51
|
Springer JM, Kalot MA, Husainat NM, Byram KW, Dua AB, James KE, Chang Lin Y, Turgunbaev M, Villa-Forte A, Abril A, Langford CA, Maz M, Chung SA, Mustafa RA. Eosinophilic Granulomatosis with Polyangiitis: A Systematic Review and Meta-Analysis of Test Accuracy and Benefits and Harms of Common Treatments. ACR Open Rheumatol 2021; 3:101-110. [PMID: 33512787 PMCID: PMC7882521 DOI: 10.1002/acr2.11194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Objective Eosinophilic granulomatosis with polyangiitis (EGPA) is part of a group of vasculitides commonly referred to as antineutrophil cytoplasmic antibody (ANCA)–associated vasculitis (AAV), in addition to granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and renal‐limited vasculitis. Patients with EGPA characteristically have asthma and marked peripheral eosinophilia with only approximately 30% to 35% of patients being myeloperoxidase (MPO)‐ANCA positive, distinguishing it from other forms of AAV (1,2). The aim of this systematic review is to support the development of the American College of Rheumatology/Vasculitis Foundation guideline for the management of EGPA. Methods A systematic review was conducted of the literature for seven forms of primary systemic vasculitis (GPA, MPA, EGPA, polyarteritis nodosa, Kawasaki disease, giant cell arteritis, and Takayasu arteritis). The search was done for articles in English using Ovid Medline, PubMed, Embase, and the Cochrane Library. Articles were screened for suitability in addressing population/patients, intervention, comparator, and outcomes (PICO) questions, with studies presenting the highest level of evidence given preference. Two independent reviewers conducted a title/abstract screen and full‐text review for each eligible study. Results The initial search, conducted in August 2019, included 13 800 articles, of which 2596 full‐text articles were reviewed. There were 190 articles (addressing 34 PICO questions) reporting on the diagnosis and management of EGPA. Conclusion This comprehensive systematic review synthesizes and evaluates the accuracy of commonly used tests for EGPA as well as benefits and toxicities of different treatment options.
Collapse
Affiliation(s)
| | | | | | | | - Anisha B Dua
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | | | | | | | - Mehrdad Maz
- University of Kansas Medical Center, Kansas City
| | | | | |
Collapse
|
52
|
Lafarge A, Joseph A, Pagnoux C, Puéchal X, Cohen P, Samson M, Hamidou M, Karras A, Quemeneur T, Ribi C, Groh M, Mouthon L, Guillevin L, Terrier B. Predictive factors of severe infections in patients with systemic necrotizing vasculitides: data from 733 patients enrolled in five randomized controlled trials of the French Vasculitis Study Group. Rheumatology (Oxford) 2021; 59:2250-2257. [PMID: 31782786 DOI: 10.1093/rheumatology/kez575] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 10/29/2019] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Infections remain a major cause of morbidity and mortality in systemic necrotizing vasculitides (SNV). We aimed to identify factors predicting severe infections (SI) in SNV. METHODS Data from five randomized controlled trials (RCTs) enrolling 733 patients were pooled. The primary end point was the occurrence of SI, defined by the need of a hospitalization and/or intravenous anti-infectious treatment and/or leading to death. RESULTS After a median follow-up of 5.2 (interquartile range 3-9.7) years, 148 (20.2%) patients experienced 189 SI, and 98 (66.2%) presented their first SI within the first 2 years. Median interval from inclusion to SI was 14.9 (4.3-51.7) months. Age ≥65 years (hazard ratio (HR) 1.49 [1.07-2.07]; P=0.019), pulmonary involvement (HR 1.82 [1.26-2.62]; P=0.001) and Five Factor Score ≥1 (HR 1.21 [1.03-1.43]; P=0.019) were independent predictive factors of SI. Regarding induction therapy, the occurrence of SI was associated with the combination of GCs and CYC (HR 1.51 [1.03-2.22]; P = 0.036), while patients receiving only GCs were less likely to present SI (HR 0.69 [0.44-1.07]; P = 0.096). Finally, occurrence of SI had a significant negative impact on survival (P<0.001). CONCLUSION SI in SNV are frequent and impact mortality. Age, pulmonary involvement and Five Factor Score are baseline independent predictors of SI. No therapeutic regimen was significantly associated with SI but patients receiving glucocorticoids and CYC as induction tended to have more SI.
Collapse
Affiliation(s)
- Antoine Lafarge
- Department of Internal Medicine.,National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital
| | | | - Christian Pagnoux
- Department of Rheumatology, Mount Sinai Hospital, Toronto, ON, Canada
| | - Xavier Puéchal
- Department of Internal Medicine.,National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital
| | - Pascal Cohen
- Department of Internal Medicine.,National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital
| | - Maxime Samson
- Department of Internal Medicine and Clinical Immunology, Hôpital François Mitterrand, Dijon
| | | | - Alexandre Karras
- Department of Nephrology, Hôpital Européen Georges Pompidou, Paris
| | - Thomas Quemeneur
- Department of Internal Medicine, Hôpital de Valenciennes, Valenciennes, France
| | - Camillo Ribi
- Department of Immunology, CHUV, Lausanne, Switzerland
| | | | - Luc Mouthon
- Department of Internal Medicine.,National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital.,Paris Descartes University, Paris 5, Paris, France
| | - Loïc Guillevin
- Department of Internal Medicine.,National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital.,Paris Descartes University, Paris 5, Paris, France
| | - Benjamin Terrier
- Department of Internal Medicine.,National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital.,Paris Descartes University, Paris 5, Paris, France
| | | |
Collapse
|
53
|
Terrier B, Darbon R, Durel CA, Hachulla E, Karras A, Maillard H, Papo T, Puechal X, Pugnet G, Quemeneur T, Samson M, Taille C, Guillevin L. French recommendations for the management of systemic necrotizing vasculitides (polyarteritis nodosa and ANCA-associated vasculitides). Orphanet J Rare Dis 2020; 15:351. [PMID: 33372616 PMCID: PMC7771069 DOI: 10.1186/s13023-020-01621-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Systemic necrotizing vasculitis comprises a group of diseases resembling polyarteritis nodosa and anti-neutrophil cytoplasmic antibody-associated vasculitis (ANCA): granulomatosis with polyangiitis, eosinophilic granulomatosis with polyangiitis, and microscopic polyangiitis. The definitive diagnosis is made in cooperation with a reference center for autoimmune diseases and rare systemic diseases or a competency center. The management goals are: to obtain remission and, in the long term, healing; to reduce the risk of relapses; to limit and reduce the sequelae linked to the disease; to limit the side effects and the sequelae linked to the treatments; to improve or at least maintain the best possible quality of life; and to maintain socio-professional integration and/or allow a rapid return to school and/or professional activity. Information and therapeutic education of the patients and those around them are an integral part of the care. All health professionals and patients should be informed of the existence of patient associations. The treatment of vasculitis is based on variable combinations of glucocorticoids and immunosuppressants, chosen and adapted according to the disease concerned, the severity and/or extent of the disease, and the underlying factors (age, kidney function, etc.). Follow-up clinical and paraclinical examinations must be carried out regularly to clarify the progression of the disease, detect and manage treatment failures and possible relapses early on, and limit sequelae and complications (early then late) related to the disease or treatment. A distinction is made between the induction therapy, lasting approximately 3–6 months and aimed at putting the disease into remission, and the maintenance treatment, lasting 12–48 months, or even longer. The role of the increase or testing positive again for ANCA as a predictor of a relapse, which has long been controversial, now seems to have greater consensus: Anti-myeloperoxidase ANCAs are less often associated with a relapse of vasculitis than anti-PR3 ANCA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas Papo
- Internal Medicine, CHU Bichat, AP-HP, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
54
|
Monti S, Quinn KA, Christensen R, Jayne D, Langford C, Lanier GE, Mahr A, Pagnoux C, Viðarsdóttir MB, Merkel PA, Tomasson G. Use and reporting of outcome measures in randomized trials for anti-neutrophil cytoplasmic antibody-associated vasculitis: a systematic literature review. Semin Arthritis Rheum 2020; 50:1314-1325. [DOI: 10.1016/j.semarthrit.2020.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 11/28/2022]
|
55
|
Rosenberg CE, Khoury P. Approach to Eosinophilia Presenting With Pulmonary Symptoms. Chest 2020; 159:507-516. [PMID: 33002503 DOI: 10.1016/j.chest.2020.09.247] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/14/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022] Open
Abstract
Eosinophilia with pulmonary involvement is characterized by the presence of peripheral blood eosinophilia, typically ≥500 cells/mm3, by pulmonary symptoms and physical examination findings that are nonspecific, and by radiographic evidence of pulmonary disease and is further supported by histopathologic evidence of tissue eosinophilia in a lung or pleura biopsy specimen and/or increased eosinophils in BAL fluid, usually >10%. Considering that there are a variety of underlying causes of eosinophilia with pulmonary manifestations and overlapping clinical, laboratory, and radiologic features, it is essential to approach the evaluation of eosinophilia with pulmonary findings systematically. In this review, we will describe a case presentation and discuss the differential diagnosis, a directed approach to the diagnostic evaluation and supporting literature, the current treatment strategies for pulmonary eosinophilia syndromes, and the levels of evidence underlying the recommendations, where available. Overall, optimal management of eosinophilic lung disease presentations are directed at the underlying cause when identifiable, and the urgency of treatment may be guided by the presence of severe end-organ involvement or life-threatening complications. When an underlying cause is not easily attributable, management of eosinophilia with pulmonary involvement largely relies on eosinophil-directed interventions, for which biologic therapies are increasingly being used.
Collapse
Affiliation(s)
- Chen E Rosenberg
- Department of Pediatrics, Division of Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Paneez Khoury
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
56
|
Mendel A, Ennis D, Go E, Bakowsky V, Baldwin C, Benseler SM, Cabral DA, Carette S, Clements-Baker M, Clifford AH, Cohen Tervaert JW, Cox G, Dehghan N, Dipchand C, Dhindsa N, Famorca L, Fifi-Mah A, Garner S, Girard LP, Lessard C, Liang P, Noone D, Makhzoum JP, Milman N, Pineau CA, Reich HN, Rhéaume M, Robinson DB, Rumsey DG, Towheed TE, Trudeau J, Twilt M, Yacyshyn E, Yeung RSM, Barra LB, Khalidi N, Pagnoux C. CanVasc Consensus Recommendations for the Management of Antineutrophil Cytoplasm Antibody-associated Vasculitis: 2020 Update. J Rheumatol 2020; 48:555-566. [PMID: 32934123 DOI: 10.3899/jrheum.200721] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE In 2015, the Canadian Vasculitis Research Network (CanVasc) created recommendations for the management of antineutrophil cytoplasm antibody (ANCA)-associated vasculitides (AAV) in Canada. The current update aims to revise existing recommendations and create additional recommendations, as needed, based on a review of new available evidence. METHODS A needs assessment survey of CanVasc members informed questions for an updated systematic literature review (publications spanning May 2014 to September 2019) using Medline, Embase, and Cochrane. New and revised recommendations were developed and categorized according to the level of evidence and strength of each recommendation. The CanVasc working group used a 2-step modified Delphi procedure to reach > 80% consensus on the inclusion, wording, and grading of each new and revised recommendation. RESULTS Eleven new and 16 revised recommendations were created and 12 original (2015) recommendations were retained. New and revised recommendations are discussed in detail within this document. Five original recommendations were removed, of which 4 were incorporated into the explanatory text. The supplementary material for practical use was revised to reflect the updated recommendations. CONCLUSION The 2020 updated recommendations provide rheumatologists, nephrologists, and other specialists caring for patients with AAV in Canada with new management guidance, based on current evidence and consensus from Canadian experts.
Collapse
Affiliation(s)
- Arielle Mendel
- A. Mendel, MD, MSc, C.A. Pineau, MD, Division of Rheumatology, Lupus and Vasculitis Clinic, McGill University, Montréal, Québec;
| | - Daniel Ennis
- D. Ennis, MD, C. Baldwin, MD, N. Dehghan, MD, N. Dhindsa, MD, Division of Rheumatology, University of British Columbia, Vancouver, British Columbia
| | - Ellen Go
- E. Go, MD, R.S. Yeung, MD, PhD, Division of Rheumatology, Hospital for Sick Children, University of Toronto, Toronto, Ontario
| | - Volodko Bakowsky
- V. Bakowsky, MD, Division of Rheumatology, QEII Health Sciences Centre, Dalhousie University, Halifax, Nova Scotia
| | - Corisande Baldwin
- D. Ennis, MD, C. Baldwin, MD, N. Dehghan, MD, N. Dhindsa, MD, Division of Rheumatology, University of British Columbia, Vancouver, British Columbia
| | - Susanne M Benseler
- S.M. Benseler, MD, PhD, M. Twilt, MD, PhD, Division of Rheumatology, Department of Pediatrics, Alberta Children's Hospital, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta
| | - David A Cabral
- D.A. Cabral, MBBS, Division of Pediatric Rheumatology, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia
| | - Simon Carette
- S. Carette, MD, MPhil, C. Pagnoux, MD, MSc, MPH, Division of Rheumatology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario
| | - Marie Clements-Baker
- M. Clements-Baker, MD, T.E. Towheed, MD, MS, Division of Rheumatology, Queen's University, Kingston, Ontario
| | - Alison H Clifford
- A.H. Clifford, MD, J.W. Cohen Tervaert, MD, PhD, E. Yacyshyn, MD, Division of Rheumatology, University of Alberta, Edmonton, Alberta
| | - Jan Willem Cohen Tervaert
- A.H. Clifford, MD, J.W. Cohen Tervaert, MD, PhD, E. Yacyshyn, MD, Division of Rheumatology, University of Alberta, Edmonton, Alberta
| | - Gerard Cox
- G. Cox, MB, Firestone Institute for Respiratory Health, McMaster University, Hamilton, Ontario
| | - Natasha Dehghan
- D. Ennis, MD, C. Baldwin, MD, N. Dehghan, MD, N. Dhindsa, MD, Division of Rheumatology, University of British Columbia, Vancouver, British Columbia
| | - Christine Dipchand
- C. Dipchand, MD, MSc, Division of Nephrology, QEII Health Sciences Centre, Dalhousie University, Halifax, Nova Scotia
| | - Navjot Dhindsa
- D. Ennis, MD, C. Baldwin, MD, N. Dehghan, MD, N. Dhindsa, MD, Division of Rheumatology, University of British Columbia, Vancouver, British Columbia
| | - Leilani Famorca
- L. Famorca, MD, S. Garner, MD, MSc, N. Khalidi, MD, Division of Rheumatology, McMaster University, Hamilton, Ontario
| | - Aurore Fifi-Mah
- A. Fifi-Mah, MD, Division of Rheumatology, University of Calgary, Calgary, Alberta
| | - Stephanie Garner
- L. Famorca, MD, S. Garner, MD, MSc, N. Khalidi, MD, Division of Rheumatology, McMaster University, Hamilton, Ontario
| | - Louis-Philippe Girard
- L.P. Girard, MD, MSc, Division of Nephrology, University of Calgary, Calgary, Alberta
| | - Clode Lessard
- C. Lessard, MD, Centre de Recherche Musculo-Squelettique, Trois-Rivières, Québec
| | - Patrick Liang
- P. Liang, MD, Division of Rheumatology, Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec
| | - Damien Noone
- D. Noone, MB, BCh, BAO, MSc, Division of Nephrology, Hospital for Sick Children, University of Toronto, Toronto, Ontario
| | - Jean-Paul Makhzoum
- J.P. Makhzoum, MD, M. Rhéaume, MD, Division of Internal Medicine, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, Québec
| | - Nataliya Milman
- N. Milman, MD, MSc, Division of Rheumatology, The Ottawa Hospital, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario
| | - Christian A Pineau
- A. Mendel, MD, MSc, C.A. Pineau, MD, Division of Rheumatology, Lupus and Vasculitis Clinic, McGill University, Montréal, Québec
| | - Heather N Reich
- H.N. Reich, MD, PhD, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario
| | - Maxime Rhéaume
- J.P. Makhzoum, MD, M. Rhéaume, MD, Division of Internal Medicine, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, Québec
| | - David B Robinson
- D.B. Robinson, MD, MSc, Section of Rheumatology, University of Manitoba, Winnipeg, Manitoba
| | - Dax G Rumsey
- D.G. Rumsey, MD, MSc, Division of Pediatric Rheumatology, University of Alberta, Edmonton, Alberta
| | - Tanveer E Towheed
- M. Clements-Baker, MD, T.E. Towheed, MD, MS, Division of Rheumatology, Queen's University, Kingston, Ontario
| | - Judith Trudeau
- J. Trudeau, MD, Division of Rheumatology, CISSS Chaudière-Appalaches, Université Laval, Québec City, Québec
| | - Marinka Twilt
- S.M. Benseler, MD, PhD, M. Twilt, MD, PhD, Division of Rheumatology, Department of Pediatrics, Alberta Children's Hospital, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta
| | - Elaine Yacyshyn
- A.H. Clifford, MD, J.W. Cohen Tervaert, MD, PhD, E. Yacyshyn, MD, Division of Rheumatology, University of Alberta, Edmonton, Alberta
| | - Rae S M Yeung
- E. Go, MD, R.S. Yeung, MD, PhD, Division of Rheumatology, Hospital for Sick Children, University of Toronto, Toronto, Ontario
| | - Lillian B Barra
- L.B. Barra, MD, Division of Rheumatology, Department of Medicine, Western University, London, Ontario, Canada
| | - Nader Khalidi
- L. Famorca, MD, S. Garner, MD, MSc, N. Khalidi, MD, Division of Rheumatology, McMaster University, Hamilton, Ontario
| | - Christian Pagnoux
- S. Carette, MD, MPhil, C. Pagnoux, MD, MSc, MPH, Division of Rheumatology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario
| |
Collapse
|
57
|
Kitching AR, Anders HJ, Basu N, Brouwer E, Gordon J, Jayne DR, Kullman J, Lyons PA, Merkel PA, Savage COS, Specks U, Kain R. ANCA-associated vasculitis. Nat Rev Dis Primers 2020; 6:71. [PMID: 32855422 DOI: 10.1038/s41572-020-0204-y] [Citation(s) in RCA: 455] [Impact Index Per Article: 113.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
The anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs) are a group of disorders involving severe, systemic, small-vessel vasculitis and are characterized by the development of autoantibodies to the neutrophil proteins leukocyte proteinase 3 (PR3-ANCA) or myeloperoxidase (MPO-ANCA). The three AAV subgroups, namely granulomatosis with polyangiitis (GPA), microscopic polyangiitis and eosinophilic GPA (EGPA), are defined according to clinical features. However, genetic and other clinical findings suggest that these clinical syndromes may be better classified as PR3-positive AAV (PR3-AAV), MPO-positive AAV (MPO-AAV) and, for EGPA, by the presence or absence of ANCA (ANCA+ or ANCA-, respectively). Although any tissue can be involved in AAV, the upper and lower respiratory tract and kidneys are most commonly and severely affected. AAVs have a complex and unique pathogenesis, with evidence for a loss of tolerance to neutrophil proteins, which leads to ANCA-mediated neutrophil activation, recruitment and injury, with effector T cells also involved. Without therapy, prognosis is poor but treatments, typically immunosuppressants, have improved survival, albeit with considerable morbidity from glucocorticoids and other immunosuppressive medications. Current challenges include improving the measures of disease activity and risk of relapse, uncertainty about optimal therapy duration and a need for targeted therapies with fewer adverse effects. Meeting these challenges requires a more detailed knowledge of the fundamental biology of AAV as well as cooperative international research and clinical trials with meaningful input from patients.
Collapse
Affiliation(s)
- A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia. .,Departments of Nephrology and Paediatric Nephrology, Monash Health, Clayton, Victoria, Australia.
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ludwig-Maximilians University, Munich, Germany
| | - Neil Basu
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Elisabeth Brouwer
- Vasculitis Expertise Centre Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Jennifer Gordon
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, PA, USA
| | - David R Jayne
- Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Paul A Lyons
- Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Peter A Merkel
- Division of Rheumatology, Department of Medicine and Division of Clinical Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline O S Savage
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Renate Kain
- Department of Pathology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
58
|
Guillevin L. Treatment of systemic necrotizing vasculitides: The 40-year experience of the French Vasculitis Study Group. Presse Med 2020; 49:104034. [PMID: 32650043 DOI: 10.1016/j.lpm.2020.104034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 05/19/2020] [Accepted: 07/02/2020] [Indexed: 10/23/2022] Open
Abstract
Treatment of vasculitides has benefited from the results of several prospective clinical trials focusing on the evaluation of new drugs, therapeutic strategies and adjuvant treatments. In the field of autoimmunity, vasculitides are the group of diseases for which the most important medical progress has been made, combining advances in understanding the pathogenetic mechanisms, classification of the various entities and willingness to evaluate treatments. Several international groups have been actively involved in these tasks. The French Vasculitis Study Group was the first to design and organize prospective trials in the field and to contribute to these medical advances. In this review, we analyze the different treatments and therapeutic strategies evaluated over the last few decades and, more precisely, the last 39 years by the French Vasculitis Study Group.
Collapse
Affiliation(s)
- Loïc Guillevin
- Department of Internal Medicine, Center for Rare Systemic and Autoimmune Diseases, Hôpital Cochin, 27, rue du Faubourg-Saint-Jacques, Paris, France.
| |
Collapse
|
59
|
ANCA-associated vasculitides: Recommendations of the French Vasculitis Study Group on the use of immunosuppressants and biotherapies for remission induction and maintenance. Presse Med 2020; 49:104031. [PMID: 32645418 DOI: 10.1016/j.lpm.2020.104031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
Treatment of vasculitides associated with anti-neutrophil cytoplasm antibodies (ANCA) (AAVs) has evolved dramatically in recent years, particularly since the demonstration of rituximab efficacy as remission induction and maintenance therapy for granulomatosis with polyangiitis and microscopic polyangiitis. In 2013, the French Vasculitis Study Group (FVSG) published recommendations for its use by clinicians. Since then, new data have made it possible to better specify and codify prescription of rituximab to treat AAVs. Herein, the FVSG Recommendations Committee, an expert panel comprised of physicians with extensive experience in the treatment and management of vasculitides, presents its consensus guidelines based on literature analysis, the results of prospective therapeutic trials and personal experience.
Collapse
|
60
|
Raffray L, Guillevin L. Updates for the treatment of EGPA. Presse Med 2020; 49:104036. [PMID: 32652104 DOI: 10.1016/j.lpm.2020.104036] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 05/19/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA, formerly Churg-Strauss syndrome) is the least frequent antineutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV). Major advances of our knowledge on its pathophysiology have revealed features of both AAV and eosinophilic disorders. The development of targeted biotherapies for both diseases opened new possibilities for EGPA management. In this review, we highlight the rationale underlying the routine treatment strategy, which relies mainly on corticosteroids, with immunosuppressant adjunction for severe disease. However, novel therapies are still needed for refractory/relapsing disease and to alleviate the corticosteroid-dependence of asthma and chronic rhinosinusitis. At present, the most promising biotherapies target either eosinophil biology, like mepolizumab, an anti-interleukin-5, or the B-cell compartment, with rituximab. Recent clinical data on new treatment options are discussed and therapeutic strategies are proposed.
Collapse
Affiliation(s)
- Loïc Raffray
- Department of Internal Medicine, Félix-Guyon University Hospital of La Réunion, CS11021, Saint Denis, Reunion
| | - Loïc Guillevin
- Referral Center for Rare Systemic and Autoimmune Diseases, Department of Internal Medicine, Hôpital Cochin, Université Paris Descartes, 27, rue du Faubourg-Saint-Jacques, 75679 Paris Cedex 14, France.
| |
Collapse
|
61
|
Abstract
INTRODUCTION Rituximab, an anti-B-cell biological therapy, has been investigated in several clinical trials on antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs). AREAS COVERED In this paper, the clinical trials and open-label studies on rituximab efficacy and safety in treating AAVs are reviewed. EXPERT OPINION Rituximab achieved high remission-induction and sustained-maintenance rates for patients with these severe diseases, thereby challenging the cornerstone treatment of corticosteroids and cyclophosphamide followed by azathioprine. Rituximab should be used as first-line therapy with corticosteroids to induce remission of severe AAVs, especially in situations in which cyclophosphamide may be problematic (relapse after cyclophosphamide, women of childbearing age, risk of malignancy). Cyclophosphamide indications are likely to be restricted in the future. Whenever possible, rituximab should be preferred to azathioprine to maintain remission. The current maintenance regimen has been extended to at least 18 months but its optimal duration remains unknown and recent data suggest the possibility to extend treatment to 4 years. Future challenges include defining the best dose regimen: at present, different schedules are used as alternatives to those recognized as standards by health authorities. In addition, it remains to identify which patients will benefit the most from long-term retreatment: potentially those with relapsing disease or anti-proteinase-3 ANCA-positivity.
Collapse
Affiliation(s)
- Loïc Raffray
- Department of Internal Medicine, Félix-Guyon University Hospital of La Réunion , Saint-Denis, France
| | - Loïc Guillevin
- Referral Center for Rare Systemic and Autoimmune Diseases, Department of Internal Medicine, Hôpital Cochin, Université Paris Descartes , Paris, France
| |
Collapse
|
62
|
Nguyen Y, Pagnoux C, Karras A, Quéméneur T, Maurier F, Hamidou M, Le Quellec A, Chiche NJ, Cohen P, Régent A, Lifermann F, Mékinian A, Khouatra C, Hachulla E, Pourrat J, Ruivard M, Godmer P, Viallard JF, Terrier B, Mouthon L, Guillevin L, Puéchal X. Microscopic polyangiitis: Clinical characteristics and long-term outcomes of 378 patients from the French Vasculitis Study Group Registry. J Autoimmun 2020; 112:102467. [PMID: 32340774 DOI: 10.1016/j.jaut.2020.102467] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To describe characteristics and long-term outcomes of patients with microscopic polyangiitis (MPA), an antineutrophil cytoplasm antibody (ANCA)-associated small-vessel necrotizing vasculitis. METHODS MPA patients from the French Vasculitis Study Group Registry satisfying the European Medicines Agency algorithm were analyzed retrospectively. Characteristics at diagnosis, treatments, relapses and deaths were analyzed to identify factors predictive of death or relapse. RESULTS Between 1966 and 2017, 378 MPA patients (median age 63.7 years) were diagnosed and followed for a mean of 5.5 years. At diagnosis, the main clinical manifestations included renal involvement (74%), arthralgias (45%), skin (41%), lung (40%) and mononeuritis multiplex (32%), with less frequent alveolar hemorrhage (16%), cardiomyopathy (5%) and severe gastrointestinal signs (4%); mean serum creatinine was 217 μmol/L. ANCA were detected in 298/347 (86%) patients by immunofluorescence and/or enzyme-linked immunosorbent assay (ELISA). Among the 293 patients with available ELISA specificities, 272 (92.8%) recognized myeloperoxidase and 13 (4.4%) proteinase-3. During follow-up, 131 (34.7%) patients relapsed and 78 (20.6%) died, mainly from infections. Respective 5-year overall and relapse-free survival rates were 84.2% and 60.4%. Multivariable analyses retained age >65 years, creatinine >130 μmol/L, severe gastrointestinal involvement and mononeuritis multiplex as independent risk factors for death. Renal impairment was associated with a lower risk of relapse. CONCLUSION Non-renal manifestations and several risk factors for death or relapse were frequent in this nationwide cohort. While mortality was low, and mainly due to treatment-related complications, relapses remained frequent, suggesting that MPA management can be further improved.
Collapse
Affiliation(s)
- Yann Nguyen
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | | | - Alexandre Karras
- Department of Nephrology, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | | | | | | | - Alain Le Quellec
- Department of Internal Medicine, Hôpital Saint-Eloi, CHU, Montpellier, France
| | | | - Pascal Cohen
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Alexis Régent
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | | | - Arsène Mékinian
- Department of Internal Medicine, Hôpital Saint-Antoine, APHP, Paris, France
| | - Chahéra Khouatra
- Department of Respiratory Medicine, National Referral Center for Rare Pulmonary Diseases, Hôpital Louis-Pradel, CHU Lyon, France
| | - Eric Hachulla
- Department of Internal Medicine, National Referral Center for Systemic Sclerosis, CHRU Claude Huriez, Lille, France
| | | | - Marc Ruivard
- Department of Internal Medicine, CHU Estaing, Clermont-Ferrand, France
| | | | | | - Benjamin Terrier
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Luc Mouthon
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Loïc Guillevin
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Xavier Puéchal
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France.
| | | |
Collapse
|
63
|
Puéchal X, Pagnoux C, Baron G, Lifermann F, Geffray L, Quémeneur T, Saraux JL, Wislez M, Cottin V, Ruivard M, Limal N, Aouba A, Bonnotte B, Néel A, Agard C, Cohen P, Terrier B, Le Jeunne C, Mouthon L, Ravaud P, Guillevin L. Non-severe eosinophilic granulomatosis with polyangiitis: long-term outcomes after remission-induction trial. Rheumatology (Oxford) 2020; 58:2107-2116. [PMID: 31056661 DOI: 10.1093/rheumatology/kez139] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/16/2019] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE In a previous controlled trial, 1-year adjunction of AZA to glucocorticoids (GC) for patients with non-severe, newly diagnosed eosinophilic granulomatosis with polyangiitis (EGPA) failed to lower remission failure, vasculitis relapse and isolated asthma/rhinosinus exacerbation rates, or cumulative GC use at month (M) 24. The aim of this study was to analyse longer-term outcomes to determine whether subsequent vasculitis relapse or isolated asthma/rhinosinus exacerbation (IARE) rates differed. METHODS After M24, patients were followed prospectively, being treated based on physicians' best judgment. Flares and reasons for increased GC dose or immunosuppressant use were recorded, and reviewed according to randomization group to distinguish vasculitis relapses from IAREs according to EGPA Task Force recommendations. RESULTS Fifty EGPA trial participants were followed for a median (interquartile range) of 6.3 (5.4-7.6) years; two (4%) died 11 months post-inclusion. By M24, vasculitis had relapsed in 21/49 (43%) patients and 14/50 (28%) had IAREs. Another patient died 4.8 years post-inclusion (infection). Among nine patients with subsequent vasculitis relapses, three had a major relapse and three had their first relapse after M24; among 25 patients with later IAREs, 17 occurred after M24. At 5 years, respective vasculitis relapse and IARE rates were 48% (95% CI 34.0, 62.6) and 56% (95% CI 41.7, 70.8), with no between-arm differences (P = 0.32 and 0.13). No entry clinical or biological parameter was associated with these outcomes during follow-up. CONCLUSION These results confirmed that 1-year AZA and GC induction obtained good overall survival but no long-term benefit for non-severe EGPA patients. Vasculitis relapses, occurring mostly during the first 2 years, and IAREs, occurring throughout follow-up, require other preventive treatments. TRIAL REGISTRATION ClinicalTrials.gov, https://clinicaltrials.gov, NCT00647166.
Collapse
Affiliation(s)
- Xavier Puéchal
- National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Christian Pagnoux
- National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France.,Vasculitis Clinic, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Gabriel Baron
- Université Paris Descartes, Hôtel-Dieu, APHP, Paris, France
| | | | | | | | | | - Marie Wislez
- Hôpital Tenon, APHP, Université Pierre et Marie Curie, Paris, France
| | - Vincent Cottin
- Hôpital Louis-Pradel, Lyon and UMR754, Université Claude Bernard Lyon 1, Lyon, France
| | - Marc Ruivard
- Centre Hospitalier Universitaire Estaing, Clermont-Ferrand, France
| | - Nicolas Limal
- Hôpital Henri-Mondor, Université Paris-Est Créteil, APHP, Créteil, France
| | - Achille Aouba
- Centre Hospitalier Universitaire Côte de Nacre, Caen, France
| | | | - Antoine Néel
- Centre Hospitalier Universitaire Hôtel-Dieu, Nantes, France
| | | | - Pascal Cohen
- National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Benjamin Terrier
- National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Claire Le Jeunne
- National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | - Luc Mouthon
- National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | | | - Loïc Guillevin
- National Referral Center for Rare Systemic Autoimmune Diseases, Hôpital Cochin, APHP, Université Paris Descartes, Paris, France
| | | |
Collapse
|
64
|
Abstract
Vasculitis is characterized by inflammation of the vascular wall. It reaches vessels of different sizes and locations, conditioning multisystem and complex manifestations that require a holistic approach. Antineutrophil cytoplasmic antibody-associated vasculitis has an annual incidence rate of 20 per million inhabitants. It is the result of the interaction of infectious, genetic and environmental factors and manifest itself with varied and unspecific symptoms, often reaching the general state, respiratory and renal systems. The combination of clinical, laboratory, imaging and fundamentally histological changes allows the diagnosis and classification of the extent / severity of the disease necessary for the decision of the therapeutics to be taken. Glucocorticoids are generally used; with methotrexate being considered when localized disease and cyclophosphamide, rituximab or azathioprine when generalized disease. A systematized approach can increase survival rates from 12% to 70% over a 5-year span.
Collapse
Affiliation(s)
- Fernando Salvador
- Autoimmune Diseases Unit, Internal Medicine Department, Centro Hospitalar de Trás-os-Montes e Alto Douro, Avenida da Noruega, 5000-508 Vila Real, Portugal; NEDAI/SPMI Autoimmune Diseases Group/Portuguese Society of Internal Medicine, Portugal.
| |
Collapse
|
65
|
Berti A, Cornec D, Casal Moura M, Smyth RJ, Dagna L, Specks U, Keogh KA. Eosinophilic Granulomatosis With Polyangiitis: Clinical Predictors of Long-term Asthma Severity. Chest 2020; 157:1086-1099. [PMID: 31958440 DOI: 10.1016/j.chest.2019.11.045] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/14/2019] [Accepted: 11/30/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The long-term clinical course of asthma in patients with eosinophilic granulomatosis with polyangiitis (EGPA) remains unclear. We aimed to characterize long-term asthma in EGPA and to identify baseline predictors of long-term asthma severity. METHODS This retrospective cohort study included patients who fulfilled standardized criteria for EGPA who were followed up in a single referral center between 1990 and 2017. Baseline and 3 (± 1) years of follow-up clinical, laboratory, and pulmonary function data were analyzed. RESULTS Eighty-nine patients with EGPA and a documented asthma assessment at baseline and at 3 years from diagnosis were included. Severe/uncontrolled asthma was observed in 42.7% of patients at diagnosis and was associated with previous history of respiratory allergy (P < .01), elevated serum total IgE levels (P < .05), and increased use of high-dose inhaled corticosteroids (ICSs; P < .05) and oral corticosteroids (OCSs; P < .001) for respiratory symptoms the year before the EGPA diagnosis. During follow-up, an improvement or worsening in asthma severity was noted in 12.3% and 10.1% of patients, respectively. Severe/uncontrolled asthma was present in 40.5% of patients at 3 years and was associated with increased airway resistance on pulmonary function tests (PFTs; P < .05). Long-term PFTs did not improve during long-term follow-up regardless of ICS or OCS therapy. Multivariate binary logistic regression results indicated that severe rhinosinusitis (P = .038), pulmonary infiltrates (P = .011), overweight (BMI ≥ 25 kg/m2; P = .041), and severe/uncontrolled asthma at vasculitis diagnosis (P < .001) independently predicted severe/uncontrolled asthma at the 3-year end point. CONCLUSIONS In patients with asthma with EGPA, long-term severe/uncontrolled asthma is associated with baseline pulmonary and ear, nose, and throat manifestations but not with clear-cut vasculitic features.
Collapse
Affiliation(s)
- Alvise Berti
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN; Rheumatology Department, Santa Chiara Hospital, and Center for Integrative Biology, University of Trento, Trento, Italy
| | - Divi Cornec
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN; INSERM UMR1227, Lymphocytes B et Autoimmunité, Université de Bretagne Occidentale, Centre Hospitalier Universitaire de Brest, Brest, France
| | - Marta Casal Moura
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Robert J Smyth
- Department of Pulmonary Medicine, Boston Medical Center, Boston, MA
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Karina A Keogh
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN.
| |
Collapse
|
66
|
Berti A, Boukhlal S, Groh M, Cornec D. Eosinophilic granulomatosis with polyangiitis: the multifaceted spectrum of clinical manifestations at different stages of the disease. Expert Rev Clin Immunol 2020; 16:51-61. [PMID: 31762336 DOI: 10.1080/1744666x.2019.1697678] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Introduction: Eosinophilic granulomatosis with polyangiitis (EGPA) usually occurs in patients with late-onset asthma and sustained peripheral blood eosinophilia and classically presents with a clinical multifaceted spectrum of manifestations, which may vary at the different stages of the natural history of the disease.Areas covered: We reviewed EGPA clinical presentation, focusing on clinical manifestations at three different phases of the disease: 1/before the development of overt vasculitis, 2/at vasculitis diagnosis and 3/during the long-term follow-up. An update on current classification criteria and recent therapeutic advances has been provided as well.Expert opinion: Asthma, chronic rhinosinusitis and blood eosinophilia could anticipate the overt vasculitis for years. An atopic background may be present in a subset of patients (25-30%), while ANCA presence varies between 10 and 40%. Systemic vasculitis rapidly occurs and clinical features demonstrating vasculitis processes (neuropathy, purpura, scleritis, alveolar hemorrhage and glomerulonephritis) develop along with systemic symptoms (50%). After vasculitis resolution, asthma remains severe in up to 50% of patients and incidence of isolated-asthma and rhinosinus exacerbations remains constantly high. Different sets of classification criteria have been published so far, and DCVAS diagnostic criteria will be presented soon. Interleukin-5 blockers seem to be promising to control the disease and to spare corticosteroids.
Collapse
Affiliation(s)
- Alvise Berti
- Department of Rheumatology, Santa Chiara Hospital, Trento, Italy.,Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Sara Boukhlal
- INSERM UMR1227, Lymphocytes B et Autoimmunité, Université de Bretagne Occidentale, CHU de Brest, Brest, France
| | - Matthieu Groh
- Department of Internal Medicine, Hôpital Foch, National Referral Center for Hypereosinophilic Syndrome (CEREO), Suresnes, France
| | - Divi Cornec
- INSERM UMR1227, Lymphocytes B et Autoimmunité, Université de Bretagne Occidentale, CHU de Brest, Brest, France
| |
Collapse
|
67
|
Abstract
BACKGROUND Renal vasculitis presents as rapidly progressive glomerulonephritis and comprises of a group of conditions characterised by acute kidney injury (AKI), haematuria and proteinuria. Treatment of these conditions involve the use of steroid and non-steroid agents in combination with plasma exchange. Although immunosuppression overall has been very successful in treatment of these conditions, many questions remain unanswered in terms of dose and duration of therapy, the use of plasma exchange and the role of new therapies. This 2019 publication is an update of a review first published in 2008 and updated in 2015. OBJECTIVES To evaluate the benefits and harms of any intervention used for the treatment of renal vasculitis in adults. SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies up to 21 November 2019 through contact with the Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Register (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA Randomised controlled trials investigating any intervention for the treatment of renal vasculitis in adults. DATA COLLECTION AND ANALYSIS Two authors independently assessed study quality and extracted data. Statistical analyses were performed using a random effects model and results expressed as risk ratio (RR) with 95% confidence intervals (CI) for dichotomous outcomes or mean difference (MD) for continuous outcomes. MAIN RESULTS Forty studies (3764 patients) were included. Studies conducted earlier tended to have a higher risk of bias due to poor (or poorly reported) study design, broad inclusion criteria, less well developed disease definitions and low patient numbers. Later studies tend to have improved in all areas of quality, aided by the development of large international study groups. Induction therapy: Plasma exchange as adjunctive therapy may reduce the need for dialysis at three (2 studies: RR 0.43, 95% CI 0.23 to 0.78; I2 = 0%) and 12 months (6 studies: RR 0.45, 95% CI 0.29 to 0.72; I2 = 0%) (low certainty evidence). Plasma exchange may make little or no difference to death, serum creatinine (SCr), sustained remission or to serious or the total number of adverse events. Plasma exchange may increase the number of serious infections (5 studies: RR 1.26, 95% CI 1.03 to 1.54; I2 = 0%; low certainty evidence). Remission rates for pulse versus continuous cyclophosphamide (CPA) were equivalent but pulse treatment may increase the risk of relapse (4 studies: RR 1.79, 95% CI 1.11 to 2.87; I2 = 0%) (low certainty evidence) compared with continuous cyclophosphamide. Pulse CPA may make little or no difference to death at final follow-up, or SCr at any time point. More patients required dialysis in the pulse CPA group. Leukopenia was less common with pulse treatment; however, nausea was more common. Rituximab compared to CPA probably makes little or no difference to death, remission, relapse, severe adverse events, serious infections, or severe adverse events. Kidney function and dialysis were not reported. A single study reported no difference in the number of deaths, need for dialysis, or adverse events between mycophenolate mofetil (MMF) and CPA. Remission was reported to improve with MMF however more patients relapsed. A lower dose of steroids was probably as effective as high dose and may be safer, causing fewer infections; kidney function and relapse were not reported. There was little of no difference in death or remission between six and 12 pulses of CPA. There is low certainty evidence that there were less relapses with 12 pulses (2 studies: RR 1.57, 95% CI 0.96 to 2.56; I2 = 0%), but more infections (2 studies: RR 0.79, 95% CI 0.36 to 1.72; I2 = 45%). One study reported severe adverse events were less in patients receiving six compared to 12 pulses of CPA. Kidney function and dialysis were not reported. There is limited evidence from single studies about the effectiveness of intravenous immunoglobulin, avacopan, methotrexate, immunoadsorption, lymphocytapheresis, or etanercept. Maintenance therapy: Azathioprine (AZA) has equivalent efficacy as a maintenance agent to CPA with fewer episodes of leucopenia. MMF resulted in a higher relapse rate when tested against azathioprine in remission maintenance. Rituximab is an effective remission induction and maintenance agent. Oral co-trimoxazole did not reduce relapses in granulomatosis with polyangiitis. There were fewer relapses but more serious adverse events with leflunomide compared to methotrexate. There is limited evidence from single studies about the effectiveness of methotrexate versus CPA or AZA, cyclosporin versus CPA, extended versus standard AZA, and belimumab. AUTHORS' CONCLUSIONS Plasma exchange was effective in patients with severe AKI secondary to vasculitis. Pulse cyclophosphamide may result in an increased risk of relapse when compared to continuous oral use but a reduced total dose. Whilst CPA is standard induction treatment, rituximab and MMF were also effective. AZA, methotrexate and leflunomide were effective as maintenance therapy. Further studies are required to more clearly delineate the appropriate place of newer agents within an evidence-based therapeutic strategy.
Collapse
Affiliation(s)
- Giles D Walters
- The Canberra HospitalDepartment of Renal MedicineYamba DriveCanberraACTAustralia2605
| | - Narelle S Willis
- The University of SydneySydney School of Public HealthSydneyNSWAustralia2006
- The Children's Hospital at WestmeadCochrane Kidney and Transplant, Centre for Kidney ResearchLocked Bag 4001WestmeadNSWAustralia2145
| | - Tess E Cooper
- The Children's Hospital at WestmeadCochrane Kidney and Transplant, Centre for Kidney ResearchLocked Bag 4001WestmeadNSWAustralia2145
| | - Jonathan C Craig
- The Children's Hospital at WestmeadCochrane Kidney and Transplant, Centre for Kidney ResearchLocked Bag 4001WestmeadNSWAustralia2145
- Flinders UniversityCollege of Medicine and Public HealthAdelaideSAAustralia5001
| | | |
Collapse
|
68
|
|
69
|
Asthma control in eosinophilic granulomatosis with polyangiitis treated with rituximab. Clin Rheumatol 2020; 39:1581-1590. [PMID: 31897956 DOI: 10.1007/s10067-019-04891-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/03/2019] [Accepted: 12/11/2019] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Rituximab (RTX) treatment is used for antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis, but its benefits in eosinophilic granulomatosis with polyangiitis (EGPA) are unclear. Our aim was to characterize asthma control and glucocorticoid (GC) sparing after RTX treatment. METHODS A retrospective, computer-assisted search was performed to identify patients with EGPA and GC-dependent asthma diagnosed between 2000 and 2017 who received RTX for remission induction. Demographic and clinical features were analyzed. RESULTS Of the 17 patients included, the majority were myeloperoxidase-ANCA positive (n = 13, 76.5%). Uncontrolled asthma symptoms and atopy were present in 13 patients (76.5%). RTX was used for initial remission induction in patients with new onset of severe disease (n = 5, 29.4%) and after failed remission induction with other immunosuppression (n = 12, 70.6%). It was used for remission maintenance in nine patients (52.9%). GCs were used for maintenance at a median dose of 25 mg/day (interquartile range, 16.25-37.5). At the end of follow-up, 13 patients (76.5%) had non-severe or controlled asthma, and remission was achieved in 12 (70.6%). Median serum eosinophil and C-reactive protein values decreased (1.06 vs 0.10 × 109/L [P = .012] and 27.0 vs 5.0 mg/dL [P = .001], respectively), whereas pulmonary function test results remained unchanged. Median GC dose was significantly reduced at 6, 12, 18, and 24 months (P < .0001). Patients receiving RTX for maintenance required less than 10 mg of GCs for asthma control. CONCLUSION RTX seems to be safe and have GC-sparing efficacy for asthma control in EGPA. Randomized controlled trials are needed for detailed study of RTX for treating EGPA.Key Points• In this retrospective study we have concluded that rituximab (RTX) might be considered for the control of severe corticosteroid-dependent asthma in eosinophilic granulomatosis polyangiitis (EGPA) patients especially when myeloperoxidase antibodies are positive.• Rituximab has not been studied particularly for asthma control in EGPA patients.• The most noticeable effect of RTX was the decrease in the use of corticosteroids for the control of asthma.
Collapse
|
70
|
|
71
|
Cohen Tervaert JW. Should proteinase-3 and myeloperoxidase anti-neutrophil cytoplasmic antibody vasculitis be treated differently: part 2. Nephrol Dial Transplant 2019; 34:384-387. [PMID: 30668794 DOI: 10.1093/ndt/gfy406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/07/2018] [Indexed: 12/26/2022] Open
|
72
|
Berti A. The severe long-term outcomes of ‘non-severe’ eosinophilic granulomatosis with polyangiitis. Rheumatology (Oxford) 2019; 58:2081-2082. [DOI: 10.1093/rheumatology/kez250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alvise Berti
- Department of Rheumatology, Santa Chiara Hospital, University of Trento, Trento, Italy
- Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
73
|
Eosinophilic Granulomatosis with Polyangiitis: Clinical Pathology Conference and Review. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 6:1496-1504. [PMID: 30197069 DOI: 10.1016/j.jaip.2018.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/29/2018] [Accepted: 07/02/2018] [Indexed: 02/06/2023]
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) (formerly Churg-Strauss syndrome) is a small vessel vasculitis associated with asthma and eosinophilia. Despite its rarity, continuous gains are being made in understanding the disease with knowledge advancements regarding its epidemiology, heterogeneous clinical manifestations, management, and outcomes. Large knowledge gaps remain, however, particularly surrounding pathophysiologic and diagnostic uncertainties. There is still an incomplete understanding of the interplay between the eosinophilic and vasculitic processes that are features of disease pathogenesis. EGPA is also a conceptually difficult disorder given its dual categorization with hypereosinophilic syndromes and systemic vasculitides and the absence of a biomarker that can reliably distinguish between the two. In addition, recent evidence points to distinct, but partly overlapping, disease phenotypes, yet there is insufficient understanding to inform phenotype-tailored therapies. EGPA also remains a diagnostic challenge in part because asthma may be the primary or predominant manifestation for years, and the chronic corticosteroid requirement may mask other disease features. Efforts are ongoing to better elucidate pathophysiologic mechanisms, resolve classification issues, better characterize disease manifestations, and further clarify disease subcategorization, all of which will translate into better diagnosis and treatment with the possibility of specifically adapted therapies.
Collapse
|
74
|
[Current treatment of eosinophilic granulomatosis with polyangiitis (Churg-Strauss syndrome)]. Z Rheumatol 2019; 78:333-338. [PMID: 30627842 DOI: 10.1007/s00393-018-0580-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
For the treatment of eosinophilic granulomatosis with polyangiitis (EGPA) much less data are available when compared to the other anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV). At the same time EGPA also differs in many aspects from AAVs. Treatment is guided by the German and international guidelines. An adapted induction therapy is chosen depending on the disease activity, manifestations and factors determining the prognosis. For patients without negative prognostic factors glucocorticoids alone may be sufficient. A medium potent immunosuppressive agent may be added in order to economize on steroids. For patients with severe organ manifestations and adverse prognostic factors, a highly potent immunosuppression usually with cyclophosphamide, is necessary. In cases of remission a maintenance therapy is recommended in the same way as for other AAVs. Recently, a biological, the IL-5 antibody mepolizumab has also become available, although its precise role still has to be established.
Collapse
|
75
|
Update on eosinophilic granulomatosis with polyangiitis. Allergol Int 2019; 68:430-436. [PMID: 31266709 DOI: 10.1016/j.alit.2019.06.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 05/24/2019] [Accepted: 06/09/2019] [Indexed: 12/23/2022] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) (formerly Churg-Strauss syndrome) is a rare form of anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis characterized by eosinophil-rich granulomatous inflammation and small to medium-size vessel vasculitis associated with bronchial asthma and eosinophilia. Its rarity and unique features such as eosinophilic inflammation have delayed progress of research regarding EGPA for several years, compared to other forms of ANCA-associated vasculitis. However, recently, attention to EGPA as a research subject has been gradually increasing. To resolve problems in existing criteria for EGPA, new classification criteria for EGPA generated by a large international cohort will be launched and is being expected to accelerate future studies. Pathogenesis and roles of ANCA in EGPA are still largely unknown; however, it has been reported that glomerulonephritis is more frequent in ANCA-positive patients than in ANCA-negative patients, while heart failure is more frequent in ANCA-negative patients than in ANCA-positive patients. In addition, a recent genome-wide association study has suggested the presence of two genetically distinct subgroups of EGPA, which correspond to ANCA-positive and -negative subgroups. Although responses to glucocorticoids in EGPA are generally good, patients with EGPA often experience a relapse. Currently, there is no standard therapy for EGPA based on accumulation of clinical trial results. Recently, clinical benefits of mepolizumab for EGPA were proved by a randomized controlled trial and mepolizumab was approved for EGPA. In addition, various new drugs are under evaluation. To find optimal use of these drugs and to resolve unmet needs, such as relapse prevention, will be needed in future.
Collapse
|
76
|
Maillet T, Goletto T, Beltramo G, Dupuy H, Jouneau S, Borie R, Crestani B, Cottin V, Blockmans D, Lazaro E, Naccache JM, Pugnet G, Nunes H, de Menthon M, Devilliers H, Bonniaud P, Puéchal X, Mouthon L, Bonnotte B, Guillevin L, Terrier B, Samson M. Usual interstitial pneumonia in ANCA-associated vasculitis: A poor prognostic factor. J Autoimmun 2019; 106:102338. [PMID: 31570253 DOI: 10.1016/j.jaut.2019.102338] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Progressive fibrosing interstitial lung disease (ILD) is rarely associated with antineutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV). This study focused on the outcomes of ILD patients with associated AAV (AAV-ILD). METHODS AAV-ILD (cases: microscopic polyangiitis (MPA) or granulomatosis with polyangiitis (GPA) with ILD) were compared to AAV patients without ILD (controls). ILD was defined as a usual interstitial pneumonia (UIP) or non-specific interstitial pneumonia (NSIP) pattern. Two controls were matched to each case for age (>or ≤65 years), ANCA status (PR3-or MPO-positive) and creatininemia (≥or <150 μmol/L). RESULTS Sixty-two cases (89% MPO-ANCA+) were included. Median age at AAV diagnosis was 66 years. ILD (63% UIP), was diagnosed before (52%) or simultaneously (39%) with AAV. Cases versus 124 controls less frequently had systemic vasculitis symptoms. One-, 3- and 5-year overall survival rates, respectively, were: 96.7%, 80% and 66% for cases versus 93.5%, 89.6% and 83.8% for controls (p = 0.008). Multivariate analyses retained age >65 years (hazard ratio (HR) 4.54; p < 0.001), alveolar haemorrhage (HR 2.25; p = 0.019) and UIP (HR 2.73; p = 0.002), but not immunosuppressant use, as factors independently associated with shorter survival. CONCLUSION For AAV-ILD patients, only UIP was associated with poorer prognosis. Immunosuppressants did not improve the AAV-ILD prognosis. But in analogy to idiopathic pulmonary fibrosis, anti-fibrosing agents might be useful and should be assessed in AAV-ILD patients with a UIP pattern.
Collapse
Affiliation(s)
- Thibault Maillet
- Department of Internal Medicine and Clinical Immunology, CHU Dijon Bourgogne, Dijon, France
| | - Tiphaine Goletto
- Department of Pulmonology, Hôpital Saint-Louis, APHP, Paris, France
| | - Guillaume Beltramo
- Respiratory and ICU Department, Referral Center for Adults Rare Pulmonary Diseases, Inserm 1231, CHU Dijon-Bourgogne, Dijon, France
| | - Henry Dupuy
- Department of Internal Medicine and Infectious Diseases, Hôpital Haut-Lévêque, Bordeaux, France
| | - Stéphane Jouneau
- Department of Pulmonology, Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR S, 1085, Rennes, France
| | - Raphael Borie
- Department of Pulmonology, Hôpital Bichat, APHP, Paris, France
| | - Bruno Crestani
- Department of Pulmonology, Hôpital Bichat, APHP, Paris, France
| | - Vincent Cottin
- Department of Pulmonology, Hôpital Louis-Pradel, Bron, France
| | - Daniel Blockmans
- Department of Internal Medicine, UZ Leuven Hospital, Leuven, Belgium
| | - Estibaliz Lazaro
- Department of Internal Medicine and Infectious Diseases, Hôpital Haut-Lévêque, Bordeaux, France
| | - Jean-Marc Naccache
- Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Tenon, Service de Pneumologie, Site Constitutif Du Centre de Référence des Maladies Pulmonaires Rares OrphaLung, Paris, France
| | - Grégory Pugnet
- Department of Internal Medicine, CHU de Toulouse, Toulouse, France
| | - Hilario Nunes
- Department of Pulmonology, Hôpital Avicenne, APHP, Bobigny, France
| | - Mathilde de Menthon
- Department of Internal Medicine, Hôpital Bicêtre, APHP, Le Kremlin-Bicêtre, France
| | - Hervé Devilliers
- Department of Internal Medicine and Systemic Diseases, CHU Dijon-Bourgogne, Dijon, France
| | - Philippe Bonniaud
- Respiratory and ICU Department, Referral Center for Adults Rare Pulmonary Diseases, Inserm 1231, CHU Dijon-Bourgogne, Dijon, France
| | - Xavier Puéchal
- Department of Internal Medicine, Referral Center for Rare Autoimmune and Systemic Diseases, Hôpital Cochin, APHP, Inserm U1016, Université Paris Descartes, Paris, France
| | - Luc Mouthon
- Department of Internal Medicine, Referral Center for Rare Autoimmune and Systemic Diseases, Hôpital Cochin, APHP, Inserm U1016, Université Paris Descartes, Paris, France
| | - Bernard Bonnotte
- Department of Internal Medicine and Clinical Immunology, CHU Dijon Bourgogne, Dijon, France
| | - Loïc Guillevin
- Department of Internal Medicine, Referral Center for Rare Autoimmune and Systemic Diseases, Hôpital Cochin, APHP, Inserm U1016, Université Paris Descartes, Paris, France
| | - Benjamin Terrier
- Department of Internal Medicine, Referral Center for Rare Autoimmune and Systemic Diseases, Hôpital Cochin, APHP, Inserm U1016, Université Paris Descartes, Paris, France
| | - Maxime Samson
- Department of Internal Medicine and Clinical Immunology, CHU Dijon Bourgogne, Dijon, France
| | | |
Collapse
|
77
|
Abstract
Polyarteritis nodosa (PAN) is a necrotizing arteritis of medium-sized vessels, which is often fatal if untreated. It frequently affects the skin (nodules and ulcers), the peripheral nervous system (mononeuritis multiplex) and the visceral vessels (stenoses and microaneurysms). The complex diagnostic work-up requires discriminating PAN from infectious, malignant, drug-induced and other inflammatory conditions. It can be subclassified into further variants (idiopathic, associated with hepatitis B, associated with hereditary inflammatory diseases or isolated cutaneous disease). While idiopathic and hereditary inflammatory variants require immunosuppressive treatment, the hepatitis B-associated variant is treated with virustatic agents and plasmapheresis. The isolated cutaneous variant has a good prognosis and rarely requires highly potent immunosuppressives.
Collapse
|
78
|
Pagnoux C, Mendel A. Treatment of systemic necrotizing vasculitides: recent advances and important clinical considerations. Expert Rev Clin Immunol 2019; 15:939-949. [DOI: 10.1080/1744666x.2019.1656527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Christian Pagnoux
- Vasculitis Clinic, Division of Rheumatology, Mount Sinai Hospital, University Health Network, Toronto, Ontario, Canada
- Canadian Vasculitis research network (CanVasc), Canada
| | - Arielle Mendel
- Vasculitis Clinic, Division of Rheumatology, Mount Sinai Hospital, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
79
|
Choi CB, Park YB, Lee SW. Eosinophilic Granulomatosis with Polyangiitis: Experiences in Korean Patients. Yonsei Med J 2019; 60:705-712. [PMID: 31347324 PMCID: PMC6660441 DOI: 10.3349/ymj.2019.60.8.705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 12/17/2022] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is one form of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Identical to what has been called Churg-Strauss syndrome, EGPA exhibits both allergic and vasculitis features. EGPA was first described as a syndrome consisting of asthma, fever, eosinophilia, and organ involvement including heart failure, neuropathy, and kidney damage, by Churg and Strauss in 1951. On the basis of the 2012 Chapel Hill Consensus Conferences Nomenclature of Vasculitis, EGPA comprises three typical allergic components, including asthma, peripheral eosinophilia, and eosinophil-rich granuloma of the respiratory tracts. EGPA has three clinical and histological stages. The first is an allergic stage composed of asthma and sinusitis, and the second is an eosinophilic stage characterised by peripheral hypereosinophilia and intra-organ infiltration of eosinophils. The last is a vasculitic stage, including necrotising inflammation of small vessels and end-organ damage. In this review, we describe the classification criteria for EGPA and recommendations for the evaluation and management of EGPA with conventional and newly suggested drugs for EGPA. Also, we discuss a variety of clinical aspects such as predictive values for prognosis and associations with other Th2-mediated diseases and hepatitis B virus.
Collapse
Affiliation(s)
- Chan Bum Choi
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Yong Beom Park
- Division of Rheumatology, Department of Internal Medicine, and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Won Lee
- Division of Rheumatology, Department of Internal Medicine, and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
80
|
Sattui SE, Spiera RF. Treatment of Antineutrophil Cytoplasmic Antibody-Associated Vasculitis: Is There Still a Role for Cyclophosphamide? Rheum Dis Clin North Am 2019; 45:379-398. [PMID: 31277751 DOI: 10.1016/j.rdc.2019.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a group of systemic necrotizing vasculitides that includes granulomatosis with polyangiitis, microscopic polyangiitis, and eosinophilic granulomatosis with polyangiitis. Treatment of these conditions has improved during the past 2 decades with better understanding of these conditions and availability of newer agents. Cyclophosphamide (CYC) was the first drug demonstrated to afford successful treatment and improvement in AAV. With the emergence of newer agents with more favorable safety profiles, CYC is no longer the cornerstone of management of AAV. This article reviews existing data for treatment and the current role of CYC in the management of AAV.
Collapse
Affiliation(s)
- Sebastian E Sattui
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA.
| | - Robert F Spiera
- Division of Rheumatology, Department of Medicine, Weill Cornell Medical College, Scleroderma, Vasculitis & Myositis Center, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| |
Collapse
|
81
|
Lee JJY, Alsaleem A, Chiang GPK, Limenis E, Sontichai W, Yeung RSM, Akikusa J, Laxer RM. Hallmark trials in ANCA-associated vasculitis (AAV) for the pediatric rheumatologist. Pediatr Rheumatol Online J 2019; 17:31. [PMID: 31242923 PMCID: PMC6595671 DOI: 10.1186/s12969-019-0343-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) refers to a complex group of systemic vasculitides that are characterized by primary small-to-medium sized blood vessel inflammation with the presence of autoantibodies known as ANCA. AAV diseases include Granulomatosis with Polyangiitis (GPA), Eosinophilic Granulomatosis with Polyangiitis (EGPA), and Microscopic Polyangiitis (MPA). AAVs are challenging conditions associated with high cumulative disease and treatment related morbidity and mortality. Given its rarity and the resulting paucity of pediatric-specific clinical trial evidence, pediatric rheumatologists have had to often extrapolate from adult literature for management and therapeutic decisions. The aim of this review is to provide a comprehensive overview of the important findings and overall conclusions of critical landmark clinical trials in the induction and maintenance treatments in adult AAV for the pediatric rheumatologist. This review also highlights the outcomes of recent pediatric AAV observational studies and discusses the future research priorities in pediatric AAV management.
Collapse
Affiliation(s)
- Jennifer J. Y. Lee
- 0000 0004 0473 9646grid.42327.30Department of Pediatrics, Division of Rheumatology, Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - Alhanouf Alsaleem
- 0000 0004 0473 9646grid.42327.30Department of Pediatrics, Division of Rheumatology, Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - Grace P. K. Chiang
- 0000 0004 0473 9646grid.42327.30Department of Pediatrics, Division of Rheumatology, Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada ,0000 0004 1772 5868grid.413608.8Department of Pediatrics and Adolescent Medicine, Alice Ho Miu Ling Nethersole Hospital, HKSAR, Tai Po, Hong Kong
| | - Elizaveta Limenis
- 0000 0004 0473 9646grid.42327.30Department of Pediatrics, Division of Rheumatology, Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - Watchareewan Sontichai
- 0000 0004 0473 9646grid.42327.30Department of Pediatrics, Division of Rheumatology, Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada ,0000 0000 9039 7662grid.7132.7Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Rae S. M. Yeung
- 0000 0004 0473 9646grid.42327.30Department of Pediatrics, Division of Rheumatology, Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - Jonathan Akikusa
- 0000 0004 0614 0346grid.416107.5Department of Rheumatology, The Royal Children’s Hospital, Melbourne, Australia
| | - Ronald M. Laxer
- 0000 0004 0473 9646grid.42327.30Department of Pediatrics, Division of Rheumatology, Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| |
Collapse
|
82
|
Teixeira V, Mohammad AJ, Jones RB, Smith R, Jayne D. Efficacy and safety of rituximab in the treatment of eosinophilic granulomatosis with polyangiitis. RMD Open 2019; 5:e000905. [PMID: 31245051 PMCID: PMC6560673 DOI: 10.1136/rmdopen-2019-000905] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 12/18/2022] Open
Abstract
Introduction Eosinophilic granulomatosis with polyangiitis (EGPA) is a subset of antineutrophil cytoplasmic antibodies (ANCA) associated vasculitis with distinct pathophysiological mechanisms, clinical features and treatment responses. Rituximab is a licensed therapy for granulomatosis with polyangiitis and microscopic polyangiitis but there is limited experience of rituximab in EGPA. Methods EGPA patients from a tertiary centre who received rituximab for mostly refractory EGPA or in whom cyclophosphamide was contra indicated were studied. A standardised dataset was collected at time of initial treatment and every 3 months for 24 months. Response was defined as a Birmingham Vasculitis Activity Score (BVAS) of 0 and partial response as ≥50% reduction in BVAS from baseline. Remission was defined as a BVAS of 0 on prednisolone dose ≤5 mg. Results Sixty-nine patients (44 female) received rituximab between 2003 and 2017. Improvement (response and partial response) was observed in 76.8% of patients at 6 months, 82.8% at 12 months and in 93.2% by 24 months, while relapses occurred in 54% by 24 months, with asthma being the most frequent manifestation. The median BVAS decreased from 6 at baseline to 1 at 6 months, and 0 at 12 and 24 months. Prednisolone dose (mg/day, median) decreased from 12.5 to 7, 7.5 and 5 at 6, 12 and 24 months, respectively. ANCA positive patients had a longer asthma/ear, nose and throat (ENT) relapse-free survival time and a shorter time to remission. Discussion Rituximab demonstrated some efficacy in EGPA and led to a reduction in prednisolone requirement, but asthma and ENT relapse rates were high despite continued treatment. The ANCA positive subset appeared to have a more sustained response on isolated asthma/ENT exacerbations.
Collapse
Affiliation(s)
- Vítor Teixeira
- Vasculitis and Lupus Clinic, Addenbrooke's Hospital, Cambridge, UK.,Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Aladdin J Mohammad
- Vasculitis and Lupus Clinic, Addenbrooke's Hospital, Cambridge, UK.,Clinical Sciences, Rheumatology, Lund University, Lund, Skåne, Sweden
| | - Rachel B Jones
- Vasculitis and Lupus Clinic, Addenbrooke's Hospital, Cambridge, UK
| | - Rona Smith
- Vasculitis and Lupus Clinic, Addenbrooke's Hospital, Cambridge, UK
| | - David Jayne
- Vasculitis and Lupus Clinic, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
83
|
Ennis D, Lee JK, Pagnoux C. Mepolizumab for the treatment of eosinophilic granulomatosis with polyangiitis. Expert Opin Biol Ther 2019; 19:617-630. [PMID: 31146595 DOI: 10.1080/14712598.2019.1623875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Eosinophilic granulomatosis with polyangiitis (EGPA) is a rare antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis characterized by asthma, hypereosinophilia, and progressive multiorgan involvement. Although not fully elucidated, advancement in our understanding of the pathophysiology of EGPA has led to the development of multiple new treatment targets. AREAS COVERED Herein we review the epidemiology, clinical manifestations, pathophysiology, treatments, and ongoing research in the management of EGPA. The central role of Interleukin-5 (IL-5) in the development and maintenance of hypereosinophilia will be discussed. The value of mepolizumab, an anti-IL-5 monoclonal antibody, in the treatment of EGPA is reviewed in detail. EXPERT OPINION The available literature supports the use of mepolizumab for the induction and maintenance of remission of refractory, relapsing, or glucocorticoid-dependent EGPA with potentially greater benefit in those who are ANCA-positive or those with greater eosinophilia ( ≥ 150 cells/ μ L). Despite these positive results, relapses remain frequent, and the need for both short- and long-term glucocorticoid use remains common. More research is needed to address these needs and determine the precise role of mepolizumab.
Collapse
Affiliation(s)
- Daniel Ennis
- a Vasculitis Clinic, Division of Rheumatology , Mount Sinai Hospital, University Health Network , Toronto , Canada
| | - Jason Kihyuk Lee
- b Division of Rheumatology, Mount Sinai Hospital, University Health Network , Toronto Allergy and Asthma Clinic , Toronto , Canada
| | - Christian Pagnoux
- a Vasculitis Clinic, Division of Rheumatology , Mount Sinai Hospital, University Health Network , Toronto , Canada.,c Division of Rheumatology, Mount Sinai Hospital, University Health Network , Canadian Vasculitis research network (CanVasc) , Toronto , Canada
| |
Collapse
|
84
|
|
85
|
Abstract
Polyarteritis nodosa (PAN) is a necrotizing vasculitis affecting medium-sized vessels whose main manifestations are weight loss, fever, peripheral neuropathy, renal, musculoskeletal, gastrointestinal tract and/or cutaneous involvement(s), hypertension and/or cardiac failure. Peripheral neuropathy is one of the most frequent and earliest symptoms, affecting 50% to 75% of PAN patients. Central nervous system involvement affects only 2% to 10% of PAN patients, often late during the disease course. Treatment relies on combining corticosteroids and an immunosuppressant (mainly cyclophosphamide) in patients with poor prognoses. In patients with hepatitis B virus-related PAN, plasma exchanges and antiviral drugs should be combined with corticosteroids.
Collapse
Affiliation(s)
- Hubert de Boysson
- Department of Internal Medicine, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Loïc Guillevin
- Vasculitides and Scleroderma, Department of Internal Medicine, Referral Center for Rare Autoimmune and Systemic Diseases, Hôpital Cochin, Université Paris Descartes, 27, rue Fg Saint-Jacques, Paris 75679 Cedex 14, France.
| |
Collapse
|
86
|
Raffray L, Guillevin L. Treatment of Eosinophilic Granulomatosis with Polyangiitis: A Review. Drugs 2019; 78:809-821. [PMID: 29766394 DOI: 10.1007/s40265-018-0920-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Eosinophilic granulomatosis with polyangiitis (formerly Churg-Strauss syndrome) is a rare type of anti-neutrophil cytoplasm antibody-associated vasculitis. Nevertheless, eosinophilic granulomatosis with polyangiitis stands apart because it has features of vasculitis and eosinophilic disorders that require targeted therapies somewhat different from those used for other anti-neutrophil cytoplasm antibody-associated vasculitides. Considerable advances have been made in understanding the underlying pathophysiology of eosinophilic granulomatosis with polyangiitis that have highlighted the key role of eosinophils and opened new therapeutic opportunities. Its conventional treatment relies mainly on agents that decrease inflammation: corticosteroids and immunosuppressant adjunction for severe manifestations. New therapeutic approaches are needed for refractory disease, relapses and issues associated with corticosteroid dependence, especially for asthma manifestations. Drugs under evaluation mostly target eosinophils and B cells. Results of low-evidence-based trials suggested possible efficacies of biologicals: B-cell-blocking rituximab and anti-immunoglobulin E omalizumab. Recently, the first large-scale randomised controlled trial on eosinophilic granulomatosis with polyangiitis proved the efficacy of anti-interleukin-5 mepolizumab. That finding opens a new era in eosinophilic granulomatosis with polyangiitis management, with mepolizumab approval but also in future drug evaluations and trial designs for eosinophilic granulomatosis with polyangiitis. Additional studies are needed to determine which patients would benefit most from targeted therapies and achieve personalised treatment for patients with eosinophilic granulomatosis with polyangiitis. Herein, we review eosinophilic granulomatosis with polyangiitis characteristics and provide an overview of established and novel pharmacological agents.
Collapse
Affiliation(s)
- Loïc Raffray
- Department of Internal Medicine, Félix-Guyon University Hospital of La Réunion, Saint Denis, Réunion Island, France
| | - Loïc Guillevin
- Department of Internal Medicine, Referral Center for Rare Systemic and Autoimmune Diseases, Hôpital Cochin, Université Paris Descartes, Paris, France.
| |
Collapse
|
87
|
Jones RB, Hiemstra TF, Ballarin J, Blockmans DE, Brogan P, Bruchfeld A, Cid MC, Dahlsveen K, de Zoysa J, Espigol-Frigolé G, Lanyon P, Peh CA, Tesar V, Vaglio A, Walsh M, Walsh D, Walters G, Harper L, Jayne D. Mycophenolate mofetil versus cyclophosphamide for remission induction in ANCA-associated vasculitis: a randomised, non-inferiority trial. Ann Rheum Dis 2019; 78:399-405. [PMID: 30612116 DOI: 10.1136/annrheumdis-2018-214245] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Cyclophosphamide induction regimens are effective for antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), but are associated with infections, malignancies and infertility. Mycophenolate mofetil (MMF) has shown high remission rates in small studies of AAV. METHODS We conducted a randomised controlled trial to investigate whether MMF was non-inferior to cyclophosphamide for remission induction in AAV. 140 newly diagnosed patients were randomly assigned to MMF or pulsed cyclophosphamide. All patients received the same oral glucocorticoid regimen and were switched to azathioprine following remission. The primary endpoint was remission by 6 months requiring compliance with the tapering glucocorticoid regimen. Patients with an eGFR <15 mL/min were excluded from the study. RESULTS At baseline, ANCA subtype, disease activity and organ involvement were similar between groups. Non-inferiority was demonstrated for the primary remission endpoint, which occurred in 47 patients (67%) in the MMF group and 43 patients (61%) in the cyclophosphamide group (risk difference 5.7%, 90% CI -7.5% to 19%). Following remission, more relapses occurred in the MMF group (23 patients, 33%) compared with the cyclophosphamide group (13 patients, 19%) (incidence rate ratio 1.97, 95% CI 0.96 to 4.23, p=0.049). In MPO-ANCA patients, relapses occurred in 12% of the cyclophosphamide group and 15% of the MMF group. In PR3-ANCA patients, relapses occurred in 24% of the cyclophosphamide group and 48% of the MMF group. Serious infections were similar between groups (26% MMF group, 17% cyclophosphamide group) (OR 1.67, 95% CI 0.68 to 4.19, p=0.3). CONCLUSION MMF was non-inferior to cyclophosphamide for remission induction in AAV, but resulted in higher relapse rate. TRIAL REGISTRATION NUMBER NCT00414128.
Collapse
Affiliation(s)
- Rachel B Jones
- Department of Renal Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Thomas F Hiemstra
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Cambridge Clinical Trials Unit, Addenbrooke's Hospital, Cambridge, UK
| | - Jose Ballarin
- Department of Nephrology, Fundació Puigvert, Barcelona, Spain
| | | | - Paul Brogan
- Department of Paediatric Rheumatology, University College London Great Ormond Street Institute of Child Health, London, UK
- Department of Paediatric Rheumatology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Annette Bruchfeld
- Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Maria C Cid
- Department of Autoimmune Diseases, Hospital Clinic, University of Barcelona, Institut d'investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Karen Dahlsveen
- Department of Renal Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Janak de Zoysa
- Renal Service, Waitemata District Health Board, Auckland, New Zealand
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Georgína Espigol-Frigolé
- Department of Autoimmune Diseases, Hospital Clinic, University of Barcelona, Institut d'investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Peter Lanyon
- Department of Rheumatology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Chen Au Peh
- Department of Renal Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Vladimir Tesar
- Department of Nephrology, Charles University and General University Hospital, Prague, Czech Republic
| | - Augusto Vaglio
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Firenze, Firenze, Italy
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, Firenze, Italy
| | - Michael Walsh
- Departments of Medicine and Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Dorothy Walsh
- Department of Renal Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Giles Walters
- Department of Renal Medicine, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Lorraine Harper
- Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - David Jayne
- Department of Renal Medicine, Addenbrooke's Hospital, Cambridge, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
88
|
Steinfeld J, Bradford ES, Brown J, Mallett S, Yancey SW, Akuthota P, Cid MC, Gleich GJ, Jayne D, Khoury P, Langford CA, Merkel PA, Moosig F, Specks U, Weller PF, Wechsler ME. Evaluation of clinical benefit from treatment with mepolizumab for patients with eosinophilic granulomatosis with polyangiitis. J Allergy Clin Immunol 2018; 143:2170-2177. [PMID: 30578883 DOI: 10.1016/j.jaci.2018.11.041] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/02/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND In a recent phase III trial (NCT02020889) 53% of mepolizumab-treated versus 19% of placebo-treated patients with eosinophilic granulomatosis with polyangiitis (EGPA) achieved protocol-defined remission. OBJECTIVE We sought to investigate post hoc the clinical benefit of mepolizumab in patients with EGPA using a comprehensive definition of benefit encompassing remission, oral glucocorticoid (OGC) dose reduction, and EGPA relapses. METHODS The randomized, placebo-controlled, double-blind, parallel-group trial recruited patients with relapsing/refractory EGPA receiving stable OGCs (prednisolone/prednisone, ≥7.5-50 mg/d) for 4 or more weeks. Patients received 300 mg of subcutaneous mepolizumab or placebo every 4 weeks for 52 weeks. Clinical benefit was defined post hoc as follows: remission at any time (2 definitions used), 50% or greater OGC dose reduction during weeks 48 to 52, or no EGPA relapses. The 2 remission definitions were Birmingham Vasculitis Activity Score of 0 plus OGC dose of 4 mg/d or less (remission 1/clinical benefit 1) or 7.5 mg/d or less (remission 2/clinical benefit 2). Clinical benefit was assessed in all patients and among subgroups with a baseline blood eosinophil count of less than 150 cells/μL, baseline OGC dosage of greater than 20 mg/d, or weight of greater than 85 kg. RESULTS With mepolizumab versus placebo, 78% versus 32% of patients experienced clinical benefit 1, and 87% versus 53% of patients experienced clinical benefit 2 (both P < .001). Significantly more patients experienced clinical benefit 1 with mepolizumab versus placebo in the blood eosinophil count less than 150 cells/μL subgroup (72% vs 43%, P = .033) and weight greater than 85 kg subgroup (68% vs 23%, P = .005); in the OGC greater than 20 mg/d subgroup, results were not significant but favored mepolizumab (60% vs 36%, P = .395). CONCLUSION When a comprehensive definition of clinical benefit was applied to data from a randomized controlled trial, 78% to 87% of patients with EGPA experienced benefit with mepolizumab.
Collapse
Affiliation(s)
- Jonathan Steinfeld
- Respiratory TAU & Flexible Discovery Unit, GlaxoSmithKline, Philadelphia, Pa
| | - Eric S Bradford
- Respiratory Therapeutic Area, GlaxoSmithKline, Research Triangle Park, NC
| | - Judith Brown
- Research and Development, Immuno-Inflammation TAU, Uxbridge, United Kingdom
| | - Stephen Mallett
- Research & Development, Statistics, Programming and Data Standards, GlaxoSmithKline, Stockley Park West, Uxbridge, United Kingdom
| | - Steven W Yancey
- Respiratory Therapeutic Area, GlaxoSmithKline, Research Triangle Park, NC
| | - Praveen Akuthota
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, Calif
| | - Maria C Cid
- Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Gerald J Gleich
- Departments of Dermatology and Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paneez Khoury
- Human Eosinophil Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Carol A Langford
- Department of Rheumatic and Immunologic Diseases, Center for Vasculitis Care and Research, Cleveland Clinic, Cleveland, Ohio
| | - Peter A Merkel
- Division of Rheumatology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pa
| | - Frank Moosig
- Rheumazentrum, Schleswig-Holstein Mitte, Neumünster, Germany
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minn
| | - Peter F Weller
- Divisions of Allergy and Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass
| | | |
Collapse
|
89
|
Puéchal X, Pagnoux C, Baron G, Lifermann F, Geffray L, Quéméneur T, Saraux J, Wislez M, Cottin V, Ruivard M, Limal N, Guillevin L. Suivi à long-terme des patients atteints de granulomatose éosinophilique avec polyangéite inclus dans l’étude CHUSPAN2 ayant évalué l’intérêt de l’adjonction de l’azathioprine à la corticothérapie en traitement d’induction. Rev Med Interne 2018. [DOI: 10.1016/j.revmed.2018.10.263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
90
|
Misra DP, Naidu GSRSNK, Agarwal V, Sharma A. Vasculitis research: Current trends and future perspectives. Int J Rheum Dis 2018; 22 Suppl 1:10-20. [PMID: 30168260 DOI: 10.1111/1756-185x.13370] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/03/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Durga P. Misra
- Department of Clinical Immunology; Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS); Lucknow India
| | - Godasi S. R. S. N. K. Naidu
- Department of Internal Medicine; Clinical Immunology and Rheumatology Services; Postgraduate Institute of Medical Education and Research (PGIMER); Chandigarh India
| | - Vikas Agarwal
- Department of Clinical Immunology; Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS); Lucknow India
| | - Aman Sharma
- Department of Internal Medicine; Clinical Immunology and Rheumatology Services; Postgraduate Institute of Medical Education and Research (PGIMER); Chandigarh India
| |
Collapse
|
91
|
Saku A, Furuta S, Hiraguri M, Ikeda K, Kobayashi Y, Kagami SI, Kurasawa K, Matsumura R, Nakagomi D, Sugiyama T, Umibe T, Watanabe N, Nakajima H. Longterm Outcomes of 188 Japanese Patients with Eosinophilic Granulomatosis with Polyangiitis. J Rheumatol 2018; 45:1159-1166. [PMID: 29907668 DOI: 10.3899/jrheum.171352] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Patients with eosinophilic granulomatosis with polyangiitis (EGPA) frequently experience relapses, which lead to cumulative organ damage. In this retrospective observational study, we aimed to reveal the risk factors for relapse in EGPA. METHODS A total of 188 Japanese patients with EGPA diagnosed between 1996 and 2015 were identified from medical records in 10 hospitals. The diagnosis was based on the American College of Rheumatology 1990 criteria or Lanham's criteria. Baseline characteristics, treatments, asthma exacerbation, and relapses were evaluated by retrospective chart review. RESULTS The median followup period was 56 months. The median age at disease onset was 59.7 years. At the disease onset, 95.2% of the patients had a history of bronchial asthma and 44.7% were positive for antineutrophil cytoplasmic antibodies. The cumulative survival and relapse-free survival rates at 5 years were 89.6% and 64.0%, respectively. Multivariate analysis with 2 models, proportional hazards, and competing risk models, was performed to identify the factors associated with relapse. The proportional hazards model identified azathioprine (AZA) maintenance therapy and high eosinophil counts at onset as independent factors with lower relapse risks, and high immunoglobulin E (IgE) levels at onset as a risk factor for relapse. The competing risk model identified no statistically significant factors. CONCLUSION Although potential benefit of AZA maintenance therapy in preventing relapse of EGPA was suggested by the proportional hazards model, there was a discrepancy in the results between the models. Eosinophil counts and IgE levels at onset were also identified as candidates of factors associated with relapse in EGPA.
Collapse
Affiliation(s)
- Aiko Saku
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Shunsuke Furuta
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan. .,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital.
| | - Masaki Hiraguri
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Kei Ikeda
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Yoshihisa Kobayashi
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Shin-Ichiro Kagami
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Kazuhiro Kurasawa
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Ryutaro Matsumura
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Daiki Nakagomi
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Takao Sugiyama
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Takeshi Umibe
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Norihiko Watanabe
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| | - Hiroshi Nakajima
- From the Department of Allergy and Clinical Immunology, Chiba University Hospital; Department of Internal Medicine, Narita Red Cross Hospital; Department of Internal Medicine, Chiba Aoba Municipal Hospital; Department of Allergy and Clinical Immunology, Asahi General Hospital, Chiba; Department of Rheumatology, Dokkyo Medical University, Tochigi; Department of Rheumatology, Allergy and Clinical Immunology, National Hospital Organization Chiba-East Hospital, Chiba; Third Department of Internal Medicine, University of Yamanashi, Yamanashi; Department of Rheumatology, National Hospital Organization Shimoshizu Hospital; Department of Internal Medicine, Matsudo City Hospital; Centre for Rheumatic Diseases, Chibaken Saiseikai Narashino Hospital, Chiba, Japan.,A. Saku, MD, Chiba University Hospital; S. Furuta, MD, PhD, Chiba University Hospital; M. Hiraguri, MD, PhD, Narita Red Cross Hospital; K. Ikeda, MD, PhD, Chiba University Hospital; Y. Kobayashi, MD, PhD, Chiba Aoba Municipal Hospital; S.I. Kagami, MD, PhD, Asahi General Hospital; K. Kurasawa, MD, PhD, Dokkyo Medical University; R. Matsumura, MD, PhD, Chiba-East Hospital; D. Nakagomi, MD, PhD, University of Yamanashi; T. Sugiyama, MD, PhD, Shimoshizu Hospital; T. Umibe, MD, PhD, Matsudo City Hospital; N. Watanabe, MD, PhD, Chibaken Saiseikai Narashino Hospital; H. Nakajima, MD, PhD, Chiba University Hospital
| |
Collapse
|
92
|
Jardel S, Puéchal X, Le Quellec A, Pagnoux C, Hamidou M, Maurier F, Aumaitre O, Aouba A, Quemeneur T, Subra JF, Cottin V, Sibilia J, Godmer P, Cacoub P, Fauchais AL, Hachulla E, Maucort-Boulch D, Guillevin L, Lega JC. Mortality in systemic necrotizing vasculitides: A retrospective analysis of the French Vasculitis Study Group registry. Autoimmun Rev 2018; 17:653-659. [PMID: 29730524 DOI: 10.1016/j.autrev.2018.01.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 01/07/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The aim of the study was to describe the evolution of mortality and cause-specific mortality over time in patients with systemic necrotizing vasculitides (SNV), including polyarteritis nodosa (PAN), granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA). METHODS Patients with SNV from the French Vasculitis Study Group registry were divided into 5 groups according to the date of diagnosis: <1980, 1980-1989, 1990-1999, 2000-2010, and ≥ 2010. The causes of death were classified as vasculitis, infection, cardiovascular, malignancy, miscellaneous, or unknown. RESULTS Among the 2217 patients included (PAN 16.1%, GPA 41.7%, EGPA 22.6%, MPA 19.6%), overall incidence of death was 2.26 per 100 person-years. The overall survival improved during each period considered. The 5-year survival rate increased from 72.2% (95% confidence interval [CI] 59.7-87.2) for patients diagnosed before 1980 to 94.5% (95% CI 90.4-98.8) after 2010 (p < 0.001). Periods of diagnosis, age, and male gender were independently associated with a poor survival with a non-significant difference between vasculitis. The incidence of mortality between the 1980s and after 2010 significantly decreased for vasculitis-related (p = 0.03) and cardiovascular-related deaths (p = 0.04). Incidence of death by infection remained stable between the 1980s and the 2000s but no death by infection occurred after 2010. The incidence of death by malignancy remained stable over time. CONCLUSION Overall survival of SNV patients has improved since the 1980s with the decrease of vasculitis- and cardiovascular-related deaths, but cancer-related mortality remained stable. These results highlight malignancy as the current target to improve the overall prognosis.
Collapse
Affiliation(s)
- Sabine Jardel
- National Referral Centre for rare Juvenile Rheumatological and Autoimmune Diseases, Department of Internal and Vascular Medicine, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, CNRS, Claude Bernard University Lyon 1, Lyon, France
| | - Xavier Puéchal
- National Referral Centre for rare Systemic Autoimmune Diseases, Hôpital Cochin, Assistance Public - Hôpitaux de Paris (AP-HP), Université Paris Descartes, Paris, France
| | - Alain Le Quellec
- Department of Internal Medicine, Centre Hospitalier Régional Universitaire de Montpellier, Hôpital St. Eloi, Montpellier 1 University, Montpellier, France
| | - Christian Pagnoux
- National Referral Centre for rare Systemic Autoimmune Diseases, Hôpital Cochin, Assistance Public - Hôpitaux de Paris (AP-HP), Université Paris Descartes, Paris, France
| | - Mohamed Hamidou
- Department of Internal Medicine, Centre Hospitalier Universitaire Hôtel-Dieu, Nantes, France
| | - François Maurier
- Department of Internal medicine and Clinical Immunology, Site Belle Isle, Metz, France
| | - Olivier Aumaitre
- Department of Internal Medicine, Centre Hospitalier Universitaire, Hôpital Gabriel Montpied, Clermont-Ferrand, France
| | - Achille Aouba
- Department of Internal Medicine, Centre Hospitalier Universitaire de Caen, Caen University, Caen, France
| | - Thomas Quemeneur
- Department of Internal Medicine, Centre Hospitalier, Valenciennes, France
| | - Jean-François Subra
- Department of Internal Medicine, Centre Hospitalier Universitaire d' Angers, Angers, France
| | - Vincent Cottin
- Department of Respiratory Medicine, National Reference Centre for Rare Pulmonary Diseases, Louis Pradel Hospital, Hospices Civils de Lyon, Claude Bernard University Lyon 1, Lyon, France
| | - Jean Sibilia
- Department of Rheumatology, National Reference Center for rare Systemic Autoimmune Diseases, Strasbourg University Hospital, Strasbourg University, Strasbourg, France
| | - Pascal Godmer
- Department of Internal Medicine, Centre Hospitalier Bretagne-Atlantique, Vannes, France
| | - Patrice Cacoub
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7211, and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75005, Paris, France; INSERM, UMRS 959, F-75013, Paris, France; CNRS, FRE3632, F-75005, Paris, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
| | - Anne Laure Fauchais
- Department of Internal Medicine, Hôpital Dupuytren, Centre Hospitalier Universitaire de Limoges Limoges University, Limoges, France
| | - Eric Hachulla
- Department of Internal Medicine, Hôpital Claude Huriez, University of Lille, Lille, France
| | - Delphine Maucort-Boulch
- Department of Biostatistics and bioinformatics, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon Claude Bernard University Lyon 1, Lyon, France
| | - Loïc Guillevin
- National Referral Centre for rare Systemic Autoimmune Diseases, Hôpital Cochin, Assistance Public - Hôpitaux de Paris (AP-HP), Université Paris Descartes, Paris, France
| | - Jean-Christophe Lega
- National Referral Centre for rare Juvenile Rheumatological and Autoimmune Diseases, Department of Internal and Vascular Medicine, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, CNRS, Claude Bernard University Lyon 1, Lyon, France.
| | | |
Collapse
|
93
|
Navarro-Mendoza EP, Tobón GJ. Eosinophilic Granulomatosis With Polyangiitis: Newer Therapies. Curr Rheumatol Rep 2018; 20:23. [PMID: 29611001 DOI: 10.1007/s11926-018-0736-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Eosinophilic granulomatosis with polyangiitis (EGPA) is a systemic disseminated vasculitis associated with extravascular granulomas in patients suffering from asthma and tissue eosinophilia. Current therapies to achieve remission and prevent relapse include glucocorticoids and immunosuppressants like cyclophosphamide. RECENT FINDINGS With the right treatment, clinical prognosis is favorable, so concerted efforts have been made in recent years to find new alternatives for treating severe EGPA. Monoclonal antibodies such as omalizumab, rituximab, and mepolizumab are among these new options. This review summarizes the pathogenesis and clinical manifestations of EGPA and critically examines current and emerging therapies.
Collapse
Affiliation(s)
- Erika P Navarro-Mendoza
- GIRAT: Grupo de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Fundación Valle del Lili, Universidad Icesi, Cl 18 #, Cali, 122-135, Colombia
| | - Gabriel J Tobón
- GIRAT: Grupo de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Fundación Valle del Lili, Universidad Icesi, Cl 18 #, Cali, 122-135, Colombia.
- Immunology Laboratory, Fundación Valle del Lili, Cali, Colombia.
| |
Collapse
|
94
|
Update on the epidemiology, risk factors, and outcomes of systemic vasculitides. Best Pract Res Clin Rheumatol 2018; 32:271-294. [DOI: 10.1016/j.berh.2018.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023]
|
95
|
Watts RA, Robson J. Introduction, epidemiology and classification of vasculitis. Best Pract Res Clin Rheumatol 2018; 32:3-20. [DOI: 10.1016/j.berh.2018.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/18/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022]
|
96
|
Conventional and biological immunosuppressants in vasculitis. Best Pract Res Clin Rheumatol 2018; 32:94-111. [DOI: 10.1016/j.berh.2018.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/18/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022]
|
97
|
Eosinophils from Physiology to Disease: A Comprehensive Review. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9095275. [PMID: 29619379 PMCID: PMC5829361 DOI: 10.1155/2018/9095275] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/27/2017] [Indexed: 12/26/2022]
Abstract
Despite being the second least represented granulocyte subpopulation in the circulating blood, eosinophils are receiving a growing interest from the scientific community, due to their complex pathophysiological role in a broad range of local and systemic inflammatory diseases as well as in cancer and thrombosis. Eosinophils are crucial for the control of parasitic infections, but increasing evidence suggests that they are also involved in vital defensive tasks against bacterial and viral pathogens including HIV. On the other side of the coin, eosinophil potential to provide a strong defensive response against invading microbes through the release of a large array of compounds can prove toxic to the host tissues and dysregulate haemostasis. Increasing knowledge of eosinophil biological behaviour is leading to major changes in established paradigms for the classification and diagnosis of several allergic and autoimmune diseases and has paved the way to a "golden age" of eosinophil-targeted agents. In this review, we provide a comprehensive update on the pathophysiological role of eosinophils in host defence, inflammation, and cancer and discuss potential clinical implications in light of recent therapeutic advances.
Collapse
|
98
|
Smitienko I, Novikov P, Moiseev S. Does the Revised Definition of Eosinophilic Granulomatosis With Polyangiitis (Churg‐Strauss) Indicate the Need for a New Treatment? Comment on the Article by Puéchal et al. Arthritis Rheumatol 2018; 70:149-151. [DOI: 10.1002/art.40335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | - Pavel Novikov
- Sechenov First Moscow State Medical University Moscow Russia
| | - Sergey Moiseev
- Sechenov First Moscow State Medical University Moscow Russia
| |
Collapse
|
99
|
Kahn JE, Groh M, Lefèvre G. (A Critical Appraisal of) Classification of Hypereosinophilic Disorders. Front Med (Lausanne) 2017; 4:216. [PMID: 29259972 PMCID: PMC5723313 DOI: 10.3389/fmed.2017.00216] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/16/2017] [Indexed: 12/17/2022] Open
Abstract
Hypereosinophilia (HE) is a heterogeneous condition that can be reported in various (namely inflammatory, allergic, infectious, or neoplastic) diseases with distinct pathophysiological pathways. In 1975, Chusid et al. published the first diagnostic criteria of hypereosinophilic syndromes (HES). Over the years, as both basic and clinical knowledge improved, several updates have been suggested, with a focus on better distinguishing isolated or asymptomatic eosinophilia from diseases with specific eosinophil-related organ damage. Moreover, underlying molecular and cellular mechanisms of eosinophilia gradually became the cornerstone of successive attempts to classify HE-related diseases. In 2011, the International Cooperative Working Group on Eosinophil Disorders criteria emerged from a multidisciplinary Working Conference on Eosinophil Disorders and Syndromes, and provided substantial contribution to the clarification of general concepts and definitions in the field of HE. Yet, owing to the low prevalence of HE/HES, to the numerous diseases encompassed in the spectrum of HE-related disorders (with sometimes overlapping phenotypes), many questions are left unanswered (e.g., the need to better standardize the use of modern molecular tools, or the clinical relevance of distinguishing different subtypes of idiopathic HES). Here, we review the current state of knowledge in the fields of classification and diagnosis criteria of HE-related diseases, with emphasis on the analysis of both strengths and weaknesses of present concepts and their usefulness in daily practice.
Collapse
Affiliation(s)
- Jean Emmanuel Kahn
- Service de Médecine Interne, Centre de Référence des Syndromes Hyperéosinophiliques-CEREO, Hôpital Foch, Université Versailles-Saint Quentin en Yvelines, Suresnes, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Saint Louis, Université Paris-Diderot, Paris, France
| | - Guillaume Lefèvre
- Université de Lille, INSERM, CHU Lille, U995 - LIRIC - Lille Inflammation Research International Center, Institut d'Immunologie, Centre de Référence des Syndromes Hyperéosinophiliques-CEREO, Unité d'Immunologie Clinique, Lille, France
| |
Collapse
|
100
|
Berti A, Specks U, Keogh KA, Cornec D. Current and Future Treatment Options for Eosinophilic Granulomatosis With Polyangiitis. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2017. [DOI: 10.1007/s40674-017-0073-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|