Self-exclusion ‘self-deferral’ of donors Retrospective analysis indicates that the risk of con-tracting AIDS from the transfusion of blood and blood components other than products of pla
Trang 1The first reported case of transfusion-associated
AIDS (TAA) turned out to be an 18-month-old infant
with severe combined immunodeficiency who had
been transfused repeatedly at birth and had received a
unit of platelets from a donor who subsequently
devel-oped AIDS (Amman et al 1983) By 1984, 38 cases
of AIDS had been reported in patients with no risk
factors other than a history of transfusion Nineteen
of the patients were adults who, during the previous
5 years, had received blood components derived from
unpooled donations In those cases in which all the
donors could be followed up, an individual in a
‘high-risk’ group could always be identified (Curran et al.
1984) An expanded study of 194 patients showed that
in most cases the high-risk donor was anti-HIV
posit-ive, and in those few cases in which the high-risk
donor lacked anti-HIV, another donor tested positive
(Peterman et al 1985) A total of 157 525 cases of
AIDS was reported in the USA by the end of 1990, and
5371 (3.4%) of these were attributed to the
trans-fusion of blood or blood components The median
incubation period, estimated as the time from
expos-ure by transfusion to diagnosis of AIDS, has been
estimated to be 34 months in adults and 22 months in
children (Peterman 1987), although later estimates
indicated a longer period with a median of 7–8 years
for adults and 3 –5 years for children (Rogers 1992)
In total, 50% of untreated subjects infected with HIV
by transfusion will develop AIDS within 7 years
com-pared with 33% of subjects infected by other routes
(Ward et al 1989).
Following the first reports, the number of cases of
TAA in the USA increased rapidly By the end of 1991,
out of a total of 206 392 cases of AIDS, there were
6060 adults and 472 infants or children who had
acquired the disease by transfusion of blood or blood
products, representing 3% and 13.6% of the total
cases in adults and children respectively (CDC 1992)
The risk of transmission of HIV by blood transfusion
is now trivial in countries without major heterosexual
spread and in which donor education, encouragement
of self-exclusion and screening for HIV antibodies
have been established since 1985 In these countries,
the risk of transmission of HIV is almost solely
attributable to donations given during the window
period (see below)
In western Europe, HIV prevalence among blood
donors has declined progressively since the onset of
systematic testing and, according to the European
Centre for Epidemiologic Monitoring of AIDS,approximated 1.3 positives per 100 000 donations
in 2002 (www.eurohiv.org) Since 1995, the ence has remained relatively constant in Belgium,Scandinavia, Ireland, the Netherlands and the UK, hasdecreased regularly in France and Spain, but hasremained above 2 per 100 000 in Italy, Greece andPortugal (Hamers and Downs 2004) In EasternEurope, prevalence has increased markedly since
preval-1995, now exceeding 30 per 100 000 in 2002 Thehighest levels are reported in the Ukraine (93), Estonia(54) followed by Azerbaijan, Georgia and the RussianFederation
Because HIV is both cell associated and present inplasma, all blood components are potentially infec-tious (Curran 1985) The viral load has been estimated
at between 1.5 × 104tissue culture infective doses in a250-ml unit of blood from an asymptomatic donor,
to 1.75 × 106from a unit drawn from a symptomatic
person (Ho et al 1989) The relative importance of the
viral strain, concurrent infection by other blood-borneagents, cellular receptors for HIV and other geneticand acquired host factors, both for infection and forclinical course of HIV in transfusion recipients, hasreceived a great deal of attention, but is still an area
of intense research (Vicenzi et al 1997; Keoshkerian
et al 2003; Zaunders et al 2004).
Follow-up studies have shown that 90–95% ofpatients receiving blood or blood components fromanti-HIV-positive donors have become infected (Ward
et al 1987; Donegan et al 1990b) The virus is well
preserved in refrigerated and frozen blood; however,components that are washed, leucoreduced or coldstored for several weeks, procedures that diminish the number of viable leucocytes or the amount of virus, reduce the likelihood of transfusion transmis-
sion (Donegan et al 1990a) Neither donor status nor
recipient characteristics affect the likelihood of HIV
transmission (Busch et al 1990a) However, when
AIDS developed in the donor shortly after donation,the period of asymptomatic infection in the recipient
was also shortened (Ward et al 1989) Albumin
pre-parations, immunoglobulins, antithrombin III andhepatitis B vaccine have not been associated with HIVinfection (Desmyter 1986; Melbye 1986; Morgenthaler1989; Cuthbertson 1991) Furthermore, when HIV isadded to plasma and the plasma is then fractionated bythe cold-ethanol process, HIV does not appear in the Igfraction (Morgenthaler 1989)
Trang 2TAA in infants, and AIDS in infants and children in
general, has a shorter incubation period than in adults
and many of the clinical manifestations of the disease
are different (Rogers 1992) Infants born to
anti-HIV-positive mothers who become infected perinatally and
infants transfused during the first years of life have the
shortest median incubation period (less than 5 years),
and usually develop AIDS in the first year of life The
increased susceptibility of infants to AIDS may be
related to their immature immune system and to the
larger viral load relative to body size Children likely to
develop TAA are those who are likely to be transfused:
premature infants and children with haemophilia,
thalassaemia and sickle cell anaemia In the USA, of
2734 children with AIDS, 250 (9.1%) were
transfu-sion associated and 138 (5.0% of the total) occurred in
children with coagulation factor deficiencies (CDC
1991)
In much of Asia and Africa, the transmission of HIV
by blood transfusion is still an important source of
infection Reasons for an alarmingly high rate of
trans-mission, reported to be up to 10% of all cases, include:
(1) the high demands for blood for inpatients with
severe anaemia and haemorrhage, mainly in obstetrics,
gynaecology and paediatrics; (2) the prevalence of
HIV infection amongst the donor population, which
can be as high as 20%; (3) the fact that HIV infection is
not confined to a minority of the population who can
be requested to refrain from blood donation; and
(4) the inability of many laboratories to test for HIV or
to perform and control the tests properly The patient
groups at greatest risk of acquiring HIV-1 or -2 by
blood transfusion in tropical Africa are children with
malaria and anaemia, patients with sickle cell anaemia
(120 000 infants with sickle cell disease are born each
year in Africa), anaemic antenatal patients, women
with severe obstetric haemorrhage and trauma victims
(Fleming 1990)
Transfusion-associated AIDS and haemophilia
To December 1996, 4674 cases of AIDS were reported
in patients with haemophilia, accounting for less
than 1% of the 581 429 AIDS cases reported in
adults and children Of the 7629 cases of AIDS
reported in children under the age of 13 years, 373
(5%) were recipients of blood or tissue and 231 (3%)
were haemophiliacs (Centers for Disease Control and
Prevention 1997) All but 39 of these infections
occurred prior to testing blood and plasma donors for HIV
Studies of patient cohorts and specimens in serumrepositories revealed that more than 90% of severehaemophiliacs (subjects with less than 1% factor VIIIactivity) treated with factor VIII concentrate had been
infected prior to 1984 (Evatt et al 1985) In a study of
a 16-centre cohort of haemophiliacs in the USA andEurope, infections first appeared in 1978, peaked
in October 1982 and declined to an estimated fourinfections per 100 person-years by July 1984 (Kroner
et al 1994) For patients who were high-dose
recipi-ents, peak risk appeared even earlier, indicating thatthe majority of patients with haemophilia wereinfected before the disease was widely recognized andlong before it was attributed to transfusion The riskwas related to each patient’s mean annual dose of clot-ting factor concentrate As clotting factor concentratesare prescribed on the basis of patient weight or plasmavolume, older patients with severe disease generallyreceived more concentrate, had more ‘donor expos-ures’ and seroconverted sooner than did children andpatients with milder disease When corrected for doseand severity of disease, the association between ageand early seroconversion disappears The cumulativeincidence of infection was 96% for high-dose recipi-ents, 92% for moderate-dose recipients and 56% forlow-dose recipients Subjects who received only single-donor products (plasma and/or cryoprecipitate) hadthe lowest cumulative incidence of infection, 16%
(Kroner et al 1994) This experience is consistent with other reports (Andes et al 1988; Gjerset et al 1991).
These startling numbers underscore the potential public health risks of using transfusion products manufactured from pools of plasma drawn from
20 000 donors or more
Although AIDS was first reported in threehaemophiliacs in 1982, studies of serum samplesstored from as far back as 1968 have shown that the first cases of the development of anti-HIV inhaemophiliacs occurred in 1978 in the USA and in
1981 in the UK (Evatt et al 1983; Machin et al 1985; Ragni et al 1986) Plasma for preparing the implicated
clotting factor concentrates may have been collected ayear prior to that The prevalence of HIV seropositiv-ity and of AIDS varies from one haemophilia centre toanother depending on the source, volume and type ofconcentrate used In a study of 13 haemophilia centres
in western Europe, Canada and Australia involving
Trang 32370 patients with haemophilia A and 434 patients
with haemophilia B, the overall incidence of
anti-HIV was 53.6% (CDC 1987a) The percentage of
haemophiliacs infected with HIV in different countries
has varied from 4% to more than 60%, with the higher
rates in those countries using mainly concentrates
from plasma imported from the USA Some batches of
factor VIII concentrate from European plasma have
also transmitted HIV (Madhok and Forbes 1990) A
clear correlation was also found between the severity
of haemophilia and HIV seropositivity (Melbye 1986;
UK Haemophilia Centre 1986) Haemophiliacs treated
before 1985 with cryoprecipitate alone have shown
a low risk of HIV infection (Ragni et al 1986; UK
Haemophilia Centre 1986)
Patients with haemophilia B fared somewhat better
(Evatt et al 1984; Mannucci et al 1985; Ragni et al.
1986; UK Haemophilia Centre 1986) The difference
may be due partly to the uneven partitioning of HIV in
infected plasma during fractionation, with HIV
sepa-rating preferentially in the cryoprecipitate fraction
(Aronson 1979; Morgenthaler 1989; Madhok and
Forbes 1990) The source of plasma used for
fractiona-tion is also partly responsible for this difference In
many countries all the factor IX is prepared locally,
whereas at least part of the factor VIII is imported
from the USA Approximately 70% of patients in the
USA with haemophilia A and 35% with haemophilia
B developed HIV antibodies before the introduction
of methods for viral inactivation in blood products
(CDC 1987b) In a multicentre study of haemophiliacs
treated in the UK between 1980 and 1984, 896 (44%)
of 2025 patients with haemophilia A were positive for
anti-HIV; 20 (6%) of 324 patients with haemophilia B
and 11 (5%) of 215 patients with von Willebrand’s
disease were seropositive
Although a large number of severe haemophiliacs in
the UK were treated up to 1983 with unheated factor
VIII concentrate imported from the USA, the incidence
of anti-HIV in haemophiliacs in the UK is much lower
than in countries such as Germany, Spain and the USA,
where the frequency of anti-HIV ranges between 68%
and 94% (Kitchen et al 1984; 1985) On the other
hand, in Groningen, in the Netherlands, only 1 out
of 18 severe haemophiliacs treated with commercial
factor VIII concentrates between 1978 and 1983
developed HIV antibodies (van der Meer et al 1986).
There has been no transmission of HBV, HCV
or HIV by US-licensed plasma derivatives since the
introduction of effective virus inactivation procedures(Tabor 1999)
Prevention of transfusion-transmitted HIVinfection
Reducing the residual risk
The reduction in risk of transfusion-transmitted HIV over the past 15 years has been dramatic and reassuring Nevertheless, enormous public concernpersists As in almost no other area, blood safety, and specifically the possibility of HIV transmission,provokes emotions and measures to further reducerisks that defy the conventional cost–benefit calculus.Actions to reduced residual risk fall into three generalcategories: (1) measures that can be introduced byblood collection facilities, such as improved donorscreening, testing, education and exclusion techniques;(2) enlightened transfusion practice, such as judicioususe of allogeneic blood components, appropriate use
of autologous blood, and alternatives to transfusion(see Chapter 17); and (3) measures that depend on thedevelopment of new technologies, such as viral inac-tivation of cellular components and safe substitutes for blood
Donor demographics have proved effective at fying and excluding donors at high risk for infection
identi-and transmission of HIV (Busch et al 1991a) Donors
with high-risk profiles include men who have had sexual contact with other men since 1977, intraven-ous drug users, residents of high prevalence regions,prisoners, prostitutes, haemophiliacs who have received
‘unsafe’ clotting factor concentrates and the sexualpartners of people in all of these groups The year 1977was chosen as the point of reference, because the firstclinical cases of AIDS in the USA were diagnosed retro-
spectively as far back as 1978 (Jaffe et al 1985) The
rate of seropositivity has been higher in paid plasmadonors than in volunteers Seven HIV positives werefound out of 35 000 plasma donors attending centres
located outside high prevalence areas (Stramer et al.
donor education are not infallible (Leitman et al.
1989) Between 1% and 2% of donors do not report
Trang 4risks that would disqualify them from blood donation,
and donation incentives such as complimentary
labor-atory testing increases this rate (Glynn et al 2001)
In a study of blood donors found positive for hepatitis
C antibody, 42% admitted to intravenous drug use on
subsequent questioning, despite denying such use on
predonation screening (Conry-Cantilena et al 1996).
In an anonymous mail survey of 50 162 volunteer
allo-geneic blood donors, 1.9% of the 34 726 respondents
reported one or more risk factors that should have led
to their deferral at the time of their last donation
(Williams et al 1997) Refinements in blood donor
screening techniques, such as the use of illustrated
risk-related activities, expansion of the screening
question-naire and interactive computer-based screening, have
been proposed, but data supporting the effectiveness
of such measures are lacking (Mayo et al 1991)
How-ever, the reasons why some volunteer blood donors
appear to disregard certain screening criteria are
un-known and may have more to do with donor
psycho-logy than with inadequate screening and education
Donors who are confirmed positive for HIV should
be counselled and referred to specialized centres for
follow-up Counselling should be performed by
spe-cially trained staff Appropriate interventions will not
only help the donor to obtain long-term supportive care
and to prevent further spread, but will also aid
transfu-sion services to understand which groups of the
popu-lation are HIV seropositive and still come forward
to donate (Lefrere et al 1992) Donor education and
selection methods can then be modified accordingly
Self-exclusion (‘self-deferral’) of donors
Retrospective analysis indicates that the risk of
con-tracting AIDS from the transfusion of blood and blood
components (other than products of plasma
fractiona-tion) prior to focused screening and testing was far
higher than the one infection per million units
trans-fused that had been estimated during the 1983–1985
interval The risk of HIV transmission from
trans-fusion in San Francisco has been calculated from its
first appearance in 1978 to rise exponentially to a peak
risk of approximately 1.1% per unit transfused in
1983 (Busch et al 1991a) A retrospective study of
heavily transfused patients with leukaemia in New
York City revealed an overall risk between 0.02% and
0.11% per component transfused (Minamoto et al.
1988) The major blood collectors published a Joint
Statement of Recommendations in January 1983
(American Association of Blood Banks et al 1983),
and the Public Health Service published tions in March 1983 (CDC 1983) that proposed suchmeasures as public education, self-deferral for donorsengaging in high-risk activity and confidential unit
recommenda-exclusion procedures (Pindyck et al 1984, 1985) These
measures proved unusually effective An estimated90% of men in high-risk cat-egories self deferred By
1984, the risk in San Francisco had dropped to lessthan 0.2% per unit Screening with anti-HIV-1 in 1985
reduced the risk to about 1 in 40 000 units (Busch et al.
1991a)
Routine screening tests for HIV in blood donors
In most countries, screening tests for anti-HIV by the enzyme immunoassay (EIA) format are now com-pulsory for all blood donations If reactive (‘repeatreactive’), additional tests for HIV using independentmethods are used to confirm the diagnosis of HIV
infection (Stramer et al 2004) The current algorithm
in the USA requires that anti-HIV-1/2 repeat reactivespecimens be further tested with the HIV-1 Westernblot (see below) and a specific anti-HIV-2 EIA EarlyELISA assays using disrupted purified virus wereplagued by false-positive reactions Current genera-tion screening assays, using recombinant and syntheticantigens, have reduced false reactives dramatically whileincreasing test sensitivity for both the predominantand the variant viral strains (Busch and Alter 1995).Less than 10% of reactive assays confirm positiveusing this strategy Western blot is notoriously subjec-tive and complicated by non-viral bands (Kleinman
et al 1998) Alternative strategies use a second EIA or
a NAT assay of HIV RNA
Approximately 25.6 million donations were screened
by the American National Red Cross from September
1999 to 30 June 2003, resulting in 17 090 HIV repeat
reactive blood donations (Stramer et al 2004) Only
4.8% of these donors (818) were Western blot positiveand approximately 90% (759) of those also tested positive by NAT Follow-up testing of the remaining10% demonstrated that almost all of these donors represent Western blot false-positive results
Although some antibodies to core and other
anti-gens of HIV-1 and HIV-2 crossreact (Sazama et al.
1992), currently available tests are designed to detectboth anti-HIV-1 and anti-HIV-2 Such combined
Trang 5assays are available as antiglobulin ELISAs and as
sandwich ELISAs and are routinely used in several
countries Seroconversion in infection with HIV-1 is
detected earlier in these combined assays than by
anti-HIV-1 assays (Gallarda et al 1994; Fiebig et al.
2003) Most of the combined assays have been found
to be less sensitive for the detection of anti-HIV-2
than most anti-HIV-2 specific tests (Christiansen et al.
1996)
Some samples with anti-HIV are repeatedly positive
in some, but negative in other screening assays If only
one screening test is used, such samples may give
false-negative results (Hancock et al 1993) One cause of
such false negatives is that antibodies against subtype
O, a variant found predominantly in West Africa, are
not recognized in all screening assays (Loussert-Ajaka
et al 1994; Schable et al 1994) At present group O
prevalence is low in the USA and in western Europe
(Sullivan et al 2000) False-negative reactions have
also been found to be due to contamination with glove
powder, inhibition by serum proteins, haemoglobin
and certain anticoagulants (Sazama 1995)
Currently available ELISAs for HIV-1 antibodies
detect HIV-1 subtype group 0 Soon after the
dis-covery of an assay for HIV-Ab, transmission of HIV by
blood from seronegative donors had been recognized
(Esteban et al 1985; Raevsky et al 1986; Ward et al.
1988; Cohen and Munoz 1989) Studies reported
detection of HIV-1 p24 antigen in analyses of stored
blood specimens from plasma donors as early as 1986,
and confirmed cases of HIV-Ag-positive, Ab-negative
blood in primary HIV infection were reported by 1988
(Allain et al 1986; Clark et al 1991; Irani et al 1991).
The utility of this test as a screening assay was not
so obvious Antigen tests are positive for only part of
the initial antibody-negative viraemic phase In some
subjects antigen can be detected as early as 2 weeks
after infection, persisting for between 3 weeks and
3 months, and is no longer detectable when anti-p24
appears in the serum, although it may reappear
inter-mittently during the asymptomatic phase (Allain et al.
1986; Fiebig et al 2003) Later, antigen may reappear
with a loss of anti-p24 A prospective study of 515 494
units donated at 13 blood centres in the USA failed
to detect a single instance of Ag-positive/Ab-negative
donated blood A retrospective analysis of 200 000
repository specimens and prospective studies of blood
donors in Europe confirmed these findings (Backer
et al 1987; Busch et al 1990a) However, after three
anti-HIV seroconversions followed transfusion of p24antigen-positive units, testing of donated blood for p24antigen was mandated in the USA in 1996 (Busch andAlter 1995) Mathematical models predicted that uni-versal antigen screening would detect eight additionalpotentially infectious units per year In fact it took
5 years before eight antigen-positive/antibody-negative
units were interdicted (Kleinman et al 1997; Kleinman
and Busch 2000) Because of this limited usefulnessand troubling frequency of false positives, HIV p24antigen screening was not adopted widely outside ofthe USA With the adoption of universal NAT for HIV
in the USA in 1999 and its licensure in 2002, HIV-Ag
screening was rendered unnecessary (Busch et al 2000).
Confirmatory tests: they do not always confirm
The Western blot is the most widely used additional
or ‘confirmatory’ test for HIV The criteria for theinterpretation of Western blot results have been re-evaluated several times because of greater sensitivityand specificity of screening assays and Western blotreagents, better insight into the serological patterns ofHIV infection, experience with patterns of non-specificreactivity in low- and high-risk populations andknowledge of the serological basis of non-specificity
(Sayre et al 1996) Samples are now considered to
be WB positive demonstrate reaction with the gp41
and gp120/160 env bands or with either of these bands and the p24 gag band The earlier requirement for a reaction with a third gene product (e.g p31 or p66 pol
bands) has been abandoned and reactivity with more
than one env antigen alone is enough for confirmation (O’Gorman et al 1991) If there is reactivity with only
one band, the result of the Western blot is considered
to be indeterminate The absence of reactivity in theWestern blot indicates that the donor has not devel-oped anti-HIV (Dodd 1991)
In the original Western blot assay, viral lysate wasused as a source of antigen In the newer assays, recom-binant or synthetic viral antigens are applied Theseassays have been found to be both more sensitive and
specific than the original WB (Soriano et al 1994).
Nevertheless, the assay is still relatively subjective and beset by indeterminate and false-positive resultswhen compared with NAT as the ‘gold standard’ orwhen investigated with sequential sampling follow-up
(Kleinman et al 1998; Mahe et al 2002; Stramer
2004)
Trang 6Detection of HIV DNA and HIV RNA
Donations during the window period constitute the
predominant risk for HIV transmission through
transfusion (Busch et al 2000) A more sensitive and
specific alternative to testing for p24 antigen is NAT
for HIV DNA in PBMC (Ou et al 1988) or HIV-1
RNA in plasma by reverse transcription PCR or a
similar amplification assay (Henrard et al 1992) All
blood in the USA, Japan and most European
coun-tries is tested by NAT in pools of 16 –90 specimens
Ultrasensitive assays can detect fewer than 10 genomic
copies/ml (Busch et al 2000) However, tests have
been optimized for HIV subtype B and may lack
sensi-tivity when applied to non-B subtypes (Triques et al.
1999) Single-donor assays are inevitable, but await the
development of fully automated combination (HIV/
HCV/HBV) assays
Current risk of transmission of HIV by blood
transfusion
Although the various measures outlined above have
dramatically reduced the risk of TAA-AIDS, a small
residual risk remains (Delwart et al 2004; Phelps et al.
2004) Most of this risk results from ‘window period’
donations Before the introduction of HIV-Ag and
NAT, prospective studies estimated this risk at about
one infection in 60 000 units (Busch et al 1991b;
Nelson et al 1992) Subsequent estimates rely on
models based on calculations of HIV incidence and
window period In the USA, residual risk has been
calculated as 1 per 2 135 000 repeat donors (Dodd
et al 2002) The incidence rate is approximately
two times greater among first-time donors In most
European countries where the prevalence of HIV in
blood donors is lower than in the USA, the residual
risk is probably lower still However, in countries with
a high percentage of infected subjects and where HIV
is spread mainly by heterosexual intercourse, the risk
of transmission of HIV by blood transfusion is still
considerable
Human T-cell leukaemia viruses types I
and II
The human T-cell leukaemia virus type I (HTLV-I), the
first human retrovirus to be described, was isolated
from cultured cells from a patient with an aggressive
variant of Mycosis fungoides and from a patient with Sézary syndrome (Poiesz et al 1980; Gallo et al 1981).
The virus has subsequently been shown to be identical
to the adult T-cell leukaemia virus (Yoshida et al 1982; Watanabe et al 1984) HTLV-I is the causative
agent of adult T-cell leukaemia (ATL) and is ciated with a chronic demyelinating neurological disease called tropical spastic paraperesis (TSP), known
asso-in Japan as HTLV-associated myelopathy (HAM)
(Vernant et al 1987; Roman and Osame 1988)
HTLV-seropositive individuals appear to have a 0.25% time risk of developing TSP, compared with a 2–5%
life-risk of developing ATL (Kaplan et al 1990) The
virus is also associated with lung infections, cancer ofother organs, monoclonal gammopathy, renal failure,
infection with Strongyloides stercoralis, intractable
non-specific dermatomycosis, lymphadenitis anduveitis These effects may be due to the immunodefici-ency induced by HTLV-I infection (Takatsuki 1996)
The association of HTLV-I with Mycosis fungoides is
controversial, as no HTLV-related DNA sequencescould be detected in patients with this disease
(Bazarbachi et al 1993; Vallejo et al 1995) In Japan,
only 2.5% of HTLV-I carriers develop ATL (Takatsuki1996)
HTLV-I and -II belong to the oncovirus subtype ofthe retrovirus family and are able to induce polyclonal
proliferation of T lymphocytes in vitro and in vivo.
Like the lentiviruses HIV-1 and -2, these viruses arelymphotropic and neurotropic, and have the essential
structural genes gag (group antigen), pol (reverse scriptase) and env (envelope) in addition to regulatory genes In HTLV-I, the gag gene codes for the structural proteins p55/24/19; pol codes for a protein of approx- imately 100 kDa, and env codes for glycoproteins
tran-gp61/46/21 In HTLV-II the structural proteins aresimilar to those in HTLV-I with a high degree of cross-
reactivity; gag encodes the polypeptides p53/24/19; pol a protein of approximately 100 kDa; and env
codes for the glycoproteins gp61/46/21
Areas endemic for HTLV have been found, larly in south-west Japan, with prevalence as high as
particu-15% (Maeda et al 1984), in the Caribbean with a 1–8% prevalence (Clark et al 1985), in regions of
Central and South America and in parts of
sub-Saharan Africa (Gessain et al 1986; Vrielink and
Reesink 2004) Populations in these areas show ent prevalence rates for anti-HTLV-I, as do emigrants
differ-from these regions (Sandler et al 1991; Vrielink and
Trang 7Reesink 2004) It has been estimated that more than
1 million Japanese people are healthy carriers of
HTLV-I Carriers of HTLV have also been found
in the USA, especially in Florida and states on the
Pacific, and in France, UK, the Netherlands and many
other countries The prevalence in blood donors has
been reported to be one in 6250 in the USA (CDC
1990), about one in 30 000 in France (Pillonel et al.
1996), one in 45 000 in the Netherlands (Zaaijer
et al 1994) and one in 20 000 in London (Brennan
1992)
HTLV-II, the second human retrovirus to be
dis-covered, has a 65% nucleotide sequence identity with
HTLV-I and a significant serological crossreactivity
(Hjelle 1991) However, HTLV-II antibodies are not
detected by all HTLV-I assays The distinction
between HTLV-I and -II can be made by DNA PCR
(Reesink et al 1994), and in the recently developed
WB assays in which specific HTLV-I and -II
recombin-ant recombin-antigens are used The relative prevalence of the
two viruses in blood donors in the USA was found to
be about equal (Glynn et al 2000), but in many other
countries HTLV-I predominates in blood donors
There is a high prevalence of HTLV-II among i.v drug
users and their sexual contacts in the USA and other
countries (Vrielink and Reesink 2004) A large
pro-portion of HTLV-II-positive subjects in the USA were
Hispanics and American Indians (Hjelle et al 1990a;
Sandler et al 1991) HTLV-II has been found to be
associated with a HAM-like neurological disease
(Hjelle et al 1992; Murphy et al 1997) Although
the virus was first found in a patient with hairy
cell leukaemia (Kalyanaraman et al 1982) and
sub-sequently in other T-cell malignancies, no viral RNA
could be detected in the malignant cells (Manns and
Blattner 1991)
Transmission of human T-cell leukaemia virus
HTLV is mainly transmitted by sexual contact, by the
sharing of infected needles and from mother to child,
particularly by breast-feeding (Kajiyama et al 1986).
If infected mothers refrain from breast-feeding,
trans-mission of HTLV to their infants is prevented in 80%
of cases (Hino et al 1996) Infection of infants is also
prevented when the milk is freeze-thawed or heated at
56°C for 30 min (Ando et al 1986; Hino et al 1987).
Transmission from mother to fetus has been
demon-strated by culture studies of cord blood lymphocytes in
2 out of 40 cord blood samples from HTLV-I-positive
mothers (Satow et al 1991).
Human T-cell leukaemia virus and bloodtransfusion
HTLV-I has been transmitted by cellular components,but not by cell-free plasma or plasma derivatives(Okochi 1985) However, HTLV-RNA is detectable inplasma from infected subjects The lack of infectivity
of HTLV-positive plasma may be explained by thepresence of neutralizing antibodies, the fact that it isintegrated in viral DNA and the requirement of cell–
cell interactions for infectivity (Rios et al 1996).
Antibodies are usually first detectable 14 –30 daysafter transfusion, although the interval may be as long
as 98 days (Inaba et al 1989; Gout et al 1990) Of 85
recipients of anti-HTLV-I-positive cell concentrates inJapan, 53 (62%) developed antibodies 3–6 weeks aftertransfusion: IgM antibodies were present only in theearly stages, whereas IgG antibodies persisted at hightitre throughout the period of follow-up (Sato andOkochi 1986) Storage of blood appears to decreasethe risk of transmission of HTLV This may explainthe lower rate of transmission reported in USA trans-fusion recipients, whose blood may have been storedfor a longer period than the units in Japan and the
Caribbean (Donegan et al 1994) Antibodies became
detectable in 79.2% of recipients of blood stored for1–5 days, but in only 55% of recipients of blood stored11–16 days (Okochi 1989)
Recipients of HTLV-I-infected concentrates may
develop HAM (Gout et al 1990; Araujo and Hall
2004) It has been estimated that 2–8% of subjectsinfected with HTLV-I by blood transfusion will even-
tually develop HAM (Murphy et al 1997; Araujo and
Hall 2004) ATL developed in two immunosuppressedpatients who had received multiple transfusions 6 and
11 years earlier (Chen et al 1989) HTLV-II has also been transmitted by blood transfusion (Hjelle et al.
Trang 8between HTLV-I and -II, the sensitivity of
anti-HTLV-I assays for detecting anti-HTLV-anti-HTLV-Ianti-HTLV-I was found to be
only 55–91% (Wiktor et al 1991; Cossen et al 1992).
As infection with HTLV-II is probably associated with
HAM and as many HTLV-positive donors (more than
50% in the USA) are HTLV-II positive, tests designed
to detect both antibodies are now used (US Food and
Drug Administration 1998) In these ELISAs,
recom-binant proteins, including HTLV-I and -II-specific
ones, have been added to lysate or are used exclusively
(Hartley et al 1991) The sensitivity of such assays has
been claimed to be 100% (Vrielink et al 1996a).
In Japan, an agglutination test in which gelatin
particles are coated with HTLV-I-antigens has been
developed and used extensively The sensitivity of
the commercially available agglutination test Serodia
HTLV-I has been claimed to be 100% for detecting
anti-HTLV-I; all of 12 anti-HTLV-II-containing sera
gave positive reactions (Vrielink et al 1996a) An
ELISA for the combined detection of anti-HIV-1 and
-2 and anti-HTLV-I and -II in which synthetic peptides
of all four viruses are used, has been developed
(McAlpine et al 1992) In a study 242 samples
from anti-HIV-1/-2 or I/-II panels, two
HTLV-II-positive samples and two very weak
anti-HIV-1-positive samples were negative The specificity of the
test was slightly less than that of specific assays
(Flanagan et al 1995).
Despite the improved specificity of anti-HTLV-I/-II
screening tests, many repeatedly positive reactions that
cannot be confirmed are still found and all repeatedly
reactive samples must therefore be tested in
confirm-atory assays
Human T-cell leukaemia virus confirmatory
assays
Confirmatory testing for HTLV-I/-II continues to be
challenging, primarily because of a dearth of licensed
reagents Western blot and radioimmunoprecipitation
assays (RIPAs) are used (Anderson et al 1989; Hartley
et al 1990) For a positive reaction, antibody
reactiv-ity with both a gag (p19 and /or p24) and an env
pro-tein (gp46 and /or gp68) are required (WHO 1990)
All other reaction patterns were considered to be
indeterminate As the sensitivity of the Western blot
in detecting antibodies against env proteins was low,
many indeterminate results had to be checked in RIPA,
a much more elaborate assay (Lillehoj et al 1990; Lal
et al 1992) A report on the transmission of HTLV-I
by blood from a donor with an indeterminate pattern
in the WB and RIPA (p19 and gp68 reactivity) to fourout of six recipients, confirmed by PCR, demonstratedthe insufficient sensitivity of these original confirma-
tion assays (Donegan et al 1992) A modified WB has
been developed in which both shared (r21e) and
speci-fic (rgp46Iand rgp46II) HTLV-I and -II recombinant
env proteins are used (Lillehoj et al 1990; Lal et al.
1992) For a positive reaction in this modified Western
blot, a reaction with at least one gag protein (p19 and /or p24) and with env r21e as well as rgp46Iorrgp46II is required All other patterns are indeter-minate This Western blot, in which a reaction withHTLV-I or -II can be distinguished, has been found
to be more sensitive and specific than the originalWestern blot Instead of 66, only two positive samplesrequired confirmation by RIPA and none of 158 inde-terminate samples (original Western blot) reacted
(Brodine et al 1993) A recombinant immunoblot
assay (RIBA) in which the same antigens are applied
gave similar results (Vrielink et al 1996b) NAT has
not been licensed for confirmatory testing
Cytomegalovirus
Characteristics of the virus and ofcytomegalovirus infectionCytomegalovirus (CMV) is a large, enveloped, double-stranded DNA, beta herpes virus that is cell associated,but may also be found free in plasma and other body
fluids (Drew et al 2003) CMV has a direct cytopathic
effect on infected cells The result may lead to tropenia, some depression of cellular immunity andinversion of T-cell subset ratios, with a consequentincrease in susceptibility to bacterial, fungal and protozoa infections in immunosuppressed patients
neu-(Grumet 1984; Landolfo et al 2003) CMV infection
causes parenchymal damage, such as retinitis, monitis, gastroenteritis and encephalitis, and canresult in substantial morbidity and mortality
pneu-CMV can cause primary acute clinical and ical infections Chronic subclinical infections may occur
subclin-in which the virus is shed subclin-in saliva and ursubclin-ine CMVremains latent in a large proportion of infected sub-jects The presence of anti-CMV does not guaranteeimmunity As with HIV and HCV infection, specificantibody is a marker of potential infectivity although
Trang 9only in the case of CMV, a relatively small proportion
of seropositive subjects seem to be infectious (Drew
et al 2003) CMV antibody-positive subjects may
infect others through sexual contact, breast-feeding,
transplacental transmission or transfusion (Tegtmeier
1986) In subjects with antibody, CMV infection may
be reactivated, or the subject may become re-infected
with exogenous strains of CMV
Primary CMV infection is generally more severe
than is re-infection (co-infection with a different strain)
or reactivation In view of the difficulty of
distinguish-ing between reactivation and re-infection, the term
recurrent infection has been coined to embrace both.
However, when necessary to distinguish between the
two, donor viral DNA can be distinguished from
recipi-ent viral DNA by restriction enzyme analysis (Glazer
et al 1979; Chou 1990) In practice, the diagnosis of
recurrent infection is limited to demonstrating a
four-fold increase in antibody titre or the presence of IgM
anti-CMV In immunosuppressed patients, serological
tests cannot be relied upon for a diagnosis of CMV
partly because the patients may not make antibody
and partly because any anti-CMV detected may have
been derived from transfused blood Viral culture is
impractical because the virus grows slowly in vitro
On the other hand, immunofluorescence techniques
to detect viral antigen using monoclonal antibodies on
biopsies or bronchial washings provide results within
hours (Griffiths 1984)
Prevalence of anti-CMV
The frequency of subjects with anti-CMV varies
widely in different populations Seroprevalence is lower
(30 –80%) in developed than in developing countries,
where the figure may reach 100% (Krech 1973;
Preiksaitis 1991) The prevalence of anti-CMV
corre-lates with age and socioeconomic status (Lamberson
1985; Tegtmeier 1986) The frequency of
anti-CMV-positive donors may vary widely within a given
coun-try, for example 25% in southern California and
70% in Nashville, Tennessee (Grumet 1984; Tegtmeier
1986)
Transmission of cytomegalovirus by transfusion
The transmission of CMV by blood transfusion was
first reported in the 1960s (Kaariainen et al 1966;
Paloheimo et al 1968; Klemola et al 1969) CMV is
now known as one of the infectious agents most quently transmitted by transfusion The pathogenesis
fre-of transfusion-transmitted CMV infection is notclearly understood In most cases CMV appears to
be transmitted in a latent, particulate state only by cellular blood components, and the virus reactivatesfrom donor leucocytes after transfusion Host as well
as donor factors are involved in CMV infection(Tegtmeier 1989; Preiksaitis 1991) CMV has beenisolated from the mononuclear and polymorpho-nuclear cells of patients with acute infections Thespecific cell type responsible for carrying the virus has not been identified, although mononuclear cellsare the favourite candidates as hosts of CMV in latentinfection Fresh blood appears more likely than storedblood to transmit CMV infection, although no con-trolled studies document this impression (Tegtmeier1986)
Some 3 –12% of units have the potential to mit CMV (Adler 1984), although most authors havereported a carrier rate of 1% or less (Tegtmeier 1986;
trans-Drew et al 2003) The discrepancy may be related to
the frequency of donor testing and the sensitivity andspecificity of the assay used Primary infection ratesdepend on the number of transfusions, age of blood,time of year and immunocompetence of the recipient
(Tegtmeier 1989; Preiksaitis et al 1988; Preiksaitis
2000) Donors with IgM anti-CMV appear to be more likely than others to transmit CMV (Lamberson
et al 1984) One large study found that only 0.5% of
antibody-positive donors have detectable CMV DNA
in their leucocytes (Roback et al 2003).
At present, no rapid, easy way to identify infectioussubjects exists Viral excretion in urine is a good index
of infectivity, but blood donors would probably rebel
at this screening strategy Virus can also be culturedfrom saliva, and PCR-based assays are available for the detection of viral genome in peripheral blood.Detection of pp65 antigen in leucocytes (pp65 anti-genaemia) is considered the ‘gold standard’ amongdiagnostic tools for diagnosing CMV infection and initiating antiviral therapy Both CMV DNA andimmediate early-messenger RNA detection have beencompared with pp65 antigenaemia, but none of themshowed advantages in terms of earlier diagnosis andbetter prognosis PCR’s major advantage is its semi-automation compared with the immunofluorescenceemployed for pp65 antigenaemia Isolation of CMV
by culture is reliable for the diagnosis of active
Trang 10infection, but is less sensitive and requires more time
for viral detection
Transfusion-transmitted cytomegalovirus
infection in immunocompetent subjects
Before the era of universal (or near universal)
leuco-reduction, some 30% of anti-CMV-negative recipients
undergoing cardiac surgery involving transfusion
developed infection, as confirmed by virus isolation or
the development of CMV In addition, some
anti-CMV-positive patients developed recurrent infection
In almost all cases, the infection is asymptomatic
Of patients who develop a primary or recurrent
CMV infection following transfusion, fewer than 10%
develop a mononucleosis-like syndrome This
syn-drome, originally termed the post-perfusion synsyn-drome,
but now referred to as the post-transfusion syndrome,
appears 3 – 6 weeks after transfusion Common
fea-tures include fever, exanthema, hepatosplenomegaly,
enlargement of lymph nodes and the presence in
peripheral blood of atypical lymphocytes resembling
those found in infectious mononucleosis (Foster 1966)
Recovery is usually complete The development of
atypical lymphocytes due to post-transfusion CMV
infection should be distinguished from the
develop-ment of atypical lymphocytes 1 week after transfusion
as a response to allogeneic lymphocytes
Consequences of transfusion in patients with
impaired immunity
During the past decade, major advances have been
achieved regarding the management of CMV infection
through the development of new diagnostic techniques
for the detection of the virus and through the
perform-ance of prospective clinical trials of antiviral agents
(Meijer et al 2003) Nevertheless, in
immunosup-pressed patients, or in fetuses and premature infants
with an immature immune system, CMV infections,
particularly primary infections, still cause severe
dis-ease that can be fatal
1 The fetus in utero Following maternal primary
CMV infection, the fetus becomes infected in 30 – 40%
of cases Approximately 5–10% of infected infants
develop sequelae such as mental retardation, hearing
loss or chorioretinitis (Stagno et al 1986).
2 Premature infants The risk of serious CMV
infection is high when the infant’s birthweight is less
than about 1300 g and when the mother is anti-CMVnegative In two large prospective studies, 25–30% ofinfants with these risk factors, transfused with a total
of 50 ml or more of blood, some of which was CMV positive, acquired CMV infection, and 25% ofthese infants died Infants transfused with anti-CMV-negative blood did not develop CMV infection (0 out
anti-of 90) (Yeager et al 1981; Adler et al 1983) A lower
incidence of CMV infection (7–9%) has been reportedfrom two other centres: in infants weighing less than
1500 g born to anti-CMV-negative mothers and fused with blood, some of which was anti-CMV posit-
trans-ive (Smith et al 1984; Tegtmeier 1984) All reports
agree that clinically significant CMV infection in newborn infants develops only when the infant is premature and of low birthweight, when the motherlacks anti-CMV and when anti-CMV-positive blood
is transfused (Tegtmeier 1986)
3 Bone marrow transplant recipients frequently
develop primary or recurrent CMV infections thatmay prove fatal (Tegtmeier 1986) Blood transfusionrepresents the main risk factor for CMV acquisition inCMV-negative patients receiving bone marrow from
a CMV-negative donor In a prospective randomizedtrial of 97 anti-CMV-negative patients, 57 receivedanti-CMV-negative marrow: 32 out of the 57 receivedanti-CMV-negative blood components and only one
of these developed a CMV infection; of the 25 whoreceived blood components unscreened for anti-CMV,eight developed CMV infection Among the 40 recipi-ents of anti-CMV-positive bone marrow, the rate ofCMV infection was no lower in those who receivedonly anti-CMV-negative blood components (Bowden
et al 1986) Granulocyte concentrates, which contain
large numbers of leucocytes that can harbour CMV,reportedly carry the greatest risk of transmitting CMV
infection (Winston et al 1980; Hersman et al 1982).
However, when only two prophylactic transfusionswere given, the risk appeared to be no higher in thosewho received granulocytes than in those who did not
(Vij et al 2003).
4 Renal transplant recipients are at high risk of
prim-ary or recurrent CMV infection; the main source ofinfection lies in the transplanted kidney (Tegtmeier1986) In anti-CMV-negative recipients of a kidneyfrom an anti-CMV-negative donor, blood transfusionplays a significant role in CMV transmission (Rubin
et al 1985).
5 Heart and heart–lung transplant recipients may
Trang 11develop severe primary CMV infection, which can
lead to opportunistic infections with fungi or bacteria
In anti-CMV-negative recipients the main sources of
infection are anti-CMV-positive transplanted organs
or organ blood donor units (Preiksaitis et al 1983)
If both donor and recipient are anti-CMV negative,
blood transfusion is a major source of CMV disease;
infection can be minimized by the use of
anti-CMV-negative red cells and platelets (Freeman et al 1990)
If a heart or heart–lung from a donor with CMV
anti-bodies is transplanted to an anti-CMV-negative
recipi-ent, prophylactic administration of specific CMV
hyperimmune IVIG seems to lessen the severity of
CMV disease (Freeman et al 1990), although data in
support of this practice are unimpressive
6 Following splenectomy due to trauma, patients
receiving massive transfusion may develop serious
CMV infections (Baumgartner et al 1982; Drew and
Miner 1982)
7 Subjects infected with HIV and especially those
with AIDS, if anti-CMV negative, may acquire
prim-ary CMV infection by transfusion (Jackson et al 1988;
Sayers et al 1992) In these patients CMV may cause
sight-threatening infection that may lead to blindness
in up to 25% of patients not receiving antiviral
therapy Because the rate of reactivation of CMV in
already infected patients is high, it is difficult to
deter-mine the contribution of transfusion-transmitted virus
(Bowden 1995)
8 Liver transplant recipients should also be
con-sidered at risk, especially children or pregnant women
Now that smaller amounts of blood and blood
com-ponents are needed in liver transplantation, it has been
possible to give only anti-CMV-negative blood and
platelets to negative recipients of
anti-CMV-negative grafts
Prevention of transfusion-transmitted
cytomegalovirus infection
Subjects who are at highest risk of severe primary
CMV infection are anti-CMV-negative patients with
impaired immunity For such patients, preventive
measures are available to reduce transmission of CMV
by transfusion The selection of CMV-seronegative
donors has proven to be effective, but not infallible
(Tegtmeier 1989; Miller et al 1991) Seropositivity,
especially the presence of IgM, is a marker of previous
infection and latent, but potentially infectious virus
However, antibody assays vary in sensitivity and asmall risk of transmission even from seronegative units
remains (Kraat et al 1993; Bowden et al 1995).
Window period infections are the most likely source ofantibody screening failures Although the windowperiods for HIV-1, HCV and HBV have been reason-ably well defined, the length of the CMV-seronegativewindow, estimated at 6–8 weeks, is less well character-ized Several recent studies using PCR technology documented CMV DNA in both plasma and cellularblood components from several weeks before sero-conversion to several months after seroconversion,although culture positivity was found for a muchshorter period in white blood cells (WBCs) and not
observed in plasma (Zanghellini et al 1999).
When only red cells are required, frozen lysed red cells can be used and have not been shown
deglycero-not to transmit CMV (Brady et al 1984; Taylor et al 1986; Sayers et al 1992).
As CMV is a white cell-associated virus, an ative approach to preventing CMV transmissioninvolves filtration removal of leucocytes from red cell
altern-and platelet concentrates (Graan-Hentzen et al 1989).
Leucoreduction by filtration may fail to prevent CMVtransmission, as 105to 106WBCs may still be trans-fused and an estimated 1 in 1000 to 1 in 10 000 WBCs
are infected by CMV during latency (Drew et al 2003).
In 10 patients whose WBCs were CMV antigen andculture positive before filtration and culture negativeafterwards, 2 out of the 10 did, however, have CMVDNA detected in leucocytes after filtration (Lipson
et al 2001) Plasma viraemia, if present, would not be
diminished by leucoreduction and might also accountfor CMV transmission following leucoreduced com-ponents Although the exact number of residual leuco-cytes that is sufficiently small to pose no risk of CMVtransmission is unknown and may not exist, a largeprospective study has shown that leucocyte removal
is as safe as selection of anti-CMV-negative donors
In this study, 502 patients were randomized prior
to bone marrow transplantation to receive filtered,three-log leucocyte-depleted cellular components orcomponents from anti-CMV-negative donors Therewas no significant difference in the probability ofCMV infection between the recipients of anti-CMV-negative or filtered concentrates, although more CMV-associated disease was observed in the filtration group (Bowden 1995) The same investigators havesubsequently questioned their finding that filtered red
Trang 12cells are equivalent to CMV-seronegative cells (Nichols
et al 2003) When granulocyte transfusions are needed,
selection of anti-CMV-negative donors is the only
solution
The success of leucocyte removal in the prevention
of CMV transmission has raised the question of the
importance of infection with a second strain of CMV
in anti-CMV-positive recipients and the value of
trans-fusing such recipients with leucocyte-depleted
concen-trates Although co-infection with a second strain of
CMV does occur (Boppana et al 2001), the clinical
consequences of such infection resulting from
trans-fusion are less clear
Tests for anti-CMV
Several tests for anti-CMV are available Complement
fixation used to be the diagnostic reference test, but it
proved too complicated for routine donor screening
Indirect immunofluorescence, solid-phase fluorescence,
ELISA and particle agglutination assays are also
avail-able Competitive ELISAs seem to be the most reliable
of the currently available screening tests (Bowden et al.
1987) CMV PCR technology has been used in clinical
diagnostics since the 1980s (Bowen et al 1997; Lipson
et al 1998) Conventional PCR requires detection and
confirmatory testing of the amplicon by gel
elec-trophoresis, incorporating an isotopically labelled or
non-radioactive hybridization assay or a nested PCR
to detect targets present in very low copy numbers
(Lipson et al 1995) A simplified, more rapid
PCR-solid-phase enzyme immunoassay (EIA) plate
tech-nology system has been developed (Davoli et al 1999).
The quantitative real-time CMV PCR assay using
TaqMan chemistry and an automated sample
prepara-tion system has also been applied to CMV detecprepara-tion
(Piiparinen et al 2004).
An inexpensive, uncomplicated CMV-Ag (pp65
antigen) assay is available and well suited for most
diagnostic microbiology laboratories (Lipson et al.
1998) Assay for the early antigen pp65 is considered
the ‘gold standard’ for the initiation of antiviral therapy
Other viruses
Epstein–Barr virus
Infection with Epstein–Barr virus (EBV), a herpes
virus-like CMV, is endemic throughout the world
EBV can cause primary symptomatic infection tious mononucleosis), but most commonly causesasymptomatic infection followed by latent infection(Henle 1985) In most countries more than 90% ofblood donors have neutralizing anti-EBV, which coex-ists with latent virus in B lymphocytes of peripheralblood and lymph nodes At least one in 107circulatinglymphocytes of carriers harbour EBV genomes, butpost-transfusion EBV infection is a rare occurrence,
(infec-and symptomatic infection is even rarer (Rocchi et al.
1977) The virus is found in three of every 104eral lymphocytes during acute infection The majority
periph-of susceptible recipients are young children Even inchildren, the chance of acquiring EBV infection fol-lowing the transfusion of anti-EBV-positive blood isminimal, because the donor’s neutralizing antibodiespersist in the recipient’s circulation long after the EBV-infected lymphocytes disappear In a study of 25 EBVantibody-negative patients aged 3 months to 15 yearsand transfused with 1–11 units of blood stored for notmore than 4 days, only one developed EBV antibodiesand this patient had no symptoms (Henle 1985)
Of five patients transfused during cardiac bypass,four who were initially anti-EBV negative producedantibody that persisted at high titre for many monthspostoperatively Two out of the four patients had concomitant CMV infection and no heterophile anti-bodies; one developed transient fever, and the otherhepatitis Only one of the four patients developed aninfectious mononucleosis-like syndrome with hetero-
phile antibodies (Gerber et al 1969).
Although most cases of ‘post-transfusion syndrome’are caused by CMV, two adult patients who were anti-EBV negative before transfusion developed post-
transfusion syndrome due to EBV (McMonigal et al.
1983) In a survey of some 800 patients, fewer than8% lacked EBV antibody before transfusion, and only5% of these developed antibodies following trans-fusion These patients suffered no clinical illness or disturbance of liver function (MRC 1974) Possibly,the discrepancy between these observations and those
of others may have been related to the use of fresh
blood in the last-named series (Gerber et al 1969).
Post-transfusion infectious mononucleosis is seenonly rarely in anti-EBV-negative immunocompetentpatients and usually occurs when only a single unit ofblood or blood component, obtained from the donorduring the incubation phase is given within 4 days ofcollection When more than 1 unit is transfused, one
Trang 13of the units is almost certain to contain anti-EBV
In the reported cases, the donors developed symptoms
of mononucleosis 2–17 days after blood donation
The incubation period in recipients has been 21–
30 days (Solem and Jorgensen 1969; Turner et al.
1972) Transfusion-transmitted EBV infection with
symptomatic mononucleosis has occasionally been
reported in patients transfused for splenectomy (Henle
1985)
Post-transfusion EBV infections may contribute to
the development of lymphomas in severely
immuno-suppressed patients such as haematopoietic graft
recipients T-lymphocyte suppression allows
EBV-infected B lymphocytes to outlive the passively
trans-fused antibodies and to proliferate (Marker et al.
1979; Hanto et al 1983).
Other herpes viruses
Herpes simplex and Herpes varicella zoster have never
been shown to be transmitted by blood transfusion;
viraemia occurs only during primary infections, which
usually occur in childhood (Henle 1985)
HHV-6 is a recently characterized human herpes
virus, originally named HBLV (human B lymphotropic
virus) for its ability to infect freshly isolated B cells
The virus was found in patients with various
lympho-proliferative disorders HHV-6 can infect monocytes,
macrophages, T cells and megakaryocytes (Ablashi
1987) The virus is cytopathic for selected T-cell lines
Infection is acquired usually within the first year of life
and the virus was found to be ubiquitous in blood
donors when tested in London and the USA (Briggs
et al 1988; Saxinger et al 1988) No
transfusion-associated disease has been reported
HHV-8, Kaposi’s sarcoma-herpes virus is white cell
associated and may be present in up to 30% of normal
donors Despite a relatively high HHV-8
seropre-valence in a Texas blood donor cohort (23%), HHV-8
DNA was not detected in any sample of donor whole
blood using a highly sensitive PCR assay (Hudnall
et al 2003) Blood components from HHV 8-infected
donors apparently carry little transfusion risk (Engels
et al 1999).
Human parvovirus B19
HPV B19 infection has long been known to cause
erythema infectiosum (Fifth disease), a common febrile
exanthem of childhood (Anderson et al 1983) HPV is
also associated with polyarthritis and rash in adults as
a result of antigen–antibody immune complex
deposi-tion in skin and joints (Reid et al 1985; White et al.
1985) Because of its specific cytotoxic effect on erythroblasts, HPV B19 can precipitate aplastic crises
in children who have haematological disorders withshortened red cell survival, such as those with sicklecell anaemia and other chronic haemolytic anaemias,
particularly hereditary spherocytosis (Pattison et al.
1981; Young and Mortimer 1984) The virus may alsocause thrombocytopenic purpura (Pattison 1987).Intrauterine infection may cause hydrops fetalis and
spontaneous abortion in early pregnancy (Brown et al 1984; Anand et al 1987).
HPV B19 was discovered by an Australian virologistwho noted viral particles in an antigen–antibody line
of detection (plate B, well 19) in an assay for hepatitis
B (Cossart et al 1975) HPV B19 is a small
single-stranded, non-enveloped, thermostable DNA member
of the Parvoviridae family (Shade et al 1986; Young
and Brown 2004) The parvoviruses are dependent
on help from host cells or other viruses to replicate.Parvovirus B19 is the type member of the erythrovirusgenus, which propagates best in erythroid progenitorcells The red cell P antigen, a globoside present on avariety of cells in addition to erythrocytes, has beendocumented as the specific receptor for HPV (Brown
et al 1994) This may account for reports of
poly-arthritis nephropathy, myocarditis and pathy (Young and Brown 2004) Persistent infectionwith anaemia occurs in immunosuppressed subjects
cardiomyo-(Kurtzman et al 1987) PCR assays have revealed the
presence of viral DNA along with the simultaneouspresence of specific IgG in 0.55–1.3% of normal blood
donors (Candotti et al 2004), but the long-term
persistence, infectivity and clinical importance of thevirus in these subjects has not been well studied.Current methods of viral inactivation may not be able
to eliminate HPV B19 completely (Williams et al.
1990a)
Infection with the virus normally occurs via atory droplets HPV B19 has been transmitted byplasma fractionation products derived from largepools of plasma, particularly by factor VIII and factor
respir-IX concentrates (Santagostino et al 1997; Blumel et al.
2002) One episode of fulminant hepatitis has beenattributed to the intravenous route of infection
(Hayakawa et al 2002) Use of recombinant products
Trang 14should eliminate this risk (Soucie et al 2004).
Transmission of the virus by single-donor components
is unusual, but a red cell unit has been associated with
HPV B19 transmission and possible cardiac
involve-ment in a 22-year-old woman with thalassaemia major
(Zanella et al 1995).
The classic diagnosis of infection with HPV B19
is based on the detection of IgM or IgG antibodies
with an EIA (Cohen et al 1983) Alternatively viral
DNA can be detected by PCR (Salimans et al 1989;
McOmish et al 1993) HPV B19 has been detected by
PCR in solvent–detergent-treated clotting factor
con-centrates (Lefrere et al 1994), in heat-treated factor
VIII and IX concentrates and in IVIG (McOmish et al.
1993; Santagostino et al 1994) and in clotting factor
concentrates prepared by different purification and
inactivation procedures (Zakrzewska et al 1992;
Blumel et al 2002) The virus has also been detected in
plasma pools designed for fractionation (in 64 out of
75 pools), in 3 out of 12 albumin preparations, in 3 out
of 15 IVIG and in three out of four IMIG preparations,
as well as in seven out of seven factor VIII
prepara-tions There is some indication that treatment of
IVIG preparations at low pH may result in removal
of detectable HPV B19 DNA (Saldanha and Minor
1996) Since 2002, major plasma fractionators have
screened plasma units with quantitative measurements
of B19 DNA to reduce the risk of transmission The
prevalence of HPV B19 in blood donors has been
estimated at 0.03% in the UK (McOmish et al 1993).
A similar prevalence was found in Japan (1 in 35 000)
but, during an epidemic of erythema infectiosum, the
prevalence was much higher (1 in 4000) (Tsujimura
et al 1995) B19 prevalence varies according to season
and from year to year (Young and Brown 2004)
A rapid test for HPV B19, suitable for large-scale
screening of donors has been developed (Sato et al.
1996) The test is based on agglutination of blood
group P-positive gluteraldehyde-treated red cells by
the virus for which the P antigen is the receptor (see
Chapter 4) Although only intact viruses can bind to P,
the test has been found to be sensitive and it could be
used to select donors for patients at risk Whether
rou-tine screening of donors is necessary is an unanswered
question Although 81.6% of 136 haemophiliacs
studied had anti-HPV B19, B19 DNA was detectable
in none and there were no signs of lasting clinical or
haematological sequelae (Ragni et al 1996).
Commercial immune globulins are a good source of
antibodies against parvovirus; a persistent B19 tion responds to a 5- or 10-day course of immuno-globulin at a dose of 0.4 g per kilogram of bodyweight, with a prompt decline in serum viral DNA, asmeasured by hybridization methods, accompanied
infec-by reticulocytosis and increased haemoglobin levels
(Kurtzman et al 1987; Frickhofen et al 1990).
West Nile VirusSince its importation into the USA in 1999, West Nilevirus (WNV) has become a significant transfusion-transmitted infection with a calculated mean risk oftransfusion transmission of 3.02 per 10 000 donations
in high-risk metropolitan areas during epidemic tions (Biggerstaff and Petersen 2003) Transfusiontransmission was first documented when four organsharvested from a common multi-transfused cadaverdonor transmitted virus to all four recipients; thedonor’s pretransfusion sample tested negative forWNV, while one of 63 blood donors tested positivewith a nucleic acid assay and developed IgM antibody
condi-to WNV over the subsequent 2 months (Iwamocondi-to et al.
2003) Using strict case definition criteria, logists document 23 transmissions and another 19inconclusive investigations of 61 case investigations
epidemio-during a 12-month period (Pealer et al 2003) The true
number of transmissions was probably higher by atleast an order of magnitude
As was the case with HIV and the hepatitis viruses,blood transfusion of WNV represents a small, buthighly visible portion of a large epidemic WNV is amosquito-borne flavivirus transmitted primarily tobirds and some small mammals Humans serve as anincidental host Approximately 80% of human infec-tions are asymptomatic; 20% result in a febrile illnessknown as West Nile fever About 1 in 150 patientsdevelop meningoencephalitis and residual neurolo-gical deficits have been reported Although there is noparticular susceptibility to mosquito-borne infection,elderly and immunosuppressed subjects appear par-ticularly vulnerable to severe, progressive disease The virus is present for a week or more during initialinfection Most symptomatic subjects describe fever,headache and malaise, although these are not suffi-ciently specific for effective donor screening Viralshedding may persist for 7–8 weeks after infection,usually in the presence of specific antibody The period
of infectivity has not been well defined
Trang 15Of the 23 well-studied patients with
transfusion-transmitted infection, 14 were identified because West
Nile virus-associated illness came to the clinician’s
attention following transfusion Overall, 15 patients
had recognized illness (13 meningoencephalitis, two
fever) The illness began between 2 and 21 days after
the implicated transfusion The highly
immunosup-pressed transplant recipients appeared to have the
longest incubation periods (median 13.5 days) Red
cells, platelets and fresh-frozen plasma (FFP) have
all been implicated in transmissions Sixteen blood
donors were implicated in transmission, with no
pre-dilection for age or gender Nine out of the sixteen
recalled symptoms compatible with a viral illness
around the time of donation, although all donors
passed the donor screening procedures Three donors
developed symptoms prior to donation, one on the day
of donation and five post donation Fever, headache
and weakness were the most common symptoms
Samples obtained at the time of donation had virus
lev-els less than 80 pfu per millilitre and all were negative
for West Nile virus IgM antibody
Since screening of blood was undertaken in the USA,
approximately 6 million units were tested during the
June–December 2003 interval, resulting in the removal
of at least 818 viraemic blood donations from the
blood supply Nevertheless, six cases of WNV
trans-mitted by transfusion occurred because of transfusion
of components containing low levels of virus not
detected by the testing of pooled specimens (Macedo
et al 2004) All blood donations in the USA are
cur-rently screened by rtPCR, and it is likely that testing of
individual donations will begin in areas of epidemic
transmission and eventually be practised universally
(Custer et al 2004).
Other flaviviruses such as St Louis encephalitis
virus, Japanese encephalitis virus and dengue virus
are likely to be blood transmissible as well, although
documentation is lacking
Simian foamy virus
Simian foamy virus (SFV) (spumaretrovirus) is a highly
prevalent primate retrovirus that has been shown to
infect human cells, replicate and produce cell-free
infectious virus Tropism is broad and includes B and
T lymphocytes, macrophages, fibroblasts, endothelial
cells and kidney cells Simian retroviruses are spread
in areas of the world such as Central Africa, where
non-human primates are hunted for food and amongpeople who handle non-human primates as pets or laboratory animals In total, 1% of people living insouthern Cameroon were found to harbour antibodies
to SFV; all had reported exposure to fresh primate
blood or body fluids (Wolfe et al 2004).
Studies from the USA CDC report a prevalence ofinfection of 2–5% with simian foamy viruses amonglaboratory and zoo workers occupationally exposed
to non-human primates (Heneine et al 1998; Switzer
et al 2004) Persistent viraemia has been detected
in peripheral blood lymphocytes (PBLs) and 11 infected workers are known to have donated blood.Six donors were confirmed positive at the time ofdonation SFV transmission through transfusion wasnot identified among four recipients of cellular bloodcomponents (two RBC, one filtered RBC, one plateletconcentrate) from one SFV-infected donor (Boneva
SFV-et al 2002) Derivatives containing plasma from that
donor tested negative for SFV
Evidence of SFV infection included seropositivity,proviral DNA detection and isolation of foamy virus.There is no evidence as yet that SFV causes human dis-ease but, recombination within the host, especially inimmunocompromised hosts that may allow persistentinfection, remains a concern
Prion proteins
A fundamental feature of prion diseases involves anormal protein constituent of human tissue, prion
protein (PrP) (Bendheim et al 1992) PrP can exist
in either a natural cellular form (PrPC) or as variantpathological forms known collectively as ‘scrapie isoforms’ or PrPSc Different prion strains result in distinct clinical and pathological diseases (Prusiner1998) Both forms of prion protein have an identicalamino acid sequence, but differ in the secondary struc-ture, beta-pleated structures in the variant formsinstead of the alpha-helical structure of the native pro-
tein (Pan et al 1993) The structural difference has two
major consequences First, the beta-pleated tion allows PrPSc molecules to form protease-resistantaggregates Second, the abnormal protein seems to becapable of suborning normal protein to the abnormalform The molecules with beta-pleated regions causekey alpha-helical regions of the native protein toassume the pleated conformation, thus converting normal protein to abnormal aggregates that cause the
Trang 16conforma-spongiform degeneration of the brain that is
character-istic of these diseases
Prion proteins are linked to the cell plasma
mem-brane by the lipid glycophosphatidyl inositol (GPI)
GPI-linked proteins are released from cells by several
different mechanisms and may be taken up by other
cells, thus facilitating dissemination (Devetten et al.
1997) Prion proteins are taken up selectively by
motile follicular dendritic cells, suggesting that
infec-tive proteins may be present in circulating blood (Klein
et al 2001).
Creutzfeldt–Jakob disease
Creutzfeldt–Jakob disease (CJD) is a rare, but
invari-ably fatal neurodegenerative disease, with an
incid-ence of about one per million of the UK and USA
population (Collinge and Rossor 1996) CJD may be
‘sporadic,’ arising from a random change in a normal
individual, ‘familial,’ arising from a point mutation
in the DNA coding for the protein, which leads to an
increased susceptibility to production of the abnormal
form, or ‘acquired,’ by the transfer of infectious
mater-ial, as in individuals injected with pituitary-derived
hormones or treated with dura mater transplants
(Brown et al 1992) A single case of transmission
from a corneal transplant has also been reported The
proportion of recipients acquiring CJD from growth
hormone varies from 0.3% to 4.4% in different
coun-tries, and acquisition from dura mater varies between
0.02% and 0.05% in Japan, where most cases occurred
(Brown et al 2000) Patient follow-up after point
exposures to contaminated materials indicate that
clinical latency for iatrogenic CJD may exceed 30 years
(Fradkin et al 1991; Brown et al 2000).
Classic CJD typically affects older subjects with
a median age of 60 years Memory loss is an early
manifestation, but the disease progresses rapidly after
the first symptoms through confusion, motor and
cere-bellar symptoms and dementia Death often occurs
within a year of the first symptom
Because of the possibility of transfer of infectious
prions by transfusion, donors have been questioned
to discover (1) whether there is any family history of
CJD; (2) whether they have received pituitary-derived
hormones; or (3) in some countries, whether they have
received corneal grafts or grafts of dura mater
How-ever, a systematic review of five case–control studies
from the UK, Europe, Japan and Australia, involving
2479 patients, failed to find an association betweenCJD and blood recipients No evidence of transmis-sion of CJD by blood transfusion exists, despite theidentification of individuals who were exposed toblood donated by people who later developed this dis-
ease (Wilson et al 2000) A study of preserved brain
samples of 25 haemophilic patients who have highexposure to blood transfusions and potentially higherexposure to blood infected with the agent responsiblefor Creutzfeldt–Jakob disease found no evidence of
the disease (Evatt et al 1998) ‘Look-back’ studies
have not identified any cases of Creutzfeldt–Jakobdisease developing in recipients who received bloodfrom a donor in whom the disease was later diagnosed.The risk of CJD transmission through transfusion isnow considered negligible, and plasma pools are nolonger discarded if a contributing donor developsCJD Deferrals designed to eliminate ‘high-risk’ donorcategories, such as growth hormone and dura materrecipients, remain in force
Variant Creutzfeldt–Jakob disease
A neurodegenerative disease affecting cattle was firstidentified in England in 1984 and subsequently branded
‘mad cow disease’ Bovine spongiform encephalopathy(BSE) is now recognized as a prion-related disorderwith more than 179 000 confirmed clinical cases ofBSE, driven by the recycling of infection through theinclusion of bovine protein in cattle feed A ban onfeeding tissues from one ruminant animal to otherruminant animals, introduced in 1988, brought theBritish BSE epidemic under control However, because
of the long delay between infection and the onset
of clinical signs of disease (5 years on average), theannual incidence of clinical cases did not peak until
1992 BSE has been found in several other Europeancountries and in Canada More rapid and accuratedescription of the epidemiology of BSE has been hampered by the absence of a diagnostic test that can be applied to live animals to detect those that areincubating the infection
The human disease equivalent, new variantCreutzfeldt–Jakob disease (vCJD), was not identifieduntil 1996, 7 years after the ban was introduced A disease-related isoform of prion resembling that ofBSE is consistent with the conclusion that BSE hascrossed the species barrier, probably from ingestion of
infected beef (Collinge and Rossor 1996; Bruce et al.
Trang 171997) Unlike classic CJD, vCJD usually affects
younger subjects (age 18–53 years) and is
charac-terized by early psychiatric and sensory symptoms
The disease progresses over 8 months to 3 years, with
progressive dementia, ataxia and myoclonus Brain
biopsy has a characteristic pattern and Western blot
analysis shows a diagnostic migration pattern of
pro-teinase-treated PrPSc Pathological PrPSc can be found
in tonsils, spleen lymph nodes and appendix (Collinge
et al 1996) Prion protein gene analysis has shown
that all cases were homozygous for methionine at
codon 129 (Ironside and Head 2004) More than 150
cases have been found in the UK and cases have been
described in France and Italy Although the number
of vCJD ‘carriers’ remains unknown, early estimates
of as many as 100 000 cases have in recent years
dwindled to a maximum of just a few hundred cases,
assuming an estimate of 15 to 20 years as the average
incubation period (Valleron et al 2001) One model
indicates that the primary epidemic in the susceptible
genotype (methionine-methionine (MM)-homozygous
at codon 129 of the prion protein gene) has now
peaked, with an estimate of 40 future deaths (Ghani
et al 2003) However, the prevalence of vCJD
infec-tion in the UK populainfec-tion, estimated to be as high as
1 per 4000 based on a study of routinely acquired
tonsils and appendices, raises the possibility of a
second and third ‘wave’ of clinical cases in subjects
heterozygous (methionine-valine) and homozygous
for valine at codon 129
Prion protein has been identified on a wide range
of circulating cells including platelets, myeloid cells,
lymphocytes and red cells (Dodelet and Cashman
1998; Holada and Vostal 2000; Bessos et al 2001;
Li et al 2001) Findings in experimental models show
that blood not only contains infective agents of prion
diseases, but that no barrier to transmission exists with
intraspecies transmission, and that the intravenous
route of exposure to prions is fairly efficient (Casaccia
et al 1989; Cervenakova et al 2003) The seminal
experiments in sheep have shown transmission of BSE
and scrapie by blood transfusion, and blood for
trans-fusion in these experiments was obtained from sheep
midway through the incubation period (Houston et al.
2000; Hunter et al 2002) Infectivity has also been
noted in the incubation period and symptomatic phase
in a rodent model of vCJD (Cervenakova et al 2003).
In 1997, the UK set up a surveillance system
between the national CJD surveillance unit and the UK
national blood services In 2004, the first probabletransfusion-associated case of vCJD was described
(Llewelyn et al 2004):
A 62-year-old man received 5 units of red cells for a surgical procedure Six and a half years later, the patient developed irritability and depression, followed by a shuffling gait, blurred vision, motor dysfunction and cognitive impairment Thirteen months after the onset of symptoms, he died of autopsy-confirmed vCJD One of the red cell units had been donated by a 24-year-old subject who developed symptoms
of vCJD 3 years and 4 months later, and subsequently died of vCJD confirmed at autopsy Red cells from a second donation were traced to a patient who died of cancer 5 months later Platelets from the donation could not be traced to a recipient The clinical presentation of the transfusion recipient was typical of vCJD, and diagnosis was confirmed by neuropatho- logical examination Transfusion took place before universal leucoreduction Of particular note in this case is the age of the recipient, the second oldest patient with vCJD, making it even less likely that this case was caused by ingestion of tainted beef.
Statistical analysis, taking account reported vCJDmortality to date and details of the recipients of vCJDdonations indicate that the probability of recording acase of vCJD in this population in the absence of trans-fusion-transmitted infection ranges between about 1 in
15 000 and 1 in 30 000 (Llewelyn et al 2004).
A case of preclinical vCJD has been reported in apatient who died from a non-neurological disorder
5 years after receiving a blood transfusion from adonor who subsequently developed vCJD:
In 1999, an elderly patient received a unit of non-leucodepleted red blood cells from a donor who developed symptoms of vCJD 18 months after donation The donor died in 2001 and vCJD was confirmed at autopsy The recipient, who had no evidence of a neurological disorder, died 5 years after receiv- ing the transfusion Western blot analysis of splenic tissue showed the presence of PrP res with the mobility and glyco- form ratio of the signals similar to those seen in spleen from patients with clinical vCJD and distinct from those of sporadic CJD cases.
Protease-resistant prion protein (PrPres) was detected
by Western blot, paraffin-embedded tissue blot andimmunohistochemistry in splenic tissue, but not in the brain However, animal studies suggest that migra-tion is inevitable given sufficient time Prion proteinwas also present in a cervical lymph node Perhaps the most disturbing observation is that this patient was
Trang 18a heterozygote at codon 129 of prion protein gene
(PRNP), suggesting that susceptibility to vCJD
infec-tion is not confined to the methionine homozygous
genotype This finding suggests that a second, later
wave of vCJD cases in heterozygotes and even a third
may appear long after the initial peak of the epidemic
(Peden et al 2004) The chance of observing vCJD
transmission in the absence of a transfusion infection
in this second recipient of blood from a donor with
vCJD is far less than the 1 in 15 000 to 1 in 30 000
chance for the first reported case
Transmissable spongiform encephalopathy (TSE)
agents are resistant to the range of physical and
chem-ical means that have been used to inactivate viruses in
plasma products However, a range of studies using
animal TSE models have demonstrated that the
pro-cesses used to purify proteins, including factor
concen-trates, over the course of plasma fractionation can also
contribute significantly to removing both abnormal
prions and infectivity (Lee et al 2001) Extensive
washing of cellular components may also deplete the
prion content; however, the risk of infection has not
been determined A prototype prion removal filter has
been designed to remove about four logs of PrP, below
the level of detection in the Western blot assay, and
has prevented infection in a hamster scrapie model
(SO Sowemino-Coker, personal communication)
Bacteria
Transmission of bacteria represents the most frequent
infectious complication of blood transfusion in the
developed world and a major cause of
transfusion-associated mortality (Andreu et al 2002) The reported
frequency of contamination varies depending upon
the nature of the blood component and the method
of study technique
Treponema pallidum
Treponema pallidum is a motile spirochaete that
spreads by sexual contact, transfusion, percutaneous
exposure and transmission from mother to infant The
incubation period from transfusion to clinical
pres-entation varies from 4 weeks to 4.5 months, averaging
9 –10 weeks, and the infected recipient usually exhibits
a typical secondary eruption Donors at any stage of
disease, including late, latent syphilis, can transmit the
infection (Hartmann and Schone 1942)
Transfusion-transmitted syphilis was once ered a serious problem Kilduffe and DeBakey (1942)identified more than 100 cases that had been pub-lished after 1915, all from direct transfusion Some
consid-138 cases had been reported by 1941 (De Schryver and Meheus 1990) Since then, few cases have beenreported in the developed world The last case pub-lished in the USA was reported from the ClinicalCenter at the National Institutes of Health (Chambers
et al 1969).
In 1966, a patient was admitted to the Clinical Center with a diagnosis of lymphoma His serological test for syphilis (STS) was negative on admission He received 5 units of RBCs and
25 units of fresh platelets, none of which reacted in the VDRL test Two months later, he developed a maculopapular rash consistent with secondary syphilis Serological tests and con- firmatory assays for syphilis turned positive The patient was treated with penicillin and the rash cleared.
Of the 30 donors, 27 tested negative repeatedly, but two could not be traced and one refused to be retested The investigators presumed that one out of those three donors, a platelet donor, was infectious for syphilis.
The chief reasons for the decline of transmitted syphilis seem to be the almost universalpractice of storing blood at 4°C before transfusion,universal donor testing and the decline in the preval-ence of syphilis in many countries since the advent
transfusion-of penicillin Other factors that have probably tributed to the decline include the administration ofantibiotics to a large proportion of patients requiringtransfusion and the exclusion of donors with high-risksexual practices
con-Spirochaetes are unlikely to survive in citrated bloodstored for more than 72 h at 4 – 6°C (Bloch 1941;Turner and Diseker 1941) However, organisms can
be detected for as long as 6 days (Selbie 1943) There
is a close relationship between the number of ponemes added to donor blood and the survival times
tre-of T pallidum determined by a sensitive assay in
rab-bits; in blood heavily contaminated with spirochaetes(1.3 × 106 and 2.5 × 107/ml blood), surviving tre-
ponema were found at 120 h (Garretta et al 1977; van der Sluis et al 1985).
Several tests are available for screening blood tions for syphilis, including the automated reagin test(ART), the rapid plasma reagin test (PRP) and theVenereal Disease Research Laboratory (VDRL) slidetechnique in which non-specific reagins (antibodies)
Trang 19dona-are detected and the T pallidum haemagglutination
assay (TPHA), the fluorescent treponemal antibodies
absorbed with Reiter’s treponeme assay (FTA-Abs)
and ELISAs for the detection of specific antibodies
Non-specific reagin tests detect antibody to lipoidal
antigen and are often considered to be insufficiently
sensitive as screening tests (Barbara et al 1982; Young
et al 1992) Positive screening tests should be tested
for FTA-Abs or some other specific assay in a reference
laboratory A lack of demonstrable T pallidum DNA
or RNA suggests that blood donors with confirmed
positive results in STS are unlikely to have circulating
T pallidum in their blood, and that that their blood
is unlikely to be infectious for syphilis (Orton et al.
2002)
Serological tests cannot prevent all cases of
trans-fusion syphilis because most remain negative in early
primary syphilis, when spirochaetaemia is most
prom-inent (Spangler et al 1964).
The rationale for continued syphilis testing relies
upon the increasing demands for fresh blood
compon-ents, especially platelets and fresh blood for exchange
transfusion in newborn infants, the very occasional
reported case (Chambers et al 1969; Soendjojo et al.
1982; Risseeuw-Appel and Kothe 1983) and its
ques-tionable value as a surrogate test to exclude donors
who are in high-risk groups for HIV and HBV
infec-tion A positive test in a transfusion recipient may
result from antibody acquired passively from the
donor (Rossi et al 2002) However, passively acquired
antibody rarely remains detectable for more than a few
months after transfusion (Ravitch et al 1949; Rossi
et al 2002).
In countries with a high incidence of syphilis, some
consultants recommend that recipients of fresh blood
receive 2 megaunits of penicillin G or its equivalent
(Bruce-Chwatt 1985) In endemic areas, subjects who
have had non-venereal trepanomatosis, such as yaws
or pima caused by T pallidum pertenue or T pallidum
carateum, may also have a positive screening test for
syphilis
Brucella abortus
This organism can survive for months in stored blood
and there are several reports of blood
transfusion-transmitted symptomatic infection, mainly in children
and splenectomized patients (Wood 1955; Tabor 1982)
Antibody-positive donors are common in Mexico,
Greece, Spain and in some rural areas of the USA,although transfusion-transmitted brucellosis has notbeen reported in the USA Infected donor blood hasvery low concentrations of brucella and poses littlerisk except for immunosuppressed patients (Fernandez
et al 1981).
After an incubation period ranging from 6 days to
4 months, recipients of infected blood may developundulant fever, headache, chills, excessive sweating,muscle pains and fatigue Hepatosplenomegaly, lym-phadenopathy, leucopenia and arthritis occur and, veryrarely, complications such as purpura, encephalitis orendocarditis develop (Tabor 1982)
In view of the chronic nature of the disease, personswith a history of brucellosis should not be used asdonors However, 80% of infections are asymp-tomatic Even in endemic areas screening tests are not practical; most subjects with high-titre brucellaantibodies do not transmit infection by transfusion
(Fernandez et al 1981).
Miscellaneous
Lyme disease The organism responsible for this disease is Borrelia burgdorferi, a tick-borne spirochaete whose primary
reservoir is the white-footed mouse Humans areinfected in the nymphal stage of the cycle and white-tailed deer are infected during the second year of life
of the tick Most cases of Lyme disease have beenreported in the USA, but thousands of cases have alsobeen reported in Europe
The disease has three stages: in the acute stage oferythema migrans, a skin rash starting at the site of the tick bite spreads locally and is often accompanied
by ‘flu-like symptoms’ If this stage is not treatedpromptly with antibiotics, the second stage progresses
to disseminated infection with cardiovascular and neurological manifestations In the third stage, arthritis develops
Although no cases of transmission of B burgdorferi
by blood transfusion have been reported so far, mission is theoretically possible Subjects in the primarystage of infection pose the most risk However, as thesepatients are generally ill and as spirochaetaemia is oflow intensity and of short duration, infectious donorsprobably do not present often for blood donation(Popovsky 1990; Westphal 1991a) The spirochaete
Trang 20trans-has been isolated and cultured from blood as old as
14 days
Four transfusion recipients of blood components
from a donor who gave blood between the
disappear-ance of erythema migrans and the second stage of
the disease have been studied; none of the recipients
showed signs of disease or developed antibodies against
B burgdorferi (Halkier et al 1990).
Mycobacterium leprae
Mycobacterium leprae is known to have been
trans-fused inadvertently from a donor incubating leprosy to
the two recipients without apparent harm M leprae
has also been injected intravenously into human
volunteers without the recipients becoming infected,
although the period of follow-up is not known (Tabor
1982)
Tick-borne rickettsial disease
Tick-borne rickettsial diseases are caused by two
groups of intracellular bacteria belonging to the order
Rickettsiales: (1) bacteria belonging to the spotted
fever group of the genus Rickettsia within the family
Rickettsiaceae; and (2) bacteria within the family
Anaplasmataceae, including several genera, such as
Anaplasma and Ehrlichia Rickettsiosis (Rocky
Mountain spotted fever) has been reported to have
been transmitted by blood from a donor incubating
the disease who subsequently died (Wells et al 1978).
The rarity of the transmission of rickettsiosis by blood
transfusion probably reflects the clinical illness
suf-fered during most of the period of rickettsial infection
that makes blood donation unlikely (Tabor 1982)
In addition to the spotted fever group rickettsioses,
human anaplasmosis (formerly human granulocytic
ehrlichiosis) has also emerged in Europe (Parola
2004) This disease was first described in the USA
in 1994 and presents most commonly as a febrile
illness occurring in summer or spring The causative
organism is Anaplasma phagocytophilum, formerly
known as Ehrlichia equi, Ehrlichia phagocytophila
and human granulocytic ehrlichia The vector in
Europe is I ricinus, which is also the vector of
Lyme borreliosis Anaplasma are well preserved in
refrigerated blood components (McKechnie et al.
2000) Transfusion-transmitted ehrlichiosis has been
reported (Eastlund et al 1999).
Exogenous and various endogenous bacteria andbacterial products contaminating stored blood orblood components
Bacteria may affect blood or blood components in one
of the following ways:
1 Bacteria may contaminate solutions or equipment
that are to be used for transfusions but which have notyet been sterilized After sterilization the solutions orequipment may remain contaminated with heat-stablebacterial products (pyrogens) capable of producingfebrile reactions when they are introduced into the cir-culation (see Chapter 15) In contaminated equipment
or solutions such as hydroxyethyl starch used in pheresis, bacteria may survive ‘sterilization’ or maycontaminate solutions that have previously been steril-ized, for example when a glass container is cracked
leuca-during shipment (Wang et al 2000) This kind of
con-tamination has become rare
2 Bacteria originating from skin flora, such as
Staphylococcus epidermidis, Micrococcus species, Sarcina species and diphtheroids, may gain entrance to
the blood pack during venesection, especially if the site
of venepuncture is scarred It is virtually impossible todisinfect the deeper layers of the skin and a skin plugoften enters the blood pack upon collection (Anderson
et al 1986; Puckett 1986a).
3 Bacteria in the environment (Pseudomonas species,
Flavobacterium species, Bacillus species) may gain
entrance to blood components through minute lesions
in the packs, during collection or processing in open
systems (Szewzyk et al 1993).
4 The ports of packs of cryoprecipitate or FFP may
become contaminated, if not protected by a secondaryplastic bag, during thawing in waterbaths contami-
nated with pseudomonas (e.g Pseudomonas cepacia,
P aeruginosa) (Casewell et al 1981).
5 Bacteria circulating in the blood of an apparently
healthy donor suffering from asymptomatic bacteraemiamay proliferate in red cell components stored at 4°C or
in platelet concentrates stored at room temperature.Bacteraemia in donors may be chronic and low grade,
as in the case of the incubation or convalescence periods
of salmonella, Yersinia enterocolitica or Campylobacter jejuni infections, or acute and transient as occurs
within the first few hours after dental extractions when
the organisms involved are usually Streptococcus dans, Bacteroides species and, less often, Staphylococcus aureus A particularly notorious case reported from the
Trang 21viri-Clinical Center at NIH involved a repeat platelet donor
with asymptomatic Salmonella cholerasuis
osteomyeli-tis, whose donations resulted in sepsis in seven patients
and death in one of these before platelets were
identi-fied as the source of infection (Rhame et al 1973).
Frequency of bacterial contamination of blood
components
Bacteria are usually prevented from growing by the
antibacterial properties of blood Complement, even
in the absence of specific antibodies may kill bacteria,
which may then be phagocytosed by leucocytes
(Högman et al 1991; Gong et al 1994) Nevertheless
positive cultures in samples from blood or blood
com-ponents are found fairly frequently In an extensive
survey in which approximately 2500 units of red cells
and an equal number of platelet concentrates were
sampled annually, over a period of 10 years the mean
rates of positive cultures were 0.3% and 0.4%
respect-ively (Goldman and Blajchman 1991) Bacterial
con-tamination of whole blood cultured after 2–20 h of
storage at room temperature ranges from 0.34% to
2.2% (Bruneau et al 2001; de Korte et al 2001).
Survival of most organisms is reduced by storage at
refrigerated temperatures However, platelet
con-centrates are stored at room temperature so that
bacterial contamination and continued growth present
an urgent clinical problem In one early study, 6 out
of 3141 (0.19%) pooled concentrates from random
donors were found to contain bacteria just prior
to transfusion (Yomtovian et al 1993) In another
prospective study, 7 out of 15 838 (0.04%)
concen-trates were confirmed positive (Blajchman et al 1994).
A higher percentage (0.79%) was found in samples
taken from freshly prepared concentrates: of 11
posit-ive cultures only one was confirmed on repeated culture
from the same concentrate (Högman and Engstrand
1996) Only 5 out of 17 928 (0.03%) single-donor
concentrates obtained by apheresis contained bacteria
(Barrett et al 1993) A 6-year experience using a
semi-automated system for routine platelet cultures in
Denmark reported an initial positive reaction in 84
samples (0.38%) from 22 057 platelet units Growth
was confirmed in 70 of these (Munksgaard et al.
2004) The risk of collecting a contaminated unit of
platelets is estimated at 1 in 2000 (Chiu et al 1994).
A prospective study using a conservative case
definition found the rate of transfusion-transmitted
bacteraemia (in events/million units) to be 9.98 for single-donor platelets, 10.64 for pooled platelets and0.21 for RBC units (1/500 000); for fatal reactions, therates were 1.94, 2.22 and 0.13 respectively (Kuehnert
et al 2001) From a 12-year retrospective analysis
of transfusion data from a single institution, tomatic sepsis from transfused platelets was estimated
symp-to occur at between 1 in 5000 (pooled derived platelets) and one in 15 000 single-donor
whole-blood-apheresis platelet transfusions (Ness et al 2001).
The incidence of clinical reactions due to ated blood is much lower than that expected from thereported rates of positive bacterial cultures Althoughsome contaminated components, especially those containing large numbers of organisms or endotoxin,result in immediate, catastrophic reactions includingshock and death, others may be quite mild with little
contamin-more than low-grade fever or chills (Yomtovian et al.
1993) As many patients who receive platelet fusions already have an infection, febrile reactions orsymptoms of septicaemia may not be ascribed to the
trans-transfusion (Chiu et al 1994) Furthermore many of
these patients are being treated with antibiotics thatblunt detection and diagnosis of septic reactions
Autologous blood
As might be anticipated, autologous donations arealso susceptible to bacterial contamination At leastfive such cases have been reported The majority of
the infected units contained Y enterocolitica, and the
donors recalled in retrospect gastrointestinal episodes
in the weeks prior to donation (Haditsch et al 1994).
Some properties of organisms that grow in stored blood
Organisms isolated from refrigerated blood are ally Gram-negative rods capable of metabolizing citrate (Pittman 1953) Many strains of organisms isolated from contaminated stored blood cause clotformation by consuming citrate, yet another reason toexamine all units carefully at the time of issue (Braude
usu-et al 1952; Pittman 1953) Blood that is heavily
con-taminated with organisms is not necessarily lysed Among bacteria isolated from stored blood, 25%
haemo-produce no haemolysis (Braude et al 1952) Many
organisms that grow in stored blood are psychrophilic– organisms that grow preferentially in the cold
Trang 22Unless blood is heavily contaminated with
organ-isms, microscopic examination is an unreliable method
of detecting infection After serial dilutions of an
inoculum of a coliform organism to blood, 24 × 105
organisms/ml could be detected readily but 24 × 104/ml
(one organism in every 100 fields) could be detected
only with difficulty (Walter et al 1957) On the other
hand, when an aliquot of 0.3 ml was cultured for 24 h,
contamination could be detected when the number of
organisms added was as low as 24/ml
The effect of antibiotics on bacteria in stored blood
has been investigated Chlortetracycline,
oxytetracy-cline and polymyxin B in concentrations as low as
10 mg in 500 ml of blood (i.e 20 µg/ml) would all
pre-vent the inocula of either cold- or warm-growing
bac-teria from multiplying in human blood either in the
cold or at room temperature Nevertheless, routine
addition of antibiotics to stored blood cannot be
recommended First, antibiotics cannot be autoclaved
and would therefore have to be added later, and this
addition might itself contaminate the collection
Second, no antibiotics are effective against all
organ-isms Third, there is the risk of immunizing patients to
particular antibiotics or of inducing a hypersensitivity
reaction in a patient already immunized
Importance of maintaining refrigeration
After the first 24 h or so of storage, strict maintenance
of refrigeration at a temperature of 4°C becomes
essential When blood has to be transported for any
considerable distance the use of refrigerated containers
can maintain blood below 10°C for as long as 48 h
Some transfusion centres routinely keep blood at
approximately 20°C for up to 24 h after collection
before it is processed The storage at room
temper-ature does not seem to affect the yields of factor VIII in
plasma subsequently frozen and fractionated or the
shelf-life of platelets or red cells (Pietersz et al 1989).
Leucocytes in fresh blood have a bactericidal effect for
a few hours after collection and before processing into
blood components with removal of the buffy coats
From experiments in which defined numbers of
colony-forming units of different bacteria were added to fresh
blood, leucocytes were shown to effect clearing of
bac-teria, the efficiency varying according to the bacterial
species: Staphylococcus epidermidis and Escherichia
coli were cleared more readily than S aureus, which
was engulfed by the leucocytes and then released after
causing cell death (Högman et al 1991) The clearing
effect of leucocytes sometimes requires as long as 24 h.Some Gram-negative bacteria are killed by plasma fac-tors, presumably antibodies and complement Certain
anaerobic bacteria such as Propionibacterium species
do not grow in stored blood, regardless of the presence
or absence of white cells Yersinia enterocolitica was
cleared temporarily, but reappeared within a few hours;
if leucocytes were removed from the blood after 5 h,the units remained sterile Although some bacteria arekilled intracellularly, others kill the cells, which then
disintegrate releasing the bacteria (Högman et al 1991).
Bacteria contaminating different blood components
The type of bacterial contamination largely depends
on the type of blood component:
1 Red cells involved in cases of bacterial infection
have been contaminated mainly with Pseudomonas fluorescens, P putida and Y enterocolitica Yersinia is
normally sensitive to complement and is phagocytosed
by leucocytes in fresh blood However, it is capable ofproducing a virulence plasmid that expresses a surfaceprotein rendering the organism resistant to comple-ment and intracellular killing after phagocytosis (Lian
et al 1987) Yersinia grows well at 4°C, uses citrate
as a source of energy and, owing to its lack ofsiderophores, requires iron for optimum growth Redcell concentrates provide an ideal culture medium Theorganism produces a potent endotoxin during storage.Transfusion of contaminated red cells can cause fatalsepticaemia In almost all serious reactions due to
Y eneterocolitica, the red cell concentrates had been
stored for more than 3 weeks Phagocytes containinglive bacteria may persist after a donor has otherwisecontained a yersinia infection During storage of redcell concentrates the leucocytes may disintegrate,releasing live bacteria, which can then multiply
(Högman et al 1992) Leucocyte-depleted red cell concentrates, previously inoculated with Y eneteroco- litica remain sterile, whereas growth develops after
2–3 weeks in leucocyte-containing concentrates (Gong
et al 1994) Between April 1987 and the end of
February 1991, eight fatalities associated with thetransfusion of red cells contaminated with bacteriawere reported to the FDA; seven of these were due to
Y enterocolitica During the same period, 10 cases
of Y enterocolitica caused by contaminated red cell
Trang 23transfusions were reported in the USA, three cases
in France, one in Belgium and one in Australia The
patients developed fever and hypotension within 50 min
of the start of transfusion In the 10 cases that occurred
in the USA, the blood had been stored in the cold for a
mean of 33 days (range 25– 42 days) Although reports
of septicaemia due to yersinia are rare, the numbers
have increased in the past 10 years If patients are
tak-ing antibiotics, infection by blood contaminated with
yersinia can be asymptomatic (Jacobs et al 1989).
2 Platelet concentrates stored at 20–24°C have caused
fatal bacterial sepsis when contaminated with any of
several Gram-negative or -positive organisms such as
staphylococci, streptococci, Serratia species,
flavo-bacteria and salmonellae (Goldman and Blajchman
1991) One plateletpheresis donor with subclinical
Salmonella cholerae suis osteomyelitis caused sepsis
in seven platelet recipients (Rhame et al 1973).
3 Cryoprecipitates and FFP can become contaminated
with P cepacia and P aeruginosa during thawing in
con-taminated waterbaths (Goldman and Blajchman 1991)
4 Any of the above (1–3) can become infected if
the exterior of blood packs is massively contaminated
during manufacture by bacteria such as Serratia
marcescens Septicaemia has been reported in
recipi-ents of such contaminated units (Heltberg et al 1992).
Effect of transfusing bacterially
contaminated blood
Shock following the injection of bacteria is
pre-sumably due to bacterial toxins, although an immune
reaction between naturally occurring antibodies and
bacteria may also produce ill effects The transfusion
of contaminated blood may produce immediate
col-lapse, followed by profound shock and hyperpyrexia;
haemorrhagic phenomena due to disseminated
intra-vascular coagulation are common; in a series of
25 cases, mortality was 58% (Habibi et al 1973).
A review of more than 30 reports of post-transfusion
sepsis due to contaminated red cells, platelets or a
few other blood components showed that septic shock
developed in many cases and that the overall mortality
rate was 26% Common signs and symptoms reported
were fever, chills, vomiting, tachycardia and
hypo-tension, often during the transfusion, but sometimes
developing a few hours later In the case of
Pseudomonas cepacia, septicaemia or wound infection
developed several days or weeks after transfusion
(Goldman and Blajchman 1991) However, atypicalpresentations have been well documented in prospect-ive studies and suggest that clinical syndromes are
under-reported (Yomtovian et al 1993).
Investigations following the suspected transfusion of injected blood
Any blood remaining in the container should be cultured at various temperatures, including 4°C and20°C, in appropriate culture media A negative cultureexcludes the possibility that the blood was heavilyinfected at the time of transfusion A positive culturedoes not enable one to say with certainty whether theblood was contaminated before the transfusion orbecame contaminated during or after transfusion, orupon sampling Techniques to exclude contaminationupon sampling have been described (Puckett 1986b)
It is still useful to culture implicated containers thathave been left at room temperature for 24 h Most sterile units remain sterile or at worst lightly con-taminated In fatal cases, blood for culture should becollected from the body (e.g by cardiac puncture) assoon as possible after death Blood contaminated with
a coliaerogenes bacterium cultured positive, althoughwhen pseudomonads were involved, blood collected atautopsy was sterile (Pittman 1953)
Specific identification of the contaminating ism can be important for determining the extent ofdonor follow-up and the donor counselling message(Haimowitz 2005)
organ-Inspection of red cells: the dark side of transfusion
Colour change in stored red cell concentrates mayindicate that the unit has been contaminated with bac-teria In 13 units of red cells that supported the growth
of Y enterocolitica, a darkening related to haemolysis and a decrease in PO2was observed in the primary
container (Kim et al 1992) The attached sample
seg-ments, which were sealed from the main unit, remainedsterile and did not darken The colour change wasapparent in all the contaminated units by day 35, or1.5 to 2 weeks after the bacteria were first detected incultures of the blood A keen observer can identify
units grossly contaminated with Y enterocolitica by
comparison of the colour of the segment tubing
with that of the unit (Kim et al 1992) Review of
Trang 24photographs on file at the Centers for Disease Control
revealed this colour change in 2 units of blood that
caused transfusion-transmitted sepsis (Enterobacter
agglomerans and an unidentified Gram-negative
bacil-lus), which demonstrates that the colour change is not
limited to contamination by Y enterocolitica.
Screening for bacterial contamination
Avoidance of bacterial contamination is preferable
to detecting organisms downstream Examination
of the donor’s arms and rigorous skin preparation
reduce quantitative bacterial growth by as much as
50% (Goldman et al 1997) (see Chapter 1) Diversion
of an aliquot of the initial blood collection removes
some of the organisms that populate the phlebotomy
site (de Korte et al 2002) Screening collected blood for
bacterial contamination is complicated by the small
initial inoculum (sampling error), differential growth
rates of various species, and the potential for
introduc-tion of organisms at the time of the screening procedure
Despite the absence of convenient and inexpensive
detection methods, routine surveillance of platelet
preparations for bacterial contamination has been
instituted in several countries The two most
com-monly used methods are culture based A sensitive and
specific automated, continuous monitoring culture
system measures carbon dioxide production as a
marker of bacterial growth (Brecher et al 2001,
2004) Both aerobic and anaerobic cultures are
pos-sible The procedure increases the storage interval
before platelets can be released and cultures may turn
positive after platelets have been issued or transfused
A second system monitors consumption of oxygen
after 24 h at room temperature in aliquots of platelet
concentrates as a marker of microbial growth (Rock
et al 2004) The disadvantages include inability to
detect anaerobic organisms, reliance on analysis at a
single time point and limited ability to detect
slow-growing bacteria However only one example of a
fatality associated with an anaerobic organism has
been reported (McDonald et al 1998) Both methods
have interdicted contaminated transfusions
Malaria
Transmission of malaria by blood transfusion
Malaria can be transmitted by the transfusion of any
blood component likely to contain even small numbers
of red cells; platelet and granulocyte concentrates,fresh plasma and cryoprecipitate have all been incrim-inated An inoculum containing as few as 10 parasites
can cause Plasmodium vivax malaria (Bruce-Chwatt
1972)
Despite intensive eradication programmes, theprevalence of malaria in tropical and subtropicalcountries is extremely high, with about 300 millioncases reported each year, and more than 1 millionannual fatalities attributed to malaria in tropicalAfrica (Wyler 1983) The increasing resistance of themosquitoes to insecticides and the increasing appear-
ance of drug-resistant strains, particularly of P parum and the recurrence of malaria in areas where
falci-the disease had previously been eradicated, have greatlyincreased the incidence of malaria in recent years.Malaria parasites of all species can remain viable instored blood for at least 1 week (Hutton and Shute
1939) Cases of P falciparum malaria have been mitted by blood stored for 14 days (Grant et al 1960) and for 19 days (De Silva et al 1988) Malaria para-
trans-sites may survive longer in blood stored in containing solutions (WHO 1986) In a review ofnearly 2000 cases of transfusion malaria due to
adenine-P malariae, in the great majority of cases the blood
had been stored for less than 5 days and cases ing the transfusion of blood stored for 2 weeks wererare (Bruce-Chwatt 1974) The parasites survive well
follow-in frozen blood (Kark 1982) Plasma that has beenfrozen or fractionated has never been known to trans-mit malaria
In non-malarious countries, mainly because of delay
in diagnosis, post-transfusion malaria has a relativelyhigh fatality rate Malaria is particularly serious inpregnant women and in splenectomized or immuno-suppressed patients (Kark 1982; Bruce-Chwatt 1985).Transfusion-transmitted malaria usually responds
to conventional drug therapy In severe cases in whichthe diagnosis has been delayed and in fulminant cases,the patient may benefit from exchange transfusion
in the period before antimalarial drugs have had time
to become effective (Yarrish et al 1982; Kramer et al 1983; Files et al 1984) However, no controlled trials
have been performed and antimalarial medical therapy
is effective in most cases (Mordmuller and Kremsner1998)
The red cells in a renal allograft and in a bone
mar-row graft have been known to transmit P falciparum
Trang 25malaria (Kark 1982; Dharmasena and Gordon-Smith
1986) The plasmodia can also be transmitted by
transplacental passage
Frequency of post-transfusion malaria
Estimates of the frequency of post-transfusion malaria
vary from less than 0.2 cases per million units of blood
transfused in non-endemic countries to 50 or more
cases per million in some endemic countries
(Bruce-Chwatt 1985) In countries where malaria is
non-endemic the frequency of reported transfusion malaria
is relatively low The UK has reported eight cases in
50 years, the USA 26 cases in 10 years from 1972 to
1981, and France 110 cases in 20 years (Bruce-Chwatt
1985), although between 1980 and 1986 only 14 cases
were observed On the other hand, nine cases of
transfusion-transmitted malaria were reported to the
Centers for Disease Control (CDC) in the USA in
1982, although only 11 cases in the following 4 years
(Westphal 1991b) In Canada, only three cases of
transfusion-transmitted malaria were reported between
1994 and1999, a rate of 0.67 cases per million
dona-tions (Slinger et al 2001) The rate of
transfusion-transmitted malaria in the USA since 1992 has
fluctuated from 0.18 to 0.6 per million units
trans-fused (Mungai et al 2001) In many malarious areas
where reporting is inefficient, the frequency of
post-transfusion malaria is unknown
Frequency with which the different parasites
are involved
In the period 1950 –72, P malariae was responsible
for about 50% of cases of transfusion malaria and
P vivax for 20% (Bruce-Chwatt 1974) In the period
1973–80 a global survey showed a large relative
increase of cases due to P vivax, which, with a few
cases of P ovale, accounted for 42% of all cases;
cases due to P malariae fell to 38% whereas those due
to P falciparum increased four-fold (Bruce-Chwatt
1982) In more than one-half of the cases reported,
the species involved was not identified The relative
increase in cases due to two of the parasites has been
attributed to the failure of donors from highly endemic
areas to comply with rules laid down by transfusion
services (Guerrero et al 1983; Shulman et al 1984).
The simultaneous transmission of two different strains
of malaria (P falciparum and P malariae) by
trans-fusion has been described (Aymard et al 1980) Of
93 cases of transfusion-transmitted malaria reported
to the CDC from 1963 to 1999, 33 (35%) were due to
P falciparum, 25 (27%) were due to P vivax, 25 (27%) were due to P malariae, 5 (5%) were due to
P ovale, 3 (3%) were mixed infections and 2 (%) were
due to unidentified species; 10 out of the 93 patients(11%) died
Incubation period
The incubation period of transfusion malaria depends
on the numbers and strain of plasmodia transfused, onthe host and on the use of antimalarial prophylaxis;
with P falciparum and P vivax it is between 1 week and 1 month, but with P malariae it may be many
months (Bruce-Chwatt 1974)
Period for which donors remain infectious
P falciparum infections, prevalent in West Africa, are
usually eliminated within 1 year but have been known
to persist for as long as 5 years after the last exposure
to infected mosquitoes (Guerrero et al 1983) P parum transmitted from asymptomatic blood donors
falci-can cause fulminant infection and death in an alreadyill, non-immune recipient in whom the diagnosis,
being unexpected, is usually delayed P vivax
infec-tions, prevalent on the Indian subcontinent, mayrelapse for up to 2.5 years after infection but only
rarely after more than 3 years With P ovale a clinical
attack may occur up to 4 years after returning from anendemic area In immune carriers who have lived in
endemic areas for most of their lives, P falciparum or
P vivax may reappear long after the limits given above.
P malariae infections persist for longer than those of
any of the other malaria parasites Transmission ofmalaria has been known to occur up to 46 years afterthe last exposure to infection (Miller 1975)
Rules to prevent transmission of malaria in non-endemic areas
In non-endemic areas, eligibility of potential blooddonors who have visited endemic areas is restricted.Regulations and standards are not harmonized andnot always consistent Two different approaches havebeen adopted: the USA relies solely on the deferral ofsubjects who have been in endemic areas, whereas in
Trang 26Europe deferral and testing for malaria parasites can
be combined
All donor clinics should have an up-to-date WHO
map showing the areas endemic for malaria together
with an alphabetical list of the countries
In the USA, those who normally live in a
non-endemic area but have travelled in an area considered
to be endemic for malaria may be accepted as regular
blood donors 1 year after returning to a non-malarious
area, provided that they have had no symptoms
Pro-spective donors who have had malaria are deferred for
3 years after the cessation of therapy or after leaving
the malarial area if they have been asymptomatic
meanwhile In the USA, of 64 implicated donors
whose country of origin was reported, 38 (59%) were
foreign born Among those for whom complete
informa-tion was available, 37 out of 60 donors (6%) would
have been excluded from donating according to
guide-lines in place since 1994, and 30 out of 48 donors
(62%) should have been excluded under the guidelines
in place at the time of donation (Mungai et al 2001).
The Council of Europe in 1995 adopted the
recom-mendation that individuals born or brought up in
endemic areas may be accepted as donors of whole
blood 6 months after their arrival in a non-endemic
area, provided that an approved immunological test
has given a negative result (Council of Europe 2003)
Others from non-endemic areas, for example
trav-ellers, can be accepted as blood donors 6 months after
their return if they have been afebrile and have not
taken antimalarial prophylaxis; those who have had
febrile illnesses may be accepted if an antibody test
is negative at 6 months Although a proportion of
antibody-positive subjects are non-infectious and are
thus unnecessarily rejected, application of these
proced-ures makes it possible to use 75 –90% of units of blood
from donors returning from malarious areas to France,
the UK and Belgium (Soulier 1984; Wells and Ala 1985)
The foregoing rules will not prevent the occasional
transmission of P malariae and, especially, of P vivax.
Donations to be used for the preparation of plasma
for fractionation may be accepted from donors who do
not meet the above criteria
Tests to detect carriers
When blood films are examined by simple microscopy,
a density of less than 100 parasites per microlitre of
blood cannot be detected, although a unit from a
donor whose blood contained even one parasite permicrolitre would contain about half a million parasites(Bruce-Chwatt 1985)
The indirect fluorescent antibody test or ELISAoffers a good chance of detecting latent malarial infec-
tion (Deroff et al 1982; Voller and Draper 1982).
ELISA is more suitable for large-scale screening in the blood transfusion service and its specificity isimproving with the use of chromogenic substrates Acommercial ELISA for malarial antibodies has under-gone a successful trial in the UK Ideally, potential car-riers who volunteer as blood donors in non-malariousareas should be screened for malaria antibodies using
a test covering three homologous antigens However,
only P falciparum is readily available for in vitro tests:
although there is some degree of crossreactivity betweenthe different strains of malaria parasites it is not known
how valuable a test based on the use of P falciparum
would be in a country such as Mexico, where none ofthe 44 cases of transfusion malaria reported in 5 years
was due to this strain (Olivares-Lopez et al 1985).
Sensitive tests, using monoclonal antibodies, arebeing developed for use in endemic areas for detection
of parasites in red cells, and soluble antigens and bodies in plasma (Soulier 1984; Prou 1985) In a study
anti-in blood donors anti-in Chandigarh, India, a comparison
of different screening techniques for malaria carriersshowed that a test for malaria antigen, using mono-clonal antibodies, had good sensitivity and specificity.The test was considerably more sensitive than the
direct examination of blood films (Choudhury et al.
1991) Serological tests for malarial antibodies and testsfor malarial antigens are useful in the identification ofdonors implicated in cases of transfusion-transmittedmalaria in non-endemic countries when blood filmsare negative PCR has been used to determine theprevalence of malaria in potential blood donorsidentified as being at high risk of having malaria Only4% of blood donors tested positive for malaria byPCR Furthermore, 42% of blood donors who werenegative on PCR testing returned to donate, and
no cases of transfusion-transmissible malaria were
reported (Shehata et al 2004).
Prevention of transfusion malaria in malarious countries
In countries such as Nigeria, Zambia and Papua NewGuinea, where up to 10% of donors have plasmodia
Trang 27detectable by simple microscopy, the contribution
of blood transfusion to the overall problem of the
transmission of malaria is negligible In such countries,
the transmission of malaria to the recipient can be
prevented by giving antimalarial treatment to donor
or recipient Officer (1945) injected blood containing
some 300 million malaria parasites into each of five
healthy volunteers All volunteers were given
anti-malarial treatment after transfusion and none
devel-oped malaria
Trypanosoma cruzi Chagas’ disease –
American trypanosomiasis
In Latin America 24 million people are infected with
Trypanosoma cruzi Infection is most commonly
transmitted to humans, usually in childhood, by
triatomid bugs Until recently, Chagas’ disease was
limited to rural areas with mud huts and thatched
roofs but, due to migration of the rural population,
infected people are also found in urban areas (Wendel
1995) The next most common mode of transmission
is by blood transfusion
Treatment of acute infection cures 50 –90% of
patients, but most acute infections are subclinical and
untreated individuals remain infected throughout life
Depending on the country, between 5% and 40% of
untreated patients may develop serious chronic
com-plications such as cardiomyopathy, megaoesophagus
and megacolon, 10 or more years after infection
(Marsden 1984; Schmunis 1991)
In chronic infection, antibodies can be detected by
complement fixation, direct or indirect
haemagglu-tination, immunofluorescence or ELISA (Schmunis
1991) ELISA appears to be more sensitive than
indirect immunofluorescence and haemagglutination
(Magnaval et al 1985) Although tests for antibody
are positive in about 95% of chronic cases and in 50%
of acute cases, they are often negative during the first
few months of infection (Wolfe 1975) At least 50% of
those infected have parasitaemia and are thus likely
to be able to transmit infection by blood transfusion
(Cerisola et al 1972) Parasitaemia is diagnosed by
allowing triatomid bugs to feed on the patient’s
blood and then examining the insects for infestation
(‘xenodiagnosis’) Most Latin American countries test
part, but not all, of their blood donations for T cruzi
antibodies; the frequency of a positive test amongst
donors varies from about 0.3% to 28% and is as high
as 62% in parts of Bolivia (Schmunis 1991) Owing tomigration to urban areas and to the existence of paiddonors, antibody-positive donors are often found incities that lack the triatomid vector When blood from
an antibody-positive donor is transfused, the incidence
of infection in recipients is between 12% and 18% andcan be as high as 50% (Schmunis 1991) The parasites
of Chagas’ disease survive well in stored blood, andblood stored for more than 10 days can still be infec-
tious (Cerisola et al 1972) T cruzi can be transmitted
by plasma as well as by whole blood: the parasite vives in plasma which has been frozen at –20°C for
sur-24 h but it does not resist lyophilization (Wendel andGonzaga 1992) The addition of 125 mg of crystal(gentian) violet to a unit of stored blood kills the para-sites after storage at 4°C for 24 h without damagingthe red cells Blood treated with gentian violet causes
no obvious toxic reaction in the recipient, but cancause minor side-effects and rouleaux formation of the red cells Artificial light and sodium ascorbateaccelerate the effect and reduce the dose of gentian violet required to kill the parasites Gentian violet
has been reported to have mutagenic effects in vitro
(Wendel and Gonzaga 1992)
Latin Americans who migrate do so mainly to NorthAmerica where more than 5 million now live Morethan 100 000 may be infected with Chagas’ disease(Wendel 1993) Chagas’ disease acquired by bloodtransfusion has been reported in the USA, Canada and
Spain (Villalba et al 1992) Six cases of transmitted T cruzi have been reported in the USA and
transfusion-Canada (four and two cases respectively), with theimplicated donor in at least five of these cases from a
T cruzi endemic country (Leiby et al 2002a) Platelets
may be the blood component that is most often implicated in transfusion-transmitted Chagas’ disease,possibly because of their relatively short storage period
at temperatures favouring parasite survival (22–24°C)
or because platelet recipients are more likely to beimmunocompromised and therefore are more likely
to manifest clinical infection than recipients of other
components (Leiby et al 1997).
Other protozoaToxoplasma gondii
Toxoplasma gondii is an obligate intracellular
proto-zoan that replicates in the intestine of members of the
Trang 28cat family, its definitive host Humans may be infected
by eating undercooked lamb or pork or by exposure to
the faeces of infected cats Seroprevalence of T gondii
rises with age and the incidence of infection varies with
the population group and geography In the USA,
almost one-quarter of the population has been infected
(Jones et al 2001) T gondii has been isolated from
donors’ blood up to 4 years after the onset of infection;
entry of parasites into the blood occurs for only a
few weeks, but as T gondii is an obligate intracellular
parasite it persists in the white cells and can survive
storage at 4°C for 4–7 weeks (Tabor 1982) Congenital
transmission occurs, particularly in women infected
shortly before or during pregnancy
Most infections with toxoplasma are asymptomatic
or result in mild fever and lymphadenopathy However,
infection in immunocompromised patients is
poten-tially life threatening Transmission of T gondii from
seropositive solid organ donor to seronegative recipient
is well documented, but for recipients of haematopoietic
stem cell transplants, reactivation of latent infection
appears to be more important Leucocyte transfusions
from donors with high levels of toxoplasma
anti-bodies have resulted in severe acute toxoplasmosis
in immunosuppressed patients (Siegel et al 1971;
Tabor 1982) As the risk of severe
transfusion-acquired toxoplasmosis is confined almost entirely to
immunosuppressed patients transfused with leucocyte
concentrates, some centres have elected to maintain a
panel of donors negative for toxoplasma antibodies
for leucapheresis and for the provision of granulocyte
concentrates for premature babies (M Contreras,
per-sonal communication)
Babesia
In North America, the parasite causing babesiosis in
humans is Babesia microti, which occurs most often
in the north-east and in Wisconsin and Minnesota
However, the geographic range of B microti is
expanding Other babesia species have been
implic-ated in transfusion transmission in the western USA,
and the mobility of blood donors and blood
com-ponents may result in the appearance of
transfusion-transmitted babesiosis in areas less familiar with these
parasites (Cable and Leiby 2003) In Europe, the
responsible parasite is Babesia bovis
Transfusion-transmitted cases have been reported only in the USA
(Popovsky 1990; Mintz et al 1991) Babesia microti,
like Anaplasma phagocytophila and B burgdorferi,
is transmitted to humans from animal reservoirs (primarily deer and mice) by black-legged ticks of the
genus Ixodes (Cable and Leiby 2003) For donor
screening purposes, self-reported tick bites are neithersensitive nor specific indicators of serological status
(Leiby et al 2002b).
B microti reproduce only in red cells, and on blood smear the organisms appear similar to Plasmodium falciparum The clinical symptoms also resemble those
of malaria The diagnosis must be confirmed by testing
for Babesia antibodies and by inoculating hamsters
with the patient’s blood Parasites can survive for up to
35 days in blood stored at 4°C, and persist in frozenred cells for years
Although normally mild and frequently tomatic, the disease can be fulminant and rapidly fatalfor immunosuppressed and splenectomized patients.Since 1980, more than 40 USA cases of post-transfusionbabesiosis, including at least two cases of WA-1, thewestern US variant, have been reported, most of them
asymp-in immunocompromised or splenectomized patients
In two splenectomized patients infected by transfusion(one by a platelet concentrate), exchange transfusionusing whole blood helped bring about control of the
disease (Jacoby et al 1980; Cahill et al 1981) As in the
case of malaria, the role of exchange transfusion is notwell documented, but the combination of clindamycinand quinine seems to eradicate the parasite from most
immunosuppressed patients (Smith et al 1986).
Microfilariae
Microfilariae of five common species have been shown
to persist in the circulation of infected subjects for
years and to survive in stored blood Wuchereria bancrofti can be detected in refrigerated blood for 3
weeks Microfilariae can be transmitted to transfusionrecipients Three varieties of filariae can cause disease
in humans: Brugia malayi in South-East Asia, Loa loa
in Africa and W bancrofti in southern Asia, tropical
Africa and some tropical areas of Latin America.However, blood transfusion-transmitted microfilariaenever reach the adult stage in non-endemic areasbecause of the absence of the vector necessary for thesecond passage to humans Recipients of infectedblood usually have no symptoms, although they mayoccasionally experience acute inflammation of thespleen, lymph nodes or lungs, sometimes with tropical
Trang 29pulmonary eosinophilia Recipients may develop
fever, headache and rash as a hypersensitivity response
to the dead microfilariae (Choudhury et al 2003).
Signs and symptoms are self-limited and so is the
sur-vival of microfilariae in transfusion recipients (Tabor
1982; Westphal 1991a)
Leishmaniasis
In 1903, Leishman and Donovan independently
described the protozoan now called Leishmania
dono-vani in patients from India with the life-threatening
disease now called visceral leishmaniasis or kala-azar.
Leishmaniasis and its major syndromes (visceral,
cutan-eous and mucosal) have changed little over the past
century, but information regarding the endemic areas,
the vectors and the transfusion risks have evolved
considerably (Herwaldt 1999) More than 20 different
species of Leishmania have been recognized, some
anthroponotic but most zoonotic The organism is
transmitted by the female phlebotomine sand fly,
of which some 30 species are vectors Visceral
leish-maniasis or kala-azar caused primarily by Leishmania
donovani is found most often in China, India and the
eastern Mediterranean L major, L tropica and
L infantum are found in the Middle East However, the
organisms are found in jungles, deserts, rural areas and
cities of some 88 countries Leishmaniasis is endemic
in much of Latin America between northern Argentina
(L braziliensis, L panamensis) and southern-central
Texas (L mexicana), and in south-western Europe
(France, Spain, Portugal, Italy, Greece) and, because
the sand fly has a limited range, microfoci of activity
are found within countries where the disease is
con-sidered endemic The cutaneous form of the disease
goes by many names, including Baghdad Aleppo boil,
Oriental sore and bush yaws These characteristics
make screening questions difficult to formulate There
is no screening test and no licensed drug therapy
No vaccine has been developed, although paediatric
inoculation using preparations from active sores is
widely practised in endemic areas
Leishmania multiply in mononuclear phagocytes
and clinical disease transmitted by blood
trans-fusion has been reported in transplant recipients, in
haemodialysis patients and in newborn infants When
present in blood, the parasite has been found in
large mononuclear cells and granulocytes (Shulman
1990) The incubation period can vary from 10 days
to 2 years, but is generally 2– 6 months and results inchronic infection Leishmania species are clearly bloodtransmitted, although fewer than 15 transfusion caseshave been reported, one of which was fatal Canine
transmission is well recognized (Owens et al 2001).
Studies of asymptomatic blood donors in southern
France indicated that L infantum circulates
intermit-tently at low levels and mononuclear cells from these
donors can infect Syrian hamsters (le Fichoux et al.
1999) Although the conventional teaching holds thatvisceral leishmaniasis poses the greatest risk of blood
transmission, ‘dermatotropic’ organisms such as L tropica, L braziliensis and L panamensis have been
found in blood decades after the patient’s exposure.Human transfusion transmission results in viscerocu-taneous disease
L tropica has been demonstrated to survive in
stored blood under blood bank conditions for 25 days
(Grogl et al 1993) A 1-year deferral was instituted by
the USA Department of Defense in 1991 and again in
2003 for service personnel stationed in endemic areas
in Iraq Deferral for diagnosed infection is indefinite.Cases of possible transmission of kala-azar by transfu-sion in India, China, Brazil and four European coun-
tries have been reported (Chung et al 1948; Cummins
et al 1995; Singh et al 1996) Subjects with a history
of visceral leishmaniasis must not be accepted asdonors (Council of Europe 1992)
African trypanosomiasis (sleeping sickness) has
rarely been transmitted by transfusion (Wolfe 1975;Tabor 1982)
Methods of inactivating pathogens in blood and blood products
Despite careful donor selection and sensitive atory testing, a small risk of transmission of infectiousagents by transfusion remains, particularly with prod-ucts obtained from large pools of plasma The frac-tions obtained at an early stage of the cold-ethanolfractionation, for example fibrinogen, factor VIII, factor IX and prothrombin complex concentrate, con-tain intact virus Plasma fractions obtained later in thefractionation process, such as the immunoglobulinsand albumin, are safer, although HCV can be detected
labor-in the fraction from which IVIG is prepared (Yei and
Yu 1992) HCV has been transmitted by IVIG
pre-parations (Weiland et al 1986; Bjoro et al 1994; Meisel
et al 1995; Power et al 1995).
Trang 30Inactivation of viruses in blood products or their
removal, or both, should further increase the safety
of transfusion and might protect against unknown
pathogens or those for which no screening tests are
available The ideal inactivation technology should
reduce the risk of infection from the blood component,
result in minimal damage to the component cell or
protein content and function, and pose minimal
toxicological risk to the recipient, particularly to
sus-ceptible patients such as gravid women and neonates
The risk–benefit estimates have clearly favoured
inactivation technologies for protein concentrates
derived from large plasma pools The calculus differs
for cellular components derived from single donors
Plasma and plasma products
Heat treatment
Pasteurization and vapour treatment of liquid clotting
factor concentrates and heat treatment of lyophilized
concentrates (‘dry heat’) have been used for decades
(Kingdon and Lundblad 2002) The efficacy of these
methods differs Hepatitis viruses have been
trans-mitted by lyophilized concentrates heated between
60°C and 68°C for periods of between 24 and 72 h
(Colombo et al 1985; Blanchette et al 1991) Even the
more heat-labile HIV has been transmitted by such
concentrates (Mariani et al 1987; Williams et al.
1990b) Dry heating at 80° for 72 h, however,
effec-tively inactivates HIV and hepatitis viruses No
evid-ence of infection with these viruses was detected in 38
previously untreated haemophiliacs treated with
dry-heated concentrates (Skidmore et al 1990; Williams
et al 1990b) Pasteurized concentrates are also free
from the risk of transmitting HIV (Schimpf et al 1987)
and in prospective studies have been found not to
transmit hepatitis viruses (Mannucci et al 1990; Azzi
et al 1992; Kreuz et al 1995) On the other hand,
sev-eral cases of hepatitis B or C have been ascribed to the
use of pasteurized concentrates in patients not enrolled
in controlled studies (Kernoff et al 1987; Brackmann
and Egli 1988; Shulman et al 1992).
A major disadvantage of the heat treatment
methods is that they may not inactivate non-enveloped
viruses such as HAV and parvovirus B19 Some
recipi-ents have developed signs of parvovirus B19 infection
(Azzi et al 1992) However, dry heat treatment at
100°C for 1 h more effectively inactivates both
lipid-enveloped and non-lipid-enveloped viruses and, whenapplied to IVIG preparations, leads to only a very
small loss of IgG (Rubinstein et al 1994).
Solvent–detergent treatment
The combination of a solvent and a detergent disruptslipid-enveloped viruses such as HIV and hepatitis
viruses A non-volatile solvent, tri-(n-butyl) phosphate
(TNBP) is used in combination with a detergent such
as sodium cholate (Prince et al 1986) The solvent–
detergent (S–D) mixture is removed at the end of theprocedure by precipitation of the product or by chro-matography The viral safety of S–D treatment hasbeen shown by clinical trials in over 100 haemophiliacsand in hundreds of thousands of transfusions (Mannucci
1993; Klein et al 1998).
The S–D method has been adapted for whole plasmaand involves incubation of pooled thawed plasma withthe organic solvent TNBP and the detergent Triton
X-100 for 4 h at 30°C (Horowitz et al 1992; Klein
et al 1998) A drawback of the S–D method is that
non-enveloped viruses are not inactivated Severalreports of transmission of HAV and parvovirus B-19
by S–D-treated concentrates have been documented
(Mannucci 1992; Temperley et al 1992; Peerlinck and
Vermylen 1993) Factor VIII or IX concentrates tain diminished levels of anti-HAV which in otherfractions, such as IVIg, neutralizes the virus
con-S–D treatment has a few other flaws The reducedlevel of alpha(2)-antiplasmin in S–D plasma has causedconcern when the product is used in patients with liver disease; however, controlled studies have not
confirmed a clinical problem (Williamson et al 1999).
S–D plasma also has reduced levels of protein S, aplasma factor with anticoagulant properties Reducedprotein S may contribute to reports of venous throm-bosis during massive plasma infusion In 68 con-secutive patients with thrombotic thrombocytopenicpurpura (TTP) (25 men, 43 women), eight documentedthromboembolic events (six deep venous thromboses,three pulmonary emboli) were identified in sevenpatients during therapeutic plasma exchange All sixwere associated with central venous lines at the site
of thrombosis (Yarranton et al 2003).
One little appreciated benefit of S–D plasma is theabsence of association with TRALI, almost certainly
as a result of the pooling of 12 000 units per batch.However, for a variety of reasons, including safety and
Trang 31commercial failure, S–D plasma is no longer available
in the USA
Heating in supension with n-heptane
Heating with n-heptane at 60°C for 20 h has been used
for factor VIII and IX concentrates but, as hepatitis
viruses have been transmitted by concentrates treated
in this way, S–D treatment is preferred
Methylene blue treatment
Methylene blue was first used clinically by Paul Ehrlich
in the nineteenth century and has been used as a
viruci-dal agent for more than half a century As with other
photoactive chemicals, methylene blue and similar
phenothiazine dyes have a high affinity for both
nucleic acids and the surface structures of viruses On
exposure to red (visible) light at 620–670 nm,
excita-tion of the dye causes chemical modificaexcita-tion of
adja-cent molecules, a process which involves oxygen
radicals (Lambrecht et al 1991) Methylene blue
treat-ment is currently the only method that can be applied
to individual units of plasma Residual dye is minimal
(1 µM) and can be further reduced 10-fold from the
final product by filtration
Methylene blue treatment does not inactivate
intra-cellular viruses efficiently but, as the plasma is frozen
and thawed, leucocytes are disrupted with release
of viral particles Neither does it effectively reduce
transmission of bacteria or protozoa For reasons that
remain unexplained, non-enveloped viruses, especially
HAV, are relatively resistant to methylene blue (Wagner
2002) An unfortunate consequence of methylene blue
treatment of plasma is the loss of 10% or more of some
clotting factors, particularly factor VIII and
fibrino-gen (Cardigan et al 2002; Williamson et al 2003).
Cryoprecipitate prepared from this plasma has not
been extensively studied and is not in current use
Commercial manufacturing systems have been
de-veloped and millions of single units of plasma treated
with methylene blue have now been transfused in
Europe without unexpected side-effects (Williamson
et al 2003) Concern remains in some quarters about
the potential mutagenic effects of methylene blue and
its derivatives, even in low concentration To date, no
immediate problems have been encountered in
trans-fusion recipients, although the number of neonates,
children and parous women treated remains small
Other methods of inactivation: at the bench but not yet at the bedside
Other methods include the use of the virucidal agent
hypericin (Lavie et al 1995) and photochemical
treat-ment with virucidal short-wavelength ultraviolet light
with the addition of rutin or psoralen (Chin et al 1995; Hambleton et al 2002).
Cellular componentsSome viruses such as CMV and HTLV are transmittedexclusively by leucocytes, whereas others, such asHIV, HBV and HCV are transmitted both by cellularcomponents and by plasma HIV can be internalized
by platelets, which may contain large amounts of the
virus (Zucker-Franklin et al 1990; Lee et al 1993).
HIV associates with red cells, probably as antibody–complement adherent complexes, but it is not clearwhether this ‘pool’ of virus is associated with transmis-
sion (Hess et al 2002) HPV B19 is known to adhere to
red cells To be effective, inactivation methods shoulddeal with free virus, virus attached to cells and intracel-lular virus
Physical methods
As mentioned above, the removal of leucocytes fromred cell and platelet concentrates reduces transmis-sion of CMV and HTLV-I and -II (Kobayashi 1993).Extensive washing and the process of freezing and deglycerolization are also effective in reducing trans-
mission (Brady et al 1984; Taylor et al 1986; Sayers
et al 1992).
Chemical and photochemical techniques
Several chemical and photochemical processes targetnucleic acid to inactivate residual contaminatingviruses, bacteria and protozoa in blood components(Klein 2005) As an added benefit, nucleic acid-targetedprocesses have the potential to inactivate residual lymphocytes and prevent transfusion-associated graft-
versus-host disease (Fast et al 2002, 2003, 2004).
Whole blood and red cells: the hurdles remain
For red cell concentrates, agents must be selected thatare photoactivated with a wavelength above that of
Trang 32haemoglobin that would otherwise absorb the agent.
Optimal properties of sensitizing dyes for use in red
cell suspensions include selection of dyes that traverse
cell and viral membranes, bind to nucleic acids, absorb
light in the red region of the spectrum, inactivate a
wide range of pathogens, produce little red cell
photo-damage from dye not bound to nucleic acid and do not
haemolyse red cells in the dark Some agents inactivate
both free and cell-associated virus, but subtle changes
in the red cell membrane may affect storage, survival or
immunogenicity (Sieber et al 1992; Wagner et al 1993).
A red cell additive, S-303 or frangible anchor linker
effector, is an alkylating agent that inactivates
nuc-leated pathogens by crosslinking DNA and RNA in a
rapid, light-independent reaction (Klein 2005) The
initial enthusiasm for this agent has been tempered
by the report of red cell antibody formation in two
subjects enrolled in clinical trials (L Corash, personal
communication) Antibody formation has also been
reported during a trial of chronic transfusion of
pati-ents with sickle cell disease using red cells treated
with PEN110 (J Chapman, personal communication)
PEN110, a binary ethyleneimine that binds covalently
to nucleic acid is also light independent and interferes
with polymerase-mediated replication (Lazo et al.
2002; Ohagen et al 2002)
A 50-µM riboflavin solution added to diluted red
cells that are exposed to visible light of 450 nm, a
wavelength with minimal absorbance by haemoglobin,
effects a two- to four-log reduction of a number of
pathogens (Klein 2005) Clinical data for riboflavin
treatment of red cells are not yet available No method
has been licensed for inactivating pathogens in whole
blood or red cells
Methods for treating platelets
PEN110 treatment, which involves covalent binding
of drug to nucleic acids (see above), requires an
extens-ive washing procedure to remove residual additextens-ive, so
the process has not been applied to plasma or platelet
components Riboflavin, an essential nutrient (vitamin
B2) that absorbs both visible and UVA light, has been
investigated with both components (Li et al 2004).
Riboflavin’s three-ringed planar structure intercalates
between bases of DNA and RNA, and upon light
exposure, oxidizes guanosine through electron transfer
reactions, resulting in single-strand breaks of nucleic
acids and formation of covalent adducts (Dardare and
Platz 2002) The process has proved effective against
a wide range of human and animal pathogens, ing bacteria, intracellular HIV-1, West Nile virus andporcine parvovirus in preclinical studies of platelets
includ-and plasma (Goodrich 2000; Ruane et al 2004).
Initial toxicology assessment of the photoproducts ofriboflavin generated under proposed treatment condi-tions have been encouraging
By far the greatest experience with pathogen tion of platelet components involves the use of pso-ralen additives and UVA light activation Psoralenspass through the cell membrane and the capsids ofviruses and intercalate reversibly into helical regions
reduc-of nucleic acids Upon illumination with UVA (300–
400 nm) covalent crosslinks to pyrimidines in RNA andDNA form and block the replication and transcription
of nucleic acids (Hanson 1992) 8-Methoxypsoralen(8-MOP) inactivates free virus and even HIV incorp-
orated in the genome (Corash et al 1992) This very
high-energy dose of UV damages platelets, but whenaminomethyl-trimethyl psoralen (amotosalen, AMT,S-59) is used, lower doses of UV can be applied forshorter periods without loss of virucidal activity
(Corash et al 1992; Benade et al 1994) When
pso-ralens are used, the platelets must be suspended inplasma-reduced medium, as plasma inhibits the inac-
tivation of virus (Moroff et al 1992) The addition of
agents that neutralize oxygen radicals further protects
the platelet membrane (Margolis-Nunno et al 1994).
Some psoralen derivatives also limit platelet damage(Goodrich 1994) Moreover, the derivatives do notappear to be mutagenic in the absence of UVA
(Wollowitz et al 1994; Yerram et al 1994) The
potential mutagenicity of psoralen is also avoided byabsorbing excess agent on a ligand, C18, fixed on sili-con as used for S–D-treated plasma (Margolis-Nunno
et al 1995) Extensive preclinical toxicology studies
for S-59-treated platelets showed no CNS, cardiac orreproductive toxicity, and no evidence of genotoxicity
or phototoxicity In a heterozygote p53 knockoutmouse model, exposure to S-59-treated plasma over
6 months or 26 weeks did not produce excess genicity This system (INTERCEPT) is licensed inEurope as a medical device
carcino-Three clinical trials in 166 thrombocytopenicpatients were conducted in Europe, two with buffycoat platelets and one with apheresis platelets Thesestudies demonstrated that, for equal platelet doses,INTERCEPT platelets provided similar platelet count
Trang 33increments to conventional platelets; the adverse
reac-tion profile appeared comparable (van Rhenen et al.
2003) A USA phase III study evaluating haemostatic
efficacy and safety of transfusions of apheresis platelet
concentrates randomized 645 patients to receive
either S-59 photochemically treated or conventional
platelets for up to 28 days Although the mean 1-h
post-transfusion platelet corrected count increment
(CCI) (11.1 × 103treatment vs 16.0 × 103control),
average number of days to next platelet transfusion
(1.9 treatment vs 2.4 control) and number of platelet
transfusions (8.4 treatment vs 6.2 control) differed,
the trial demonstrated equivalence in prevention and
treatment of grade 2 and higher grade bleeding
accord-ing to WHO criteria (McCullough et al 2004).
References
Aach RD, Stevens CE, Hollinger FB et al (1991) Hepatitis C
virus infection in post-transfusion hepatitis An analysis
with first- and second-generation assays N Engl J Med
325: 1325–1329
Ablashi DV, Salahuddin SZ, Josephs SF et al (1987) HBLV
(or HHV-6) in human cell lines (Letter) Nature (Lond)
329: 207.
Adler SP (1984) International forum: Transfusion
trans-mitted CMV infections Vox Sang 46: 387–414
Adler SP, Chandrika T, Lawrence L et al (1983)
Cyto-megalovirus infections in neonates acquired by blood
transfusions Pediatr Infect Dis 2: 114–118
Aledort LM, Levine PH, Hilgartner M et al (1985) A study of
liver biopsies and liver disease among hemophiliacs Blood
66: 367–372
Allain JP (2000) Genomic screening for blood-borne viruses
in transfusion settings Clin Lab Haematol 22: 1–10
Allain JP (2004) Occult hepatitis B virus infection:
implica-tions in transfusion Vox Sang 86: 83–91
Allain JP, Laurian Y, Paul DA et al (1986) Serological
markers in early stages of human immunodeficiency virus
infection in haemophiliacs Lancet ii: 1233–1236
Allain, J-P, Coghlan PJ, Kenrick KG et al (1996) Safety and
efficacy of anti-hepatitis C virus screening of blood donors
with two sequential screening assays Transfusion 36: 401–
406
Alter HJ (1975) Hepatitis B surface antigen and the health
care professions In: Transmissable Disease and Blood
Transfusion TJ Greenwalt, GA Jamieson (eds) New York,
NY: Grune & Stratton
Alter HJ (1992) New kit on the block: evaluation of second
generation assays for detection of antibody to hepatitis C
virus Hepatology 15: 350–353
Alter HJ, Houghton M (2000) Clinical Medical Research Award Hepatitis C virus and eliminating post-transfusion hepatitis Nature Med 6: 1082–1086
Alter HJ, Tabor E, Meryman HT et al (1978a) Transmission
of hepatitis B virus infection by transfusion of deglycerolized red blood cells N Engl J Med 298: 637
frozen-Alter HJ, Purcell RH, Holland PV et al (1978b)
Transmis-sible agent in non-A, non-B hepatitis Lancet 1: 459–463
Alter HJ, Purcell RG, Shih JW et al (1989a) Detection of
antibody to hepatitis C virus in prospectively followed transfusion recipients with acute and chronic non-A, non-B hepatitis N Engl J Med 321: 1495–1500
Alter MJ, Hadler SC, Judson FN et al (1990) Risk factors
for acute non-A, non-B hepatitis in the United States and association with hepatitis C virus infection JAMA 264: 2231–2235
Alter HJ, Nakatsuji Y, Melpolder J et al (1997) The
incid-ence of transfusion-associated hepatitis G virus infection and its relation to liver disease N Engl J Med 336: 747– 754
American Association of Blood Banks, American Red Cross, Council of Community Blood Centers (1983) Joint state- ment on acquired immune deficiency syndrome (AIDS) related to transfusion (1983a) Transfusion 23: 87–88
Amman AJ, Cowan MJ, Wara DM et al (1983) Acquired
immunodeficiency in an infant: possible transmission by means of blood products Lancet i: 956–957
Anahane Y, Aikawa T, Sugai Y et al (1992) Transmission of
HCV between spouses Lancet 339: 1059–1060
Anand A, Gray ES, Brown T et al (1987) Human parvovirus
infection in pregnancy and hydrops fetalis N Engl J Med 316: 183–186
Anderson MJ, Jones SE, Fisher-Hoch SP et al (1983) Human
parvovirus The cause of erythema infectiosum (Fifth ease) Lancet i: 1378
dis-Anderson KC, Lew MA, Gorgone BC et al (1986)
Transfusion-related sepsis after prolonged platelet storage.
Am J Med 81: 405–411
Anderson DW, Epstein JS, Lee TH et al (1989) Serological
confirmation of human T-lymphocytotropic virus type I infection in healthy blood and plasma donors Blood 74: 2585–2591
Andes WA, Daul CB, deShazo RD et al (1988)
Sero-conversion to human immunodeficiency virus (HIV) in hemophiliacs Relation to lymphadenopathy Transfusion 28: 98–102
Ando Y, Nakano S, Saito K et al (1986) Prevention of
HTLV-I transmission through the breast milk by a drying process Jap J Cancer Res (Gann) 77: 974 –977
freeze-Andreu G, Morel P, Forestier F et al (2002) Hemovigilance
network in France: organization and analysis of immediate transfusion incident reports from 1994 to 1998 Trans- fusion 42: 1356–1364
Trang 34Araujo A, Hall WW (2004) Human T-lymphotropic virus
type II and neurological disease Ann Neurol 56: 10 –19
Aronson DL (1979) Factor IX complex Semin Thromb
Hemost 6: 28–43
Aymard JP, Vinet E, Lederlin P et al (1980) Paludisme
post-transfusionnel: un cas de double infestation a Plasmodium
falciparum et Plasmodium malariae Rev Fr Transfusion
Immunohematol 23: 491– 493
Azzi A, Ciappi S, Zakvrezeska K (1992) Human parvovirus B
19 infection in hemophiliacs first infused with two
high-purity virally attenuated factor VIII concentrates Am J
Hematol 39: 228–230
Backer U, Weinauer F, Gathof G et al (1987) HIV antigen
screening in blood donors (Letter) Lancet ii: 177–180
Barbara JAJ (1983) Microbiology In: Blood Transfusion.
Bristol: John Wright
Barbara JAJ, Briggs M (1981) Follow-up of HBsAg-positive
donors to determine the proportion undergoing acute
infections (Abstract) Transfusion 21: 605–606
Barbara JAJ, Mijovic V, Cleghorn et al (1978) Liver enzyme
concentrations as a measure of possible infectivity in chronic
asymptomatic carriers of hepatitis B BMJ 12: 1600–1602
Barbara JAJ, Salker R, Lalji F et al (1982) TPHA compared
with cardiolipin tests for serological detection of early
primary syphilis J Clin Pathol 35: 1394–1395
Barre-Sinoussi F, Chermann JC, Rey F et al (1983) Isolation
of a T-lymphotropic retrovirus from a patient at risk for
acquired immune deficiency syndrome (AIDS) Science 220:
868–871
Barrett BB, Andersen JW, Anderson KC (1993) Strategies for
the avoidance of bacterial contamination of blood
com-ponents Transfusion 33: 228–233
Baumgartner JD, Glauser MP, Burgo-Black AL et al (1982)
Severe cytomegalovirus infection in multiply transfused,
splenectomised, trauma patients Lancet ii: 63–66
Bazarbachi A, Soal F, Laroche L et al (1993) HTLV-I
provirus and mycosis fungoides Science 259: 1470
Benade LE, Shumaker J, Xu Y et al (1994) Inactivation of
free and cell-associated human immunodeficiency virus in
platelet suspensions by aminomethyltrimethylpsoralen and
ultraviolet light Transfusion 34: 680–684
Bendheim PE, Brown HR, Rudelli RD et al (1992) Nearly
ubiquitous tissue distribution of the scrapie agent
pre-cursor protein Neurology 42: 149–156
Berger EA, Murphy PM, Farber JM (1999) Chemokine
recep-tors as HIV-1 coreceprecep-tors: roles in viral entry, tropism, and
disease Annu Rev Immunol 17: 657–700
Bergman GE (1995) Transmission of hepatitic C virus by
monoclonal-purified viral-attenuated factor VII
concen-trate Lancet 346: 1296–1297
Bessos H, Drummond O, Prowse C et al (2001) The release
of prion protein from platelets during storage of apheresis
platelets Transfusion 41: 61–66
Biggerstaff BJ, Petersen LR (2003) Estimated risk of mission of the West Nile virus through blood transfusion
trans-in the US, 2002 Transfusion 43: 1007–1017
Biswas R, Tabor E, Hsia CC et al (2003) Comparative
sensitivity of HBV NATs and HBsAg assays for detection
of acute HBV infection Transfusion 43: 788–798
Bjoro K, Froland SS, Yun Z et al (1994) Hepatitis C infection
in patients with primary hypogammaglobulinemia after treatment with contaminated immunoglobulin N Engl J Med 331: 1607–1611
Blajchman MA, Ali AM, Richardson HL (1994) Bacterial contamination of blood components Vox Sang 67, S3: 25–33
Blanchette VS, Vorstman E, Shore A et al (1991) Hepatitis C
infection in children with hemophilia A and B Blood 78: 285–289
Bloch O (1941) Loss of Treponema pallidum in citrated
blood at 5 °C Bull Johns Hopkins Hosp 68: 412 Blum HE, Vyas GN (1982) Non-A, non-B hepatitis: a con- temporary assessment Haematologia 15: 162–183 Blumberg BS, Sutnick AI, London WT (1968) Hepatitis and leukaemia: their relation to Australia antigen Bull NY Acad Med 44: 1566
Blumel J, Schmidt I, Effenberger W et al (2002) Parvovirus
B19 transmission by heat-treated clotting factor trates Transfusion 42: 1473–1481
concen-Boneva RS, Grindon AJ, Orton SL et al (2002) Simian
foamy virus infection in a blood donor Transfusion 42:
886 – 891
Boppana SB, Rivera LB, Fowler KB et al (2001) Intrauterine
transmission of cytomegalovirus to infants of women with preconceptional immunity N Engl J Med 344: 1366 –1371 Bowden RA (1995) Transfusion-transmitted cytomegalovirus infection Immunol Invest 24: 117–128
Bowden RA, Sayers M, Flournoy N et al (1986)
Cyto-megalovirus immune globulin and seronegative blood products to prevent primary cytomegalovirus infection after marrow transplantation N Engl J Med 314: 1006 – 1010
Bowden RA, Sayers M, Gleaves et al (1987)
Cyto-megalovirus-seronegative blood components for the prevention of primary cytomegalovirus infection after marrow transplantation Considerations for blood banks Transfusion 27: 478 – 481
Bowden RA, Slichter SJ, Sayers M et al (1995) A comparison
of filtered leukocyte-reduced and cytomegalovirus (CMV) seronegative blood products for the prevention of trans- fusion-associated CMV infection after marrow transplant Blood 86: 3598–3603
Bowen EF, Sabin CA, Wilson P et al (1997) Cytomegalovirus
(CMV) viraemia detected by polymerase chain reaction identifies a group of HIV-positive patients at high risk of CMV disease AIDS 11: 889–893
Trang 35Brackmann HH, Egli H (1988) Acute hepatitis B infection
after treatment with heat-inactivated Factor VIII
concen-trate Lancet ii: 967
Bradley DW, McCaustland, Karen A et al (1985)
Post-transfusion non-A, non-B hepatitis in chimpanzees.
Physicochemical evidence that the tubule-forming agent is
a small, enveloped virus Gastroenterology 88: 773–779
Brady MT, Milam JD, Anderson DC et al (1984) Use of
deglycerolized red blood cells to prevent posttransfusion
infection with cytomegalovirus in neonates J Infect Dis
150: 334 –339
Braude AJ, Sandford JP, Bartlett JE et al (1952) Effects and
clinical significance of bacterial contaminants in transfused
blood J Lab Clin Med 39: 902
Brecher ME, Means N, Jere CS et al (2001) Evaluation of an
automated culture system for detecting bacterial
contam-ination of platelets: an analysis with 15 contaminating
organisms Transfusion 41: 477–482
Brecher ME, Hay SN, Rothenberg SJ (2004) Validation of
BacT/ALERT plastic culture bottles for use in testing of
whole-blood-derived leukoreduced
platelet-rich-plasma-derived platelets Transfusion 44: 1174 –1178
Brennan MT, Runganga J, Barbara JAJ et al (1992) The
prevalence of anti-HTLV in North London blood donors.
HTLV Symposium (Abstract), Montpellier, France
Bresters D, Mauser-Bunschoten EP, Reesink HW et al (1993)
Sexual transmission of hepatitis C virus Lancet 342: 210 –
211
Briggs M, Fox J, Tedder RS (1988) Age prevalence of antibody
to human herpes virus 6 (Letter) Lancet i: 1058–1059
Brodine SK, Kaime EM, Roberts C et al (1993) Simultaneous
confirmation and differentiation of human T-lymphotropic
virus types I and II infection by modified Western blot
containing recombinant envelope proteins Transfusion
33: 925–929
Brouwer J, Nevens F, Kleter G et al (1993) Which hepatitis
patient will benefit from interferon? Multivariate analysis
of 350 patients treated in a Benelux multicentre study
J Hepatol 18 (Suppl 1): S10
Brown KE, Hibbs JR, Gallinella G et al (1994) Resistance to
parvovirus B19 infection due to lack of virus receptor
(erythrocyte P antigen) N Engl J Med 330: 1192–1196
Brown P, Preece MA, Will RG (1992) Friendly fire in
medicine: hormones, homografts and Creutzfeld-Jacob
disease Lancet 340: 24 –27
Brown P, Preece M, Brandel JP et al (2000) Iatrogenic
Creutzfeldt-Jakob disease at the millennium Neurology
55: 1075–1081
Brown T, Anaud A, Ritchie JP et al (1984) Intrauterine
par-vovirus infection in pregnancy and hydrops fetalis Lancet
ii: 1033–1034
Bruce-Chwatt LJ (1972) Blood transfusion and tropical
disease Trop Dis Bull 69: 825
Bruce-Chwatt LJ (1974) Transfusion malaria Bull WHO 50: 337
Bruce-Chwatt LJ (1982) Transfusion malaria revisited Trop Dis Bull 79: 827–840
Bruce-Chwatt LJ (1985) Transfusion associated parasitic infections In: Infection, Immunity and Blood Transfusion.
RY Dodd, LF Barker (ed.) New York: Alan R Liss,
pp 101–125
Bruce ME, Will RG, Ironside JW et al (1997) Transmissions
to mice indicate that ‘new variant’ CJD is caused by the BSE agent Nature 389: 498–501
Brucker G, Brun-Vezinet F, Rosenheim M et al (1987) HIV-2
infection in two homosexual men in France Lancet i: 223 Brun-Vezinet F, Rey MA, Katlama C (1987) Lymphadenopathy-associated virus type 2 in AIDS and AIDS related complex Clinical and virological features in four patients Lancet i: 128–132
Bruneau C, Perez P, Chassaigne M et al (2001) Efficacy of a
new collection procedure for preventing bacterial ination of whole-blood donations Transfusion 41: 74 – 81 Bryant ML, Ratner L (1990) Myristoylation-dependent repli- cation and assembly of human immunodeficiency virus 1 Proc Natl Acad Sci USA 87: 523 –527
contam-Busch MP, Alter HJ (1995) Will human immunodeficiency virus p24 antigen screening increase the safety of the blood supply and, if so, at what cost? Transfusion 35: 536 –539
Busch MP, Donegan E, Stuart M et al (1990a) Donor HIV-1
p24 antigenaemia and course of infection in recipients Transfusion Safety Study Group Lancet 335: 1342
Busch MP, Taylor PE, Lenes BA et al (1990b) Screening of
selected male blood donors for p24 antigen of human immunodeficiency virus type 1 N Engl J Med 323: 1308–1312
Busch MP, Young MJ, Samson SM et al (1991a) Risk of
human immunodeficiency virus (HIV) transmission by blood transfusions before the implementation of HIV-1 antibody screening The Transfusion Safety Study Group Transfusion 31: 4–11
Busch MP, Eble BE, Khayam-Bashi H et al (1991b)
Evaluation of screened blood donations for human immunodeficiency virus type 1 infection by culture and DNA amplification of pooled cells N Engl J Med 325: 1–5
Busch MP, Korelitz SH, Kleinman SR et al (1995) Declining
value of alanine aminotransferase in screening of blood donors to prevent posttransfusion hepatitis B and C virus infection Transfusion 35: 903–910
Busch MP, Kleinman SH, Jackson B et al (2000) Committee
report Nucleic acid amplification testing of blood donors for transfusion-transmitted infectious diseases: Report
of the Interorganizational Task Force on Nucleic Acid Amplification Testing of Blood Donors Transfusion 40: 143–159
Trang 36Cable RG, Leiby DA (2003) Risk and prevention of
transfusion-transmitted babesiosis and other tick-borne
diseases Curr Opin Hematol 10: 405–411
Cahill KM, Benach JL, Reich LM et al (1981) Red cell
exchange: treatment of babesiosis in a splenectomized
patient Transfusion 21: 193–198
Candotti D, Etiz N, Parsyan A et al (2004) Identification and
characterization of persistent human erythrovirus infection
in blood donor samples J Virol 78: 12169–12178
Cardigan R, Allford S, Williamson L (2002) Levels of
von Willebrand factor-cleaving protease are normal in
methylene blue-treated fresh-frozen plasma Br J Haematol
117: 253–254
Carman WF, Jacyna MR, Hadziyannis S et al (1989)
Mutation preventing formation of hepatitis e antigen in
patients with chronic hepatitis B virus infection Lancet ii:
588–591
Carman WF, Zanetti AR, Karayiannis P et al (1990) Vaccine
induced escape mutant of hepatitis B virus Lancet ii:
325–329
Carman WF, Fagan EA, Hadziyannis S et al (1991)
Association of a precore variant of hepatitis B virus with
fulminant hepatitis Hepatology 14: 219–222
Casaccia P, Ladogana A, Xi YG et al (1989) Levels of
infec-tivity in the blood throughout the incubation period of
hamsters peripherally injected with scrapie Arch Virol
108: 145–149
Casewell MW, Slater NG, Cooper JE (1981) Operating
theatre water-baths as a cause of pseudomonas septicaemia.
J Hosp Infect 2: 237–247
CDC (1982) Pneumocystis carinii pneumonia among persons
with haemophilia A MMWR 31: 365–367
CDC (1983) Prevention of acquired immune deficiency
syn-drome (AIDS): Report of interagency recommendations.
MMWR 32: 101–103
CDC (1984) Delta hepatitis – Massachusetts MMWR 33:
493–494
CDC (1987a) Survey of non-US haemophilia treatment
centers for HIV seroconversion following therapy with
heat treated factor concentrates MMWR 36: 121–124
CDC (1987b) Human immunodeficiency virus infection in
the United States MMWR 36: 801–804
CDC (1990) Human T-lymphotropic virus type I screening in
volunteer blood donors – United States, 1989 MMWR 39:
915–924
CDC (1991) HIV/AIDS Surveillance Report Atlanta:
CDC (1992) HIV/AIDS Surveillance report MMWR 9:
CDC (1997) HIV/AIDS Surveillance Report 8(2): 10
Cerisola JA, Rabinovich A, Alvarez M (1972) Enfermedad de
Chagas y la transfusión de sangre Bol Of Sanit Panam 73:
203–206
Cervenakova L, Yakovleva O, McKenzie C et al (2003)
Similar levels of infectivity in the blood of mice infected
with human-derived vCJD and GSS strains of transmissible spongiform encephalopathy Transfusion 43: 1687–1694 Chambers RW, Foley HT, Schmidt PJ (1969) Transmission
of syphilis by fresh blood components Transfusion 9: 32–34
Chen Y-C, Wang C-H, Su I-J et al (1989) Infection of human
T-cell leukemia virus type Land Development of human T-cell leukemia/lymphoma in patients with hematologic neoplasms: a possible linkage to blood transfusion Blood 74: 388–394
Cherubin CE, Prince AM (1971) Serum hepatitis specific gen (SH) in commercial and volunteer sources of blood Transfusion 11: 25
anti-Chin S, Williams B, Cottlieb P et al (1995) Virucidal short
wavelength ultraviolet light treatment of plasma and Factor VIII concentrate: protection of proteins by antioxidants Blood 86: 4331–4336
Chiu EKW, Yuen KY, Lie AKW et al (1994) A
pro-spective study of symptomatic bacteremia following platelet transfusion and its management Transfusion 34: 950–954
Choo Q-L, Kuo G, Weiner AJ et al (1989) Isolation of a
cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome Science 244: 359–361
Choo Q-L, Richmond KH, Han JH (1991) Genetic tion and diversity of the hepatitis C virus Proc Natl Acad Sci USA 88: 2451–2455
organiza-Chou SW (1990) Differentiation of cytomegalovirus strains
by restriction analysis of DNA sequences amplified from clinical specimens J Infect Dis 162: 738–742
Choudhury N, Jolly JG, Mahajan RC et al (1991) Malaria
screening to prevent transmission by transfusion: an ation of techniques Med Lab Sci 48: 206–211
evalu-Choudhury N, Murthy PK, Chatterjee RK et al (2003)
Transmission of filarial infection through blood fusion Indian J Pathol Microbiol 46: 367–370
trans-Christiansen CB, Jessen TE, Nielsen C et al (1996) False
negative anti-HIV-1/HIV-2 ELISAs in acute HIV-2 infection Vox Sang 70: 144–147
Chung H-L, Chow H-K, Lu J-P (1948) The first two cases of transfusion kala-azar Chinese Med J 66: 325–326 Clark J, Saxinger C, Gibbs WN (1985) Seroepidemiologic studies of human T-cell leukemia/lymphoma virus type I in Jamaica Int J Cancer 36: 37–41
Clark SJ, Saag MS, Decker WD et al (1991) High titers of
cytopathic virus in plasma of patients with symptomatic primary HIV-1 infection N Engl J Med 324: 954 –960
Clavel F, Guetard D, Brun-Vezinet F et al (1986) Isolation of
a new human retrovirus from West African patients with AIDS Science 233: 343–346
Cohen BJ, Mortimer PP, Pereira MS (1983) Diagnostic assays with monoclonal antibodies for the human serum parvovirus-like virus (SPLV) J Hyg 91: 113–130
Trang 37Cohen ND, Munoz AR (1989) Transmission of retroviruses
by transfusion of screened blood in patients undergoing
cardiac surgery N Engl J Med 320: 1172–1176
Collinge J, Rossor M (1996) A new variant of Prion disease.
Lancet 347: 1996–1997
Collinge J, Sidle KCL, Meads J et al (1996) Molecular
ana-lysis of prion strain variation and the aetiology of ‘new
variant’ CJD Nature (Lond) 383: 685–690
Colombo M, Mannucci PM, Carnelli V et al (1985)
Transmission of non-A, non-B hepatitis by heat-treated
Factor VIII concentrate Lancet ii: 1–4
Conry-Cantilena C, VanRaden M, Gibble J et al (1996)
Routes of infection, viremia, and liver disease in blood
donors found to have hepatitis C virus infection N Engl
J Med 334: 1691–1696
Contreras M, Barbara JAJ, Anderson CC et al (1991) Low
incidence of non-A, non-B post-transfusion hepatitis in
London confirmed by hepatitis C virus serology Lancet i:
753 –757
Corash L, Lin L, Wiesehahn G (1992) Use of
8-methoxypso-ralen and longwavelength ultraviolet irradiation for
decon-tamination of platelet concentrates Blood Cells 18: 57–74
Corrao G, Arico S (1998) Independent and combined action
of hepatitis C virus infection and alcohol consumption on
the risk of symptomatic liver cirrhosis Hepatology 27:
914 –919
Cossart YE, Field AM, Cant B et al (1975) Parvovirus-like
particles in human sera Lancet 1: 72–73
Cossen C, Hagens D, Fukuchi R et al (1992) Comparison of
six commercial human T-cell lymphotropic virus type I
(HTLV-I) enzyme immunoassay kits for detection of
anti-body to HTLV-I and -II J Clin Microbiol 30: 724–725
Council of Europe (1995) Guide to the Preparation, Use
and Quality Assurance of Blood Components, 9th edn.
Strasbourg: Council of Europe Press, p 45
Coursaget P, Yvonnet B, Chotard J et al (1987) Age- and
sex-related study of hepatitis B virus chronic carrier state
in infants from an endemic area (Senegal) J Med Virol 22:
1–5
Craig AS, Schaffner W (2004) Clinical practice Prevention of
hepatitis A with the hepatitis A vaccine N Engl J Med 350:
476 – 481
Cummins D, Amin S, Halil O et al (1995) Visceral
leishmania-sis after cardiac surgery Arch Dis Child 72: 235–236
Curran JW, Lawrence DN, Jaffe H et al (1984) Acquired
immunodeficiency syndrome (AIDS) associated with
trans-fusions N Engl J Med 310: 69–75
Curran JW, Jaffe HW, Peterman TA et al (1985)
Epidemio-logic aspects of acquired immunodeficiency syndrome
(AIDS) in the United States: cases associated with
trans-fusions In: Infection, Immunity and Blood Transfusion.
RY Dodd, LF Barker (eds) New York: Alan R Liss,
pp 259–269
Custer B, Tomasulo PA, Murphy EL et al (2004) Triggers for
switching from minipool testing by nucleic acid technology
to individual-donation nucleic acid testing for West Nile virus: analysis of 2003 data to inform 2004 decision making Transfusion 44: 1547–1554
Cuthbertson B (1991) Viral contamination of human plasma and procedures for preventing virus transmission by plasma products In: Blood Separation and Plasma Fractionation.
JR Harris (ed.) New York: Wiley-Liss, pp 385– 435
Dalgleish AG, Beverley PCL, Clapham PR et al (1984) The
CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus Nature (Lond) 312: 763–767
Damen M, Zaaijer HL, Cuypers HTM et al (1995) Reliability
of the third-generation recombinant immunoblot assay for hepatitis C virus Transfusion 35: 745–749
Damond F, Worobey M, Campa P et al (2004) Identification
of a highly divergent HIV type 2 and proposal for a change
in HIV type 2 classification AIDS Res Hum Retroviruses 20: 666–672
Dane DS, Cameron CH, Briggs M (1970) Virus-like particles
in serum of patients with Australia-antigen associated hepatitis Lancet i: 695–698
Dardare N, Platz MS (2002) Binding affinities of commonly employed sensitizers of viral inactivation Photochem Photobiol 75: 561–564
Davoli EH, Lipson SM, Match ME et al (1999) Evaluation of
the PrimeCapture CMV DNA detection plate system for detection of cytomegalovirus in clinical specimens J Clin Microbiol 37: 2587–2591
Dawson GJ, Chan KH, Cabal CM et al (1992) Solid-phase
enzyme-linked immunosorbent assay for hepatitis E virus IgG and IgM antibodies utilising recombinant antigens and synthetic peptides J Virol Methods 38: 174 –186 Deinhardt F, Zuckerman AJ (1985) Immunization against hepatitis B: report on a WHO meeting on viral hepatitis in Europe J Med Virol 17: 209–217
Deinhardt F, Holmes AW, Capps RB et al (1967) Studies on
the transmission of human viral hepatitis to marmoset monkeys: transmission of disease: serial passages and description of liver lesions J Exp Med 125: 673 – 687
Delwart EL, Kalmin ND, Jones TS et al (2004) First report
of human immunodeficiency virus transmission via an RNA-screened blood donation Vox Sang 86: 171–177
Deng HK, Lin R, Ellmeyer W et al (1996) Identification of a
major co-receptor for primary isolates of HIV-1 Nature (Lond) 381: 661–666
Denis F, Ranger-Rogez S, Nicot T (1996) Les nouveaux virus des hepatites Transfusion Clin Biol 1: 19–25
Deroff P, Regner M, Simitzis AM et al (1982) Screening of
blood donors likely to transmit falciparum malaria Rev Fr Transfusion Immunohematol 25: 3 –10
Desmyter J (1986) AIDS and blood transfusion Vox Sang 51 (Suppl.): 1, 21
Trang 38De Schryver A, Meheus A (1990) Syphilis and blood
trans-fusion: a global perspective Transfusion 30: 844 –847
De Silva M, Contreras M, Barbara J (1988) Two cases of
transfusion-transmitted malaria (TTM) in the UK (letter).
Transfusion 28: 86
Devetten MP, Liu JM, Ling V et al (1997) Paroxysmal
nocturnal hemoglobinuria: new insights from murine
Pig-a-deficient hematopoiesis Proc Assoc Am Phys 109:
99–110
Dharmasena F, Gordon-Smith EC (1986) Transmission of
malaria by bone marrow transplantation Transplantation
42: 228
Dienstag JL, Purcell HR, Alter HJ et al (1977) Non-A, non-B
post-transfusion hepatitis Lancet 1: 560–562
Dodd RY (1991) Donor testing and its impact on
transfusion-transmitted infection In: Transfusion Transmitted Infections.
DM Smith, RY Dodd (eds) Chicago: ASCP, pp 243–269
Dodd RY, Notari EP, Stramer SL (2002) Current prevalence
and incidence of infectious disease markers and estimated
window-period risk in the American Red Cross blood
donor population Transfusion 42: 975–979
Dodelet VC, Cashman NR (1998) Prion protein expression in
human leukocyte differentiation Blood 91: 1556 –1561
Donegan E, Lenes BA, Tomasulo PA et al (1990a)
Trans-mission of HIV-1 by component type and duration of shelf
storage before transfusion Transfusion 30: 851–852
Donegan E, Stuart M, Niland JC et al (1990b) Infection with
human immunodeficiency virus type 1 (HIV-1) among
recipients of antibody-positive blood donations Ann
Intern Med 113: 733–739
Donegan E, Pell P, Lee H (1992) Transmission of human
T-lymphotropic virus type I by blood components from a
donor lacking anti-p24: a case report Transfusion 32:
68–71
Donegan E, Lee H, Operskalski EA et al (1994) Transfusion
transmission of retroviruses: human T-lymphotropic virus
types I and II compared with human immunodeficiency
virus type 1 Transfusion 34: 478– 483
Dow BC, MacVarish I, Barr A et al (1980) Significance of
tests for HBeAg and anti-HBe in HBsAg positive blood
donors J Clin Pathol 33: 1106 –1109
Dragic T, Litwin V, Allaway GP et al (1996) HIV entry into
CD4+ cells is mediated by the chemokine receptor for
CC-CKR-5 Nature (Lond) 381: 667– 673
Drew WL, Miner RC (1982) Transfusion-related
cytome-galovirus infection following noncardiac surgery JAMA
247: 2389–2391
Drew WL, Tegtmeier G, Alter HJ et al (2003) Frequency and
duration of plasma CMV viremia in seroconverting blood
donors and recipients Transfusion 43: 309–313
Eastlund T, Persing D, Mathiesen D et al (1999) Human
granulocytic ehrlichiosis after red cell transfusion
Trans-fusion 39 (Suppl.): 117S
Eddleston A (1990) Modern vaccines Hepatitis Lancet i: 1142–1145
Engels EA, Eastman H, Ablashi DV et al (1999) Risk of
transfusion-associated transmission of human herpesvirus
8 J Natl Cancer Inst 91: 1773 –1775
Esteban JI, Shih JW, Tai CC et al (1985) Importance of
western blot analysis in predicting infectivity of HTLV-III/LAV positive blood Lancet 2: 1083–1086
anti-Esteban JI, Estban R, Viladomiu L et al (1989) Hepatitis C
virus antibodies among risk groups in Spain Lancet ii:
294 –297
Esteban JI, Gonzalez A, Hernandez M et al (1990)
Evaluation of antibodies to hepatitis C virus in a study of transfusion associated hepatitis N Engl J Med 323: 1107–1112
Evatt B, Austin H, Barnhart E et al (1998) Surveillance for
Creutzfeldt-Jakob disease among persons with hemophilia Transfusion 38: 817–820
Evatt BL, Stein SF, Francis DP et al (1983) Antibodies to
human T-cell leukemia virus-associated membrane gens in hemophiliacs: evidence for infection before 1980 Lancet ii: 698
anti-Evatt BL, Ramsey RB, Lawrence DN et al (1984) The
acquired immunodeficiency syndrome in patients with hemophilia Ann Intern Med 100: 499
Evatt BL, Gomperts ED, McDougal JS et al (1985)
Coincidental appearance of LAV/HTLV-III antibodies in hemophiliacs and the onset of the AIDS epidemic N Engl
J Med 312: 483–486 Fagan EA, Williams R (1990) Fulminant viral hepatitis Br Med Bull 46: 462–480
Fang CT, Tobler LH, Haesche C et al (2003) Fluctuation
of HCV viral load before seroconversion in a healthy volunteer blood donor Transfusion 43: 541–544
Farci P (2001) Hepatitis C virus The importance of viral heterogeneity Clin Liver Dis 5: 895–916
Farci P, Alter HJ, Govindaragan S et al (1993) Lack of
pro-tective immunity against reinfection with hepatitis C virus Science 258: 135–140
Fast LD (2003) The effect of exposing murine splenocytes to UVB light, psoralen plus UVA light, or gamma-irradiation
on in vitro and in vivo immune responses Transfusion 43:
576–583
Fast LD, DiLeone G, Edson CM et al (2002) PEN110
treat-ment functionally inactivates the PBMNCs present in RBC units: comparison to the effects of exposure to gamma irra- diation Transfusion 42: 1318 –1325
Fast LD, Semple JW, DiLeone G et al (2004) Inhibition of
xenogeneic GVHD by PEN110 treatment of donor human PBMNCs Transfusion 44: 282–285
Feng Y, Broder CC, Kennedy PE et al (1996) HIV entry
co-factor; functional cDNA cloning of a seven-transmembrane
G protein-coupled receptor Science 272: 872–877
Trang 39Fernandez MN, Daza RM, Orden B et al (1981) Anticuerpos
a Brucela en donantes de sangre Sangre 26: 360–366
Ferrero S, Lungaro P, Bruzzone BM et al (2003) Prospective
study of mother-to-infant transmission of hepatitis C virus:
a 10-year survey (1990–2000) Acta Obstet Gynecol Scand
82: 229–234
le Fichoux Y, Quaranta JF, Aufeuvre JP et al (1999)
Occurrence of Leishmania infantum parasitemia in
asymp-tomatic blood donors living in an area of endemicity in
southern France J Clin Microbiol 37: 1953–1957
Fiebig EW, Wright DJ, Rawal BD et al (2003) Dynamics of
HIV viremia and antibody seroconversion in plasma
donors: implications for diagnosis and staging of primary
HIV infection AIDS 17: 1871–1879
Files JC, Case CJ, Morrison FS (1984) Automated
erythro-cyte exchange in fulminant falciparum malaria Ann Intern
Med 100: 396–397
Flanagan P, McAlpine L, Ramskill SJ et al (1995) Evaluation
of a combined HIV-1/2 and HTLV-I/II assay for screening
blood donors Vox Sang 68: 220–224
Fleming AF (1990) AIDS in Africa In: Haematology in HIV
Disease C Costello (ed.) Baillières Clinical Haematology
3: 177–206
Foster KM (1966) Post-transfusion mononucleosis Aust Ann
Med 15: 305
Fradkin JE, Schonberger LB, Mills JL et al (1991)
Creutzfeldt-Jakob disease in pituitary growth hormone
recipients in the United States JAMA 265: 880 –884
Freeman R, Gould FK, McMaster A (1990) Management of
cytomegalovirus antibody negative patients undergoing
heart transplantation J Clin Pathol 43: 373–376
Frickhofen N, Abkowitz JL, Safford M et al (1990)
Per-sistent B19 parvovirus infection in patients infected
with human immunodeficiency virus type 1 (HIV-1): a
treatable cause of anemia in AIDS Ann Intern Med 113:
926–933
Gaeta GB, Stornaiuolo G, Precone DF et al (2003)
Epidemiological and clinical burden of chronic hepatitis B
virus/hepatitis C virus infection A multicenter Italian
study J Hepatol 39: 1036–1041
Gallarda JL, Henrard DR, Liu D et al (1994) Early detection
of antibody to human immuno-deficiency virus type 1 by
using an antigen conjugate immunoassay correlated with
the presence of IgM antibody J Clin Microbiol 30:
2379–2384
Gallo RC (2003) A journey with T cells, primate/human
retroviruses and other persisting human T-cell tropic
viruses Rev Clin Exp Hematol 7: 329–335
Gallo RC, De-The GB, Ito Y (1981) Kyoto workshop on
some specific recent advances in human tumor virology.
Cancer Res 41: 4738–4739
Gallo RC, Salahuddin SZ, Popovic M et al (1984) Frequent
detection and isolation of cytopathic retroviruses
(HTLV-III) from patients with AIDS and at risk for AIDS Science 224: 500–503
Ganem D, Prince AM (2004) Hepatitis B virus infection – natural history and clinical consequences N Engl J Med 350: 1118–1129
Garretta M, Paris-Hamelin A, Vaismam A (1977) Syphilis
et transfusion Rév Fr Transfusion Immunohematol 2: 287–308
Gerber P, Walsh JH, Rosenblum EN et al (1969) Association
of EB-virus infection with the post-perfusion syndrome Lancet i: 593
Gessain A, Francis H, Sonan T et al (1986) HTLV-1 and
tropical spastic paraparesis in Africa Lancet ii: 698
Ghani AC, Donnelly CA, Ferguson NM et al (2003) Updated
projections of future vCJD deaths in the UK BMC Infect Dis 3: 4
Ghany MG, Kleiner DE, Alter H et al (2003) Progression
of fibrosis in chronic hepatitis C Gastroenterology 124: 97–104
Gjerset GF, Clements MJ, Counts RB et al (1991) Treatment
type and amount influenced human immunodeficiency virus seroprevalence of patients with congenital bleeding disorders Blood 78: 1623–1627
Glazer JP, Friedman HM, Grossman RA et al (1979) Live
cytomegalovirus vaccination of renal transplant ates: a preliminary trial Ann Intern Med 91: 676–683
candid-Glynn SA, Kleinman SH, Schreiber GB et al (2000) Trends in
incidence and prevalence of major transfusion-transmissible viral infections in US blood donors, 1991 to 1996 Retrovirus Epidemiology Donor Study (REDS) JAMA 284: 229–235
Glynn SA, Smith JW, Schreiber GB et al (2001) Repeat
whole-blood and plateletpheresis donors: unreported deferrable risks, reactive screening tests, and response to incentive programs Transfusion 41: 736–743
Gocke DJ (1972) A prospective study of post-transfusion hepatitis The role of Australia antigen JAMA 219: 1165
Goedert JJ, Eyster ME, Lederman MM et al (2002)
End-stage liver disease in persons with hemophilia and transfusion-associated infections Blood 100: 1584 –1589 Goldman MB, Blajchman MA (1991) Blood product-associated bacterial sepsis Transfusion Med Rev 5: 73–83
Goldman M, Roy G, Frechette N et al (1997) Evaluation
of donor skin disinfection methods Transfusion 37: 309– 312
Gong J, Rawal BD, Högman CF et al (1994) Complement killing of Yersinia enterocolitica and retention of the bac-
teria by leucocyte removal filters Vox Sang 66: 166 –170 Goodrich RP (2000) The use of riboflavin for the inactivation
of pathogens in blood products Vox Sang 78 (Suppl 2): 211–215
Goodrich RP, Yerram NR, Tay-Goodrich BH et al (1994)
Selective inactivation of viruses in the presence of human
Trang 40platelets: UV sensitization with psoralen derivatives Proc
Natl Acad Sci USA 91: 5552–5556
Gout O, Baulac M, Gessain A et al (1990) Rapid
develop-ment of myelopathy after HTLV-1 infection acquired by
transfusion during cardiac transplantation N Engl J Med
322: 383–388
Graan-Hentzen YC, Gratama JW, Mudde GC et al (1989)
Prevention of primary cytomegalovirus infection in
patients with hematologic malignancies by intensive white
cell depletion of blood products Transfusion 29: 757–760
Grady GF, Lee VA (1975) Prevention of hepatitis from
accid-ental exposure among medical workers N Engl J Med
293: 1067
Grant DB, Perinpanayagam MS, Shute PG et al (1960) A case
of malignant tertian (Plasmodium falciparum) malaria
after blood-transfusion Lancet ii: 469
Greene WC, Peterlin BM (2002) Charting HIV’s remarkable
voyage through the cell: Basic science as a passport to
future therapy Nature Med 8: 673–680
Griffiths PD (1984) Diagnostic techniques for
cytome-galovirus infection Clin Haematol 13: 631–644
Grogl M, Daugirda JL, Hoover DL et al (1993) Survivability
and infectivity of viscerotropic Leishmania tropica from
Operation Desert Storm participants in human blood
products maintained under blood bank conditions Am J
Trop Med Hyg 49: 308–315
Grumet FC (1984) In International Forum:
Transfusion-transmitted CMV infections Vox Sang 46: 387– 414
Guerrero IC, Weniger BC, Schultz MG (1983) Transfusion
malaria in the United States 1972–1981 Ann Intern Med
99: 221–226
Högman CF, Gong J, Erikkson L et al (1991) Transfusion
transmitted bacterial infection (TTBI) (Abstracts) 2 nd
Congr W Hong Kong: Pacific Region Int Soc Blood Transf
Högman CF, Gong J, Hamkraens A et al (1992) The role of
leucocytes in the transmission of Yersinia enterocolitica
with blood components Transfusion 32: 654 – 657
Habibi B, Kleinknecht D, Vachon F et al (1973) Le choc
transfusionnel par contamination bacté‚ rienne du sang
conservé Analyse de 25 observations Rev Fr Transfusion
16: 41
Haditsch M, Binder L, Gabriel C et al (1994) Yersinia
enterocolitica septicemia in autologous blood transfusion.
Transfusion 34: 907–909
Haimowitz MD, Hernandez LA, Herron RM Jr (2005) A
blood donor with bacteremia Lancet 365: 1596
Halkier-Sørensen, L, Kragballe K, Nedergaard ST et al.
(1990) Lack of transmission of Borrelia burgdorferi by
blood transfusion Lancet i: 550
Hall AJ, Alveyn CG, Winter PD et al (1988) Mortality of
hepatitis B-positive blood donors in England and Wales.
In: Viral Hepatitis and Liver Disease AJ Zuckerman (ed.).
New York: Alan R Liss, pp 192–194
Hambleton J, Wages D, Radu-Radulescu L et al (2002)
Pharmacokinetic study of FFP photochemically treated with amotosalen (S-59) and UV light compared to FFP in healthy volunteers anticoagulated with warfarin Trans- fusion 42: 1302–1307
Hamers FF, Downs AM (2004) The changing face of the HIV epidemic in western Europe: what are the implications for public health policies? Lancet 364: 83–94
Hancock JS, Taylor RN, Johnson CE et al (1993) Quality of
laboratory performance in testing for human deficiency virus type I antibodies: identification of variables associated with laboratory performance Arch Pathol Lab Med 117: 1148–1155
immuno-Hanson CV (1992) Photochemical inactivation of viruses with psoralenes: an overview Blood Cells 18: 7–25
Hanto DW, Gajl-Peczalska KJ, Frizzera G et al (1983)
Epstein-Barr virus (EBV)-induced polyclonal and clonal B-cell lymphoproliferative diseases occurring after renal transplantation Ann Surg 198: 356–369
mono-Harris DR, Gonin R, Alter HJ et al (2001) The relationship
of acute transfusion-associated hepatitis to the ment of cirrhosis in the presence of alcohol abuse Ann Intern Med 134: 120–124
develop-Harrison TJ, Zuckerman AJ (1992) Variants of hepatitis B virus Vox Sang 63: 161–167
Harrison TJ, Bal V, Wheeler EG et al (1985) Hepatitis B
virus DNA and e antigen in serum from blood donors in the United Kingdom positive for hepatitis B surface antigen BMJ 290: 663 – 664
Hartley TM, Khabbaz RF, Cannon RO et al (1990)
Characterization of antibody reactivity to human T-cell lymphotropic virus types I and II using immunoblot and radioimmunoprecipitation assays J Clin Microbiol 28:
646 – 650
Hartley TM, Malone GE, Khabbatz RF et al (1991)
Evalu-ation of a recombinant T-cell lymphotropic virus type I (HTLV-I) p21e antibody detection enzyme immunoassay
as a supplementary test in HTLV-I/II antibody testing rithms J Clin Microbiol 29: 1125–1129
algo-Hartmann O, Schone R (1942) Syfilis overfort ved fusion Nord T Milit-Med 45: 1
blodtrans-Hawkins A, Davidson F, Simmonds P (1997) Comparison of plasma virus loads among individuals infected with hepatitis
C virus (HCV) genotypes 1, 2, and 3 by quantiplex HCV RNA assay versions 1 and 2, Roche Monitor assay, and
an in-house limiting dilution method J Clin Microbiol 35: 187–192
Hay CR, Preston FE, Triger DR et al (1985) Progressive liver
disease in haemophilia: an understated problem? Lancet 1: 1495–1498
Hayakawa F, Imada K, Towatari M et al (2002)
Life-threatening human parvovirus B19 infection transmitted by intravenous immune globulin Br J Haematol 118: 1187–1189