You are on page 1of 18

CJASN ePress. Published on October 17, 2017 as doi: 10.2215/CJN.

00620117

Thrombotic Microangiopathy and the Kidney


Vicky Brocklebank,*† Katrina M. Wood,‡ and David Kavanagh*†

Abstract
Thrombotic microangiopathy can manifest in a diverse range of diseases and is characterized by thrombocytopenia,
microangiopathic hemolytic anemia, and organ injury, including AKI. It can be associated with significant
morbidity and mortality, but a systematic approach to investigation and prompt initiation of supportive
management and, in some cases, effective specific treatment can result in good outcomes. This review considers the *National Renal
classification, pathology, epidemiology, characteristics, and pathogenesis of the thrombotic microangiopathies, Complement
and outlines a pragmatic approach to diagnosis and management. Therapeutics Centre,
Newcastle upon Tyne,
Clin J Am Soc Nephrol 13: ccc–ccc, 2018. doi: https://doi.org/10.2215/CJN.00620117
Hospitals National
Health Service
Foundation Trust,
Newcastle upon Tyne,
Introduction nomenclature make interpretation difficult: aHUS UK; †Institute of
Cellular Medicine,
Thrombotic microangiopathy (TMA), a pathologic de- may refer specifically to complement mediated Newcastle University,
scription, is characterized by a clinical presentation with TMA, or be more loosely applied to any TMA that Newcastle upon Tyne,
thrombocytopenia, microangiopathic hemolytic anemia is not TTP or STEC-HUS. In this review, we adopt the UK; and ‡Department
(MAHA), and organ injury (1,2). It can manifest in a term complement-mediated aHUS when the cause is of Cellular Pathology,
diverse range of conditions and presentations, but AKI Newcastle upon Tyne
defined as such, and use aHUS where the cause is ill
Hospitals National
is a common prominent feature because of the apparent defined. Current classifications describe primary Health Service
propensity of the glomerular circulation to endothelial TMAs, known as either acquired (e.g., factor H Foundation Trust,
damage and occlusion (3). Early recognition is impor- (FH) autoantibodies, ADAMTS13 autoantibodies) or Newcastle upon Tyne,
tant: TMAs are associated with significant mortality and inherited (e.g., complement mutations, ADAMTS13 UK
morbidity, including ESRD, although prompt initiation mutations); secondary TMAs; and infection-associated
of supportive and specific management can transform Correspondence:
TMAs (2) (Figure 1). Although a useful framework
Prof. David Kavanagh,
outcome. In particular, the prognoses of thrombotic for discussion, these terms do not account for the National Renal
thrombocytopenic purpura (TTP) and atypical hemo- increasing recognition that patients with an under- Complement
lytic uremic syndrome (aHUS) have been revolutionized lying complement risk factor often require a second- Therapeutics Centre,
following the elucidation of the pathogenic mechanisms ary trigger for TMA to manifest. Additionally, with Atypical Haemolytic
Uremic Syndrome
and the introduction of effective therapy. Challenges and increasing reports of eculizumab nonresponse, a more Service, Building 26,
controversies remain, however. In this review, we will clinically orientated classification such as eculizumab- Royal Victoria
discuss the classification, pathology, epidemiology, char- responsive and eculizumab-resistant aHUS are likely to Infirmary, Queen
acteristics, and pathogenesis of the TMAs, and outline an be introduced. Victoria Road,
approach to diagnosis and management. Newcastle upon Tyne,
NE1 4LP, United
Pathology Kingdom. Email:
Classification of Thrombotic Microangiopathies TTP is characterized by unusually large multimers david.kavanagh@
The classification of the TMAs is challenging and of vWf- and platelet-rich thrombi in capillaries and newcastle.ac.uk
constantly evolving. Historical classifications were on arterioles (5), although with current practice, pathologic
the basis of clinical findings: TTP for predominant specimens are rarely available. In complement-mediated
neurologic involvement and hemolytic uremic syndrome aHUS and other TMAs with more pronounced AKI,
(HUS) for kidney dominant disease. Classifications kidney biopsy is more frequently performed, although
evolved with greater understanding of the molecular is not requisite for the diagnosis. The pathologic find-
basis of disease: TTP was defined by severe ADAMTS13 ings reflect tissue responses to endothelial injury, in-
deficiency, hemolytic uremic syndrome caused by shiga cluding endothelial swelling and mesangiolysis in active
toxin–producing Escherichia coli (STEC-HUS) was de- lesions, and double contours of the basement mem-
fined by the presence of shiga toxin–producing bacte- brane in chronic lesions (Figure 2) (reviewed by
ria, and aHUS was broadly used for all other causes Goodship et al. (6)). Overt fibrin platelet thrombosis
of TMA. The discovery of the role of complement may be absent from renal biopsies of TMA, which has
dysregulation in a proportion of patients with aHUS recently led to a suggested reclassification to micro-
subsequently led to the acceptance of the term angiopathy with or without thrombosis (6). Evidence
complement-mediated TMA to refer to this subgroup of TMA has also been reported in a number of glomer-
(2,4). It should be recognized that differences in the ular diseases (7) and autoimmune diseases; however,
historical and contemporary literature over in published clinicopathologic studies, only a small

www.cjasn.org Vol 13 March, 2018 Copyright © 2018 by the American Society of Nephrology 1
2 Clinical Journal of the American Society of Nephrology

Figure 1. | Thrombotic microangiopathies are classified into: Inherited or acquired primary; secondary; or infection associated TMAs. Current classi-
fications define primary TMAs as hereditary (mutations in ADAMTS13, MMACHC (cb1c deficiency), or genes encoding complement proteins) or acquired
(autoantibodies to ADAMTS13, or autoantibodies to complement FH, which is associated with homozygous CFHR3/1 deletion). TMA is associated with
various infections: in STEC-HUS and pneumococcal HUS, distinct mechanisms result in TMA; in other infections, the processes are not defined and in some
cases the infection may trigger manifestation of a primary TMA. Secondary TMAs occur in a spectrum of conditions, and in many cases the pathogenic
mechanismsaremultifactorialorunknown.Theclassificationpresentedhereisnotunequivocal:insomesecondaryTMAs,for examplepregnancy-associated
TMA or de novo TMA after transplantation, a significant proportion of individualswill have a genetic predisposition to a primary TMA. AAV, ANCA-associated
vasculitis; ADAMTS13, a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13; aHUS, atypical hemolytic uremic syndrome;
C3G, C3 glomerulopathy; CAPS, catastrophic antiphospholipid syndrome; cblC, cobalamin C type; DGKE, gene encoding diacylglycerol kinase «; FH, factor
H; HELLP, syndrome of hemolysis, elevated liver enzymes, and low platelets; HUS, hemolytic uremic syndrome; IgAN, IgA nephropathy; MN, membranous
nephropathy; MPGN, membranoproliferative GN; SRC, scleroderma renal crisis; STEC, shiga toxin–producing Escherichia coli; TMA, thrombotic micro-
angiopathy; TTP, thrombotic thrombocytopenic purpura.

proportion of individuals with pathologic evidence of TMA in and MAHA, identified by evidence of erythrocyte fragmen-
these contexts had concurrent clinical and laboratory evidence tation on peripheral blood film microscopy, which occurs in
(8,9). It is not possible, on the basis of current knowledge, to areas of turbulent flow in the microcirculation due to partial
establish the cause of TMA from the histopathologic mor- occlusion by platelet aggregates (1). Raised lactate dehy-
phology, although this may change with further research (6). drogenase is a result of cell lysis and tissue ischemia (1),
haptoglobin is low, and the direct antiglobulin (Coombs)
test is negative (except in pneumococcal HUS). Presen-
Clinical and Laboratory Features
tation with AKI reflects the consequences of ischemia in
The clinical presentation of TMA reflects hemolysis
the kidney.
and ischemic organ dysfunction, and depends on the
Once routine biochemical and hematologic analysis have
underlying disease etiology; AKI is a common manifesta-
demonstrated a TMA, investigations are aimed at determining
tion of TMAs, although rarely a severe feature of TTP (10).
the underlying disease cause and excluding other differential
Extrarenal manifestations are reported in thrombotic
diagnoses, with the most urgent test being an ADAMTS13
microangiopathies, with the published data referring
assay (Figure 3). The epidemiology, pathogenesis, and man-
primarily to those observed in complement-mediated aHUS
agement of TMAs are summarized in Table 2.
and STEC-HUS (Table 1), although it is not known whether
they are a consequence of the TMA, a direct effect of comple-
ment activation or shiga toxin, or complications of AKI, such as Primary TMAs
severe hypertension and uremia (6). Complement-Mediated aHUS
The defining laboratory features comprise thrombocyto- Complement-mediated aHUS is prototypical of disease
penia, resulting from platelet aggregation and consumption, occurring as a consequence of complement dysregulation.
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018 TMA and the Kidney, Brocklebank et al. 3

Figure 2. | The pathological features of thrombotic microangiopathies reflect the tissue responses to endothelial injury. (A) Glomerular
paralysis with capillary loops containing abundant erythrocytes (silver, 3400). (B) Thrombus in an artery (hematoxylin and eosin, 3400). (C)
Glomerular capillary lumina containing fibrin thrombi (red) and erythrocytes (yellow) (Martius Scarlet Blue, 3400). (D) Erythrocyte fragments
within the arterial vessel wall (arrow) (hematoxylin and eosin, 3400). (E) Mucoid thickening and obliteration of the lumen of a small artery
(hematoxylin and eosin, 3400). (F) Myxoid intimal thickening of small artery (hematoxylin and eosin, 3400). (G) Fibrinoid necrosis of arterial
wall (Martius Scarlet Blue, 3400). (H) Reduplication of glomerular basement membrane (arrow) and fibrillary mesangium (periodic acid–Schiff,
3400). (I) Glomerulus with endothelial swelling and erythrocyte fragmentation (arrow) (hematoxylin and eosin, 3400). (J) Electron micrograph
demonstrating fibrin tactoids (black) in glomerular capillary (310,000).
4 Clinical Journal of the American Society of Nephrology

Hereditary Complement-Mediated aHUS


Table 1. Extrarenal manifestations reported in thrombotic Molecular evidence that CFH mutations are associated
microangiopathies with aHUS was first described in genetic research published
in 1998 (15), and since then a multitude of studies have
* † Neurologic involvement, including seizures and determined heterozygous pathogenic activating mutations in
altered consciousness the genes encoding the alternative pathway components C3
* † Pancreatitis
and factor B, and loss-of-function mutations in the genes
* † Cardiac involvement/myocardial infarction
* † Gastrointestinal involvement (including diarrhea, encoding the regulators FH (including CFH/CFHR fusions),
vomiting, abdominal pain) FI, and CD46 (reviewed in Kavanagh et al. [14]).
* Cerebral artery thrombosis/stenosis These genetic mutations are not causative but are instead
* Extracerebral artery stenosis predisposing, with penetrance incomplete. Penetrance of
* Digital gangrene/skin disease is age-related and has been reported to be as high as
* Ocular involvement 64% by the age of 70 years for individuals carrying a single
* Hepatitis genetic mutation (6). This suggests that additional disease
* Pulmonary involvement risk modifiers are important. Around 3% of patients have
more than one mutation, with increased penetrance per
The published data refers primarily to those observed in additional mutation. Haplotypes (particular combinations of
complement-mediated atypical hemolytic uremic syndrome (*)
single nucleotide polymorphisms) in CFH and CD46 also
and hemolytic uremic syndrome caused by shiga toxin–producing
modify penetrance (14). Together, these still do not answer
Escherichia coli (†) (6,14,27,59).
the question of why some individuals do not develop dis-
ease until later in life. This is best explained by the need
The complement system is a regulated cascade network of for an environmental trigger (e.g., pregnancy or infection) to
signaling and amplification that incorporates .30 plasma unmask a latent complement defect. Complement activation
and cell surface–bound proteins, and was reviewed in detail is a common factor in many of these triggering events: it
earlier in this series (11). A key physiologic role involves occurs in the placenta at the fetomaternal interface in normal
stimulating the inflammatory response and opsonization pregnancy and is the normal physiologic response to in-
and lysis of pathogens to protect the host from infection fection. Counseling of family members and offering genetic
(encapsulated organisms in particular); it is a fundamental screening are important considerations.
component of the innate immune system, and modulates Historically the outcome of complement mediated aHUS
the adaptive immune system. In addition, it facilitates the at the initial presentation was very poor, as were outcomes
disposal of potentially injurious immune complexes and after kidney transplantation: recurrence was 68% and
damaged host cells (12). 5-year death-censored graft survival was 51% (16). The out-
Complement activation is initiated by three pathways: come of kidney transplantation is predicted largely by the
the classical pathway is activated by immune complexes and underlying genetic abnormality, with the highest risk asso-
other molecules (e.g., C-reactive protein), and activation of ciated with CFH, CFB, and C3 mutations and the lowest with
the lectin pathway results from protein interactions with CD46 mutations (17). In the pre-eculizumab era, one option for
pathogens (11). The alternative pathway may also be initiated individuals with a defect in a complement protein predomi-
by pattern recognition molecules, but crucially, it is a positive nantly synthesized in the liver (FH, FI, factor B, and C3) was
feedback loop that is constitutively active due to spontaneous combined liver and kidney transplantation (14); however,
hydrolysis (tickover) of C3, and is recruited by the classical short-term risks were significant and international experience
and lectin pathways; this enables a rapid response against was limited (18), and some people were therefore considered
pathogens, but leaves the host vulnerable to bystander to be untransplantable.
damage if the amplification loop is unchecked. The system
is therefore tightly regulated by plasma proteins, including FH Acquired Complement-Mediated aHUS
and factor I (FI), and cell surface proteins, such as membrane aHUS associated with autoantibodies against FH was
cofactor protein (CD46) (13). Defects in these regulators or in first reported in 2005 (19), and functional analyses have
the alternative pathway components can lead to complement demonstrated disruption of complement regulation by mul-
dysregulation, with activation of the terminal complement tiple mechanisms (20). There is a strong association with
pathway and generation of the anaphylatoxin C5a and the homozygous deletion of CFHR3 and CFHR1, which encode the
membrane attack complex (C5b-9), resulting in a complement- proteins FHR3 and FHR1 (21), although the mechanism is not
mediated aHUS (Figure 4). The relative roles of these effector understood; CFHR3/1 deletion is a common polymorphism,
molecules in causing disease remains to be established. and it is not present in all individuals who develop FH
In complement-mediated aHUS, dysregulated comple- autoantibodies (22). It predominantly presents in childhood,
ment activation primarily occurs on endothelial cell sur- frequently with a gastrointestinal prodrome. Autoantibodies
faces, and although abnormal serum levels of complement against FI have also been reported, but they are rare and their
components such as C3 may be observed, normal levels do functional relevance remains to be established (23).
not exclude complement-mediated disease (6,14). There is
evidence of complement activation (plasma levels and Plasma Exchange
tissue staining) in many other TMAs, but whether this Plasma exchange (PE) still remains the initial treat-
is a primary event, a disease modifier, or an inconsequen- ment of choice until ADAMTS13 activity is available to
tial bystander phenomenon has not yet been definitively exclude TTP as a diagnosis. It should be initiated in adults
established (13). as soon as the diagnosis of TMA is suspected. In addition to
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018 TMA and the Kidney, Brocklebank et al. 5

Figure 3. | A diagnostic algorithm for the investigation and management of a patient presenting with thrombotic microangiopathy. Supportive
treatment is essential. This comprises fluid management and, if indicated, judicious packed red blood cell transfusion, intensive care unit admission,
and RRT. Platelet transfusion may worsen the TMA and so should be avoided if possible. The complete evaluation of a patient presenting with TMA
comprises complement analysis and investigations for all conditions in which TMA can manifest, and usually enables the disease cause to be
established. However, collating the results of the requisite investigations may take considerable time. The clinical evaluation during the acute
presentation of a TMA requires some time-critical decision-making, which should focus initially on the consideration of TTP, because immediate
management is imperative given the high mortality rate if untreated, and therefore in adults, PE should be instituted on the presumption that it is TTP
unless other evidence is available that strongly suggests an alternative cause. In children, in whom TTP is rarer, first-line treatment with eculizumab
should be considered if complement-mediated aHUS is suspected and should not be delayed while ADAMTS13 activity is determined. In the
absence of a defined cause, complement-mediated aHUS is presumed and treatment with eculizumab is recommended, pending the complete
evaluation. Eculizumab is not universally available; in these circumstances, treatment should comprise PE, and there may be a role for liver
transplantation. *Full complement evaluation is recommended (Kidney Disease: Improving Global Outcomes consensus [6]) in individuals with
pregnancy-associated aHUS and de novo transplantation-associated aHUS because of the high prevalence of rare genetic variants described in
these subgroups, and in cases of STEC-HUS that result in ESRD, as rare genetic variants have been described after HUS recurrence in a subsequent
kidney transplant. Additionally, it is recognized that infection can trigger manifestation of complement-mediated aHUS. In other secondary cases
of aHUS, insufficient evidence exists to recommend a full genetic evaluation, although rare genetic variants have been described in many of these
6 Clinical Journal of the American Society of Nephrology

removing ADAMTS13 autoantibodies and replacing autoantibody to FH, all had an improvement in eGFR,
ADAMTS13 in TTP, PE will also replace faulty complement whereas 27% of individuals without an identified comple-
regulators and remove FH autoantibodies and hyperfunc- ment abnormality failed to show an improvement. It is not
tional complement components in complement-mediated clear whether this represents late presentation of disease or
aHUS. In children, PE is associated with a high rate of true nonresponse. A polymorphism in the C5 gene (p.R885H)
complications (24) and TTP is rare, therefore PE is not has been demonstrated to prevent eculizumab binding to
considered as a first-line treatment when eculizumab is avail- C5 (35), and nonresponders to eculizumab should have
able. In many parts of the world, the cost of eculizumab genetic screening for this single nucleotide polymorphism
precludes its use, and PE remains the only treatment for and alternative treatment strategies considered. More re-
complement-mediated aHUS (25). cently, individuals presenting with a TMA with failure to
respond to eculizumab have been demonstrated to have
Complement-Inhibiting Therapy genetic variants in the noncomplement genes DGKE (36) and
The evidence for the central role of primary complement INF2 (37).
defects in pathogenesis summarized above provided the The primary concern with terminal complement blockade is
mechanistic rationale for treating complement-mediated increased susceptibility to infection with encapsulated organ-
aHUS with complement-inhibiting therapy. Eculizumab is a isms, particularly Neisseria infections (33,38). For this reason,
recombinant humanized mAb that functionally blocks C5, meningococcal vaccination is considered mandatory; how-
and trials published in 2013 demonstrated its efficacy (26). ever, serologic response is variable (39), and the efficacy of
Although these were single-arm studies rather than random- vaccination in the context of complement blockade is un-
ized, controlled trials, the historically poor outcomes (up to certain (6). Long-term antibiotic prophylaxis is therefore
77% of patients with CFH mutations had died or reached recommended for the duration of treatment and up to 3
ESRD by 3–5 years) justified such study designs (27,28). These months after withdrawal (6), although meningococcal in-
findings have been replicated in subsequent extension studies fection can occur despite these precautions (40,41). Thus,
(29), prospective (nonrandomized) studies (30,31), and cohort patient education regarding vigilance is essential. Other
analysis (32). In the prospective trials, complete TMA re- concerns may become apparent as use of complement-
sponse was achieved in approximately 65% after 26 weeks of inhibiting therapy in clinical practice expands: hepatotox-
eculizumab therapy in both adults (26) and children (31). icity in association with eculizumab has been reported in
Eculizumab was approved by the US Food and Drug children (42), and deposition of eculizumab has been
Administration and the European Medicines Agency for reported in individuals with C3 glomerulopathy (C3G)
use in aHUS in 2013 (see Table 3 for practical consider- (but not yet in aHUS), although the clinical significance is
ations) (33). In those presenting late in the course of disease unclear (43).
who do not recover kidney function, the high recurrence The use of complement-inhibiting therapy in TMAs other
rate after kidney transplantation necessitates preemptive than complement-mediated aHUS is controversial; case
eculizumab (Table 4) (6,16). The trials included patients in studies have reported success but the inherent publication
whom no complement mutation or FH autoantibody had bias needs to be considered when interpreting these, and
been identified, and the majority responded to complement- given that the role of complement in the pathogenesis of
inhibiting therapy; therefore, negative genetic and autoan- other TMAs has not yet been defined, interventional trials are
tibody investigations do not exclude the diagnosis of likely to be needed before a consensus can be achieved (13).
complement-mediated aHUS.
The current license for eculizumab is for lifelong treatment, Thrombotic Thrombocytopenic Purpura
but this is not evidence based. Withdrawal of eculizumab TTP is mediated by ADAMTS13 deficiency: in hereditary
has been described in a large series of patients with aHUS, with TTP, this results from recessive mutations (homozygous or
relapse reported in around one third, exclusively in those compound heterozygous) in the ADAMTS13 gene, and in
with complement mutations (34). That rapid reintroduction acquired TTP, ADAMTS13 activity is inhibited by autoan-
of eculizumab returned kidney function to baseline suggests tibodies (2). ADAMTS13 is a metalloproteinase enzyme
that a disease-driven intermittent regime could replace long- that functions to cleave vWf; deficiency results in unusu-
term therapy, although prospective trials are required. ally large vWf multimers and consequent occlusive micro-
With increased clinical use, evidence is emerging of vascular platelet aggregation (2). The incidence of TTP is
nonresponse to eculizumab. In the recent pediatric higher in adults than in children (2), and there is a female
trial, Greenbaum et al. (31) highlighted that, for those predominance (44). Neurologic manifestations are common
with a rare genetic variant in the complement system or an and laboratory characteristics in comparison to aHUS are

presentations. In cases where the role of complement is as yet unclear, the clinician should decide on the evaluation on the basis of the potential
clinical consequences of a positive result (e.g., listing for renal transplantation). 1ve, positive; AAV, ANCA-associated vasculitis; Ab, antibody;
ACA, anticentromere antibody; ACEI, angiotensin-converting enzyme inhibitor; ADAMTS13, a disintegrin and metalloproteinase with a
thrombospondin type 1 motif, member 13; Ag, antigen; aHUS, atypical hemolytic uremic syndrome; ANA antinuclear antibody; anti-ds DNA,
antidouble-stranded DNA; anti–Scl70, anti-topoisomerase I antibody; BMT; bone marrow transplant; C3G, C3 glomerulopathy; CAPS, cata-
strophic antiphospholipid syndrome; DGKE, gene encoding diacylglycerol kinase «; DIC, disseminated intravascular coagulation; FH, factor H;
FI, factor I; Hb, hemoglobin; HUS, hemolytic uremic syndrome; LDH, lactate dehydrogenase; MAHA, microangiopathic hemolytic anemia; MN,
membranous nephropathy; MPGN, membranoproliferative GN; PE, plasma exchange; SRC, scleroderma renal crisis; STEC, shiga toxin–producing
Escherichia coli; Stx, shiga toxin; T-Ag, Thomsen–Friedenreich antigen; TMA, thrombotic microangiopathy; TTP, thrombotic thrombocytopenic purpura.
Table 2. The thrombotic microangiopathies: epidemiology, pathogenesis, and management

TMA Epidemiology Pathogenesis Management Recommendations

Complement- Incidence (UK): 0.42 per million per yr (32) Complement dysregulation caused by: Eculizumab
mediated aHUS Hereditary: incomplete penetrance; can Hereditary: heterozygous mutations in CFH, CFI, If eculizumab is not available: PE, liver
present at any age; “trigger” required CD46, C3, and CFB (27,28) transplantation may be considered.
Acquired: anti-FH Ab aHUS more common Acquired: anti-FH Ab (19,21)
in children

TTP/ADAMTS13 Incidence (US): 0.37 per 100,000 per yr (46); ADAMTS13 deficiency caused by: PE
deficiencymediated adults 2.9 per million per yr, children 0.1 Hereditary: recessive (homozygous or
TMA per million per yr (2) compound heterozygous) ADAMTS13
Female.Male (44) mutations
Adults . children (2) Acquired: autoantibody-mediated inhibition Immunosuppression (steroids/
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018

rituximab) indicated if acquired

Cobalamin C Incidence of cblC deficiency: 1:37,000– Disorder of cblC metabolism results from recessive Metabolic therapy recommended:
deficiency 1:100,000 births (50) (homozygous or compound heterozygous) efficacy of hydroxocobalamin and
mutations in the MMACHC gene betaine established (50)
Can present in adulthood as well as Pathogenic mechanisms causing TMA remain
childhood undetermined

DGKE TMA Rare Recessive (homozygous or compound Insufficient evidence to determine


heterozygous) mutations in DGKE (36) optimal management
Presents aged ,1 yr (36) Loss of DGKE results in prothrombotic state Reports of both nonresponse (36) and
independently of complement activation (51) response (31) to eculizumab

STEC-HUS E. coli O157 predominant causative Enteric infection with Stx-producing pathogens Supportive care is recommended; high
pathogen: incidence (UK): 7.1 per million proportion may require dialysis. No
per yr; Latin America, 10–17 per 100,000 intervention evaluated in an RCT was
children per yr (113) superior to supportive care for any
outcome (systematic review [114])
Peak incidence in children ,5 yr (115) and Stx binds to Gb3, which is highly expressed in the Antibiotics: controversy remains,
approximately 15% of children with kidney, is internalized, and inhibits protein studies inconclusive, may worsen
enteric infection develop HUS (59) synthesis (63) outcome, therefore currently not
recommended; RCT of azithromycin
ongoing (NCT02336516)
E. coli O104: European outbreak in 2011 The consequent endothelial injury results in PE: not recommended, no benefit
exceptional for high HUS occurrence rate intravascular fibrin generation (59) established (no RCTs) (46)
(24%), severity, and high proportion of
adults (60)
Other Stx producing pathogens e.g., Shigella Complement activation observed (116) but role is Eculizumab: not recommended; no
dysenteriae not defined benefit demonstrated in retrospective
Outbreaks and sporadic cases analyses (60,68,69); RCT ongoing
(NCT02205541)
TMA and the Kidney, Brocklebank et al.
7
8

Table 2. (Continued)

TMA Epidemiology Pathogenesis Management Recommendations

Pneumococcal HUS Rare. Limited data available from published Neuraminidase cleaves sialic acid residues from Supportive management and treatment
cohorts, and incidence unknown glycoproteins on erythrocyte, platelet, and of infection recommended
(117,118). 10-yr cumulative incidence rate endothelial cell membranes, exposing the
of 1.2 per 100,000 children reported (NZ) cryptic T antigen to which IgM in the plasma
(119). Associated with pneumonia and can then bind, resulting in cell damage and
empyema in children ,2 yr (72) TMA (73).

HIV-associated TMA Pre-HAART era: incidence of 1.5%–7% Pathogenic mechanisms remain undetermined Supportive management and treatment
(75,120) of infection recommended
HAART era: incidence 0.3% (77)

Pregnancy- Complement-mediated aHUS: pregnancy Complement gene mutations in up to 86% (81) Eculizumab; start immediately (do not
associated TMA triggers complement-mediated TMA in wait for genetic analysis) if TTP has
approximately 20% of women with aHUS been excluded and presentation is not
Clinical Journal of the American Society of Nephrology

(81) suggestive of HELLP

A proportion are TTP: 10%–36% of women with TTP present vWf increases in normal pregnancy and PE
primary TMAs; during pregnancy (44) consumes ADAMTS13; in women with a
differential genetic predisposition, its activity can fall low
diagnosis includes enough for TTP to manifest (81)
complement-
mediated aHUS,
TTP, and HELLP
HELLP: incidence: 0.5%–0.9% of all Mechanisms are poorly understood (80) Definitive treatment is delivery (86). No
pregnancies; complicates 5%–10% of Increased levels of endoglin and sFlt-1 may play a role for PE (46)
cases of severe preeclampsia (121) role in endothelial dysfunction (79,86).

Drug-mediated TMA True incidence unknown Immune mediated: drug-dependent antibodies, Recommendation is drug
e.g., quinine (2) discontinuation
Reported incidences of TMA include: Toxicity mediated: multiple mechanisms, e.g., Ticlopidine-associated TMA: as per TTP
CNIs, IFN, chemotherapy agents, VEGF (46,92) (PE)
inhibitors (2)
Approximately 1:1000 patient-yr for high- Ticlopidine-associated TMA is mediated by
dose IFN-b (90) acquired ADAMTS13 deficiency (92)
2%–10% of patients treated with mitomycin
(122)
1%–4.7% of patients treated with tacrolimus
(122)
VEGF inhibitors: unknown (91)
Table 2. (Continued)

TMA Epidemiology Pathogenesis Management Recommendations

De novo TMA after Incidence after kidney transplantation: 0.8% Multifactorial Treatment of precipitating factors, e.g.,
solid organ in USRDS data (123); up to 14% in single- AMR, infection, drug withdrawal
transplant center studies (94)
Incidence: 4% in liver transplant and 2.3% in Complement gene mutations reported in up to Eculizumab where complement-
lung transplant recipients (93) 29% (renal transplants) (96) mediated aHUS is possible

TMA after bone Multisystem TMA complicates 10%–40% of Multifactorial No evidence-based effective
marrow transplant allogenic BMTs (97,98) management strategy
No established benefit with PE (46)
Favorable outcomes with eculizumab
compared with historical controls
reported in retrospective analysis
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018

(101), but prospective trials needed to


establish consensus

Severe hypertension- Incidence: variously reported; case series of Pathogenic mechanisms unclear No evidence supporting any strategy
associated TMA severe hypertension have identified TMA other than BP management
in 27%–44% (13,124–127)

Malignancy- Unknown Multifactorial Discontinuation of causative


associated TMA chemotherapy agents

TMA with TMA can occur in association with IgA Pathogenic mechanisms unclear Limited evidence for management of the
glomerular diseases nephropathy, ANCA-associated TMA
vasculitis, membranous nephropathy, Hereditary and acquired complement defects in Role of complement inhibition in C3G/
FSGS, and MPGN/C3G (13) MPGN/C3G aHUS crossover unclear

TMA with SLE: concurrent TMA reported in up to 8%– Pathogenic mechanisms unclear SLE: no evidence for specific TMA
autoimmune 15% of biopsies (9,128) management in addition to SLE
conditions management as recommended in
international guidelines
CAPS: TMA in 14% in international registry Some evidence of complement activation in SLE CAPS: efficacy of eculizumab suggested
(112) and CAPS (13) in case reports and series; prospective
trial (not RCT) to enable renal
transplantation ongoing (NCT01029587)
SRC: occurs in approximately 10% of people SRC: ACEIs significantly reduce
with SSc; TMA in 45%–50% (111,129) mortality (111)

aHUS, atypical hemolytic uremic syndrome; UK, United Kingdom; PE, plasma exchange; FH, factor H; Ab, antibody; TTP, thrombotic thrombocytopenic purpura; ADAMTS13, a disintegrin and
metalloproteinase with a thrombospondin type 1 motif, member 13; TMA, thrombotic microangiopathy; US, United States; cblC, cobalamin C type; STEC-HUS, hemolytic uremic syndrome
caused by shiga toxin–producing Escherichia coli; Stx, shiga toxin; RCT, randomized controlled trial; HUS, hemolytic uremic syndrome; Gb3, globotriaosylceramide; NZ, New Zealand; T antigen,
Thomsen–Friedenreich antigen; HAART, highly active antiretroviral therapy; HELLP, syndrome of hemolysis, elevated liver enzymes, and low platelets; CNI, calcineurin inhibitor; VEGF,
vascular endothelial growth factor; USRDS, United States Renal Data System; AMR, antibody-mediated rejection; BMT, bone marrow transplants; MPGN, mesangioproliferative GN; C3G, C3
TMA and the Kidney, Brocklebank et al.

glomerulopathy; CAPS, catastrophic antiphospholipid syndrome; SRC, scleroderma renal crisis; SSc, systemic sclerosis; ACEI, angiotensin-converting enzyme inhibitor.
9
10 Clinical Journal of the American Society of Nephrology

Figure 4. | Unfettered complement activation ultimately results in thrombus formation, platelet consumption, vascular occlusion and
mechanical hemolysis. Complement is activated by the alternative, classic, and lectin pathways. The alternative pathway of complement is a
positive amplification loop. C3b interacts with factor B, which is then cleaved by factor D to form the C3 convertase C3bBb. Unchecked, this leads to
activation of the terminal complement pathway with generation of the effector molecules, the anaphylatoxin C5a and the membrane attack complex
(C5b-9). To protect host cells from bystander damage, the alternative pathway is downregulated by complement regulators including factor H (FH),
factor I (FI), and CD46. In complement-mediated aHUS, activating mutations in C3 and CFB and loss-of-function mutations in CFH, CFI, and CD46, in
addition to autoantibodies to FH, result in overactivation of the alternative pathway. This leads to immune cell and platelet activation and endothelial cell
damage and swelling, with consequent thrombus formation, platelet consumption, vascular occlusion, and mechanical hemolysis. There is evidence
that complement is activated in other TMAs, but whether this is a causative, disease modifier, or bystander phenomenon has not yet been elucidated.
Eculizumab is a humanized mAb that binds to C5 and prevents activation of the terminal pathway; it thereby inhibits the generation of the effector
molecules that cause TMA in individuals in whom a primary defect in complement underlies the TMA pathogenesis. Its role in the treatment of other
TMAs is undefined. aHUS, atypical hemolytic uremic syndrome; TMA, thrombotic microangiopathy.

reportedly more severe thrombocytopenia (,303109/L) by complement-independent mechanisms (51). Only a small
and less severe AKI (serum creatinine, 1.7–2.3 mg/dl) (45), number of cases have been published, but it appears to
but these are not absolute and should not be relied upon in present in patients aged ,1 year and commonly results
clinical practice (10), where the diagnosis is confirmed by in progressive CKD and ESRD (36). There is insufficient ev-
ADAMTS13 activity ,10% (5). TTP was historically almost idence to determine optimal management; there are reports
universally fatal, but after the introduction of PE treatment, of both response (31) and nonresponse (36) to eculizumab,
mortality decreased to ,10% (46). In acquired TTP, immuno- as well as response (52,53) and nonresponse (36) to plas-
suppressive therapies (e.g., glucocorticoids and rituximab) ma therapy. Concomitant mutations in complement genes
have reduced the relapse rate (47–49). have been reported (54). Genetic pleiotropy is seen, whereby
DGKE mutations have also been associated with mesangio-
proliferative GN (MPGN) (55).
Cobalamin C Deficiency
TMA can manifest in methylmalonic aciduria and homo-
cystinuria, cobalamin C type, which is the most common in- Other Genetic Associations
herited form of functional cobalamin (vitamin B12) deficiency Genetic variants in thrombomodulin (THBD) have been
(50). It is caused by recessive (homozygous or compound reported in association with aHUS (56,57), although this
heterozygous) mutations in the MMACHC gene, can pre- finding was not replicated by Fremeaux-Bacchi et al. (27).
sent in adulthood as well as childhood, and the phenotype Bu et al. (58) reported rare genetic variants in plasminogen
may comprise developmental, ophthalmologic, neurologic, (PLG) in aHUS; however, replication analysis will be
cardiac, and kidney manifestations, although severity varies. required to clarify this disease association. Functionally
The pathophysiologic mechanisms that cause endothelial significant mutations in INF2 have been seen to segregate in
damage and subsequent TMA have not yet been determined families with TMAs, but it remains to be seen whether this
(50). Mortality is high if untreated or if there is cardiopulmo- is a primary TMA or secondary phenomenon in association
nary involvement, but metabolic therapy (hydroxocobalamin with FSGS (37).
and betaine) is very effective (50).

DGKE TMA Infection-Associated TMAs


Recessive (homozygous or compound heterozygous) STEC-HUS
DGKE mutations causing TMA was first reported in 2013 STEC-HUS is more common than aHUS, with a peak
(36), and this defined a cohort of patients with aHUS caused incidence in children aged ,5 years (Table 2). The
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018 TMA and the Kidney, Brocklebank et al. 11

presentation (6). Therefore, all individuals with TMA


Table 3. Eculizumab: practical considerations should be investigated for STEC-HUS (Figure 3). Rarely,
complement gene mutations are detected, and in these cases
Eculizumab: Practical Considerations
the clinical picture is unusually severe (13,65). In STEC-HUS
Before administration resulting in ESRD, it is recommended to screen for mutations
Meningococcal vaccination mandatory before transplantation. STEC-HUS is considered to be a self-
Tetravalent ACWY conjugated vaccine 1 limiting illness, with good outcomes relative to other TMAs:
multicomponent serogroup B vaccine 70% fully recover, the mortality rate is approximately 1%–4%,
Prophylaxis ESRD occurs in 3% of patients, and CKD occurs in 9%–18% of
Long-term antibiotic prophylaxis recommended patients (66). There is some evidence of complement activation
Administration (13), but the role of complement-inhibiting therapy has not yet
Intravenous infusion
been defined. A case series in 2011 first reported efficacy of
Maintenance therapy is administered every 14 d
Monitoring eculizumab in three children with severe disease (67), and
CH50 and AH50 ,10% a significant proportion of those affected in the 2011 E. coli
Eculizumab trough level 100 mg/ml O104 outbreak were treated with eculizumab; no benefit over
Hematologic indicators of TMA supportive care or PE was demonstrated in retrospective
Patient education analyses (60,68,69), although a direct comparison is difficult
Vigilance regarding meningococcal infection because of the inherent disadvantages of retrospective anal-
Counseling family members yses, for example there were no controls, patients received
Genetic screening multiple therapies administered with inconsistency, and those
When to stop? treated with eculizumab had more severe disease. In this
Continue during intercurrent illness, unless infection
self-limiting condition, only a randomized, controlled trial
with encapsulated organism, due to high risk of
TMA relapse in this context will resolve this controversy; one is underway in France
Withdrawal (Clinicaltrials.gov identifier: NCT02205541) and another is
Systematic investigation in clinical trials is being due to start recruiting in the United Kingdom. For now, the
undertaken recommendation remains supportive management.
May be appropriate in some patientsa, with
monitoring: liaise with specialist center Pneumococcal HUS
TMA may occur in adults and children in the context of
This guidance is based on our practice in the United Kingdom, invasive Streptococcus pneumoniae infection, and the high
and the 2015 Kidney Disease: Improving Global Outcomes morbidity and mortality rate usually reflects the severity of
Controversies Conference consensus recommendations (6).
the infection (70,71). In a recently published North American
Eculizumab (Soliris; Alexion Pharmaceuticals) is a recombinant
humanized mAb that functionally blocks C5, and is approved by
cohort, pneumococcal HUS was most commonly observed
the US Food and Drug Administration and the European in children aged ,2 years with pneumonia and empyema
Medicines Agency for use in atypical hemolytic uremic syn- who were extremely unwell and frequently required
drome. ACWY, meningococcal serotypes; CH50, total comple- prolonged hospitalization and intensive care unit admis-
ment hemolytic activity; AH50, alternative pathway hemolytic sion; the mortality rate was 3%, and 33% developed ESRD
activity; TMA, thrombotic microangiopathy; aHUS, atypical (72). The hypothesized mechanism is that pneumococci
hemolytic uremic syndrome. produce neuraminidase, which cleaves sialic residues from
a
For example, if there is no renal recovery after 6 months of glycoproteins on erythrocyte, platelet, and endothelial cell
treatment. membranes, exposing the cryptic Thomsen–Friedenreich
antigen (T antigen), to which IgM in the plasma can then
bind, resulting in cell damage and TMA (73). The Coombs
predominant causative pathogen is E. coli O157; enteric test is positive. Additionally, it has been suggested that
infection can occur sporadically or in the context of an cleavage of sialic acid may reduce FH binding, resulting in
outbreak, and around 15% of children with enteric infection impaired endothelial complement regulation, thus contrib-
develop HUS (59). The E. coli O104 outbreak in Europe in 2011 uting to disease pathogenesis (74). Supportive management
was exceptional because of the high STEC-HUS occurrence and treatment of the infection should be the focus.
rate (24%) and the high proportion of adults affected (60).
Shiga toxin translocates through the intestinal epithelium HIV-Associated TMA
and is thought to bind to leukocytes and circulate to TMA has been reported in association with HIV, more
the kidneys (61). The shiga toxin is then internalized via the commonly in the pre-highly active antiretroviral therapy
Gb3 receptor and is cleaved to release a protease into the era (75–77), and in association with lower CD41 cell counts
cytoplasm. This protease inhibits ribosomal function and and higher viral RNA levels (77). The pathogenic mecha-
protein synthesis, leading to cell death. It can also activate nisms are poorly understood, although endothelial damage
signaling pathways, inducing an inflammatory response in is thought to be the primary event. Again, there is no evidence
affected cells (62,63). for any treatment strategy other than supportive care and
The distinction between STEC-HUS and complement- antiretroviral treatment.
mediated aHUS may not be clinically obvious: approxi-
mately 5% of patients with STEC-HUS do not have Other Infections
diarrhea (64), and approximately 30% of patients with TMA has been reported in association with a wide range
complement-mediated aHUS have concurrent diarrhea at of viral, bacterial, fungal, and parasitic infections, although
12 Clinical Journal of the American Society of Nephrology

Table 4. Approach to kidney transplantation when thrombotic microangiopathy results in established renal disease

Complete Complement Evaluation before Renal Transplant Listing Is Recommended if Thrombotic Microangiopathy
Results in ESRDa

Risk Stratification Inclusion Criteria Management Strategy

High Pathogenic complement mutations Prophylaxis with eculizumab (KDIGO global


Previous early recurrence panel suggest plasma exchange and liver
transplantation may also be considered)
Moderate No complement mutation, or variant
of unknown significance
Isolated CFI mutation
Detectable anti-factor H antibody

Low Isolated CD46 mutationb No prophylaxis


Previously positive but now consistently
negative anti-factor H antibody

Delay transplantation until at least 6 months after starting dialysis as late renal recovery with eculizumab treatment has been reported.
Living related kidney donation should only be considered if a genetic or acquired cause is identified in the recipient and is not present in
the intended donor. Recommendations are on the basis of the 2015 Kidney Disease: Improving Global Outcomes (KDIGO) Controversies
Conference consensus recommendations (6).
a
Complete complement evaluation should include: serum levels of C3, C4, factor H, factor I; anti-factor H antibody; CD46 FACS; and
CFH, CFI, CD46, C3, and CFB genetics.
b
Allograft cells express functional CD46.

it is frequently unclear if this is a direct effect of the pathogen, manifest (81). PE is recommended on the basis of obser-
a side effect of treatment, or a trigger that unmasks a latent vational data and knowledge of pathogenesis (85).
complement defect (reviewed by Nester et al. [4]). For instance, The pathogenesis of preeclampsia and HELLP are poorly
Caprioli et al. (78) reported undefined infectious triggers in understood (80), although there is some evidence to sug-
.70% of patients with complement-mediated aHUS and gest that increased circulating levels of the syncytiotrophoblast-
CFH, CFI, or CD46 mutations. Supportive management derived antiangiogenic factors, soluble endoglin and the soluble
with appropriate treatment of the infection is recommended, form of the vascular endothelial growth factor receptor (sFlt-1),
and complement evaluation should be undertaken. may contribute to the observed endothelial dysfunction (79,86).
Although there is significant overlap in the clinical presentations
of HELLP and TMA, in HELLP, the predominant glomerular
Secondary TMAs
pathology is endotheliosis (79). In contrast to pregnancy-
Pregnancy-Associated TMA
The differential diagnosis of TMA in pregnancy includes associated aHUS, only a minority (8%–10%) of patients with
the primary TMAs complement-mediated aHUS and TTP, preeclampsia and HELLP syndrome harbor complement
as well as the TMA-like presentation occurring in the gene variants, mostly of unknown or nonpathogenic signif-
syndrome of hemolysis, elevated liver enzymes, and low icance (87). There is some evidence that complement is
platelets (HELLP) (79), which is part of the clinical spec- activated in preeclampsia and HELLP, although it is un-
trum of preeclampsia (80). clear whether it plays a role in pathogenesis; of note, pre-
In around 20% of women with aHUS, the disease onset eclampsia has occurred in women taking eculizumab for
appears to be triggered by pregnancy, occurring most often paroxysmal nocturnal hemoglobinuria (83) and aHUS (84).
in the postpartum period (81,82). Outcomes were histor- Management should be supportive, and the definitive
ically poor, with 76% developing ESRD despite PE (81). treatment of HELLP is expedited delivery, although expec-
More recently, it has become clear that a high proportion tant management may be considered if the woman is not at
will have identifiable complement mutations (.50%), and or near term (86).
that pregnancy acts as a trigger in those with an underlying In clinical practice, a pragmatic approach is usually taken
genetic predisposition (81). In the era of eculizumab, this in peripartum cases of TMA: if expedited delivery does not
high prevalence of complement mutations in pregnancy- result in resolution of the TMA then standard management
associated HUS provides the biologic rationale for com- is instituted (Figure 3).
plement inhibition. Increasing data in both paroxysmal
nocturnal hemoglobinuria (83) and aHUS (84) suggests that Drug-Mediated TMA
eculizumab is safe during pregnancy. In published reports TMA has been associated with a
Similarly, a significant proportion of women with TTP large number of drugs, although definitive causality has
present during pregnancy (80), particularly in the second only been established in relatively few (reviewed by Al-
and third trimesters (81). This might be explained by the Nouri et al. [88]). Drug-mediated TMA occurs by two main
physiologic increase in vWf during pregnancy, which mechanisms: immune-mediated damage and direct toxicity
consumes ADAMTS13, so in women with a genetic pre- (Table 5). For example, quinine induces the development of
disposition, its activity can fall low enough for TMA to autoantibodies reactive with either platelet glycoprotein
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018 TMA and the Kidney, Brocklebank et al. 13

Ib/IX or IIb/IIIa complexes, or both (89). In contrast, IFN-b The optimal treatment strategy remains controversial;
(90) and bevacizumab (91) cause TMAs by a dose-dependent favorable outcomes with eculizumab have been described
toxicity. There are no trials to guide management, and the in uncontrolled retrospective analyses (101) and there is
recommendation is supportive care and discontinuation of some evidence to suggest that complement is activated (13),
the causative drug. However, ticlopidine has been reported but prospective trials are likely needed in order to establish
to be associated with anti-ADAMTS13 antibodies with a consensus.
resultant TTP, and therefore PE is recommended (92). It is
crucial that a full evaluation is undertaken (Figure 3) even Severe Hypertension-Associated TMA
if drug-mediated TMA is suspected, including an urgent TMA can occur in association with severe hypertension,
ADAMTS13 assay. and should be managed with antihypertensive and sup-
portive treatment. The pathogenic mechanisms that result
De Novo TMA after Solid Organ Transplant in endothelial damage and TMA are unclear (102,103). Con-
De novo TMA has been reported to occur after kidney, versely, any patient with a TMA can have severe hypertension,
liver, pancreas, lung, and heart transplantation (93,94). The thus a clinical conundrum can arise in the initial evaluation of a
pathogenic mechanisms are not well understood, but it is patient presenting acutely with TMA. In practice, failure of BP
likely to be multifactorial, with ischemia-reperfusion in- control and supportive management to control the TMA will
jury, antibody-mediated rejection, viral infections such as often result in the pragmatic initiating of PE or eculizumab until
cytomegalovirus, and immunosuppressant drugs, especially complement evaluation is available. In the majority of patients
calcineurin inhibitors (CNIs), contributing to an “endothelial with TMA associated with severe hypertension, renal function
damaging milieu” (95). In one series of de novo TMA after and MAHA usually recover with management of BP (102,104).
kidney transplantation, complement mutations were iden- In a retrospective case series, genetic analysis identified rare
tified in 29% (96), providing a rationale for complement- variants in complement genes in patients for whom TMA was
inhibiting therapy especially where the primary diagnosis initially attributed to severe hypertension; eight out of nine
was not incompatible with complement-mediated aHUS. patients progressed to ESRD despite management of hyper-
In many cases, however, supportive treatment and address- tension (103). This may be particularly relevant when consid-
ing the precipitating factors (CNI discontinuation or dose ering kidney transplant assessment in an individual with ESRD
reduction, treatment of antibody-mediated rejection and viral attributed to severe hypertension with TMA, or if TMA occurs
infections) may be sufficient to stop the TMA (13). again after transplantation.

TMA after Bone Marrow Transplant Malignancy-Associated TMA


A multisystem TMA complicates 10%–40% of allogenic When TMA occurs in association with malignancy it can
bone marrow transplants (97,98) and is associated with be difficult to distinguish between TMA caused by chemo-
significant mortality, variously reported as 21%–75% (13). therapy and TMA caused by malignancy (105). It is possible
Again, this is likely to be multifactorial, with risk factors that the causative mechanism in disseminated malignancy
including CNIs, graft versus host disease, HLA mismatch, involves erythrocyte shearing after direct contact with
chemotherapy, radiation therapy, and infections (99). Rare microvascular embolic tumor cells (106). Prognosis is poor
genetic variants in aHUS associated genes (98) and FH because of malignancy-related mortality (107,108), and there
autoantibodies (100) have been infrequently reported in is no evidence to support any treatment strategy other than
post bone marrow transplant TMA. withdrawal of causative chemotherapy agents.

Table 5. Drugs with evidence supporting a causal association with thrombotic microangiopathya

Immune-Mediated TMA Direct Drug-Induced Toxicity Other

Quinine: Drug-dependent Immunosuppressive agents, Ticlopidine: ADAMTS13


antibodies e.g., calcineurin inhibitors: autoantibodyb
ciclosporin and tacrolimus Sirolimus
IFN-a, IFN-b
VEGF inhibitors, e g., bevacizumab,
sunitinib
Chemotherapeutic agents, e.g., gemcitabine,
mitomycin
Recreational drugs, e.g., cocaine

Even if drug-mediated TMA is suspected, a full evaluation should still be undertaken as shown in Figure 3, including an urgent
ADAMTS13 assay. TMA, thrombotic microangiopathy; VEGF, vascular endothelial growth factor; TTP, thrombotic thrombocytopenic
purpura; ADAMTS13, a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13.
a
TMA has been reported in association with many drugs, but definite causality has been established in relatively few (2,88,90,91), some
examples of which are included in this table.
b
Ticlopidine association with TTP reported, with severe ADAMTS13 deficiency due to inhibitory autoantibodies. This should be
managed as for TTP, with plasma exchange (46,92).
14 Clinical Journal of the American Society of Nephrology

TMA Associated with Glomerular Diseases required. The acute decision-making is time-critical: the
TMA can occur in association with IgA nephropathy, initial priority should be the consideration of TTP, because
ANCA-associated vasculitis, membranous nephropa- urgent management is imperative given the high mortality if
thy, FSGS, and MPGN/C3G, although it may be a his- untreated, and therefore in adults, PE should be instituted
topathologic finding without biochemical or clinical (after obtaining a sample for ADAMTS13 activity testing) on
manifestation (13). the presumption that it is TTP unless other evidence is avai-
In MPGN/C3G, the hereditary and acquired comple- lable that strongly suggests an alternative cause. If the
ment defects are similar, although subtly different to those ADAMTS13 result excludes TTP, then complement-mediated
seen in complement-mediated aHUS, and it is perhaps aHUS is presumed and treatment with eculizumab is rec-
unsurprising that concurrent (7) and sequential (109) ommended, pending the complete evaluation. In children, in
manifestation of C3G and TMA has been reported. Muta- whom PE may not be appropriate and is high risk, treatment
tions in CFH are observed in both complement-mediated with eculizumab has been recommended by Kidney Disease:
aHUS and C3G; the reason for this genetic pleiotropy is not Improving Global Outcomes before the availability of the
fully understood, but the location of the mutation within ADAMTS13 result, with the caveat that clinical deterioration
the gene may be important. In aHUS, the majority of mutations on eculizumab should necessitate immediate plasma therapy
are located at the C-terminal of FH, which binds to C3b and (6). An algorithm for the real-time investigation and man-
glycosaminoglycans on host cells to mediate cell surface agement of a patient presenting with TMA is illustrated in
protection (14), whereas in C3G, mutations are more often Figure 3. Once the full evaluation has been completed, the
located at the N-terminal of FH, which mediates comple- indication for eculizumab can be reviewed.
ment regulation in the fluid phase (110).
Genetic pleiotropy has also been reported in INF2- and
DGKE-mediated disease. In addition to the FSGS normally
Summary
seen in INF2-mediated disease (37) and the MPGN seen in In summary, TMA can manifest in a diverse range of
DGKE-mediated disease (55), TMAs have been reported in diseases and can be associated with significant morbidity
both cases that seemed nonresponsive to eculizumab. and mortality. For some forms of TMA, such as TTP and
complement-mediated aHUS, research has now defined the
TMA Associated with Autoimmune Diseases molecular mechanisms of disease leading to targeted ther-
TMA can occur in association with autoimmune diseases apy and improved patient outcomes. However, the opti-
including SLE, catastrophic antiphospholipid syndrome mal eculizumab treatment duration and the specific patient
(CAPS), and scleroderma renal crisis (SRC); again, the populations who will benefit remain undefined.
mechanisms remain unclear, although there is some evi-
dence of complement activation in SLE and CAPS (13). For Acknowledgment
SLE, CAPS, and SRC with TMA, treatment should be of the V.B. has received funding from the Northern Counties Kidney
underlying condition. The influence of TMA on outcome of Research Fund and is a Medical Research Council/Kidney Research
SLE is uncertain, as case series have reported both no UK Clinical Research Training Fellow. D.K. has received funding
difference and worse outcome (13); treatment should be from the Wellcome Trust, The Medical Research Council and Kidney
immunosuppression according to international guidelines, Research UK.
and there is no evidence to suggest that any additional
treatment specifically directed at the TMA is beneficial. The Disclosures
high mortality of SRC is significantly reduced with angiotensin- D.K. has received honoraria for consultancy work from Alexion
converting enzyme inhibitors (111). In CAPS, the interna- Pharmaceuticals, and is a director of and scientific advisor to Gyroscope
tional registry (522 episodes) reported the incidence of TMA Therapeutics.
to be 14%, and overall mortality was 37%, with no subgroup
analysis according to treatment. The most frequent treatment References
regimens used were anticoagulation plus glucocorticoids 1. Moake JL: Thrombotic microangiopathies. N Engl J Med 347:
(19%) or anticoagulation, glucocorticoids and PE with or 589–600, 2002
without intravenous Ig (18%), with only 0.2% receiving 2. George JN, Nester CM: Syndromes of thrombotic micro-
eculizumab (112). There is some evidence from mouse angiopathy. N Engl J Med 371: 654–666, 2014
3. Kerr H, Richards A: Complement-mediated injury and pro-
models and observational clinical data that complement
tection of endothelium: Lessons from atypical haemolytic
may be involved in the pathophysiology of CAPS; success- uraemic syndrome. Immunobiology 217: 195–203, 2012
ful use of eculizumab has been reported in case reports and 4. Nester CM, Barbour T, de Cordoba SR, Dragon-Durey MA,
series, and a prospective trial of preemptive eculizumab to Fremeaux-Bacchi V, Goodship TH, Kavanagh D, Noris M,
enable renal transplantation is ongoing (Clinicaltrials.gov Pickering M, Sanchez-Corral P, Skerka C, Zipfel P, Smith RJ:
Atypical aHUS: State of the art. Mol Immunol 67: 31–42, 2015
identifier: NCT01029587) (13).
5. Scully M, Cataland S, Coppo P, de la Rubia J, Friedman KD,
Kremer Hovinga J, Lammle B, Matsumoto M, Pavenski K, Sadler
E, Sarode R, Wu H; International Working Group for Thrombotic
Diagnosis and Management Thrombocytopenic Purpura: Consensus on the standardization
Establishing the cause of a TMA is usually possible after a of terminology in thrombotic thrombocytopenic purpura and
complete evaluation. However, the results of the requisite related thrombotic microangiopathies. J Thromb Haemost 15:
312–322, 2017
investigations are not available immediately, and in clinical 6. Goodship TH, Cook HT, Fakhouri F, Fervenza FC, Fremeaux-
practice, the scenario is often one of a severely ill patient Bacchi V, Kavanagh D, Nester CM, Noris M, Pickering MC,
with TMA for whom immediate management decisions are Rodriguez de Cordoba S, Roumenina LT, Sethi S, Smith RJ;
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018 TMA and the Kidney, Brocklebank et al. 15

Conference Participants: Atypical hemolytic uremic syndrome N, Nailescu C, Goodship TH, Marchbank KJ: Factor I autoan-
and C3 glomerulopathy: Conclusions from a “Kidney Disease: tibodies in patients with atypical hemolytic uremic syndrome:
Improving Global Outcomes” (KDIGO) controversies confer- Disease-associated or an epiphenomenon? Clin J Am Soc
ence. Kidney Int 91: 539–551, 2017 Nephrol 7: 417–426, 2012
7. Manenti L, Gnappi E, Vaglio A, Allegri L, Noris M, Bresin E, Pilato 24. Loirat C, Fakhouri F, Ariceta G, Besbas N, Bitzan M, Bjerre A,
FP, Valoti E, Pasquali S, Buzio C: Atypical haemolytic uraemic Coppo R, Emma F, Johnson S, Karpman D, Landau D, Langman
syndrome with underlying glomerulopathies. A case series and a CB, Lapeyraque AL, Licht C, Nester C, Pecoraro C, Riedl M, van
review of the literature. Nephrol Dial Transplant 28: 2246– de Kar NC, Van de Walle J, Vivarelli M, Frémeaux-Bacchi V; HUS
2259, 2013 International: An international consensus approach to the
8. El Karoui K, Hill GS, Karras A, Jacquot C, Moulonguet L, management of atypical hemolytic uremic syndrome in chil-
Kourilsky O, Frémeaux-Bacchi V, Delahousse M, Duong Van dren. Pediatr Nephrol 31: 15–39, 2016
Huyen JP, Loupy A, Bruneval P, Nochy D: A clinicopathologic 25. Hofer J, Giner T, Safouh H: Diagnosis and treatment of the he-
study of thrombotic microangiopathy in IgA nephropathy. J Am molytic uremic syndrome disease spectrum in developing re-
Soc Nephrol 23: 137–148, 2012 gions. Semin Thromb Hemost 40: 478–486, 2014
9. Barrera-Vargas A, Rosado-Canto R, Merayo-Chalico J, Arreola- 26. Legendre CM, Licht C, Muus P, Greenbaum LA, Babu S,
Guerra JM, Mejı́a-Vilet JM, Correa-Rotter R, Gómez-Martı́n D, Bedrosian C, Bingham C, Cohen DJ, Delmas Y, Douglas K, Eitner
Alcocer-Varela J: Renal thrombotic microangiopathy in pro- F, Feldkamp T, Fouque D, Furman RR, Gaber O, Herthelius M,
liferative lupus nephritis: Risk factors and clinical outcomes: A Hourmant M, Karpman D, Lebranchu Y, Mariat C, Menne J,
Case-Control study. J Clin Rheumatol 22: 235–240, 2016 Moulin B, Nürnberger J, Ogawa M, Remuzzi G, Richard T,
10. Phillips EH, Westwood JP, Brocklebank V, Wong EK, Tellez JO, Sberro-Soussan R, Severino B, Sheerin NS, Trivelli A,
Marchbank KJ, McGuckin S, Gale DP, Connolly J, Goodship TH, Zimmerhackl LB, Goodship T, Loirat C: Terminal complement
Kavanagh D, Scully MA: The role of ADAMTS-13 activity and inhibitor eculizumab in atypical hemolytic-uremic syndrome.
complement mutational analysis in differentiating acute throm- N Engl J Med 368: 2169–2181, 2013
botic microangiopathies. J Thromb Haemost 14: 175–185, 2016 27. Fremeaux-Bacchi V, Fakhouri F, Garnier A, Bienaimé F, Dragon-
11. Thurman JM, Nester CM: All things complement. Clin J Am Soc Durey MA, Ngo S, Moulin B, Servais A, Provot F, Rostaing L,
Nephrol 11: 1856–1866, 2016 Burtey S, Niaudet P, Deschênes G, Lebranchu Y, Zuber J, Loirat C:
12. Ricklin D, Hajishengallis G, Yang K, Lambris JD: Complement: A Genetics and outcome of atypical hemolytic uremic syndrome:
key system for immune surveillance and homeostasis. Nat Im- A nationwide French series comparing children and adults.
munol 11: 785–797, 2010 Clin J Am Soc Nephrol 8: 554–562, 2013
13. Brocklebank VKD: Complement C5 inhibiting therapy for the 28. Noris M, Caprioli J, Bresin E, Mossali C, Pianetti G, Gamba S,
thrombotic microangiopathies: Accumulating evidence, but Daina E, Fenili C, Castelletti F, Sorosina A, Piras R, Donadelli R,
not a panacea. Clin Kidney J, 2017 Maranta R, van der Meer I, Conway EM, Zipfel PF, Goodship TH,
14. Kavanagh D, Goodship TH, Richards A: Atypical hemolytic Remuzzi G: Relative role of genetic complement abnormalities
uremic syndrome. Semin Nephrol 33: 508–530, 2013 in sporadic and familial aHUS and their impact on clinical
15. Warwicker P, Goodship TH, Donne RL, Pirson Y, Nicholls A, phenotype. Clin J Am Soc Nephrol 5: 1844–1859, 2010
Ward RM, Turnpenny P, Goodship JA: Genetic studies into in- 29. Licht C, Greenbaum LA, Muus P, Babu S, Bedrosian CL, Cohen
herited and sporadic hemolytic uremic syndrome. Kidney Int 53: DJ, Delmas Y, Douglas K, Furman RR, Gaber OA, Goodship T,
836–844, 1998 Herthelius M, Hourmant M, Legendre CM, Remuzzi G, Sheerin
16. Le Quintrec M, Zuber J, Moulin B, Kamar N, Jablonski M, Lionet N, Trivelli A, Loirat C: Efficacy and safety of eculizumab in
A, Chatelet V, Mousson C, Mourad G, Bridoux F, Cassuto E, Loirat atypical hemolytic uremic syndrome from 2-year extensions of
C, Rondeau E, Delahousse M, Frémeaux-Bacchi V: Complement phase 2 studies. Kidney Int 87: 1061–1073, 2015
genes strongly predict recurrence and graft outcome in adult 30. Fakhouri F, Hourmant M, Campistol JM, Cataland SR, Espinosa
renal transplant recipients with atypical hemolytic and uremic M, Gaber AO, Menne J, Minetti EE, Provôt F, Rondeau E,
syndrome. Am J Transplant 13: 663–675, 2013 Ruggenenti P, Weekers LE, Ogawa M, Bedrosian CL, Legendre
17. Kavanagh D, Richards A, Goodship T, Jalanko H: Transplantation CM: Terminal complement inhibitor eculizumab in adult pa-
in atypical hemolytic uremic syndrome. Semin Thromb Hemost tients with atypical hemolytic uremic syndrome: A single-arm,
36: 653–659, 2010 open-label trial. Am J Kidney Dis 68: 84–93, 2016
18. Saland J: Liver-kidney transplantation to cure atypical HUS: Still 31. Greenbaum LA, Fila M, Ardissino G, Al-Akash SI, Evans J,
an option post-eculizumab? Pediatr Nephrol 29: 329–332, 2014 Henning P, Lieberman KV, Maringhini S, Pape L, Rees L, van de
19. Dragon-Durey MA, Loirat C, Cloarec S, Macher MA, Blouin J, Kar NC, Vande Walle J, Ogawa M, Bedrosian CL, Licht C:
Nivet H, Weiss L, Fridman WH, Frémeaux-Bacchi V: Anti-Factor Eculizumab is a safe and effective treatment in pediatric patients
H autoantibodies associated with atypical hemolytic uremic with atypical hemolytic uremic syndrome. Kidney Int 89:
syndrome. J Am Soc Nephrol 16: 555–563, 2005 701–711, 2016
20. Blanc C, Roumenina LT, Ashraf Y, Hyvärinen S, Sethi SK, Ranchin 32. Sheerin NS, Kavanagh D, Goodship TH, Johnson S: A national
B, Niaudet P, Loirat C, Gulati A, Bagga A, Fridman WH, Sautès- specialized service in England for atypical haemolytic uraemic
Fridman C, Jokiranta TS, Frémeaux-Bacchi V, Dragon-Durey syndrome-the first year’s experience. QJM 109: 27–33, 2016
MA: Overall neutralization of complement factor H by auto- 33. Wong EK, Kavanagh D: Anticomplement C5 therapy with
antibodies in the acute phase of the autoimmune form of atypical eculizumab for the treatment of paroxysmal nocturnal hemo-
hemolytic uremic syndrome. J Immunol 189: 3528–3537, 2012 globinuria and atypical hemolytic uremic syndrome. Transl Res
21. Moore I, Strain L, Pappworth I, Kavanagh D, Barlow PN, Herbert 165: 306–320, 2015
AP, Schmidt CQ, Staniforth SJ, Holmes LV, Ward R, Morgan L, 34. Fakhouri F, Fila M, Provôt F, Delmas Y, Barbet C, Châtelet V, Rafat
Goodship TH, Marchbank KJ: Association of factor H autoan- C, Cailliez M, Hogan J, Servais A, Karras A, Makdassi R, Louillet
tibodies with deletions of CFHR1, CFHR3, CFHR4, and with F, Coindre JP, Rondeau E, Loirat C, Frémeaux-Bacchi V: Patho-
mutations in CFH, CFI, CD46, and C3 in patients with atypical genic variants in complement genes and risk of atypical he-
hemolytic uremic syndrome. Blood 115: 379–387, 2010 molytic uremic syndrome relapse after eculizumab
22. Brocklebank V, Johnson S, Sheerin TP, Marks SD, Gilbert RD, discontinuation. Clin J Am Soc Nephrol 12: 50–59, 2017
Tyerman K, Kinoshita M, Awan A, Kaur A, Webb N, Hegde S, 35. Nishijima Y, Hirata H, Himeno A, Kida H, Matsumoto M,
Finlay E, Fitzpatrick M, Walsh PR, Wong EKS, Booth C, Kerecuk Takahashi R, Otani Y, Inoue K, Nagatomo I, Takeda Y, Kijima T,
L, Salama AD, Almond M, Inward C, Goodship TH, Sheerin NS, Tachibana I, Fujimura Y, Kumanogoh A: Drug-induced throm-
Marchbank KJ, Kavanagh D: Factor H: Autoantibody associated botic thrombocytopenic purpura successfully treated with re-
atypical haemolytic uraemic syndrome in children in the United combinant human soluble thrombomodulin. Intern Med 52:
Kingdom and Ireland [published online ahead of print July 24, 1111–1114, 2013
2017]. Kidney Int doi:10.1016/j.kint.2017.04.028 36. Lemaire M, Frémeaux-Bacchi V, Schaefer F, Choi M, Tang WH,
23. Kavanagh D, Pappworth IY, Anderson H, Hayes CM, Moore I, Le Quintrec M, Fakhouri F, Taque S, Nobili F, Martinez F, Ji W,
Hunze EM, Bennaceur K, Roversi P, Lea S, Strain L, Ward R, Plant Overton JD, Mane SM, Nürnberg G, Altmüller J, Thiele H, Morin D,
16 Clinical Journal of the American Society of Nephrology

Deschenes G, Baudouin V, Llanas B, Collard L, Majid MA, Simkova Remuzzi G, Noris M: Characterization of a new DGKE intronic
E, Nürnberg P, Rioux-Leclerc N, Moeckel GW, Gubler MC, Hwa J, mutation in genetically unsolved cases of familial atypical he-
Loirat C, Lifton RP: Recessive mutations in DGKE cause atypical molytic uremic syndrome. Clin J Am Soc Nephrol 10: 1011–
hemolytic-uremic syndrome. Nat Genet 45: 531–536, 2013 1019, 2015
37. Challis RC, Ring T, Xu Y, Wong EK, Flossmann O, Roberts IS, 53. Westland R, Bodria M, Carrea A, Lata S, Scolari F, Fremeaux-
Ahmed S, Wetherall M, Salkus G, Brocklebank V, Fester J, Strain Bacchi V, D’Agati VD, Lifton RP, Gharavi AG, Ghiggeri GM,
L, Wilson V, Wood KM, Marchbank KJ, Santibanez-Koref M, Sanna-Cherchi S: Phenotypic expansion of DGKE-associated
Goodship TH, Kavanagh D: Thrombotic microangiopathy in diseases. J Am Soc Nephrol 25: 1408–1414, 2014
inverted formin 2-mediated renal disease. J Am Soc Nephrol 28: 54. Sánchez Chinchilla D, Pinto S, Hoppe B, Adragna M, Lopez L,
1084–1091, 2017 Justa Roldan ML, Pe~ na A, Lopez Trascasa M, Sánchez-Corral P,
38. Benamu E, Montoya JG: Infections associated with the use of Rodrı́guez de Córdoba S: Complement mutations in dia-
eculizumab: Recommendations for prevention and prophylaxis. cylglycerol kinase-«-associated atypical hemolytic uremic
Curr Opin Infect Dis 29: 319–329, 2016 syndrome. Clin J Am Soc Nephrol 9: 1611–1619, 2014
39. Alashkar F, Vance C, Herich-Terhürne D, Preising N, Dührsen U, 55. Ozaltin F, Li B, Rauhauser A, An SW, Soylemezoglu O, Gonul II,
Röth A: Serologic response to meningococcal vaccination in Taskiran EZ, Ibsirlioglu T, Korkmaz E, Bilginer Y, Duzova A, Ozen
patients with paroxysmal nocturnal hemoglobinuria (PNH) S, Topaloglu R, Besbas N, Ashraf S, Du Y, Liang C, Chen P, Lu D,
chronically treated with the terminal complement inhibitor Vadnagara K, Arbuckle S, Lewis D, Wakeland B, Quigg RJ,
eculizumab. Ann Hematol 96: 589–596, 2017 Ransom RF, Wakeland EK, Topham MK, Bazan NG, Mohan C,
40. Cullinan N, Gorman KM, Riordan M, Waldron M, Goodship TH, Hildebrandt F, Bakkaloglu A, Huang CL, Attanasio M: DGKE
Awan A: Case report: Benefits and challenges of long-term variants cause a glomerular microangiopathy that mimics
eculizumab in atypical hemolytic uremic syndrome. Pediatrics membranoproliferative GN. J Am Soc Nephrol 24: 377–384,
135: e1506–e1509, 2015 2013
41. Struijk GH, Bouts AH, Rijkers GT, Kuin EA, ten Berge IJ, 56. Maga TK, Nishimura CJ, Weaver AE, Frees KL, Smith RJ: Mu-
Bemelman FJ: Meningococcal sepsis complicating eculizumab tations in alternative pathway complement proteins in American
treatment despite prior vaccination. Am J Transplant 13: 819– patients with atypical hemolytic uremic syndrome. Hum Mutat
820, 2013 31: E1445–E1460, 2010
42. Hayes W, Tschumi S, Ling SC, Feber J, Kirschfink M, Licht C: 57. Delvaeye M, Noris M, De Vriese A, Esmon CT, Esmon NL, Ferrell
Eculizumab hepatotoxicity in pediatric aHUS. Pediatr Nephrol G, Del-Favero J, Plaisance S, Claes B, Lambrechts D, Zoja C,
30: 775–781, 2015 Remuzzi G, Conway EM: Thrombomodulin mutations in atyp-
43. Herlitz LC, Bomback AS, Markowitz GS, Stokes MB, Smith RN, ical hemolytic-uremic syndrome. N Engl J Med 361: 345–357,
Colvin RB, Appel GB, D’Agati VD: Pathology after eculizumab in
2009
dense deposit disease and C3 GN. J Am Soc Nephrol 23: 1229– 58. Bu F, Maga T, Meyer NC, Wang K, Thomas CP, Nester CM, Smith
1237, 2012
RJ: Comprehensive genetic analysis of complement and co-
44. George JN: The association of pregnancy with thrombotic
agulation genes in atypical hemolytic uremic syndrome. J Am
thrombocytopenic purpura-hemolytic uremic syndrome. Curr
Soc Nephrol 25: 55–64, 2014
Opin Hematol 10: 339–344, 2003
59. Tarr PI, Gordon CA, Chandler WL: Shiga-toxin-producing Es-
45. Zuber J, Fakhouri F, Roumenina LT, Loirat C, Frémeaux-Bacchi V;
cherichia coli and haemolytic uraemic syndrome. Lancet 365:
French Study Group for aHUS/C3G: Use of eculizumab for
1073–1086, 2005
atypical haemolytic uraemic syndrome and C3 glomer-
60. Kielstein JT, Beutel G, Fleig S, Steinhoff J, Meyer TN, Hafer C,
ulopathies. Nat Rev Nephrol 8: 643–657, 2012
Kuhlmann U, Bramstedt J, Panzer U, Vischedyk M, Busch V, Ries
46. Schwartz J, Padmanabhan A, Aqui N, Balogun RA, Connelly-
Smith L, Delaney M, Dunbar NM, Witt V, Wu Y, Shaz BH: W, Mitzner S, Mees S, Stracke S, Nürnberger J, Gerke P, Wiesner
Guidelines on the use of therapeutic apheresis in clinical M, Sucke B, Abu-Tair M, Kribben A, Klause N, Schindler R,
practice-evidence-based approach from the writing committee Merkel F, Schnatter S, Dorresteijn EM, Samuelsson O,
of the american society for apheresis: The seventh special issue. Brunkhorst R; Collaborators of the DGfN STEC-HUS registry:
J Clin Apher 31: 149–162, 2016 Best supportive care and therapeutic plasma exchange with or
47. Scully M, McDonald V, Cavenagh J, Hunt BJ, Longair I, Cohen H, without eculizumab in Shiga-toxin-producing E. coli O104:H4
Machin SJ: A phase 2 study of the safety and efficacy of rituximab induced haemolytic-uraemic syndrome: An analysis of the
with plasma exchange in acute acquired thrombotic thrombo- German STEC-HUS registry. Nephrol Dial Transplant 27: 3807–
cytopenic purpura. Blood 118: 1746–1753, 2011 3815, 2012
48. Clark WF, Rock G, Barth D, Arnold DM, Webert KE, Yenson PR, 61. Brigotti M, Caprioli A, Tozzi AE, Tazzari PL, Ricci F, Conte R,
Kelton JG, Li L, Foley SR; members of Canadian Apheresis Carnicelli D, Procaccino MA, Minelli F, Ferretti AV, Paglialonga
Group: A phase-II sequential case-series study of all patients F, Edefonti A, Rizzoni G: Shiga toxins present in the gut and in the
presenting to four plasma exchange centres with presumed re- polymorphonuclear leukocytes circulating in the blood of
lapsed/refractory thrombotic thrombocytopenic purpura treated children with hemolytic-uremic syndrome. J Clin Microbiol 44:
with rituximab. Br J Haematol 170: 208–217, 2015 313–317, 2006
49. Benhamou Y, Paintaud G, Azoulay E, Poullin P, Galicier L, 62. Obrig TG: Escherichia coli Shiga toxin mechanisms of action in
Desvignes C, Baudel JL, Peltier J, Mira JP, Pene F, Presne C, Saheb renal disease. Toxins (Basel) 2: 2769–2794, 2010
S, Deligny C, Rousseau A, Feger F, Veyradier A, Coppo P; French 63. Hughes AK, Ergonul Z, Stricklett PK, Kohan DE: Molecular
Reference Center for Thrombotic M: Efficacy of a rituximab basis for high renal cell sensitivity to the cytotoxic effects of
regimen based on B cell depletion in thrombotic thrombocy- shigatoxin-1: Upregulation of globotriaosylceramide expres-
topenic purpura with suboptimal response to standard treat- sion. J Am Soc Nephrol 13: 2239–2245, 2002
ment: Results of a phase II, multicenter non-comparative study. 64. Brandt JR, Fouser LS, Watkins SL, Zelikovic I, Tarr PI, Nazar-
Am J Hematol 91: 1246–1251, 2016 Stewart V, Avner ED: Escherichia coli O 157:H7-associated
50. Beck BB, van Spronsen F, Diepstra A, Berger RM, Komhoff M: hemolytic-uremic syndrome after ingestion of contaminated
Renal thrombotic microangiopathy in patients with cblC defect: hamburgers. J Pediatr 125: 519–526, 1994
Review of an under-recognized entity. Pediatr Nephrol 32: 733– 65. Dowen FWK, Brown AL, Palfrey J, Kavanagh D, Brocklebank V:
741, 2017 Rare genetic variants in Shiga toxin–associated haemolytic
51. Bruneau S, Néel M, Roumenina LT, Frimat M, Laurent L, uraemic syndrome: Genetic analysis prior to transplantation is
Frémeaux-Bacchi V, Fakhouri F: Loss of DGK« induces endo- essential. Clin Kidney J 10: 490–493, 2017
thelial cell activation and death independently of complement 66. Spinale JM, Ruebner RL, Copelovitch L, Kaplan BS: Long-term
activation. Blood 125: 1038–1046, 2015 outcomes of Shiga toxin hemolytic uremic syndrome. Pediatr
52. Mele C, Lemaire M, Iatropoulos P, Piras R, Bresin E, Bettoni S, Nephrol 28: 2097–2105, 2013
Bick D, Helbling D, Veith R, Valoti E, Donadelli R, Murer L, 67. Lapeyraque AL, Malina M, Fremeaux-Bacchi V, Boppel T,
Neunhäuserer M, Breno M, Frémeaux-Bacchi V, Lifton R, Kirschfink M, Oualha M, Proulx F, Clermont MJ, Le Deist F,
Clin J Am Soc Nephrol 13: ccc–ccc, March, 2018 TMA and the Kidney, Brocklebank et al. 17

Niaudet P, Schaefer F: Eculizumab in severe Shiga-toxin- 83. Kelly RJ, Höchsmann B, Szer J, Kulasekararaj A, de Guibert S,
associated HUS. N Engl J Med 364: 2561–2563, 2011 Röth A, Weitz IC, Armstrong E, Risitano AM, Patriquin CJ, Terriou
68. Menne J, Nitschke M, Stingele R, Abu-Tair M, Beneke J, L, Muus P, Hill A, Turner MP, Schrezenmeier H, Peffault de Latour
Bramstedt J, Bremer JP, Brunkhorst R, Busch V, Dengler R, R: Eculizumab in pregnant patients with paroxysmal nocturnal
Deuschl G, Fellermann K, Fickenscher H, Gerigk C, Goettsche hemoglobinuria. N Engl J Med 373: 1032–1039, 2015
A, Greeve J, Hafer C, Hagenmüller F, Haller H, Herget-Rosenthal 84. Servais A, Devillard N, Fremeaux-Bacchi V, Hummel A,
S, Hertenstein B, Hofmann C, Lang M, Kielstein JT, Klostermeier Salomon L, Contin-Bordes C, Gomer H, Legendre C, Delmas Y:
UC, Knobloch J, Kuehbacher M, Kunzendorf U, Lehnert H, Atypical haemolytic uraemic syndrome and pregnancy: Out-
Manns MP, Menne TF, Meyer TN, Michael C, Münte T, come with ongoing eculizumab. Nephrol Dialysis Transplant
Neumann-Grutzeck C, Nuernberger J, Pavenstaedt H, Ramazan 31: 2122–2130, 2016
L, Renders L, Repenthin J, Ries W, Rohr A, Rump LC, Samuelsson 85. Scully M, Thomas M, Underwood M, Watson H, Langley K,
O, Sayk F, Schmidt BM, Schnatter S, Schöcklmann H, Schreiber Camilleri RS, Clark A, Creagh D, Rayment R, Mcdonald V, Roy A,
S, von Seydewitz CU, Steinhoff J, Stracke S, Suerbaum S, van de Evans G, McGuckin S, Ni Ainle F, Maclean R, Lester W, Nash M,
Loo A, Vischedyk M, Weissenborn K, Wellhöner P, Wiesner M, Scott R, O Brien P; collaborators of the UK TTP Registry:
Zeissig S, Büning J, Schiffer M, Kuehbacher T; EHEC-HUS Thrombotic thrombocytopenic purpura and pregnancy: Pre-
consortium: Validation of treatment strategies for entero- sentation, management, and subsequent pregnancy outcomes.
haemorrhagic Escherichia coli O104:H4 induced haemolytic Blood 124: 211–219, 2014
uraemic syndrome: Case-control study. BMJ 345: e4565, 2012 86. Steegers EA, von Dadelszen P, Duvekot JJ, Pijnenborg R: Pre-
69. Loos S, Ahlenstiel T, Kranz B, Staude H, Pape L, Härtel C, Vester eclampsia. Lancet 376: 631–644, 2010
U, Buchtala L, Benz K, Hoppe B, Beringer O, Krause M, Müller D, 87. Salmon JE, Heuser C, Triebwasser M, Liszewski MK, Kavanagh
Pohl M, Lemke J, Hillebrand G, Kreuzer M, König J, Wigger M, D, Roumenina L, Branch DW, Goodship T, Fremeaux-Bacchi V,
Konrad M, Haffner D, Oh J, Kemper MJ: An outbreak of Shiga Atkinson JP: Mutations in complement regulatory proteins
toxin-producing Escherichia coli O104:H4 hemolytic uremic predispose to preeclampsia: A genetic analysis of the PROMISSE
syndrome in Germany: Presentation and short-term outcome in cohort. PLoS Med 8: e1001013, 2011
children. Clin Infect Dis 55: 753–759, 2012 88. Al-Nouri ZL, Reese JA, Terrell DR, Vesely SK, George JN: Drug-
70. Johnson S, Waters A: Is complement a culprit in infection- induced thrombotic microangiopathy: A systematic review of
induced forms of haemolytic uraemic syndrome? Immunobi- published reports. Blood 125: 616–618, 2015
ology 217: 235–243, 2012 89. Gottschall JL, Elliot W, Lianos E, McFarland JG, Wolfmeyer K,
71. Spinale JM, Ruebner RL, Kaplan BS, Copelovitch L: Update on Aster RH: Quinine-induced immune thrombocytopenia asso-
Streptococcus pneumoniae associated hemolytic uremic syn- ciated with hemolytic uremic syndrome: A new clinical entity.
drome. Curr Opin Pediatr 25: 203–208, 2013 Blood 77: 306–310, 1991
72. Banerjee R, Hersh AL, Newland J, Beekmann SE, Polgreen PM, 90. Kavanagh D, McGlasson S, Jury A, Williams J, Scolding N,
Bender J, Shaw J, Copelovitch L, Kaplan BS, Shah SS; Emerging Bellamy C, Gunther C, Ritchie D, Gale DP, Kanwar YS, Challis R,
Infections Network Hemolytic-Uremic Syndrome Study Group: Buist H, Overell J, Weller B, Flossmann O, Blunden M, Meyer EP,
Streptococcus pneumoniae-associated hemolytic uremic syn- Krucker T, Evans SJ, Campbell IL, Jackson AP, Chandran S, Hunt
drome among children in North America. Pediatr Infect Dis DP: Type I interferon causes thrombotic microangiopathy by a
J 30: 736–739, 2011 dose-dependent toxic effect on the microvasculature. Blood
73. Kavanagh D, Goodship TH, Richards A: Atypical haemolytic 128: 2824–2833, 2016
uraemic syndrome. Br Med Bull 77-78: 5–22, 2006 91. Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M,
74. Blaum BS, Hannan JP, Herbert AP, Kavanagh D, Uhrı́n D, Stehle Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH,
T: Structural basis for sialic acid-mediated self-recognition by Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE: VEGF
complement factor H. Nat Chem Biol 11: 77–82, 2015 inhibition and renal thrombotic microangiopathy. N Engl J Med
75. Alpers CE: Light at the end of the TUNEL: HIV-associated 358: 1129–1136, 2008
thrombotic microangiopathy. Kidney Int 63: 385–396, 2003 92. Bennett CL, Jacob S, Dunn BL, Georgantopoulos P, Zheng XL,
76. Peraldi MN, Maslo C, Akposso K, Mougenot B, Rondeau E, Sraer Kwaan HC, McKoy JM, Magwood JS, Qureshi ZP, Bandarenko N,
JD: Acute renal failure in the course of HIV infection: A single- Winters JL, Raife TJ, Carey PM, Sarode R, Kiss JE, Danielson C,
institution retrospective study of ninety-two patients and sixty Ortel TL, Clark WF, Ablin RJ, Rock G, Matsumoto M, Fujimura Y:
renal biopsies. Nephrol Dial Transplant 14: 1578–1585, 1999 Ticlopidine-associated ADAMTS13 activity deficient throm-
77. Becker S, Fusco G, Fusco J, Balu R, Gangjee S, Brennan C, botic thrombocytopenic purpura in 22 persons in Japan: A report
Feinberg J; Collaborations in HIVORUSC: HIV-associated from the Southern Network on Adverse Reactions (SONAR).
thrombotic microangiopathy in the era of highly active anti- Br J Haematol 161: 896–898, 2013
retroviral therapy: An observational study. Clin Infect Dis 39 93. Verbiest A, Pirenne J, Dierickx D: De novo thrombotic micro-
[Suppl 5]: S267–S275, 2004 angiopathy after non-renal solid organ transplantation. Blood
78. Caprioli J, Noris M, Brioschi S, Pianetti G, Castelletti F, Rev 28: 269–279, 2014
Bettinaglio P, Mele C, Bresin E, Cassis L, Gamba S, Porrati F, 94. Ponticelli C, Banfi G: Thrombotic microangiopathy after kidney
Bucchioni S, Monteferrante G, Fang CJ, Liszewski MK, Kavanagh transplantation. Transpl Int 19: 789–794, 2006
D, Atkinson JP, Remuzzi G; International Registry of Recurrent 95. Zuber J, Le Quintrec M, Morris H, Frémeaux-Bacchi V, Loirat
and Familial HUS/TTP: Genetics of HUS: The impact of MCP, C, Legendre C: Targeted strategies in the prevention and
CFH, and IF mutations on clinical presentation, response to management of atypical HUS recurrence after kidney
treatment, and outcome. Blood 108: 1267–1279, 2006 transplantation. Transplant Rev (Orlando) 27: 117–125,
79. Fakhouri F, Vercel C, Frémeaux-Bacchi V: Obstetric nephrology: 2013
AKI and thrombotic microangiopathies in pregnancy. Clin J Am 96. Le Quintrec M, Lionet A, Kamar N, Karras A, Barbier S, Buchler
Soc Nephrol 7: 2100–2106, 2012 M, Fakhouri F, Provost F, Fridman WH, Thervet E, Legendre C,
80. George JN, Nester CM, McIntosh JJ: Syndromes of thrombotic Zuber J, Frémeaux-Bacchi V: Complement mutation-associated
microangiopathy associated with pregnancy. Hematology (Am de novo thrombotic microangiopathy following kidney trans-
Soc Hematol Educ Program) 2015: 644–648, 2015 plantation. Am J Transplant 8: 1694–1701, 2008
81. Fakhouri F: Pregnancy-related thrombotic microangiopathies: 97. Chapin J, Shore T, Forsberg P, Desman G, Van Besien K, Laurence
Clues from complement biology. Transfus Apher Sci 54: 199– J: Hematopoietic transplant-associated thrombotic micro-
202, 2016 angiopathy: Case report and review of diagnosis and treatments.
82. Bruel A, Kavanagh D, Noris M, Delmas Y, Wong EKS, Bresin E, Clin Adv Hematol Oncol 12: 565–573, 2014
Provôt F, Brocklebank V, Mele C, Remuzzi G, Loirat C, 98. Jodele S, Zhang K, Zou F, Laskin B, Dandoy CE, Myers KC, Lane
Frémeaux-Bacchi V, Fakhouri F: Hemolytic uremic syndrome in A, Meller J, Medvedovic M, Chen J, Davies SM: The genetic
pregnancy and postpartum. Clin J Am Soc Nephrol 12: 1237– fingerprint of susceptibility for transplant-associated thrombotic
1247, 2017 microangiopathy. Blood 127: 989–996, 2016
18 Clinical Journal of the American Society of Nephrology

99. Dhakal P, Bhatt VR: Is complement blockade an acceptable 115. Lynn RM, O’Brien SJ, Taylor CM, Adak GK, Chart H, Cheasty T,
therapeutic strategy for hematopoietic cell transplant-associated Coia JE, Gillespie IA, Locking ME, Reilly WJ, Smith HR, Waters A,
thrombotic microangiopathy? Bone Marrow Transplant 52: Willshaw GA: Childhood hemolytic uremic syndrome, United
352–356, 2017 Kingdom and Ireland. Emerg Infect Dis 11: 590–596, 2005
100. Jodele S, Licht C, Goebel J, Dixon BP, Zhang K, Sivakumaran TA, 116. Noris M, Mescia F, Remuzzi G: STEC-HUS, atypical HUS and
Davies SM, Pluthero FG, Lu L, Laskin BL: Abnormalities in the TTP are all diseases of complement activation. Nat Rev Nephrol
alternative pathway of complement in children with hemato- 8: 622–633, 2012
poietic stem cell transplant-associated thrombotic micro- 117. Waters AM, Kerecuk L, Luk D, Haq MR, Fitzpatrick MM, Gilbert
angiopathy. Blood 122: 2003–2007, 2013 RD, Inward C, Jones C, Pichon B, Reid C, Slack MP, Van’t Hoff W,
101. Jodele S, Dandoy CE, Myers KC, El-Bietar J, Nelson A, Wallace G, Dillon MJ, Taylor CM, Tullus K: Hemolytic uremic syndrome
Laskin BL: New approaches in the diagnosis, pathophysiology, associated with invasive pneumococcal disease: The United
and treatment of pediatric hematopoietic stem cell transplantation- kingdom experience. J Pediatr 151: 140–144, 2007
associated thrombotic microangiopathy. Transfus Apher Sci 54: 118. Cochran JB, Panzarino VM, Maes LY, Tecklenburg FW:
181–190, 2016 Pneumococcus-induced T-antigen activation in hemolytic ure-
102. Van Laecke S, Van Biesen W: Severe hypertension with renal mic syndrome and anemia. Pediatr Nephrol 19: 317–321, 2004
thrombotic microangiopathy: What happened to the usual 119. Prestidge C, Wong W: Ten years of pneumococcal-associated
suspect? Kidney Int 91: 1271–1274, 2017 haemolytic uraemic syndrome in New Zealand children.
103. Timmermans SAMEG, Abdul-Hamid MA, Vanderlocht J, J Paediatr Child Health 45: 731–735, 2009
Damoiseaux JGMC, Reutelingsperger CP, van Paassen P; 120. Feinberg JE, Hurwitz S, Cooper D, Sattler FR, MacGregor RR,
Limburg Renal Registry: Patients with hypertension-associated Powderly W, Holland GN, Griffiths PD, Pollard RB, Youle M, Gill
thrombotic microangiopathy may present with complement MJ, Holland FJ, Power ME, Owens S, Coakley D, Fry J, Jacobson
abnormalities. Kidney Int 91: 1420–1425, 2017 MA: A randomized, double-blind trial of valaciclovir pro-
104. Shavit L, Reinus C, Slotki I: Severe renal failure and micro- phylaxis for cytomegalovirus disease in patients with ad-
angiopathic hemolysis induced by malignant hypertension–case vanced human immunodeficiency virus infection. AIDS
series and review of literature. Clin Nephrol 73: 147–152, 2010 Clinical Trials Group Protocol 204/Glaxo Wellcome 123-014
105. Izzedine H, Perazella MA: Thrombotic microangiopathy, can- International CMV Prophylaxis study group. J Infect Dis 177:
cer, and cancer drugs. Am J Kidney Dis 66: 857–868, 2015 48–56, 1998
106. George JN: Systemic malignancies as a cause of unexpected 121. Crovetto F, Borsa N, Acaia B, Nishimura C, Frees K, Smith RJ,
Peyvandi F, Palla R, Cugno M, Tedeschi S, Castorina P,
microangiopathic hemolytic anemia and thrombocytopenia.
Somigliana E, Ardissino G, Fedele L: The genetics of the alter-
Oncology (Williston Park) 25: 908–914, 2011
native pathway of complement in the pathogenesis of HELLP
107. Lesesne JB, Rothschild N, Erickson B, Korec S, Sisk R, Keller J,
syndrome. J Matern Fetal Neonatal Med 25: 2322–2325, 2012
Arbus M, Woolley PV, Chiazze L, Schein PS: Cancer-associated
122. Pisoni R, Ruggenenti P, Remuzzi G: Drug-induced thrombotic
hemolytic-uremic syndrome: Analysis of 85 cases from a na-
microangiopathy: Incidence, prevention and management.
tional registry. J Clin Oncol 7: 781–789, 1989
Drug Saf 24: 491–501, 2001
108. Oberic L, Buffet M, Schwarzinger M, Veyradier A, Clabault K,
123. Reynolds JC, Agodoa LY, Yuan CM, Abbott KC: Thrombotic
Malot S, Schleinitz N, Valla D, Galicier L, Bengrine-Lefèvre L, microangiopathy after renal transplantation in the United States.
Gorin NC, Coppo P; Reference Center for the Management of Am J Kidney Dis 42: 1058–1068, 2003
Thrombotic Microangiopathies: Cancer awareness in atypical 124. van den Born BJ, Honnebier UP, Koopmans RP, van Montfrans
thrombotic microangiopathies. Oncologist 14: 769–779, 2009 GA: Microangiopathic hemolysis and renal failure in malignant
109. Cooper M, McGraw ME, Unsworth DJ, Mathieson P: Familial hypertension. Hypertension 45: 246–251, 2005
mesangio-capillary glomerulonephritis with initial presentation 125. van den Born BJ, Koopmans RP, van Montfrans GA: The renin-
as haemolytic uraemic syndrome. Nephrol Dial Transplant 19: angiotensin system in malignant hypertension revisited: plasma
230–233, 2004 renin activity, microangiopathic hemolysis, and renal failure in
110. Zipfel PF, Skerka C, Chen Q, Wiech T, Goodship T, Johnson S, malignant hypertension. Am J Hypertens 20: 900–906, 2007
Fremeaux-Bacchi V, Nester C, de Córdoba SR, Noris M, 126. Akimoto T, Muto S, Ito C, Takahashi H, Takeda S, Ando Y, Kusano
Pickering M, Smith R: The role of complement in C3 glomer- E: Clinical features of malignant hypertension with thrombotic
ulopathy. Mol Immunol 67: 21–30, 2015 microangiopathy. Clin Exp Hypertens 33: 77–83, 2011
111. Woodworth TG, Suliman YA, Furst DE, Clements P: Scleroderma 127. Amraoui F, Bos S, Vogt L, van den Born BJ: Long-term renal
renal crisis and renal involvement in systemic sclerosis. Nat Rev outcome in patients with malignant hypertension: A retro-
Nephrol 12: 678–691, 2016 spective cohort study. BMC Nephrol 13: 71, 2012
112. Rodrı́guez-Pintó I, Moitinho M, Santacreu I, Shoenfeld Y, Erkan D, 128. Barber C, Herzenberg A, Aghdassi E, Su J, Lou W, Qian G, Yip J,
Espinosa G, Cervera R; CAPS Registry Project Group (European Fo- Nasr SH, Thomas D, Scholey JW, Wither J, Urowitz M, Gladman
rumon AntiphospholipidAntibodies):Catastrophicantiphospholipid D, Reich H, Fortin PR: Evaluation of clinical outcomes and renal
syndrome (CAPS): Descriptive analysis of 500 patients from the in- vascular pathology among patients with lupus. Clin J Am Soc
ternational CAPS registry. Autoimmun Rev 15: 1120–1124, 2016 Nephrol 7: 757–764, 2012
113. Fakhouri F, Zuber J, Frémeaux-Bacchi V, Loirat C: Haemolytic 129. Ghossein C, Varga J, Fenves AZ: Recent developments in the
uraemic syndrome [published online ahead of print February 25, classification, evaluation, pathophysiology, and management of
2017]. Lancet doi:10.1016/S0140-6736(17)30062-4 scleroderma renal crisis. Curr Rheumatol Rep 18: 5, 2016
114. Michael M, Elliott EJ, Craig JC, Ridley G, Hodson EM: Inter-
ventions for hemolytic uremic syndrome and thrombotic
thrombocytopenic purpura: A systematic review of randomized Published online ahead of print. Publication date available at www.
controlled trials. Am J Kidney Dis 53: 259–272, 2009 cjasn.org.

You might also like