Platelet transfusions are commonly administered for the prevention or treatment of bleeding in patients with acquired thrombocytopenia across a range of clinical contexts. Recent data, including randomized trials, have highlighted uncertainties in the risk-benefit balance of this therapy, which is the subject of this review. Hemovigilance systems report that platelets are the most frequently implicated component in transfusion reactions. There is considerable variation in platelet count increment after platelet transfusion, and limited evidence of efficacy for clinical outcomes, including prevention of bleeding. Bleeding events commonly occur despite the different policies for platelet transfusion prophylaxis. The underlying mechanisms of harm reported in randomized trials may be related to the role of platelets beyond hemostasis, including mediating inflammation. Research supports the implementation of a restrictive platelet transfusion policy. Research is needed to better understand the impact of platelet donation characteristics on outcomes, and to determine the optimal thresholds for platelet transfusion before invasive procedures or major surgery (eg, laparotomy). Platelet transfusion policies should move toward a risk-adapted approach that does not focus solely on platelet count.

The purpose of this article is to provide a practical, evidence-based approach for the decision to administer platelet transfusion in patients with acquired thrombocytopenia. Platelet counts remain remarkably stable during life, and reference ranges for thrombocytopenia are typically defined by the lower limits: normal platelet count, 150 × 109/L; moderate thrombocytopenia, <50 × 109/L; and severe thrombocytopenia, <20 × 109/L. Approximately 100 billion platelets are produced daily by the adult bone marrow, many of which are stored in the spleen.1,2 By comparison the average yield of platelets in a platelet transfusion unit is ∼3 × 1011. Platelets are essential for primary hemostasis and maintaining vascular integrity. Therefore, treating thrombocytopenia and reducing bleeding risk with a platelet transfusion has biological plausibility, as first reported in a case study of a young man with life-threatening epistaxis in in 1910.3 Advances in processing and storage of platelet components have underpinned the expansion of platelet banking.4 Annually, >2 million platelet transfusions are administered in the United States5 and 300 000 in the United Kingdom at considerable cost, yet there are concerns about security of supply, as has been apparent during the COVID-19 pandemic.6 There is a need for a better understanding of the role of platelet transfusions for both patients (recipients) and donors.

The case studies described herein review the common underlying assumptions that drive our current platelet transfusion practice, which can be summarized as follows:

  • Thrombocytopenia predicts bleeding.

  • Platelet transfusions consistently raise platelet counts (efficacy).

  • Platelet transfusions prevent or treat clinical bleeding (clinical/cost effectiveness), without causing harm (safety).

The heterogeneity of the platelet component is often underappreciated. Platelet units are either whole blood–derived (by the buffy coat method in the United Kingdom and Canada or from platelet-rich plasma in the United States; 4 to 6 donations are pooled for an adult dose) or by apheresis (obtained from a single donor). Nearly all platelet units undergo leukocyte reduction before storage. During storage in plasma or different media, platelets undergo varying degrees of biochemical, structural, and functional changes, commonly known as the storage lesion.7,8 There is ongoing research interest into the efficacy of cold-stored platelets by comparison with standard room temperature storage.9 There is variation in platelet count between normal individuals that will affect yield after donation. Platelets from different donors exhibit many biological differences, for example the degree of responsiveness (ie, donors with highly responsive platelets have a higher level of activated platelets).10,11 In a proof-of-principle, semirandomized trial, patients with nonbleeding thrombocytopenia with myelodysplasia were randomly allocated to receive a platelet transfusion from a high- or low-response donor.12 It was hypothesized that platelets donated by a high responder would be cleared more quickly, but no differences in platelet count increments were seen after transfusion between high- and low-responder donations, although the lack of change in count may reflect the study population under evaluation and important differences may apply, for example, in patients with acute bleeding.

Other important donor/donation characteristics may affect affect recipient outcomes. Donor age and sex may affect platelet count, size, and function.13 Inflammatory cytokines may be higher in platelets from female donors.14 Because of the pressures on platelet availability and supply, substitution of platelets of a different blood group may be needed for transfusion, such that various proportions (up to a half) of all administered platelet transfusions may not be fully ABO identical.15,16 Yet, this may not represent ideal practice, and better ABO matching, however defined, may have important clinical benefits.17 Pathogen reduction technology has been applied to platelets to reduce current and future infection risks, and research is addressing safety and efficacy in different groups of recipients, and effects on platelet refractoriness.18 

A 47-year-old man with myeloma received an autologous stem cell transplant (SCT). He had experienced minor reactions to platelet transfusions and minor bleeding episodes in the past, but reported no spontaneous bleeding today. His posttransplant platelet count of 13 × 109/L yesterday was 7 × 109/L the next day. What are the factors to consider in this case when deciding on the need for a platelet transfusion?

Clinicians often assume that a low platelet count predicts the risk of spontaneous (or nontraumatic) bleeding, but the strength of this relationship is unclear. In The Platelet Dose (PLADO) randomized controlled trial (RCT), there was no pattern of decreased bleeding with increased platelet count in the range of 6 × 109/L to 80 × 109/L in adult or pediatric patients.19-21 Even at counts <5 × 109/L, the increased uptick in bleeding rates was minimal. Bleeding was more common among allograft SCT recipients and in children, suggesting that clinical factors other than platelet count are important determinants of bleeding risk.22 A secondary analysis of The Trial of Prophylactic vs No-prophylactic Platelet Transfusions in Patients with Hematological Malignancies (TOPPS) explored risk factors for bleeding, including platelet count.23,24 The results indicated that a range of clinical factors are relevant to an increased number of days of bleeding, such as treatment plan (allogeneic hematopoietic SCT/chemotherapy), female sex, and pyrexia. The number of days with a platelet count <10 × 109/L was associated with developing a World Health Organization (WHO) grade 2 to 4 bleed, perhaps suggesting that the cumulative burden of thrombocytopenia, rather than isolated low platelet counts, alongside a history of recent bleeding events, would be a better guide for the decision for a prophylactic platelet transfusion. The lack of a clear relationship between severity of thrombocytopenia and risk of spontaneous bleeding also extends to other patient populations, with a poor correlation between the degree or severity of thrombocytopenia and the risk of bleeding or interventricular hemorrhage (IVH) in neonates (see later section “Case 2: pediatric patients and preterm neonates”).

Platelets are the most commonly implicated component associated with transfusion reactions (Figure 1).25 Febrile nonhemolytic transfusion reactions and allergic reactions may occur at a reported frequency of 1 in 14 and 1 in 50 per transfusion unit, respectively.5 Although some of these reactions may be seen as minor by clinicians, recipients of platelet transfusion say that these episodes can be distressing, particularly when repeated. There is no evidence that prophylactic antipyretics or antihistamines reduce the incidence of transfusion reactions,26,27 but data suggest that fewer reactions occur in platelet products stored in platelet additive solutions, which contain less plasma.28 Sepsis from a bacterially contaminated platelet unit is the most frequent infectious complication from transfusing any blood product.29 

Figure 1.

Number of reactions reported per 10 000 components issued in the United Kingdom from 2011 through 2020. Although red blood cells are the most common blood component transfused, platelets account for the highest number of reactions. Convalescent plasma is not included. Reproduced with permission from SHOT.25 FFP, fresh frozen plasma.

Figure 1.

Number of reactions reported per 10 000 components issued in the United Kingdom from 2011 through 2020. Although red blood cells are the most common blood component transfused, platelets account for the highest number of reactions. Convalescent plasma is not included. Reproduced with permission from SHOT.25 FFP, fresh frozen plasma.

Close modal

Although threshold-driven platelet transfusion forms the basis of current practice, it is essential to recall that early investigators were never able to observe a threshold effect.30 Available data (3 RCTs, 499 participants) suggest that lower (standard thresholds) of 10 × 109/L are not associated with more bleeding than are higher thresholds (20 × 109/L to 30 × 109/L).31 The threshold of 10 × 109/L is now commonly recommended by clinical guidelines (see Table 1).6 Yet, it is debatable whether some of these trials were adequately powered, particularly when bleeding is used as an outcome measure. A reanalysis of 1 trial suggested that differences may become apparent in a larger study.32 An important issue is what constitutes clinically important bleeding in patients with bone marrow failure or chemotherapy-induced thrombocytopenia. The WHO system for grading the severity of different bleeding events is often used, although it was developed as a system for reporting adverse events.33 Limitations include broad categories that may miss small changes in bleeding and definitions, such as bleeding requiring a transfusion, which, although pragmatic, may limit standardization, as thresholds for transfusion vary. Clinically significant bleeding in RCTs has often been applied as WHO grade ≥2, but such an outcome is a composite of different grades of bleeding.34 Overall, RCTs have reported highly different baseline rates of bleeding, raising further questions about the methodology and reporting of bleeding events.32,33 There are also likely to be differences between patients’ and clinicians’ perceptions of bleeding severity. The impact of bleeding on patients’ quality of life is unclear, as few studies have evaluated this aspect.35 Of course, the more severe types of bleeding events (eg, intracranial or intracerebral) are accepted as significant to patients and clinicians, but these events remain uncommon. Personal practices and review of severe bleeding events in RCTs reveals that major bleeding events often occur at platelet counts above the thresholds that would be considered indications for platelet transfusion. Finally, it is unclear whether less severe grades of bleeding predict more severe episodes. If there were data to support this progression of bleeding, it would have important implications for how we may use prophylactic platelet transfusions. Secondary analyses from the TOPPS RCT, found no evidence that minor bleeding predicted WHO grade 2 to 4 bleeding episodes,24 although an analysis of a different earlier data set showed different findings.36 

Table 1.

Examples of various recommendations in transfusion guidelines relevant to prophylactic platelet transfusions

Society/recommendationsStrength of recommendationQuality of evidence
European Society of Intensive Care Medicine 202061    
We suggest not using platelet transfusion to treat thrombocytopenia unless the platelet count falls below 10 × 109/L. Conditional Very low 
We make no recommendation regarding prophylactic platelet transfusion before invasive procedures for platelet counts between 10 × 109/L and 50 × 109/L. Research recommendation  
We suggest not giving prophylactic platelet transfusion before percutaneous tracheostomy or central venous catheter insertion for platelet counts between 50 100 × 109/L and 100 × 109/L. Conditional Very low 
Society of Interventional Radiology 2019124    
Consider platelet transfusion if platelet count is <20 × 109/L for procedures with low bleeding risk (eg, central venous access, including PICC placement, dialysis access, lumbar puncture, paracentesis, thoracentesis, transjugular liver biopsy, or superficial abscess drainage). Weak Limited (evidence level D) 
Consider platelet transfusion if platelet count is <50 × 109/L for procedures with high bleeding risk (eg, deep abscess drainage, solid organ biopsies, arterial intervention <7 French sheath, gastrostomy, urinary tract interventions [nephrostomy, stone removal], or transjugular intrahepatic portosystemic shunt). Weak Limited (evidence level D) 
British Society for Haematology 201787    
Consider performing the following procedures above the platelet count threshold indicated:   
Central venous lines, >20 × 109/L (using ultrasound) Strong Moderate 
Major surgery, >50 × 109/L Strong Low 
Lumbar puncture, ≥ 40 × 109/L Weak Low 
Insertion/removal of epidural catheter, ≥80 × 109/L Weak Low 
Neurosurgery or posterior segment ophthalmic surgery, >100 × 109/L Strong Low 
Percutaneous liver biopsy, >50 × 109/L (consider transjugular biopsy if platelet count is lower) Weak Moderate 
Give prophylactic platelet transfusions (platelet transfusions to patients who do not have clinically significant bleeding and do not require a procedure) to patients with reversible bone marrow failure (eg, general critical illness, receiving intensive chemotherapy, or undergoing hematopoietic stem cell transplantation) at or above 10 × 109/L. Strong Moderate 
Consider increasing the threshold for prophylactic platelet transfusion to between 10 × 109/L and 20 × 109/L in patients judged to have additional risk factors for bleeding (eg, sepsis). Weak Low 
American Association of Blood Banks (AABB) 20155    
Suggest prophylactic platelet transfusion for patients having elective central venous catheter placement with a platelet count <20 × 109/L. Weak Low 
Suggest prophylactic platelet transfusion for patients having elective diagnostic lumbar puncture with a platelet count <50 × 109/L. Weak Very low 
Suggest prophylactic platelet transfusion for patients having elective neuraxial anesthesia with a platelet count <50 × 109/L. Weak Very low 
Recommends against routine prophylactic platelet transfusion for patients who are nonthrombocytopenic and have cardiac surgery with cardiopulmonary bypass. Weak Very low 
Recommends transfusing hospitalized patients with a platelet count <10 × 109/L to reduce the risk of spontaneous bleeding. Strong Moderate 
Society/recommendationsStrength of recommendationQuality of evidence
European Society of Intensive Care Medicine 202061    
We suggest not using platelet transfusion to treat thrombocytopenia unless the platelet count falls below 10 × 109/L. Conditional Very low 
We make no recommendation regarding prophylactic platelet transfusion before invasive procedures for platelet counts between 10 × 109/L and 50 × 109/L. Research recommendation  
We suggest not giving prophylactic platelet transfusion before percutaneous tracheostomy or central venous catheter insertion for platelet counts between 50 100 × 109/L and 100 × 109/L. Conditional Very low 
Society of Interventional Radiology 2019124    
Consider platelet transfusion if platelet count is <20 × 109/L for procedures with low bleeding risk (eg, central venous access, including PICC placement, dialysis access, lumbar puncture, paracentesis, thoracentesis, transjugular liver biopsy, or superficial abscess drainage). Weak Limited (evidence level D) 
Consider platelet transfusion if platelet count is <50 × 109/L for procedures with high bleeding risk (eg, deep abscess drainage, solid organ biopsies, arterial intervention <7 French sheath, gastrostomy, urinary tract interventions [nephrostomy, stone removal], or transjugular intrahepatic portosystemic shunt). Weak Limited (evidence level D) 
British Society for Haematology 201787    
Consider performing the following procedures above the platelet count threshold indicated:   
Central venous lines, >20 × 109/L (using ultrasound) Strong Moderate 
Major surgery, >50 × 109/L Strong Low 
Lumbar puncture, ≥ 40 × 109/L Weak Low 
Insertion/removal of epidural catheter, ≥80 × 109/L Weak Low 
Neurosurgery or posterior segment ophthalmic surgery, >100 × 109/L Strong Low 
Percutaneous liver biopsy, >50 × 109/L (consider transjugular biopsy if platelet count is lower) Weak Moderate 
Give prophylactic platelet transfusions (platelet transfusions to patients who do not have clinically significant bleeding and do not require a procedure) to patients with reversible bone marrow failure (eg, general critical illness, receiving intensive chemotherapy, or undergoing hematopoietic stem cell transplantation) at or above 10 × 109/L. Strong Moderate 
Consider increasing the threshold for prophylactic platelet transfusion to between 10 × 109/L and 20 × 109/L in patients judged to have additional risk factors for bleeding (eg, sepsis). Weak Low 
American Association of Blood Banks (AABB) 20155    
Suggest prophylactic platelet transfusion for patients having elective central venous catheter placement with a platelet count <20 × 109/L. Weak Low 
Suggest prophylactic platelet transfusion for patients having elective diagnostic lumbar puncture with a platelet count <50 × 109/L. Weak Very low 
Suggest prophylactic platelet transfusion for patients having elective neuraxial anesthesia with a platelet count <50 × 109/L. Weak Very low 
Recommends against routine prophylactic platelet transfusion for patients who are nonthrombocytopenic and have cardiac surgery with cardiopulmonary bypass. Weak Very low 
Recommends transfusing hospitalized patients with a platelet count <10 × 109/L to reduce the risk of spontaneous bleeding. Strong Moderate 

Coming back to our case, what would happen if we omitted a platelet transfusion?

The PLADO trial demonstrated that a high-dose platelet policy did not decrease rates of bleeding or the number of transfusion episodes per participant. Put another way, there was no evidence of a dose effect. A higher dose was associated, unsurprisingly, with an increase in the number of transfusion-related adverse events.19,31 This work is informing national discussions about minimum threshold specifications for platelet content, given ongoing concerns about supply and inventory management.37 Two later RCTs compared outcomes in patients allocated to a protocol of routine prophylaxis or no prophylaxis (only therapeutic).23,38 These trials were considered to challenge the dogma of the time, given that they supported a protocol of no-platelet transfusions irrespective of platelet count. Both trials reached recruitment targets, perhaps indicating that any risks that may have been found were more likely to be on the lower side. Noninferiority for rates of WHO grade 2 to 4 bleeding was close to being declared in the larger TOPPS trial. Moreover, in patients who underwent autologous transplantation (the largest subgroup), rates of WHO grade 2 to 4 bleeding were identical. The TOPPS trial was not powered to assess differences in severe bleeding at WHO grade 3 or 4, but this information was reported, as in all RCTs (6 cases in the no-prophylaxis group vs 1 case in the prophylaxis group; see later analysis).

Case management

  • Many patients who receive platelet transfusions, irrespective of transfusion policy, will continue to experience bleeding, and the impact of platelet transfusions on bleeding on subsequent days is unclear.20 

  • Clinical factors other than platelet count are important determinants of bleeding.

  • Certain subgroups of patients (eg, autologous SCT) may not require prophylactic platelet transfusions, irrespective of platelet count.

  • A risk-adapted approach to platelet transfusions may be more prudent in our case, rather than applying a transfusion threshold platelet count of 10 × 109/L and this patient may not benefit from a prophylactic platelet transfusion.

A preterm female neonate, born at 27 weeks gestational age, needed minimal respiratory support at postnatal day 4. Clinical examination revealed minimal oozing at the umbilical cord stump. The platelet count was 35 × 109/L.

IVH is a catastrophic complication in preterm neonates and is associated with a high likelihood of death or disability.39 To prevent this occurrence, neonatologists have traditionally adopted a more liberal approach toward platelet transfusion, with surveys suggesting that many clinicians apply prophylactic transfusion at platelet counts >50 × 109/L.40-42 Until recently, neonatologists had few data from RCTs but the Platelets for Neonatal Transfusion Study 2 (PLaNet-2/MATISSE) trial has now provided information on this question.43 Preterm neonates in the liberal threshold arm (<50 × 109/L) had a significantly higher rate of death or major bleeding within 28 days after randomization, when compared with restrictive transfusion (<25 × 109/L). A secondary analysis reported that the 25 × 109/L threshold was associated with absolute-risk reduction of different baseline risks across all groups. Another small trial compared a liberal (100 × 109/L) vs standard (20 × 109/L to 100 × 109/L) threshold for platelet transfusion in preterm infants with a hemodynamically significant patent ductus arteriosus.44 A liberal transfusion policy did not hasten closure of the patent ductus arteriosus but resulted in a higher incidence of IVH. In summary, available data favor a lower transfusion threshold for platelet transfusion in nonbleeding preterm neonates.45 

Possible hypotheses to explain the mechanisms of harm in participants randomly allocated to liberal transfusion groups include the fluid shifts associated with transfusion volume and the proinflammatory effects of platelets, including inflammatory mediators that accumulate in platelets during storage and disruption of cerebral blood flow.39,45,46 An additional way to understand the risks of platelet transfusions would be to use hemovigilance systems. Unfortunately, most hemovigilance systems fail to clearly report pediatric and neonatal transfusions. Only 8% of reports submitted to SHOT (Serious Hazards of Transfusion) are pediatric cases, although these may be more common proportionately relative to adults.47 In the most recent 2020 report, there was an increase in reports related to febrile, allergic, and hypotensive reactions which appeared to be largely related to an unexplained increase in pediatric platelet transfusions.

Although there are no comparable RCT data in older children, international cohort studies in critically ill children indicate that most transfusions are given as prophylaxis to nonbleeding children, with significant variation in platelet thresholds and increments after transfusion.48 Many of these children, who will never develop bleeding complications, may be exposed unnecessarily to the risks of platelet transfusion.

Case management

  • RCT data support a lower platelet transfusion threshold (25 × 109/L) in nonbleeding preterm neonates.

  • Better hemovigilance reporting is needed for platelet transfusions in children.

  • In this case, with the absence of any clinically significant bleeding, it would be reasonable to withhold a platelet transfusion.

A 70-year-old man was admitted to the intensive care unit (ICU) with pneumococcal pneumonia. He was placed on a ventilator and treated with intravenous antibiotics. The platelet count was 17 × 109/L. The patient required insertion of a central venous catheter.

Thrombocytopenia is common in critically ill adults, and 5% to 20% will develop severe thrombocytopenia (<50 × 109/L) at some point in their ICU stay.49-51 The underlying etiology is multifactorial,52-56 but thrombocytopenia within the first 24 hours of ICU admission appears to be associated with increased 28-day mortality,57 along with a dysregulated host immune response.58 After patients with cancer, critically ill patients are the second largest group of platelet users.15 In a study of 29 ICUs in the United Kingdom, 9% of all patients received a platelet transfusion at some point during their ICU stay, many as prophylaxis. The range of platelet counts over which platelet transfusions are given to critically ill patients is wide,50,59,60 usually within 10 × 109/L to 50 × 109/L; the variation most likely reflects a lack of supporting evidence for best practice. Clinical guidelines have made inconsistent recommendations, often based on low-quality evidence (Table 1). Recent European Society of Intensive Care Medicine transfusion guidelines did not have enough evidence to make a recommendation regarding prophylactic platelet transfusion before an invasive procedure for platelet counts between 10 × 109/L and 50 × 109/L, and trials are urgently needed.61 

Other guidelines5 have recommended transfusion thresholds of 10 × 109/L to 20 × 109/L, largely based on studies in patients with a hypoproliferative bone marrow as described in case 1. However, critically ill patients may also have acquired platelet dysfunction related to accompanying conditions (renal failure, trauma, and antiplatelet drugs), and bleeding may occur even with platelet counts >50 × 109/L.62 Given the routine use of ultrasound to guide insertion of central venous catheters, the incidence of major procedure-related bleeding is very low at ∼0.05% to 1%,63 which has implications for sample sizes for future studies.64 A substudy of a large RCT found that prophylactic platelet transfusions given to critically ill patients with thrombocytopenia were not associated with a reduction in the risk of major bleeding compared with that in patients without transfusion.65 One observational study found that preprocedural platelet transfusion in patients with thrombocytopenia (<100 × 109/L) scheduled to undergo interventional radiology procedures was not associated with a reduced risk of bleeding complications (defined as a requirement for a periprocedural red cell transfusion).66 

The expected increase in platelet count from 1 platelet transfusion is between 12 × 109/L and 20 × 109/L,15,67 but this is highly variable in critically illness.48 Patients with underlying bone marrow failure may experience smaller increments in platelet count when compared with those without marrow failure.68 Although the absolute count may increase, it is unclear whether these transfused platelets function effectively, and there are few data on critically ill patients.69 

Platelet transfusions in critically ill patients are associated with risks, including acute respiratory distress syndrome, nosocomial infection, worsening organ failure, venous and arterial thrombosis, longer ICU stays, and increased mortality70-73 although there may be residual confounding by indication (sicker patients receive more platelet transfusions). The rates and mechanisms of harm need further investigation in critical illness but may result from proinflammatory mediators contained in platelet transfusions, including platelet-derived microvesicles and inflammatory cytokine release7,74-77 (Figure 2). Worsening oxygenation may also be associated with the mostly platelet-derived soluble form of CD40 ligand.73,78,79 

Figure 2.

A summary of putative mechanisms underlying the potential benefits and risks of platelet transfusions. Product and donation characteristics that may modify the efficacy and safety of platelet transfusions include ABO matching between donor and recipient, processing methods (eg, pathogen reduction technology, and storage media), and storage duration. Platelet and leukocyte activation leads to accumulation of proinflammatory cytokines (IL-1, -6, and -8 and transforming growth factor-β), soluble CD40 ligand, and formation of microvesicles. Platelet microvesicles become more numerous and injurious during storage and may trigger a recipient reaction, mediated by their molecular cargo, resulting in further inflammatory cytokine release. Platelet microparticles may downregulate macrophages and impair the reactivity of dendritic cells.

Figure 2.

A summary of putative mechanisms underlying the potential benefits and risks of platelet transfusions. Product and donation characteristics that may modify the efficacy and safety of platelet transfusions include ABO matching between donor and recipient, processing methods (eg, pathogen reduction technology, and storage media), and storage duration. Platelet and leukocyte activation leads to accumulation of proinflammatory cytokines (IL-1, -6, and -8 and transforming growth factor-β), soluble CD40 ligand, and formation of microvesicles. Platelet microvesicles become more numerous and injurious during storage and may trigger a recipient reaction, mediated by their molecular cargo, resulting in further inflammatory cytokine release. Platelet microparticles may downregulate macrophages and impair the reactivity of dendritic cells.

Close modal

The effects of platelet transfusion on inflammation and hemostasis in critically ill patients may be further modified by donation characteristics of the platelet unit, but this has been poorly studied in critical illness. In the setting of prophylaxis in hematological cancers, a secondary analysis of the PLADO trial showed that platelet source, ABO compatibility, and duration of storage did not affect bleeding rates, although platelet increments were generally higher with transfusions of apheresis platelets, ABO-identical platelets, and platelets stored 3 days vs 4 to 5 days.80 In other studies, ABO-incompatible transfusions have been associated with poor platelet recovery and increased mortality.80-82 Data support possible associations between donation characteristics and outcomes and this reiterates the importance for further study of how donation characteristics impact on outcomes including in settings such as critical illness.83-86 

Viscoelastic hemostatic assays (VHAs) such as thromboelastography (TEG) and rotational thromboelastometry (ROTEM) are increasingly being used to guide transfusion therapy in critical care, liver disease, cardiac surgery, and obstetrics.87 Such tests provide a global assessment of coagulation at the bedside to deliver targeted transfusion of blood products. Limited evidence suggests that VHA-guided therapy may reduce transfusion requirements in patients who undergo cardiac or liver surgery or have obstetric hemorrhage.87 In a small number of patients with thrombocytopenia and hematological malignancy, ROTEM-measured clot firmness and TEG-measured α angle have been shown to correlate better with bleeding than platelet count.88,89 A systematic review of the use of TEG/ROTEM in patients with sepsis found that these tests may be useful for diagnosing alterations in coagulation in sepsis, such as impaired fibrinolysis, when compared with standard laboratory tests.90 Larger studies are needed to answer whether correcting abnormal TEG/ROTEM values is associated with improvements in clinical outcomes in critically ill patients, including use of platelets.

Case management

  • Platelet transfusions may be associated with increased morbidity and mortality in critically ill patients, with limited evidence of benefit, and RCTs are needed.

  • The role of TEG/ROTEM in stable, nonbleeding, critically ill patients requires further investigation.

  • It would be reasonable to withhold a platelet transfusion in this case. The procedure should be performed by an experienced operator, using ultrasound guidance, to minimize the risk of bleeding.

In case 4, a 25-year-old man was admitted to the emergency department after a serious traffic accident. The massive hemorrhage protocol (MHP) was activated. Despite an initial normal platelet count of 140 × 109/L and an intraoperative transfusion of 1 dose of platelets, the repeated platelet count was 49 × 109/L.

Trauma-induced coagulopathy is an overall failure of the coagulation system mediated by protein C activation, hyperfibrinolysis secondary to release of tissue plasminogen activator, and rapid depletion of fibrinogen.91-93 Platelet dysfunction is common after major trauma and may be associated with increased mortality, even when the platelet count is within the normal reference range.94 Significant thrombocytopenia is considered a late event in major hemorrhage. The cornerstones of management include timely and balanced administration of blood components with control of bleeding (either surgical or radiological).91 As a pragmatic approach, guidelines often recommend that platelet transfusions be given to maintain the platelet count at >50 × 109/L in trauma bleeding.95,96 MHPs may have improved outcomes in many observational studies, including mortality, but these studies did have significant survivorship or immortal time bias (ie, participants must be alive long enough to receive the intervention).91,97,98 

The Pragmatic, Randomized Optimal Platelet and Plasma Ratios (PROPPR) trial randomly allocated 680 patients with traumatic bleeding to receive either high or low ratios of plasma and platelets to red blood cells (1:1:1 vs 1:1:2). There was no difference in all-cause mortality between the groups, but in secondary analyses, patients in the high-plasma and platelet ratio group (1:1:1) had a reduced risk of dying from exsanguination in the first 24 hours, although not a prespecified outcome.99 A post hoc analysis suggested that early platelet administration was associated with improved hemostasis and reduced mortality.100 

TEG/ROTEM-directed transfusion algorithms have been used to guide a more individualized approach for blood components and platelets in major traumatic bleeding. These have been widely reported, and the most common abnormality observed is a reduction in clot strength.101 Two small, single-center RCTs have reported reductions in mortality and clinically relevant bleeding using TEG102 and ROTEM.103 However, the recent Implementing Treatment Algorithms for the Correction of Trauma-Induced Coagulopathy (ITACTIC)104 multicenter RCT compared standard MHPs, using conventional coagulation tests vs VHA guided algorithms, but found no difference in the number of patients who were alive and free of massive transfusion at 24 hours.104 

Case management

  • Trauma-induced coagulopathy can lead to platelet loss and consumption.

  • The impact of TEG/ROTEM on clinical outcomes is unclear.

  • In our case, priority should be given to identifying and controlling the source of bleeding, either radiologically or surgically. The platelet count should (pragmatically) be maintained at 50 × 109/L with platelet transfusions.

In case 5, a 78-year-old woman was admitted to the emergency department with increasing confusion. She was taking clopidogrel. A computed tomography scan revealed an acute subdural hematoma but neurosurgery is not currently planned. Her platelet count was 271 × 109/L.

Approximately 2 million nontraumatic (spontaneous) intracerebral hemorrhages (ICHs) occur worldwide each year,105 and in high-income countries more than a quarter of patients who experience an ICH will be taking antiplatelet therapy.106 Platelet transfusions have been commonly used (and continue to be) in those patients to reduce ICH volume. However, the results of the Platelet Transfusion in Cerebral Hemorrhage (PATCH) RCT have challenged this indication.107 Patients with a supratentorial ICH, use of antiplatelet medication for at least 7 days prior, and Glasgow Coma Scale >8 were randomly allocated to receive standard care or standard care with platelet transfusion within 90 minutes of diagnostic brain imaging. Platelet transfusion was associated with an increased the risk of death or dependence in patients receiving antiplatelet therapy and presenting with an acute ICH. It is unclear whether the findings are generalizable to an increasing number of patients who are now taking agents such as clopidogrel, and various methodological limitations have been described,108 but this trial again shows the potential harm of platelet transfusions.

Case management

  • Despite its limitations, PATCH is the best available RCT evidence on this topic, and a platelet transfusion is not indicated.

  • Further studies are needed on patients prescribed antiplatelet medication and the underlying mechanisms between platelet transfusions and clinical outcomes.

This review has highlighted the uncertainty about the perceived benefits of platelet transfusions alongside risks. To further illustrate this uncertainty, we undertook an exploratory meta-analysis of major bleeding (grade ≥3) and mortality (Figure 3) across all randomized trials, recruiting 100 patients or more, irrespective of clinical setting. The results of this analysis are hypothesis generating, but the pooled estimates of effect suggest no consistent impact of platelet transfusions in reducing bleeding or mortality, although the confidence intervals are wide and may encompass potentially important clinical differences. Platelets clearly have many biological roles beyond hemostasis, and we need a better understanding of the clinical consequences of these immune effects.109,110 Given the uncertainties regarding the role of platelet transfusions, there is interest in alternatives to platelet transfusion111,112 that may include drugs and factors that stimulate endogenous production of platelets or von Willebrand factor (eg, thrombopoietin, desmopressin),113 target fibrinolysis (eg, tranexamic acid),114 or increase fibrin and fibrinogen (eg, fibrinogen concentration, recombinant factor VIIa) or use of artificial platelets or platelet membranes (eg, nanoparticles).115 Of note, a recent trial reported no evidence of an effect of tranexamic acid in reducing WHO grade 2+ bleeding in adult patients with thrombocytopenia who undergo therapy for hematological malignancy116; results of an ongoing trial are awaited.117 

Figure 3.

Exploratory forest plots of the effect of 2 strategies. Restrictive or no prophylaxis strategy (as defined by the study authors) vs a liberal strategy (as defined by the study authors) on major bleeding (A) and all-cause mortality (B) from randomized trials of platelet transfusions recruiting ∼100 patients or >100 patients. Study definitions vary and analysis included all settings although more commonly hematological cancers. No prophylaxis strategies for platelet transfusion were applied unless there was evidence of clinically significant bleeding. Restrictive transfusion strategies advocated platelet transfusions at thresholds ranging from 10 × 109/L to 25 × 109/L. Slichter et al19 compared 3 different doses of platelets; for the purposes of this analysis, we selected the low- and high-dose arms.

Figure 3.

Exploratory forest plots of the effect of 2 strategies. Restrictive or no prophylaxis strategy (as defined by the study authors) vs a liberal strategy (as defined by the study authors) on major bleeding (A) and all-cause mortality (B) from randomized trials of platelet transfusions recruiting ∼100 patients or >100 patients. Study definitions vary and analysis included all settings although more commonly hematological cancers. No prophylaxis strategies for platelet transfusion were applied unless there was evidence of clinically significant bleeding. Restrictive transfusion strategies advocated platelet transfusions at thresholds ranging from 10 × 109/L to 25 × 109/L. Slichter et al19 compared 3 different doses of platelets; for the purposes of this analysis, we selected the low- and high-dose arms.

Close modal

There are gaps in research and development for the platelet product, including the role of cryopreserved and cold-storage platelets, particularly in military settings or remote hospitals where the ability to provide standard platelets is challenging because of their short shelf life. Recent research has highlighted the uncertain clinical impact of the transfusion of platelets with qualitative defects in donations.118 Cryopreserved platelets have been approved for general civilian use and for military use in some countries.91 Small pilot RCTs of cryopreserved119 or cold-stored platelets120 in patients who undergo major cardiac surgery have shown no signs of harm and may have added hemostatic benefits. A large confirmatory trial of cold-stored platelets is ongoing (registered on https://clinicaltrials.gov as #NCT04834414).

Work on larger data sets, using advanced statistical techniques, would enable us to identify the patient at lower or greater risk of bleeding, allowing us to target interventions like platelet transfusions. Sufficient platelets would be administered to optimize vascular integrity and improve hemostasis and improve patient outcomes, while avoiding unnecessary routine transfusions of platelets with associated risks and costs.121,122 A high platelet count unit containing platelets that are more primed for activation and aggregation (hyperreactive) may be preferentially allocated for major bleeding. In contrast, a low platelet count unit may be indicated in the setting of nonbleeding prophylaxis, where these platelets support and maintain endothelial function, without adding to prothrombotic and proinflammatory risks.123 

We must design more cost-efficient adaptive trials, rather than those based on comparisons between only 2 arguably arbitrarily defined thresholds, which may not identify the actual “sweet spot” or optimal threshold where platelet transfusions have maximal benefit in a specific patient subgroup. The future offers opportunities for a precision-medicine–based approach for platelet transfusions with optimized donor-recipient matching.

The authors thank Darrell Triulzi, Rebecca Cardigan, and Peter Watkinson for constructive feedback and discussions that refined the manuscript.

Contribution: Both authors designed, reviewed, and approved the final submission.

Conflict-of-interest disclosure: S.J.S. reports receiving funds from government sources (National Institutes of Health Research and NHS Blood and Transplant) for research in the field of platelet transfusion. S.J.S. and A.S. are co-investigators on the NIHR-funded Threshold for Platelet (T4P) trial (NIHR 131822).

Correspondence: Simon J. Stanworth, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom and NHS Blood and Transplant, Level 2, John Radcliffe Hospital, Headley Way, Headington, Oxford OX3 9BQ, United Kingdom; e-mail: simon.stanworth@nhsbt.nhs.uk.

1.
Moss
P
. Essential Haematology.
Hoboken, NJ
:
Wiley-Blackwell
;
2011
.
2.
Silberstein
LE
,
Anastasi
J
. Hematology: Basic Principles and Practice E-Book.
Amsterdam, The Netherlands
:
Elsevier Health Sciences
;
2017
.
3.
Duke
WW
.
The relation of blood platelets to hemorrhagic disease: description of a method for determining the bleeding time and coagulation time and report of three cases of hemorrhagic disease relieved by transfusion
.
JAMA
.
1910
;
55
(
14
):
1185
-
1192
.
4.
Murphy
S
,
Gardner
FH
.
Maintenance of platelet viability and functional integrity during storage
.
Vox Sang
.
1971
;
20
(
5
):
427
-
428
.
5.
Kaufman
RM
,
Djulbegovic
B
,
Gernsheimer
T
, et al
.
AABB. Platelet transfusion: a clinical practice guideline from the AABB
.
Ann Intern Med
.
2015
;
162
(
3
):
205
-
213
.
6.
Estcourt
LJ
,
Birchall
J
,
Allard
S
, et al
.
British Committee for Standards in Haematology. Guidelines for the use of platelet transfusions
.
Br J Haematol
.
2017
;
176
(
3
):
365
-
394
.
7.
Ng
MSY
,
Tung
JP
,
Fraser
JF
.
Platelet storage lesions: what more do we know now?
.
Transfus Med Rev
.
2018
;
32
(
3
):
144
-
154
.
8.
Gulliksson
H
.
Platelet storage media
.
Vox Sang
.
2014
;
107
(
3
):
205
-
212
.
9.
Mack
JP
,
Miles
J
,
Stolla
M
.
Cold-stored platelets: review of studies in humans
.
Transfus Med Rev
.
2020
;
34
(
4
):
221
-
226
.
10.
Garner
SF
,
Jones
CI
,
Stephens
J
, et al;
BLOODOMICS Consortium
.
Apheresis donors and platelet function: inherent platelet responsiveness influences platelet quality
.
Transfusion
.
2008
;
48
(
4
):
673
-
680
.
11.
Rinder
HM
,
Murphy
M
,
Mitchell
JG
,
Stocks
J
,
Ault
KA
,
Hillman
RS
.
Progressive platelet activation with storage: evidence for shortened survival of activated platelets after transfusion
.
Transfusion
.
1991
;
31
(
5
):
409
-
414
.
12.
Kelly
AM
,
Garner
SF
,
Foukaneli
T
, et al
.
The effect of variation in donor platelet function on transfusion outcome: a semirandomized controlled trial
.
Blood
.
2017
;
130
(
2
):
214
-
220
.
13.
Eicher
JD
,
Lettre
G
,
Johnson
AD
.
The genetics of platelet count and volume in humans
.
Platelets
.
2018
;
29
(
2
):
125
-
130
.
14.
Dzieciatkowska
M
,
D’Alessandro
A
,
Burke
TA
, et al
.
Proteomics of apheresis platelet supernatants during routine storage: gender-related differences
.
J Proteomics
.
2015
;
112
:
190
-
209
.
15.
Gottschall
J
,
Wu
Y
,
Triulzi
D
, et al
.
NHLBI Recipient Epidemiology and Donor Evaluation (REDS-III) Study. The epidemiology of platelet transfusions: an analysis of platelet use at 12 US hospitals
.
Transfusion
.
2020
;
60
(
1
):
46
-
53
.
16.
Cardigan
R
,
New
HV
,
Estcourt
L
, et al
.
International forum on policies and practice for transfusion of ABO and RhD non-identical platelets: summary
.
Vox Sang
.
2022
;
117
(
1
):
136
-
144
.
17.
Cardillo
A
,
Heal
JM
,
Henrichs
K
, et al
.
Reducing the need for HLA-matched platelet transfusion
.
N Engl J Med
.
2021
;
384
(
25
):
2451
-
2452
.
18.
Lu
W
,
Fung
M
.
Platelets treated with pathogen reduction technology: current status and future direction
.
F1000 Res
.
2020
;
9
:
F1000
. Faculty Rev-40.
19.
Slichter
SJ
,
Kaufman
RM
,
Assmann
SF
, et al
.
Dose of prophylactic platelet transfusions and prevention of hemorrhage
.
N Engl J Med
.
2010
;
362
(
7
):
600
-
613
.
20.
Uhl
L
,
Assmann
SF
,
Hamza
TH
,
Harrison
RW
,
Gernsheimer
T
,
Slichter
SJ
.
Laboratory predictors of bleeding and the effect of platelet and RBC transfusions on bleeding outcomes in the PLADO trial
.
Blood
.
2017
;
130
(
10
):
1247
-
1258
.
21.
Josephson
CD
,
Granger
S
,
Assmann
SF
, et al
.
Bleeding risks are higher in children versus adults given prophylactic platelet transfusions for treatment-induced hypoproliferative thrombocytopenia
.
Blood
.
2012
;
120
(
4
):
748
-
760
.
22.
Slichter
SJ
.
Relationship between platelet count and bleeding risk in thrombocytopenic patients
.
Transfus Med Rev
.
2004
;
18
(
3
):
153
-
167
.
23.
Stanworth
SJ
,
Estcourt
LJ
,
Powter
G
, et al
.
TOPPS Investigators. A no-prophylaxis platelet-transfusion strategy for hematologic cancers
.
N Engl J Med
.
2013
;
368
(
19
):
1771
-
1780
.
24.
Stanworth
SJ
,
Hudson
CL
,
Estcourt
LJ
,
Johnson
RJ
,
Wood
EM
.
TOPPS study investigators. Risk of bleeding and use of platelet transfusions in patients with hematologic malignancies: recurrent event analysis
.
Haematologica
.
2015
;
100
(
6
):
740
-
747
.
25.
Narayan
S (Ed)
,
Poles
D
, et al;
on behalf of the Serious Hazards of Transfusion (SHOT) Steering Group
. The 2021 Annual SHOT Report.
2022
.
26.
Kennedy
LD
,
Case
LD
,
Hurd
DD
,
Cruz
JM
,
Pomper
GJ
.
A prospective, randomized, double-blind controlled trial of acetaminophen and diphenhydramine pretransfusion medication versus placebo for the prevention of transfusion reactions
.
Transfusion
.
2008
;
48
(
11
):
2285
-
2291
.
27.
Tobian
AA
,
King
KE
,
Ness
PM
.
Transfusion premedications: a growing practice not based on evidence
.
Transfusion
.
2007
;
47
(
6
):
1089
-
1096
.
28.
Tobian
AA
,
Fuller
AK
,
Uglik
K
, et al
.
The impact of platelet additive solution apheresis platelets on allergic transfusion reactions and corrected count increment (CME)
.
Transfusion
.
2014
;
54
(
6
):
1523
-
1529
. quiz 1522.
29.
Stramer
SL
.
Current risks of transfusion-transmitted agents: a review
.
Arch Pathol Lab Med
.
2007
;
131
(
5
):
702
-
707
.
30.
Schiffer
CA
.
Commentary on the prescient observations made by Emil J Freireich in effectiveness of platelet transfusion in leukemia and aplastic anemia.
. [published correction appears in Transfusion. 2022;62(3):539].
Transfusion
.
2022
;
62
(
2
):
267
-
272
.
31.
Estcourt
LJ
,
Stanworth
S
,
Doree
C
, et al
.
Different doses of prophylactic platelet transfusion for preventing bleeding in people with haematological disorders after myelosuppressive chemotherapy or stem cell transplantation
.
Cochrane Database Syst Rev
.
2015
;
10
:
CD010984
.
32.
Heddle
NM
,
Cook
RJ
,
Webert
KE
,
Sigouin
C
,
Rebulla
P
.
Biomedical Excellence for Safer Transfusion Working Party of the International Society for Blood Transfusion. Methodologic issues in the use of bleeding as an outcome in transfusion medicine studies
.
Transfusion
.
2003
;
43
(
6
):
742
-
752
.
33.
Estcourt
LJ
,
Heddle
N
,
Kaufman
R
, et al
.
Biomedical Excellence for Safer Transfusion Collaborative. The challenges of measuring bleeding outcomes in clinical trials of platelet transfusions
.
Transfusion
.
2013
;
53
(
7
):
1531
-
1543
.
34.
Balitsky
AK
,
Liu
Y
,
Van der Meer
PF
,
Heddle
NM
,
Arnold
DM
.
Exploring the components of bleeding outcomes in transfusion trials for patients with hematologic malignancy
.
Transfusion
.
2021
;
61
(
1
):
286
-
293
.
35.
Estcourt
LJ
,
Pinchon
D
,
Symington
E
, et al
.
Does bleeding affect patient-reported outcome measures in patients with myelodysplasia or hematologic malignancies: a systematic review
.
Transfusion
.
2014
;
54
(
4
):
1166
-
1179
.
36.
Webert
K
,
Cook
RJ
,
Sigouin
CS
,
Rebulla
P
,
Heddle
NM
.
The risk of bleeding in thrombocytopenic patients with acute myeloid leukemia
.
Haematologica
.
2006
;
91
(
11
):
1530
-
1537
.
37.
Benjamin
RJ
,
Katz
L
,
Gammon
R
,
Quinley
E
.
Consortium for Blood Availability. The argument(s) for lowering the US minimum required content of apheresis platelet components
.
Transfusion
.
2019
;
59
(
4
):
1404
.
38.
Wandt
H
,
Schaefer-Eckart
K
,
Wendelin
K
, et al
.
Study Alliance Leukemia. Therapeutic platelet transfusion versus routine prophylactic transfusion in patients with haematological malignancies: an open-label, multicentre, randomised study
.
Lancet
.
2012
;
380
(
9850
):
1309
-
1316
.
39.
Moore
CM
,
Curley
AE
.
Neonatal platelet transfusions: starting again
.
Transfus Med Rev
.
2021
;
35
(
3
):
29
-
35
.
40.
Cremer
M
,
Sola-Visner
M
,
Roll
S
, et al
.
Platelet transfusions in neonates: practices in the United States vary significantly from those in Austria, Germany, and Switzerland
.
Transfusion
.
2011
;
51
(
12
):
2634
-
2641
.
41.
Sparger
KA
,
Assmann
SF
,
Granger
S
, et al
.
Platelet transfusion practices among very-low-birth-weight infants
.
JAMA Pediatr
.
2016
;
170
(
7
):
687
-
694
.
42.
Andrew
M
,
Vegh
P
,
Caco
C
, et al
.
A randomized, controlled trial of platelet transfusions in thrombocytopenic premature infants
.
J Pediatr
.
1993
;
123
(
2
):
285
-
291
.
43.
Curley
A
,
Stanworth
SJ
,
Willoughby
K
, et al;
PlaNeT2 MATISSE Collaborators
.
Randomized trial of platelet-transfusion thresholds in neonates
.
N Engl J Med
.
2019
;
380
(
3
):
242
-
251
.
44.
Kumar
J
,
Dutta
S
,
Sundaram
V
,
Saini
SS
,
Sharma
RR
,
Varma
N
.
Platelet transfusion for PDA closure in preterm infants: a randomized controlled trial
.
Pediatrics
.
2019
;
143
(
5
):
e20182565
.
45.
Hasan
R
,
Saifee
NH
.
Benefits of lower neonatal platelet transfusion thresholds
.
Transfusion
.
2021
;
61
(
6
):
1672
-
1675
.
46.
Sola-Visner
M
,
Leeman
KT
,
Stanworth
SJ
.
Neonatal platelet transfusions: new evidence and the challenges of translating evidence-based recommendations into clinical practice
.
J Thromb Haemost
.
2022
;
20
(
3
):
556
-
564
.
47.
Stainsby
D
,
Jones
H
,
Wells
AW
,
Gibson
B
,
Cohen
H
;
SHOT Steering Group
.
Adverse outcomes of blood transfusion in children: analysis of UK reports to the serious hazards of transfusion scheme 1996-2005
.
Br J Haematol
.
2008
;
141
(
1
):
73
-
79
.
48.
Nellis
ME
,
Karam
O
,
Mauer
E
, et al
.
Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) network, Pediatric Critical Care Blood Research Network (BloodNet), and the P3T Investigators. Platelet transfusion practices in critically Ill children
.
Crit Care Med
.
2018
;
46
(
8
):
1309
-
1317
.
49.
Hui
P
,
Cook
DJ
,
Lim
W
,
Fraser
GA
,
Arnold
DM
.
The frequency and clinical significance of thrombocytopenia complicating critical illness: a systematic review
.
Chest
.
2011
;
139
(
2
):
271
-
278
.
50.
Stanworth
SJ
,
Walsh
TS
,
Prescott
RJ
,
Lee
RJ
,
Watson
DM
,
Wyncoll
DL
.
Intensive Care Study of Coagulopathy Investigators. Thrombocytopenia and platelet transfusion in UK critical care: a multicenter observational study
.
Transfusion
.
2013
;
53
(
5
):
1050
-
1058
.
51.
Jonsson
AB
,
Rygård
SL
,
Hildebrandt
T
,
Perner
A
,
Møller
MH
,
Russell
L
.
Thrombocytopenia in intensive care unit patients: a scoping review
.
Acta Anaesthesiol Scand
.
2021
;
65
(
1
):
2
-
14
.
52.
Greinacher
A
,
Selleng
S
.
How I evaluate and treat thrombocytopenia in the intensive care unit patient
.
Blood
.
2016
;
128
(
26
):
3032
-
3042
.
53.
Levi
M
,
Opal
SM
.
Coagulation abnormalities in critically ill patients
.
Crit Care
.
2006
;
10
(
4
):
222
.
54.
Larkin
CM
,
Santos-Martinez
MJ
,
Ryan
T
,
Radomski
MW
.
Sepsis-associated thrombocytopenia
.
Thromb Res
.
2016
;
141
:
11
-
16
.
55.
Bedet
A
,
Razazi
K
,
Boissier
F
, et al
.
Mechanisms of thrombocytopenia during septic shock: a multiplex cluster analysis of endogenous sepsis mediators
.
Shock
.
2018
;
49
(
6
):
641
-
648
.
56.
Thachil
J
,
Warkentin
TE
.
How do we approach thrombocytopenia in critically ill patients?
.
Br J Haematol
.
2017
;
177
(
1
):
27
-
38
.
57.
Thiery-Antier
N
,
Binquet
C
,
Vinault
S
,
Meziani
F
,
Boisramé-Helms
J
,
Quenot
JP
.
EPIdemiology of Septic Shock Group. Is thrombocytopenia an early prognostic marker in septic shock?
.
Crit Care Med
.
2016
;
44
(
4
):
764
-
772
.
58.
Claushuis
TA
,
van Vught
LA
,
Scicluna
BP
, et al
.
Molecular Diagnosis and Risk Stratification of Sepsis Consortium. Thrombocytopenia is associated with a dysregulated host response in critically ill sepsis patients
.
Blood
.
2016
;
127
(
24
):
3062
-
3072
.
59.
de Bruin
S
,
Scheeren
TWL
,
Bakker
J
,
van Bruggen
R
,
Vlaar
APJ
.
Cardiovascular Dynamics Section and Transfusion Guideline Task Force of the ESICM. Transfusion practice in the non-bleeding critically ill: an international online survey-the TRACE survey
.
Crit Care
.
2019
;
23
(
1
):
309
.
60.
Shah
A
,
Gould
DW
,
Doidge
J
, et al
.
Threshold for Platelets (T4P) Investigators. A UK national survey of prophylactic platelet transfusion thresholds in non-bleeding, critically ill adults
.
Transfus Med
.
2020
;
30
(
6
):
515
-
517
.
61.
Vlaar
AP
,
Oczkowski
S
,
de Bruin
S
, et al
.
Transfusion strategies in non-bleeding critically ill adults: a clinical practice guideline from the European Society of Intensive Care Medicine
.
Intensive Care Med
.
2020
;
46
(
4
):
673
-
696
.
62.
Yaguchi
A
,
Lobo
FL
,
Vincent
JL
,
Pradier
O
.
Platelet function in sepsis
.
J Thromb Haemost
.
2004
;
2
(
12
):
2096
-
2102
.
63.
Cabrini
L
,
Pappacena
S
,
Mattioli
L
, et al
.
Administration of blood products to prevent bleeding complications associated with central venous catheter insertion in patients at risk: a systematic review
.
Br J Anaesth
.
2017
;
118
(
4
):
630
-
634
.
64.
Estcourt
LJ
,
Desborough
M
,
Hopewell
S
,
Doree
C
,
Stanworth
SJ
.
Comparison of different platelet transfusion thresholds prior to insertion of central lines in patients with thrombocytopenia
.
Cochrane Database Syst Rev
.
2015
;
12
:
CD011771
.
65.
Arnold
DM
,
Lauzier
F
,
Albert
M
, et al
.
PROTECT Investigators, the Canadian Critical Care Trials Group and the Australian and New Zealand Intensive Care Society Clinical Trials Group. The association between platelet transfusions and bleeding in critically ill patients with thrombocytopenia
.
Res Pract Thromb Haemost
.
2017
;
1
(
1
):
103
-
111
.
66.
Warner
MA
,
Woodrum
D
,
Hanson
A
,
Schroeder
DR
,
Wilson
G
,
Kor
DJ
.
Preprocedural platelet transfusion for patients with thrombocytopenia undergoing interventional radiology procedures is not associated with reduced bleeding complications
.
Transfusion
.
2017
;
57
(
4
):
890
-
898
.
67.
Lieberman
L
,
Bercovitz
RS
,
Sholapur
NS
,
Heddle
NM
,
Stanworth
SJ
,
Arnold
DM
.
Platelet transfusions for critically ill patients with thrombocytopenia
.
Blood
.
2014
;
123
(
8
):
1146
-
1151
. quiz 1280.
68.
O’Bryan
LJ
,
Bedford
J
,
Redfern
OC
,
Hatch
RA
,
Young
JD
,
Watkinson
PJ
.
Prophylactic use of platelets in critically ill patients with thrombocytopaenia: A retrospective two-centre observational study
.
J Crit Care
.
2020
;
57
:
157
-
167
.
69.
Kander
T
,
Tanaka
KA
,
Norström
E
,
Persson
J
,
Schött
U
.
The effect and duration of prophylactic platelet transfusions before insertion of a central venous catheter in patients with bone marrow failure evaluated with point-of-care methods and flow cytometry
.
Anesth Analg
.
2014
;
119
(
4
):
882
-
890
.
70.
Warner
MA
,
Chandran
A
,
Frank
RD
,
Kor
DJ
.
Prophylactic platelet transfusions for critically ill patients with thrombocytopenia: a single-institution propensity-matched cohort study
.
Anesth Analg
.
2019
;
128
(
2
):
288
-
295
.
71.
Kasotakis
G
,
Starr
N
,
Nelson
E
, et al
.
Inflammation and Host Response to Injury Investigators. Platelet transfusion increases risk for acute respiratory distress syndrome in non-massively transfused blunt trauma patients
.
Eur J Trauma Emerg Surg
.
2019
;
45
(
4
):
671
-
679
.
72.
Khan
H
,
Belsher
J
,
Yilmaz
M
, et al
.
Fresh-frozen plasma and platelet transfusions are associated with development of acute lung injury in critically ill medical patients
.
Chest
.
2007
;
131
(
5
):
1308
-
1314
.
73.
Khan
SY
,
Kelher
MR
,
Heal
JM
, et al
.
Soluble CD40 ligand accumulates in stored blood components, primes neutrophils through CD40, and is a potential cofactor in the development of transfusion-related acute lung injury
.
Blood
.
2006
;
108
(
7
):
2455
-
2462
.
74.
McVey
MJ
,
Weidenfeld
S
,
Maishan
M
, et al
.
Platelet extracellular vesicles mediate transfusion-related acute lung injury by imbalancing the sphingolipid rheostat
.
Blood
.
2021
;
137
(
5
):
690
-
701
.
75.
Blumberg
N
,
Gettings
KF
,
Turner
C
,
Heal
JM
,
Phipps
RP
.
An association of soluble CD40 ligand (CD154) with adverse reactions to platelet transfusions
.
Transfusion
.
2006
;
46
(
10
):
1813
-
1821
.
76.
Silliman
CC
,
Boshkov
LK
,
Mehdizadehkashi
Z
, et al
.
Transfusion-related acute lung injury: epidemiology and a prospective analysis of etiologic factors
.
Blood
.
2003
;
101
(
2
):
454
-
462
.
77.
Kapur
R
,
Kim
M
,
Aslam
R
, et al
.
T regulatory cells and dendritic cells protect against transfusion-related acute lung injury via IL-10
.
Blood
.
2017
;
129
(
18
):
2557
-
2569
.
78.
Henn
V
,
Slupsky
JR
,
Gräfe
M
, et al
.
CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells
.
Nature
.
1998
;
391
(
6667
):
591
-
594
.
79.
Vanichakarn
P
,
Blair
P
,
Wu
C
,
Freedman
JE
,
Chakrabarti
S
.
Neutrophil CD40 enhances platelet-mediated inflammation
.
Thromb Res
.
2008
;
122
(
3
):
346
-
358
.
80.
Triulzi
DJ
,
Assmann
SF
,
Strauss
RG
, et al
.
The impact of platelet transfusion characteristics on posttransfusion platelet increments and clinical bleeding in patients with hypoproliferative thrombocytopenia
.
Blood
.
2012
;
119
(
23
):
5553
-
5562
.
81.
Nellis
ME
,
Goel
R
,
Karam
O
, et al
.
P3T Investigators. Effects of ABO matching of platelet transfusions in critically ill children
.
Pediatr Crit Care Med
.
2019
;
20
(
2
):
e61
-
e69
.
82.
Magid-Bernstein
J
,
Beaman
CB
,
Carvalho-Poyraz
F
, et al
.
Impacts of ABO-incompatible platelet transfusions on platelet recovery and outcomes after intracerebral hemorrhage
.
Blood
.
2021
;
137
(
19
):
2699
-
2703
.
83.
Aubron
C
,
Flint
AWJ
,
Ozier
Y
,
McQuilten
Z
.
Platelet storage duration and its clinical and transfusion outcomes: a systematic review
.
Crit Care
.
2018
;
22
(
1
):
185
.
84.
Norol
F
,
Kuentz
M
,
Cordonnier
C
, et al
.
Influence of clinical status on the efficiency of stored platelet transfusion
.
Br J Haematol
.
1994
;
86
(
1
):
125
-
129
.
85.
Roeloffzen
WW
,
Kluin-Nelemans
HC
,
Veeger
NJ
,
Bosman
L
,
de Wolf
JT
.
Transfused stored platelets have the same haemostatic function as circulating native platelets
.
Vox Sang
.
2010
;
99
(
2
):
123
-
130
.
86.
Nellis
ME
,
Spinella
PC
,
Tucci
M
, et al
.
Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) network, Pediatric Critical Care Blood Research Network (BloodNet), and the P3T Investigators†. Effect of platelet storage duration on clinical outcomes and incremental platelet change in critically ill children
.
Transfusion
.
2020
;
60
(
12
):
2849
-
2858
.
87.
Curry
NS
,
Davenport
R
,
Pavord
S
, et al
.
The use of viscoelastic haemostatic assays in the management of major bleeding: a British Society for Haematology Guideline
.
Br J Haematol
.
2018
;
182
(
6
):
789
-
806
.
88.
Opheim
EN
,
Apelseth
TO
,
Stanworth
SJ
,
Eide
GE
,
Hervig
T
.
Thromboelastography may predict risk of grade 2 bleeding in thrombocytopenic patients
.
Vox Sang
.
2017
;
112
(
6
):
578
-
585
.
89.
Estcourt
LJ
,
Stanworth
SJ
,
Harrison
P
, et al
.
Prospective observational cohort study of the association between thromboelastometry, coagulation and platelet parameters and bleeding in patients with haematological malignancies- the ATHENA study
.
Br J Haematol
.
2014
;
166
(
4
):
581
-
591
.
90.
Müller
MC
,
Meijers
JC
,
Vroom
MB
,
Juffermans
NP
.
Utility of thromboelastography and/or thromboelastometry in adults with sepsis: a systematic review
.
Crit Care
.
2014
;
18
(
1
):
R30
.
91.
Curry
NS
,
Davenport
R
.
Transfusion strategies for major haemorrhage in trauma
.
Br J Haematol
.
2019
;
184
(
4
):
508
-
523
.
92.
Cohen
MJ
.
Acute traumatic coagulopathy: clinical characterization and mechanistic investigation
.
Thromb Res
.
2014
;
133
(
suppl 1
):
S25
-
S27
.
93.
Davenport
RA
,
Brohi
K
.
Cause of trauma-induced coagulopathy
.
Curr Opin Anaesthesiol
.
2016
;
29
(
2
):
212
-
219
.
94.
Kutcher
ME
,
Redick
BJ
,
McCreery
RC
, et al
.
Characterization of platelet dysfunction after trauma
.
J Trauma Acute Care Surg
.
2012
;
73
(
1
):
13
-
19
.
95.
Stanworth
SJ
,
Dowling
K
,
Curry
N
, et al
.
Transfusion Task Force of the British Society for Haematology. Haematological management of major haemorrhage: a British Society for Haematology Guideline
.
Br J Haematol
.
2022
;
198
(
4
):
654
-
667
.
96.
Klein
AA
,
Arnold
P
,
Bingham
RM
, et al
.
AAGBI guidelines: the use of blood components and their alternatives 2016
.
Anaesthesia
.
2016
;
71
(
7
):
829
-
842
.
97.
Hallet
J
,
Lauzier
F
,
Mailloux
O
, et al
.
The use of higher platelet: RBC transfusion ratio in the acute phase of trauma resuscitation: a systematic review
.
Crit Care Med
.
2013
;
41
(
12
):
2800
-
2811
.
98.
Yadav
K
,
Lewis
RJ
.
Immortal Time Bias in Observational Studies
.
JAMA
.
2021
;
325
(
7
):
686
-
687
.
99.
Holcomb
JB
,
Tilley
BC
,
Baraniuk
S
, et al;
PROPPR Study Group
.
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
(
5
):
471
-
482
.
100.
Cardenas
JC
,
Zhang
X
,
Fox
EE
, et al;
PROPPR Study Group
.
Platelet transfusions improve hemostasis and survival in a substudy of the prospective, randomized PROPPR trial
.
Blood Adv
.
2018
;
2
(
14
):
1696
-
1704
.
101.
Khan
S
,
Davenport
R
,
Raza
I
, et al
.
Damage control resuscitation using blood component therapy in standard doses has a limited effect on coagulopathy during trauma hemorrhage
.
Intensive Care Med
.
2015
;
41
(
2
):
239
-
247
.
102.
Gonzalez
E
,
Moore
EE
,
Moore
HB
, et al
.
Goal-directed hemostatic resuscitation of trauma-induced coagulopathy: a pragmatic randomized clinical trial comparing a viscoelastic assay to conventional coagulation assays
.
Ann Surg
.
2016
;
263
(
6
):
1051
-
1059
.
103.
Innerhofer
P
,
Fries
D
,
Mittermayr
M
, et al
.
Reversal of trauma-induced coagulopathy using first-line coagulation factor concentrates or fresh frozen plasma (RETIC): a single-centre, parallel-group, open-label, randomised trial
.
Lancet Haematol
.
2017
;
4
(
6
):
e258
-
e271
.
104.
Baksaas-Aasen
K
,
Gall
LS
,
Stensballe
J
, et al
.
Viscoelastic haemostatic assay augmented protocols for major trauma haemorrhage (ITACTIC): a randomized, controlled trial
.
Intensive Care Med
.
2021
;
47
(
1
):
49
-
59
.
105.
Feigin
VL
,
Lawes
CM
,
Bennett
DA
,
Barker-Collo
SL
,
Parag
V
.
Worldwide stroke incidence and early case fatality reported in 56 population-based studies: a systematic review
.
Lancet Neurol
.
2009
;
8
(
4
):
355
-
369
.
106.
Lovelock
CE
,
Molyneux
AJ
,
Rothwell
PM
;
Oxford Vascular Study
.
Change in incidence and aetiology of intracerebral haemorrhage in Oxfordshire, UK, between 1981 and 2006: a population-based study
.
Lancet Neurol
.
2007
;
6
(
6
):
487
-
493
.
107.
Baharoglu
MI
,
Cordonnier
C
,
Al-Shahi Salman
R
, et al
.
PATCH Investigators. Platelet transfusion versus standard care after acute stroke due to spontaneous cerebral haemorrhage associated with antiplatelet therapy (PATCH): a randomised, open-label, phase 3 trial
.
Lancet
.
2016
;
387
(
10038
):
2605
-
2613
.
108.
Baharoglu
MI
,
Al-Shahi Salman
R
,
Cordonnier
C
, et al
.
PATCH trial: explanatory analyses
.
Blood
.
2020
;
135
(
16
):
1406
-
1409
.
109.
Sut
C
,
Tariket
S
,
Aubron
C
, et al
.
The non-hemostatic aspects of transfused platelets
.
Front Med (Lausanne)
.
2018
;
5
:
42
.
110.
Maouia
A
,
Rebetz
J
,
Kapur
R
,
Semple
JW
.
The immune nature of platelets revisited
.
Transfus Med Rev
.
2020
;
34
(
4
):
209
-
220
.
111.
Desborough
MJ
,
Smethurst
PA
,
Estcourt
LJ
,
Stanworth
SJ
.
Alternatives to allogeneic platelet transfusion
.
Br J Haematol
.
2016
;
175
(
3
):
381
-
392
.
112.
Desborough
M
,
Hadjinicolaou
AV
,
Chaimani
A
, et al
.
Alternative agents to prophylactic platelet transfusion for preventing bleeding in people with thrombocytopenia due to chronic bone marrow failure: a meta-analysis and systematic review
.
Cochrane Database Syst Rev
.
2016
;
10
:
CD012055
.
113.
Desborough
MJR
,
Al-Shahi Salman
R
,
Stanworth
SJ
, et al
.
Desmopressin for reversal of Antiplatelet drugs in Stroke due to Haemorrhage (DASH): protocol for a phase II double-blind randomised controlled feasibility trial
.
BMJ Open
.
2020
;
10
(
11
):
e037555
.
114.
Bouillon-Minois
JB
,
Croizier
C
,
Baker
JS
, et al
.
Tranexamic acid in non-traumatic intracranial bleeding: a systematic review and meta-analysis
.
Sci Rep
.
2021
;
11
(
1
):
15275
.
115.
Sekhon
UDS
,
Swingle
K
,
Girish
A
, et al
.
Platelet-mimicking procoagulant nanoparticles augment hemostasis in animal models of bleeding
.
Sci Transl Med
.
2022
;
14
(
629
):
eabb8975
.
116.
Gernsheimer
TB
,
Brown
SP
,
Triulzi
DJ
, et al
.
Prophylactic tranexamic acid in patients with hematologic malignancy: a placebo controlled, randomized clinical trial
.
Blood
.
2022
. [published online ahead of print 6 June 2022].
117.
Estcourt
LJ
,
McQuilten
Z
,
Powter
G
, et al
.
TREATT Trial Collaboration (provisional). The TREATT Trial (TRial to EvaluAte Tranexamic acid therapy in Thrombocytopenia): safety and efficacy of tranexamic acid in patients with haematological malignancies with severe thrombocytopenia: study protocol for a double-blind randomised controlled trial
.
Trials
.
2019
;
20
(
1
):
592
.
118.
Pedini
P
,
Baudey
J.-B.
,
Pouymayou
K
, et al
.
Screening platelet function in blood donors
.
Transfusion
.
2022
;
62
(
8
):
1643
-
1651
.
119.
Reade
MC
,
Marks
DC
,
Bellomo
R
, et al
.
Cryopreserved vs Liquid Platelet (CLIP) Investigators, the Australian and New Zealand College of Anaesthetists Clinical Trials Network, and the Australian and New Zealand Intensive Care Society Clinical Trials Group. A randomized, controlled pilot clinical trial of cryopreserved platelets for perioperative surgical bleeding: the CLIP-I trial (Editorial, p. 2759)
.
Transfusion
.
2019
;
59
(
9
):
2794
-
2804
.
120.
Strandenes
G
,
Sivertsen
J
,
Bjerkvig
CK
, et al
.
A pilot trial of platelets stored cold versus at room temperature for complex cardiothoracic surgery
.
Anesthesiology
.
2020
;
133
(
6
):
1173
-
1183
.
121.
Kaiser
R
,
Escaig
R
,
Kranich
J
, et al
.
Procoagulant platelet sentinels prevent inflammatory bleeding through GPIIBIIIA and GPVI
.
Blood
.
2022
;
140
(
2
):
121
-
139
.
122.
Slichter
SJ
,
Gernsheimer
T
,
LeBlanc
R
, et al
.
Fecal blood loss: a quantitative method of evaluating hemostasis in patients with thrombocytopenia
.
Transfusion
.
2021
;
61
(
2
):
393
-
403
.
123.
Nicolai
L
,
Schiefelbein
K
,
Lipsky
S
, et al
.
Vascular surveillance by haptotactic blood platelets in inflammation and infection [published correction appears in Nat Commun. 2022;13(1):4645]
.
Nat Commun
.
2020 Nov 13
;
11
(
1
):
5778
.
124.
Patel
I
,
Rahim
S
,
Davidson
J
, et al
.
Society of Interventional Radiology consensus guidelines for the periprocedural management of thrombotic and bleeding risk in patients undergoing percutaneous image-guided interventions - part II: recommendations
.
J Vasc Interv Radiol
.
2019
;
30
(
8
):
1168
-
1184
.
Sign in via your Institution