1-s2.0-S0268960X01901511-main

download 1-s2.0-S0268960X01901511-main

of 7

Transcript of 1-s2.0-S0268960X01901511-main

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    1/15

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    2/15

    incompatible transfusions in which complement fixation

    results in formation of the C5b-9 membrane attack complex.

     Accordingly, only rarely do delayed reactions cause intravas-

    cular hemolysis with associated hemoglobinemia and hemo-

    globinuria. Cytokines, which play a central role in the

    pathophysiology of ABO incompatible reactions, may also

    contribute to DHTR.12 This is particularly true in more severe

    delayed reactions in which elevations of inflammatory medi-ators like tumor necrosis factor, IL-6, and IL-8 have been

    described.13

     Antibodies most commonly implicated in DHTR include

    those directed against Kidd (Jk), Rh (E, C, c), Kell (K), and

    Duffy (Fy) blood group antigens (Table 2).Anti-Jk a, in particu-

    lar, is often implicated in these reactions.14 Certain antibod-

    ies, such as those with Kidd and Duffy specificity, are more

    frequently identified in delayed hemolytic reactions in which 

    the transfused cells are clearly removed.15 Delayed serologic

    transfusion reactions are defined as the presence of a new 

    red cell antibody in a patient, without evidence of red cell

    removal.16 Patients with DHTR who require further transfu-

    sions should receive red cells that do not carry the associ-ated antigen, so-called ‘antigen negative’ units. Patients

    previously sensitized to red cell antigens must be clearly 

    identified in blood bank information systems to avoid subse-

    quent reactions caused by an anamnestic response to further 

    exposures. In addition, most blood bank standards demand

    that pre-transfusion testing of recently transfused or preg-

    nant patients be performed on samples obtained within 3

    days of a transfusion.This requirement is based on the fre-

    quent observation that these patients develop clinically sig-

    nificant red cell antibodies associated with immediate and

    delayed hemolysis.17 DHTR are particularly prevalent in

    patients with diseases that require frequent red cell transfu-

    sion.18

    Patients with sickle cell disease (SCD) are at a high risk of DHTR because of differences in red cell antigen frequency 

    between the donor population and the recipients. For exam-

    ple, black SCD patients are often Fy a and Fy b negative.This

    phenotype is uncommon in white blood donors.Thus, black 

    patients are very likely to be exposed to Fy positive blood,

    and thus, may develop anti-Fy antibodies.There are also sig-

    nificant differences in the Rh system between black and

     white individuals.Many centers attempt to avoid these prob-

    lems by providing ‘antigen matched’red cells when possible.For example, the recipients red cells are typed for Rh and

    Kell antigens,after which they receive units that do not carry 

    antigens that they do not possess. Finally, hemolytic transfu-

    sion reactions (HTR) can be particularly dangerous in

    patients with sickle cell disease – the ‘sickle cell HTR syn-

    drome’.19 These may exhibit the typical manifestations of an

    acute or delayed hemolytic transfusion reaction. In addition,

    patients will have symptoms suggestive of a sickle cell crisis,

    marked reticulocytopenia, and may develop a more severe

    anemia following transfusion than was previously present.

    However, serologic evaluation may not reveal a new red cell

    antibody or a newly positive DAT.20 Serologic studies are

    often complicated by the presence of other red cell antibod-ies that were evident before the transfusion.These patients

    may be very difficult to transfuse.

    FEBRILE NON-HEMOLYTIC TRANSFUSION REACTIONS

    Febrile non-hemolytic transfusion reactions (FNHTR) are

    most commonly encountered during transfusions of red

    cells, platelets, or plasma. They typically occur during the

    transfusion, but may present minutes or several hours after 

    the transfusion is completed.The frequency of febrile reac-

    tions is higher following transfusion of platelets (4% to

    30%)21 than of red cells (0.5%).Most FNHTRs are self-limited

    and not life-threatening. The most common signs include

    fever (> 1˚C elevation) and shaking chills, and these may be

    accompanied by nausea, vomiting, dyspnea, and hypoten-

    sion. Oxygen saturation may decrease during rigors, but

    should return to baseline as the reaction resolves.The sever-

    ity of symptoms can be related to number of leukocytes in

    the product and/or the rate of transfusion. Pre-medicating

    transfusion recipients with an antipyretic like aceta-

    minophen [Paracetamol] may help to minimize FNHTR.

     Antihistamines are not useful in preventing or treating

    FNHTR and are not indicated unless there is a clear allergic

    component.Corticosteroids may also minimize FNHTRs,but

    these should be administered several hours before transfu-

    sion to be maximally effective.Severe rigors can be promptly 

    Perrotta and Snyder 

    70Blood Reviews (2001) 15 , 69–83   2001 Harcourt Publishers Ltd 

    Table 2 Antibodies commonly implicated in delayed

    hemolytic reactions

    Blood group system Specific antibody

    Kidd (Jk) Jk  a > Jk b

    Rh E > C > c

    Kell K

    Duffy (Fy) Fya

    Glycophorin S > s

    Table 1 Non-infectious complications of transfusion therapy

    Acute immune hemolytic reactions Metabolic disturbances

    Delayed extravascular hemolysis Air embolism

    Febrile non-hemolytic reactions Hypothermia

    Allergic reactions (urticarial, anaphylactic) Hypotensive reactions and ACE inhibitors

    Non-immune red cell hemolysis Red-eye syndrome

    Post-transfusion purpura Transfusion-associated sepsisCirculatory overload Transfusion-associated graft-versus-host disease

    Iron overload Transfusion-associated acute lung injury

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    3/15

    resolved with intravenous meperidine [Pethidine].22

    Intravenous corticosteroids can be considered, but these

     will not produce as rapid relief as meperidine. If symptoms

    are especially severe or do not resolve within 4–6 h,

    other causes of the fever should be considered, including

    transfusion-related sepsis.The transfusion should be stopped

     when a FNHTR develops and should not be restartedusing the causative donation/component.This is because the

    most common sign of an acute hemolytic reaction is also

    fever.

    FNHTR are likely related to interactions between a recipi-

    ent’s cytotoxic antibodies and HLA and/or leukocyte specific

    antigens on donor white blood cells. Formation of leukocyte

    antigen-antibody complexes results in complement binding

    and release of endogenous pyrogens like TNF-α, IL-1, and IL-

    6. Direct activity of various biological response modifiers

    including cytokines also appear to play a role in these reac-

    tions.23 For instance, FNHTR are seen in patients who have

    not been previously transfused or pregnant.24 The higher 

    incidence of febrile reactions with platelet transfusion may be related to the duration of storage. When platelets are

    prepared by the buffy coat method, there is a progressive

    increase in the relative risk of developing predominantly 

    febrile reactions as platelets are stored for their 5 day

    shelf-life.25 During platelet storage, there is continued elabo-

    ration of biologically active cytokines by residual white cells.

    Reaction rates are as much as double in 3 to 5 day-old

    platelets as compared to 1 or 2 day-old products.26 This find-

    ing is attributed to reactive substances contained in

    the plasma portion of platelet concentrates. In particular,

    a strong positive correlation between supernatant IL-1β

    and IL-6 levels and the frequency of febrile reactions has

    been observed. Similar associations between the durationof storage of non-filtered pooled platelet concentrates

    and febrile reactions has been described by other

    groups.27

    Patients with recurrent febrile reactions to platelet or red

    cell transfusions should receive leukoreduced (LR) products

    and pre-transfusion antipyretics. In countries which do not

    routinely leukocyte deplete all blood components,plasma is

    not filtered for white cell reduction, although significant

    numbers of white cells can be found in freshly frozen

    plasma.28 Third generation leukoreduction filters eliminate

    99.9% of the white cells found in a unit of blood.In addition

    to reducing the incidence of FNHTR, LR can also decrease

    transmission of cytomegalovirus29

    and alloimmunization toHLA antigens.30 Pre-storage leukoreduction of red cells can

    further reduce the likelihood of FNHTR as compared to bed-

    side filtration.31 LR decreases the incidence and severity of 

    FNHTR at least partly by minimizing production of cytokines

    by residual leukocytes during storage.In particular,IL-8 levels

    of platelets prepared by the platelet-rich plasma method and

    leukoreduced shortly after collection are significantly lower 

    than in non-LR PCs stored for 5 days.32 Prestorage leukore-

    duction of platelet concentrates, as compared to platelets

    leukoreduced at the bedside, does not appear to further 

    reduce the incidence of FTRs.33 Interestingly, both febrile

    nonhemolytic and allergic reactions can follow autologous

    blood transfusions.34

     ALLERGIC REACTIONS, URTICARIAL AND

     ANAPHYLACTIC

    Mild allergic reactions, triggered by exposure to soluble sub-

    stances in donor plasma, are common following plasma,

    platelet, and red cell transfusions. Typical cutaneous hyper-

    sensitivity reactions present as pruritis and/or urticaria in

    the absence of fever. Allergic reactions are classically IgEmediated and symptoms are attributed to histamine release.

    Symptoms and signs include pruritis, erythema, papular 

    rashes, and weals.Distinguishing allergic and febrile transfu-

    sion reactions can be difficult when urticarial symptoms are

    accompanied by low-grade fever.Treatment of mild allergic

    reactions consists of temporarily interrupting the transfusion

    and administering 25–50 mg diphenhydramine or other anti-

    histamines. In mild allergic reactions not associated with 

    fever or vasomotor instability, the transfusion can be contin-

    ued if the symptoms promptly resolve. If symptoms recur 

    after the transfusion is restarted, a new unit should be

    obtained. Pre-medicating patients with an antihistamine

    before blood transfusion may help to minimize milder aller-gic reactions.It is difficult to predict which patients are a risk 

    for allergic reactions, but in general, the allergic predisposi-

    tion of the recipient and/or donor may be important.35

    However, screening blood donors for a history of atopy does

    not seem warranted.36 Allergic reactions could also occur on

    exposure to drugs that a blood donor is taking prior to the

    donation.37

    Fortunately, severe anaphylactic reactions following

    blood transfusion are rare.Anaphylaxis is a severe, systemic

    reaction caused by the release of histamine and other bio-

    logic mediators.The most serious symptoms include laryn-

    geal edema, lower-airway obstruction, and hypotension.

    Reactions are IgE-mediated responses to plasma proteins.Severe reactions can occur in susceptible individuals on

    exposure to latex found in blood containers. Latex is not

    found within the plastic bag itself, however, there may be

    small amounts of latex in side arms,ports,or caps attached to

    the blood container.The supplier of the plastic container in

    question should know if latex was used in the manufacturing

    process. As in other allergic responses, symptoms are not

    dose-related and severe manifestations can occur following

    small exposures.Treatment of anaphylaxis includes prompt

    administration of epinephrine as epinephrine reverses the

    actions of histamine within minutes.Vasopressors and airway 

    support may be required. An H1-receptor antagonist and

    intravenous steroids can be given to reduce the risk of pro-tracted anaphylaxis.Washed red cells in which the residual

    donor plasma has been removed and replaced by saline may 

    benefit patients with repeated or severe allergic reactions.

    Plasma can be removed from platelet concentrates to mini-

    mize allergic reactions, but these procedures usually cause

    the loss of significant numbers of platelets.38 Red cells leuko-

    reduced by filtration do not prevent allergic reactions

    because they do not remove the offending soluble stimuli.

    The duration of platelet storage, which is related to febrile

    non-hemolytic reactions, is not related to increased allergic

    reactions.27 Individuals with congenital IgA deficiency may 

    develop class-specific antibodies to IgA. Upon exposure to

    IgA found in blood products,immediate generalized reactions

    Non-infectious transfusion reactions

    71 2001 Harcourt Publishers Ltd  Blood Reviews (2001) 15 , 69–83

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    4/15

    can occur. IgA-deficient patients who have had a severe ana-

    phylactic reaction should receive blood components that

    lack IgA.These must be obtained from larger blood centers

    that maintain a supply of products collected from IgA-defi-

    cient donors. If large volumes of red cells are required, they 

    should be thoroughly washed to remove as much residual

    plasma as possible.39

    Finally, allergic-type reactions candevelop in individuals who are exposed to ethylene oxide

    gas. Ethylene oxide is used to sterilize plastic equipment in

    dialysis and apheresis instruments. Rare individuals have

    developed IgE antibodies directed against ethylene oxide

    and, subsequently, acute hypersensitivity reactions during

    hemodialysis or platelet collection by apheresis.40

    NON-IMMUNE RED CELL HEMOLYSIS

    Red cell hemolysis occurs through a variety of mechanisms,

    many of which do not require activation of the immune sys-

    tem (Table 3). Most of these involve the physical and/or 

    chemical destruction of red cells. Many medications andintravenous fluids lyse red cells through an osmotic effect

    that allows entry of water into cells.Hypotonic fluids such as

    5% dextrose in water will cause gross hemolysis on exposure

    to red cells.41 Since no hemolysis occurs when red cells are

    mixed with normal saline, this is the only fluid that can be

    used in intravenous administration tubing during transfu-

    sion. Intravenous medications should never  be added to a

    unit of red cells. Life-threatening hemolysis may occur if 

    patients are accidentally given large infusions of water by 

    rapid intravenous injection. Red blood cell damage also may 

    result from either overheating or freezing blood.Thus, blood

    should be warmed only using devices that are specifically 

    designed for this purpose. Placing red cells in hot water baths or common microwave ovens is unacceptable and can

    produce significant hemolysis.42 Red cells are irreversibly 

    damaged when warmed above 50˚C. Accordingly, blood

     warmers do not heat blood above 42˚C when functioning

    properly. During refrigerated storage there is lysis of a small

    fraction of red cells.However, significant hemolysis occurs if 

    blood is inadvertently stored frozen without a cryopreserv-

    ing agent such as glycerol.

    Mechanical hemolysis can occur as red cells traverse pumps

    used in cardiac bypass surgery and when red cells are forced

    through a small-gauge needle or other narrow orifice using a

    pressure cuff.43 Factors that influence hemolysis include lumen

    diameter, pressure and velocity of the infused blood, shape of 

    the needle tip,and age of the red cell unit.44Thus,red cell dam-

    age is minimized by using larger diameter needles and slower 

    infusion rates.45 Significant reactions from mechanical hemoly-

    sis are uncommon but must be distinguished from immune or 

    intravascular hemolysis which has more life-threatening

    implications. Staff must be trained in the proper use of 

    equipment (pumps, catheters) used to transfuse blood. Of 

    practical concern, blood forced through commonly used

    leukocyte reduction filters can produce significant hemoly-

    sis.46 While bacterial contamination of red cells is reportedly 

    quite common (on the order of 1 in 2000 units collected),the incidence of septic reactions to red cells is rare (esti-

    mated 1 in 500,000 red cell transfusions).47 Red cell units

    that are contaminated by certain bacteria may appear grossly 

    hemolyzed. Transfusing such units may cause hemoglobin-

    uria, but these effects are often overshadowed by the fever,

    hypotension,and pain produced by endotoxins generated by 

    gram-negative bacteria (see ‘Transfusion associated sepsis’).

    Uncommonly, a blood donor may have an intrinsic red cell

    defect like glucose-6-phosphatase dehydrogenase (G6PD)

    deficiency that predisposes the cells to hemolysis. Blood

    donors are not routinely screened for these conditions.

    Patients transfused with G6PD-deficient red cells may 

    develop a mild transient hemolysis with an unconjugatedhyperbilirubinemia and increased lactate dehydrogenase.

    The degree of hemolysis can be exacerbated if a recipient is

    concurrently receiving medications like primaquine or nitro-

    furantoin.48

    Patients who receive hemolyzed blood may or may not

    develop the classic signs and symptoms of an immune

    hemolytic reaction.The degree of hemolysis encountered in

    stored red cells is usually not harmful to the recipient as free

    hemoglobin in small quantities is rapidly removed from

    plasma through mechanisms involving binding with hapto-

    globin and oxidation to methemoglobin. Frozen stored red

    cells contain glycerol to prevent hemolysis during the freez-

    ing process. Following thawing, however, these units willcontain some free hemoglobin and more importantly, glyc-

    erol. Glycerol content is decreased by washing the red cells

    and resuspending the cells in isotonic saline.49 If red cells are

    not adequately deglycerolized,they may lyse on contact with 

    plasma as water enters cells faster than glycerol can diffuse

    out of the cell. Transfusing large amounts of hemolyzed

    blood,however, can cause hypotension,shock, and renal dys-

    function. Laboratory evidence of non-immune hemolysis

    includes hemoglobinuria and plasma-free hemoglobin.

     Assuming that the pre-transfusion direct antiglobulin test

    (DAT, direct Coombs) was negative, the DAT should be nega-

    tive as immunoglobulins are not coating red cells.The DAT is

    usually reactive following an immune-mediated hemolytic

    reaction in which recipient red cell antibodies bind to

    incompatible transfused donor red cells. Depending on the

    degree of hemolysis and the etiology of the hemolysis, treat-

    ment would include measures taken following an acute

    intravascular hemolytic reaction caused by an ABO incom-

    patible blood transfusion.Vital signs should be closely moni-

    tored. Cardiac and airway support are provided, and urine

    output is maintained with a saline diuresis with or without a

    loop diuretic. Dialysis should be considered in patients with 

    renal failure. Biological response modifiers including proin-

    flammatory cytokines (IL-1, TNF-α ), chemokines (IL-8) and

    complement fragments (C3a, C5a) play a role in the patho-

    physiology of immune hemolytic reactions.50 However, their 

    role in non-immune mediated hemolysis has not been

    Perrotta and Snyder 

    72Blood Reviews (2001) 15 , 69–83   2001 Harcourt Publishers Ltd 

    Table 3 Causes of non-immune red cell hemolysis

    Over warming of red cell unit

    Infusing larger volumes of older stored red cell units

    Adding incompatible intravenous fluids or drugs to red cells

    Improper freezing and thawing units of red cells

    Mechanical disruption of red cells by extracorporeal instruments

    Bacterial contamination of red cells

    Pre-existing red cell membrane or enzyme defect in the donor

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    5/15

    extensively studied. Most importantly, non-immune red cell

    hemolysis can be prevented by adhering to proper procedures

    for collecting,processing,storing,and infusing blood products.

    POST-TRANSFUSION PURPURA

    Post-transfusion purpura (PTP) is a rare complication of blood transfusion that results in profound thrombocytope-

    nia.51 It is characterized by acute thrombocytopenia (platelet

    count < 10,000/ µl;10 × 109 /l) occurring 5–10 days following

    red cell transfusion.52,53 PTP is often not immediately recog-

    nized because of the interval between the transfusion and

    the onset of thrombocytopenia. PTP is considered an

    immune thrombocytopenia in which anti-platelet antibodies,

    most often with specificity to HPA-la (PL AI ), are identified in

    the recipient.54  Antibodies directed against other broad

    platelet antigens like GPIIb/IIIa are also observed,55 as are

    multiple antibody specificities.56 Most patients are women

     who have been sensitized to platelets through pregnancies

    as described in one of the larger series of cases.57

    Sensitization can also occur in men,presumably through pre-

     vious blood exposure.58,59 The specificity of the platelet anti-

    bodies can be determined by platelet-ELISA and monoclonal

    antibody-specific immobilization of platelet antigen assay.60

    The extent of reactivity may decrease as the platelet count

    recovers, however, it is not usually practical or necessary to

    follow antibody titers.

    Treatment with high-dose intravenous immunoglobulin

    (IVIG) is the most common therapy and can increase platelet

    counts to > 100,000 µl in 4–5 days as described in observa-

    tional studies.61,62 As in other diseases in which IVIG is used,

    mechanisms of action are believed to involve Fc receptor 

    blockade and/or non-specific binding of immunoglobulin toplatelet surfaces.63 The beneficial effects of IVIG in antibody-

    mediated autoimmune disease have been attributed to the

    ability of exogenous IgG to accelerate the rate of IgG catabo-

    lism.64 Plasma exchange,often used to treat diseases thought

    to be mediated by pathogenic autoantibodies or immune

    complexes,was used to treat PTP before IVIG was found effi-

    cacious.65 The role of steroids in treating PTP is unclear,

    although patients who develop PTP are often on chemother-

    apy which may include corticosteroids. Splenectomy has

    been performed in very few patients who do not respond to

    the primary treatments.66 The disease is usually self-limited

    and the prognosis of patients with PTP is good; platelet

    counts usually recover within 21 days.Patients are at risk for 

    significant, e.g. intracranial bleeding, when platelet counts

    are extremely low, thus treatment should be considered.67

    Platelet transfusions may be given,often before the diagnosis

    of PTP is established. Transfused platelets typically survive

    poorly, even if they do not carry the implicated antigen.IVIG

    therapy may also help the survival of transfused platelets

    during the period of severe thrombocytopenia.68,69

    Interestingly, recurrence after later red cell transfusions is

    uncommon.Transfusion with washed red cells and/or cells

    from antigen (HPA-1a) donors has been advocated,70 although 

    the clear value of these practices is unclear.The HPA-1a anti-

    gen is carried by over 95% of blood donors and thus, there are

    only small reserves of antigen negative components. In fact,

    platelet counts may recover with IVIG therapy to levels that

    obviate prophylactic transfusion before antigen negative

    units can be obtained.71

    CIRCULATORY OVERLOAD

    During acute hemorrhage,it is difficult to overload the circu-

    latory system of patients with normal cardiac function.First,the amount of blood lost in severe trauma is often underesti-

    mated as intravascular volume is maintained by intravenous

    fluids and blood products. Second, temporary increases in

    blood volume which may cause small increases in venous

    pressure are well-tolerated by most patients. Major physio-

    logic responses to anemia include increases in cardiac out-

    put and increases in 2,3-DPG content of red cells.The latter 

    shifts the oxygen dissociation curve to the right which facili-

    tates the release of oxygen to surrounding tissues.In chroni-

    cally anemic patients who have increased their cardiac

    output, there is a risk that attempts to raise the arterial oxy-

    gen content by transfusion will overload the circulatory sys-

    tem.This is particularly true in patients with compromisedcardiac status who may not be able to tolerate increased

    intravascular volume. Therefore, the risks and benefits of 

    blood transfusion therapy must be examined before any 

    transfusion. Most physicians no longer use ‘transfusion trig-

    gers’ to determine when a patient should be transfused and

    in general,patients are now transfused at lower hematocrits.

    The risk/benefit ratio is less clear in patients with significant

    cardiac disease, e.g. post-infarction and congestive heart fail-

    ure, and the decision to transfuse is more complex. Recent

    concerns have been expressed about the risks of under 

    transfusing patients with coronary disease.72 Specifically,

    there is evidence that the hematocrit of patients with cardiac

    disease should be maintained close to 30%.Once cardiac output cannot be maintained, circulatory 

    overload can result in pulmonary edema. Symptoms of circu-

    latory overload include chest tightness and cough and wors-

    ening dyspnea as pulmonary edema progresses. Volume

    overload often occurs during or after infusions of both 

    plasma and red cells.Whole blood, rarely used today, carried

    a higher risk of volume overload because of the increased

    amount of plasma.By removing most of the plasma from col-

    lected blood, the same oxygen carrying capacity is main-

    tained in a smaller total volume. Each unit of red cells

    measures between 300 and 350 mL and contains red cells,

    some residual plasma, and the anticoagulant/preservative

    solution. The volume of each unit of plasma is typically 

    between 180 and 300 mL.Plasma is often used to emergently 

    reverse the effects of coumarins (e.g. Warfarin) before vita-

    min K has an effect. In the event of Coumarin overdose, large

     volumes of plasma may be infused to correct the prothrom-

    bin time. Circulatory overload is much less of a problem in

    factor VIII or factor IX replacement therapy as highly puri-

    fied and concentrated products are used instead of large vol-

    umes of fresh frozen plasma or cryoprecipitate.An additional

    benefit of factor concentrates and recombinant products are

    decreased risks of transfusion-transmitted viral infections.

    The use of plasma in thrombotic thrombocytopenic purpura

    should not result in volume overload because plasma

    exchange using apheresis instruments is largely isovolaemic.

    Patients at risk for circulatory overload may benefit from

    Non-infectious transfusion reactions

    73 2001 Harcourt Publishers Ltd  Blood Reviews (2001) 15 , 69–83

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    6/15

    slower rates of transfusion,and may require additional diure-

    sis to minimize excess fluid.The rate of transfusion can be

    lowered so that a single red cell unit is transfused over 4 h,

     which is the maximum time allowed to infuse a unit. Only 

    rarely does it seem necessary to infuse volumes smaller than

    a single red cell unit (volume 300–350 mL) to normally-sized

    adult patients. However, clinicians occasionally request thatred cells units be ‘split’ so that only one-half unit is infused

    over 3–4 h. Diuretics should be considered in patients with 

    any degree of cardiac or renal failure. Furosemide, or other 

    diuretics, should never be added directly to red cells or 

    infused through the same line as the blood product to avoid

    hemolysis. Invasive cardiac monitoring, including measure-

    ments of central venous pressure, cardiac filling pressures,

    and pulmonary vessel pressures,can help to identify impend-

    ing circulatory overload. In addition to patients with 

    impaired cardiac function, patients with neurologic condi-

    tions associated with autonomic dysfunction may have diffi-

    culty regulating their blood pressure during blood

    transfusion and plasmapheresis, as can other individuals with impaired renal function.73

    IRON OVERLOAD IN CHRONICALLY TRANSFUSED

    PATIENTS

    Patients who receive numerous red cell transfusions may 

    develop iron overload. Each unit of red cells contributes 250

    mg iron, whereas daily iron excretion is only about 1 mg in

    the absence of bleeding.Excess iron in patients who receive

    few red cell transfusion is typically harmless.However, trans-

    ferrin may become saturated after only 10–15 units of 

    blood,74 after which iron may be deposited in tissue

    parenchyma.

    75

    It is the non-transferrin-bound iron thatappears in the serum of individuals with iron overload that

    has been implicated in the biologic damage associated with 

    iron overload.76 Favored sites of iron deposition include the

    liver, pancreas and heart. Hepatic iron overload may initially 

    cause histologic evidence of injury like fibrosis, which may 

    then progress to overt cirrhosis.77 Cardiac toxicity may cause

    a cardiomyopathy and arrhythmias.However, signs of clinical

    toxicity are typically not apparent until total body iron

    reaches 400–1000 mg/kg body weight. Other suggested

    detrimental effects of iron overload include accelerated

    development of diabetic nephropathy 78 and increased sus-

    ceptibility to infectious disease79 in transfusion-dependent

    beta thalassemia. Iron overload is most commonly seen in

    conditions that require frequent intermittent red cell transfu-

    sion like the thalassemias, hemoglobinopathies, and aplastic

    anemias. Periodic blood transfusion, in particular, is being

    used more frequently to prevent and treat major complica-

    tions of sickle cell disease like cerebrovascular accidents.

    Long-term transfusion effectively reduces hemoglobin S lev-

    els and can prevent recurrent stroke, however, it eventually 

     will result in iron overload.

    Iron-chelating therapy should be considered in these and

    other chronically transfused patients to minimize the potential

    long-term effects of iron overload. Subcutaneous deferoxam-

    ine injections80 can limit,or in some cases reverse, cardiac and

    hepatic injury in patients with thalassemia.81,82 This treatment

    is most effective when started early. Newer orally active iron

    chelators like deferiprone have been shown to maintain

    lower serum ferritin levels in patients with transfusion-

    dependent iron overload.83,84 Other alternatives to periodic

    blood transfusion like hydroxyurea may reduce the inci-

    dence of iron overload in sickle cell disease by reducing

    transfusion needs.85 Iron overload can also present problems

    for chronically transfused oncology patients who are surviv-ing longer with newer therapies.86 Massively transfused

    patients with hemorrhagic blood loss are at a much lower 

    risk for iron overload because transfused iron is balanced

     with iron lost by bleeding. Periodic erythrocytapheresis in

     which the patient’s red cells are efficiently exchanged with 

    normal donor blood has been examined as an adjunct or 

    alternative to regular chelation therapy.87 Although erythro-

    cytapheresis can reduce total iron burden,the safety and effi-

    cacy of this treatment as compared to standard chelation

    therapy is unclear.88 In some cases, measurements of iron

    stores like ferritin may not accurately predict end-organ dam-

    age.77 Liver biopsy with quantitative iron determination and

    histochemistry remain the reference methods for assessingiron overload. In general, liver iron contents over 15 mg/g

    dry weight are associated with a high risk of cardiac disease.

    Non-invasive measurements of body iron status like MRI

    have been used to accurately quantify liver hemosiderosis in

    multiply transfused thalassemic patients.89 However, MR 

    imaging is extremely sensitive, and iron deposition can be

    seen in the hearts and livers of transfused patients before

    there is clear evidence of organ dysfunction.90

    METABOLIC DISTURBANCES

    Electrolyte imbalances may develop in massively transfused

    patients.Modern fluid replacement strategies and electrolytemonitoring have minimized this complication in the majority 

    of adult patients.In fact,post-transfusion acidosis and hyper-

    kalemia may be more likely related to inadequate resuscita-

    tion from shock than to blood administration. However,

    potentially dangerous metabolic changes can occur in small

    children and neonates.Electrolyte disturbances that are most

    commonly cited include elevated potassium or ammonia lev-

    els and acidosis (Table 4). Many metabolic problems related

    to red cell transfusion are attributed to changes that occur 

    during red cell storage.These changes include increases in

    plasma potassium and hemoglobin related to red cell lysis

    that progress during refrigerated storage. Specific changes in

    red cells – termed the ‘storage lesion’ – include decreasing

    intracellular adenosine triphosphate (ATP), 2,3-diphospho-

    glycerate (2,3-DPG),pH and potassium.Preservative solutions

    Perrotta and Snyder 

    74Blood Reviews (2001) 15 , 69–83   2001 Harcourt Publishers Ltd 

    Table 4 Changes in red cells and supernatant plasma with

    storage

    Red cells Supernatant

    Decreasing ATP Increasing potassium

    Decreasing 2,3-DPG Decreasing pH

    Increasing hemolysis Decreasing sodium

    Increasing lactate Decreasing glucose

    Increasing ammonia

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    7/15

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    8/15

    severe air embolism based on their ability to improve tissue

    oxygenation.106

    HYPOTHERMIA

    Hypothermia may occur when large volumes of cold blood

    components are transfused over a short period, most com-monly in the emergency department or operating room.

    Transfusion-related hypothermia is also dangerous to small

    children and newborns who undergo exchange transfusion.

    Red cells are stored in the liquid state at 1–6˚C and are

    removed from the refrigerator only at the time of transfusion.

    Thus, blood may be transfused ‘ice-cold’ in emergency situa-

    tions. Fresh frozen plasma is stored at

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    9/15

    occurred in patients who received red cells in which white

    cells were removed using specific lots of the LeukoNet

    Prestorage Leukoreduction Filtration System (Hemasure,

    Marlborough, Massachusetts). Possible explanations include

    allergic responses to unidentified allergens in the filtration

    system, or other reactions to a chemical, material, or break-

    down product contained in the leukoreducing system.Reports of acute periocular reactions decreased after these

    leukoreduction filters were removed from inventories.

     TRANSFUSION-ASSOCIATED SEPSIS

    Bacterial sepsis is a long-recognized complication of blood

    transfusion therapy.Blood products may be contaminated by 

    bacteria if a donor is bacteremic during the blood collection

    or if the arm is improperly prepared before venipuncture.120

    Use of single-use, closed, sterilized collection systems and

    limiting the duration of red cell storage to 35–42 days has

    contributed to the low incidence of bacterial contamination.However, there remain occasional reports of severe, often

    fatal reactions related to bacteria in a blood component.The

    risk of transfusion-associated sepsis (TAS) is difficult to esti-

    mate because blood recipients are often immunocompro-

    mised and have other risk factors for sepsis. TAS was

    implicated in three of 28 deaths in the United Kingdom’s

    serious hazards of transfusion study (SHOT)121. In the United

    States, 10 fatal transfusion reactions reported from January 

    1998 through June 1999 as part of the bacterial contamina-

    tion (BACON) study were likely caused by TAS. Organisms

    commonly implicated in septic transfusion reactions (STR)

    include gram-positive (Staphylococcus sp.) and gram-nega-

    tive (Yersinia, Enterobacter, Pseudomonas sp.) bacteria.122

     Yersinia enterocolitica in particular, is capable of growing at

    colder temperatures (4–6˚C) and elaborated endotoxins can

    cause shock.123 Blood donors must be in good health on the

    day of donation. This requirement, however, does not

    exclude asymptomatic donors who may have had a short-

    lasting, gastroenteritis or mild diarrhea 5–14 days prior to

    blood donation. These donors may have a longer than

    expected period of asymptomatic bacteria that allows trans-

    mission of organisms on donation.124,125 Other less common

    blood contaminants like Serratia liquefaciens consistently 

    cause severe morbidity and are associated with a high death 

    rate.126 Autologous transfusion does not protect againt bacte-

    rial contamination.

    127

    Bacterial contamination of platelet concentrates, espe-

    cially by gram-positive microorganisms, is more common

    because platelets are stored at room temperature.128,129 For 

    this reason, platelets are stored for a maximum of 5 days.

    Storing platelets at lower temperatures will reduce bacterial

    growth, however, platelets are damaged when refrigerated

    and efforts to use cryoprotectants to protect platelet during

    cold storage continue.130 Similarly, antibiotics cannot be

    added directly to platelet concentrates because they will

    damage cell membranes.Transfusing blood products contam-

    inated by bacteria is dangerous and may cause profound

    hypotension and shock. However, fatal STR are relatively 

    rare.This may be explained by the slow growth of many con-

    taminating bacteria which do not produce toxins.There are

    no screening tests available at this time to detect contami-

    nated units. Commercially available multiple-reagent urine

    dipsticks have been used to detect falling glucose levels and

    pH in platelet units experimentally contaminated with bacte-

    ria.131 However, the sensitivity of reagent strips may be lim-

    ited by the normal changes in pH and glucose encountered

    during prolonged platelet storage. Short-term bacterial cul-ture of blood products using automated bacterial systems

    can detect contaminated blood products,but again this prac-

    tice is not widespread.132 Other efforts are underway to

    develop both screening tests and means for destroying bac-

    teria within individual blood units. Several approaches to

    pathogen inactivation are in pre-clinical or clinical trials.

    Most involve use of chemicals or photochemical methods to

    destroy blood pathogens by targeting nucleic acids.133

     Although originally designed to inactivate viruses like HIV,

    they are also effective against bacteria and parasitic organ-

    isms.The efficacy of pathogen inactivation systems is usually 

    first determined in non-cellular products (plasma), and if 

    results are promising, studies are extended to cellular com-ponents (red cells, platelets). Phase 3 trials are currently 

    underway in the United States and Europe using psoralen, S-

    59 medicated pathogen inactivation of platelet concen-

    trates.134  Alternative photochemical treatments that utilize

    riboflavin are also being developed to inactivate bacteria.135

     Although most bacteria are effectively inactivated by these

    techniques, some bacteria can survive decontamination.

    Leukodepletion of blood components may decrease the

    growth of some,but not all bacteria, and is not considered an

    effective means of eliminating septic transfusion reactions.

    Blood cultures should be drawn from patients who

    develop high fevers during or following transfusion, espe-

    cially if they become hypotensive. Septic reactions can be dif-ferentiated from more common febrile reactions in that the

    latter are generally self-limited and lack profound hypoten-

    sion. Clearly, it is critical to distinguish febrile and septic reac-

    tions in immunosuppressed patients. A temperature rise of 

    more than 2˚C following platelet transfusion makes a septic

    reaction more likely.136 Rare patients who receive contami-

    nated products may develop no, or mild febrile symptoms.

    Many symptoms are attributed to preformed endotoxins and

    cytokines.These include skin f lushing, rigors,and rapidly pro-

    gressive cardiovascular collapse.Symptoms may ensue during,

    or minutes to hours after the transfusion is completed.Clinical

    severity is related to the elaboration of endotoxins by gram-

    negative organisms and other virulence factors that permit

    bacterial growth. The load of bacteria infused is directly 

    related to the time of storage and the volume of the compo-

    nent. Host characteristics including concomitant antibiotic

    administration, degree of immunosuppression and overall

    medical status of the patient will also influence clinical

    severity.Gram stains of suspected products may help when

    organisms are seen. Blood component bags are not rou-

    tinely retained at most hospitals, but suspected units

    should be cultured if possible.The patient should also be

    cultured to confirm that the infection was caused by the

    blood transfusion and to exclude other sources of infec-

    tion. Importantly, an acute hemolytic transfusion reaction

    should be excluded. Treatment includes broad-spectrum

    antibiotics,fluids, and cardiorespiratory support.

    Non-infectious transfusion reactions

    77 2001 Harcourt Publishers Ltd  Blood Reviews (2001) 15 , 69–83

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    10/15

     TRANSFUSION-ASSOCIATED GRAFT VERSUS HOST

    DISEASE

    Transfusion-associated graft versus host disease (TA-GVHD)

    is a rare complication of blood transfusion that is fatal in

    approximately 90% of patients.The risk of TA-GVHD is diffi-

    cult to estimate but is related to the number of viable T lym-

    phocytes transfused, the recipient’s immune status, and theHLA disparity between donor and recipient.137 Therefore,

    multiply transfused patients who receive cells from donors

     who share HLA haplotypes (haploidentical) with the recipi-

    ent are at greatest risk.The classic scenario for the develop-

    ment of TA-GVHD in immunocompetent individuals is

    homozygosity of the donor for an HLA type shared by the

    recipient.138 Thus, TA-GVHD is more common in societies

    like Japan where there is a higher likelihood for HLA 

    homozygosity.TA-GVHD occurs when donor immunocompe-

    tent T and NK cells attack recipient cells because these recip-

    ient cells appear foreign due to differences in major or minor 

    histocompatibility antigens.139 GVHD is usually seen follow-

    ing allogeneic bone marrow transplant (BMT),but may alsooccur in immunodeficient or immunosuppressed patients

    following blood transfusion.Clinically,TA-GVHD is character-

    ized by the acute onset of rash, abdominal pain, diarrhea,

    liver function abnormalities, and bone marrow suppression

    beginning 2 to 30 days following transfusion. The macu-

    lopapular rash seen is similar to that observed in acute

    GVHD following BMT. Biopsy of the skin can confirm the

    diagnosis.Immunohistochemical studies of skin biopsies will

    show an infiltrate of CD3+ T lymphocytes, and populations

    of T lymphocyte subsets (CD4+, CD8+) that are donor 

    derived.140 Pancytopenia in TA-GVHD may be severe and is

    attributed to destruction of recipient marrow stem cells by 

    donor lymphocytes.Immunosuppressive therapy using corticosteroids,

    cyclosporine A and anti-CD3 monoclonal antibody (OKT3)

    has been used in cases of TA-GVHD.141,142 The benefits of 

    these treatments are unclear, however, therapeutic modali-

    ties are proposed based on the presumed mechanism of the

    disease.143 These strategies are designed to reduce the cyto-

    toxic T-lymphocyte mediated tissue injury through apoptotic

    pathways, as well as reducing the production of inflamma-

    tory cytokines like tumor necrosis factor.144 Fortunately,TA-

    GVHD can be prevented by irradiating products prior to

    transfusion. Specifically, irradiating cellular blood products

     with 2500 cGy inactivates donor lymphocytes and is the

    most effective method for preventing TA-GVHD.145This radia-tion dose is required to completely inactive T cells found in

    blood collected and stored in common plastic containers

    used today. Platelets and granulocytes are not damaged by 

    this radiation dose, but red cells sustain detectable damage.

    For this reason, the maximum storage time for irradiated red

    cells is 28 days. Patients who should receive irradiated blood

    components include neonates who may have immature or 

    abnormal immune systems,patients with hematologic malig-

    nancies, and cancer patients who are marrow/stem cell

    transplant candidates or are receiving high-dose chemother-apy (Table 5). Directed donations from first-degree relatives

    must be irradiated, although most blood centers will extend

    this requirement to all blood relatives. For the same reason,

    apheresis platelets matched for HLA class I antigens should

    be irradiated.146 Radiochromic film can be used as a dosime-

    ter to verify that an effective dose of radiation has been deliv-

    ered to the product.147 Leukoreduction is not adequate to

    prevent TA-GVHD as there are sufficient residual lympho-

    cytes present to cause GVHD.148 Photochemical treatment

    (PCT) of blood products using psoralen S-59 and long-wave-

    length ultraviolet light has been shown to prevent TA-GVHD

    in a murine parent to F1 transfusion model.149 In this study,

    mice received allogeneic splenic leukocytes that wereuntreated,gamma irradiated (2500 cGy),or PCT treated (150

    µM S-59, 2.1 J/cm2 UVA). Mice that received gamma irradi-

    ated or PCT treated leukocytes did not develop clinical or 

    histologic evidence of TA-GVHD,whereas those that received

    untreated leukocytes developed clear evidence of TA-GVHD.

     As PCT is also effective in inactivating contaminating viruses

    and bacteria – and is being developed for this purpose – PCT

    may also provide protection against TA-GVHD.

     TRANSFUSION-RELATED ACUTE LUNG INJURY

    Transfusion-related acute lung injury (TRALI) is a rare but

    serious complication of blood transfusion that presents asnon-cardiogenic pulmonary edema.150 It typically occurs

     within 6 h of transfusion and is clinically similar to the adult

    respiratory distress syndrome. The most common clinical

    findings include the rapid onset of dyspnea, tachypnea,

    cyanosis,fever,and hypotension.151 The incidence of TRALI is

    unclear because it is often overlooked or not reported, but

    the estimated frequency is one in 5000 transfusions.152 Lung

    auscultation reveals diffuse crackling and decreased breath 

    sounds. Invasive hemodynamic monitoring may be required

    to differentiate TRALI from pulmonary edema secondary to

    cardiac failure or volume overload and to guide therapy.

    Specifically, cardiac monitoring shows normal cardiac pres-

    sures and function with hypoxemia and decreased pul-monary compliance. Radiographic findings include diffuse,

    fluffy infiltrates characteristic of pulmonary edema. The

    etiology of many cases of TRALI appears to involve an

    Perrotta and Snyder 

    78Blood Reviews (2001) 15 , 69–83   2001 Harcourt Publishers Ltd 

    Table 5 General indications for irradiated blood components

    Infants

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    11/15

    immune-mediated reaction of HLA antibodies or other 

    leukoagglutins with white cells.153 According to this model,

    granulocytes are first activated in the peripheral circulation

    by HLA or other Ag-Ab complexes.Activated leukocytes then

    migrate to the lungs where they bind to the pulmonary cap-

    illary bed via integrins and other cell adhesion molecules.Ex

     vivo animal models of TRALI provide some support for thishypothesis.154 Proteolytic enzymes are then released that

    destroy tissue,resulting in a capillary leak syndrome and pul-

    monary edema. Massive pulmonary edema with granulocyte

    aggregation within the pulmonary microvasculature and

    alveolar extravasation have been noted in patients who die

    of TRALI.155 However, antibody specificity cannot be identi-

    fied in many cases of TRALI, suggesting that other mecha-

    nisms are involved. More recently, reactive lipid products

    released from donor cell membranes have been associated

     with the development of TRALI using an isolated, perfused

    rat lung model.156

    TRALI should be suspected in patients with rapid onset

    respiratory distress following transfusion therapy, or pul-monary edema without hypervolemia and congestive heart

    failure.The identification of HLA and/or granulocyte antibod-

    ies in either the donor’s or recipient’s serum is highly sug-

    gestive of TRALI in the appropriate clinical context. Ideally,

    the corresponding antigens are found on the recipient’s or 

    donor’s leukocytes.This specialized testing is performed in

    few specialized laboratories and will not be available during

    the acute episode. Products obtained from donors impli-

    cated in TRALI should not be used for blood transfusion,

    although their plasma is suitable for fractionation and for 

    producing plasma-free components.157 It has been suggested

    that transfusing products obtained from female blood donors

     who are sensitized to HLA antigens by pregnancy may increase the risk of TRALI.However, cases of TRALI could not

    be found in a large series of platelet transfusions using

    apheresis platelets collected from woman,158 despite the fact

    that 17% of the female donors demonstrated HLA sensitiza-

    tion. Thus, prospective screening of blood donors for HLA 

    sensitization is not recommended. Unlike acute respiratory 

    distress syndrome, approximately 80–90% of patients with 

    TRALI will survive with supportive care consisting of 

    aggressive respiratory support, supplemental oxygen and

    mechanical ventilation when necessary. Based on the pre-

    sumed pathogenesis of TRALI, leukoreduced blood products

    could potentially decrease the incidence of TRALI. Drugs

    used to treat TRALI have included corticosteroids and

    diuretics, but there are no controlled studies demonstrating

    the efficacy of these or other agents versus supportive care

    alone.

    CONCLUSIONS

     Vast improvements in each phase of blood transfusion ther-

    apy have markedly reduced, but not eliminated, the inci-

    dence of adverse events. Significant technologic innovations

    in the equipment used to collect, process, and store blood

    products have paralleled our increased understanding of the

    physiology of transfusion. Despite many safeguards and

    a clear understanding of red cell immunology, hemolytic

    reactions caused by transfusing ABO incompatible blood

    remain the most common cause of immediate fatality. Less

    common fatal reactions attributed to blood transfusion

    include septic transfusion reactions, transfusion associated

    graft-versus-host disease (TA-GVHD), and transfusion related

    acute lung injury.159 Therefore, decisions to transfuse a

    patient should be made by individuals with a knowledge of 

    the most relevant risks of blood component therapy and anability to weigh risk/benefit ratios. In many countries,

    informed consent must be obtained by health care person-

    nel who are able to communicate these risks before blood

    products are transfused. Most patients are concerned about

    contracting HIV or hepatitis C from donor blood.However, it

    is difficult for patients to understand that the risk of viral

    infection is exceedingly small. In some respects, the non-

    infectious risks of blood transfusion are even more difficult

    to quantify because they are often related to the training and

    experience of the transfusion team. In many cases, risks dif-

    fer among individual patients. Cardiac patients are often at

    increased risk for volume overload; multiply transfused

    patients frequently develop multiple red cell antibodies,andseverely immunocompromised patients may develop TA-

    GVHD if blood is not irradiated. Despite technological

    advances in warming devices, hypothermia continues to

    adversely affect massively-transfused patients. Autologous

    blood – often considered quite safe – does not eliminate all

    risks of transfusion therapy such as bacterial contamination

    and fluid overload. A better understanding of the mecha-

    nisms leading to the described non-infectious complications

    of blood component therapy has lead to strategies designed

    to reduce, eliminate, and treat these adverse effects.

    However, continued vigilance is needed to prevent the most

    avoidable untoward reactions.

    References

    1. Dodd RY. Current viral risks of blood and blood products.Ann Med

    2000; 32: 469–474.

    2. Busch MP. HIV, HBV and HCV: new developments related to

    transfusion safety.Vox Sang 2000; 78: 253–256.

    3. Dodd RY.Transmission of parasites and bacteria by blood

    components.Vox Sang 2000; 78: 239–242.

    4. Williamson LM,Lowe S,Love EM et al. Serious hazards of 

    transfusion (SHOT) initiative: analysis of the first two annual

    reports. BMJ 1999; 319:16–19.

    5. Brand A. Immunological aspects of blood transfusions. Blood Rev

    2000; 14: 130–144.

    6. Capon SM, Goldfinger D.Acute hemolytic transfusion reaction,a

    paradigm of the systemic inflammatory response:new insights into

    pathophysiology and treatment.Transfusion 1995;35: 513–520.

    7. Solanki D,McCurdy PR. Delayed hemolytic transfusion reactions.An

    often-missed entity. Jama 1978;239: 729–731.

    8. Moore SB,Taswell HF, Pineda AA, Sonnenberg CL. Delayed hemolytic

    transfusion reactions. Evidence of the need for an improved

    pretransfusion compatibility test.Am J Clin Pathol 1980;74: 94–97.

    Non-infectious transfusion reactions

    79 2001 Harcourt Publishers Ltd  Blood Reviews (2001) 15 , 69–83

    Correspondence to: P. L. Perrotta,State University of New York @ Stony

    Brook, University Hospital,Laboratory Administration L3-532, Stony Brook,

    NY 11794, USA.Tel.: +1 631 444 2601; Fax: +1 631 444 2653; E-mail:

    [email protected]

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    12/15

    9. Pineda AA,Vamvakas EC, Gorden LD,Winters JL, Moore SB.Trends

    in the incidence of delayed hemolytic and delayed serologic

    transfusion reactions.Transfusion 1999;39: 1097–1103.

    10. Salama A, Mueller-Eckhardt C. Delayed hemolytic transfusion

    reactions. Evidence for complement activation involving allogeneic

    and autologous red cells.Transfusion 1984;24: 188–193.

    11. Patten E, Reddi CR,Riglin H,Edwards J. Delayed hemolytictransfusion reaction caused by a primary immune response.

    Transfusion 1982; 22: 248–250.

    12. Davenport RD.The role of cytokines in hemolytic transfusion

    reactions. Immunol Invest 1995;24: 319–331.

    13. von Zabern I, Ehlers M,Grunwald U, Mauermann K,

    Greinacher A. Release of mediators of systemic inflammatory

    response syndrome in the course of a severe delayed hemolytic

    transfusion reaction caused by anti-D.Transfusion 1998; 38:

    459–468.

    14. Pineda AA,Taswell HF, Brzica SM, Jr.Transfusion reaction.An

    immunologic hazard of blood transfusion.Transfusion 1978;18:

    1–7.

    15. Vamvakas EC, Pineda AA,Reisner R, Santrach PJ, Moore SB.Thedifferentiation of delayed hemolytic and delayed serologic

    transfusion reactions: incidence and predictors of hemolysis.

    Transfusion 1995; 35: 26–32.

    16. Ness PM,Shirey RS,Thoman SK, Buck SA.The differentiation of 

    delayed serologic and delayed hemolytic transfusion reactions:

    incidence, long-term serologic findings, and clinical significance.

    Transfusion 1990; 30: 688–693.

    17. Shulman IA,Nelson JM, Nakayama R.When should antibody

    screening tests be done for recently transfused patients?

    Transfusion 1990; 30: 39–41.

    18. Syed SK, Sears DA,Werch JB, Udden MM, Milam JD. Case reports:

    delayed hemolytic transfusion reaction in sickle cell disease. Am J

    Med Sci 1996; 312: 175–181.

    19. Petz LD, Calhoun L,Shulman IA, Johnson C,Herron RM.The sickle

    cell hemolytic transfusion reaction syndrome.Transfusion 1997;37:

    382–392.

    20. Cullis JO,Win N, Dudley JM, Kaye T. Post-transfusion

    hyperhaemolysis in a patient with sickle cell disease: use of steroids

    and intravenous immunoglobulin to prevent further red cell

    destruction.Vox Sang 1995; 69: 355–357.

    21. Heddle NM,Klama LN, Griffith L et al.A prospective study to

    identify the risk factors associated with acute reactions to platelet

    and red cell transfusions.Transfusion 1993; 33: 794–797.

    22. Winqvist I. Meperidine (pethidine) to control shaking chills and

    fever associated with non-hemolytic transfusion reactions. Eur J

    Haematol 1991; 47:154–155.

    23. Perrotta PL, Feldman D, Snyder EL. Biological response modifiers in

    platelet transfusion therapy. In: Seghatchian J,Snyder EL, Krailadsiri P,

    eds. Platelet therapy (First ed).Amsterdam: Elsevier 2000;227–262.

    24. Chambers LA, Kruskall MS, Pacini DG, Donovan LM. Febrile

    reactions after platelet transfusion:the effect of single versus

    multiple donors.Transfusion 1990;30: 219–221.

    25. Riccardi D, Raspollini E, Rebulla P et al. Relationship of the time of 

    storage and transfusion reactions to platelet concentrates from

    buffy coats.Transfusion 1997; 37: 528–530.

    26. Heddle NM,Klama L, Singer J et al.The role of the plasma from

    platelet concentrates in transfusion reactions. N Engl J Med 1994;

    331: 625–628.

    27. Sarkodee-Adoo CB, Kendall JM,Sridhara R,Lee EJ, Schiffer CA.The

    relationship between the duration of platelet storage and the

    development of transfusion reactions.Transfusion 1998; 38:

    229–235.

    28. Neumuller J, Schwartz DW, Mayr WR.Demonstration by flow

    cytometry of the numbers of residual white blood cells and

    platelets in filtered red blood cell concentrates and plasma

    preparations.Vox Sang 1997; 73: 220–229.

    29. Bowden RA,Slichter SJ,Sayers M et al.A comparison of filteredleukocyte-reduced and cytomegalovirus (CMV) seronegative blood

    products for the prevention of transfusion-associated CMV

    infection after marrow transplant. Blood 1995; 86: 3598–3603.

    30. Leukocyte reduction and ultraviolet B irradiation of platelets to

    prevent alloimmunization and refractoriness to platelet

    transfusions.The Trial to Reduce Alloimmunization to Platelets

    Study Group.N Engl J Med 1997; 337: 1861–1869.

    31. Federowicz I, Barrett BB,Andersen JW et al.Characterization of 

    reactions after transfusion of cellular blood components that are

    white cell reduced before storage.Transfusion 1996;36: 21–28.

    32. Stack G,Snyder EL.Cytokine generation in stored platelet

    concentrates.Transfusion 1994;34: 20–25.

    33. Kluter H, Bubel S, Kirchner H,Wilhelm D. Febrile and allergictransfusion reactions after the transfusion of white cell-poor

    platelet preparations.Transfusion 1999;39: 1179–1184.

    34. Domen RE.Adverse reactions associated with autologous blood

    transfusion: evaluation and incidence at a large academic hospital.

    Transfusion 1998; 38: 296–300.

    35. Wilhelm D, Kluter H,Klouche M, Kirchner H. Impact of allergy

    screening for blood donors: relationship to nonhemolytic

    transfusion reactions.Vox Sang 1995;69: 217–221.

    36. Stern A, van Hage-Hamsten M, Sondell K, Johansson SG. Is allergy

    screening of blood donors necessary? A comparison between

    questionnaire answers and the presence of circulating IgE

    antibodies.Vox Sang 1995; 69: 114–119.

    37. Sharon R, Kidroni G,Michel J. Presence of aspirin in blood units.

    Vox Sang 1980;38: 284–287.

    38. Buck SA, Kickler TS,McGuire M,Braine HG,Ness PM.The utility of 

    platelet washing using an automated procedure for severe platelet

    allergic reactions.Transfusion 1987;27: 391–393.

    39. Davenport RD, Burnie KL, Barr RM.Transfusion management of 

    patients with IgA deficiency and anti-IgA during liver

    transplantation.Vox Sang 1992; 63: 247–250.

    40. Nicholls A. Ethylene oxide and anaphylaxis during haemodialysis. Br

    Med J (Clin Res Ed) 1986; 292: 1221–1222.

    41. Ryden SE,Oberman HA.Compatibility of common intravenous

    solutions with CPD blood.Transfusion 1975;15: 250–255.

    42. Staples PJ, Griner PF. Extracorporeal hemolysis of blood in a

    microwave blood warmer. N Engl J Med 1971;285: 317–319.

    43. Beauregard P, Blajchman MA. Hemolytic and pseudo-hemolytic

    transfusion reactions:an overview of the hemolytic transfusion

    reactions and the clinical conditions that mimic them.Transfus Med

    Rev 1994;8: 184–199.

    44. Moss G,Staunton C.Needles and hemolysis.N Engl J Med 1970;

    283:598.

    45. Mollison PL, Engelfriet CP, Contreras M. Blood transfusion in

    clinical medicine (10th ed). Oxford [UK];Malden,MA,USA:

    Blackwell Science; 1997.

    46. Ma SK,Wong KF, Siu L. Hemoglobinemia and hemoglobinuria

    complicating concomitant use of a white cell filter and a pressure

    infusion device.Transfusion 1995; 35: 180.

    47. Blajchman MA. Reducing the risk of bacterial contamination of 

    cellular blood components.Dev Biol Stand 2000; 102: 183–193.

    Perrotta and Snyder 

    80Blood Reviews (2001) 15 , 69–83   2001 Harcourt Publishers Ltd 

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    13/15

    48. Beutler E. Glucose-6-phosphate dehydrogenase deficiency. N Engl J

    Med 1991;324:169–174.

    49. Meryman HT, Hornblower M.A method for freezing and washing

    red blood cells using a high glycerol concentration.Transfusion

    1972; 12: 145–156.

    50. Capon SM, Goldfinger D.Acute hemolytic transfusion reaction,a

    paradigm of the systemic inflammatory response:new insights intopathophysiology and treatment.Transfusion 1995;35: 513–520.

    51. Morrison FS, Mollison PL. Post-transfusion purpura.N Engl J Med

    1966; 275:243–248.

    52. Waters AH. Post-transfusion purpura.Blood Rev 1989;3: 83–87.

    53. Taaning E, Svejgaard A.Post-transfusion purpura: a survey of 12

    Danish cases with special reference to immunoglobulin G

    subclasses of the platelet antibodies.Transfus Med 1994;4: 1–8.

    54. Mueller-Eckhardt C, Lechner K, Heinrich D et al. Post-transfusion

    thrombocytopenic purpura: immunological and clinical

    studies in two cases and review of the literature. Blut 1980;40:

    249–257.

    55. Pegels JG,Bruynes EC,Engelfriet CP, von dem Borne AE.Post-

    transfusion purpura:a serological and immunochemical study. Br JHaematol 1981;49: 521–530.

    56. Chapman JF, Murphy MF, Berney SI et al. Post-transfusion purpura

    associated with anti-Baka and anti-PIA2 platelet antibodies and

    delayed haemolytic transfusion reaction.Vox Sang 1987; 52:

    313–317.

    57. Kroll H, Kiefel V, Mueller-Eckhardt C. [Post-transfusion purpura:

    clinical and immunologic studies in 38 patients]. Infusionsther

    Transfusionmed 1993; 20: 198–204.

    58. Zeigler Z,Murphy S, Gardner FH. Post-transfusion purpura: a

    heterogeneous syndrome.Blood 1975;45: 529–536.

    59. Dainer PM, Canada ED.Post-transfusion purpura and multiple

    transfusions. Br Med J 1977; 2: 999.

    60. Taaning E,Tonnesen F. Pan-reactive platelet antibodies in post-

    transfusion purpura.Vox Sang 1999;76: 120–123.

    61. Mueller-Eckhardt C, Kiefel V. High-dose IgG for post-transfusion

    purpura-revisited. Blut 1988;57: 163–167.

    62. Becker T, Panzer S, Maas D et al. High-dose intravenous

    immunoglobulin for post-transfusion purpura.Br J Haematol 1985;

    61: 149–155.

    63. Salama A, Mueller-Eckhardt C, Kiefel V. Effect of intravenous

    immunoglobulin in immune thrombocytopenia.Lancet 1983;2:

    193–195.

    64. Yu Z, Lennon VA. Mechanism of intravenous immune globulin

    therapy in antibody-mediated autoimmune diseases.N Engl J Med

    1999; 340:227–228.

    65. Cimo PL,Aster RH. Post-transfusion purpura: successful treatment

    by exchange transfusion.N Engl J Med 1972; 287: 290–292.

    66. Cunningham CC, Lind SE.Apparent response of refractory post-

    transfusion purpura to splenectomy.Am J Hematol 1989; 30:

    112–113.

    67. Vogelsang G, Kickler TS, Bell WR.Post-transfusion purpura: a

    report of five patients and a review of the pathogenesis and

    management.Am J Hematol 1986;21: 259–267.

    68. Kekomaki R, Elfenbein G, Gardner R et al. Improved response of 

    patients refractory to random-donor platelet transfusions by

    intravenous gamma globulin.Am J Med 1984; 76: 199–203.

    69. Gabriel A, Lassnigg A, Kurz M, Panzer S. Post-transfusion purpura

    due to HPA-Ia immunization in a male patient: response to

    subsequent multiple HPA-Ia-incompatible red-cell transfusions.

    Transfus Med 1995; 5: 131–134.

    70. Win N,Peterkin MA,Watson WH.The therapeutic value of HPA-Ia-

    negative platelet transfusion in post-transfusion purpura complicated

    by life-threatening haemorrhage.Vox Sang 1995;69:138–139.

    71. Brecher ME, Moore SB, Letendre L.Posttransfusion purpura: the

    therapeutic value of P1A1-negative platelets.Transfusion 1990; 30:

    433–435.

    72. Hebert PC,Wells G, Blajchman MA et al.A multicenter,randomized, controlled clinical trial of transfusion requirements in

    critical care.Transfusion Requirements in Critical Care

    Investigators,Canadian Critical Care Trials Group.N Engl J Med

    1999; 340:409–417.

    73. Couriel D,Weinstein R. Complications of therapeutic plasma

    exchange:a recent assessment. J Clin Apheresis 1994; 9: 1–5.

    74. Ley TJ, Griffith P, Nienhuis AW.Transfusion haemosiderosis and

    chelation therapy. Clin Haematol 1982; 11: 437–464.

    75. Marcus RE, Huehns ER.Transfusional iron overload.Clin Lab

    Haematol 1985; 7: 195–212.

    76. Breuer W, Ronson A,Slotki IN et al.The assessment of serum

    nontransferrin-bound iron in chelation therapy and iron

    supplementation.Blood 2000;95: 2975–2982.77. Harmatz P, Butensky E, Quirolo K et al. Severity of iron overload in

    patients with sickle cell disease receiving chronic red blood cell

    transfusion therapy. Blood 2000;96: 76–79.

    78. Loebstein R,Lehotay DC,Luo X et al. Diabetic nephropathy in

    hypertransfused patients with beta-thalassemia.The role of 

    oxidative stress. Diabetes Care 1998; 21: 1306–1309.

    79. Cunningham-Rundles S,Giardina PJ, Grady RW et al. Effect of 

    transfusional iron overload on immune response. J Infect Dis 2000;

    182 Suppl 1: S115–121.

    80. Propper RD, Cooper B, Rufo RR et al. Continuous subcutaneous

    administration of deferoxamine in patients with iron overload. N

    Engl J Med 1977; 297: 418–423.

    81. Wolfe L,Olivieri N, Sallan D et al. Prevention of cardiac disease by

    subcutaneous deferoxamine in patients with thalassemia major. N

    Engl J Med 1985; 312: 1600–1603.

    82. Maurer HS, Lloyd-Still JD,Ingrisano C, Gonzalez-Crussi F, Honig

    GR.A prospective evaluation of iron chelation therapy in children

    with severe beta-thalassemia.A six-year study.Am J Dis Child

    1988; 142:287–292.

    83. Kersten MJ, Lange R, Smeets ME et al.Long-term treatment of 

    transfusional iron overload with the oral iron chelator deferiprone

    (L1): a Dutch multicenter trial.Ann Hematol 1996; 73: 247–252.

    84. Hoffbrand AV, F AL-R, Davis B et al. Long-term trial of deferiprone

    in 51 transfusion-dependent iron overloaded patients.Blood 1998;

    91: 295–300.

    85. Ware RE,Zimmerman SA, Schultz WH.Hydroxyurea as an

    alternative to blood transfusions for the prevention of recurrent

    stroke in children with sickle cell disease.Blood 1999;94: 3022–3026.

    86. Lichtman SM,Attivissimo L, Goldman IS, Schuster MW, Buchbinder

    A. Secondary hemochromatosis as a long-term complication of the

    treatment of hematologic malignancies.Am J Hematol 1999; 61:

    262–264.

    87. Singer ST, Quirolo K, Nishi K et al. Erythrocytapheresis for

    chronically transfused children with sickle cell disease: an effective

    method for maintaining a low hemoglobin S level and reducing iron

    overload. J Clin Apheresis 1999; 14: 122–125.

    88. Adams DM, Schultz WH,Ware RE,Kinney TR.Erythrocytapheresis

    can reduce iron overload and prevent the need for chelation

    therapy in chronically transfused pediatric patients. J Pediatr

    Hematol Oncol 1996;18: 46–50.

    Non-infectious transfusion reactions

    81 2001 Harcourt Publishers Ltd  Blood Reviews (2001) 15 , 69–83

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    14/15

  • 8/9/2019 1-s2.0-S0268960X01901511-main

    15/15

    128. Anderson KC, Lew MA,Gorgone BC et al.Transfusion-related

    sepsis after prolonged platelet storage.Am J Med 1986; 81:

    405–411.

    129. Engelfriet CP, Reesink HW, Blajchman MA et al.Bacterial

    contamination of blood components.Vox Sang 2000;78: 59–67.

    130. Lozano M, Escolar G, Mazzara R et al. Effects of the addition of 

    second-messenger effectors to platelet concentrates separatedfrom whole-blood donations and stored at 4 degrees C or -80

    degrees C.Transfusion 2000; 40: 527–534.

    131. Burstain JM,Brecher ME,Workman K et al.Rapid identification

    of bacterially contaminated platelets using reagent strips: glucose

    and pH analysis as markers of bacterial metabolism.Transfusion

    1997; 37:255–258.

    132. Liu HW,Yuen KY, Cheng TS et al.Reduction of platelet

    transfusion-associated sepsis by short-term bacterial culture.

    Vox Sang 1999;77: 1–5.

    133. Lin L, Londe H, Janda JM, Hanson CV, Corash L. Photochemical

    inactivation of pathogenic bacteria in human platelet

    concentrates.Blood 1994;83: 2698–2706.

    134. Corash L. Inactivation of viruses, bacteria,protozoa andleukocytes in platelet and red cell concentrates.Vox Sang 2000;

    78:205–210.

    135. Goodrich RP.The use of riboflavin for the inactivation of 

    pathogens in blood products.Vox Sang 2000;78: 211–215.

    136. Chiu EK,Yuen KY, Lie AK et al.A prospective study of 

    symptomatic bacteremia following platelet transfusion and of its

    management.Transfusion 1994; 34: 950–954.

    137. Yasuura K, Okamoto H,Matsuura A.Transfusion-associated

    graft-versus-host disease with transfusion practice in cardiac

    surgery. J Cardiovasc Surg (Torino) 2000;41: 377–380.

    138. Ohto H,Anderson KC.Survey of transfusion-associated graft-

    versus-host disease in immunocompetent recipients.Transfus

    Med Rev 1996;10: 31–43.

    139. Vogelsang GB,Hess AD. Graft-versus-host disease: new

    directions for a persistent problem. Blood 1994; 84: 2061–2067.

    140. Tanei R,Ohta Y, Ishihara S et al.Transfusion-associated graft-

    versus-host disease: an in situ hybridization analysis of the

    infiltrating donor-derived cells in the cutaneous lesion.

    Dermatology 1999;199: 20–24.

    141. Yasukawa M. [Treatment of transfusion-associated graft-versus-

    host disease]. Nippon Rinsho 1997;55: 2290–2295.

    142. Sakurai M, Moizumi Y, Uchida S, Imai Y,Tabayashi K.Transfusion-

    associated graft-versus-host disease in immunocompetent

    patient: early diagnosis and therapy.Am J Hematol 1998; 58:

    84–86.

    143. Saigo K,Ryo R.Therapeutic strategy for post-transfusion graft-

    vs.-host disease. Int J Hematol 1999;69: 147–151.

    144. Nishimura M,Uchida S,Mitsunaga S et al.Characterization of T-

    cell clones derived from peripheral blood lymphocytes of a

    patient with transfusion-associated graft-versus-host disease:

    Fas-mediated killing by CD4+ and CD8+ cytotoxic T-cell clones

    and tumor necrosis factor beta production by CD4+ T-cell

    clones. Blood 1997; 89:1440–1445.

    145. Williamson LM,Warwick RM.Transfusion-associated graft-versus-host disease and its prevention. Blood Rev 1995; 9: 251–261.

    146. Grishaber JE,Birney SM,Strauss RG. Potential for transfusion-

    associated graft-versus-host disease due to apheresis platelets

    matched for HLA class I antigens.Transfusion 1993;33: 910–914.

    147. Butson MJ,Yu PK,Cheung T et al.Dosimetry of blood irradiation

    with radiochromic film.Transfus Med 1999;9: 205–208.

    148. Akahoshi M,Takanashi M,Masuda M et al.A case of transfusion-

    associated graft-versus-host disease not prevented by white cell-

    reduction filters.Transfusion 1992; 32: 169–172.

    149. Grass JA,Wafa T, Reames A et al. Prevention of transfusion-

    associated graft-versus-host disease by photochemical

    treatment. Blood 1999; 93:3140–3147.

    150. Philipps E, Fleischner FG. Pulomonary edema in the course of ablood transfusion without overloading the circulation.Dis Chest

    1966; 50: 619–623.

    151. Popovsky MA,Moore SB. Diagnostic and pathogenetic

    considerations in transfusion-related acute lung injury.

    Transfusion 1985; 25: 573–577.

    152. Popovsky MA,Chaplin HC,Moore SB.Transfusion-related acute

    lung injury: a neglected, serious complication of hemotherapy.

    Transfusion 1992; 32: 589–592.

    153. Silliman CC.Transfusion-related acute lung injury.Transfus Med

    Rev 1999;13: 177–186.

    154. Seeger W, Schneider U,Kreusler B et al.Reproduction of 

    transfusion-related acute lung injury in an ex vivo lung model.

    Blood 1990; 76: 1438–1444.

    155. Dry SM,Bechard KM, Milford EL,Churchill WH, Benjamin RJ.

    The pathology of transfusion-related acute lung injury.Am J Clin

    Pathol 1999; 112:216–221.

    156. Silliman CC,Voelkel NF,Allard JD et al. Plasma and lipids from

    stored packed red blood cells cause acute lung injury in an

    animal model. J Clin Invest 1998; 101: 1458–1467.

    157. Ramanathan RK,Triulzi DJ, Logan TF.Transfusion-related acute

    lung injury following random donor platelet transfusion: a report

    of two cases.Vox Sang 1997; 73: 43–45.

    158. Densmore TL, Goodnough LT,Ali S,Dynis M,Chaplin H.

    Prevalence of HLA sensitization in female apheresis donors.

    Transfusion 1999; 39: 103–106.

    159. Sazama K. Reports of 355 transfusion-associated deaths: 1976

    through 1985.Transfusion 1990;30: 583–590.

    Non-infectious transfusion reactions

    83 2001 Harcourt Publishers Ltd Blood Reviews (2001) 15 69–83