Você está na página 1de 20

Journal of Internal Medicine 2006; 260: 285–304 doi:10.1111/j.1365-2796.2006.01697.

REVIEW

Clinical aspects of parvovirus B19 infection


K. BROLIDEN, T. TOLFVENSTAM & O. NORBECK
From the Department of Medicine, Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University
Hospital, Stockholm, Sweden

Abstract. Broliden K, Tolfvenstam T, Norbeck O and can mimic or trigger autoimmune inflammatory
(Karolinska Institutet, Karolinska University Hospi- disorders. Another important clinical aspect to
tal, Stockholm, Sweden). Clinical aspects of parvo- consider is the risk of infection through B19-con-
virus B19 infection (Review). J Intern Med 2006; taminated blood products. Recent advances in
260: 285–304. diagnosis and pathogenesis, new insights in the
cellular immune response and newly discovered
Parvovirus B19 is a significant human pathogen
genotypes of human parvoviruses form a platform
that causes a wide spectrum of clinical complica-
for the development of modern therapeutic and
tions ranging from mild, self-limiting erythema in-
prophylactic alternatives.
fectiosum in immunocompetent children to lethal
cytopenias in immunocompromised patients and Keywords: B19, clinical management, diagnosis,
intrauterine foetal death in primary infected preg- parvovirus, pathogenesis, pregnancy
nant women. The infection may also be persistent

reaction (PCR) assays [9–13]. Recently, another


Viral characteristics
human parvovirus provisionally named human
Parvovirus B19 (‘B19’) is a member of the erythro- bocavirus was cloned by molecular screening of
viruses, named so because of a pronounced tropism respiratory tract samples from children with lower
for erythroid precursor cells. It is a single-stranded respiratory tract infections [14]. Although these
nonenveloped DNA virus and one of the smallest novel genotypes are capable of human infection and
viruses known to infect mammalian cells [1]. B19 is were initially detected in individuals with B19-
genetically stable, and sequenced isolates have related disease, the potential pathology has yet to be
shown low variability in the range of a few established [8, 12, 15].
percentage for the two capsid proteins VP1 and
VP2, and even lower for the nonstructural protein
Infection and tropism
NS1 [2–4] (Fig. 1). Differences in clinical manifes-
tations of B19 infection have not been explained by B19 is thought to exclusively infect humans, and
B19 sequence variability [5–8]. Novel viral isolates shows a pronounced tropism for erythroid precur-
have now also been sequenced that are similar to sors [16–18]. The virus is very stable and often
B19 only to about 88–90%, and are proposed to survives in blood products despite standard proce-
compose two novel genotypes within the erythrovi- dures for viral elimination [19, 20]. B19 uses at least
rus genus. These variants are not readily detected three cellular receptors for cell attachment and
by available B19 serology and polymerase chain entry. The first to be identified was the glycolipid
 2006 Blackwell Publishing Ltd 285
286 K . B R O L I D E N et al.

seroprevalence is high, viraemia or presence of viral


DNA is rare in healthy individuals. The frequency
of B19 viraemia in voluntary blood donors has
been estimated at rates of 1 : 167 to 1 : 35 000 [35,
39–43]. The frequency varies greatly depending on
epidemic periods and sensitivity of the methodology
used. Although B19 viraemia in blood is rare,
presence of B19 DNA in bone marrow (BM) samples
can be found by PCR in 2% of healthy individuals
and in up to 10% of children with haematologic
malignancies without concomitant viraemia [31,
44, 45]. The persistency of B19 DNA may represent
both infectious virus and residual DNA from remote
infection.
The incidence of infection shows a seasonal
Fig. 1 Parvoviruses are symmetrical icosahedral particles. They variation in temperate climates, being more com-
form small capsids and contain a DNA genome. The viral genome
mon during winter and early spring [46]. Epidemics
encodes only three proteins with known function, the nonstruc-
tural protein NS-1 and two capsid proteins VP1 and VP2. are noted at intervals of about 3–4 years, with
outbreaks of erythema infectiosum (EI) and B19-
globoside, also known as the blood group P antigen related disease. B19 is normally transmitted through
(P-ag) [21, 22]. P-ag is present on the haematopoi- the respiratory route, but can also be transmitted
etic precursors, erythroblasts and megakaryocytes vertically from the mother to the foetus, through BM
that B19 shows tropism for and also on a variety of and organ transplantations, and via transfused
other cells including endothelial cells, foetal myo- blood products [42, 47–51]. As most infections
cytes, and placental trophoblasts [23–27]. These occur in children aged 5–15 years, adults at risk are
latter cells have been thought to represent nonper- parents of children in that age group, or those
missive cells into which B19 can enter but not working at day care centres or schools [52–54]. The
produce complete virions. For a productive infection secondary attack rate during epidemics of EI is about
to occur, a co-receptor has been defined, the a5b1- 50% in susceptible children and 25% in susceptible
integrin. This integrin is involved in cell adhesion teachers [53, 55]. Nosocomial transmission may
and is expressed on erythroid progenitors, which occur and although rare, is a potential risk in
might explain the tropism of B19 and the active paediatric wards for immunocompromised children
replication seen in these cells [28, 29]. Another [56–58].
molecule, the Ku80 autoantigen, has also been
suggested as a co-receptor for B19 allowing entry
Blood product safety
into cells [30].
The genomic stability and the absence of a lipid
envelope make B19 resistant to heat inactivation
Epidemiology
and solvent detergents normally used to inactivate
B19 is a common virus that is spread worldwide, viral concomitants in blood products. The risk of
and the seroprevalence increases with age, so that transmission through blood transfusions and plas-
15% of preschool children, 50% of younger adults ma-derived products has been known for many
and about 85% of the elderly show serologic years [59–63] and many manufacturers of plasma
evidence of past infection [31–35]. In developing derivatives screen their products by quantitative
countries the seroprevalence has been shown to be a PCR [64]. Quantitative measurements of B19 DNA
little higher, probably because of poor and crowded should also be considered in blood transfusions
living standards, whereas in isolated tribal com- intended for immunosuppressed individuals and
munities seroprevalence figures are below 10% other risk groups, which has been reviewed by
[36–38]. Infection appears to confer lifelong immu- Corcoran and Doyle [65]. Indeed, recommendations
nity to immunocompetent hosts. Although the have been made in the Netherlands that blood
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
REVIEW: PARVOVIRUS B19 INFECTION 287

donors with high anti-B19 immunoglobulin G (IgG) children with low-grade fever, malaise and a char-
titres should be used for donations to high-risk acteristic facial rash that has given rise to the name
recipients [66]. ‘Slapped cheek syndrome’. Arthralgia may occur in
We thus still need to learn more about risk some children with EI but is not as common as in
judgement in recipients of blood products. The adults. The slapped-cheek appearance is followed by
susceptibility depends not only on host factors but the spread of a maculopapular rash on the trunk,
also on the relation of viral load and anti-B19 IgG back and extremities (Fig. 2). The infection is
titres in the blood product [67, 68]. Life-threatening normally self-limiting within a week or two but
B19 infection has even been transmitted by intra- the rash can be recurrent for some months following
venous immunoglobulin (IVIG) [69]. exposure to sunlight, heat, emotion, or exercise [75].
The rash and the joint symptoms are most likely
caused by immune complex deposition.
Clinical manifestations
After the discovery in 1974, it was not until 1981 a
Arthropathy
distinct disease was associated with infection of B19
when it was detected in a sickle-cell anaemic patient On average, 50% of adult cases of EI have associated
with transient aplastic crisis (TAC) [70]. It was joint manifestations that may persist for weeks to
subsequently shown to be the cause of EI in 1983, a months, and in a few cases for years. The typical facial
disease first described in 1799 [71]. exanthema and fever of EI is only seen in a minority of
The features of B19 infection have been studied in
experimental infections of healthy individuals [72,
73]. After intranasal inoculation of B19, there was a
biphasic clinical course. The viraemia peaked after
8–9 days at 1011 virions per mL, with viral excre-
tion through the respiratory tract, but not via urine
or faeces, accompanied with mild symptoms of fever,
malaise, myalgia and pruritus. About three weeks
after infection, a second phase of symptoms devel-
oped, with typical maculopapular rash for the B19
infection and in some cases arthralgia. The BM was
normal after 6 days, but on day 10 there was an
almost complete loss of erythroid precursors, and a
subsequent marked drop in the reticulocyte counts
was noted in peripheral blood. The effects on the
haemoglobin level and other blood cell counts were
less pronounced.
Since the time of these experiments, the wide
clinical spectrum of B19-associated complications
has been dissected and the different outcomes
depend heavily on the immune status of the host.

Erythema infectiosum
Fig. 2 The pathogenesis of erythema infectiosum is probably a
Erythema infectiosum is the most common clinical result of antibody-antigen immune complex depositions in skin,
manifestation of B19 infection in immunocompetent blood vessels and synovia. The rash typically appears on the
hosts although asymptomatic infection is seen in cheeks followed by a lace-like maculopapular rash on the upper
25–50% of infected individuals [55, 74]. Another part of the body. Joint symptoms are more common in adults than
in children. In addition to deposition of immune complexes, the
name for EI is the ‘Fifth disease’, referring to a fifth inflammatory response in synovial tissue may be a result of the
place in a listing of the common infections dur- secreted phospholipase A2 motif in the unique region of the B19
ing childhood. Classically, EI affects school-aged minor capsid protein [89].

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


288 K . B R O L I D E N et al.

adult cases. The arthropathy is particularly common disorders is not clear and in some cases the infection
in middle-aged women, and is characterized by a may be a pure coincidence and in other cases it can
polyarthritis typically involving the metacarpopha- be a triggering or even a rare aetiological factor. In
langeal joints, knees, wrists, or ankles [76–79]. Both any case, the infection is an important differential
arthralgia and inflammatory arthritis may occur and diagnosis for several autoimmune disorders both in
the arthropathy sometimes even mimics classical clinical presentation and in immunological tests. In
rheumatoid arthritis (RA). The joint involvement is conclusion, in individuals with immunogenetic pre-
however not erosive and is probably immune-medi- disposing factors B19 may indeed be capable of
ated as it appears simultaneously with circulating causing at least arthropathy associated with severe,
antibodies. B19 DNA has indeed been detected in long-lasting morbidity.
synovial fluid and biopsies in both acute and remotely Many viral infections including B19 induce the
infected individuals [80–83]. Although B19 infection production of autoantibodies. Although these re-
may mimic RA, and even trigger a positive test for sponses are normally of short duration they can
rheumatoid factor, its role in the aetiology of this create diagnostic difficulties. As reviewed by Meyer
disease has not been proved [84, 85]. [98], B19 can, for example, induce antibodies to
Kerr et al. [86] have described an association double-stranded DNA, anti-nuclear soluble antigens,
between development of symptoms during B19 cardiolipin and rheumatoid factor. The autoanti-
infection and carriage of the HLA-DRB1*01, *04, body production most likely results from both
and *07 alleles. B19-associated arthritis may thus at polyclonal stimulation of immune responses and
least partly be genetically associated and has, in production of polyspecific anti-B19 antibodies [92].
earlier studies, been shown to be more common in
individuals with HLA DR4 or B27 [87, 88]. The
Patients with increased red cell turnover
mechanism of the arthropathy is partly unknown.
In addition to immune-mediated inflammation, one In patients with either decreased production or
hypothesis include the activation of synoviocytes by increased loss of erythrocytes, there is a potential
the secreted phospholipase A2 motif in the B19 VP1 risk that B19 infection leads to TAC. When the red
unique region. This activity would thus accelerate cell turnover is increased, the suppression of the BM
the inflammatory response in synovial tissue [89]. caused by B19 leads to a severe drop in the
The B19 NS1 protein causes the secretion of haemoglobin level that can be fatal. This was first
proinflammatory cytokines, which could contribute described in patients with haemolytic anaemia, but
to the arthritis and inflammatory and autoimmune can occur in patients with a wide range of disorders,
disorders associated with the infection [90, 91]. including thalassemia, hereditary spherocytosis,
Furthermore, peptides derived from VP2 may be sickle-cell anaemia, malaria and even iron deficiency
pathogenic in B19 arthropathy as they can induce and haemorrhage [70, 99–104]. In addition to a
cross-reactive autoantibodies against human kera- cessation of the erythroid production, other blood
tin, collagen, and cardiolipin [92]. cell lineages can be affected with clinically signifi-
cant thrombocytopenia, neutropenia or pancytope-
nia as the result (Fig. 3) [105–107].
Autoimmune disorders
Once the immune response clears the infection,
Apart from RA, B19 infection has been associated the red cell production resumes and eventually
with the onset of numerous autoimmune disorders normalizes followed by lifelong immunity in most
including systemic lupus erythematosus (SLE) [93, cases. However, the aplastic crisis can cause severe
94], other connective tissue diseases and systemic and even fatal anaemia resulting in congestive heart
vasculitides. Although a few cases of erosive RA and failure, cerebrovascular events, and acute splenic
SLE have been associated with B19 infection, the sequestration [108].
virus is probably an extremely rare cause of these
diseases. Systemic vasculitides including for example
Other B19 associated disorders
Henoch-Schönlein pupura [95], periarteritis nodosa
[96], and giant cell arteritis [97] can occur after B19 has a cardiotropic potential as P-ag is
acute B19 infection. The role of B19 in these expressed by myocytes and B19 DNA has indeed
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
REVIEW: PARVOVIRUS B19 INFECTION 289

Fig. 3 B19 binds to immature erythroblasts thereby arresting production of mature erythropoietic cells. Following acute infection, the
reticulocyte count in peripheral blood is zero and if the patients have an underlying disorder with pathologic red cell survival, the number of
erythocytes may fall dramatically in peripheral blood. The pathogenesis of thrombocytopenia is thought to be explained by the cytotoxicity
of the NS1 protein [238].

been detected in heart tissue [24, 43, 109]. Cases about the aetiology of B19 in rheumatic and other
of sometimes fatal myocarditis and heart failure in disorders.
both children and adults have been reported [110–
115].
B19 infection in immunocompromised individuals
Other B19-associated disorders include hepatitis
[116–118], transient erythroblastopenia of child- The clinical picture of acute and persistent B19
hood [119], neutropenia [120], trombocytopenia infection in the immunocompromised host differs
[121], Kawasaki disease [122], Gloves-and-sock significantly from immunocompetent subjects. In
syndrome [123], neurological disease including the absence of an efficient humoral and/or cellular
meningitis and encephalitis [124, 125], fibromyal- immune response, the infection can cause persistent
gia and chronic fatigue syndrome [126, 127]. BM suppression manifested by chronic anaemia but
Several other manifestations of B19 infection have not supposedly immune-mediated symptoms such as
been reported and have been reviewed in detail by rash and arthralgia [132]. Predisposing conditions
Heegaard and Brown [128], but the evidence for a include congenital immunodeficiencies, leukaemia,
clear causality, as stated above, is sometimes scarce. lymphoma, myelodysplastic syndrome, BM and solid
It must be pointed out that apart from BM which is organ transplantation, chemotherapy, and infection
the primary site of viral replication, B19 DNA is with human immunodeficiency virus [50, 51, 132–
found in tissues like synovia [83, 129], liver [130] 142].
and in skin [131] in both B19-associated clinical About 5% of adult patients and 10% of children
disorders and in healthy controls. This emphasize undergoing chemotherapy for haematological
that caution should be taken in drawing conclusions malignancies are persistently infected with the virus
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
290 K . B R O L I D E N et al.

resulting in severe and even lethal cytopenias which is associated with poor prognosis (K. Broliden,
whereas about 1–2% of organ and stem cell trans- unpublished observation). Early diagnosis of parvo-
planted patients have life-threatening complications virus infection is therefore of extreme importance
caused by parvovirus infection [45, 50]. Cases of and the infection can be treated with either blood
parvovirus-associated pancytopenias or isolated transfusion or intravenous immunoglobulin.
‘penias’ of different haematological cell lineages Many immunodeficiencies affect the production of
can be misinterpreted as relapse of the underlying neutralizing antibodies against the virus resulting in
malignant disorder or of other microbial infections, persistent infections associated with chronic anae-
graft failure or drug toxicity. In Fig. 4, a case of B19 mia [141, 143]. However, conditions or chemother-
infection in a child suffering from acute lymphatic apies leading to deficient cellular immune responses
leukaemia is shown. In a group of children with may also result in persistent infections. The contri-
acute leukaemia, the number of days of unwanted bution of the cellular immune response for elimin-
treatment interruptions was significantly higher in ation of B19 may thus explain why IVIG therapy
parvovirus infected than in uninfected individuals does not always clear infection in some cases and

Fig. 4 The clinical and laboratory outcome of a 10-year-old girl with acute lymphocytic leukaemia and B19 infection, adapted from
Broliden et al. [45]. The infection was not diagnosed until the end of December and thus the B19 diagnostic parameters were performed in
retrospect. During maintenance chemotherapy the patient developed a rash on her cheeks in April. The rash reappeared two times during
the observation time. During the ongoing B19 viraemia she had several periods of severe cytopenias that required blood transfusions. A
relapse of the leukaemia was suspected and a new bone marrow aspirate was performed in December. At this time the B19 diagnosis was
finally suspected and upon additional blood transfusions and three weekly periods of interruptions of the chemotherapy (November–
December) the infection spontaneously resolved. It can be speculated that early diagnosis already at time of the first rash and cytopenic
period in April followed by IVIG treatment could have led to immediate elimination of the B19 infection and avoidance of the critical course
of infection. It should also be noted that serology is unreliable in immunodeficient patients as she did not develop IgG and IgM until the end
of the infection when the immune system recovered following treatment interruption.

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


REVIEW: PARVOVIRUS B19 INFECTION 291

why chronic infection is seen in the presence of immunocompetent subjects together with an altered
neutralizing antibodies without viral clearance. Yet cytokine profile (K. Broliden, unpublished data)
another case demonstrating that neutralizing anti- [149]. However, the role of this aberrant cellular
bodies may be complemented by a cellular immune immune response in relation to clinical symptoms or
response was an AIDS patient with persistent B19 establishment of the persistent infection has not
infection who showed an initial remission of B19 been proved.
infection in the absence of a specific antibody
response [144].
B19 and pregnancy: vertical transmission and foetal
hydrops
B19 persistence in immunocompetent patients
The association between foetal B19 infection and the
In immunocompromised individuals, B19 is known development of non-immune foetal hydrops was first
to cause persistent infection with potentially severe proposed by Brown et al., and it is estimated that
and chronic anaemia as the result. In addition, B19 about 15–20% of cases of non-immune hydrops
seems to be capable of persisting in a subset of fetalis are caused by B19 [150–152]. About 50% of
apparently immunocompetent individuals. B19 is pregnant women are susceptible to B19 infection,
thus reported to be present in about 2% of BM and maternal infection is reported to occur in a few
samples from healthy individuals [31]. The persist- percentage of pregnancies [153, 154]. There is
ence in these individuals is, however, not only approximately a 30% risk of vertical transmission to
seemingly perpetual but also associated with various the foetus, and the over-all risk of an abnormal
long-lasting symptoms such as fatigue, fever, arth- outcome after maternal infection is estimated to 5–
ralgia, and myalgia [145]. The symptom complex is 10% [49, 151, 155–158]. Transmission over the
sometimes similar to the chronic fatigue syndrome, placenta is reportedly most likely to occur in the first
and B19 has been proposed to play a role in this or second trimester, as placental P-ag, which may be
disease [127, 146, 147]. Whereas no specific necessary for transmission, becomes less frequent
sequence variations in the viral genome [3] or with increasing gestational age [23]. The infection
abnormal humoral B19 epitope specificities [148] or causes anaemia, hypoalbuminaemia, inflammation
lack of functional neutralizing antibodies [145] of the liver and possible myocarditis, leading to
could explain the persistence, a selective defect cardiac failure and the development of foetal hy-
in the cellular immune response was seen in drops (Fig. 5) [159]. The condition may resolve but

Fig. 5 Vertical transmission of B19 from a primary infected mother may cause foetal infection. Pathogenic mechanisms include devel-
opment of acute anaemia upon infection of foetal haematopoietic cells. In early pregnancy haematopoiesis is seen in the liver and in later
pregnancy this shifts to the bone marrow. The anaemia may resolve spontaneously or proceed by causing cardiac failure and development
of hydrops fetalis and in rare cases foetal death. The virus may also cause myocarditis and heart arrest by direct infection of myocardial
tissue. Modified from Anderson and Young [239].

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


292 K . B R O L I D E N et al.

can also result in foetal death, with a mean time selecting for hydrops or serologically evident infec-
span between maternal infection and foetal symp- tion [172]. On the other hand, a partly revised
toms of six weeks, but it may be several months in clinical picture was proposed by our studies on B19
rare cases [151, 160–162]. infection in late gestation [170]. The foetuses in
Foetal B19 infection may also be asymptomatic, third trimester IUFD associated with B19 infection
and there are several observations of infants born were rarely hydropic and recent maternal infection
healthy despite evidence of intrauterine infection was not commonly evident by the detection of B19-
diagnosed by the presence of IgM in umbilical cord specific IgM. Maternal IgG may be present, or
blood [163]. Malformations as a consequence of seroconversion may occur months later following
intrauterine B19 infection have been reported in a IUFD [168]. The diagnosis was not readily estab-
few isolated cases [164–166], but has not been lished by histopathological methods but viral DNA
concluded to be a common feature of this infection was detected in foetal tissues and placenta.
[49, 167]. It is interesting to speculate in the mechanisms
underlying these different pictures at various time of
gestation. The proposed pathogenic mechanism in
B19 and pregnancy: intrauterine foetal death (IUFD)
foetal infection is summarized in Fig. 5 as mentioned
There has been a prevailing apprehension that foetal above. In the second trimester, P-ag is present on
demise caused by B19 infection in the third trimester the trophoblast layer in the placenta to allow the
is extremely rare or even nonexistent, but B19 is a vertical transmission of B19 to the foetus from the
common cause of IUFD also in late gestation, but infected mother [23]. The haematopoiesis is at this
without foetal hydrops in a majority of cases [168– time located in the liver and is extremely active to
170]. By investigating cases of IUFD during a 6-year increase the erythrocyte cell mass 34-fold to match
period we found B19 DNA by PCR in 14% the raised demand from the growing foetus. At the
representing both second and third trimester cases. same time, the lifespan of the red blood cells is
Table 1 summarizes the different pictures of B19- decreased to 45–70 days, making the foetus very
associated IUFD that have emerged from our own vulnerable to any pause in the haematopoietic
studies. In the first trimester B19 DNA was found in production [43]. The destruction of late erythroid
3% of cases of spontaneous abortion and an precursors, caused by the B19 infection, leads to
etiological link could not be proved [171]. In the severe anaemia that in combination with the
second trimester, a serologically evident recent hepatic inflammation and possibly myocarditis due
infection could be demonstrated in maternal sera. to B19 infection of cardiac myocytes, results in heart
The foetuses were normally hydropic and B19 DNA failure and the development of hydrops fetalis. In
was detected by PCR in foetal tissues and placenta contrast, in the third trimester, the haematopoiesis
and histopathological investigation often confirmed migrates to the BM, the need for a quickly increasing
the diagnosis [168, 169]. This picture of second red blood cell mass is relieved, and the lifespan of
trimester IUFD cases is often reported in studies the erythrocytes is normalized. The cellular receptor

Table 1 Clinical complications associated with B19 infection during pregnancy

Trimester

First Second Third

Clinical foetal complication Spontaneous abortion Hydrops fetalis and/ Foetal death
or foetal death
Frequency of B19-DNA positivity 3% 12% 7%
in placenta and/or foetal tissue
Maternal symptoms None None or erythema infectiosum None
Maternal B19-IgM reactivity Negative Positive Negative

The table summarizes a generalized picture of more than 150 cases of spontaneous abortions and IUFDs representing different time-points
(trimesters) during pregnancy. The studies were performed by a nested B19 PCR technique for detection of qualitative PCR in placental and/
or foetal tissue [168–171].

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


REVIEW: PARVOVIRUS B19 INFECTION 293

P-ag is virtually nonpresent in the third trimester that both the humoral and the cellular immune
[23]. These factors act in concert to make the systems are mounted to battle B19.
development of anaemia and subsequent heart It was first reported that no cellular immune
failure and hydrops less likely. A low-grade persist- responses were mounted in B19 infection, but
ent foetal and/or maternal infection may also cause CD4+ T cell proliferative responses directed to VP1
IUFD at a later time-point, and in the case of and VP2 have now been shown in seropositive
placental dysfunction even without foetal infection. individuals [185–189]. Analogous to the humoral
Degenerative lesions have been shown in the response, CD4+ T cell responses to NS1 may be
placenta due to the inflammatory response to B19 associated with acute and persistent arthropathy
infection [109, 159, 173, 174]. In the foetus, [190]. In contrast, our studies of the CD8+ T-cell
myocarditis may be the cause of death caused by response have revealed a predominance of NS1-
dysrhythmia or cardiac arrest without the develop- directed responses in acute and remote infection
ment of anaemia or hydrops [159, 175]. Indeed, whereas a skewing of the response towards VP2
investigation of endomyocardial biopsy specimens was seen in persistent infection [184].
revealed B19-associated inflammatory changes in In 2001, Tolfvenstam et al. [184] defined the first
15% of cases of peripartum cardiomyopathy [176]. B19-specific CD8+ T-cell epitope in an individual
with remote infection, providing the first evidence of
an existent cytotoxic cellular response to this infec-
Immune responses in B19 infection
tion. In another report [191], this finding was
B19 is regarded to show a ‘hit-and-run’ mode of explored by following individuals with documented
infection, which in the normal host is believed to primary B19 infection during the acute phase and a
quickly resolve with successful eradication of the thorough phenotypic analysis was later performed
virus. Prolonged detection of B19 DNA peripherally, on the CD8+ T cells [192]. The CD8+ T-cell
in BM and in other compartments is now reported responses were mounted against three to five
by several investigators, indicating that it may take epitopes per individual with no sign of changing
longer to eradicate the virus than previously specificities over time, as for example seen in
thought [80, 177–180]. Viral and host properties infection with the latent cytomegalovirus virus
have thus been explored to explain this failure to (CMV) [193]. The finding of predominantly NS1
eventually clear the virus. specificities in acutely infected patients contrasts
what is known about the humoral response to B19,
where the neutralizing epitopes have been shown to
Immune responses to acute infection
be located mainly in the structural proteins [194–
The humoral immune response is thought to be the 196]. The most unexpected finding was the lack of
most important in infections with short replication rapid contraction of the expanded CD8+ T-cell clones
cycles, characterized by quick lysis of the infected thought to be the hallmark of resolving infections of
cells and release of free virions readily exposed to the ‘hit-and-run’ type [197, 198]. Sustained re-
antibodies that can bind to them and facilitate their sponses were shown that in most cases peaked after
destruction [181]. On the other hand, nonlytic and 1 year and were still detectable after 2 years
persistent viruses such as human hepatitis B virus (Fig. 6). Maintenance of the responses seems un-
(HBV), are less exposed to the humoral immune likely in the absence of antigenic drive, which means
system and the host is therefore more dependent on that B19, although not a classically persistent virus,
the cellular immune response to localize and kill manage to persist in the body for an extended period
infected cells [182, 183]. of time; and B19 may thus be more of a ‘hit-and-
The humoral immune response has been the focus hang’ type of infecting virus. Indeed, we detected
of most studies on B19 and immunity, whereas the peripheral B19 DNA by PCR for over 1 year in some
cellular response and the CD8+ T cell response in of the patients, one of whom was still DNA positive
particular, has been detected and investigated only in the last sample collected after a little more than
recently [184]. Being a virus that shares character- 2 years. It is plausible that B19 persists even longer
istics both with classically lytic resolving viruses and in the BM, although undetectable in peripheral
nonlytic and persistent viruses, it is not surprising blood [145].
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
294 K . B R O L I D E N et al.

Fig. 6 B19-specific CD8+ T-cell responses in relation to B19 viraemia in a healthy immunocompetent individual who acquired acute B19
infection with the onset of symptoms (rash, arthritis and fever) at time-point zero. B19 IgG and B19 IgM were present at onset of symptoms
and the B19 IgM response disappeared after a few months. The B19-specific CD8+ response was measured in an ELISpot assay (gamma-
interferon secretion) by using overlapping B19-specific peptides covering the whole B19 genome, adapted from Norbeck et al. [191]. Thus,
this patient showed sustained levels of cells with three different specificities against the NS-1 protein (as represented by the blue, green and
red line, respectively). No response was seen against the capsid proteins VP1 or VP2. The CD8+ T-cell response was subsequently confirmed
by tetramer staining and the phenotypic markers were followed over time by multicolour staining using flow cytometry [192]. The
corresponding viral titres are also shown (black line) [180]. The Y-axis indicates number of spot-forming cells per 106 PBMC.

the infection with high viral load as a result


Immune responses in persistently infected individuals
[200]. In this situation clones with lower sensi-
Recent data thus suggested that there is a skewing tivity, possibly represented by the VP-specific CD8+
of the CD8+ T-cell responses in persistently infec- T cells in our study, have a better chance to avoid
ted individuals to the structural proteins when exhaustion but may not achieve viral clearance.
compared with acutely infected and healthy indi- In persistent HBV infection, low T-cell numbers is
viduals [149]. This is the first time a discrepancy reported and is possibly the result of immunologic
in the immune response was shown between exhaustion [201]. The situation in persistent B19
healthy and persistently B19-infected individuals, infection may thus resemble that of HBV infection,
and the lack of an efficient NS1 response may and the increased T-cell activity seen after inter-
allow the virus to establish persistence. However, feron treatment of HBV infection, suggests that a
the other way around should be considered, similar approach might prove beneficial also in
namely that the persistence and the continuous B19-persistent infection [202]. Recently, Corcoran
antigenic stimulation results in exhaustion of the et al. [203] showed persistent infection in a child
NS1 response, a phenomenon reported to occur in with acute lymphatic leukaemia despite the treat-
persistent infection [199]. The failed NS1 response ment with intravenous immunoglobulin. Interest-
may in turn favour the expansion of VP-specific ingly, resolution of the infection was associated
CD8+ T-cell clones. It has been reported that with the simultaneous strengthening of antigen-
clones of CD8+ T cells with high antigenic specific B-cell memory against B19 VP2 and
sensitivity are activated first, but succumb due to diminution in the memory response against B19
exhaustion if they for some reason fail to control NS1.
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
REVIEW: PARVOVIRUS B19 INFECTION 295

serological assays are sometimes needed for an accu-


Diagnosis
rate diagnosis and timing of maternal infection dur-
Modern diagnostics of B19 infection usually include ing pregnancy, which has recently been reviewed by
measurement of B19 IgG and IgM antibodies in de Jong et al. [213] and Enders et al. [214].
blood and B19 DNA in blood or tissue samples by
PCR. Morphologically, BM aspirates show no mature
Polymerase chain reaction
erythroid precursors and with characteristic giant
pronormoblasts at time of acute infection. Immu- Serology must in many cases be complemented by
nohistochemistry may also be very specific and a PCR analysis of B19 DNA. In immunocompromised
complement to the more sensitive PCR assay in patients a positive PCR test in blood indicates
cases of placental or foetal infection. ongoing acute or persistent infection whereas a
positive PCR test in BM may indicate either acute or
remote infection as about 2% of healthy individuals
Serology
with remote infection are PCR positive [31]. In cases
Specific IgM antibodies directed to VP2 are present of intrauterine foetal complications it can be very
after 10–12 days after infection and usually disap- useful to analyse amniotic fluid or cord blood for
pear within 3–4 months, even though they may viral DNA or RNA by PCR for differential diagnostic
occasionally remain detectable longer [204]. The purposes including B19 [215–218]. To learn more
maturation and shift to IgG occurs shortly after the about the clinical significance of viral load in foetal
advent of IgM, and these high-avidity antibodies infection is of high priority as therapeutic interven-
are believed to mediate lifelong immunity, with tion can be necessary in severe cases.
slowly decreasing titres boosted by subsequent Detection of B19 DNA can thus be detected by
encounters with B19. IgA antibodies are also PCR in serum, BM and other tissues for diagnostic
detectable for a short period and may play a purposes. Whilst B19 DNA may be positive also in
protective role in the respiratory tract [205]. In tissues of healthy individuals as previously men-
addition, long-term B19-specific IgE antibodies tioned, the development of a quantitative PCR has
have been shown, but with unclear biological been helpful in determining the viral load and
function [206]. The IgG is directed to both VP1 should be used routinely. This also allow differenti-
and VP2 with the majority of linear epitopes ation of human parvovirus variants [219, 220]. In
located in the VP1ur and the junction between healthy individuals with acute B19 infection, viral
VP1 and VP2, and these specificities are reported to titres as high as 1012 geq mL)1 are detectable in
represent the most effective immune response blood [61]. However, a marked drop is seen at time
[194]. However, others have shown that IgG of immunoglobulin production and onset of symp-
directed to VP2 is maintained even when IgG toms and titres remain at about 103 to 105 for
directed to linear epitopes within VP1 is lost, months and even a few years following acute
indicating immunodominance for VP2 [187, 195, infection [180, 221]. Future studies will hopefully
196]. Either way, the humoral responses seem to determine the concentrations of viral load in differ-
be directed to the structural proteins in normal ent tissues and clinical settings to guide the
infection, with persistence of IgG antibodies direc- clinicians in therapeutic choices.
ted to conformational epitopes. Antibodies directed
to NS1 have been described to be a feature only of
Treatment and prophylaxis
acute and persistent infection [207–209], but this
has not been confirmed in other studies [210–212]. There is no specific antiviral drug against B19
Caution should be made when interpreting serol- infection but a number of alternative options to
ogy in immunodeficient individuals and in pregnant eliminate the virus can be recommended. The choice
women. Due to their immune status they are not of treatment of B19 infection must however take
always able to mount an antibody response to host factors into account as the virus yields different
pathogens at all. Furthermore, blood products pathogenesis in different risk groups of patients
or treatment with immunoglobulins may yield depending on underlying diseases and immunodefi-
false-positive B19 IgG responses. Supplementary ciency status (Table 2).
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
296 K . B R O L I D E N et al.

Table 2 Parvovirus B19-infection pathogenesis in different risk groups

Proposed Clinical and


Risk group pathogenesis laboratory signs Treatment

Immunocompetent Immune complex Erythema infectiosum, Antiinflammatory


individuals deposition arthropathy drugs
Patients with Reduced erythropoeisis Transient aplastic crisis Blood transfusion
increased
turnover of
red blood cells
Immunodeficient Deficient humoral and/or Chronic anemia IVIG
patients cellular immune response or cytopenia
Fetuses Deficient humoral and/or Hydrops fetalis, IUFD, anemia, Blood transfusion,
cellular immune response cardiac failure IVIG
Reduced erythropoiesis

The clinical and laboratory signs of B19 infection and corresponding therapeutic strategies is dependent on host factors and underlying
clinical disorders. For example, the pathogenesis in the B19-infected foetus is a result of the physiologically higher red blood cell turnover
and the relative immunodeficiency of the foetus. It must be pointed out that the infection resolves spontaneously in many cases in all risk
groups and treatment is therefore only given to severe cases.

antibodies does not respond well to IVIG treatment,


Immunocompetent hosts
but it may induce a transient remission [145]. In
There is no specific antiviral drug against B19 and patients subject to chemotherapy or otherwise
the infection does not normally need treatment in iatrogenically immunosuppressed, a persistent B19
the immunocompetent host. Nonsteroidal anti- infection usually resolves upon cessation of the
inflammatory drugs are sometimes useful for pro- therapy [224, 225].
nounced symptoms such as arthralgia.
Pregnancy
Transient aplastic crisis
If B19 infection is confirmed in the pregnant
Transient aplastic crisis usually requires hospitaliza- woman, monitoring of the foetus by weekly ultra-
tion and erythrocyte transfusions, but has good sound examinations are usually recommended. It
prognosis if treated correctly with restored haema- must be emphasized though that foetal death may
topoiesis within 1–10 days [103]. In a study inclu- occur several months postmaternal infection and
ding 62 patients with sickle-cell anaemia and B19- without foetal hydrops, or even in the absence of
induced TAC, 87% required transfusion therapy and laboratory or clinical signs of maternal infection
63% were hospitalized; one patient died before the [160, 168, 226]. If hydrops and/or anaemia is
initiation of transfusion therapy [222]. diagnosed by Doppler ultrasound and cordocentesis,
intrauterine erythrocyte transfusions have been
shown to reduce the mortality rate from about
Immunosuppressed patients
50–18% [227–231]. Maternally administrated IVIG
In immunosuppressed patients lacking neutralizing or intrauterine therapy using B19 IgG-rich high titre
antibodies, IVIG has proved to be useful for treatment gamma-globulin has also been tried but needs
of persistent B19 infection. Administration of 0.4 g further evaluation [232, 233].
kg)1 · 5 days or 1 g kg)1 · 3 days induces an in-
crease in reticulocytecount with an accompanied raise
Vaccine development
in the haemoglobulin level, and is often curative in that
B19 is cleared from the body [58, 137, 142, 223]. A candidate vaccine has been tested consisting of
25% VP1 and 75% VP2, that with adjuvant elicits a
strong humoral response, and is well tolerated [234,
Persistent infection in immunocompetent individuals
235]. The vaccine is optimized with a humoral
Persistent B19 infection in apparently immunocom- response in mind, which clearly has a well-docu-
petent individuals who already possess neutralizing mented role in B19 infection. However, in some
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
REVIEW: PARVOVIRUS B19 INFECTION 297

situations the infection is not controlled despite the diagnostic and prognostic tool, as well as be of
presence of neutralizing antibodies, and evidence of guidance for therapeutic intervention. However, we
the importance of the cellular immune responses is still need to learn more about how to interpret
growing. Therefore, cellular immune responses quantitative PCR results in blood and tissue samples
should be thought of when discussing responsiveness in relation to the immune status of the host.
to a future B19 vaccine. Our findings of a predom- B19 elicits a strong multi-specific CD8+ T-cell
inant NS1-specific CD8+ T-cell response in normal response in the acute phase of the infection that is
infection and the aberrant CD8+ T-cell response in sustained for up to 2 years. The specificity of the
persistent infection may warrant the inclusion of response does not change during the course of
NS1 epitopes using T-cell vaccine techniques. Unfor- infection, and the response is probably antigen-
tunately, commercial interest rather than lack of driven as B19 DNA is also durably detectable by PCR
efficacy and safety has limited the development of a in peripheral blood indicative of continued replica-
B19 vaccine [236]. The vaccine would primarily be tion. This finding, unexpected in a virus typically
intended for certain risk groups of severe B19 associated with ‘hit-and-run’ infection, reveals a
infection. For example it could be used to prevent novel pattern of immune reactivity and mode of
TAC in patients with sickle-cell disease and pure red infection for which the paraphrase ‘hit-and-hang’
cell anaemia (PRCA) in immunodeficient patients. may be more suitable. In contrast to the humoral
Whether seronegative women of childbearing age and CD4+ T-cell responses, the majority of the CD8+
should be included is a matter of controversy. The T cell responses are directed to epitopes within NS1 in
risk of fatal outcome following maternal primary normal infection. Based on the recent definition of
infection may be too low to warrant a generalized novel CD8+ T-cell epitopes, multimeric major histo-
vaccination programme. compatibility complexes can be used for the detailed
study of B19-specific CD8+ T cells with regard to
frequency, phenotype and function. In this way, the
Conclusions and future perspectives
maturation of the CD8+ T-cell response in different
B19 infection causes a wide range of symptoms with clinical manifestations of B19 infection can be
the most severe clinical outcome in foetuses and assessed. Such investigations could provide further
immunosuppressed individuals. The infection can be characterization of the concept of persistent versus
both acute as well as establish persistency. Although normal viral clearance patterns, and hopefully pro-
BM is the major target organ for virus replication, the vide knowledge about the underlying mechanisms for
persistent infections seen in some individuals for the unsuccessful clearance despite the presence of
years and even decades indicate that the virus may neutralizing antibodies and specific CD8+ T cells.
reside in additional immunoprotected compart- Furthermore, the frequency, phenotype and function
ments. The recently launched quantitative PCR of specific CD8+ T cells, peripherally and locally in the
techniques for measurement of B19 DNA titres have placenta, will possibly reveal some new clues as to
significantly improved the diagnostic repertoire as what determines the outcome in foetal infection.
serology and qualitative PCR is insufficient in some In summary, measurement of quantitative viral
cases. For example, increase in viral load could be titres and increased knowledge about the immune
monitored prior to relapse of anaemia in an AIDS response to the virus are new developments to study
patient with persistent B19 infection [237]. Likewise, B19 pathogenesis with the aim of improving the
the possibility of monitoring transplanted patients for medical care of infected patients.
B19 viral load with the aim of initiating preemptive
therapy may be a future strategy to combat viral
Conflict of interest statement
complications in this group of specific immunosup-
pressed patients. This strategy has been successful in The authors have no conflict of interest.
the case of CMV infections in BM-transplanted
patients. Furthermore, by performing quantitative
Acknowledgements
PCR in amniotic fluid, maternal sera and cord blood
we could correlate viral load to clinical outcome, in We hereby acknowledge the Swedish Children
which case the quantitative PCR could serve as a Cancer Foundation, the Swedish Cancer Society,
 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304
298 K . B R O L I D E N et al.

the Swedish Medical Research Council and the parvovirus in vitro increases with differentiation. Blood
Tobias Foundation. 1990; 75: 603–10.
17 Mortimer PP, Humphries RK, Moore JG, Purcell RH, Young
NS. A human parvovirus-like virus inhibits haematopoietic
colony formation in vitro. Nature 1983; 302: 426–9.
References 18 Ozawa K, Kurtzman G, Young N. Productive infection by
1 Cossart YE, Field AM, Cant B, Widdows D. Parvovirus-like B19 parvovirus of human erythroid bone marrow cells in
particles in human sera. Lancet 1975; 1: 72–3. vitro. Blood 1987; 70: 384–91.
2 Gallinella G, Venturoli S, Gentilomi G, Musiani M, Zerbini M. 19 Solheim BG, Rollag H, Svennevig JL, Arafa O, Fosse E,
Extent of sequence variability in a genomic region coding for Bergerud U. Viral safety of solvent/detergent-treated plasma.
capsid proteins of B19 parvovirus. Arch Virol 1995; 140: Transfusion 2000; 40: 84–90.
1119–25. 20 Schwarz TF, Serke S, Von Brunn A et al. Heat stability of
3 Erdman DD, Durigon EL, Wang QY, Anderson LJ. Genetic parvovirus B19: kinetics of inactivation. Zentralbl Bakteriol
diversity of human parvovirus B19: sequence analysis of the 1992; 277: 219–23.
VP1/VP2 gene from multiple isolates. J Gen Virol 1996; 77: 21 Brown KE, Anderson SM, Young NS. Erythrocyte P antigen:
2767–74. cellular receptor for B19 parvovirus. Science 1993; 262:
4 Haseyama K, Kudoh T, Yoto Y, Suzuki N, Chiba S. Analysis 114–7.
of genetic diversity in the VP1 unique region gene of human 22 Brown KE, Hibbs JR, Gallinella G et al. Resistance to par-
parvovirus B19 using the mismatch detection method and vovirus B19 infection due to lack of virus receptor (eryth-
direct nucleotide sequencing. J Med Virol 1998; 56: 205–9. rocyte P antigen). N Engl J Med 1994; 330: 1192–6.
5 Umene K, Nunoue T. Genetic diversity of human parvovirus 23 Jordan JA, DeLoia JA. Globoside expression within the hu-
B19 determined using a set of restriction endonucleases man placenta. Placenta 1999; 20: 103–8.
recognizing four or five base pairs and partial nucleotide 24 Cooling LL, Koerner TA, Naides SJ. Multiple glycosphingol-
sequencing: use of sequence variability in virus classifica- ipids determine the tissue tropism of parvovirus B19. J Infect
tion. J Gen Virol 1991; 72: 1997–2001. Dis 1995; 172: 1198–205.
6 Umene K, Nunoue T. The genome type of human parvovirus 25 von dem Borne AE, Bos MJ, Joustra-Maas N et al. A murine
B19 strains isolated in Japan during 1981 differs from types monoclonal IgM antibody specific for blood group P antigen
detected in 1986 to 1987: a correlation between genome (globoside). Br J Haematol 1986; 63: 35–46.
type and prevalence. J Gen Virol 1990; 71: 983–6. 26 Rouger P, Gane P, Salmon C. Tissue distribution of H, Lewis
7 Hemauer A, von Poblotzki A, Gigler A et al. Sequence and P antigens as shown by a panel of 18 monoclonal anti-
variability among different parvovirus B19 isolates. J Gen bodies. Rev Fr Transfus Immunohematol 1987; 30: 699–708.
Virol 1996; 77: 1781–5. 27 Srivastava A, Bruno E, Briddell R et al. Parvovirus B19-
8 Gallinella G, Venturoli S, Manaresi E, Musiani M, Zerbini M. induced perturbation of human megakaryocytopoiesis in
B19 virus genome diversity: epidemiological and clinical vitro. Blood 1990; 76: 1997–2004.
correlations. J Clin Virol 2003; 28: 1–13. 28 Weigel-Kelley KA, Yoder MC, Srivastava A. Recombinant
9 Nguyen QT, Wong S, Heegaard ED, Brown KE. Identification human parvovirus B19 vectors: erythrocyte P antigen is
and characterization of a second novel human erythrovirus necessary but not sufficient for successful transduction of
variant, A6. Virology 2002; 301: 374–80. human hematopoietic cells. J Virol 2001; 75: 4110–6.
10 Nguyen QT, Sifer C, Schneider V et al. Novel human eryth- 29 Weigel-Kelley KA, Yoder MC, Srivastava A. Alpha5beta1
rovirus associated with transient aplastic anemia. J Clin integrin as a cellular coreceptor for human parvovirus B19:
Microbiol 1999; 37: 2483–7. requirement of functional activation of beta1 integrin for
11 Nguyen QT, Sifer C, Schneider V, Bernaudin F, Auguste V, viral entry. Blood 2003; 102: 3927–33.
Garbarg-Chenon A. Detection of an erythrovirus sequence 30 Munakata Y, Saito-Ito T, Kumura-Ishii K et al. Ku80 auto-
distinct from B19 in a child with acute anaemia. Lancet antigen as a cellular coreceptor for human parvovirus B19
1998; 352: 1524. infection. Blood 2005; 106: 3449–56.
12 Servant A, Laperche S, Lallemand F et al. Genetic diversity 31 Heegaard ED, Petersen BL, Heilmann CJ, Hornsleth A. Pre-
within human erythroviruses: identification of three geno- valence of parvovirus B19 and parvovirus V9 DNA and
types. J Virol 2002; 76: 9124–34. antibodies in paired bone marrow and serum samples from
13 Hokynar K, Soderlund-Venermo M, Pesonen M et al. A new healthy individuals. J Clin Microbiol 2002; 40: 933–6.
parvovirus genotype persistent in human skin. Virology 32 Anderson LJ, Tsou C, Parker RA et al. Detection of antibodies
2002; 302: 224–8. and antigens of human parvovirus B19 by enzyme-linked
14 Allander T, Tammi MT, Eriksson M, Bjerkner A, Tiveljung- immunosorbent assay. J Clin Microbiol 1986; 24: 522–6.
Lindell A, Andersson B. Cloning of a human parvovirus by 33 Kelly HA, Siebert D, Hammond R, Leydon J, Kiely P, Maskill
molecular screening of respiratory tract samples. Proc Natl W. The age-specific prevalence of human parvovirus
Acad Sci U S A 2005; 102: 12891–6. immunity in Victoria, Australia compared with other parts
15 Sanabani S, Neto WK, Pereira J, Sabino EC. Sequence of the world. Epidemiol Infect 2000; 124: 449–57.
variability of human erythroviruses present in bone 34 Cohen BJ, Buckley MM. The prevalence of antibody to hu-
marrow of Brazilian patients with various parvovirus B19- man parvovirus B19 in England and Wales. J Med Microbiol
related hematological symptoms. J Clin Microbiol 2006; 44: 1988; 25: 151–3.
604–6. 35 Tsujimura M, Matsushita K, Shiraki H, Sato H, Okochi K,
16 Takahashi T, Ozawa K, Takahashi K, Asano S, Takaku F. Maeda Y. Human parvovirus B19 infection in blood donors.
Susceptibility of human erythropoietic cells to B19 Vox Sang 1995; 69: 206–12.

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


REVIEW: PARVOVIRUS B19 INFECTION 299

36 Tolfvenstam T, Enbom M, Ghebrekidan H et al. Seropreva- 55 Woolf AD, Campion GV, Chishick A et al. Clinical manifes-
lence of viral childhood infections in Eritrea. J Clin Virol tations of human parvovirus B19 in adults. Arch Intern Med
2000; 16: 49–54. 1989; 149: 1153–6.
37 Schwarz TF, Gurtler LG, Zoulek G, Deinhardt F, Roggendorf 56 Miyamoto K, Ogami M, Takahashi Y et al. Outbreak of hu-
M. Seroprevalence of human parvovirus B19 infection in man parvovirus B19 in hospital workers. J Hosp Infect 2000;
Sao Tome and Principe, Malawi and Mascarene Islands. 45: 238–41.
Zentralbl Bakteriol 1989; 271: 231–6. 57 Evans JP, Rossiter MA, Kumaran TO, Marsh GW, Mortimer
38 Nascimento JP, Buckley MM, Brown KE, Cohen BJ. The PP. Human parvovirus aplasia: case due to cross infection in
prevalence of antibody to human parvovirus B19 in Rio de a ward. Br Med J (Clin Res Ed) 1984; 288: 681.
Janeiro, Brazil. Rev Inst Med Trop Sao Paulo 1990; 32: 41–5. 58 Frickhofen N, Chen ZJ, Young NS, Cohen BJ, Heimpel H,
39 Thomas I, Di Giambattista M, Gerard C et al. Prevalence of Abkowitz JL. Parvovirus B19 as a cause of acquired chronic
human erythrovirus B19 DNA in healthy Belgian blood pure red cell aplasia. Br J Haematol 1994; 87: 818–24.
donors and correlation with specific antibodies against 59 Eis-Hubinger AM, Sasowski U, Brackmann HH, Kaiser R,
structural and non-structural viral proteins. Vox Sang 2003; Matz B, Schneweis KE. Parvovirus B19 DNA is frequently
84: 300–7. present in recombinant coagulation factor VIII products.
40 Yoto Y, Kudoh T, Haseyama K et al. Incidence of human Thromb Haemost 1996; 76: 1120.
parvovirus B19 DNA detection in blood donors. Br J Hae- 60 Williams MD, Cohen BJ, Beddall AC, Pasi KJ, Mortimer PP,
matol 1995; 91: 1017–8. Hill FG. Transmission of human parvovirus B19 by coagu-
41 Wakamatsu C, Takakura F, Kojima E et al. Screening of lation factor concentrates. Vox Sang 1990; 58: 177–81.
blood donors for human parvovirus B19 and characteriza- 61 Prowse C, Ludlam CA, Yap PL. Human parvovirus B19 and
tion of the results. Vox Sang 1999; 76: 14–21. blood products. Vox Sang 1997; 72: 1–10.
42 Jordan J, Tiangco B, Kiss J, Koch W. Human parvovirus B19: 62 Santagostino E, Mannucci PM, Gringeri A et al. Transmis-
prevalence of viral DNA in volunteer blood donors and sion of parvovirus B19 by coagulation factor concentrates
clinical outcomes of transfusion recipients. Vox Sang 1998; exposed to 100 degrees C heat after lyophilization. Transfu-
75: 97–102. sion 1997; 37: 517–22.
43 Berry PJ, Gray ES, Porter HJ, Burton PA. Parvovirus infec- 63 Plentz A, Hahn J, Knoll A, Holler E, Jilg W, Modrow S.
tion of the human fetus and newborn. Semin Diagn Pathol Exposure of hematologic patients to parvovirus B19 as a
1992; 9: 4–12. contaminant of blood cell preparations and blood products.
44 Lundqvist A, Tolfvenstam T, Brytting M, Stolt CM, Hedman Transfusion 2005; 45: 1811–5.
K, Broliden K. Prevalence of parvovirus B19 DNA in bone 64 Aberham C, Pendl C, Gross P, Zerlauth G, Gessner M. A
marrow of patients with haematological disorders. Scand J quantitative, internally controlled real-time PCR assay for
Infect Dis 1999; 31: 119–22. the detection of parvovirus B19 DNA. J Virol Methods 2001;
45 Broliden K, Tolfvenstam T, Ohlsson S, Henter JI. Persistent 92: 183–91.
B19 parvovirus infection in pediatric malignancies. Med 65 Corcoran A, Doyle S. Advances in the biology, diagnosis and
Pediatr Oncol 1998; 31: 66–72. host-pathogen interactions of parvovirus B19. J Med
46 Anderson MJ, Cohen BJ. Human parvovirus B19 infections Microbiol 2004; 53: 459–75.
in United Kingdom 1984–86. Lancet 1987; 1: 738–9. 66 Groeneveld K, van der Noordaa J. Blood products and par-
47 Heegaard ED, Laub Petersen B. Parvovirus B19 transmitted vovirus B19. Neth J Med 2003; 61: 154–6.
by bone marrow. Br J Haematol 2000; 111: 659–61. 67 Blumel J, Schmidt I, Willkommen H, Lower J. Inactivation of
48 Koenigbauer UF, Eastlund T, Day JW. Clinical illness due parvovirus B19 during pasteurization of human serum
to parvovirus B19 infection after infusion of solvent/ albumin. Transfusion 2002; 42: 1011–8.
detergent-treated pooled plasma. Transfusion 2000; 40: 68 Daly P, Corcoran A, Mahon BP, Doyle S. High-sensitivity
1203–6. PCR detection of parvovirus B19 in plasma. J Clin Microbiol
49 PHLS. Prospective study of human parvovirus (B19) infec- 2002; 40: 1958–62.
tion in pregnancy. Public Health Laboratory Service Work- 69 Hayakawa F, Imada K, Towatari M, Saito H. Life-threatening
ing Party on Fifth Disease. BMJ 1990; 300: 1166–70. human parvovirus B19 infection transmitted by intravenous
50 Broliden K. Parvovirus B19 infection in pediatric solid-organ immune globulin. Br J Haematol 2002; 118: 1187–9.
and bone marrow transplantation. Pediatr Transplant 2001; 70 Pattison JR, Jones SE, Hodgson J et al. Parvovirus infections
5: 320–30. and hypoplastic crisis in sickle-cell anaemia. Lancet 1981; 1:
51 Egbuna O, Zand MS, Arbini A, Menegus M, Taylor J. A 664–5.
cluster of parvovirus B19 infections in renal transplant 71 Anderson MJ, Jones SE, Fisher-Hoch SP et al. Human par-
recipients: a prospective case series and review of the lit- vovirus, the cause of erythema infectiosum (fifth disease)?
erature. Am J Transplant 2006; 6: 225–31. Lancet 1983; 1: 1378.
52 Anderson LJ. Role of parvovirus B19 in human disease. 72 Potter CG, Potter AC, Hatton CS et al. Variation of erythroid
Pediatr Infect Dis J 1987; 6: 711–8. and myeloid precursors in the marrow and peripheral blood
53 Anderson LJ, Gillespie SM, Torok TJ, Hurwitz ES, Tsou CJ, of volunteer subjects infected with human parvovirus (B19).
Gary GW. Risk of infection following exposures to human J Clin Invest 1987; 79: 1486–92.
parvovirus B19. Behring Inst Mitt 1990; 85: 60–3. 73 Anderson MJ, Higgins PG, Davis LR et al. Experimental par-
54 Gillespie SM, Cartter ML, Asch S et al. Occupational risk of voviral infection in humans. J Infect Dis 1985; 152: 257–65.
human parvovirus B19 infection for school and day-care 74 Cartter ML, Farley TA, Rosengren S et al. Occupational risk
personnel during an outbreak of erythema infectiosum. factors for infection with parvovirus B19 among pregnant
JAMA 1990; 263: 2061–5. women. J Infect Dis 1991; 163: 282–5.

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


300 K . B R O L I D E N et al.

75 Musiani M, Manaresi E, Gallinella G, Cricca M, Zerbini M. 94 Magro CM, Dawood MR, Crowson AN. The cutaneous
Recurrent erythema in patients with long-term parvovirus manifestations of human parvovirus B19 infection. Hum
B19 infection. Clin Infect Dis 2005; 40: e117–9. Pathol 2000; 31: 488–97.
76 Woolf AD. Human parvovirus B19 and arthritis. Behring Inst 95 Ferguson PJ, Saulsbury FT, Dowell SF, Torok TJ, Erdman DD,
Mitt 1990; 85: 64–8. Anderson LJ. Prevalence of human parvovirus B19 infection
77 Reid DM, Reid TM, Brown T, Rennie JA, Eastmond CJ. Hu- in children with Henoch-Schonlein purpura. Arthritis Rheum
man parvovirus-associated arthritis: a clinical and laborat- 1996; 39: 880–1.
ory description. Lancet 1985; 1: 422–5. 96 Corman LC, Dolson DJ. Polyarteritis nodosa and parvovirus
78 White DG, Woolf AD, Mortimer PP, Cohen BJ, Blake DR, B19 infection. Lancet 1992; 339: 491.
Bacon PA. Human parvovirus arthropathy. Lancet 1985; 1: 97 Gabriel SE, Espy M, Erdman DD, Bjornsson J, Smith TF,
419–21. Hunder GG. The role of parvovirus B19 in the pathogenesis
79 Naides SJ. Rheumatic manifestations of parvovirus B19 of giant cell arteritis: a preliminary evaluation. Arthritis
infection. Rheum Dis Clin North Am 1998; 24: 375–401. Rheum 1999; 42: 1255–8.
80 Saal JG, Steidle M, Einsele H, Muller CA, Fritz P, Zacher J. 98 Meyer O. Parvovirus B19 and autoimmune diseases. Joint
Persistence of B19 parvovirus in synovial membranes of pa- Bone Spine 2003; 70: 6–11.
tients with rheumatoid arthritis. Rheumatol Int 1992; 12: 99 Graeve JA, Elliott SC. Neurologic symptoms, iron deficiency
147–51. anemia, and parvovirus infection. J Pediatr 1991; 118: 830.
81 Zakrzewska K, Azzi A, De Biasi E et al. Persistence of par- 100 Lortholary O, Eliaszewicz M, Dupont B, Courouce AM. Par-
vovirus B19 DNA in synovium of patients with haemophilic vovirus B19 infection during acute Plasmodium falciparum
arthritis. J Med Virol 2001; 65: 402–7. malaria. Eur J Haematol 1992; 49: 219.
82 Dijkmans BA, van Elsacker-Niele AM, Salimans MM, van 101 Serjeant GR, Topley JM, Mason K et al. Outbreak of aplastic
Albada-Kuipers GA, de Vries E, Weiland HT. Human par- crises in sickle cell anaemia associated with parvovirus-like
vovirus B19 DNA in synovial fluid. Arthritis Rheum 1988; agent. Lancet 1981; 2: 595–7.
31: 279–81. 102 Lefrere JJ, Courouce AM, Bertrand Y, Girot R, Soulier JP.
83 Soderlund M, von Essen R, Haapasaari J, Kiistala U, Ki- Human parvovirus and aplastic crisis in chronic hemolytic
viluoto O, Hedman K. Persistence of parvovirus B19 DNA in anemias: a study of 24 observations. Am J Hematol 1986;
synovial membranes of young patients with and without 23: 271–5.
chronic arthropathy. Lancet 1997; 349: 1063–5. 103 Harris JW. Parvovirus B19 for the hematologist. Am J He-
84 Speyer I, Breedveld FC, Dijkmans BA. Human parvovirus matol 1992; 39: 119–30.
B19 infection is not followed by inflammatory joint disease 104 Rao SP, Miller ST, Cohen BJ. Transient aplastic crisis in
during long term follow-up. A retrospective study of 54 patients with sickle cell disease. B19 parvovirus studies
patients. Clin Exp Rheumatol 1998; 16: 576–8. during a 7-year period. Am J Dis Child 1992; 146: 1328–30.
85 Nikkari S, Luukkainen R, Mottonen T et al. Does parvovirus 105 Muir K, Todd WT, Watson WH, Fitzsimons E. Viral-associ-
B19 have a role in rheumatoid arthritis? Ann Rheum Dis ated haemophagocytosis with parvovirus-B19-related pan-
1994; 53: 106–11. cytopenia. Lancet 1992; 339: 1139–40.
86 Kerr JR, Mattey DL, Thomson W, Poulton KV, Ollier WE. 106 Rao SP, Desai N, Miller ST. B19 parvovirus infection and
Association of symptomatic acute human parvovirus B19 transient aplastic crisis in a child with sickle cell anemia.
infection with human leukocyte antigen class I and II alleles. J Pediatr Hematol Oncol 1996; 18: 175–7.
J Infect Dis 2002; 186: 447–52. 107 Saarinen UM, Chorba TL, Tattersall P et al. Human
87 Jawad AS. Persistent arthritis after human parvovirus B19 parvovirus B19-induced epidemic acute red cell aplasia in
infection. Lancet 1993; 341: 494. patients with hereditary hemolytic anemia. Blood 1986; 67:
88 Klouda PT, Corbin SA, Bradley BA, Cohen BJ, Woolf AD. 1411–7.
HLA and acute arthritis following human parvovirus infec- 108 Smith-Whitley K, Zhao H, Hodinka RL et al. Epidemiology of
tion. Tissue Antigens 1986; 28: 318–9. human parvovirus B19 in children with sickle cell disease.
89 Lu J, Zhi N, Wong S, Brown KE. Activation of synoviocytes Blood 2004; 103: 422–7.
by the secreted phospholipase A2 motif in the VP1-unique 109 Morey AL, Keeling JW, Porter HJ, Fleming KA. Clinical and
region of parvovirus B19 minor capsid protein. J Infect Dis histopathological features of parvovirus B19 infection in the
2006; 193: 582–90. human fetus. Br J Obstet Gynaecol 1992; 99: 566–74.
90 Moffatt S, Tanaka N, Tada K et al. A cytotoxic nonstructural 110 Beghetti M, Gervaix A, Haenggeli CA, Berner M, Rimens-
protein, NS1, of human parvovirus B19 induces activation berger PC. Myocarditis associated with parvovirus B19
of interleukin-6 gene expression. J Virol 1996; 70: 8485–91. infection in two siblings with merosin-deficient congenital
91 Mitchell LA. Parvovirus B19 nonstructural (NS1) protein as muscular dystrophy. Eur J Pediatr 2000; 159: 135–6.
a transactivator of interleukin-6 synthesis: common path- 111 Enders G, Dotsch J, Bauer J et al. Life-threatening parvovirus
way in inflammatory sequelae of human parvovirus infec- B19-associated myocarditis and cardiac transplantation as
tions? J Med Virol 2002; 67: 267–74. possible therapy: two case reports. Clin Infect Dis 1998; 26:
92 Lunardi C, Tiso M, Borgato L et al. Chronic parvovirus B19 355–8.
infection induces the production of anti-virus antibodies 112 Saint-Martin J, Choulot JJ, Bonnaud E, Morinet F. Myocar-
with autoantigen binding properties. Eur J Immunol 1998; ditis caused by parvovirus. J Pediatr 1990; 116: 1007–8.
28: 936–48. 113 Chia JK, Jackson B. Myopericarditis due to parvovirus B19 in
93 Hsu TC, Tsay GJ. Human parvovirus B19 infection in pa- an adult. Clin Infect Dis 1996; 23: 200–1.
tients with systemic lupus erythematosus. Rheumatology 114 Papadogiannakis N, Tolfvenstam T, Fischler B, Norbeck O,
(Oxford) 2001; 40: 152–7. Broliden K. Active, fulminant, lethal myocarditis associated

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


REVIEW: PARVOVIRUS B19 INFECTION 301

with parvovirus B19 infection in an infant. Clin Infect Dis 133 Weiland HT, Salimans MM, Fibbe WE, Kluin PM, Cohen BJ.
2002; 35: 1027–31. Prolonged parvovirus B19 infection with severe anaemia in a
115 Gutersohn A, Zimmermann U, Bartel T, Erbel R. A rare case bone marrow transplant patient. Br J Haematol 1989; 71: 300.
of acute ‘infective’ myocardial infarction triggered by acute 134 Frickhofen N, Arnold R, Hertenstein B, Wiesneth M, Young
parvovirus B19 myocarditis. Nat Clin Pract Cardiovasc Med NS. Parvovirus B19 infection and bone marrow transplan-
2005; 2: 167–71. tation. Ann Hematol 1992; 64(Suppl.): A121–4.
116 Langnas AN, Markin RS, Cattral MS, Naides SJ. Parvovirus 135 Hasle H, Heegaard E, Kerndrup G, Jensen IM, Peterslund
B19 as a possible causative agent of fulminant liver failure and NA, Hornsleth A. Parvovirus B19 infection infrequently
associated aplastic anemia. Hepatology 1995; 22: 1661–5. involved in children and adults with myelodysplastic syn-
117 Sokal EM, Melchior M, Cornu C et al. Acute parvovirus B19 drome. Leuk Res 1996; 20: 81–3.
infection associated with fulminant hepatitis of favourable 136 Heegaard ED, Myhre J, Hornsleth A, Gundestrup M, Boye H.
prognosis in young children. Lancet 1998; 352: 1739–41. Parvovirus B19 infections in patients with chronic anemia.
118 Hillingso JG, Jensen IP, Tom-Petersen L. Parvovirus B19 and Haematologica 1997; 82: 402–5.
acute hepatitis in adults. Lancet 1998; 351: 955–6. 137 Frickhofen N, Abkowitz JL, Safford M et al. Persistent B19
119 Nagai K, Morohoshi T, Kudoh T, Yoto Y, Suzuki N, Matsu- parvovirus infection in patients infected with human
naga Y. Transient erythroblastopenia of childhood with immunodeficiency virus type 1 (HIV-1): a treatable cause of
megakaryocytopenia associated with human parvovirus anemia in AIDS. Ann Intern Med 1990; 113: 926–33.
B19 infection. Br J Haematol 1992; 80: 131–2. 138 Azzi A, Fanci R, Ciappi S, Zakrzewska K, Bosi A. Human
120 Scheurlen W, Ramasubbu K, Wachowski O, Hemauer A, parvovirus B19 infection in bone marrow transplantation
Modrow S. Chronic autoimmune thrombopenia/neutrope- patients. Am J Hematol 1993; 44: 207–9.
nia in a boy with persistent parvovirus B19 infection. J Clin 139 Nour B, Green M, Michaels M et al. Parvovirus B19 infection
Virol 2001; 20: 173–8. in pediatric transplant patients. Transplantation 1993; 56:
121 Heegaard ED, Rosthoj S, Petersen BL, Nielsen S, Karup 835–8.
Pedersen F, Hornsleth A. Role of parvovirus B19 infection in 140 Graeve JL, de Alarcon PA, Naides SJ. Parvovirus B19
childhood idiopathic thrombocytopenic purpura. Acta Pae- infection in patients receiving cancer chemotherapy: the
diatr 1999; 88: 614–7. expanding spectrum of disease. Am J Pediatr Hematol Oncol
122 Nigro G, Zerbini M, Krzysztofiak A et al. Active or recent 1989; 11: 441–4.
parvovirus B19 infection in children with Kawasaki disease. 141 Kurtzman GJ, Cohen BJ, Field AM, Oseas R, Blaese RM,
Lancet 1994; 343: 1260–1. Young NS. Immune response to B19 parvovirus and an
123 Smith SB, Libow LF, Elston DM, Bernert RA, Warschaw KE. antibody defect in persistent viral infection. J Clin Invest
Gloves and socks syndrome: early and late histopathologic 1989; 84: 1114–23.
features. J Am Acad Dermatol 2002; 47: 749–54. 142 Kurtzman G, Frickhofen N, Kimball J, Jenkins DW, Nienhuis
124 Barah F, Vallely PJ, Chiswick ML, Cleator GM, Kerr JR. AW, Young NS. Pure red-cell aplasia of 10 years’ duration
Association of human parvovirus B19 infection with acute due to persistent parvovirus B19 infection and its cure with
meningoencephalitis. Lancet 2001; 358: 729–30. immunoglobulin therapy. N Engl J Med 1989; 321: 519–23.
125 Kerr JR, Barah F, Chiswick ML et al. Evidence for the role of 143 Young NS. Parvovirus infection and its treatment. Clin Exp
demyelination, HLA-DR alleles, and cytokines in the Immunol 1996; 104(Suppl. 1): 26–30.
pathogenesis of parvovirus B19 meningoencephalitis and 144 Chen MY, Hung CC, Fang CT, Hsieh SM. Reconstituted
its sequelae. J Neurol Neurosurg Psychiatry 2002; 73: 739– immunity against persistent parvovirus B19 infection in a
46. patient with acquired immunodeficiency syndrome after
126 Jacobson SK, Daly JS, Thorne GM, McIntosh K. Chronic highly active antiretroviral therapy. Clin Infect Dis 2001; 32:
parvovirus B19 infection resulting in chronic fatigue syn- 1361–5.
drome: case history and review. Clin Infect Dis 1997; 24: 145 Lundqvist A, Tolfvenstam T, Bostic J, Soderlund M, Broliden
1048–51. K. Clinical and laboratory findings in immunocompetent
127 Kerr JR, Bracewell J, Laing I et al. Chronic fatigue syndrome patients with persistent parvovirus B19 DNA in bone mar-
and arthralgia following parvovirus B19 infection. J Rheu- row. Scand J Infect Dis 1999; 31: 11–6.
matol 2002; 29: 595–602. 146 Matano S, Kinoshita H, Tanigawa K, Terahata S, Sugimoto
128 Heegaard ED, Brown KE. Human parvovirus B19. Clin T. Acute parvovirus B19 infection mimicking chronic fati-
Microbiol Rev 2002; 15: 485–505. gue syndrome. Intern Med 2003; 42: 903–5.
129 Nikkari S, Roivainen A, Hannonen P et al. Persistence of 147 Kerr JR, Tyrrell DA. Cytokines in parvovirus B19 infection as
parvovirus B19 in synovial fluid and bone marrow. Ann an aid to understanding chronic fatigue syndrome. Curr Pain
Rheum Dis 1995; 54: 597–600. Headache Rep 2003; 7: 333–41.
130 Wong S, Young NS, Brown KE. Prevalence of parvovirus 148 Tolfvenstam T, Lundqvist A, Levi M, Wahren B, Broliden K.
B19 in liver tissue: no association with fulminant hepatitis Mapping of B-cell epitopes on human parvovirus B19 non-
or hepatitis-associated aplastic anemia. J Infect Dis 2003; structural and structural proteins. Vaccine 2000; 19: 758–
187: 1581–6. 63.
131 Vuorinen T, Lammintausta K, Kotilainen P, Nikkari S. 149 Isa A, Norbeck O, Hirbod T et al. Aberrant cellular immune
Presence of parvovirus B19 DNA in chronic urticaric and responses in humans infected persistently with parvovirus
healthy human skin. J Clin Virol 2002; 25: 217–21. B19. J Med Virol 2006; 78: 129–33.
132 Koch WC, Massey G, Russell CE, Adler SP. Manifestations 150 Jordan JA. Identification of human parvovirus B19 infection
and treatment of human parvovirus B19 infection in in idiopathic nonimmune hydrops fetalis. Am J Obstet Gyn-
immunocompromised patients. J Pediatr 1990; 116: 355–9. ecol 1996; 174: 37–42.

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


302 K . B R O L I D E N et al.

151 Yaegashi N, Niinuma T, Chisaka H et al. The incidence of, 170 Norbeck O, Papadogiannakis N, Petersson K, Hirbod T,
and factors leading to, parvovirus B19-related hydrops Broliden K, Tolfvenstam T. Revised clinical presentation of
fetalis following maternal infection; report of 10 cases and parvovirus B19-associated intrauterine fetal death. Clin In-
meta-analysis. J Infect 1998; 37: 28–35. fect Dis 2002; 35: 1032–8.
152 Brown T, Anand A, Ritchie LD, Clewley JP, Reid TM. 171 Nyman M, Tolfvenstam T, Petersson K, Krassny C, Skjolde-
Intrauterine parvovirus infection associated with hydrops brand-Sparre L, Broliden K. Detection of human parvovirus
fetalis. Lancet 1984; 2: 1033–4. B19 infection in first-trimester fetal loss. Obstet Gynecol
153 Kerr JR, O’Neill HJ, Coyle PV, Thompson W. An outbreak of 2002; 99: 795–8.
parvovirus B19 infection; a study of clinical manifestations 172 Enders M, Weidner A, Zoellner I, Searle K, Enders G. Fetal
and the incidence of fetal loss. Ir J Med Sci 1994; 163: 65–7. morbidity and mortality after acute human parvovirus B19
154 Harger JH, Adler SP, Koch WC, Harger GF. Prospective infection in pregnancy: prospective evaluation of 1018
evaluation of 618 pregnant women exposed to parvovirus cases. Prenat Diagn 2004; 24: 513–8.
B19: risks and symptoms. Obstet Gynecol 1998; 91: 413–20. 173 Jordan JA, Huff D, DeLoia JA. Placental cellular immune
155 Anand A, Gray ES, Brown T, Clewley JP, Cohen BJ. Human response in women infected with human parvovirus B19
parvovirus infection in pregnancy and hydrops fetalis. N during pregnancy. Clin Diagn Lab Immunol 2001; 8: 288–92.
Engl J Med 1987; 316: 183–6. 174 Jordan JA, Butchko AR. Apoptotic activity in villous
156 Eis-Hubinger AM, Dieck D, Schild R, Hansmann M, Sch- trophoblast cells during B19 infection correlates with clin-
neweis KE. Parvovirus B19 infection in pregnancy. Intervi- ical outcome: assessment by the caspase-related M30 Cyto-
rology 1998; 41: 178–84. death antibody. Placenta 2002; 23: 547–53.
157 Enders G, Biber M. Parvovirus B19 infections in pregnancy. 175 von Kaisenberg CS, Bender G, Scheewe J et al. A case of fetal
Behring Inst Mitt 1990; 85: 74–8. parvovirus B19 myocarditis, terminal cardiac heart failure,
158 Rodis JF, Borgida AF, Wilson M et al. Management of par- and perinatal heart transplantation. Fetal Diagn Ther 2001;
vovirus infection in pregnancy and outcomes of hydrops: a 16: 427–32.
survey of members of the Society of Perinatal Obstetricians. 176 Bultmann BD, Klingel K, Nabauer M, Wallwiener D, Kandolf
Am J Obstet Gynecol 1998; 179: 985–8. R. High prevalence of viral genomes and inflammation in
159 Garcia AG, Pegado CS, Cubel Rde C, Fonseca ME, Sloboda I, peripartum cardiomyopathy. Am J Obstet Gynecol 2005;
Nascimento JP. Feto-placentary pathology in human par- 193: 363–5.
vovirus B19 infection. Rev Inst Med Trop Sao Paulo 1998; 40: 177 Musiani M, Zerbini M, Gentilomi G, Plazzi M, Gallinella G,
145–50. Venturoli S. Parvovirus B19 clearance from peripheral blood
160 Sheikh AU, Ernest JM, O’Shea M. Long-term outcome in fetal after acute infection. J Infect Dis 1995; 172: 1360–3.
hydrops from parvovirus B19 infection. Am J Obstet Gynecol 178 Cassinotti P, Siegl G. Quantitative evidence for persistence of
1992; 167: 337–41. human parvovirus B19 DNA in an immunocompetent
161 Nunoue T, Kusuhara K, Hara T. Human fetal infection with individual. Eur J Clin Microbiol Infect Dis 2000; 19: 886–7.
parvovirus B19: maternal infection time in gestation, viral 179 Cassinotti P, Burtonboy G, Fopp M, Siegl G. Evidence for
persistence and fetal prognosis. Pediatr Infect Dis J 2002; 21: persistence of human parvovirus B19 DNA in bone marrow.
1133–6. J Med Virol 1997; 53: 229–32.
162 Nyman M, Skjoldebrand-Sparre L, Broliden K. Non-hydropic 180 Lindblom A, Isa A, Norbeck O et al. Slow clearance of hu-
intrauterine fetal death more than 5 months after primary man parvovirus B19 viremia following acute infection. Clin
parvovirus B19 infection. J Perinat Med 2005; 33: 176–8. Infect Dis 2005; 41: 1201–3.
163 Kinney JS, Anderson LJ, Farrar J et al. Risk of adverse out- 181 McMichael AJ, Gotch FM, Noble GR, Beare PA. Cytotoxic T-
comes of pregnancy after human parvovirus B19 infection. cell immunity to influenza. N Engl J Med 1983; 309: 13–7.
J Infect Dis 1988; 157: 663–7. 182 Kagi D, Ledermann B, Burki K et al. Cytotoxicity mediated
164 Katz VL, McCoy MC, Kuller JA, Hansen WF. An association by T cells and natural killer cells is greatly impaired in
between fetal parvovirus B19 infection and fetal anomalies: perforin-deficient mice. Nature 1994; 369: 31–7.
a report of two cases. Am J Perinatol 1996; 13: 43–5. 183 Lechner F, Wong DK, Dunbar PR et al. Analysis of successful
165 Tiessen RG, van Elsacker-Niele AM, Vermeij-Keers C, Oepkes immune responses in persons infected with hepatitis C virus.
D, van Roosmalen J, Gorsira MC. A fetus with a parvovirus J Exp Med 2000; 191: 1499–512.
B19 infection and congenital anomalies. Prenat Diagn 1994; 184 Tolfvenstam T, Oxenius A, Price DA et al. Direct ex vivo
14: 173–6. measurement of CD8(+) T-lymphocyte responses to human
166 Weiland HT, Vermey-Keers C, Salimans MM, Fleuren GJ, parvovirus B19. J Virol 2001; 75: 540–3.
Verwey RA, Anderson MJ. Parvovirus B19 associated with 185 Franssila R, Hokynar K, Hedman K. T helper cell-mediated
fetal abnormality. Lancet 1987; 1: 682–3. in vitro responses of recently and remotely infected subjects
167 Guidozzi F, Ballot D, Rothberg AD. Human B19 parvovirus to a candidate recombinant vaccine for human parvovirus
infection in an obstetric population. A prospective study B19. J Infect Dis 2001; 183: 805–9.
determining fetal outcome. J Reprod Med 1994; 39: 36–8. 186 von Poblotzki A, Gerdes C, Reischl U, Wolf H, Modrow S.
168 Skjoldebrand-Sparre L, Tolfvenstam T, Papadogiannakis N, Lymphoproliferative responses after infection with human
Wahren B, Broliden K, Nyman M. Parvovirus B19 infection: parvovirus B19. J Virol 1996; 70: 7327–30.
association with third-trimester intrauterine fetal death. 187 Corcoran A, Doyle S, Waldron D, Nicholson A, Mahon BP.
BJOG 2000; 107: 476–80. Impaired gamma interferon responses against parvovirus B19
169 Tolfvenstam T, Papadogiannakis N, Norbeck O, Petersson K, by recently infected children. J Virol 2000; 74: 9903–10.
Broliden K. Frequency of human parvovirus B19 infection in 188 Franssila R, Auramo J, Modrow S et al. T helper cell-medi-
intrauterine fetal death. Lancet 2001; 357: 1494–7. ated interferon-gamma expression after human parvovirus

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


REVIEW: PARVOVIRUS B19 INFECTION 303

B19 infection: persisting VP2-specific and transient VP1u- 206 Bluth MH, Norowitz KB, Chice S et al. Detection of IgE anti-
specific activity. Clin Exp Immunol 2005; 142: 53–61. parvovirus B19 and increased CD23+ B cells in parvovirus
189 Lindner J, Barabas S, Saar K et al. CD4(+) T-cell responses B19 infection: relation to Th2 cytokines. Clin Immunol 2003;
against the VP1-unique region in individuals with recent 108: 152–8.
and persistent parvovirus B19 infection. J Vet Med B Infect 207 von Poblotzki A, Hemauer A, Gigler A et al. Antibodies to
Dis Vet Public Health 2005; 52: 356–61. the nonstructural protein of parvovirus B19 in persistently
190 Mitchell LA, Leong R, Rosenke KA. Lymphocyte recognition infected patients: implications for pathogenesis. J Infect Dis
of human parvovirus B19 non-structural (NS1) protein: 1995; 172: 1356–9.
associations with occurrence of acute and chronic arthro- 208 von Poblotzki A, Gigler A, Lang B, Wolf H, Modrow S.
pathy? J Med Microbiol 2001; 50: 627–35. Antibodies to parvovirus B19 NS-1 protein in infected
191 Norbeck O, Isa A, Pohlmann C et al. Sustained CD8+ T-cell individuals. J Gen Virol 1995; 76: 519–27.
responses induced after acute parvovirus B19 infection in 209 Hemauer A, Gigler A, Searle K et al. Seroprevalence of par-
humans. J Virol 2005; 79: 12117–21. vovirus B19 NS1-specific IgG in B19-infected and uninfected
192 Isa A, Kasprowicz V, Norbeck O et al. Prolonged activation of individuals and in infected pregnant women. J Med Virol
virus-specific CD8+ T cells after acute B19 infection. PLoS 2000; 60: 48–55.
Med 2005; 2: e343. 210 Jones LP, Erdman DD, Anderson LJ. Prevalence of antibodies
193 Hislop AD, Annels NE, Gudgeon NH, Leese AM, Rickinson to human parvovirus B19 nonstructural protein in persons
AB. Epitope-specific evolution of human CD8(+) T cell re- with various clinical outcomes following B19 infection.
sponses from primary to persistent phases of Epstein-Barr J Infect Dis 1999; 180: 500–4.
virus infection. J Exp Med 2002; 195: 893–905. 211 Searle K, Schalasta G, Enders G. Development of antibodies
194 Saikawa T, Anderson S, Momoeda M, Kajigaya S, Young NS. to the nonstructural protein NS1 of parvovirus B19 during
Neutralizing linear epitopes of B19 parvovirus cluster in the acute symptomatic and subclinical infection in pregnancy:
VP1 unique and VP1-VP2 junction regions. J Virol 1993; implications for pathogenesis doubtful. J Med Virol 1998; 56:
67: 3004–9. 192–8.
195 Kerr S, O’Keeffe G, Kilty C, Doyle S. Undenatured parvovirus 212 Venturoli S, Gallinella G, Manaresi E, Gentilomi G,
B19 antigens are essential for the accurate detection of Musiani M, Zerbini M. IgG response to the immunoreac-
parvovirus B19 IgG. J Med Virol 1999; 57: 179–85. tive region of parvovirus B19 nonstructural protein by
196 Manaresi E, Gallinella G, Zerbini M, Venturoli S, Gentilomi immunoblot assay with a recombinant antigen. J Infect Dis
G, Musiani M. IgG immune response to B19 parvovirus VP1 1998; 178: 1826–9.
and VP2 linear epitopes by immunoblot assay. J Med Virol 213 de Jong EP, de Haan TR, Kroes AC, Beersma MF, Oepkes D,
1999; 57: 174–8. Walther FJ. Parvovirus B19 infection in pregnancy. J Clin
197 Badovinac VP, Porter BB, Harty JT. Programmed contraction Virol 2006; 36: 1–7.
of CD8(+) T cells after infection. Nat Immunol 2002; 3: 619– 214 Enders M, Schalasta G, Baisch C et al. Human parvovirus
26. B19 infection during pregnancy – value of modern
198 Hou S, Hyland L, Ryan KW, Portner A, Doherty PC. Virus- molecular and serological diagnostics. J Clin Virol 2006; 35:
specific CD8+ T-cell memory determined by clonal burst size. 400–6.
Nature 1994; 369: 652–4. 215 Skjoldebrand-Sparre L, Nyman M, Broliden K, Wahren B. All
199 Zhou S, Ou R, Huang L, Price GE, Moskophidis D. Differential cases of intrauterine fetal death should be evaluated for
tissue-specific regulation of antiviral CD8+ T-cell immune parvovirus B19 viral deoxyribonucleic acid. Am J Obstet
responses during chronic viral infection. J Virol 2004; 78: Gynecol 1999; 180: 1595–6.
3578–600. 216 Petersson K, Norbeck O, Westgren M, Broliden K. Detection
200 Probst HC, Tschannen K, Gallimore A et al. Immunodomi- of parvovirus B19, cytomegalovirus and enterovirus infec-
nance of an antiviral cytotoxic T cell response is shaped by tions in cases of intrauterine fetal death. J Perinat Med 2004;
the kinetics of viral protein expression. J Immunol 2003; 32: 516–21.
171: 5415–22. 217 Wolff K, Broliden K, Marsk A, Tolfvenstam T, Papa-
201 Nayersina R, Fowler P, Guilhot S et al. HLA A2 restricted dogiannakis N, Westgren M. One stillborn and one severely
cytotoxic T lymphocyte responses to multiple hepatitis B hydropic twin due to parvovirus B19 infection; successful
surface antigen epitopes during hepatitis B virus infection. outcome of the surviving twin. Acta Obstet Gynecol Scand
J Immunol 1993; 150: 4659–71. 1999; 78: 828–30.
202 Rehermann B, Lau D, Hoofnagle JH, Chisari FV. Cytotoxic T 218 Knoll A, Louwen F, Kochanowski B et al. Parvovirus B19
lymphocyte responsiveness after resolution of chronic infection in pregnancy: quantitative viral DNA analysis
hepatitis B virus infection. J Clin Invest 1996; 97: 1655–65. using a kinetic fluorescence detection system (TaqMan PCR).
203 Corcoran A, Crowley B, Dewhurst C, Pizer BL, Doyle S. J Med Virol 2002; 67: 259–66.
Establishment of functional B cell memory against parvovi- 219 Hokynar K, Norja P, Laitinen H et al. Detection and differ-
rus B19 capsid proteins may be associated with resolution of entiation of human parvovirus variants by commercial
persistent infection. J Med Virol 2006; 78: 125–8. quantitative real-time PCR tests. J Clin Microbiol 2004; 42:
204 Shneerson JM, Mortimer PP, Vandervelde EM. Febrile illness 2013–9.
due to a parvovirus. Br Med J 1980; 280: 1580. 220 Liefeldt L, Plentz A, Klempa B et al. Recurrent high level
205 Erdman DD, Usher MJ, Tsou C et al. Human parvovirus B19 parvovirus B19/genotype 2 viremia in a renal trans-
specific IgG, IgA, and IgM antibodies and DNA in serum plant recipient analyzed by real-time PCR for simulta-
specimens from persons with erythema infectiosum. J Med neous detection of genotypes 1 to 3. J Med Virol 2005;
Virol 1991; 35: 110–5. 75: 161–9.

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304


304 K . B R O L I D E N et al.

221 Lefrere JJ, Servant-Delmas A, Candotti D et al. Persistent B19 232 Selbing A, Josefsson A, Dahle LO, Lindgren R. Parvovirus
infection in immunocompetent individuals: implications for B19 infection during pregnancy treated with high-dose
transfusion safety. Blood 2005; 106: 2890–5. intravenous gammaglobulin. Lancet 1995; 345: 660–1.
222 Goldstein AR, Anderson MJ, Serjeant GR. Parvovirus asso- 233 Matsuda H, Sakaguchi K, Shibasaki T, Takahashi H, Ka-
ciated aplastic crisis in homozygous sickle cell disease. Arch wakami Y, Furuya K. Intrauterine therapy for parvovirus
Dis Child 1987; 62: 585–8. B19 infected symptomatic fetus using B19 IgG-rich high
223 Koduri PR, Kumapley R, Valladares J, Teter C. Chronic pure titer gammaglobulin. J Perinat Med 2005; 33: 561–3.
red cell aplasia caused by parvovirus B19 in AIDS: use of 234 Ballou WR, Reed JL, Noble W, Young NS, Koenig S. Safety
intravenous immunoglobulin – a report of eight patients. and immunogenicity of a recombinant parvovirus B19
Am J Hematol 1999; 61: 16–20. vaccine formulated with MF59C.1. J Infect Dis 2003; 187:
224 Carstensen H, Ornvold K, Cohen BJ. Human parvovirus B19 675–8.
infection associated with prolonged erythroblastopenia in a 235 Bansal GP, Hatfield JA, Dunn FE et al. Candidate recom-
leukemic child. Pediatr Infect Dis J 1989; 8: 56. binant vaccine for human B19 parvovirus. J Infect Dis 1993;
225 Smith MA, Shah NR, Lobel JS, Cera PJ, Gary GW, Anderson 167: 1034–44.
LJ. Severe anemia caused by human parvovirus in a leuke- 236 Young NS, Brown KE. Parvovirus B19. N Engl J Med 2004;
mia patient on maintenance chemotherapy. Clin Pediatr 350: 586–97.
(Phila) 1988; 27: 383–6. 237 Hung CC, Lee KL, Chen MY. Increase in B19 viral load prior
226 Brennand JE, Cameron AD. Human parvovirus B19 in to relapse of anaemia in an AIDS patient with persistent B19
pregnancy. Hosp Med 2000; 61: 93–6. infection. J Infect 2001; 43: 150–2.
227 von Kaisenberg CS, Jonat W. Fetal parvovirus B19 infection. 238 Pallier C, Greco A, Le Junter J, Saib A, Vassias I, Morinet F.
Ultrasound Obstet Gynecol 2001; 18: 280–8. The 3¢ untranslated region of the B19 parvovirus capsid
228 Schild RL, Bald R, Plath H, Eis-Hubinger AM, Enders G, protein mRNAs inhibits its own mRNA translation in non-
Hansmann M. Intrauterine management of fetal parvovirus permissive cells. J Virol 1997; 71: 9482–9.
B19 infection. Ultrasound Obstet Gynecol 1999; 13: 161–6. 239 Anderson LJ, Young NS (eds). Human Parvovirus B.19, Vol.
229 Smoleniec JS, Pillai M. Management of fetal hydrops asso- 20. Basel: Karger, 1997.
ciated with parvovirus B19 infection. Br J Obstet Gynaecol
1994; 101: 1079–81. Correspondence: Prof. Kristina Broliden, Department of Medicine,
230 Miller E, Fairley CK, Cohen BJ, Seng C. Immediate and long Solna, Unit of Infectious Diseases, B2:00, Center for Molecular
term outcome of human parvovirus B19 infection in preg- Medicine, Karolinska Institutet, Karolinska University Hospital,
nancy. Br J Obstet Gynaecol 1998; 105: 174–8. Solna, 171 76 Stockholm, Sweden.
231 Forestier F, Tissot JD, Vial Y, Daffos F, Hohlfeld P. Haema- (fax: +46 8 7178501; e-mail: kristina.broliden@karolinska.se).
tological parameters of parvovirus B19 infection in 13
fetuses with hydrops foetalis. Br J Haematol 1999; 104: 925–
7.

 2006 Blackwell Publishing Ltd Journal of Internal Medicine 260: 285–304

Você também pode gostar