeline of possible future therapeutic options. It is based on research of clinical trials involving novel, unestablished methods of treatment. Increasing knowledge of the processes and mediators involved in systemic scleroderma has led to the initiation of drug trials with therapeutic targets of CD28-CD80/86, CD19, CCL24, CD20, CD30, tumor necrosis factor (TNF), transforming growth factor β (TGF-β), B-cell activating factor (BAFF), lysophosphatidic acid receptor 1 (LPA1 receptor), soluble guanylate cyclase (sGC), Janus kinases (JAK), interleukin 6 (IL-6), endothelin receptor, and autotaxin. Data from clinical trials on these drugs indicate a significant potential for several new therapeutic options for systemic sclerosis in the upcoming future.
Collapse
Affiliation(s)
- Anna Bohdziewicz
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland; (A.B.); (K.K.P.); (R.A.-M.); (J.C.); (L.R.)
| | - Katarzyna Karina Pawlik
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland; (A.B.); (K.K.P.); (R.A.-M.); (J.C.); (L.R.)
| | - Magdalena Maciejewska
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland; (A.B.); (K.K.P.); (R.A.-M.); (J.C.); (L.R.)
| | - Mariusz Sikora
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland;
| | - Rosanna Alda-Malicka
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland; (A.B.); (K.K.P.); (R.A.-M.); (J.C.); (L.R.)
| | - Joanna Czuwara
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland; (A.B.); (K.K.P.); (R.A.-M.); (J.C.); (L.R.)
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland; (A.B.); (K.K.P.); (R.A.-M.); (J.C.); (L.R.)
| |
Collapse
|
2
|
Mendoza FA, Piera-Velazquez S, Jimenez SA. Tyrosine kinases in the pathogenesis of tissue fibrosis in systemic sclerosis and potential therapeutic role of their inhibition. Transl Res 2021; 231:139-158. [PMID: 33422651 DOI: 10.1016/j.trsl.2021.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/09/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Systemic sclerosis (SSc) is an idiopathic autoimmune disease with a heterogeneous clinical phenotype ranging from limited cutaneous involvement to rapidly progressive diffuse SSc. The most severe SSc clinical and pathologic manifestations result from an uncontrolled fibrotic process involving the skin and various internal organs. The molecular mechanisms responsible for the initiation and progression of the SSc fibrotic process have not been fully elucidated. Recently it has been suggested that tyrosine protein kinases play a role. The implicated kinases include receptor-activated tyrosine kinases and nonreceptor tyrosine kinases. The receptor kinases are activated following specific binding of growth factors (platelet-derived growth factor, fibroblast growth factor, or vascular endothelial growth factor). Other receptor kinases are the discoidin domain receptors activated by binding of various collagens, the ephrin receptors that are activated by ephrins and the angiopoetin-Tie-2s receptors. The nonreceptor tyrosine kinases c-Abl, Src, Janus, and STATs have also been shown to participate in SSc-associated tissue fibrosis. Currently, there are no effective disease-modifying therapies for SSc-associated tissue fibrosis. Therefore, extensive investigation has been conducted to examine whether tyrosine kinase inhibitors (TKIs) may exert antifibrotic effects. Here, we review the role of receptor and nonreceptor tyrosine kinases in the pathogenesis of the frequently progressive cutaneous and systemic fibrotic alterations in SSc, and the potential of TKIs as SSc disease-modifying antifibrotic therapeutic agents.
Collapse
Affiliation(s)
- Fabian A Mendoza
- Rheumatology Division, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania; Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
3
|
Zhao M, Wu J, Wu H, Sawalha AH, Lu Q. Clinical Treatment Options in Scleroderma: Recommendations and Comprehensive Review. Clin Rev Allergy Immunol 2021; 62:273-291. [PMID: 33449302 DOI: 10.1007/s12016-020-08831-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
There are two major clinical subsets of scleroderma: (i) systemic sclerosis (SSc) is a complex systemic autoimmune disorder characterized by inflammation, vasculopathy, and excessive fibrosis of the skin and multiple internal organs and (ii) localized scleroderma (LoS), also known as morphea, is confined to the skin and/or subcutaneous tissues resulting in collagen deposition and subsequent fibrosis. SSc is rare but is associated with significant morbidity and mortality compared with other rheumatic diseases. Fatal outcomes in SSc often originate from organ complications of the disease, such as lung fibrosis, pulmonary artery hypertension (PAH), and scleroderma renal crisis (SRC). Current treatment modalities in SSc have focused on targeting vascular damage, fibrosis, and regulation of inflammation as well as autoimmune responses. Some drugs previously used in an attempt to suppress fibrosis, like D-penicillamine (D-Pen) or colchicine, have been disappointing in clinical practice despite anecdotal evidence of their advantages. Some canonical medications, including glucocorticoids, immunosuppressants, and vasodilators, have had some success in treating various manifestations in SSc patients. Increasing evidence suggests that some biologic agents targeting collagen, cytokines, and cell surface molecules might have promising therapeutic effects in SSc. In recent years, hematopoietic stem cell transplantation (HSCT), mostly autologous, has made great progress as a promising treatment option in severe and refractory SSc. Due to the complexity and heterogeneity of SSc, there are currently no optimal treatments for all aspects of the disease. As for LoS, local skin-targeted therapy is generally used, including topical application of glucocorticoids or other immunomodulatory ointments and ultraviolet (UV) irradiation. In addition, systemic immunosuppressants are also utilized in several forms of LoS. Here, we comprehensively discuss current treatment options for scleroderma, encompassing old, new, and future potential treatment options. In addition, we summarize data from new clinical trials that have the potential to modify the disease process and improve long-term outcomes in SSc.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Immune-Related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences, Changsha, China
| | - Jiali Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Immune-Related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Immune-Related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences, Changsha, China
| | - Amr H Sawalha
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China. .,Research Unit of Key Technologies of Immune-Related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences, Changsha, China. .,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
4
|
Antin-Ozerkis D, Hinchcliff M. Connective Tissue Disease-Associated Interstitial Lung Disease: Evaluation and Management. Clin Chest Med 2020; 40:617-636. [PMID: 31376896 DOI: 10.1016/j.ccm.2019.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interstitial lung disease is common among patients with connective tissue disease and is an important contributor to morbidity and mortality. Infection and drug toxicity must always be excluded as the cause of radiographic findings. Immunosuppression remains a mainstay of therapy despite few controlled trials supporting its use. When a decision regarding therapy initiation is made, considerations include an assessment of disease severity as well as a determination of the rate of progression. Because patients may have extrathoracic disease activity, a multidisciplinary approach is crucial and should include supportive and nonpharmacologic management strategies.
Collapse
Affiliation(s)
- Danielle Antin-Ozerkis
- Section of Pulmonary and Critical Care Medicine, Yale School of Medicine, PO Box 208057, New Haven, CT 06520-8057, USA.
| | - Monique Hinchcliff
- Section of Rheumatology, Allergy and Immunology, Yale School of Medicine, PO Box 208031, New Haven, CT 06520-8031, USA
| |
Collapse
|
5
|
Wermuth PJ, Jimenez SA. Abrogation of transforming growth factor-β-induced tissue fibrosis in TBRIcaCol1a2Cre transgenic mice by the second generation tyrosine kinase inhibitor SKI-606 (Bosutinib). PLoS One 2018; 13:e0196559. [PMID: 29718973 PMCID: PMC5931634 DOI: 10.1371/journal.pone.0196559] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/16/2018] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor-β (TGF-β) plays a crucial role in the pathogenesis of Systemic Sclerosis (SSc) and other fibrotic disorders. TGF-β-mediated c-Abl and Src kinase activation induces strong profibrotic cascade signaling. The purpose of this study was to test in vivo the antifibrotic activity of Bosutinib (SKI-606), a second generation c-Abl and Src kinase inhibitor, on TGF-β induced cutaneous and pulmonary fibrosis. For this purpose, we employed the TBRIcaCol1a2Cre transgenic mice expressing an inducible constitutively active TGF-β receptor 1 constitutively activated by Col1a promoter-mediated Cre recombinase. The mice were treated parenterally with 2.5, 5.0 or 10.0 mg/kg/day of Bosutinib for 42 days. Skin and lungs from control and Bosutinib-treated mice (n = 6 per group) were assessed by histopathology, measurement of tissue hydroxyproline content, PCR analysis of tissue fibrosis associated gene expression, and evidence of myofibroblast activation. Mice with constitutive TGF-β-1 signaling displayed severe cutaneous and pulmonary fibrosis. Bosutinib administration decreased collagen deposition and hydroxyproline content in the dermis and lungs in a dose-dependent manner. Bosutinib also reversed the marked increase in profibrotic and myofibroblast activation-associated gene expression. These results demonstrate that constitutive TGF-β-1-signaling-induced cutaneous and pulmonary fibrosis were abrogated in a dose-related manner following parenteral administration of the c-Abl and Src tyrosine kinase inhibitor, Bosutinib. These results indicate that Bosutinib may be a potential therapeutic agent for tissue fibrosis in SSc and other fibroproliferative disorders.
Collapse
Affiliation(s)
- Peter J Wermuth
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States of America
| |
Collapse
|
6
|
Heldin CH, Lennartsson J, Westermark B. Involvement of platelet-derived growth factor ligands and receptors in tumorigenesis. J Intern Med 2018; 283:16-44. [PMID: 28940884 DOI: 10.1111/joim.12690] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Platelet-derived growth factor (PDGF) isoforms and their receptors have important roles during embryogenesis, particularly in the development of various mesenchymal cell types in different organs. In the adult, PDGF stimulates wound healing and regulates tissue homeostasis. However, overactivity of PDGF signalling is associated with malignancies and other diseases characterized by excessive cell proliferation, such as fibrotic conditions and atherosclerosis. In certain tumours, genetic or epigenetic alterations of the genes for PDGF ligands and receptors drive tumour cell proliferation and survival. Examples include the rare skin tumour dermatofibrosarcoma protuberance, which is driven by autocrine PDGF stimulation due to translocation of a PDGF gene, and certain gastrointestinal stromal tumours and leukaemias, which are driven by constitute activation of PDGF receptors due to point mutations and formation of fusion proteins of the receptors, respectively. Moreover, PDGF stimulates cells in tumour stroma and promotes angiogenesis as well as the development of cancer-associated fibroblasts, both of which promote tumour progression. Inhibitors of PDGF signalling may thus be of clinical usefulness in the treatment of certain tumours.
Collapse
Affiliation(s)
- C-H Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - J Lennartsson
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - B Westermark
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
7
|
Sacchetti C, Bottini N. Protein Tyrosine Phosphatases in Systemic Sclerosis: Potential Pathogenic Players and Therapeutic Targets. Curr Rheumatol Rep 2017; 19:28. [PMID: 28397126 DOI: 10.1007/s11926-017-0655-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW The pathogenesis of systemic sclerosis depends on a complex interplay between autoimmunity, vasculopathy, and fibrosis. Reversible phosphorylation on tyrosine residues, in response to growth factors and other stimuli, critically regulates each one of these three key pathogenic processes. Protein tyrosine kinases, the enzymes that catalyze addition of phosphate to tyrosine residues, are known players in systemic sclerosis, and tyrosine kinase inhibitors are undergoing clinical trials for treatment of this disease. Until recently, the role of tyrosine phosphatases-the enzymes that counteract the action of tyrosine kinases by removing phosphate from tyrosine residues-in systemic sclerosis has remained largely unknown. Here, we review the function of tyrosine phosphatases in pathways relevant to the pathogenesis of systemic sclerosis and their potential promise as therapeutic targets to halt progression of this debilitating rheumatic disease. RECENT FINDINGS Protein tyrosine phosphatases are emerging as important regulators of a multitude of signaling pathways and undergoing validation as molecular targets for cancer and other common diseases. Recent advances in drug discovery are paving the ways to develop new classes of tyrosine phosphatase modulators to treat human diseases. Although so far only few reports have focused on tyrosine phosphatases in systemic sclerosis, these enzymes play a role in multiple pathways relevant to disease pathogenesis. Further studies in this field are warranted to explore the potential of tyrosine phosphatases as drug targets for systemic sclerosis.
Collapse
Affiliation(s)
- Cristiano Sacchetti
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California, San Diego, 9500 Gilman Drive MC #0656, La Jolla, CA, 92093, USA
| | - Nunzio Bottini
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California, San Diego, 9500 Gilman Drive MC #0656, La Jolla, CA, 92093, USA.
| |
Collapse
|
8
|
Haddon DJ, Wand HE, Jarrell JA, Spiera RF, Utz PJ, Gordon JK, Chung LS. Proteomic Analysis of Sera from Individuals with Diffuse Cutaneous Systemic Sclerosis Reveals a Multianalyte Signature Associated with Clinical Improvement during Imatinib Mesylate Treatment. J Rheumatol 2017; 44:631-638. [PMID: 28298564 PMCID: PMC5860882 DOI: 10.3899/jrheum.160833] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2017] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Imatinib has been investigated for the treatment of systemic sclerosis (SSc) because of its ability to inhibit the platelet-derived growth factor receptor and transforming growth factor-β signaling pathways, which have been implicated in SSc pathogenesis. In a 12-month open-label clinical trial assessing the safety and efficacy of imatinib in the treatment of diffuse cutaneous SSc (dcSSc), significant improvements in skin thickening were observed. Here, we report our analysis of sera collected during the clinical trial. METHODS We measured the levels of 46 cytokines, chemokines, and growth factors in the sera of individuals with dcSSc using Luminex and ELISA. Autoantigen microarrays were used to measure immunoglobulin G reactivity to 28 autoantigens. Elastic net regularization was used to identify a signature that was predictive of clinical improvement (reduction in the modified Rodnan skin score ≥ 5) during treatment with imatinib. The signature was also tested using sera from a clinical trial of nilotinib, a tyrosine kinase inhibitor that is structurally related to imatinib, in dcSSc. RESULTS The elastic net algorithm identified a signature, based on levels of CD40 ligand, chemokine (C-X-C motif) ligand 4 (CXCL4), and anti-PM/Scl-100, that was significantly higher in individuals who experienced clinical improvement than in those who did not (p = 0.0011). The signature was validated using samples from a clinical trial of nilotinib. CONCLUSION Identification of patients with SSc with the greatest probability of benefit from treatment with imatinib has the potential to guide individualized treatment. Validation of the signature will require testing in randomized, placebo-controlled studies. Clinicaltrials.gov NCT00555581 and NCT01166139.
Collapse
Affiliation(s)
- D James Haddon
- From the Division of Immunology and Rheumatology, Stanford University School of Medicine; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford; Department of Rheumatology, Palo Alto VA Health Care System, Palo Alto, California; Department of Rheumatology, Hospital for Special Surgery, New York, New York, USA
- D.J. Haddon, PhD, Research Associate, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine; H.E. Wand, BS, Genetic Counseling Candidate, Division of Immunology and Rheumatology, Stanford University School of Medicine; J.A. Jarrell, PhD Candidate, Immunology, Division of Immunology and Rheumatology, Stanford University School of Medicine; R.F. Spiera, MD, Professor of Clinical Medicine, Rheumatology and Director, Vasculitis and Scleroderma Program, Department of Rheumatology, Hospital for Special Surgery; P.J. Utz, MD, Professor of Medicine, Immunology and Rheumatology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine; J.K. Gordon, MD, Assistant Professor of Medicine, Rheumatology, Department of Rheumatology, Hospital for Special Surgery, New York; L.S. Chung, MD, MS, Associate Professor of Medicine, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, and Department of Rheumatology, Palo Alto VA Health Care System
| | - Hannah E Wand
- From the Division of Immunology and Rheumatology, Stanford University School of Medicine; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford; Department of Rheumatology, Palo Alto VA Health Care System, Palo Alto, California; Department of Rheumatology, Hospital for Special Surgery, New York, New York, USA
- D.J. Haddon, PhD, Research Associate, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine; H.E. Wand, BS, Genetic Counseling Candidate, Division of Immunology and Rheumatology, Stanford University School of Medicine; J.A. Jarrell, PhD Candidate, Immunology, Division of Immunology and Rheumatology, Stanford University School of Medicine; R.F. Spiera, MD, Professor of Clinical Medicine, Rheumatology and Director, Vasculitis and Scleroderma Program, Department of Rheumatology, Hospital for Special Surgery; P.J. Utz, MD, Professor of Medicine, Immunology and Rheumatology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine; J.K. Gordon, MD, Assistant Professor of Medicine, Rheumatology, Department of Rheumatology, Hospital for Special Surgery, New York; L.S. Chung, MD, MS, Associate Professor of Medicine, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, and Department of Rheumatology, Palo Alto VA Health Care System
| | - Justin A Jarrell
- From the Division of Immunology and Rheumatology, Stanford University School of Medicine; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford; Department of Rheumatology, Palo Alto VA Health Care System, Palo Alto, California; Department of Rheumatology, Hospital for Special Surgery, New York, New York, USA
- D.J. Haddon, PhD, Research Associate, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine; H.E. Wand, BS, Genetic Counseling Candidate, Division of Immunology and Rheumatology, Stanford University School of Medicine; J.A. Jarrell, PhD Candidate, Immunology, Division of Immunology and Rheumatology, Stanford University School of Medicine; R.F. Spiera, MD, Professor of Clinical Medicine, Rheumatology and Director, Vasculitis and Scleroderma Program, Department of Rheumatology, Hospital for Special Surgery; P.J. Utz, MD, Professor of Medicine, Immunology and Rheumatology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine; J.K. Gordon, MD, Assistant Professor of Medicine, Rheumatology, Department of Rheumatology, Hospital for Special Surgery, New York; L.S. Chung, MD, MS, Associate Professor of Medicine, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, and Department of Rheumatology, Palo Alto VA Health Care System
| | - Robert F Spiera
- From the Division of Immunology and Rheumatology, Stanford University School of Medicine; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford; Department of Rheumatology, Palo Alto VA Health Care System, Palo Alto, California; Department of Rheumatology, Hospital for Special Surgery, New York, New York, USA
- D.J. Haddon, PhD, Research Associate, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine; H.E. Wand, BS, Genetic Counseling Candidate, Division of Immunology and Rheumatology, Stanford University School of Medicine; J.A. Jarrell, PhD Candidate, Immunology, Division of Immunology and Rheumatology, Stanford University School of Medicine; R.F. Spiera, MD, Professor of Clinical Medicine, Rheumatology and Director, Vasculitis and Scleroderma Program, Department of Rheumatology, Hospital for Special Surgery; P.J. Utz, MD, Professor of Medicine, Immunology and Rheumatology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine; J.K. Gordon, MD, Assistant Professor of Medicine, Rheumatology, Department of Rheumatology, Hospital for Special Surgery, New York; L.S. Chung, MD, MS, Associate Professor of Medicine, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, and Department of Rheumatology, Palo Alto VA Health Care System
| | - Paul J Utz
- From the Division of Immunology and Rheumatology, Stanford University School of Medicine; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford; Department of Rheumatology, Palo Alto VA Health Care System, Palo Alto, California; Department of Rheumatology, Hospital for Special Surgery, New York, New York, USA
- D.J. Haddon, PhD, Research Associate, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine; H.E. Wand, BS, Genetic Counseling Candidate, Division of Immunology and Rheumatology, Stanford University School of Medicine; J.A. Jarrell, PhD Candidate, Immunology, Division of Immunology and Rheumatology, Stanford University School of Medicine; R.F. Spiera, MD, Professor of Clinical Medicine, Rheumatology and Director, Vasculitis and Scleroderma Program, Department of Rheumatology, Hospital for Special Surgery; P.J. Utz, MD, Professor of Medicine, Immunology and Rheumatology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine; J.K. Gordon, MD, Assistant Professor of Medicine, Rheumatology, Department of Rheumatology, Hospital for Special Surgery, New York; L.S. Chung, MD, MS, Associate Professor of Medicine, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, and Department of Rheumatology, Palo Alto VA Health Care System
| | - Jessica K Gordon
- From the Division of Immunology and Rheumatology, Stanford University School of Medicine; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford; Department of Rheumatology, Palo Alto VA Health Care System, Palo Alto, California; Department of Rheumatology, Hospital for Special Surgery, New York, New York, USA
- D.J. Haddon, PhD, Research Associate, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine; H.E. Wand, BS, Genetic Counseling Candidate, Division of Immunology and Rheumatology, Stanford University School of Medicine; J.A. Jarrell, PhD Candidate, Immunology, Division of Immunology and Rheumatology, Stanford University School of Medicine; R.F. Spiera, MD, Professor of Clinical Medicine, Rheumatology and Director, Vasculitis and Scleroderma Program, Department of Rheumatology, Hospital for Special Surgery; P.J. Utz, MD, Professor of Medicine, Immunology and Rheumatology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine; J.K. Gordon, MD, Assistant Professor of Medicine, Rheumatology, Department of Rheumatology, Hospital for Special Surgery, New York; L.S. Chung, MD, MS, Associate Professor of Medicine, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, and Department of Rheumatology, Palo Alto VA Health Care System
| | - Lorinda S Chung
- From the Division of Immunology and Rheumatology, Stanford University School of Medicine; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford; Department of Rheumatology, Palo Alto VA Health Care System, Palo Alto, California; Department of Rheumatology, Hospital for Special Surgery, New York, New York, USA.
- D.J. Haddon, PhD, Research Associate, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine; H.E. Wand, BS, Genetic Counseling Candidate, Division of Immunology and Rheumatology, Stanford University School of Medicine; J.A. Jarrell, PhD Candidate, Immunology, Division of Immunology and Rheumatology, Stanford University School of Medicine; R.F. Spiera, MD, Professor of Clinical Medicine, Rheumatology and Director, Vasculitis and Scleroderma Program, Department of Rheumatology, Hospital for Special Surgery; P.J. Utz, MD, Professor of Medicine, Immunology and Rheumatology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine; J.K. Gordon, MD, Assistant Professor of Medicine, Rheumatology, Department of Rheumatology, Hospital for Special Surgery, New York; L.S. Chung, MD, MS, Associate Professor of Medicine, Immunology and Rheumatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, and Department of Rheumatology, Palo Alto VA Health Care System.
| |
Collapse
|
9
|
Kang HS, Rhee CK, Lee HY, Yoon HK, Kwon SS, Lee SY. Different anti-remodeling effect of nilotinib and fluticasone in a chronic asthma model. Korean J Intern Med 2016; 31:1150-1158. [PMID: 27764539 PMCID: PMC5094918 DOI: 10.3904/kjim.2015.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 07/05/2015] [Accepted: 08/16/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND/AIMS Inhaled corticosteroids are the most effective treatment currently available for asthma, but their beneficial effect against airway remodeling is limited. The tyrosine kinase inhibitor nilotinib has inhibitory activity against c-kit and the platelet-derived growth factor receptor. We compared the effects of fluticasone and nilotinib on airway remodeling in a chronic asthma model. We also examined whether co-treatment with nilotinib and fluticasone had any synergistic effect in preventing airway remodeling. METHODS We developed a mouse model of airway remodeling, including smooth muscle thickening, in which ovalbumin (OVA)-sensitized female BALB/c-mice were repeatedly exposed to intranasal OVA administration twice per week for 3 months. Mice were treated with fluticasone and/or nilotinib intranasally during the OVA challenge. RESULTS Mice chronically exposed to OVA developed eosinophilic airway inflammation and showed features of airway remodeling, including thickening of the peribronchial smooth muscle layer. Both fluticasone and nilotinib attenuated airway smooth muscle thickening. However, only nilotinib suppressed fibrotic changes, demonstrating inhibition of collagen deposition. Fluticasone reduced pro-inflammatory cells, such as eosinophils, and several cytokines, such as interleukin 4 (IL-4), IL-5, and IL-13, induced by repeated OVA challenges. On the other hand, nilotinib reduced transforming growth factor β1 levels in bronchoalveolar lavage fluid and inhibited fibroblast proliferation significantly. CONCLUSIONS These results suggest that fluticasone and nilotinib suppressed airway remodeling in this chronic asthma model through anti-inflammatory and anti-fibrotic pathways, respectively.
Collapse
Affiliation(s)
- Hye Seon Kang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Chin Kook Rhee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Hea Yon Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Hyoung Kyu Yoon
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Soon Seok Kwon
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Bucheon St. Mary’s Hospital, The Catholic University of Korea, Bucheon, Korea
| | - Sook Young Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
- Correspondence to Sook Young Lee, M.D. Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-6061 Fax: +82-2-596-2158 E-mail:
| |
Collapse
|
10
|
Artemov A, Aliper A, Korzinkin M, Lezhnina K, Jellen L, Zhukov N, Roumiantsev S, Gaifullin N, Zhavoronkov A, Borisov N, Buzdin A. A method for predicting target drug efficiency in cancer based on the analysis of signaling pathway activation. Oncotarget 2016; 6:29347-56. [PMID: 26320181 PMCID: PMC4745731 DOI: 10.18632/oncotarget.5119] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 07/24/2015] [Indexed: 02/07/2023] Open
Abstract
A new generation of anticancer therapeutics called target drugs has quickly developed in the 21st century. These drugs are tailored to inhibit cancer cell growth, proliferation, and viability by specific interactions with one or a few target proteins. However, despite formally known molecular targets for every "target" drug, patient response to treatment remains largely individual and unpredictable. Choosing the most effective personalized treatment remains a major challenge in oncology and is still largely trial and error. Here we present a novel approach for predicting target drug efficacy based on the gene expression signature of the individual tumor sample(s). The enclosed bioinformatic algorithm detects activation of intracellular regulatory pathways in the tumor in comparison to the corresponding normal tissues. According to the nature of the molecular targets of a drug, it predicts whether the drug can prevent cancer growth and survival in each individual case by blocking the abnormally activated tumor-promoting pathways or by reinforcing internal tumor suppressor cascades. To validate the method, we compared the distribution of predicted drug efficacy scores for five drugs (Sorafenib, Bevacizumab, Cetuximab, Sorafenib, Imatinib, Sunitinib) and seven cancer types (Clear Cell Renal Cell Carcinoma, Colon cancer, Lung adenocarcinoma, non-Hodgkin Lymphoma, Thyroid cancer and Sarcoma) with the available clinical trials data for the respective cancer types and drugs. The percent of responders to a drug treatment correlated significantly (Pearson's correlation 0.77 p = 0.023) with the percent of tumors showing high drug scores calculated with the current algorithm.
Collapse
Affiliation(s)
- Artem Artemov
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR.,D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexander Aliper
- D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,First Oncology Research and Advisory Center, Moscow, Russia
| | | | | | - Leslie Jellen
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nikolay Zhukov
- D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,First Oncology Research and Advisory Center, Moscow, Russia.,Pirogov Russian National Research Medical University, Department of Oncology, Hematology and Radiotherapy, Moscow, Russia
| | - Sergey Roumiantsev
- D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,Pirogov Russian National Research Medical University, Department of Oncology, Hematology and Radiotherapy, Moscow, Russia
| | - Nurshat Gaifullin
- Moscow State University, Faculty of Fundamental Medicine, Moscow, Russia
| | - Alex Zhavoronkov
- Insilico Medicine, Inc., ETC, Johns Hopkins University, Baltimore, MD, USA
| | | | - Anton Buzdin
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR.,D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
11
|
Bartscht T, Rosien B, Rades D, Kaufmann R, Biersack H, Lehnert H, Gieseler F, Ungefroren H. Dasatinib blocks transcriptional and promigratory responses to transforming growth factor-beta in pancreatic adenocarcinoma cells through inhibition of Smad signalling: implications for in vivo mode of action. Mol Cancer 2015; 14:199. [PMID: 26588899 PMCID: PMC4654868 DOI: 10.1186/s12943-015-0468-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/08/2015] [Indexed: 12/12/2022] Open
Abstract
Background We have previously shown in pancreatic ductal adenocarcinoma (PDAC) cells that the SRC inhibitors PP2 and PP1 effectively inhibited TGF-β1-mediated cellular responses by blocking the kinase function of the TGF-β type I receptor ALK5 rather than SRC. Here, we investigated the ability of the clinically utilised SRC/ABL inhibitor dasatinib to mimic the PP2/PP1 effect. Methods The effect of dasatinib on TGF-β1-dependent Smad2/3 phosphorylation, general transcriptional activity, gene expression, cell motility, and the generation of tumour stem cells was measured in Panc-1 and Colo-357 cells using immunoblotting, reporter gene assays, RT-PCR, impedance-based real-time measurement of cell migration, and colony formation assays, respectively. Results In both PDAC cell lines, dasatinib effectively blocked TGF-β1-induced Smad phosphorylation, activity of 3TPlux and pCAGA(12)-luc reporter genes, cell migration, and expression of individual TGF-β1 target genes associated with epithelial-mesenchymal transition and invasion. Moreover, dasatinib strongly interfered with the TGF-β1-induced generation of tumour stem cells as demonstrated by gene expression analysis and single cell colony formation. Dasatinib also inhibited the high constitutive migratory activity conferred on Panc-1 cells by ectopic expression of kinase-active ALK5. Conclusions Our data suggest that the clinical efficiency of dasatinib may in part be due to cross-inhibition of tumour-promoting TGF-β signalling. Dasatinib may be useful as a dual TGF-β/SRC inhibitor in experimental and clinical therapeutics to prevent metastatic spread in late-stage PDAC and other tumours. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0468-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tobias Bartscht
- First Department of Medicine, UKSH, Campus Lübeck, 23538, Lübeck, Germany
| | - Benjamin Rosien
- First Department of Medicine, UKSH, Campus Lübeck, 23538, Lübeck, Germany
| | - Dirk Rades
- Department of Radiation Oncology, UKSH, Campus Lübeck, D-23538, Lübeck, Germany
| | - Roland Kaufmann
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, D-07747, Jena, Germany
| | - Harald Biersack
- First Department of Medicine, UKSH, Campus Lübeck, 23538, Lübeck, Germany
| | - Hendrik Lehnert
- First Department of Medicine, UKSH, Campus Lübeck, 23538, Lübeck, Germany
| | - Frank Gieseler
- First Department of Medicine, UKSH, Campus Lübeck, 23538, Lübeck, Germany
| | - Hendrik Ungefroren
- First Department of Medicine, UKSH, Campus Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
12
|
Update on etiopathogenesis of systemic sclerosis. REVISTA BRASILEIRA DE REUMATOLOGIA 2015; 53:516-24. [PMID: 24477730 DOI: 10.1016/j.rbr.2013.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 02/28/2013] [Indexed: 02/06/2023] Open
Abstract
Systemic Sclerosis (SSc) is an autoimmune disease of multifactorial etiology, triggered by a combination of genetic and environmental factors. Its varied clinical expression results from the complex physiopathogenic interaction of three main elements: proliferative vasculopathy, immune dysregulation and abnormal deposition and remodeling of the extracellular matrix (ECM), of which the characteristic disease fibrosis is the result. Early physiopathogenic events appear to be endothelial injury and imbalance in vascular repair with the activation of endothelial cells, the immune system and platelets, with the release of multiple mediators such as TH2 proinflammatory cytokines and growth factors, triggering a sequence of simultaneous or cascading events that involve several intracellular signaling pathways. The most important result of these events is the hyperactivation of fibroblasts, the main effector cells of fibrosis, which will then produce large amounts of ECM constituents and secrete multiple growth factors and cytokines that perpetuate the process. In this article we review the main factors potentially involved in the etiology of SSc and reexamine the current knowledge about the most important mechanisms involved in the development of lesions that are characteristic of the disease. A better understanding of these physiopathogenic mechanisms will help identify potential therapeutic targets, which may result in advances in the management of this complex and debilitating disease.
Collapse
|
13
|
Cong L, Xia ZK, Yang RY. Targeting the TGF-β receptor with kinase inhibitors for scleroderma therapy. Arch Pharm (Weinheim) 2014; 347:609-15. [PMID: 24917246 DOI: 10.1002/ardp.201400116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/28/2014] [Accepted: 05/05/2014] [Indexed: 11/12/2022]
Abstract
Scleroderma (systemic sclerosis) is a connective tissue disease that affects various organ systems; the treatment of scleroderma is still difficult and remains a challenge to the clinician. Recently, kinase inhibitors have shown great potential against fibrotic diseases and, specifically, the transforming growth factor-β receptor (TGF-βR) was found as a new and promising target for scleroderma therapy. In the current study, we propose that the large pool of existing kinase inhibitors could be exploited for inhibiting the TGF-βR to suppress scleroderma. In this respect, we developed a modeling protocol to systematically profile the inhibitory activities of 169 commercially available kinase inhibitors against the TGF-βR, from which five promising candidates were selected and tested using a standard kinase assay protocol. Consequently, two molecular entities, namely the PKB inhibitor MK-2206 and the mTOR C1/C2 inhibitor AZD8055, showed high potency when bound to the TGF-βR, with IC50 values of 97 and 86 nM, respectively, which are close to those of the recently developed TGF-βR selective inhibitors SB525334 and LY2157299 (IC50 = 14.3 and 56 nM, respectively). We also performed atomistic molecular dynamics simulations and post-molecular mechanics/Poisson-Boltzmann surface area analyses to dissect the structural basis and energetic properties of intermolecular interactions between the TGF-βR kinase domain and these potent compounds, highlighting intensive nonbonded networks across the tightly packed interface of non-cognate TGF-βR-inhibitor complexes.
Collapse
Affiliation(s)
- Lin Cong
- Department of Dermatology, General Hospital of Beijing Military Command, Beijing, China; Graduate School, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|
14
|
Kitayama J, Emoto S, Yamaguchi H, Ishigami H, Watanabe T. CD90+ mesothelial-like cells in peritoneal fluid promote peritoneal metastasis by forming a tumor permissive microenvironment. PLoS One 2014; 9:e86516. [PMID: 24466130 PMCID: PMC3897715 DOI: 10.1371/journal.pone.0086516] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/13/2013] [Indexed: 01/09/2023] Open
Abstract
The peritoneal cavity is a common target of metastatic gastrointestinal and ovarian cancer cells, but the mechanisms leading to peritoneal metastasis have not been fully elucidated. In this study, we examined the roles of cells in peritoneal fluids on the development of peritoneal metastasis. We found that a minor subset of human intraperitoneal cells with CD90(+)/CD45(−) phenotype vigorously grew in culture with mesothelial-like appearance. The mesothelial-like cells (MLC) displayed the characteristics of mesenchymal stem cell, such as differentiating into adipocytes, osteocytes, and chondrocytes, and suppressing T cell proliferation. These cells highly expressed type I collagen, vimentin, α-smooth muscle actin and fibroblast activated protein-α by the stimulation with TGF-β, which is characteristic of activated myofibroblasts. Intraperitoneal co-injection of MLCs with the human gastric cancer cell line, MKN45, significantly enhanced the rate of metastatic formation in the peritoneum of nude mice. Histological examination revealed that many MLCs were engrafted in metastatic nodules and were mainly located at the fibrous area. Dasatinib, a potent tyrosine kinase inhibitor, strongly inhibited the proliferation of MLCs but not MKN45 in vitro. Nevertheless, oral administration of Dasatinib significantly inhibited the development of peritoneal metastasis of MKN45, and resulted in reduced fibrillar formation of metastatic nodules. These results suggest floating MLCs in the peritoneal fluids support the development of peritoneal metastasis possibly through the production of the permissive microenvironment, and thus the functional blockade of MLCs is a reasonable strategy to treat recurrent abdominal malignancies.
Collapse
Affiliation(s)
- Joji Kitayama
- Department of Surgical Oncology, University of Tokyo, Tokyo, Japan
- * E-mail:
| | - Shigenobu Emoto
- Department of Surgical Oncology, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
15
|
Heldin CH. Targeting the PDGF signaling pathway in the treatment of non-malignant diseases. J Neuroimmune Pharmacol 2013; 9:69-79. [PMID: 23793451 DOI: 10.1007/s11481-013-9484-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/05/2013] [Indexed: 12/13/2022]
Abstract
Platelet-derived growth factor (PDGF) is a family of mesenchymal mitogens with important functions during the embryonal development and in the control of tissue homeostasis in the adult. The PDGF isoforms exert their effects by binding to α-and β-tyrosine kinase receptors. Overactivity of PDGF signaling has been linked to the development of certain malignant and non-malignant diseases, including atherosclerosis and various fibrotic diseases. Different types of PDGF antagonists have been developed, including inhibitory monoclonal antibodies and DNA aptamers against PDGF isoforms and receptors, and receptor tyrosine kinase inhibitors. Beneficial effects have been recorded using such inhibitors in preclinical models and in patients with certain malignant as well as non-malignant diseases. The present communication summarizes the use of PDGF antagonists in the treatment of non-malignant diseases.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research Ltd, Science for Life Laboratory, Uppsala University, Box 595, SE-75124, Uppsala, Sweden,
| |
Collapse
|
16
|
Hamama S, Delanian S, Monceau V, Vozenin MC. Therapeutic management of intestinal fibrosis induced by radiation therapy: from molecular profiling to new intervention strategies et vice et versa. FIBROGENESIS & TISSUE REPAIR 2012; 5:S13. [PMID: 23259677 PMCID: PMC3368760 DOI: 10.1186/1755-1536-5-s1-s13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Chronic toxicities of locoregional and systemic oncological treatments commonly develop in long-term cancer survivors. Amongst these toxicities, post-radiotherapeutic complications alter patient's quality of life. Reduction of exposure of normal tissues can be achieved by optimization of radiotherapy. Furthermore, understanding of the fibrogenic mechanisms has provided targets to prevent, mitigate, and reverse late radiation-induced damages. This mini-review shows how (i) global molecular studies using gene profiling can provide tools to develop new intervention strategies and (ii) how successful clinical trials, conducted in particular with combined pentoxifylline-vitamin E, can take benefice of biological and molecular evidences to improve our understanding of fibrogenic mechanisms, enhance the robustness of proposed treatments, and lead ultimately to better treatments for patient's benefice.
Collapse
Affiliation(s)
- Saad Hamama
- INSERM U-1030 "Molecular Radiotherapy" Institut Gustave Roussy, Villejuif, France ; "Molecular Radiotherapy", Université Paris Sud Paris XI, France
| | - Sylvie Delanian
- INSERM U-1030 "Molecular Radiotherapy" Institut Gustave Roussy, Villejuif, France ; "Molecular Radiotherapy", Université Paris Sud Paris XI, France ; Unité de Radiopathologie, Service Oncologie-Radiothérapie, Hôpital Saint-Louis, APHP, Paris, France
| | - Virginie Monceau
- INSERM U-1030 "Molecular Radiotherapy" Institut Gustave Roussy, Villejuif, France
| | - Marie-Catherine Vozenin
- INSERM U-1030 "Molecular Radiotherapy" Institut Gustave Roussy, Villejuif, France ; "Molecular Radiotherapy", Université Paris Sud Paris XI, France
| |
Collapse
|
17
|
Ratcliffe AJ. The Drug Discovery and Development of Kinase Inhibitors Outside of Oncology. KINASE DRUG DISCOVERY 2011. [DOI: 10.1039/9781849733557-00218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
18
|
Current world literature. Curr Opin Rheumatol 2011; 23:620-5. [PMID: 21960037 DOI: 10.1097/bor.0b013e32834ccef6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Almeida I, Faria R, Vita P, Vasconcelos C. Systemic sclerosis refractory disease: from the skin to the heart. Autoimmun Rev 2011; 10:693-701. [PMID: 21575745 DOI: 10.1016/j.autrev.2011.04.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Systemic sclerosis or scleroderma (SSc) is an heterogeneous disease involving the connective tissue and the microvasculature with fibrosis and vascular occlusion. It is difficult to define refractory SSc once it is itself a paradigm of a refractory condition: there is no evidence of when to act to stop the progression to fibrosis and irreversible microvascular damage. There is no definition of refractory disease in SSc and to propose a definition we used mainly the Medsger severity index and the EULAR 2009 treatment recommendations from the skin to the heart through peripheral vascular, musculoskeletal, gastrointestinal, renal, pulmonary hypertension and interstitial lung disease. We used some clinical setting reflecting the different reasoning when there is probable refractory disease and finally we briefly pointed out some available treatment options to refractory disease. With this reflection, we would like to open paths to a broader discussion.
Collapse
Affiliation(s)
- Isabel Almeida
- Unidade de Imunologia Clínica, Hospital de Santo António, Centro Hospitalar do Porto, Portugal.
| | | | | | | |
Collapse
|