INTRODUCTION

Plasma is the liquid component of blood that contains many dissolved proteins including albumin, immunoglobulins, coagulation/clotting factors, inhibitors of coagulation (anticoagulants) and complement factors. 
The most common rationale for plasma transfusion is to provide a source of pro-coagulant factors in patients with presumed deficiencies, as tested for by standard laboratory-based coagulation assays, typically the prothrombin time (PT) and/or viscoelastic haemostatic assays (VHAs). 
Plasma transfusions may be given to treat active bleeding attributable to coagulation factor deficiencies (therapeutic transfusion) or to prevent bleeding in patients considered at heightened risk of bleeding (prophylactic transfusion), where specific factor replacement is not available.  Plasma may also be used to replace other elements missing due to disease or its therapy, and is used as the first-line replacement fluid for plasma exchange in the treatment of TTP. Plasma may less commonly be used to replace natural anticoagulants, such as in the setting of congenital protein C deficiency, if specific plasma-derived or recombinant alternatives are not available. 
Data to support plasma transfusion practice are scarce, yet widespread use of plasma in varying clinical settings occurs without substantiating evidence, and audit data show that many transfusions are given for inappropriate indications or at inappropriate doses. 

TYPES AND DESCRIPTION

Plasma components for transfusion are preferably prepared from blood donors meeting eligibility criteria for voluntary donation, either by centrifugation of whole blood or by apheresis collection. To reduce the risk of transfusion-associated lung injury (TRALI), which is linked to the presence of human leucocyte antigen (HLA) and human neutrophil antigen (HNA) alloantibodies, many centres/countries utilize male donor plasma or plasma from never-pregnant women. 

Plasma is typically frozen within a few hours to maintain coagulation factor activity and prolong storage duration. When needed, it is thawed for clinical use. However, there are a number of different plasma components available internationally. Each product varies in collection method, manufacturing process (including time to freezing), product volume, diluent, anticoagulant properties, potency of coagulation proteins, storage and thawing requirements (Table 1) [1]. For this reason, the specific standards and relevant local guidelines for the type of plasma manufactured in each country should be referenced. 

Factor VIII level is used as a quality measure of plasma for transfusion because it is the most labile factor, and immediately after thawing, standard fresh frozen plasma (FFP) must have a level of at least 70% of normal fresh plasma. However, it remains debatable whether factor VIII levels provide the most clinically relevant information for quality control. To preserve the activity of the labile coagulation factors (factor V and factor VIII), plasma is frozen soon after collection, typically within 6 to 8 h of collection for FFP or 24 h for FP24.

Table 1. Types of plasma transfusion

Plasma product Production Special information about coagulation activity
FFP Frozen to ≤ –18°C within 6–8 h 
Stored at ≤ –18°C 
Transfused within 24 h of thawing
Normal amounts of coagulation factors including VWF
Normal antithrombin and ADAMTS-13 levels
Plasma frozen within 24 h (FP24) Frozen within 24 h of collection 
Stored at 4–6°C for immediate use and then frozen to ≤ –18°C 
Reduced FV and FVIII levels 
Cryo-depleted plasma FFP is thawed to 1–6°C, allowing the cold-induced precipitate to separate by centrifugation
Remaining supernatant plasma is refrozen to ≤ –18°C
Deficient in fibrinogen, FVIII, FXIII and VWF
Thawed plasma FFP or FP24 that has been thawed
Stored at 1–6°C 
Expires 5 days after thawing 
Deficient in FV and FVIII
Liquid (never frozen) plasma Made from WB within 5 days of donation
Stored at 1–6°C
Expires up to 5 days after WB’s expiry date
Deficient FV and FVIII
Cryoprecipitate Cold insoluble protein that precipitates when FFP is thawed to 1–6°C
Cryoprecipitate is then refrozen to ≤ –18°C
Concentrated levels of fibrinogen, FVIII, FXIII and VWF
Dried plasma Pooled plasma is frozen and dehydrated by sublimation under vacuum
Stored between 2°C and 25°C.
Pathogen reduction strategies vary depending on product
Normal coagulation factors
 

Abbreviations: FFP, fresh frozen plasma; FV, factor V; FVIII, factor VIII; FXIII, factor XIII; VWF, von Willebrand factor; WB, whole blood.

Pathogen-inactivated plasma

Pathogen-reduced plasma products are used in some countries to provide an additional level of risk reduction against viral transmission, by reducing the infectivity of residual pathogens in blood components. Major available pathogen inactivation systems currently in place include: methylene blue and light (M/B) treatment, solvent/detergent (S/D) treatment, amotosalen and riboflavin treatment (Table 2) [2].

The most commonly used methods of pathogen reduction are effective at eliminating the risk of transmission of lipid-enveloped viruses, such as human immunodeficiency virus (HIV) and hepatitis C. However, S/D and M/B treatments have variable efficacy on non-lipid-enveloped viruses such as hepatitis A and parvovirus. 

All the pathogen-inactivation strategies involve some loss or degradation of coagulation factors and reduced plasma quality. For example, decreased factor VIII activity to levels of 67–78% and fibrinogen levels of 65–84% have been reported [2].

Table 2. Pathogen-reduction techniques

Plasma product Plasma product Special information about coagulation activity
S/D-treated FFP  Pharmaceutical product 
Large plasma pools, many donors
 
Reduced FV, FVIII, protein S, VWF activity and high VWF multimers
Solvent detergent technology with 1% tri-n-butyl phosphate and 1% Triton X -100
M/B-treated FFP Single donor plasma donation
 
Reduced FVIII and fibrinogen level with altered fibrinogen structure
Plasma treated with methylene blue and exposed to white light Methylene blue is removed by special filter
Amotosalen 
(InterceptTM)
Single donor plasma donation
 
Reduced FVII, FVIII and fibrinogen
Plasma treated with amotosalen followed by ultraviolet illumination
Amotosalen is removed by adsorption
Riboflavin
(MirasolTM)
Single donor plasma donation  Reduced FV, FVIII, FIX, FXI and fibrinogen 
Plasma treated with riboflavin (vitamin B2) followed by ultraviolet illumination 
Riboflavin does not require specific removal 

Abbreviations: FFP, fresh frozen plasma, FV, factor V; FVII, factor VII, FVIII, factor VIII; FIX, factor IX; FXI, factor XI; M/B, methylene blue and light; S/D, solvent/detergent; VWF, von Willebrand factor.

Dried plasma

Plasma may be lyophilized or spray-dried to preserve the plasma’s coagulation properties and allow transport and storage in areas where freezers are not readily available. Lyophilized plasma can be stored at room temperature, has a longer shelf life, has a rapid reconstitution time, and is pathogen-reduced [3].

SELECTION OF PLASMA COMPONENTS

Plasma selected for transfusion must be compatible with the recipient’s red cells. Whilst the first choice of plasma for a patient is plasma that is ABO-identical with the recipient, if not possible, then plasma of a different ABO group may be acceptable if the product has been shown not to possess ‘high-titre’ anti-A or anti-B. Group O plasma should only ever be given to group O patients.

AB plasma is the universal plasma donor group as it does not contain anti-A or anti-B, and may be used when a patient’s ABO type is unknown, for example, in initial trauma resuscitation where immediate transfusion is required. It is important that a patient’s ABO group is determined as soon as possible, so that group-specific plasma may be provided when practical. Some hospitals, especially trauma services, keep pre-thawed group AB plasma available for these scenarios. However, the challenge is that group AB plasma is a scarce resource and pre-thawing plasma for emergency use may lead to unnecessary wastage. For this reason, some transfusion services will utilize group A plasma in initial trauma resuscitation regardless of the patient’s ABO group. A multi-centre retrospective study examining the use of group A plasma during initial resuscitation of trauma patients of unknown ABO group, found no increase in early or in-hospital mortality or length of hospital stay between group B or AB patients compared to group A who received at least one unit of group A plasma during their resuscitation [4].

RISKS OF PLASMA TRANSFUSION

Allergic reactions can occur following plasma transfusion. Usually these are mild reactions but occasionally severe allergic reactions or anaphylaxis can occur following plasma transfusion.  Other risks include transfusion-associated circulatory overload (TACO) and TRALI, and transmission of infectious agents. See Table 3 for important risks related to plasma transfusion. Avoidance of unnecessary plasma transfusion is the best measure to reduce risk. 

Table 3 Risks of plasma transfusion 

Risks of plasma transfusion Saadah et al., 2018 [43]
ISTARE haemovigilance data (estimate rounded to the nearest 100)
Saadah et al., 2017 [42]
Systematic review and meta-analysis (estimate rounded to nearest 100)
Allergic reactions 1/3600 1/1100 (FFP)a
Anaphylaxis Estimate not provided 1/125,000
Febrile non-haemolytic transfusion reaction 1/21,900 1/8333 (FFP)b
TACO 1/128,2000 1/16,900
Transfusion-associated dyspnoea 1/144,900 Not reported
TRALI 1/ 212,800 1/55,600c–1/1,000,000d
Hypotensive reactions 1/227,300 Not reported
Acute or delayed haemolytic transfusion reaction 1/833,300 Not reported
Transfusion-transmitted infection (viral or bacterial) Combined estimate not provided Not reported

aLower rates for solvent-detergent (S/D)-treated plasma (1/3100) and methylene-blue (MB)-treated plasma (1/7100).
bNo reports of febrile non-haemolytic transfusion reaction with SD or MB plasma. 
cMixed-sex plasma.
dMale-only plasma.
Abbreviations: FFP, fresh frozen plasma; TACO, transfusion-associated circulatory overload; TRALI, transfusion-associated lung injury.

THERAPEUTIC PLASMA TRANSFUSIONS FOR BLEEDING

Plasma transfusion in the setting of trauma and massive haemorrhage

Plasma transfusion in the setting of trauma and massive haemorrhage

Plasma is integral to the transfusion-supportive management pathways defined by massive transfusion protocols (MTPs). The biological rationale is correction of multi-factor coagulopathy. Acute traumatic coagulopathy (ATC) refers to the coagulation derangement that develops rapidly following significant traumatic insult, and is evident in over 25% of trauma patients arriving to the hospital emergency department (before fluid resuscitation) [5]. Patients who arrive at hospital with ATC are four times more likely to die compared to those without acute coagulopathy; they have significantly higher transfusion requirements, in addition to an increased incidence of organ dysfunction [5]. Many studies report that resuscitation without plasma, or with low-dose plasma compared to the number of red blood cells (RBCs) transfused, is associated with poorer clinical outcomes [6–8].

MTPs have been developed to provide rapid blood product resuscitation in the setting of major haemorrhage. The number, type, timing and ratio of blood components issued in each MTP pack will depend on the clinical setting, institution and local guidelines. Plasma is important in initial haemostatic resuscitation of trauma patients and may be given as part of MTP packs in fixed-ratio component resuscitation.  Fixed-ratio component transfusion attempts to mimic resuscitation with whole blood. For each RBC unit a set number of plasma and platelet unit(s) is administered.  In contrast, goal-directed component therapy is often based on the use of VHAs, rotational thromboelastography (ROTEM) and thromboelastography (TEG). These allow early diagnosis and management of ATC by providing global and functional assessments of coagulation and allow directed transfusion therapy. However, the Implementing Treatment Algorithms for the Correction of Trauma-Induced Coagulopathy (iTACTIC) trial, a multicentre randomized controlled trial (RCT), evaluated trauma patients who received MTPs augmented by VHAs compared with conventional coagulation tests and found no difference in mortality or other clinical outcomes between study arms [9].

Robust clinical trial evidence to support a clear role for high-dose or empiric plasma transfusion remains a subject of debate. The Pragmatic, Randomized Optimal Platelets and Plasma Ratios (PROPPR) trial, a large multicentre RCT, was conducted to evaluate the effect of two different fixed ratios on massive haemorrhage. Trauma patients were randomized to receive a ratio of 1:1:1 or 1:1:2 of platelets: plasma and RBCs, with the primary outcome being mortality [10]. There was no statistically significant difference in 24-h or 30-day mortality between transfusion strategies.  However, in post hoc analyses, there were fewer deaths from bleeding at 24 h with the 1:1:1 transfusion strategy [10]. More recently, trials have focused on plasma transfusion delivery prior to hospital.  The Pre-hospital Air Medical Plasma (PAMPer) trial was a multicentre, cluster-randomized trial, designed to determine the efficacy and safety of pre-hospital plasma compared with standard crystalloid resuscitation [6]. The study enrolled 501 severely injured patients and reported 30-day mortality was almost 10% lower in the plasma group compared to the standard-care group. However, interpretation is limited by incomplete outcome data, non-standardized fluid management in the standard-care group, and many patients seemingly not requiring plasma (or platelets) after admission and there were no effects on 24-h mortality (after adjustment) [6]. The Control of Major Bleeding After Trauma Trial (COMBAT) was a single-centre placebo-controlled RCT in 125 trauma patients with haemorrhagic shock [11]. There was no statistical difference in mortality at 28 days between the plasma group (15%) and crystalloid group (10%). Coagulation factors, transfusion requirements and safety outcomes were similar between groups [11]. However, comparing the two trials, hospital transit times were much shorter in the COMBAT trial, and only 32% of patients in the COMBAT plasma group had received the full dose of 2 units of plasma pre-hospital, compared with 84% in the PAMPer trial, which also featured longer transit times [6, 11].

Some transfusion services will maintain an inventory of thawed group AB or group A plasma for immediate transfusion.  The availability of thawed AB plasma in the emergency department has been demonstrated to expedite the time to transfusion of plasma components in severely injured patients and, in a single-centre retrospective study, was associated with better survival [12].  As noted above, high wastage rates are often observed unless the thawed plasma has an extended shelf-life.

Ongoing plasma transfusion requirements in the trauma and critical bleeding setting will be guided by clinical assessment and response, in addition to results of standard laboratory-based coagulation testing or VHA testing. 

Small RCTs have compared FFP with four-factor PCC in the management of bleeding cardiac surgery patients [13, 14]. Karkouti et al. included 101 patients randomized to FFP (median dose 12.5 mL/kg) or PCC (median dose 24.9 IU/kg). Patients receiving PCC had less chest tube drainage and reduced red cell transfusion requirements in the first 24 h after surgery, without safety concerns [14]. The PROPHESY study, a pilot feasibility trial where bleeding adult cardiac surgery patients were randomized to 15 mL/kg FFP or four-factor PCC (500 IU weight <60 kg; 1000 IU if 61–90 kg; and 1500 IU if >90 kg). The study demonstrated feasibility for recruitment in a larger clinical trial with no safety concerns, but due to its size it was not powered to make any conclusions about bleeding or transfusion outcomes [13]. Larger, well-designed clinical trials are warranted to evaluate the role, efficacy, and safety of PCCs compared with plasma in cardiac surgery. 

Disseminated intravascular coagulopathy

Disseminated intravascular coagulopathy

Disseminated intravascular coagulopathy (DIC) is a poorly defined entity. The International Society of Thrombosis and Haemostasis consensus guidelines define acute coagulopathy as a prolonged PT/activated partial thromboplastin time (APTT; >1.5 times normal) or decreased fibrinogen (<1.5 g/L) [15]. DIC is an acquired syndrome characterized by coagulation and haemostatic dysfunction, with risks of both haemorrhage and thrombosis. There is systemic coagulation activation with coagulation factor and platelet consumption and a propensity to bleed, in addition to microvascular thrombosis and a risk of end-organ ischaemia. DIC may occur as a complication of sepsis, solid tumours, pregnancy complications, trauma, severe liver disease and vascular abnormalities, as well as others.  Diagnosis involves both clinical features and abnormalities of laboratory parameters: PT, platelet count, fibrinogen and fibrin-related markers, such as fibrinogen degradation products and D-dimer [15].

The mainstay of treatment for established DIC is aggressive and timely treatment of the underlying clinical condition [15]. Plasma transfusions may have a role in the treatment of active bleeding associated with acute coagulopathy and prior to invasive procedures in patients at risk of bleeding complications [15].

Response to plasma transfusion should be monitored clinically and be followed by reassessment of the patient’s coagulation profile.  

Warfarin reversal

Warfarin reversal

Warfarin is the most commonly prescribed oral vitamin K antagonist and is used to prevent arterial and venous thromboembolism in a range of settings. It exhibits its anticoagulant effect by inhibiting the vitamin K-facilitated carboxylation of factors II, VII, IX and X. 

A key component of any urgent warfarin reversal strategy is prompt intravenous vitamin K (phytonadione) administration, which allows synthesis of new, functional vitamin K-dependent coagulation proteins.  However, in the setting of life- or limb-threatening bleeding, more rapid reversal is required to restore the coagulation potential of plasma. PCCs are virally inactivated products containing concentrated amounts of vitamin K-dependent factors either formulated with four factors (factors II, VII, IX and X) or three factors (II, IX and X). PCCs have the advantage of a small volume for infusion, rapid reconstitution, ease of administration, fast onset of action, no requirements for ABO compatibility, and minimal risk of viral transmission, TRALI or TACO.  

Two RCTs have evaluated urgent warfarin reversal with FFP compared to PCC in patients undergoing cardiac surgery [16] or with mechanical heart valves [17]. Demeyere randomized 40 patients to receive two units (2 × 200 mL) S/D-treated FFP or four-factor PCC (dose individualized based on international normalized ratio [INR] and weight) prior to cardiopulmonary bypass and after bypass. PCC was found to reverse warfarin-related coagulopathy quicker and more patients in the PCC group reached the target INR than in the FFP group [16]. Fariborz Farsad et al. compared three-factor PCC (dose determined by INR and weight) versus FFP (10–15 mL/kg) in patients with mechanical heart valves requiring urgent reversal and found PCCs were more effective at reversal to a target INR of <2.5 than plasma and more patients in the plasma arm required additional doses to reach the target INR [17].

The INCH trial randomized patients with vitamin K antagonist-associated intracranial haemorrhage, to four-factor PCC (30 IU/kg) versus FFP (20 mL/kg) in addition to 10 mg vitamin K, with a primary endpoint of INR ≤1.2 within 3 h of treatment [18]. The study was stopped prematurely due to concern about increased haematoma expansion in the FFP arm.  Analysis supported earlier correction of the INR with PCC and less haematoma expansion in those patients treated with PCC compared with FFP [18].

PCC (preferably four-factor) in addition to vitamin K should be used first-line for warfarin reversal. In cases of severe bleeding, such as intracranial haemorrhage, if PCC is not available, then plasma transfusion is appropriate to allow adequate factor VII replacement. However, reassessment of the INR and follow-up plasma or PCC doses may be needed. 

PROPHYLACTIC PLASMA TRANSFUSIONS TO PREVENT BLEEDING

A considerable amount of plasma is transfused in the absence of clinical bleeding to treat mild abnormalities of standard tests or prior to invasive procedures [19]. Its intention is to prevent peri-procedural bleeding, yet there is very limited evidence to show it is effective in normalizing abnormal coagulation tests or reducing bleeding. Indeed, it has been well described that conventional doses of plasma transfusion cannot even correct mild abnormalities of PT [20].

Preoperative coagulation testing and plasma transfusion

Preoperative coagulation testing and plasma transfusion

The two most common global coagulation tests used in clinical practice are the PT and APTT. The PT detects deficiencies in factors II, VII, IX and X, and varies depending on the laboratory coagulation platform and type of thromboplastin used.  The INR was developed to standardize the assessment and monitoring of warfarin and is the ratio of the patient’s PT compared to a normal PT and corrects for thromboplastin sensitivity. The APTT was developed to screen for congenital bleeding disorders, specifically factor VIII, IX or XI deficiency. It will also be prolonged with factor XII deficiency (not associated with bleeding), in the setting of coagulation factor inhibitors, heparin effect and phospholipid inhibitors (lupus anticoagulants). These laboratory tests were never developed to assess perioperative bleeding risk: the INR was developed to monitor warfarin and the APTT was designed to screen for bleeding disorders and monitor heparin.

Unselected coagulation testing with PT/INR and APTT prior to surgery or invasive procedures is not recommended, as these tests have not been validated to predict bleeding risk in surgical patients [20, 21]. Coagulation results may become abnormal with mildly reduced factor deficiencies at factor levels that are not associated with bleeding. Attempts to treat or correct these perceived laboratory-defined coagulopathic values are largely inappropriate and mildly increased INRs and mildly prolonged PTs do not normalize when treated with therapeutic doses of plasma [19, 22]. Plasma transfusions will only impact the INR when there is a relatively large difference between the patient’s INR and the coagulation activity of the plasma. Therefore, the higher the INR the more effective plasma is in reducing the INR [19, 23]. The effect of a plasma transfusion on an INR can be transient and therefore if plasma is used to correct an INR preoperatively, the plasma transfusion should occur immediately prior to the procedure to maximize its effect [24]. 

Plasma transfusion should not be given to correct abnormal coagulation tests in the absence of bleeding in neonates, children and adults. Evidence from clinical trials (RCTs and observational studies) has failed to support prophylactic plasma transfusions in minimizing bleeding outcomes in the following patients undergoing procedures: critically ill adults with an INR of 1.5 to 3.0 [25]; non-cardiac surgery and INR >1.5 [26]; patients with cirrhotic liver disease and central venous catheter placement [27]; and those undergoing procedures outside the operating room and an INR of 1.5 to 2.5 [28].

Transfusion practice based on  PT/INR thresholds for use of prophylactic plasma is likely to have very little impact on reducing the PT/INR when the abnormalities are mild/ moderate (e.g., INR of 1.5–2.4) [23]. Plasma is unlikely to be warranted in most surgical patients when the PT/INR is prolonged. There is very little evidence to support the use of prophylactic plasma transfusion perioperatively.  

Liver disease

Liver disease

The liver serves an important role in the manufacture of most pro-coagulant and anticoagulant proteins, in addition to proteins involved in fibrinolysis. Due to the reorganized balance of coagulation factors and associated anticoagulants, haemostasis is largely preserved in patients with chronic liver disease. Patients will frequently have prolongations of the PT and INR, yet as discussed previously, these tests poorly predict bleeding risk. A prospective observational study of critically ill patients with liver cirrhosis found that, instead of INR, fibrinogen levels <0.6 g/L, platelet count <30 × 109/L and APTT >100 s were the strongest predictors of major bleeding [29]. The conventional coagulation testing (PT/INRs and APTTs) do not reflect the parallel and compensatory reductions in the anticoagulants protein C, protein S and antithrombin. 

Global tests of haemostasis, such as thrombin generation potential and VHAs, are preserved in patients with chronic liver disease and may offer better predictors of bleeding risk, although they are yet to be validated in this setting [30]. In a small open-label RCT, 60 patients with cirrhosis and significant coagulopathy undergoing invasive procedures were randomized to a TEG-guided transfusion strategy or standard of care (guided by INR and platelet count). Those who received the TEG-guided transfusion strategy received significantly fewer blood products, without an increased risk of bleeding [31].

Plasma is frequently used prophylactically prior to procedures in patients with chronic liver disease, despite the absence of evidence from clinical trials to support its benefit.  Plasma transfusions at standard doses are largely ineffective in correcting the coagulopathy associated with chronic liver disease [32] and for this reason prophylactic plasma transfusions should not be used to correct abnormal coagulation tests in non-bleeding patients or prior to low-risk bleeding procedures. An RCT of cirrhosis patients undergoing ultrasonography-guided central venous catheter insertion, randomized to standard laboratory-based coagulation test-guided plasma transfusion 10 mL/kg when the INR was >1.5 or APTT was >32 s, TEG-guided transfusion when the CT – EXTEM was >80 s, or restrictive plasma transfusion when the INR was >5.0, found no difference in bleeding outcomes. [27].

Large volumes of plasma are required to significantly increase clotting factor levels in chronic liver disease, and this can increase portal vein pressures, which may in turn lead to an increased risk of gastrointestinal bleeding.  Plasma transfusions have a limited role of treating the coagulopathy associated with liver disease and should be saved as an adjunctive therapy for those patients with active bleeding. 

BLEEDING DISORDERS

Rare inherited bleeding disorders

Rare inherited bleeding disorders

The treatment of choice for inherited bleeding disorders is treatment with the specific fractionated factor concentrate, where available.  Single factor plasma-derived or recombinant concentrates are available for factors I, VII, VIII, IX, XI and XIII, although access to these products is limited in some countries [33]. Factor V deficiency is the only single-factor deficiency where a factor concentrate does not currently exist [33]. Plasma transfusion is indicated for the treatment of factor V deficiency to treat active bleeding or prevent bleeding prior to a procedure. 

Vitamin K deficiency bleeding

Vitamin K deficiency bleeding

Vitamin K is an essential vitamin, necessary to help the blood clot. Newborns have very low levels of vitamin K compared with adults, and deficiency in vitamin K can result in life-threatening bleeding in the first few hours to months of life. Early (<24 h after birth) vitamin K deficiency bleeding (VKDB) can occur in infants whose mothers are taking medications that interact with vitamin K metabolism, such as anticoagulants, anti-convulsants and anti-tuberculosis medications. Classic VKDB occurs between days 2 and 14 of life and may be due to poor feeding.  Early and late VKDB are associated with bleeding, most commonly gastrointestinal or umbilical bleeding.  However, late VKDB, onset between weeks 2 and 12, typically occurs in exclusively breast-fed babies, who most frequently have received no or inadequate vitamin K or have intestinal malabsorption defects. The typical presentation of late VKDB is with intracranial haemorrhage [34]. To prevent VKDB it is standard of care that all newborns receive vitamin K supplementation. The American Academy of Pediatrics recommends that all newborns receive a single dose of intramuscular vitamin K at birth [35]. In cases of suspected VKDB, immediate treatment with intravenous vitamin K is necessary and, in life-threatening cases, plasma transfusion may be necessary to rapidly replenish coagulation factors [36].

PROTEIN DEFICIENCIES

Protein C and S deficiency

Purpura fulminans is a rare and life-threatening condition characterized by multi-site, cutaneous purpuric lesions and acute DIC. Congenital purpura fulminans is due to homozygous protein C or protein S deficiency, or compound heterozygous states. Due to the associated DIC, these neonates typically have a prolonged PT and APTT, with low fibrinogen levels, elevated D-dimer and thrombocytopenia [37].

Empiric short-term treatment with plasma transfusion is appropriate for suspected inherited protein C or S deficiency and for treatment of post-infectious purpura fulminans until diagnosis is confirmed [37]. Commercially available plasma-derived, viral-inactivated protein C concentrate is licensed in some countries for treatment of congenital deficiency, but no specific protein S product is currently available. 

Other

Plasma transfusion has been reported in the setting of hereditary angioedema caused by C1 esterase inhibitor deficiency. First-line treatment is with a specific C1 esterase inhibitor, whenever available, however, if unavailable, plasma transfusion may be a temporizing alternative. 

THERAPEUTIC PLASMA EXCHANGE

TPE for many clinical indications typically involves removal of 1.0 or 1.5 patient plasma volumes during each procedure, with albumin replacement for most indications.  
Plasma is indicated as the fluid replacement in the treatment of TTP and is often considered when there is a risk of coagulation factor depletion (particularly hypofibrinogenaemia) after daily TPE. Similarly, plasma may be preferred if a patient has underlying coagulopathy, such as in the setting of acute liver failure or bleeding. See Table 4, for conditions where TPE is considered first- or second-line therapy and plasma is the suggested replacement fluid [38].

TTP is due to severe deficiency (<10%) of ADAMTS-13, the von Willebrand factor (VWF)-cleaving metalloprotease [39]. In most cases this is autoimmune in nature, and the presence of an autoantibody against ADAMTS-13 will be detected. First-line treatment for suspected TTP is daily TPE, and it should be instituted immediately to reduce the risk of death. TPE allows reduction in the high molecular VWF multimers, removal of the autoantibody and replacement with plasma containing normal ADAMTS-13 levels. Treatment continues until there is a demonstrated clinical response with sustained normalization of the platelet count and improved lactate dehydrogenase levels (<1.5 upper limit of normal) after cessation of TPE [39]. If immediate TPE cannot be commenced promptly, a plasma infusion may provide temporary increase in ADAMTS-13 levels. For patients with congenital TTP, intermittent plasma infusion may be appropriate, as there is no autoantibody to be removed. Specialist advice should be sought for the management of TTP.

Table 4 Indications where therapeutic plasma exchange is accepted as first-line or second-line treatment and plasma is the first-line replacement fluid. Adapted from the American Society for Apheresis 2019 Guideline [38].

Plasma is considered first-line replacement fluid
Condition Indication
Acute liver failure (high-volume TPE) Moderate to severe coagulopathy
Anti-glomerular basement membrane disease (Goodpasture’s syndrome) Diffuse alveolar haemorrhage
Catastrophic anti-phospholipid syndrome  
Hyperviscosity in hypergammaglobulinaemia Symptomatic or prophylaxis for rituximab if daily plasma exchange 
Thrombotic microangiopathy, complement-mediated Factor H autoantibodies
Thrombotic microangiopathy, drug-associated Ticlopidine
TTP  
Thyroid storm   
Vasculitis, ANCA-associated (AAV) Diffuse alveolar haemorrhage
Wilson’s disease, fulminant Fulminant

Abbreviations: ANCA, anti-neutrophil cytoplasmic antibody; TPE, therapeutic plasma exchange; TTP, thrombotic thrombocytopenic purpura.

CONDITIONS OR CASES WHERE PLASMA TRANSFUSION IS NOT INDICATED

Plasma transfusion is not indicated for the treatment of hypovolaemia or to be used as a replacement intravenous fluid. It does not have any role in enhancing wound healing, in treatment of hypoproteinaemia or immune deficiencies. 
Convalescent plasma (plasma from individuals recovered from coronavirus 19 [COVID-19] infection) does not reduce mortality or improve clinical outcomes in unselected patients with moderate-severe COVID-19 infection [40, 41]. Trials are ongoing in patients with immunodeficiencies at the time of writing.

ABBREVIATIONS

APTT activated partial thromboplastin time 
ATC acute traumatic coagulopathy
DIC disseminated intravascular coagulopathy
FFP fresh frozen plasma
INR international normalized ratio
M/B methylene blue and light
MTP massive transfusion protocol
PCC  prothrombin complex concentrate
PT    prothrombin time
RBC red blood cell
RCT randomized controlled trial
S/D solvent/detergent
TACO transfusion-associated circulatory overload
TEG thromboelastography
TPE  therapeutic plasma exchange
TTP  thrombotic thrombocytopenic purpura
VKDB vitamin K deficiency bleeding
VHA viscoelastic haemostatic assay
VWF von Willebrand factor

SUGGESTED READINGS

General

Therapeutic Plasma Transfusion

Disseminated intravascular coagulopathy

Warfarin reversal

Prophylactic transfusions

​​​​​​​Coagulation testing

Liver disease

Bleeding disorders

Protein deficiencies

Therapeutic plasma exchange

REFERENCES 

  1. Benjamin RJ, McLaughlin LS. Plasma components: properties, differences, and uses. Transfusion. 2012;52 (Suppl 1):9S-19S.
  2. Rock G. A comparison of methods of pathogen inactivation of FFP. Vox Sang. 2011;100:169-78.
  3. Pusateri AE, Given MB, Schreiber MA, Spinella PC, Pati S, Kozar RA, et al. Dried plasma: state of the science and recent developments. Transfusion. 2016;56 (Suppl 2):S128–39.
  4. Dunbar NM, Yazer MH, Biomedical Excellence for Safer Transfusion (BEST) Collaborative and the STAT Study Investigators. Safety of the use of group A plasma in trauma: the STAT study. Transfusion. 2017;57:1879–84.
  5. Brohi K, Cohen MJ, Davenport RA. Acute coagulopathy of trauma: mechanism, identification and effect. Curr Opin Crit Care. 2007;13:680–5.
  6. Sperry JL, Guyette FX, Brown JB, Yazer MH, Triulzi DJ, Early-Young BJ, et al. Prehospital Plasma during Air Medical Transport in Trauma Patients at Risk for Hemorrhagic Shock. N Engl J Med. 2018;379:315–26.
  7. Holcomb JB, del Junco DJ, Fox EE, Wade CE, Cohen MJ, Schreiber MA, et al. The prospective, observational, multicenter, major trauma transfusion (PROMMTT) study: comparative effectiveness of a time-varying treatment with competing risks. JAMA Surg. 2013;148:127–36.
  8. Borgman MA, Spinella PC, Perkins JG, Grathwohl KW, Repine T, Beekley AC, et al. The ratio of blood products transfused affects mortality in patients receiving massive transfusions at a combat support hospital. J Trauma. 2007;63:805–13.
  9. Baksaas-Aasen K, Gall LS, Stensballe J, Juffermans NP, Curry N, Maegele M, et al. Viscoelastic haemostatic assay augmented protocols for major trauma haemorrhage (ITACTIC): a randomized, controlled trial. Intensive Care Med. 2021;47:49–59.
  10. Holcomb JB, Tilley BC, Baraniuk S, Fox EE, Wade CE, Podbielski JM, et al. Transfusion of plasma, platelets, and red blood cells in a 1:1:1 vs a 1:1:2 ratio and mortality in patients with severe trauma: the PROPPR randomized clinical trial. JAMA. 2015;313:471–82.
  11. Moore HB, Moore EE, Chapman MP, McVaney K, Bryskiewicz G, Blechar R, et al. Plasma-first resuscitation to treat haemorrhagic shock during emergency ground transportation in an urban area: a randomised trial. Lancet. 2018;392:283–91.
  12. Radwan ZA, Bai Y, Matijevic N, del Junco DJ, McCarthy JJ, Wade CE, et al. An emergency department thawed plasma protocol for severely injured patients. JAMA Surg. 2013;148:170–5.
  13. Green L, Roberts N, Cooper J, Field J, Gill R, Klein A, et al. A pragmatic pilot phase II randomised controlled trial of prothrombin complex concentrates (PCC) versus fresh frozen plasma (FFP) in adult patients who are undergoing heart surgery (PROPHESY). Trials. 2019;20:684.
  14. Karkouti K, Bartoszko J, Grewal D, Bingley C, Armali C, Carroll J, et al. Comparison of 4-Factor Prothrombin Complex Concentrate With Frozen Plasma for Management of Hemorrhage During and After Cardiac Surgery: A Randomized Pilot Trial. JAMA Netw Open. 2021;4:e213936.
  15. Wada H, Thachil J, Di Nisio M, Mathew P, Kurosawa S, Gando S, et al. Guidance for diagnosis and treatment of DIC from harmonization of the recommendations from three guidelines. J Thromb Haemost. 2013; doi: 10.1111/jth.12155. Online ahead of print.
  16. Demeyere R, Gillardin S, Arnout J, Strengers PF. Comparison of fresh frozen plasma and prothrombin complex concentrate for the reversal of oral anticoagulants in patients undergoing cardiopulmonary bypass surgery: a randomized study. Vox Sang. 2010;99:251–60.
  17. Fariborz Farsad B, Golpira R, Najafi H, Totonchi Z, Salajegheh S, Bakhshandeh H, et al. Comparison between Prothrombin Complex Concentrate (PCC) and Fresh Frozen Plasma (FFP) for the Urgent Reversal of Warfarin in Patients with Mechanical Heart Valves in a Tertiary Care Cardiac Center. Iran J Pharm Res. 2015;14:877–85.
  18. Steiner T, Poli S, Griebe M, Husing J, Hajda J, Freiberger A, et al. Fresh frozen plasma versus prothrombin complex concentrate in patients with intracranial haemorrhage related to vitamin K antagonists (INCH): a randomised trial. Lancet Neurol. 2016;15:566–73.
  19. Stanworth SJ, Grant-Casey J, Lowe D, Laffan M, New H, Murphy MF, et al. The use of fresh-frozen plasma in England: high levels of inappropriate use in adults and children. Transfusion. 2011;51:62–70.
  20. Segal JB, Dzik WH, Transfusion Medicine/Hemostasis Clinical Trials Network. Paucity of studies to support that abnormal coagulation test results predict bleeding in the setting of invasive procedures: an evidence-based review. Transfusion. 2005;45:1413–25.
  21. Chee YL, Crawford JC, Watson HG, Greaves M. Guidelines on the assessment of bleeding risk prior to surgery or invasive procedures. British Committee for Standards in Haematology. Br J Haematol. 2008;140:496–504.
  22. Abdel-Wahab OI, Healy B, Dzik WH. Effect of fresh-frozen plasma transfusion on prothrombin time and bleeding in patients with mild coagulation abnormalities. Transfusion. 2006;46:1279–85.
  23. Karam O, Demaret P, Shefler A, Leteurtre S, Spinella PC, Stanworth SJ, et al. Indications and Effects of Plasma Transfusions in Critically Ill Children. Am J Respir Crit Care Med. 2015;191:1395–402.
  24. Bryan AW Jr, Staley EM, Kennell T Jr, Feldman AZ, Williams LA 3rd, Pham HP. Plasma Transfusion Demystified: A Review of the Key Factors Influencing the Response to Plasma Transfusion. Lab Med. 2017;48:108–112.
  25. Muller MC, Arbous MS, Spoelstra-de Man AM, Vink R, Karakus A, Straat M, et al. Transfusion of fresh-frozen plasma in critically ill patients with a coagulopathy before invasive procedures: a randomized clinical trial (CME). Transfusion. 2015;55:26–35; quiz 25.
  26. Jia Q, Brown MJ, Clifford L, Wilson GA, Truty MJ, Stubbs JR, et al. Prophylactic plasma transfusion for surgical patients with abnormal preoperative coagulation tests: a single-institution propensity-adjusted cohort study. Lancet Haematol. 2016;3:e139–48.
  27. Rocha LL, Neto AS, Pessoa CMS, Almeida MD, Juffermans NP, Crochemore T, et al. Comparison of three transfusion protocols prior to central venous catheterization in patients with cirrhosis: A randomized controlled trial. J Thromb Haemost. 2020;18:560–70.
  28. Carson JL, Ness PM, Pagano MB, Philipp CS, Bracey AW, Jr, Brooks MM, et al. Plasma trial: Pilot randomized clinical trial to determine safety and efficacy of plasma transfusions. Transfusion. 2021;61:2025–34.
  29. Drolz A, Horvatits T, Roedl K, Rutter K, Staufer K, Kneidinger N, et al. Coagulation parameters and major bleeding in critically ill patients with cirrhosis. Hepatology. 2016;64:556–68.
  30. Mallett SV, Chowdary P, Burroughs AK. Clinical utility of viscoelastic tests of coagulation in patients with liver disease. Liver Int. 2013;33:961–74.
  31. De Pietri L, Bianchini M, Montalti R, De Maria N, Di Maira T, Begliomini B, et al. Thrombelastography-guided blood product use before invasive procedures in cirrhosis with severe coagulopathy: A randomized, controlled trial. Hepatology. 2016;63:566–73.
  32. Youssef WI, Salazar F, Dasarathy S, Beddow T, Mullen KD. Role of fresh frozen plasma infusion in correction of coagulopathy of chronic liver disease: a dual phase study. Am J Gastroenterol. 2003;98:1391–4.
  33. World Federation of Hemophilia. Rare Clotting Factor Deficiencies - Treatment Options: World Federation of Hemophilia; 2018 [cited 2018]. Available from https://www.wfh.org/en/page.aspx?pid=668.
  34. Loughnan PM, McDougall PN. Epidemiology of late onset haemorrhagic disease: a pooled data analysis. J Paediatr Child Health. 1993;29:177–81.
  35. American Academy of Pediatrics Committee on Fetus and Newborn. Controversies concerning vitamin K and the newborn. American Academy of Pediatrics Committee on Fetus and Newborn. Pediatrics. 2003;112(1 Pt 1):191–2.
  36. Williams MD, Chalmers EA, Gibson BE, Haemostasis, Thrombosis Task Force BCfSiH. The investigation and management of neonatal haemostasis and thrombosis. Br J Haematol. 2002;119:295–309.
  37. Chalmers E, Cooper P, Forman K, Grimley C, Khair K, Minford A, et al. Purpura fulminans: recognition, diagnosis and management. Arch Dis Child. 2011;96:1066–71.
  38. Padmanabhan A, Connelly-Smith L, Aqui N, Balogun RA, Klingel R, Meyer E, et al. Guidelines on the Use of Therapeutic Apheresis in Clinical Practice - Evidence-Based Approach from the Writing Committee of the American Society for Apheresis: The Eighth Special Issue. J Clin Apher. 2019;34:171–354.
  39. Scully M, Cataland S, Coppo P, de la Rubia J, Friedman KD, Kremer Hovinga J, et al. Consensus on the standardization of terminology in thrombotic thrombocytopenic purpura and related thrombotic microangiopathies. J Thromb Haemost. 2017;15:312–22.
  40. Piechotta V, Iannizzi C, Chai KL, Valk SJ, Kimber C, Dorando E, et al. Convalescent plasma or hyperimmune immunoglobulin for people with COVID‐19: a living systematic review. Cochrane Database Syst Rev. 2021; 5(5):CD013600.
  41. Axfors C, Janiaud P, Schmitt AM, Van't Hooft J, Smith ER, Haber NA, et al. Association between convalescent plasma treatment and mortality in COVID-19: a collaborative systematic review and meta-analysis of randomized clinical trials. BMC Infect Dis. 2021;21:1170.
  42. Saadah NH, van Hout FMA, Schipperus MR, le Cessie S, Middelburg RA, Wiersum-Osselton JC, et al. Comparing transfusion reaction rates for various plasma types: a systematic review and meta-analysis/regression. Transfusion. 2017;57:2104–14.
  43. Saadah NH, van der Bom JG, Wiersum-Osselton JC, Richardson C, Middelburg RA, Politis C, et al. Comparing transfusion reaction risks for various plasma products - an analysis of 7 years of ISTARE haemovigilance data. Br J Haematol. 2018;180:727–34.

THE AUTHORS

Gemma L. Crighton

Gemma L. Crighton

Royal Children’s Hospital, Melbourne, Victoria, Australia, Transfusion Research Unit, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia

Rajendra Chaudhary

Rajendra Chaudhary

Department of Transfusion Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India

Simon J. Stanworth

Simon J. Stanworth

John Radcliffe Hospital, NHS Blood and Transplant Oxford, Oxford, UK, Oxford University Hospitals NHS Foundation Trust, Oxford, UK, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK

 

Other chapters

Platelet transfusion

Read more

 

Principles of appropriate use of blood

Read more