Вы находитесь на странице: 1из 18

CHAPTER

53  VASCULITIS
J. Charles Jennette, Eric T. Weimer, Jason Kidd

INTRODUCTION, 1016 Polyarteritis Nodosa, 1020 IgA Vasculitis (Henoch-Schönlein


Kawasaki Disease, 1021 Purpura), 1028
DIAGNOSTIC CLASSIFICATION
OF VASCULITIS, 1016 Hypocomplementemic Urticarial
SMALL VESSEL  
Vasculitis, 1029
LARGE VESSEL   VASCULITIS, 1022
Rheumatoid Vasculitis, 1030
VASCULITIS, 1017 ANCA-Associated Small Vessel
Takayasu Arteritis, 1018 Vasculitis, 1022 SUMMARY, 1031
Giant Cell Arteritis, 1019 Anti-Glomerular Basement SELECTED REFERENCES, 1031
Membrane Disease, 1026
MEDIUM VESSEL   Cryoglobulinemic Vasculitis, 1027
VASCULITIS, 1020

lar and cellular events that are caused by specific pathogenic events. This
KEY POINTS approach is difficult to apply to all forms of vasculitis because the etiology
• Vasculitis affects blood vessels of any type in any organ or tissue and and pathogenesis of some forms are unknown. Vasculitis can be catego-
thus causes extremely diverse signs and symptoms of disease that rized into infectious vasculitis that is known to be caused by vessel wall
overlap with many other diseases. invasion by pathogens that incite the inflammation (Pagnoux et al, 2006),
and noninfectious vasculitis that is not known to be caused by infection
• Even if it is not the final diagnosis, vasculitis often is in the differential
in vessel walls (Jennette et al, 2013) (Box 53-1). For example, the infec-
diagnosis in a patient with evidence for single organ or systemic
inflammatory disease. tious vasculitis of Rocky Mountain spotted fever (RMSF) is caused by
proliferation of Rickettsia rickettsii in endothelial cells and smooth muscle
• Laboratory tests for vasculitis identify a specific etiologic factor or cells in small vessels in many tissues (Silverman & Bond, 1984). The
measure perturbations in physiological molecular and cellular events immune response to vessel wall rickettsiae causes vascular inflammation
that result from specific pathogenic events. that results in hemorrhage that produces the skin spots that are typical
• Clinical laboratory tests for the diagnosis of vasculitis include tests for RMSF. Bacterial sepsis can cause small vessel vasculitis that can mimic
that document active inflammation (e.g., C reactive protein assay), noninfectious immune-mediated vasculitis ( Jain et al, 2007) and clearly
demonstrate injury to a specific organ (e.g., urinalysis), or implicate a must be diagnosed correctly and treated with antimicrobial drugs and not
specific category of vasculitis (e.g., cryoglobulin assay). immunosuppression.
• Vasculitis is categorized into infectious vasculitis that is caused by Noninfectious vasculitis is caused by inflammation that is not known
vessel wall invasion by pathogens and noninfectious vasculitis that is to be caused by pathogens in vessel walls. However, it is important to
not known to be caused by infection in vessel walls. realize that the etiology and pathogenesis of some forms of vasculitides are
currently not known, so an infectious etiology cannot be completely ruled
• Noninfectious vasculitis is categorized as immune complex–mediated, out. For example, although the etiology is not known for Kawasaki disease,
antineutrophil cytoplasmic autoantibody (ANCA)–mediated, or
giant cell arteritis, and Takayasu arteritis, some theories propose that an
cell-mediated immune vasculitis.
unidentified pathogen may be the cause. For example, Mycobacterium
• Clinical diagnosis of vasculitis is based on both the presence and tuberculosis has been incriminated but not proven to be involved in the
absence of multiple clinical, pathologic, and laboratory features. pathogenesis of Takayasu arteritis and a Burkholderia bacterium in the
• Accurate and precise diagnosis of a specific category of vasculitis is pathogenesis of giant cell arteritis (van Timmeren et al, 2014).
essential for optimum patient outcome. Noninfectious vasculitis can be caused indirectly by an infection that
initiates a sequence of pathogenic events that causes vasculitis. For example,
cryoglobulinemic vasculitis can be caused by hepatitis C virus infection in
the liver, which induces polyclonal, oligoclonal, and monoclonal B cell
proliferations that produce rheumatoid factors (antiantibodies) that bind
INTRODUCTION to other immunoglobulins to form pathogenic immune complexes (Tede-
Vasculitis is inflammation of blood vessel walls. Blood vessels of any type schi et al, 2007). Laboratory tests for cryoglobulinemic vasculitis comprise
in any organ or tissue may be affected. Thus the signs and symptoms of identification of the infectious agent (hepatitis C virus serology) that initi-
disease are extremely diverse and overlap with many other diseases. As a ated the autoimmune response, the autoantibodies (rheumatoid factor
consequence, vasculitis often is in the differential diagnosis for both single assay), the pathogenic immune complexes (cryoglobulin assay), secondary
organ and systemic disease, especially in a patient with evidence for an disturbances in inflammatory mediators (e.g., hypocomplementemia), and
inflammatory condition. Clinical laboratory tests (i.e., tests for biomarkers) the evidence for end organ damage (hematuria and proteinuria).
that are used for the diagnosis of vasculitis fall into three categories: those Laboratory testing for RMSF provides a good example of testing strate-
that document the presence of an inflammatory process (e.g., C reactive gies for infectious vasculitis. The most direct laboratory test for RMSF is
protein assay), those that demonstrate injury to a specific organ (urinalysis), immunohistologic visualization of R. rickettsii in vessel walls in a skin
and those that are indicative of a specific category of vasculitis (e.g., cryo- biopsy specimen. This test has excellent specificity and 70% sensitivity
globulin assay). This chapter reviews the clinical and pathologic features during the acute phase before antibiotic treatment is started. Serologic
of the major categories of vasculitis and discusses the role of laboratory testing for antibodies to R. rickettsii has poor sensitivity during the first 10
testing in the diagnosis and management of these diseases. to 12 days but increases to more than 90% after 2 weeks (Nathavitharana
& Mitty, 2015). The most definitive serologic finding for RMSF is a rise
DIAGNOSTIC CLASSIFICATION in titer over several weeks. Analogous testing is applicable to other infec-
tious vasculitis—that is, testing to identify the etiologic infection and
OF VASCULITIS testing to demonstrate a specific immune response to the pathogen.
Laboratory tests for a disease often are based on identification of a specific Based on known or putative pathogenic mechanisms, noninfectious
etiologic factor or measurement of perturbations in physiological molecu- vasculitis can be categorized as immune complex–mediated, antineutrophil

1016
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
BOX 53-1  BOX 53-2 
Putative Pathogenic Mechanisms for Vasculitis Names for Vasculitides Adopted by the 2012 International
Chapel Hill Consensus Conference on the Nomenclature
Vasculitis Caused by Vessel Wall Infection
of Vasculitides
Bacterial (e.g., neisserial vasculitis, syphilitic aortitis)
Rickettsial (e.g., Rocky Mountain spotted fever) Large Vessel Vasculitis (LVV)
Fungal (e.g., cryptococcal vasculitis) Takayasu arteritis (TAK)
Viral (e.g., cytomegalovirus vasculitis) Giant cell arteritis (GCA)
Parasitic (e.g., toxoplasmosis vasculitis
Medium Vessel Vasculitis (MVV)
Immune Complex–Mediated Vasculitis
Polyarteritis nodosa (PAN)
Cryoglobulinemic vasculitis Kawasaki disease (KD)
Polyarteritis nodosa
Small Vessel Vasculitis (SVV)
IgA vasculitis (Henoch-Schönlein)
Hypocomplementemic urticarial vasculitis Antineutrophil cytoplasmic antibody (ANCA)–associated vasculitis (AAV)
Lupus vasculitis Microscopic polyangiitis (MPA)
Rheumatoid vasculitis Granulomatosis with polyangiitis (Wegener’s) (GPA)
Drug-induced immune complex vasculitis Eosinophilic granulomatosis with polyangiitis (Churg-Strauss) (EGPA)
Cancer-induced (paraneoplastic) immune complex vasculitis Immune complex SVV
Anti-glomerular basement membrane disease Anti-glomerular basement membrane (anti-GBM) disease
Cryoglobulinemic vasculitis (CV)
Antineutrophil Cytoplasmic Antibody (ANCA)–Mediated Vasculitis
IgA vasculitis (Henoch-Schönlein) (IgAV)
Microscopic polyangiitis Hypocomplementemic urticarial vasculitis (HUV) (anti-C1q vasculitis)
Granulomatosis with polyangiitis (Wegener’s)
Variable Vessel Vasculitis (VVV)
Eosinophilic granulomatosis with polyangiitis (Churg-Strauss)
Drug-induced ANCA vasculitis Behçet’s disease (BD)
Cogan’s syndrome (CS)
Cell-Mediated Immune Vasculitis
Single-Organ Vasculitis (SOV)
Takayasu arteritis

PART 6
Giant cell arteritis Cutaneous leukocytoclastic angiitis
Sarcoid vasculitis Cutaneous arteritis
Primary central nervous system vasculitis
Isolated aortitis
Others
Vasculitis Associated with Systemic Disease
cytoplasmic autoantibody (ANCA)–mediated, or cell-mediated immune
Lupus vasculitis
vasculitis (see Box 53-1). Immune complex vasculitis can be caused by
Rheumatoid vasculitis
antibodies directed against a non–self-antigen (e.g., hepatitis B immune
Sarcoid vasculitis
complex vasculitis caused by antibodies to viral antigens) or autoantibodies Others
directed against an autoantigen (e.g., lupus vasculitis caused by autoanti-
bodies against nuclear antigens). Cell-mediated immunity also can cause Vasculitis Associated with Probable Etiology
vascular inflammation. The pattern of inflammatory injury in giant Hepatitis C virus–associated cryoglobulinemic vasculitis
cell arteritis and in Takayasu arteritis suggests possible cell-mediated Hepatitis B virus–associated vasculitis
immune injury; however, definitive self or non-self targets have not been Syphilis-associated aortitis
conclusively identified. In idiopathic vasculitides, another possibility is an Drug-associated immune complex vasculitis
autoinflammatory process that is mediated by a disturbance in the regula- Drug-associated ANCA-associated vasculitis
Cancer-associated (paraneoplastic) vasculitis
tion of the innate immune response in the absence of a specific antigen
Others
target.
The absence of identifiable etiologies or distinctive pathogenic mecha- From Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised International Chapel Hill
nisms in many forms of vasculitis does not allow diagnosis or classification Consensus Conference Nomenclature of Vasculitides, Arthritis Rheum 65:1–11,
of vasculitides based only on these characteristics. Alternatively, classifica- 2013.
tion and diagnosis is most often based on the presence and absence of
multiple clinical, pathologic, and laboratory features. The most widely
used approach to categorizing vasculitis is based on the nomenclature of as one of the systemic vasculitides (e.g., cutaneous arteritis later becoming
vasculitis proposed by the Chapel Hill Consensus Conferences (CHCC) systemic polyarteritis nodosa). Vasculitis associated with systemic disease
held in 1994 ( Jennette et al, 1994) and modified in 2012 ( Jennette et al, should have a prefix term specifying the systemic disease (e.g., rheumatoid
2013). The CHCC nomenclature provides consensus names (Box 53-2; vasculitis, lupus vasculitis, etc.). Likewise, vasculitis associated with prob-
Fig. 53-1) and definitions for many, but not all, forms of systemic and able etiology should have a prefix term specifying the association (e.g.,
single organ vasculitis. Three major categories are large vessel vasculitis hydralazine-associated microscopic polyangiitis, hepatitis B virus–
(LVV), medium vessel vasculitis (MVV), and small vessel vasculitis (SVV). associated vasculitis, hepatitis C virus–associated cryoglobulinemic vascu-
This terminology can be misleading if it is taken too literally. In general, litis, etc.).
LVV more often affects the aorta and its major branches than any other
form of vasculitis, MVV is confined to arteries, with a preference for major
visceral arteries, and SVV involves not only arteries but also vessels smaller
LARGE VESSEL VASCULITIS
than arteries. Especially with respect to arterial involvement, there is CHCC 2012 defines LVV as vasculitis affecting the aorta and its major
substantial overlap among the categories. For example, although large branches more often than other vasculitides, although any size artery may
vessel involvement is the most specific finding, giant cell arteritis can affect be affected ( Jennette et  al, 2013). In fact, in some patients the number
any size artery from the aorta to the smallest arterial vessels, such as those of medium and small arteries affected is greater than the number of large
in the uveal tract of the eye (Waldman et al, 2013). arteries affected. For example, in giant cell arteritis, very few branches of
In addition to SVV, MVV, and LVV, the 2012 CHCC also recognized the carotid arteries may be affected, although there is involvement of
variable vessel vasculitis (VVV), single organ vasculitis (SOV), vasculitis numerous small arteries extending into the head and neck (Waldman
associated with systemic disease, and vasculitis associated with probable et  al, 2013).
etiology ( Jennette et al, 2013). VV is vasculitis with no predominant type The two major variants of LVV are Takayasu arteritis (TA) and giant
of vessel involved that can affect vessels of any size (small, medium, and cell arteritis (GCA) (Table 53-1). Both have similar pathologic features,
large) and type (arteries, veins, and capillaries), such as Behçet’s disease and so they cannot be distinguished based on pathologic features alone
Cogan’s syndrome. SOV is vasculitis in arteries or veins of any size in a ( Jennette et al, 2013). Demographic and clinical features are required to
single organ that has no features that indicate that it is a limited expression make the distinction, especially age. The differential diagnosis for LVV
of a systemic vasculitis. Some patients originally diagnosed with SOV will includes syphilitic aortitis, which is an infectious vasculitis caused by
develop additional disease manifestations that warrant redefining the case tertiary syphilis, and very rarely by secondary syphilis (Dietrich et al,

1017
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Immune complex small vessel vasculitis
53  Vasculitis
IgA vasculitis
Cryoglobulinemic vasculitis
Hypocomplementemic urticarial vasculitis

Medium vessel vasculitis Anti-GBM disease


Polyarteritis nodosa
Kawasaki disease

Capillary
Venule
Arteriole
Vein
Artery

Artery
Aorta

ANCA-associated small vessel vasculitis


Microscopic polyangiitis
Granulomatosis with polyangiitis
Eosinophilic granulomatosis with polyangiitis

Large vessel vasculitis


Takayasu arteritis
Giant cell arteritis
Figure 53-1  The predominant distribution of vessel involvement by large vessel vasculitis, medium vessel vasculitis,
and small vessel vasculitis. Note that all categories of vasculitis can affect all types of arteries. The aorta and the largest
arteries are most often affected by large vessel vasculitis. Medium vessel vasculitis most often affects medium arteries,
although large and small arteries may be affected. Small vessel vasculitis preferentially affects venules and capillaries, although
arteries and veins may be affected. ANCA (antineutrophil cytoplasmic antibody)–associated vasculitis affects the broadest
spectrum of vessels, whereas immune complex vasculitis usually is restricted to capillaries or venules or both. (Modified from
Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vascu-
litides, Arthritis Rheum 65:1–11, 2013, with permission.)

TABLE 53-1  40 years of age. TAK occurs worldwide with an estimated incidence of 1.2
Names and Definitions for Large Vessel Vasculitis Adopted by to 2.6/million per year in the Western countries. TAK occurs in all races,
the 2011-2012 International Chapel Hill Consensus but the incidence is higher in Southeast Asia, Central and South America,
and Africa (Vaideeswar & Deshpande, 2013).
Conference Nomenclature of the Vasculitides
Large vessel vasculitis Vasculitis affecting large arteries more often
Pathology
(LVV) than other vasculitides. Large arteries are the The pathology of TAK in the acute active phase is characterized by inflam-
aorta and its major branches. Any size artery mation of the aorta or larger arteries with a predominance of mononuclear
may be affected. leukocytes, often, but not always, accompanied by multinucleated giant
Takayasu arteritis Arteritis, often granulomatous, predominantly cells ( Jennette, 2015; Vaideeswar & Deshpande, 2013). Even if disease
affecting the aorta and/or its major branches. appears more localized clinically, postmortem pathologic examination
Onset usually in patients younger than 50. usually shows more extensive involvement. Involved vessels may have well-
Giant cell arteritis Arteritis, often granulomatous, usually affecting demarcated skip areas resulting in localized narrowing or dilation of
the aorta and/or its major branches, with a lumens. This inflammation causes arterial stenosis, thrombosis, and aneu-
predilection for the branches of the carotid rysms. There are at least two classification systems based on distribution
and vertebral arteries. Often involves the of vessel involvement (Vaideeswar & Deshpande, 2013). One classifies
temporal artery. Onset usually in patients TAK into five types: type I, aortic arch and its branches; type II, descending
older than 50 and often associated with thoracic and abdominal aorta; type III, aortic arch and thoraco-abdominal
polymyalgia rheumatica. aorta; type IV, pulmonary artery in addition to the aforementioned types;
and type V, coronary artery in addition to the other types (Gulati & Bagga,
From Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised International Chapel Hill
2010). Another classifies TAK into six types: type I, aortic arch and its
Consensus Conference Nomenclature of Vasculitides, Arthritis Rheum 65:1–11,
2013. branches; type IIa, ascending aorta, arch, and its branches; type IIb,
ascending aorta, arch, and its branches, and descending thoracic aorta;
type III, thoracic aorta and abdominal aorta; type IV, abdominal aorta; and
2014). The resulting aortic inflammation may lead to aneurysms of the type V, combination of type IIb and type IV (Hata et al, 1996).
aorta that can be life-threatening or fatal if not treated appropriately with The pathologic differential diagnosis includes syphilitic aortitis that
antibiotics. also has inflammatory infiltrates with predominantly mononuclear leuko-
cytes, including plasma cells. The only diagnostic pathologic finding that
TAKAYASU ARTERITIS can identify syphilitic aortitis is immunohistochemical identification of
Treponema pallidum.
Definition and Epidemiology
Takayasu arteritis (TAK), is often granulomatous, with a predilection for Etiology and Pathogenesis
the aorta and its major branches ( Jennette et al, 2013). Onset is usually in An autoimmune or infectious etiology that is influenced by genetic and
patients younger than 50. TAK most often affects women between 10 and environmental factors and results in cell-mediated immune injury to

1018
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
vessels is incriminated in the pathogenesis of TAK; however, specific etio-
logic factors have not been identified (Vaideeswar & Deshpande, 2013).
Clinical Laboratory Testing
In part because the specific etiology and unique pathogenic mechanisms
are not known for TAK, no laboratory tests have been identified that are
specific for this disease. Erythrocyte sedimentation rate (ESR) and
C-reactive protein (CRP) are the laboratory tests that are used most often
to support a diagnosis of TAK and to follow disease activity and response
to therapy. The ESR is elevated in 75% to 100% of patients with active
disease and tends to return to normal over time. ESR is thought to be a
reliable index of inflammatory activity and has been used as a tool to
monitor the effectiveness of therapy. Resolution of the disease, both
symptomatically and angiographically, has been found to correlate with
decline of the ESR, although one surgical series observed that 44% of
patients thought to be quiet clinically were not upon arterial biopsy
(Langford, 2003).
In a study of TAK treatment response, ESR decreased from 55.5 +/−
14.7 mm/h to 21.9 +/− 9.5 mm/h within 3 months (p < 0.001) and further
to 20.8 +/− 15.2 at 1 year (p = NS). CRP concentrations fell from 4.8
+/− 5.2 mg/dL to 0.5 +/− 0.2 mg/dL at 3 months (p = 0.004) and remained
at 0.5 +/− 0.3 mg/dL at 1 year (p = NS) (Valsakumar et al, 2003). Figure 53-2  Histopathologic features in the media of the aorta of a patient
In addition to ESR and CRP, multiple biomarkers have been evaluated with giant cell aortitis. Note that the elastic fibers are spread apart by numerous
as diagnostic and prognostic tests in TAK: interleukin (IL)-6, serum infiltrating mononuclear leukocytes with admixed multinucleated giant cells (H&E
stain, ×500).
amyloid A, fibrinogen, complement split fragments, B cell activating
factor (BAFF), interleukin (IL)-12, matrix metalloproteinase 9 (MMP-9),
and pentraxin 3 (PTX3) (Chatterjee et al, 2014). To date, none of these

PART 6
have been adopted for widespread use. leukocytes often accompanied by multinucleated giant cells ( Jennette,
Laboratory testing to rule out syphilitic aortitis includes the VDRL 2015) (Fig. 53-2). The chronic inactive phase has nonspecific chronic
(Venereal Disease Research Laboratory) test, TPPA (Treponema pallidum inflammation and scarring. Although minor focal necrosis may occur,
particle agglutination assay), and IgG and IgM FTA-Abs (fluorescent extensive fibrinoid necrosis, especially in arteries, should raise the possibil-
treponemal antibody-absorption) (Dietrich et al, 2014). A definitive test is ity of another variant of arteritis. The concurrence of another pathologic
positive immunohistochemical staining for Treponema pallidum organisms process that causes chronic inflammation and scarring (e.g., atherosclero-
in tissue specimens (e.g., a resected aneurysm). A negative test is not as sis) complicates the pathologic diagnosis of chronic LVV.
predictive. The most common pathologic pattern in active GCA is transmural
inflammation, which is identified in more than 75% of patients, although
Diagnosis, Treatment, and Prognosis a minority of specimens will have inflammation confined to the adventi-
The diagnosis of TAK is based primarily on clinical signs and symptoms tial tissues, including the vasa vasorum (Cavazza et al, 2014). In a study
and on imaging studies to document involved vessels. Unlike in GCA, of 274 temporal artery biopsies that had transmural inflammation, most
pathologic examination of biopsies or resected vessels is not a routine as a result of GCA, 100% had inflammation with predominantly mono-
component of a diagnostic workup for TAK. TAK has three phases of nuclear leukocytes (lymphocytes, monocytes macrophages, and plasma
progression (Vaideeswar & Deshpande, 2013). The first is characterized cells), 75% giant cells, 8% admixed eosinophils, and 2% admixed neutro-
by low-grade fever, malaise, night sweats, arthralgia, anorexia, and weight phils (Cavazza et al, 2014). Although small zones of laminar degeneration
loss, with no clinical evidence for large vessel narrowing. The second phase (necrosis) may occur in areas of medial inflammation, overt fibrinoid
is the vasculitic stage in which the first-phase constitutional symptoms are necrosis, especially if accompanied by neutrophils and/or neutrophils
accompanied by evidence for large vessel inflammation, such as tenderness with leukocytoclasia, should raise the possibility of a vasculitis other than
or pain over vessels (angiodynia). These first two phases may last for GCA, such as polyarteritis nodosa or ANCA vasculitis (Hamidou et al,
several months before signs of more chronic injury appear. The final phase, 2003; Cavazza et al, 2014).
which may evolve over months or years, has various manifestations of
chronic sclerosing vasculopathy affecting the aorta and its major branches, Etiology and Pathogenesis
including pulseless disease, related to arterial stenosis or occlusion. Cell-mediated immune mechanisms are incriminated in the pathogenesis
In patients with active disease, corticosteroid therapy is the initial treat- of GCA, although the initiating etiologic factors are not known. For
ment of choice. If active disease persists despite corticosteroids, cyclophos- example, in GCA, investigations have demonstrated that dendritic cells in
phamide or methotrexate may be administered. Vasodilators, anticoagulants, vessel walls initiate the recruitment of T cells and macrophages that are
and nonsteroidal anti-inflammatory agents are used for symptomatic relief. the basis for the granulomatous inflammation (Weyand & Goronzy, 2014).
Surgical intervention may be necessary for symptomatic vascular stenosis Two major immune-response networks have been identified in inflamed
or occlusion (Hoffman, 2003). arteries: the interleukin-12, type 1 helper T cell (Th1), and interferon-γ
axis; and the interleukin-6, type 17 helper T cell (Th17), and interleukin-17
GIANT CELL ARTERITIS or interleukin-21 axis. Cytokines released by these inflammatory pathways
activate leukocytes as well as endothelial cells, vascular smooth muscle
Definition and Epidemiology cells, and fibroblasts, leading to intimal and medial hyperplasia and fibrosis.
Giant cell arteritis (GCA) is arteritis, often granulomatous, with a predilec- Elastolytic and proteolytic enzymes (e.g., matrix metalloproteinases) and
tion for the aorta and its major branches ( Jennette et al, 2013). In contrast growth factors (e.g., vascular endothelial growth factor and platelet derived
to TAK, GCA occurs in patients more than 50 years old. GCA is more growth factor) promote both degenerative and proliferative and sclerosing
common in Western countries with the highest incidence and prevalence changes that cause pathologic damage in vessel walls (Weyand & Goronzy,
in Caucasians of Scandinavian descent (González-Gay & Pina, 2015). 2014). A large-scale genetic analysis comprising 1651 patients with GCA
There is a north-south gradient in the incidence of GCA in Europe, with and 15,306 unrelated control subjects from six different European coun-
high rates in Sweden and Norway, which has an incidence of over 20 new tries of European ancestry identified multiple HLA loci, PTPN22, and
cases per 100,000 individuals aged 50 and older per year, and a lower other loci that influence Th1, Th17, and Treg cell function as risk factors
incidence in southern Europe, which has an annual incidence of 10 cases for GCA (Carmona et al, 2015). The involvement of these genes supports
per 100,000 people aged 50 years and older in northwestern Spain a role for a pathogenic autoimmune response in the pathogenesis of GCA.
(González-Gay & Pina, 2015).
Clinical Laboratory Testing
Pathology Temporal artery biopsy is the current gold standard for diagnosis of GCA;
The pathology of GCA in the acute active phase is characterized by inflam- however, a biopsy that is not considered diagnostic does not exclude GCA.
mation of the aorta or larger arteries, with a predominance of mononuclear False-negative results are estimated to occur in up to 15% of patients and

1019
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
are influenced by the length of the biopsy specimen and the duration of TABLE 53-2
53  Vasculitis
corticosteroid treatment prior to biopsy (Achkar et al, 1994; Poller et al, Names and Definitions for Medium Vessel Vasculitis Adopted
2000; Kermani et al, 2013). by the 2011-2012 International Chapel Hill Consensus
Especially in the absence of a tissue diagnosis, laboratory data are very Conference Nomenclature of the Vasculitides
important in reaching an actionable diagnosis of GCA. Laboratory find-
ings that support a diagnosis of GCA are increased erythrocyte sedimenta- Medium vessel Vasculitis predominantly affecting medium arteries
tion rate (ESR) and C-reactive protein (CRP), and thrombocytosis vasculitis (MVV) defined as the main visceral arteries and their
(Kermani et al, 2012; Waldman et al, 2013). An elevated platelet count is branches. Any size artery may be affected.
an early laboratory marker that can be detected a year before GCA is Inflammatory aneurysms and stenoses are
diagnosed (Lincoff et al, 2000). In a study of patients with GCA confirmed common.
by temporal artery biopsy, elevated CRP and elevated ESR had a sensitivity Polyarteritis nodosa Necrotizing arteritis of medium or small arteries
of 87% and 84%, respectively, and 4% had a normal ESR and CRP without glomerulonephritis or vasculitis in
(Kermani et al, 2012). ACR-EULAR Classification Criteria for GCA arterioles, capillaries, or venules, and not
(Dasgupta et al, 2012) require, in part, abnormal elevations in CRP, ESR, associated with ANCA.
or both. An additional laboratory criterion is absence of rheumatoid factor Kawasaki disease Arteritis associated with the mucocutaneous lymph
or antibodies to cyclic citrullinated peptides. node syndrome and predominantly affecting
medium and small arteries. Coronary arteries are
Diagnosis, Treatment, and Prognosis often involved. Aorta and large arteries may be
The diagnosis of GCA is most accurate when it is to be based on clinical involved. Usually occurs in infants and young
manifestations, laboratory tests, and confirmation by biopsy of the tempo- children.
ral artery or other affected artery (Waldman et al, 2013; Weyand &
From Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised International Chapel Hill
Goronzy, 2014). Bilateral temporal artery biopsies increase diagnostic sen- Consensus Conference Nomenclature of Vasculitides, Arthritis Rheum 65:1–11,
sitivity (Durling et al, 2014). Corticosteroid therapy should not be discon- 2013.
tinued after a negative biopsy result if there is enough evidence to make
the diagnosis. GCA with normal ESR and/or normal CRP levels is rare
but does occur; thus laboratory testing cannot completely rule out GCA
(Kermani et al, 2012).
Imaging studies of large arteries and the aorta—for example, with
positron emission tomography (PET)—are a helpful adjunct to pathology
and laboratory testing for making a diagnosis and assessing the status of
patients with GCA (Prieto-Gonzalez et al, 2014). One-quarter of patients
with GCA have involvement of the aorta and its major branches that can
be detected with imaging studies (Weyand & Goronzy, 2014). PET also
is useful in the early diagnosis of syphilitic aortitis (Treglia et al, 2013).
Most cases of GCA are effectively managed with glucocorticoid mono-
therapy (Weyand & Goronzy, 2014). Disease flares are common when the
glucocorticoids are tapered and often require a short-term increase to
induce and maintain remission. Patients with visual impairment or evidence
of unstable blood supply to the eyes benefit from high doses of intravenous
glucocorticoids. If disease flares, patients may need to be treated with
glucocorticoid-sparing agents, including methotrexate or infliximab.
Corticosteroids are the treatment of choice for TAK but may not be
sufficiently effective 50% of the time. An immunosuppressive regimen of
azathioprine and prednisolone is safe, well tolerated, and effective in ame-
liorating systemic symptoms and laboratory abnormalities in TA, and it
halts the progression in lesions based on angiographic studies, although it
does not seem to lead to complete resolution of arterial lesions (Valsaku-
mar et al, 2003). Other agents may be considered, including cyclophos-
phamide and methotrexate.
The differential diagnosis for LVV also includes variable vessel vascu- Figure 53-3  Necrotizing arteritis caused by polyarteritis nodosa. Note the
litis (VVV), including Behçet’s disease and Cogan’s syndrome, and by replacement of most of the media by deeply acidophilic fibrinoid necrosis and the
infectious vasculitis (e.g., aortitis caused by tertiary syphilis) and lympho- perivascular inflammation with a mixture of polymorphonuclear leukocytes and
mononuclear leukocytes (H&E stain, ×300).
plasmocytic disease (e.g., aortitis as a component of IgG4-related disease)
(Miller & Maleszewski, 2011). There is controversy about whether isolated
aortitis or isolated giant cell arteritis of a visceral artery is a limited expres-
sion of Takayasu arteritis or GCA or a distinct form of single organ vas- associated with ANCA ( Jennette et al, 2013). PAN is a diagnosis of exclu-
culitis. When single organ LVV is detected, follow-up should include sion that is made after omitting many other categories of large, medium,
vigilance for additional organ involvement. and small vessel vasculitis that can affect medium arteries ( Jennette & Falk,
2007). PAN affects patients of all ages, genders, and racial backgrounds,
although it is more common in older adults. The annual incidence ranges
MEDIUM VESSEL VASCULITIS from 0 to 1.6 cases/million in European countries, and its prevalence is
MVV is vasculitis that predominantly affects medium arteries defined as about 31 cases/million (Hernández-Rodríguez et al, 2014). Before vaccina-
the main visceral arteries and their branches; however, any size artery may tion against HBV was available, more than one-third of adults with PAN
be affected ( Jennette et al, 2013). The two major categories are polyar- were infected by HBV; however, currently, less than 5% of patients with
teritis nodosa (PAN) and Kawasaki disease (KD) (Table 53-2). Inflamma- PAN are infected with HBV in developed countries (Mahr et al, 2004).
tory aneurysms and stenoses are common. It is important to note that
medium arteries also can be affected by large vessel vasculitis and small Pathology
vessel vasculitis. Thus multiple clinical, laboratory, and pathologic findings Any type of artery in any organ can be affected; however, main visceral
must be taken into consideration to make an accurate diagnosis of the arteries and their initial branches in the parenchyma are the most frequent
disease that is causing arteritis. targets, such as the major mesenteric arteries, renal arteries, hepatic arter-
ies, and coronary arteries. Epineural arteries of peripheral nerves and
POLYARTERITIS NODOSA subcutaneous arteries of the skin also are common targets. When main
arteries are involved, nodular inflammatory lesions and pseudoaneurysms
Definition and Epidemiology can be observed grossly or with imaging studies (or palpated in the skin).
PAN is necrotizing arteritis of medium or small arteries without glomeru- The inflammation has a predilection for arterial branch points. The his-
lonephritis or vasculitis in arterioles, capillaries, or venules, and it is not topathologic features of PAN (Fig. 53-3) begin as fibrinoid necrosis with

1020
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
are not features of PAN and raise the possibility of a small vessel vasculitis
such as microscopic polyangiitis. Involvement of the gastrointestinal tract,
kidneys, heart, and central nervous system is associated with higher mortal-
ity. Laboratory abnormalities reveal a prominent acute phase response but
are nonspecific. Histologic confirmation of vasculitis in medium-sized
arteries is valuable for diagnosis. Positive pathologic finds are more likely
if biopsies are obtained from symptomatic organs if feasible. Skin or
muscle and nerve are preferred because of higher diagnostic yield and
safety. If biopsies are negative or cannot be obtained, visceral angiography
may reveal multiple micro-aneurysms, supporting the diagnosis of PAN.
Current treatment includes high-dose corticosteroids, which are com-
bined with immunosuppressive agents when critical organ involvement or
life-threatening complications occur. IV pulse cyclophosphamide in the
remission induction phase, later switched to a safer immunosuppressant
for remission maintenance, is a frequently used therapeutic approach
(Hernández-Rodríguez et al, 2014).

KAWASAKI DISEASE
Definition and Epidemiology
Kawasaki disease (KD) is arteritis associated with the mucocutaneous
lymph node syndrome and predominantly affecting medium and small
arteries ( Jennette et al, 2013). Coronary arteries are often involved. Aorta
and large arteries may be involved. KD usually occurs in infants and young
children. Clinical manifestations include persistent fever for a week or
more, erythematous rash, conjunctivitis, oropharyngeal erythema, and
lymphadenopathy. Over 80% of KD cases occur between the ages of 6

PART 6
months and 4 years. The annual incidence varies from approximately 3 per
100,000 children under 5 years of age in South America to 4 in Australia,
8 in the United Kingdom, 4 to 15 in the United States, 20 to 30 in China,
Figure 53-4  Cross section of a kidney from a patient with polyarteritis nodosa 50 in Taiwan, 90 in Korea, and more than 130 in Japan (Burgner &
showing multiple pseudoaneurysms (black arrow) and infarcts (white arrow).
Harnden, 2005). Kawasaki disease is more common in boys, with a
male : female ratio of 1.6 : 1.
A number of epidemiological observations suggest but do not prove an
intense acute neutrophil-rich inflammation, evolve through chronic infectious etiology for KD (Burgner & Harnden, 2005). Geographical
inflammation and fibroplasia, and eventually result in fibrosis that may clustering of KD cases and epidemics has been reported. The onset of KD
completely or partially impede blood flow ( Jennette & Falk, 2007; Jen- has seasonal variations, but the predominant season varies in different
nette, 2015). Acute occlusion causing infarction may result from throm- countries. In the United Kingdom, Australia, and the United States, KD
bosis of arteries at sites of segmental acute inflammation, necrosis, and is most common in the winter and spring; in China, in the spring and
pseudoaneurysm formation (Fig. 53-4). summer; and in Korea, during the summer months.
Etiology and Pathogenesis Pathology
The etiology and pathogenesis of PAN are unknown. A role for infections Active KD arteritis is characterized by segmental necrotizing arteritis (Fig.
has been incriminated. During the 1970s, about half of the patients with 53-5). In the acute phase, in general, KD has more infiltration by mono-
PAN were infected with HBV, and HBe antigen to anti-HBe antibody cytes, whereas PAN has more infiltration by neutrophils ( Jennette & Falk,
seroconversion usually paralleled recovery (Pagnoux et al, 2006). This 2007). Fibrinoid necrosis occurs with both PAN and KD, but it is less
suggested that PAN was caused by hepatitis B virus infection. However, conspicuous in KD. The chronic sclerotic phase is characterized by seg-
the frequency of HBV-associated PAN has declined from 35% in the 1980s mental transmural chronic inflammation and scarring. Special stains for
to less than 5% in the early 2000s (Mahr et al, 2004). Additional infections elastica demonstrate focal disruption of the internal elastica.
that have been incriminated as but not proven to be etiologies for PAN The acute lesion of KD is characterized by transmural necrotizing
include hepatitis C virus (HCV), human immunodeficiency virus (HIV), inflammation that is rich in neutrophils and monocytes (Orenstein et al,
cytomegalovirus, and parvovirus B19 (Hernández-Rodríguez et al, 2014). 2012; Jennette, 2015). Degeneration of artery walls results in aneurysm
Most cases are idiopathic. and pseudoaneurysm formation, with the latter having complete erosion
of the necrotizing process into the perivascular tissues. Thrombosis may
Clinical Laboratory Testing occur at sites of arteritis, especially sites of aneurysm/pseudoaneurysm
Laboratory testing in patients with PAN often demonstrates thrombocy- formation. Acute arteritis wanes by 2 weeks after fever onset, and it is
tosis, low serum albumin, and ESR and CRP. Liver enzymes may be replaced by varying degrees of subacute and chronic inflammation, includ-
abnormal in patients who have viral hepatitis. Testing for HBV, HCV, and ing intimal myofibroblast proliferation (Orenstein et al, 2012; Reindel
HIV in patients with necrotizing arteritis is important because antiviral et al, 2014).
treatment must be added to immunosuppressive treatment for optimum
outcome (Guillevin, 2013). ANCA testing is important to rule out ANCA Etiology and Pathogenesis
vasculitis with a component of necrotizing arteritis that is mimicking PAN. Infection also has been suspected as a cause of KD because concurrent
Other laboratory abnormalities are dictated by the organ systems that are infection with pathogens has been detected by culture, molecular methods,
involved, such as kidneys, pancreas, skeletal muscle, heart, gastrointestinal or serological testing in patients with KD; however, although more than
tract, and so on. 20 different pathogens have been incriminated, none have been proven to
be a direct cause of KD, and many are infectious diseases that are common
Diagnosis, Treatment, and Prognosis in young children (Burgner & Harnden, 2005; Principi et al, 2013). Medi-
Clinical manifestations of PAN are protean and depend on the organs that ation by an immune response to a superantigen (possibly of microbial
are affected (Hernández-Rodríguez et al, 2014). Most patients have non- origin) has been studied extensively. KD shares many clinical features
specific constitutional symptoms, such as fever, malaise, arthralgias, myal- with superantigen-mediated diseases—for example, rash, conjunctivitis,
gias, and weight loss. Peripheral neuropathy occurs in approximately 75% and skin sloughing—and KD has been reported in children with toxic
of patients, and gastrointestinal involvement in approximately half. The shock syndrome, which can be caused by superantigens (Burgner &
major cutaneous manifestations are inflammatory nodules, infarction, and Harnden, 2005). However, there is no proof that superantigens are
livido reticularis. Renal involvement manifests as flank pain and hematuria involved in the pathogenesis of KD. Multiple inflammatory pathways are
and, rarely, as retroperitoneal hemorrhage from rupture of an aneurysm. upregulated in the inflamed arteries of KD, including a pathway that
Greater than 2 g/day proteinuria and hematuria with red blood cell casts involves the lymphocyte activation molecule CD84 (Reindel et al, 2014).

1021
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
TABLE 53-3
53  Vasculitis
Names and Definitions for ANCA-Associated Small Vessel
Vasculitis Adopted by the 2011-2012 International Chapel Hill
Consensus Conference Nomenclature of the Vasculitides
ANCA-associated Necrotizing vasculitis, with few or no immune
vasculitis (AAV) deposits, predominantly affecting small vessels
(i.e., capillaries, venules, arterioles, and small
arteries), associated with MPO-ANCA or
PR3-ANCA. Not all patients have ANCA. Add a
prefix indicating ANCA reactivity—for example,
PR3-ANCA, MPO-ANCA, ANCA-negative.
Microscopic Necrotizing vasculitis, with few or no immune
polyangiitis (MPA) deposits, predominantly affecting small vessels
(i.e., capillaries, venules, or arterioles). Necrotizing
arteritis involving small and medium arteries may
be present. Necrotizing glomerulonephritis is very
common. Pulmonary capillaritis often occurs.
Granulomatous inflammation is absent.
Granulomatosis with Necrotizing granulomatous inflammation usually
polyangiitis involving the upper and lower respiratory tract,
(Wegener’s) (GPA) and necrotizing vasculitis affecting predominantly
small to medium vessels (e.g., capillaries, venules,
arterioles, arteries, and veins). Necrotizing
glomerulonephritis is common.
Eosinophilic Eosinophil-rich and necrotizing granulomatous
granulomatosis inflammation often involving the respiratory tract,
with polyangiitis and necrotizing vasculitis predominantly affecting
(Churg Strauss) small to medium vessels, and associated with
Figure 53-5  Kawasaki disease arteritis showing segmental transmural necro-
tizing inflammation with residual areas of intact muscularis adjacent to intense (EGPA) asthma and eosinophilia. ANCA is more frequent
medial inflammation that is continuous with intimal inflammation and adven- when glomerulonephritis is present.
titial inflammation (H&E stain, ×400).
From Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised International Chapel Hill
Consensus Conference Nomenclature of Vasculitides, Arthritis Rheum 65:1–11,
2013.
CD84 was upregulated 16-fold in acute coronary arteritis caused by KD
and 32-fold in chronic coronary arteritis caused by KD (5 months to years
after onset). CD84 was detected in leukocytes in KD tissues.
pathogen reveals the etiologic infection ( Jain et al, 2007; D’Alessandro
Clinical Laboratory Testing et al, 2014).
Diagnosis of KD is based primarily on clinical signs and symptoms. Non- Based on immunopathologic features, noninfectious SVV is divided
specific laboratory indicators of acute inflammation (i.e., acute phase reac- into ANCA-associated vasculitis with few or no vessel-immune deposits,
tants, including CRP and ESR) are consistently elevated in the acute phase and immune complex SVV with moderate to marked vessel wall deposits
of KD. One study suggests that elevated serum levels of the cardiac natri- of immunoglobulin and/or complement components.
uretic peptide called N-terminal of the prohormone brain natriuretic
peptide (NT-pro-BNP) might be a useful diagnostic tool for KD, espe- ANCA-ASSOCIATED SMALL VESSEL VASCULITIS
cially in patients with cardiac injury and atypical clinical presentations
(Cho et al, 2011). The cutoff level of NT-pro-BNP that best distinguished Definition and Epidemiology
KD from other childhood febrile illnesses was 235.2 pg/mL. The same ANCA-associated vasculitis is necrotizing vasculitis, with few or no
study concluded that serum levels of high-sensitivity CRP are not useful immune deposits, that predominantly affects small vessels (i.e., capillaries,
in distinguishing KD from other childhood febrile illnesses. venules, arterioles, and small arteries) and is associated with myeloper-
oxidase (MPO)–ANCA, or proteinase 3 (PR3)–ANCA ( Jennette et  al,
Diagnosis, Treatment, and Prognosis 2013) (Table 53-3). However, not all patients have MPO-ANCA or PR3-
Features of the mucocutaneous lymph node syndrome, especially in a child ANCA, at least using current analytic methods. The few or no immune
5 years of age or younger, are important for diagnosis of KD, but some deposits distinguishes ANCA vasculitis from immune complex vasculitis
patients present with other KD manifestations before full-blown mucocu- that has moderate to marked vessel wall deposits of immunoglobulin
taneous lymph node syndrome, including signs and symptoms of myocar- and/or complement components ( Jennette et  al, 2013). Because of this
dial ischemia caused by coronary arteritis. Although timely treatment with paucity of immune deposits, ANCA vasculitis is also called pauci-immune
intravenous gamma globulin (IVIG) plus aspirin has profoundly reduced vasculitis.
the frequency and severity of coronary arteritis in KD, approximately 10% ANCA SVV is classified based on clinicopathologic phenotypes and by
to 20% of patients develop clinically significant sequelae of coronary arte- the antigen-specificity of ANCA. The clinicopathologic variants are
ritis (Orenstein et al, 2012). Myocardial infarction occurs in approximately microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA)
2% of KD patients with coronary arteritis (Burgner & Harnden, 2005). (formerly called Wegener’s granulomatosis), eosinophilic granulomatosis
with polyangiitis (EGPA) (formerly called Churg-Strauss syndrome), and
renal limited vasculitis (RLV) ( Jennette et al, 2013). An additional variant
SMALL VESSEL VASCULITIS is drug-induced ANCA vasculitis, which may manifest as MPA, GPA, or
Small vessel vasculitis (SVV) is vasculitis predominantly affecting small RLV (Pendergraft & Niles, 2014). An optimum diagnosis should include
vessels, defined as small intraparenchymal arteries, arterioles, capillaries, both the ANCA serology status and the clinicopathologic phenotype—for
and venules ( Jennette et al, 2013). Medium arteries and veins may be example, MPO-ANCA MPA, PR3-ANCA GPA, ANCA-negative EGPA,
affected. and so on ( Jennette et al, 2013).
As with other vasculitides, an important diagnostic dichotomy is MPA is multiorgan pauci-immune small vessel vasculitis with no
between infectious SVV and immune-mediated SVV. Viral infection and granulomatous inflammation or asthma. MPA often has necrotizing glo-
bacterial infection can mimic immune-mediated SVV, and the diagnostic merulonephritis and pulmonary capillaritis. GPA has necrotizing granu-
distinction between these forms of vasculitis is critically important for lomatous inflammation usually involving the upper and lower respiratory
appropriate treatment with antimicrobial therapy versus immunosuppres- tract, as well as vasculitic lesions indistinguishable from those of MPA.
sive therapy. Detection of the pathogen by culture or by genomic or EGPA has eosinophil-rich and necrotizing granulomatous inflammation
proteomic assays or serologic detection of an immune response to the often involving the respiratory tract, as well as asthma and blood

1022
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
A B

PART 6
C D
Figure 53-6  ANCA disease can cause many forms of vascular injury. These include: A, dermal leukocytoclastic angiitis
with intense neutrophil-rich infiltrates centered in small dermal vessels (H&E stain, ×400); B, hemorrhagic pulmonary alveolar
capillaritis with destruction of alveolar walls by neutrophil-rich inflammation (H&E stain, ×500); C, glomerulonephritis with
fibrinoid necrosis of glomerular capillaries (white arrow) and adjacent cellular crescent formation (black arrow) (Masson tri-
chrome stain, ×500); and D, arteritis with extensive irregular fibrinoid necrosis (Masson trichrome stain, ×400).

eosinophilia. Although there appears to be a prevasculitic phase that is 45 per million older adults (over age 65). Ninety-one percent of the
ANCA-negative, ANCA-positive EGPA patients typically have pauci- patients were MPO-ANCA positive, but none were PR3-ANCA positive,
immune small vessel vasculitis and pauci-immune necrotizing and cres- and none had GPA or EGPA (Fujimoto et al, 2006).
centic glomerulonephritis that is indistinguishable from MPA and GPA.
ANCA vasculitis confined to a single organ may occur. One example is Pathology
ANCA renal-limited vasculitis (RLV) causing pauci-immune necrotizing ANCA vasculitis can affect arteries (usually small arteries), arterioles, capil-
and crescentic glomerulonephritis. laries, venules, and veins (usually small veins). In all organs and in all
The incidence of ANCA vasculitis increases with age, with a peak clinicopathologic variants (e.g., MPA, GPA, EGPA), acute ANCA arteritis
around 60 years old, although children can develop ANCA vasculitis. is characterized by segmental fibrinoid necrosis (Fig. 53-6), and mural
Gender distribution is about equal, with a possible slight male predomi- and perivascular neutrophil-rich infiltration ( Jennette & Falk, 2007;
nance. The incidence and prevalence of ANCA vasculitis varies among the Jennette, 2015). Eosinophils may be numerous, especially in EGPA.
different clinicopathologic phenotypes and among different geographic Within several days, the neutrophil-rich inflammation is replaced by pre-
locations (Watts et al, 2015). Roughly, the incidence of all forms of ANCA dominantly mononuclear leukocytes, with numerous monocytes and mac-
vasculitis is 15 to 20/million, and the prevalence is roughly 50 to 150/ rophages and fewer lymphocytes. Within a week, the active inflammatory
million. There is a higher ratio of GPA : MPA in northern Europe (e.g., process wanes and vasculitic lesions develop progressive sclerosis/fibrosis.
Germany and Scandinavia) and northern U.S. states (e.g., Montana and In a given tissue, such as skin, gut, or kidney, vasculitic lesions of different
Minnesota), and a lower ratio of GPA : MPA in southern Europe (e.g., ages often are observed because persistently active ANCA vasculitis has
Greece and Spain) and southern U.S. states (e.g., North Carolina). This multiple acute vasculitic lesions developing while prior active lesions are
is accompanied by the expected differences in ratio of PR3:MPO ANCA undergoing scarring. Patients in complete remission should have only
specificity in accord with more frequent PR3-ANCA with GPA and more chronic quiescent lesions. ANCA arteritis is histopathologically indistin-
frequent MPO-ANCA with MPA. The precise ratios are not known, but guishable from other forms of necrotizing arteritis (e.g., PAN).
in general there is a reversal of preponderance of MPO-ANCA MPA When ANCA vasculitis affects venules—for example, in the dermis—
versus PR3-ANCA GPA from north to south in both Europe and North acute lesions have less conspicuous fibrinoid necrosis and more prominent
America. There is a striking preponderance of MPO-ANCA and of MPA leukocytoclasia (Fig. 53-6, A). This pattern of injury may be called leuko-
in Asia. For example, in one study of ANCA-associated vasculitis in Japan, cytoclastic angiitis, and it can also be caused by other forms of SVV,
the estimated annual incidence was 15 per million adults (over age 15) and especially immune complex SVV.

1023
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
53  Vasculitis

A B
Figure 53-7  Pulmonary lesions in GPA and EGPA include necrotizing granulomatous inflammation and vasculitis.
A, Necrotizing granulomatous inflammation in a GPA patient showing neutrophil-rich necrotic debris in the upper left with
a marginal zone of macrophages, including a giant cell in the lower right (H&E stain, ×500). B, Necrotizing arteritis in the
lung of a patient with EGPA showing intense eosinophil-rich inflammation (H&E stain, ×400).

Acute pulmonary alveolar capillaritis is characterized by accumulation Clinical observations that support a pathogenic role include the high
of neutrophils in alveolar capillary lumens, destruction of capillary base- frequency of ANCA in the blood, the general correlation of ANCA titers
ment membranes (seen with silver stains), and hemorrhage into alveolar with disease activity (especially if capture assays are used), increased risk
air spaces (Fig. 53-6, B). for disease when there are higher levels of ANCA antigens on the surface
The necrotizing glomerulonephritis that often is a component of of circulating neutrophils, induction of ANCA as well as vasculitis by
ANCA vasculitis and that may occur as renal limited vasculitis is character- certain drugs (e.g., hydralazine, propylthiouracil, penicillamine, minocy-
ized by segmental fibrinoid necrosis with adjacent cellular crescent forma- cline), and the therapeutic efficacy of apheresis and targeted B cell
tion in the acute phase (Fig. 53-6, C) and segmental sclerosis with adjacent therapy (e.g., rituximab).
fibrous crescents in the chronic phase. The pathology of ANCA glomeru- The best experimental evidence is induction of ANCA-mediated pauci-
lonephritis is the same in MPA, GPA, EGPA, and renal limited disease. In immune crescentic glomerulonephritis and small-vessel vasculitis in exper-
addition to necrotizing and crescentic glomerulonephritis, kidney lesions imental animals. The first animal model that was described used intravenous
in ANCA disease also include necrotizing arteritis (most often affecting injection of mouse anti-MPO IgG (derived from immunizing MPO
interlobular and arcuate arteries) (Fig. 53-6, D) and leukocytoclastic med- knockout mice with purified mouse MPO) into wild-type mice, resulting
ullary angiitis affecting the vasa rectae. in the induction of a pauci-immune glomerulonephritis with segmental
Immunohistology demonstrates an absence or paucity of immuno- fibrinoid necrosis and crescent formation that closely resembled human
globulin deposition, which distinguishes ANCA vasculitis from immune ANCA glomerulonephritis (Xiao et al, 2002). Some of the mice that
complex vasculitis. However, most specimens, including glomeruli affected received anti-MPO IgG also developed systemic small-vessel vasculitis,
by ANCA disease, have at least a small amount of immune complex–type including alveolar capillaritis and necrotizing arteritis that was histologi-
staining for immunoglobulins and complement, and a small amount of cally indistinguishable from human ANCA disease.
immune complex–type electron-dense deposits by electron microscopy. Numerous in vitro and in vivo experiments support the hypothesis that
ANCA disease also can occur concurrently with anti-GBM disease or ANCA in the circulation can interact with ANCA antigens (MPO, PR3)
immune complex disease, and in this context, there is substantial immu- on the surface of neutrophils that have been primed by cytokines or
nostaining for immunoglobulins and complement. complement fragments (e.g., C5a) (Jennette & Falk, 2014). ANCA-
The necrotizing granulomatous inflammation of GPA and EGPA activated neutrophils release factors that activate the alternative comple-
begins as a focus of neutrophil-rich inflammation that may resemble a ment pathway, which in turn recruits more neutrophils and primes and
microabscess. EGPA lesions also contain numerous eosinophils ( Jennette, activates neutrophils by engaging the C5a receptors (Xiao et al, 2014).
2011). These lesions may be small and spherical, or they may be large Neutrophils that are activated in the circulation adhere to and penetrate
and irregular, creating so-called geographic necrosis. Within days, the vessel walls and release destructive enzymes and oxygen radicals that cause
focus of acute inflammation is surrounded by a rim of monocytes vascular injury. Hypothetically, primed neutrophils in the extravascular
and macrophages, including multinucleated giant cells (Fig. 53-7, A). The tissues that are activated by ANCA are the cause for the granulomatous
central zone of inflammation becomes completely necrotic and evolves inflammation of GPA and EGPA ( Jennette & Falk, 2014).
from basophilic or eosinophilic cellular fragments (derived primarily from Drugs can induce the development of ANCA, especially MPO-ANCA
degenerated neutrophils and eosinophils) to amorphous necrotic debris. (Pendergraft & Niles, 2014). Hydralazine, minocycline, propylthiouracil,
The adjacent granulomatous inflammation becomes more organized with and levamisole-adulterated cocaine are major causes. A hypothesis has
palisades of elongated macrophages adjacent to the necrosis. Eventually, been proposed to explain the induction of ANCA by hydralazine. Patients
the central zone forms a cavity or becomes fibrotic. Necrotizing granulo- with ANCA vasculitis have circulating neutrophils that aberrantly tran-
matous inflammation occurs most often in the upper and lower respira- scribe and express neutrophil granule proteins, including the MPO and
tory tract, but occasionally in other tissues, such as the nasal tissues, orbit, PR3, because of altered epigenetic regulation of gene expression (McInnis
skin, and kidneys. The differential diagnosis for pulmonary granuloma- et al, 2015). Hydralazine can function as a nonnucleoside DNA methyla-
tous inflammation includes mycobacterial and fungal infectious disease. tion inhibitor, so it can reverse epigenetic silencing of PR3 and MPO,
The presence of pulmonary vasculitis along with the granulomatous which could result in increased expression of both autoantigens and loss
inflammation is more specific evidence for GPA or EPGA. Numerous of tolerance (Pendergraft & Niles, 2014).
eosinophils in the granulomatous inflammation and the vasculitis favor Except for the small minority of cases caused by drugs, the etiology of
EGPA (Fig. 53-7, B). ANCA disease is not known. Various theories postulate the result of infec-
tions (e.g., Staphylococcus aureus), environmental exposures (e.g., silica), or
Etiology and Pathogenesis dysregulated gene expression (e.g., of PR3 antisense peptides) ( Jennette &
Clinical and experimental observations support a direct role of ANCA in Falk, 2014; Kallenberg, 2014). One novel theory posits that a protein that
the pathogenesis of vasculitis ( Jennette & Falk, 2014; Kallenberg, 2014). is complementary to the autoantigen PR3 triggers the PR3-ANCA

1024
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
A B C
Figure 53-8  IIF assay using ethanol-fixed, normal human neutrophils as substrate showing: A, C-ANCA cytoplasmic stain-
ing; B, P-ANCA perinuclear staining; and C, atypical-ANCA showing heaped-up, very perinuclear staining (×1000).

autoimmune response through an anti-idiotypic network (Pendergraft pathogenic and nonpathogenic ANCA may change this in the future
et al, 2004). This complementary protein could be an endogenous protein (Roth et  al, 2013).
resulting from age-related dysregulation of antisense gene expression or First-generation ANCA assays were direct noncompetitive enzyme-

PART 6
an exogenous protein derived from a pathogen that makes an antisense linked immunosorbent assay (ELISA). The second-generation tests
peptide mimic that is an inhibitor of the antimicrobial function of comple- (capture ELISA) or third-generation tests (anchor ELISA) are more sensi-
mentary PR3. tive and specific for ANCA testing (Cohen Tervaert & Damoiseaux, 2012).
Fourth-generation assays may be epitope specific and more effective in
Clinical Laboratory Testing guiding therapy (Roth et al, 2013).
In 1999, an International Consensus Statement on Testing and Reporting Chemiluminescence assays (CIAs) have been used to detect PR3-
of Antineutrophil Cytoplasmic Antibodies recommended that testing in ANCA and MPO-ANCA with comparable results to ELISAs (Mahler
new patients should be by indirect immunofluorescence (IIF) assay and et al, 2012). A combination of cell- and microbead-based digital indirect
that samples that are positive by IIF should be tested by ELISAs for PR3- immunofluorescence analysis of ANCA in one reaction environment by
ANCA and MPO-ANCA (Savige et al, 1999). The recommendation for the novel multiplexing bead technology for simultaneous screening and
optimum testing of patients suspected of having ANCA disease was to test confirmatory ANCA testing has been developed (Sowa et al, 2014). ANCA
by ELISA for PR3-ANCA and MPO-ANCA on all serum samples irre- results by classical testing (IIF and ELISA) compared with multiplex bead
spective of IIF results. Although immunoassays have become more effec- analysis were very good (κ = 0.831) with no significant difference (p =
tive since this recommendation was made, most laboratories continue to 0.735). Automated ANCA titer detection in one dilution demonstrated
employ both IIF and antigen-specific immunoassays for MPO and PR3 very good agreement with classical analysis requiring dilution of samples
for ANCA testing. An automated ANCA-pattern recognition system (κ = 0.985).
(Damoiseaux et al, 2012) and a digital indirect immunofluorescence assay Even using current ANCA testing capabilities, patients with clinical
(Sowa et al, 2014) have been evaluated as alternatives to IIF, but they are features and pathologic findings consistent with ANCA vasculitis can test
unlikely to replace conventional IIF microscopy assays anytime soon. negative for ANCA. In a study of 539 patients with a clinical diagnosis of
IIF assay using ethanol-fixed, normal human neutrophils as substrate ANCA vasculitis, positive ANCA serology was demonstrated in 98% of
produces two different staining patterns: cytoplasmic (C-ANCA) (Fig. MPA patients, 96% of GPA patients, 94% of renal-limited pauci-immune
53-8, A) and perinuclear (P-ANCA) (Fig. 53-8, B) (Savige et al, 2000). The crescentic glomerulonephritis patients, and 64% of EGPA patients (Kal-
perinuclear staining is an artifact of substrate preparation that results from lenberg, 2014). Of the 364 patients with GPA in this analysis, 12 tested
diffusion of the cationic MPO antigen from the cytoplasm to the nucleus negative for ANCA, but 10 had GPA limited to the ear, nose, and throat.
during substrate preparation. This perinuclear relocation does not occur EGPA patients usually have P-ANCA with MPO-ANCA specificity
when formalin is used to fix plasma protein in place before preparing (Sinico et al, 2005). EGPA patients with a positive ANCA have more
neutrophils for IIF. Thus a true P-ANCA will produce cytoplasmic stain- glomerulonephritis, pulmonary capillaritis, peripheral neuropathy, and
ing on formalin-fixed neutrophils, whereas an antinuclear antibody will biopsy-proven vasculitis, whereas patients with a negative ANCA have
stain the nucleus in both ethanol- and formalin-fixed preparations (Falk & more myocarditis (Sablé-Fourtassou et al, 2005; Sinico et al, 2005). All
Jennette, 1988). EGPA patients who have biopsy-proven pauci-immune necrotizing and
When the antigen specificity is determined by immunoassays, PR3- crescentic glomerulonephritis are ANCA positive (Sinico et al, 2006). It
ANCA typically produces the C-ANCA staining pattern by IIF, and MPO- appears that EGPA may have two serologically defined phenotypes:
ANCA typically produces a P-ANCA staining pattern. ANCAs that cause ANCA-positive EGPA with vasculitis and ANCA-negative EGPA without
a P-ANCA pattern in IIF assays are not restricted to MPO and include a vasculitis.
variety of autoantibodies that occur in inflammatory diseases, including ANCA specificity correlates with clinical symptoms. Ninety percent of
ulcerative colitis, sclerosing cholangitis, autoimmune hepatitis, rheuma- patients with ANCA who have destructive upper respiratory tract disease
toid arthritis, and Felty’s syndrome (Savige et al, 2000). Positive ANCA (e.g., saddle-nose deformity) have PR3-ANCA, whereas most patients with
IIF results in serum from patients with inflammatory bowel disease and exclusively or predominantly renal involvement have MPO-ANCA
other nonvasculitic inflammatory diseases may have an atypical pattern (Lionaki et al, 2012). Dual positivity for both MPO-ANCA and PR3-
that shows more intense and heaped-up perinuclear staining (very peri- ANCA is rare except in the setting of drug-induced ANCA (Pendergraft
nuclear staining) (Fig. 53-8, C). Antibodies that produce an atypical ANCA & Niles, 2014).
IIF pattern usually are not specific for MPO or PR3 (Savige et al, 2000). ANCAs are in approximately one third of patients with anti-GBM
Antigen-specific, quantitative immunoassays are the standard of care disease ( Jennette, 2003; Rutgers et al, 2005). The dual anti-GBM and
for diagnosis and some management decisions in patients with ANCA ANCA specificity is the result of two different antibody populations rather
disease. ANCA serotype may be more important than clinicopathologic than one cross-reacting population (Rutgers et al, 2005), and the antigen
classification for predicting outcome, including the likelihood of recur- specificities of anti-GBM antibodies are the same in patients with anti-
rence after induction of remission (Kallenberg, 2014). Detection of GBM antibodies only or combined ANCA and anti-GBM antibodies
ANCA to monitor disease activity and adjust treatment remains contro- (Hellmark et al, 1997). Compared with patients with anti-GBM alone,
versial; however, epitope-specific assays that can distinguish between patients with both autoantibodies are older, often have features of systemic

1025
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
small-vessel vasculitis, and have a better prognosis for renal survival with TABLE 53-4
53  Vasculitis
treatment. Names and Definitions for Immune Complex Small Vessel
The possibility of drug-induced disease should be considered in Vasculitis Adopted by the 2011-2012 International Chapel Hill
patients with ANCA vasculitis. The most common are hydralazine, mino- Consensus Conference Nomenclature of the Vasculitides
cycline, propylthiouracil, and levamisole-adulterated cocaine. If cocaine
abuse is a possibility, the urine should be tested for cocaine, and if positive, Immune complex Vasculitis with moderate to marked vessel wall
should be tested for levamisole (Pendergraft & Niles, 2014). Most drug- vasculitis deposits of immunoglobulin and/or
induced vasculitis is not caused by ANCA. A recent study of 239 patients complement components predominantly
with all forms of drug-associated cutaneous vasculitis demonstrated that affecting small vessels (i.e., capillaries, venules,
the most common laboratory abnormalities were increased ESR (40%) and arterioles, and small arteries).
cryoglobulins (26%), while leukocytosis and positive ANA were found in Glomerulonephritis is frequent.
less than 25% (Ortiz-Sanjuán et al, 2014). Only rare examples of drug- Anti-GBM disease Vasculitis affecting glomerular capillaries,
induced immune complex vasculitis have been described, and most of these pulmonary capillaries, or both, with basement
have been caused by therapeutic agents that are proteins, such as mono- membrane deposition of antibasement
clonal antibodies (Baldo, 2013). membrane autoantibodies. Lung involvement
The diagnostic value in patients with vasculitis of ANCA with other causes pulmonary hemorrhage, and renal
specificities has been investigated; however, to date, the only routine involvement causes glomerulonephritis with
testing that has been adopted is for MPO-ANCA and PR3-ANCA. ANCA necrosis and crescents.
with specificity for lysosomal membrane protein 2 (LAMP2) has been Cryoglobulinemic Vasculitis with cryoglobulin immune deposits
reported to have high sensitivity and specificity for pauci-immune SVV vasculitis affecting small vessels (predominantly
(Kain et al, 2008); however, not all investigators have confirmed this capillaries, venules, or arterioles) and associated
finding (Roth et al, 2012). with cryoglobulins in serum. Skin, glomeruli,
and peripheral nerves are often involved.
Diagnosis, Treatment, and Prognosis IgA vasculitis (Henoch- Vasculitis, with IgA1-dominant immune deposits
The diagnosis of ANCA-associated vasculitides as MPA, GPA, or EGPA Schönlein) affecting small vessels (predominantly
is based on algorithms that have been derived from the Chapel Hill Con- capillaries, venules, or arterioles). Often
sensus Conference definitions (Watts et al, 2007; Abdulkader et al, 2013). involves skin and gut, and frequently causes
The diagnostic evaluation of patients with ANCA disease should include arthritis. Glomerulonephritis indistinguishable
from IgA nephropathy may occur.
both testing for ANCA specificity—that is, for proteinase 3 (PR3-ANCA)
or myeloperoxidase (MPO-ANCA)—as well as classification as MPA, Hypocomplementemic Vasculitis accompanied by urticaria and
GPA, EGPA, or renal-limited vasculitis (RLV). Assessment of the disease urticarial vasculitis hypocomplementemia affecting small vessels
stage and severity also is important for tailoring treatment. (anti-C1q vasculitis) (i.e., capillaries, venules, or arterioles), and
associated with anti-C1q antibodies.
Treatment of ANCA disease begins with induction of remission, fol-
Glomerulonephritis, arthritis, obstructive
lowed by maintenance of remission (Kallenberg, 2014). In patients with
pulmonary disease, and ocular inflammation
active generalized disease, remission is most often induced with an immu-
are common.
nosuppressive agent (e.g., cyclophosphamide or rituximabrituximab) and
high-dose glucocorticoids. Rituximab has been shown to be noninferior to From Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised International Chapel Hill
cyclophosphamide for induction of remission. Patients presenting with Consensus Conference Nomenclature of Vasculitides, Arthritis Rheum 65:1–11,
severe kidney failure (i.e., requiring dialysis or an estimated GFR less than 2013.
15) or diffuse alveolar hemorrhage should receive plasma exchange as well
as the usual induction therapy. Mild localized disease, especially with no
renal or pulmonary disease, may be controlled with methotrexate and
glucocorticoids. When disease is in remission, patients should be transi- Pathology
tioned to oral azathioprine, although the use of rituximab as remission The light microscopic features of anti-GBM glomerulonephritis (GN) are
maintenance therapy is increasing (Kallenberg, 2014). Once a patient is in indistinguishable from ANCA glomerulonephritis, although on average
remission, glucocorticoids should be tapered within 6 months to reduce the extent of injury is more severe in anti-GBM GN compared to ANCA
the likelihood of infectious complications (McGregor et al, 2012). Other GN. For example, at the time of biopsy, anti-GBM GN has an average of
treatment options for refractory disease are IVIg, infliximab, alemtu- 77% crescents compared to 49% for ANCA GN ( Jennette, 2003). Con-
zumab, and deoxyspergualin (Kallenberg, 2014). current anti-GBM plus ANCA GN has an average of 62% crescents. The
Drug-induced ANCA vasculitis may require not only cessation of the activity and chronicity of anti-GBM GN lesions tend to be more synchro-
drug and supportive care but also immunosuppression, plasma exchange nous than the lesions of ANCA GN. That is, anti-GBM lesions are either
in severe cases, and dialysis as needed (Pendergraft & Niles, 2014). mostly acute or mostly chronic, whereas ANCA GN lesions more often
have a mixture of acute and chronic lesions. This is in accord with the
ANTI-GLOMERULAR BASEMENT   clinical and serologic observations that anti-GBM disease and anti-GBM
MEMBRANE DISEASE seropositivity typically occur as one episode with uncommon relapses,
whereas ANCA disease and ANCA seropositivity often are persistent with
Definition and Epidemiology multiple recurrences even after initial induction of remission.
Anti-glomerular basement membrane (anti-GBM) disease is an autoim- Anti-GBM pulmonary disease is characterized by extensive hemor-
mune disease caused by autoantibodies that target the noncollagen domain rhage into the alveolar air spaces but with less overt influx of neutrophils
of the α3 chain of type IV collagen (Cui & Zhao, 2011; Greco et al, 2015). into alveolar capillaries than is observed with ANCA pulmonary capil-
This form of vasculitis can be grouped with immune complex vasculitis laritis. Identification of arteritis in the kidney or lung of a patient with
because it results from immune complexes forming in situ along capillary anti-GBM disease is strong evidence for concurrent anti-GBM plus ANCA
basement membranes (Table 53-4). The glomerular capillaries of the disease.
kidneys and alveolar capillaries of the lungs contain this autoantigen, and The most definitive pathology finding is demonstration by direct
either or both can be injured in anti-GBM disease. When both are immunofluorescence microscopy of linear staining for IgG along glomeru-
involved, the resultant pulmonary-renal syndrome is called Goodpasture lar capillary basement membranes or pulmonary alveolar capillary base-
syndrome. Most patients with anti-GBM disease have rapidly progressive ment membranes (Fig. 53-9). This usually is accompanied by staining for
glomerulonephritis, approximately half have pulmonary disease, and only C3 and other complement components and variable staining for IgM and
a few have lung disease. IgA. This is distinct from the granular staining for immunoglobulin in
The incidence of anti-GBM disease is estimated to be 1 per million vessel walls with immune complex vasculitis or the paucity of immuno-
per year (Cui & Zhao, 2011; Greco et al, 2015). It occurs more commonly globulin staining in vessel walls in ANCA vasculitis ( Jennette et al, 2013).
in Caucasians than African Americans. Age distribution is bimodal, with
the most common ages being between 20 and 30 and between 60 and 70. Etiology and Pathogenesis
The younger patients are predominantly men, and there is a higher fre- Both humoral and cellular autoimmune responses have been incriminated
quency of pulmonary disease. The older patients are predominantly in the pathogenesis of anti-GBM disease (Pusey, 2003; Cui & Zhao, 2011).
women, and there is a lower frequency of pulmonary disease. There is compelling evidence that anti-GBM antibodies are pathogenic,

1026
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
A B
Figure 53-9  Linear immunofluorescence staining for IgG along glomerular basement membranes (A) and pulmonary
alveolar capillary basement membranes (B); demonstration by direct immunofluorescence microscopy (FITC rabbit antihu-
man IgG, ×400).

PART 6
which includes (1) the close association of circulating and basement is the result of two different antibody populations rather than one cross-
membrane–bound anti-GBM antibodies with disease; (2) relatively good reacting population (Rutgers et al, 2005).
correlation of autoantibody levels with activity of disease; (3) clinical
response to therapy that reduces circulating anti-GBM levels, such as Diagnosis, Treatment, and Prognosis
apheresis and immunosuppressive therapy; (4) recurrence of disease in Standard treatment is a combined therapy of plasmapheresis, predniso-
kidney transplants in the presence of residual circulating anti-GBM anti- lone, and cyclophosphamide (Pusey, 2003; Cui & Zhao, 2011). Anecdotal
bodies in the recipient; (5) development of anti-GBM glomerulonephritis reports of success with anti-CD20 targeted B cell therapy have emerged
in patients with hereditary type IV collagen abnormalities who make neph- but are not conclusive. The important determinant for the response to
ritogenic autoantibodies directed against donor GBM; and (6) experimen- therapy and long-term prognosis for anti-GBM disease is early diagnosis
tal animal studies, including direct transfer of glomerulonephritis to to prevent end-stage kidney disease and life-threatening pulmonary hem-
primates by intravenous injection of human anti-GBM antibodies. orrhage. If treatment is started before the serum creatinine is above 5 mg/
Although anti-GBM antibodies appear to be major pathogenic factors, dL, there is a good chance of renal survival. Even in patients with advanced
T cells also play an important role. T cell help is required for class- renal disease, treatment may result in improvement and may also be ben-
switching to produce high-affinity IgG antibodies, and T cell involvement eficial in preventing or ameliorating concurrent pulmonary disease.
is indicated by MHC associations. Patients should not be transplanted until circulating anti-GBM autoanti-
The etiology of anti-GBM disease is not known. One hypothesis pro- bodies can no longer be detected by standard assays.
poses that anti-GBM disease is an autoimmune “conformeropathy” that
involves pathogenic conformational changes in the alpha 3 and alpha 5 CRYOGLOBULINEMIC VASCULITIS
noncollagen domains of type IV collagen, revealing cryptic epitopes that
in turn elicit an autoimmune response (Pedchenko et al, 2010). Definition and Epidemiology
Another hypothesis is that autoimmunity to the alpha 3 chain of type Cryoglobulinemic vasculitis (CV) is vasculitis with cryoglobulin-immune
IV collagen is triggered by “autoantigen complementarity” that begins deposits affecting small vessels (predominantly capillaries, venules, or arte-
with an immune response against a peptide that is antisense or comple- rioles) and is associated with serum cryoglobulins (Tedeschi et al, 2007;
mentary to the autoantigen (Reynolds et al, 2015). Autoantigen comple- Jennette et al, 2013). Skin, glomeruli, and peripheral nerves are often
mentarity also is incriminated as a cause for ANCA disease (Pendergraft involved.
et al, 2004). Cryoglobulins are immunoglobulins that reversibly precipitate in the
cold (Damoiseaux, 2014). Type I cryoglobulins consist of a monoclonal
Clinical Laboratory Testing immunoglobulin, generally either IgM or IgG. Type II cryoglobulins
Anti-GBM assays using ELISAs, chemiluminescence assays, dot blots, and contain a monoclonal immunoglobulin with rheumatoid factor (RF) activ-
Western blots are highly specific (>90%) and sensitive (>95%) for anti- ity and polyclonal immunoglobulins. Most often this is a combination of
GBM disease (Pusey, 2003). Most commercial assays use bovine or sheep a monoclonal IgM with polyclonal IgG. Type III cryoglobulins contain
GBM enriched for the noncollagen domain of the α3 chain of type IV polyclonal immunoglobulins, typically IgM and IgG, with RF activity in
collagen (Pusey, 2003). Positive results can be confirmed by Western blot- the IgM fraction. Types II and III are called mixed cryoglobulins.
ting using collagenase-solubilized human GBM. Indirect immunofluores- Type I cryoglobulins often are associated with B cell lymphoprolifera-
cence microscopy assays using normal human or primate kidney as tive diseases, such as Waldenström’s macroglobulinemia or multiple
substrate were used historically, but they are not sensitive enough for myeloma. Type II cryoglobulins are often associated with HCV infection,
current clinical use. as well as other chronic viral infections and systemic autoimmune diseases.
Rapid screening assays are available for concurrently detecting PR3- Type III cryoglobulins often are associated with chronic viral infections
ANCA, MPO-ANCA, and anti-GBM antibodies in the same assay for and systemic autoimmune diseases and may be a transient state between
rapid diagnosis of patients with clinical evidence for rapidly progressive polyclonal hypergammaglobulinemia and type II cryoglobulinemia
glomerulonephritis, hemorrhagic pulmonary capillaritis, or both, using (Damoiseaux, 2014).
either ELISA (Westman et al, 1997) or dot-blot technology (Rutgers et al,
2005). A rapid, random access chemiluminescence assay (CIA) also has Pathology
been developed to detect anti-GBM antibodies with 95.6% sensitivity and CV usually is confined to capillaries (e.g., glomerular capillaries), venules
99.6% specificity (Mahler et al, 2012). (e.g., dermal venules), and arterioles or very small arteries (e.g., epineural
All patients with anti-GBM disease should be tested for ANCA to rule arteries in peripheral nerves). Involvement of medium arteries is extremely
out concurrent anti-GBM plus ANCA disease, which occurs in approxi- rare. The acute inflammation is neutrophil-rich with leukocytoclasia. The
mately one third of patients with anti-GBM disease ( Jennette, 2003; inflammation may be accompanied by luminal or vessel wall deposits
Rutgers et al, 2005). The double anti-GBM and ANCA reactivity in assays of PAS-positive hyaline material that is a coagulum of cryoglobulins.

1027
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
53  Vasculitis

A B
Figure 53-10  Cryoglobulinemic glomerulonephritis with “hyaline thrombi” seen by light microscopy (A, Masson tri-
chrome stain, ×500) and by direct immunofluorescence microscopy using FITC-labeled rabbit antihuman C3 (B, ×500).

Cryoglobulinemic glomerulonephritis occurs as a renal-limited process or et al, 2011). In addition to the detection of cryoglobulins, at least two
as a component of systemic CV. Cryoglobulinemic glomerulonephritis additional patient-reported symptoms, clinical observations, or laboratory
usually has a membranoproliferative pattern but may have other patterns findings must be documented to make a diagnosis of CV.
of proliferative glomerulonephritis. A useful diagnostic finding is hyaline Antiviral therapy with pegylated interferon-α and ribavirin is recom-
material in glomerular capillary lumens (“hyaline thrombi”) that are com- mended for induction therapy for HCV-CV with mild to moderate disease
posed of cryoglobulins (Fig. 53-10). By immunofluorescence microscopy, severity and activity (Cacoub et al, 2014). In patients with more severe
these deposits contain the immunoglobulins of the cryoglobulins as well disease, an immunosuppressive treatment regimen is required for the
as complement components that have been activated by the cryoglobulin induction phase while awaiting the response to antiviral treatments—for
immune complexes (see Fig. 53-10). example, pegylated interferon-α and ribavirin plus steroids and cyclophos-
phamide or rituximab. If there is life-threatening disease or rapidly pro-
Etiology and Pathogenesis gressive glomerulonephritis, apheresis may be beneficial.
Hepatitis C virus (HCV) is the main etiologic agent of mixed cryoglobu- The 1-year, 3-year, 5-year, and 10-year survival rates for HCV-CV are
linemic vasculitis (Cacoub et al, 2014). In some European settings, mixed 95%, 85%, 75%, and 65%, respectively. Mortality is mainly related to
cryoglobulinemia has been associated with HCV infection in more than infections and end-stage liver disease. Factors associated with a poor prog-
80% of the patients, although only a minority develop signs and symptoms nosis are severe liver fibrosis (HR 5.31), central nervous system involve-
of vasculitis (Pagnoux et al, 2006). Although the association of CV with ment (HR 2.74), kidney involvement (HR 1.91), and heart involvement
HCV is approximately 85% in the Mediterranean region, it varies from (HR 4.2) (Terrier et al, 2011).
30% to 100% in other geographic locations (Damoiseaux, 2014).
Deposition of cryoglobulins in vessel walls incites vasculitis by comple-
ment activation. Monoclonal cryoglobulins (type I) are not as effective at IgA VASCULITIS (HENOCH-SCHÖNLEIN
activating inflammatory mediator systems as are mixed cryoglobulins PURPURA)
(types II and III), which are immune complexes composed of antiantibod-
ies (rheumatoid factors) bound to target antibodies. An important cause Definition and Epidemiology
for cryoglobulinemia is hepatitis C infection. In HCV-CV, immune IgA vasculitis (IgAV) (formerly called Henoch-Schönlein purpura) is vas-
complex deposits contain viral proteins (Damoiseaux, 2014). culitis with IgA1-dominant immune deposits affecting small vessels (pre-
dominantly capillaries, venules, or arterioles) ( Jennette et al, 2013). IgAV
Clinical Laboratory Testing often involves the skin and gastrointestinal tract, and it frequently causes
Clinical laboratory testing for cryoglobulins is divided into three phases: arthritis. Glomerulonephritis indistinguishable from IgA nephropathy may
the preanalytic phase, the analytic phase, and the typing phase (Damoi- occur.
seaux, 2014). The preanalytic phase entails collecting the sample in pre- IgA vasculitis is most common in children, especially children under
warmed (37° C) tubes, transporting at 37° C, allowing coagulation of the 10 years old, which differs from ANCA vasculitis and CV, which are more
blood sample at 37° C for at least 1 hour, and centrifugation at 37° C to common in older patients. In fact, IgAV is the most common systemic
separate the serum. The analytic phase entails incubating two fractions of small-vessel vasculitis in children (Chen & Mao, 2015). Depending on
the sample at 4° C for 3 to 7 days, visual assessment of precipitation, incu- ethnic background, 6 to 24 per 100,000 children younger than 17 years of
bating one fraction at 37° C to assess resolution, and isolating the cryo- age will develop IgAV. In Asia, the incidence is as high as approximately
globulin from the other fraction for typing. The typing phase includes 70 cases per 100,000 children every year. IgAV is rare in adults, with an
determining the immunoglobulin composition (e.g., by immunofixation or annual incidence of 0.1 to 1.8 per 100,000 individuals (Audemard-Verger
immunoelectrophoresis) and quantifying the cryoglobulin by assessing et al, 2015). Disease is more frequent in males (male : female ratio of 5 : 1).
cryocrit, total protein content, and/or content of immunoglobulin isotypes
(Damoiseaux, 2014). Pathology
In addition to a positive cryoglobulin assay, in a patient with clinical The histopathologic features of inflammation in venules, arterioles, and
evidence for SVV, laboratory detection of low C4, rheumatoid factor, small arteries, caused by IgAV, are indistinguishable from other forms of
and evidence for HCV infection add additional support for a diagnosis of SVV. In the dermis, this takes the form of a leukocytoclastic angiitis affect-
CV (Lamprecht et al, 2001). ing predominantly venules and arterioles. The presence of hyaline aggre-
gates in areas of inflammation or the lumen increases the likelihood of CV.
Diagnosis, Treatment, and Prognosis Arteritis affecting arteries in the deep dermis, and especially arteries in
Detection of mixed cryoglobulins, especially accompanied by low C4, the subcutaneous tissue, increases the likelihood of ANCA vasculitis.
along with purpura, peripheral neuropathy, and/or glomerulonephritis, is Numerous eosinophils increase the likelihood of hypersensitivity drug-
strong evidence for CV. induced vasculitis or EGPA. However, it is best to correlate the histopa-
One set of classification criteria for CV requires the detection of cryo- thology with immunostaining for IgA, IgG, IgM, C3, and C1q, and with
globulins in at least two determinations more than 12 weeks apart (DeVita serologic testing for ANCA, cryoglobulins, HCV antigen and antibody,

1028
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
complexes may be of value in monitoring disease activity. Serum levels of
galactose-deficient IgA1 and IgA1-IgG immune complexes are associated
with disease activity of IgA nephropathy (Suzuki et al, 2014).
To date, these research observations on abnormal IgA1 glycosylation
and anti-IgA1 antibodies in IgAV and IgAN have not been translated into
analytically and clinically validated diagnostic tests, although multiple
research laboratories are working on this.
Diagnosis, Treatment, and Prognosis
Until alternative clinical tests are validated, pathologic identification of
IgA-dominant immune deposits in vessel walls or glomeruli by direct
immunofluorescence microscopy is the gold standard method for confirm-
ing a diagnosis of IgAV or IgAN. Immunostaining is required in biopsy
specimens because the light microscopic patterns of injury are not as
conclusive for distinguishing among the many forms of SVV. For example,
a skin biopsy that has leukocytoclastic angiitis by light microscopy could
be found by immunofluorescence microscopy to be IgAV with IgA-
dominant deposits, CV with IgM and C3 deposits, or ANCA vasculitis
with a paucity of immunoglobulin and complement deposits.
The classic triad of symptoms and signs for IgAV are purpura, arthral-
gia, and abdominal pain. The frequency of renal disease varies from 45%
to 85% in different cohorts (Audemard-Verger et al, 2015). The European
League Against Rheumatism (EULAR), the Paediatric Rheumatology
International Trials Organization, and the Paediatric Rheumatology Euro-
pean Society have proposed classification criteria for pediatric patients
Figure 53-11  Direct immunofluorescence microscopy of small vessels in the
upper dermis of a patient with IgA vasculitis demonstrating coarsely granular
(Audemard-Verger et al, 2015). Based on these criteria, a patient is classi-
fied as IgAV if there are purpura with lower-extremity predominance plus

PART 6
deposits of IgA in vessel walls (FITC-labeled rabbit antihuman C3, ×600).
one of the following four criteria: abdominal pain, histopathology demon-
strating IgA-dominant deposits in vessels or glomeruli, arthritis or arthral-
gia, or renal involvement. These criteria have not been adopted for adults
anti-C1q, ANA, C3, and C4 before reaching a diagnosis. Patients with and would be problematic in adults because the prevalence of IgAV is much
IgAV have immune complex deposits in small dermal vessels that stain lower than the prevalence of other forms of SVV that cause similar signs
brightly by direct immunofluorescence microscopy using antibodies spe- and symptoms, especially ANCA disease. For example, a patient who has
cific for IgA (Fig. 53-11). purpura, arthralgias, and nephritis fulfills the EULAR criteria for IgAV.
The glomerulonephritis of IgAV is indistinguishable from IgA Such a patient could also have ANCA vasculitis, cryoglobulinemic vascu-
nephropathy (IgAN), although glomerular necrosis and crescents are litis, or other forms of SVV. Especially if applied to adults, the pediatric
somewhat more frequent in IgA vasculitis. Because glomerulonephritis is EULAR criteria should be combined with testing for ANCA, cryoglobu-
a frequent component of many forms of immune complex SVV, immuno- lins, HCV infection, and hypocomplementemia.
pathologic features are very helpful in distinguishing among different Because the clinical course of IgAV is relatively benign in most patients,
forms of SVV—for example, IgAV with IgA1-dominant glomerular depos- treatment is usually conservative and directed at symptom relief. Patients
its, CV with membranoproliferative lesions and IgG/IgM deposits, anti- who develop severe gastrointestinal complications may require steroids or/
GBM disease with necrosis and linear IgG, and ANCA vasculitis with and immunosuppressive drugs. When kidney disease develops with IgAV,
necrosis and a paucity of immunoglobulin deposits in glomeruli. the following Kidney Disease Improving Global Outcome (KDIGO)
guidelines can be used (Chen & Mao, 2015). Persistent proteinuria should
Etiology and Pathogenesis be treated with angiotensin-converting enzyme inhibitors or angiotensin
Patients with IgAV and IgAN have increased serum levels of IgA1 with receptor blockers. If the proteinuria persists and the glomerular filtration
abnormal glycosylation of the hinge region (Suzuki et al, 2009, 2011; rate is greater than 50 mL/min per 1.73 m2, a 6-month course of cortico-
Mestecky et al, 2013). This abnormality is caused by combined genetic and steroid therapy should be given. If severe renal failure and crescentic
environmental/infectious influences. There is strong evidence that IgAV glomerulonephritis are documented, immunosuppressive therapy similar
and IgAN are caused by vessel wall deposition of aggregates of abnormally to that used for other forms of crescentic glomerulonephritis should be
glycosylated IgA1 molecules or by deposition of immune complexes com- provided.
posed of IgA1 complexed to anti-IgA1 IgG that recognizes the abnormal
hinge region glycosylation (Suzuki et al, 2009, 2011; Mestecky et al, 2013).
Abnormally glycosylated IgA1 molecules also could induce disease HYPOCOMPLEMENTEMIC  
through a variety of other mechanisms, such as reduced clearance from URTICARIAL VASCULITIS
the circulation because of lack of Fc receptor engagement by the abnormal
IgA, increased self-aggregation of IgA1 in the circulation resulting in Definition and Epidemiology
deposition in vessel lumens and vessel walls, and increased affinity of the Hypocomplementemic urticarial vasculitis (HUV) (anti-C1q vasculitis) is
abnormal IgA1 for perivascular and glomerular mesangial matrix. vasculitis accompanied by urticaria and hypocomplementemia affecting
Vascular deposits containing IgA1 appear to mediate inflammation by small vessels (i.e., capillaries, venules, or arterioles), and it is often associ-
activating complement via the lectin pathway and the alternative pathway. ated with anti-C1q antibodies ( Jennette et al, 2013). Glomerulonephritis,
arthritis, obstructive pulmonary disease, and ocular inflammation are com-
Clinical Laboratory Testing monly present in HUV, and HUV has overlapping clinical and laboratory
Abnormally glycosylated IgA1 has been detected in serum using lectin- features with SLE, though it also occurs independently of SLE (Venzor
binding assays and proteomic mass spectroscopy analysis. For example, a et al, 2002; Kallenberg, 2008; Grotz et al, 2009).
lectin that recognizes N-acetylgalactosamine was used in an ELISA to HUV is a rare disease. The incidence of all forms of leukocytoclastic
measure serum galactose-deficient IgA1 (Moldoveanu et al, 2007). This vasculitis affecting the skin in a population-based study in Minnesota from
study concluded that the assay for abnormal IgA1 had diagnostic sensitivity 1996 to 2010 identified an incidence rate of 4.5 per 100,000 person-years
of 76.5%, specificity of 94%, positive predictive value of 88.6%, and nega- (Arora et al, 2014). The cohort included 38 patients with leukocytoclastic
tive predictive value of 78.9% for IgAN. vasculitis not otherwise specified (45%), 25 with IgA vasculitis (30%), 10
Anti-IgA antibodies with specificity for the IgA1 hinge region have with urticarial vasculitis (12%), 8 with ANCA vasculitis (10%), and 3 with
been detected in serum using monoclonal human IgA1 as substrate and by cryoglobulinemic vasculitis (4%) (Arora et al, 2014). Thus HUV has an
identifying IgA1 in circulating immune complexes. One study concluded incidence of less than 0.5 per 100,000 person-years in this population.
that IgA1 glycan-specific IgG antibodies have 88% specificity and 95% HUV affects women more frequently than men (ratio 2 : 1) and has a
sensitivity for IgA nephropathy (Suzuki, 2009). In addition to diagnosis, peak incidence in the fifth decade of life, although HUV has been observed
serologic tests for abnormally glycosylated IgA1 and IgA1 immune in children (Grotz et al, 2009).

1029
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
cytoclastic vasculitis, arthralgia and arthritis, uveitis or episcleritis,
53  Vasculitis
Pathology glomerulonephritis, and abdominal pain (Grotz et al, 2009).
The dermal vasculitis in HUV is a leukocytoclastic angiitis that cannot be Glucocorticoids are standard therapy for HUV. Depending on clinical
readily distinguished from other forms of SVV by histology alone. Direct severity, additional immunosuppressive agents such as methotrexate, aza-
immunofluorescence microscopy demonstrates complement-rich immune thioprine, cyclophosphamide, cyclosporine, and mycophenolate mofetil
complex deposits with prominent C1q in dermal vessels and along the may be added. Apheresis is reserved for very aggressive disease (Grotz et al,
dermal-epidermal junction. 2009).
By immunofluorescence microscopy, glomerulonephritis in HUV has
extensive capillary wall and mesangial immune complex deposits contain- RHEUMATOID VASCULITIS
ing immunoglobulin and complement, including prominent C1q. The
most frequent histologic pattern is membranoproliferative glomerulone- Definition and Epidemiology
phritis with thickened capillary walls and endocapillary hypercellularity, Rheumatoid vasculitis is an uncommon extra-articular complication of
although focal or diffuse proliferative glomerulonephritis may also occur. rheumatoid arthritis (RA) and primarily affects small to medium vessels
In this respect, HUV glomerulonephritis resembles lupus nephritis, and (Ntatsaki et al, 2014; Makol et al, 2015). There has been a decrease in the
patients with HUV who also meet clinical criteria for SLE should be incidence of rheumatoid vasculitis in the last decade in both Europe and
considered to have lupus nephritis. the United States (Ntatsaki et al, 2014). The average annual incidence of
rheumatoid vasculitis in the United Kingdom declined from 9.1 per
Etiology and Pathogenesis million between 1988 and 2002 to 3.9 per million between 2001 and 2010
Experimental studies strongly support a pathogenic role for anti-C1q anti- (Ntatsaki et al, 2014). This reduction may be the result of the use of
bodies in forming pathogenic immune complexes in SLE and HUV, and current disease-modifying antirheumatic drugs (DMARDs), including
anti-C1q antibodies may interfere with the clearance of apoptotic cells and methotrexate.
enhance induction of autoimmunity (Kallenberg, 2008). The hypocomple-
mentemia appears to be secondary to classical complement pathway activa- Pathology
tion with reduced C1, C2, C4, and C3 (Kallenberg, 2008). The extensive Rheumatoid vasculitis involves small and medium vessels, including
deposition of immune complexes in the walls of inflamed vessels—for medium and small arteries and venules. It is a necrotizing arteritis that is
example, dermal venules and glomerular capillaries—supports an immune histologically indistinguishable from other forms of necrotizing arteritis,
complex pathogenesis for the inflammation. such as PAN and MPA. Its venulitis, especially in the dermis, is histologi-
cally indistinguishable from the leukocytoclastic angiitis of ANCA or
Clinical Laboratory Testing immune complex disease. In patients with rheumatoid vasculitis, dermal
The typical laboratory test profile is elevated anti-C1q antibodies (essen- necrotizing venulitis has been identified as the cause of palpable purpura,
tially 100% sensitive) and hypocomplementemia with low C1q, C3, and maculopapular erythema, erythema elevatum diutinum, and hemorrhagic
C4. In patients who have HUV without overt SLE, ANA often is present blisters; arteritis has been identified as the cause of subcutaneous nodules,
without anti–double-stranded DNA. Positive testing for cryoglobulins livedo reticularis, and deep ulcers (Chen et al, 2002). Some patients have
supports a diagnosis of CV rather than HUV. both patterns of vasculitis.
C1q has a collagen-like portion attached to globular heads, resembling
a bouquet of tulips (Venzor et al, 2002; Kallenberg, 2008). Immune Etiology and Pathogenesis
complex Fc regions bind to these globular portions, whereas anti-C1q There is a strong association between rheumatoid vasculitis and particular
autoantibodies instead bind to the collagen-like portion. Anti-C1q usually HLA genotypes that suggests a role for immune recognition of antigens
is detected by ELISA using human C1q as the substrate. High ionic in its pathogenesis. A role for immune complexes containing autoantibod-
strength conditions (0.5 to 1.0 M NaCl) are used to inhibit binding of ies has been proposed based on the observed high prevalence and high
immune complexes (Kohro-Kawata et al, 2002). Anti-C1q is detected in a titer of rheumatoid factor (RF), anticitrullinated peptide antibodies
variety of inflammatory diseases, including HUV (100%), rheumatoid (ACPA), antinuclear antibodies, and circulating or tissue-deposited
vasculitis (77%), Felty syndrome (76%), lupus nephritis (63%), membra- immune complexes in patients with rheumatoid vasculitis (Makol et al,
noproliferative glomerulonephritis (54%), and SLE without nephritis 2015). However, many RA patients with high titers of RF and ACPA do
(33%) (Seelen et al, 2003; Kallenberg, 2008). HUV patients who do not not develop rheumatoid vasculitis. Immunoglobulin and complement have
have overt SLE may have low-titer antinuclear antibodies, rheumatoid been detected by immunofluorescence in dermal vessels of patients with
factor, lupus anticoagulant, and cryoglobulins. dermal rheumatoid vasculitis (Chen et al, 2002).
While the presence of anti-C1q antibodies is highly sensitive for HUV
(found in 100% of patients), they are neither sensitive nor specific for SLE. Clinical Laboratory Testing
Anti-C1q antibodies are more prevalent in SLE patients with lupus nephri- Rheumatoid vasculitis should be suspected in a patient with clinical or
tis than in patients without nephritis. In a study of 43 patients with SLE, pathologic evidence for vasculitis who has rheumatoid factor (RF), anti­
anti-C1q antibodies were identified in 82% of patients with nephritis, citrullinated peptide antibodies (ACPA), antinuclear antibodies, and circu-
compared to only 38% of patients who did not have nephritis (Coremans lating or tissue-deposited immune complexes (Makol et al, 2015). Less
et al, 1995). In a study of 228 patients with lupus nephritis, the evaluation specific features that are often observed include anemia, leukocytosis,
of anti-C1q, C3, and C4 provided the best prediction of renal flares (Sinico thrombocytosis, elevated erythrocyte sedimentation rate, increased
et al, 2009). However, some SLE patients with high titers of anti-C1q C-reactive protein, and polyclonal hypergammaglobulinemia. The absence
antibodies do not develop nephritis. of RF and ACPA has a high negative predictive value for rheumatoid
vasculitis (Makol et al, 2015).
Diagnosis, Treatment, and Prognosis In a study of 19 patients with rheumatoid vasculitis, all of the patients
HUV is characterized by recurrent episodes of urticaria with underlying were seropositive for RF, no patients were C-ANCA positive, 5 patients
leukocytoclastic vasculitis. Skin manifestations include painful, tender, were P-ANCA positive by IIF but had negative ELISA for MPO-ANCA,
burning, or pruritic urticarial skin lesions that persist for greater than 1 patient with P-ANCA was positive for PR3-ANCA by ELISA, all of the
24 hours, often with central areas of hemorrhage. Once the urticaria patients were negative for ANA, and 1 patient was positive for antimito-
resolves, there may be residual purpura. Additional manifestations chondrial antibodies (Ntatsaki et al, 2014). Anti-C1q antibody testing is
include angioedema, laryngeal edema, pulmonary involvement, arthritis, positive in approximately 75% of patients with rheumatoid vasculitis
arthralgia, glomerulonephritis, and uveitis. Compared to patients with (Seelen et al, 2003; Kallenberg, 2008). This raises the possibility of a
urticaria and vasculitis who do not have hypocomplementemia, HUV relationship with HUV, but anti-C1q positive patients with rheumatoid
patients are more likely to have abdominal pain and chronic obstructive vasculitis typically do not have other features of HUV, including no hypo-
pulmonary disease (Mehregan et  al, 1992). Chronic obstructive pulmo- complementemia, or urticarial or immune complex glomerulonephritis.
nary disease and acute laryngeal edema are important causes for morbid-
ity and mortality. Around 30% of HUV patients suffer gastrointestinal Diagnosis, Treatment, and Prognosis
symptoms: pain, nausea, vomiting, diarrhea, and sometimes ascites in The Scott and Bacon diagnostic criteria for rheumatoid vasculitis require
connection with serositis, hepatomegaly, and splenomegaly (Grotz et  al, the presence of one or more of the following in a patient with RA: mono-
2009). neuritis multiplex or peripheral neuropathy, peripheral gangrene, biopsy
The two major diagnostic criteria are chronic urticaria and hypo- evidence of acute necrotizing arteritis plus systemic illness (e.g., fever and
complementemia. Minor criteria include anti-C1q antibodies, leuko­ weight loss), deep cutaneous ulcers or extra-articular disease (e.g., pleurisy,

1030
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
pericarditis, scleritis) if associated with typical digital infarcts, or biopsy patient demographics, clinical signs and symptoms, laboratory tests, and
evidence of vasculitis (Scott & Bacon, 1984). pathologic findings. Positive as well as negative laboratory test results are
Hydroxychloroquine and low-dose aspirin are associated with lower critically important in the diagnostic evaluation of patients with suspected
odds of developing rheumatoid vasculitis among RA patients, suggesting vasculitis, and they are useful for prognostication, predicting and monitor-
a possible protective effect (Makol et al, 2015). Among 86 patients treated ing response to therapy, and predicting or detecting relapse. Over the past
for rheumatoid vasculitis at Mayo Clinic between 2000 and 2010, 99% 20 years, there has been a valuable increase in the menu of laboratory tests
received glucocorticoids, 29% received cyclophosphamide, 55% received that facilitate the care of patients with vasculitis (e.g., ANCA). More are
another disease-modifying antirheumatic drug (methotrexate, azathio- currently on the verge of entering clinical practice (e.g., IgA1 glycosylation
prine, mycophenolate mofetil, hydroxychloroquine, minocycline), and and anti-IgA1 antibody assays), and undoubtedly more will be discovered
28% received a biologic agent (anti-TNF, rituximab, abatacept, anakinra) before the next edition of this text is published.
(Makol et al, 2014). However, despite aggressive use of cyclophosphamide
and biologics, rheumatoid vasculitis remained difficult to treat, with high REFERENCES
relapse rates and mortality rates of 12% at 1 year and 60% at 5 years.
Access the complete reference list online at ExpertConsult.com.

SUMMARY
The diagnosis of vasculitis is difficult. Accurate and precise diagnosis
requires knowledgeable integration of evidence from the medical history,

SELECTED REFERENCES
Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised Makol A, Matteson EL, Warrington KJ: Rheumatoid Pagnoux C, Cohen P, Guillevin L: Vasculitides second-
International Chapel Hill Consensus Conference vasculitis: An update, Curr Opin Rheumatol 27:63–70, ary to infections, Clin Exp Rheumatol 24(2 Suppl
Nomenclature of Vasculitides, Arthritis Rheum 65:1– 2015. 41)):S71–S81, 2006.
11, 2013. Although rheumatoid vasculitis is a rare extra-articular Recognition of infectious vasculitides is critically impor-
This excellent comprehensive review of giant cell arteritis manifestation of rheumatoid arthritis, it has high morbid- tant because appropriate therapy can be very effective, and
and Takayasu arteritis is by one of the major leaders in the ity and mortality when it occurs. This timely update pre­ failure to make a correct diagnosis can be devastating. These
field of large vessel vasculitis. Epidemiology, pathology, sents the clinical manifestations, management, and distinguished authors provide an excellent review of the

PART 6
putative pathogenesis, and clinical management are outcomes of rheumatoid vasculitis in the current era of variety of infectious pathogens that can cause vasculitis by
reviewed in depth with expert commentary. disease-modifying antirheumatic drugs (DMARDS). multiple pathogenic mechanisms and summarize approaches
Kallenberg CG: Key advances in the clinical approach Mestecky J, Raska M, Julian BA, et al: IgA nephropathy: for diagnosis and treatment.
to ANCA-associated vasculitis, Nat Rev Rheumatol Molecular mechanisms of the disease, Annu Rev Pathol Pusey CD: Anti-glomerular basement membrane
10:484–493, 2014. Mech Dis 8:217–240, 2013. disease, Kidney Int 64:1535–1550, 2003.
This insightful review of ANCA vasculitis covers This authoritative detailed review describes the discovery This article is written by the preeminent physician scien-
pathogenesis, clinical presentations, development of of abnormal glycosylation of IgA1 in IgAV and IgAN, tist who has been at the forefront of clinical and basic
diagnostic and classification criteria, and advances in which is one of the most impactful advances in the under- research on anti-GBM disease. It covers pathobiology and
therapy. This expert overview is informed by Professor standing of the pathogenesis of vasculitis that has emerged clinical aspects of this aggressive category of immune-medi-
Kallenberg’s participation in many important studies on over the past 10 or more years. This discovery not only has ated vascular injury. This is a rare disease, but it illustrates
the pathogenesis and clinical management of ANCA elucidated the pathobiology of these diseases, but it also sets important concepts of autoimmune vascular inflammation
vasculitis. the stage for valuable new clinical laboratory tests. and strategies for their treatment.

1031
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
REFERENCES
Abdulkader R, Lane SE, Scott DG, et al: Classification Miyazaki Prefecture: The first population-based, ret- Mahler M, Radice A, Sinico RA, et al: Performance
of vasculitis: EMA classification using CHCC 2012 rospective, epidemiologic survey in Japan, Clin J Am evaluation of a novel chemiluminescence assay for
definitions, Ann Rheum Dis 72:1888, 2013. Soc Nephrol 1(5):1016–1022, 2006. detection of anti-GBM antibodies: An international
Achkar AA, Lie JT, Hunder GG, et al: How does previ- González-Gay MA, Pina T: Giant cell arteritis and poly- multicenter study, Nephrol Dial Transplant 27(1):243–
ous corticosteroid treatment affect the biopsy findings myalgia rheumatica: An update, Curr Rheumatol Rep 252, 2012.
in giant cell (temporal) arteritis?, Ann Intern Med 17(2):6, 2015. Mahr A, Guillevin L, Poissonnet M, et al: Prevalences
120:987–992, 1994. Greco A, Rizzo MI, De Virgilio A, et al: Goodpasture’s of polyarteritis nodosa, microscopic polyangiitis,
Arora A, Wetter DA, Gonzalez-Santiago TM, et al: syndrome: A clinical update, Autoimmun Rev Wegener’s granulomatosis, and Churg-Strauss syn-
Incidence of leukocytoclastic vasculitis, 1996 to 2010: 14(3):246–253, 2015. drome in a French urban multiethnic population in
A population-based study in Olmsted County, Min- Grotz W, Baba HA, Becker JU, et al: Hypocomplement- 2000: A capture-recapture estimate, Arthritis Rheum
nesota, Mayo Clin Proc 89(11):1515–1524, 2014. emic urticarial vasculitis syndrome: An interdisciplin- 51:92–99, 2004.
Audemard-Verger A, Pillebout E, Guillevin L, et al: IgA ary challenge, Dtsch Arztebl Int 106(46):756–763, Makol A, Crowson CS, Wetter DA, et al: Vasculitis asso-
vasculitis (Henoch-Schönlein purpura) in adults: 2009. ciated with rheumatoid arthritis: A case-control study,
Diagnostic and therapeutic aspects, Autoimmun Rev Guillevin L: Infections in vasculitis, Best Pract Res Clin Rheumatology (Oxford) 53:890–899, 2014.
14(7):579–585, 2015. Rheumatol 27:19–31, 2013. McGregor JG, Hogan SL, Hu Y, et al: Glucocorticoids
Baldo BA: Adverse events to monoclonal antibodies Gulati A, Bagga A: Large vessel vasculitides, Pediatr and relapse and infection rates in anti-neutrophil cyto-
used for cancer therapy: Focus on hypersensitivity Nephrol 25:1037–1048, 2010. plasmic antibody disease, Clin J Am Soc Nephrol 7:240–
responses, Oncoimmunology 2(10):e26333, 2013. Hamidou MA, Moreau A, Toquet C, et al: Temporal 247, 2012.
Burgner D, Harnden A: Kawasaki disease: What is the arteritis associated with systemic necrotizing vasculi- McInnis EA, Badhwar AK, Muthigi A, et al: Dysreg­
epidemiology telling us about the etiology?, Int J Infect tis, J Rheumatol 30:2165–2169, 2003. ulation of autoantigen genes in ANCA-associated
Dis 9:185–194, 2005. Hata A, Noda M, Moriwaki R, et al: Angiographic find- vasculitis involves alternative transcripts and new
Cacoub P, Terrier B, Saadoun D: Hepatitis C virus- ings in Takayasu arteritis: New classification, Int J protein synthesis, J Am Soc Nephrol 26:390–399,
induced vasculitis: Therapeutic options, Ann Rheum Cardiol 54:S155–S163, 1996. 2015.
Dis 73(1):24–30, 2014. Hellmark T, Niles JL, Collins AB, et al: Comparison of Mehregan DR, Hall MJ, Gibson LE: Urticarial vasculi-
Carmona FD, Mackie SL, Martín JE, et al: A large-scale anti-GBM antibodies in sera with or without ANCA, tis: A histopathologic and clinical review of 72 cases,
genetic analysis reveals a strong contribution of the J Am Soc Nephrol 8:376, 1997. J Am Acad Dermatol 26:441–448, 1992.
HLA class II region to giant cell arteritis susceptibility, Hernández-Rodríguez J, Alba MA, Prieto-González S, Miller DV, Maleszewski JJ: The pathology of large-
Am J Hum Genet 96(4):565–580, 2015. et al: Diagnosis and classification of polyarteritis vessel vasculitides, Clin Exp Rheumatol 29(Suppl 64):
Cavazza A, Muratore F, Boiardi L, et al: Inflamed tem- nodosa, J Autoimmun 48-49:84–89, 2014. S92–S98, 2011.
poral artery: Histologic findings in 354 biopsies, with Hoffman GS: Large-vessel vasculitis: Unresolved issues, Moldoveanu Z, Wyatt RJ, Lee JY, et al: Patients with

PART 6
clinical correlations, Am J Surg Pathol 38:1360–1370, Arthritis Rheum 48:2406–2414, 2003. IgA nephropathy have increased serum galactose-
2014. Jain S, Win HN, Chalam V, et al: Disseminated gono- deficient IgA1 levels, Kidney Int 71:1148–1154, 2007.
Chatterjee S, Flamm SD, Tan CD, et al: Clinical diag- coccal infection presenting as vasculitis: A case report, Nathavitharana RR, Mitty JA: Diseases from North
nosis and management of large vessel vasculitis: J Clin Pathol 60:90–91, 2007. America: Focus on tick-borne infections, Clin Med
Takayasu arteritis, Curr Cardiol Rep 16(7):499, 2014. Jennette JC: Rapidly progressive crescentic glomerulo- 15:74–77, 2015.
Chen JY, Mao JH: Henoch-Schönlein purpura nephritis nephritis, Kidney Int 63:1164, 2003. Ntatsaki E, Mooney J, Scott DG, et al: Systemic rheu-
in children: Incidence, pathogenesis and management, Jennette JC: Nomenclature and classification of vasculi- matoid vasculitis in the era of modern immunosup-
World J Pediatr 11(1):29–34, 2015. tis: Lessons learned from granulomatosis with polyan- pressive therapy, Rheumatology (Oxford) 53:145–152,
Chen KR, Toyohara A, Suzuki A, et al: Clinical and giitis (Wegner’s granulomatosis), Clin Exp Immunol 2014.
histopathological spectrum of cutaneous vasculitis in 164(Suppl 1):7–10, 2011. Orenstein JM, Shulman ST, Fox LM, et al: Three linked
rheumatoid arthritis, Br J Dermatol 147(5):905–913, Jennette JC, Falk RJ: Pathologic classification of vascu- vasculopathic processes characterize Kawasaki disease:
2002. litis, Pathology Case Reviews 12:179–185, 2007. A light and transmission electron microscopic study,
Cho SY, Kim Y, Cha SH, et al: Adjuvant laboratory Jennette JC, Falk RJ: Pathogenesis of antineutrophil PLoS ONE 7(6):e38998, 2012.
marker of Kawasaki disease: NT-pro-BNP or cytoplasmic autoantibody-mediated disease, Nature Ortiz-Sanjuán F, Blanco R, Hernández JL, et al: Drug-
hs-CRP?, Ann Clin Lab Sci 41:360–363, 2011. Rev Rheumatol 10:463–473, 2014. associated cutaneous vasculitis: Study of 239 patients
Cohen Tervaert JW, Damoiseaux J: Antineutrophil cyto- Jennette JC, Falk RJ, Andrassy K, et al: Nomenclature from a single referral center, J Rheumatol 41(11):2201–
plasmic autoantibodies: How are they detected and of systemic vasculitides: The proposal of an interna- 2207, 2014.
what is their use for diagnosis, classification and tional consensus conference, Arthritis Rheum 37:187– Pedchenko V, Bondar O, Fogo AB, et al: Molecular
follow-up?, Clin Rev Allergy Immunol 43(3):211–219, 192, 1994. architecture of the Goodpasture autoantigen in
2012. Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised anti-GBM nephritis, N Engl J Med 363(4):343–354,
Coremans IE, Spronk PE, Bootsma H, et al: Changes in International Chapel Hill Consensus Conference 2010.
antibodies to C1q predict renal relapses in systemic Nomenclature of Vasculitides, Arthritis Rheum 65:1– Pendergraft WF III, Niles JL: Trojan horses: Drug
lupus erythematosus, Am J Kidney Dis 26(4):595–601, 11, 2013. culprits associated with antineutrophil cytoplasmic
1995. Kain R, Exner M, Brandes R, et al: Molecular mimicry autoantibody (ANCA) vasculitis, Curr Opin Rheumatol
Cui Z, Zhao MH: Advances in human antiglomerular in pauci-immune focal necrotizing glomerulonephri- 26(1):42–49, 2014.
basement membrane disease, Nat Rev Nephrol 7(12): tis, Nat Med 14(10):1088–1096, 2008. Pendergraft WF, Preston GA, Shah RR, et al: cPR3105-
697–705, 2011. Kallenberg CG: Anti-C1q autoantibodies, Autoimmun 206, a protein complementary to the autoantigen
D’Alessandro M, Buoncompagni A, Minoia F, et al: Rev 7:612–615, 2008. proteinase 3, triggers autoimmunity, Nature Med 10:
Cytomegalovirus-related necrotising vasculitis mim- Kermani TA, Schmidt J, Crowson CS, et al: Utility of 72–79, 2004.
icking Henoch-Schönlein syndrome, Clin Exp Rheu- erythrocyte sedimentation rate and C-reactive protein Poller DN, van Wyk Q, Jeffrey MJ: The importance of
matol 32(3 Suppl 82):S73–S75, 2014. for the diagnosis of giant cell arteritis, Semin Arthritis skip lesions in temporal arteritis, J Clin Pathol 53:137–
Damoiseaux J: The diagnosis and classification of the Rheum 41:866–871, 2012. 139, 2000.
cryoglobulinemic syndrome, Autoimmun Rev 13(4– Kermani TA, Warrington KJ, Crowson CS, et al: Prieto-Gonzalez S, Depetris M, Garcia-Martinez A,
5):359–362, 2014. Large-vessel involvement in giant cell arteritis: A et al: Positron emission tomography assessment of
Damoiseaux J, Mallet K, Vaessen M, et al: Automatic population-based cohort study of the incidence— large vessel inflammation in patients with newly diag-
reading of ANCA-slides: Evaluation of the AKLIDES trends and prognosis, Ann Rheum Dis 72:1989–1994, nosed, biopsy-proven giant cell arteritis: A prospec-
system, Clin Dev Immunol 2012:762874, 2012. 2013. tive, case-control study, Ann Rheum Dis 73:1388–1392,
Dasgupta B, Cimmino MA, Kremers HM, et al: 2012 Kohro-Kawata J, Wener MH, Mannik M: The effect of 2014.
Provisional classification criteria for polymyalgia high salt concentration on detection of serum immune Principi N, Rigante D, Esposito S: The role of infection
rheumatica: A European League Against Rheumatism/ complexes and autoantibodies to C1q in patients with in Kawasaki syndrome, J Infect 67:1–10, 2013.
American College of Rheumatology collaborative ini- systemic lupus erythematosus, J Rheumatol 29:84–89, Reindel R, Bischof J, Kim KY, et al: CD84 is markedly
tiative, Arthritis Rheum 64:943–954, 2012. 2002. up-regulated in Kawasaki disease arteriopathy, Clin
DeVita S, Soldano F, Isola M, et al: Preliminary classi- Lamprecht P, Moosig F, Gause A, et al: Immunological Exp Immunol 177:203–211, 2014.
fication criteria for the cryoglobulinaemic vasculitis, and clinical follow-up of hepatitis C virus associated Reynolds J, Preston GA, Pressler BM, et al: Autoim-
Ann Rheum Dis 70:1183–1190, 2011. cryoglobulinaemic vasculitis, Ann Rheum Dis 60:385– munity to the alpha 3 chain of type IV collagen in
Dietrich A, Gauglitz GG, Pfluger TT, et al: Syphilitic 390, 2001. glomerulonephritis is triggered by “autoantigen com-
aortitis in secondary syphilis, JAMA Dermatol 150: Langford CA: Vasculitis, J Allergy Clin Immunol 111: plementarity”, J Autoimmun 59:8–18, 2015.
790–791, 2014. S602–S612, 2003. Roth AJ, Brown MC, Smith RN, et al: Anti-LAMP-2
Durling B, Toren A, Patel V, et al: Incidence of discor- Lincoff NS, Erlich PD, Brass LS: Thrombocytosis in antibodies are not prevalent in patients with antineu-
dant temporal artery biopsy in the diagnosis of giant temporal arteritis rising platelet counts: A red flag for trophil cytoplasmic autoantibody glomerulonephritis,
cell arteritis, Can J Ophthalmol 49:157–161, 2014. giant cell arteritis, J Neuro-Ophthalmol 20:67–72, J Am Soc Nephrol 23:545–555, 2012.
Falk RJ, Jennette JC: Anti-neutrophil cytoplasmic auto- 2000. Roth AJ, Ooi J, Hess JJ, et al: ANCA epitope specificity
antibodies with specificity for myeloperoxidase in Lionaki S, Blyth ER, Hogan SL, et al: Classification of determines pathogenicity, detectability and clinical
patients with systemic vasculitis and idiopathic necro- antineutrophil cytoplasmic autoantibody vasculitides: predictive value, J Clin Invest 123:1773–1783, 2013.
tizing and crescentic glomerulonephritis, N Engl J The role of antineutrophil cytoplasmic autoantibody Rutgers A, Slot M, van Paassen P, et al: Coexistence of
Med 318:1651–1657, 1988. specificity for myeloperoxidase or proteinase 3 in anti-glomerular basement membrane antibodies and
Fujimoto S, Uezono S, Hisanaga S, et al: Incidence disease recognition and prognosis, Arthritis Rheum myeloperoxidase-ANCAs in crescentic glomerulone-
of ANCA-associated primary renal vasculitis in the 64:3452–3462, 2012. phritis, Am J Kidney Dis 46:253, 2005.

1031.e1
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Sablé-Fourtassou R, Cohen P, Mahr A, et al: Antineu- vasculitis-specific ANCA, PLoS ONE 9(9):e107743, van Timmeren MM, Heeringa P, Kallenberg CG: Infec-
53  Vasculitis
trophil cytoplasmic antibodies and the Churg-Strauss 2014. tious triggers for vasculitis, Curr Opin Rheumatol 26:
syndrome, Ann Intern Med 143:632–638, 2005. Suzuki H, Fan R, Zhang Z, et al: Aberrantly glycosylated 416–423, 2014.
Savige J, Davies D, Falk RJ, et al: Antineutrophil cyto- IgA1 in IgA nephropathy patients is recognized by Venzor J, Lee WL, Huston DP: Urticarial vasculitis,
plasmic antibodies and associated diseases: A review of IgG antibodies with restricted heterogeneity, J Clin Clin Rev Allergy Immunol 23(2):201–216, 2002.
the clinical and laboratory features, Kidney Int 57:846, Invest 119:1668–1677, 2009. Waldman CW, Waldman SD, Waldman RA: Giant cell
2000. Suzuki H, Kiryluk K, Novak J, et al: The pathophysiol- arteritis, Med Clin North Am 97:329–335, 2013.
Savige J, Gillis D, Davies D, et al: International consen- ogy of IgA nephropathy, J Am Soc Nephrol 22:1795– Watts R, Lane S, Hanslik T, et al: Development and
sus statement on testing and reporting of antineutro- 1803, 2011. validation of a consensus methodology for the classi-
phil cytoplasmic antibodies (ANCA), Am J Clin Pathol Suzuki Y1, Matsuzaki K, Suzuki H, et al: Serum levels fication of the ANCA-associated vasculitides and
111:507–513, 1999. of galactose-deficient immunoglobulin (Ig) A1 and polyarteritis nodosa for epidemiological studies, Ann
Scott DG, Bacon PA: Intravenous cyclophosphamide related immune complex are associated with disease Rheum Dis 66:222–227, 2007.
plus methylprednisolone in treatment of systemic activity of IgA nephropathy, Clin Exp Nephrol 18(5): Watts RA, Mahr A, Mohammad AJ, et al: Classifica­
rheumatoid vasculitis, Am J Med 76:377–384, 1984. 770–777, 2014. tion, epidemiology and clinical subgrouping of anti-
Seelen MA, Trouw LA, Daha MR: Diagnostic and prog- Tedeschi A, Baratè C, Minola E, et al: Cryoglobuline- neutrophil cytoplasmic antibody (ANCA)-associated
nostic significance of anti-C1q antibodies in systemic mia, Blood Rev 21:183–200, 2007. vasculitis, Nephrol Dial Transplant 30(Suppl 1):i14–i22,
lupus erythematosus, Curr Opin Nephrol Hypertens Terrier B, Semoun O, Saadoun D, et al: Prognostic 2015.
12:619–624, 2003. factors in patients with hepatitis C virus infection and Westman KW, Bygren PG, Eilert I, et al: Rapid screen-
Silverman DJ, Bond SB: Infection of human vascular systemic vasculitis, Arthritis Rheum 63:1748–1757, ing assay for anti-GBM antibody and ANCAs: An
endothelial cells by Rickettsia rickettsii, J Infect Dis 2011. important tool for the differential diagnosis of pulmo-
149:201–206, 1984. Treglia G, Taralli S, Maggi F, et al: Usefulness of (18) nary renal syndromes, Nephrol Dial Transplant 12(9):
Sinico RA, Di Toma L, Maggiore U, et al: Prevalence F-FDG PET/CT in disease extent and treatment 1863–1868, 1997.
and clinical significance of antineutrophil cytoplasmic response assessment in a patient with syphilitic aorti- Weyand CM, Goronzy JJ: Giant cell arteritis and poly-
antibodies in Churg-Strauss syndrome, Arthritis tis, Clin Nucl Med 38:e185–e187, 2013. myalgia rheumatica, N Engl J Med 371:50–57, 2014.
Rheum 52:2926–2935, 2005. Vaideeswar P, Deshpande JR: Pathology of Takayasu Xiao H, Dairaghi DJ, Powers JP, et al: C5a receptor
Sinico RA, Di Toma L, Maggiore U, et al: Renal arteritis: A brief review, Ann Pediatr Cardiol 6(1):52– (CD88) blockade protects against MPO-ANCA glo-
involvement in Churg-Strauss syndrome, Am J Kidney 58, 2013. merulonephritis, J Am Soc Nephrol 25:225–231, 2014.
Dis 47:770–779, 2006. Valsakumar AK, Valappil UC, Jorapur V, et al: Role of Xiao H, Heeringa P, Hu P, et al: Antineutrophil cyto-
Sinico RA, Rimoldi L, Radice A, et al: Anti-C1q immunosuppressive therapy on clinical, immunologi- plasmic autoantibodies specific for myeloperoxidase
autoantibodies in lupus nephritis, Ann N Y Acad Sci cal, and angiographic outcome in active Takayasu’s cause glomerulonephritis and vasculitis in mice, J Clin
1173:47–51, 2009. arteritis, J Rheumatol 30:1793–1798, 2003. Invest 110:955–963, 2002.
Sowa M, Grossmann K, Knütter I, et al: Simultaneous
automated screening and confirmatory testing for

1031.e2
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en junio 26, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.

Вам также может понравиться