Abstract
Background
Platelet transfusion is essential for preventing and treating hemorrhage in oncology patients and can markedly improve quality of life and survival. However, access to platelet concentrates is often limited by global shortages and logistical constraints, especially in low-resource settings.
Summary
Maintaining product quality requires stringent donor screening, pathogen-reduction technologies, and optimized storage conditions to preserve hemostatic function and reduce adverse reactions. Common transfusion-related complications (e.g., alloimmunization, non-hemolytic reactions, and circulatory overload) underscore the importance of real-time monitoring and individualized transfusion protocols. Emerging thrombopoietin receptor agonists, such as romiplostim and eltrombopag, exhibited benefit in reducing transfusion dependency and merit further study in cancer-associated thrombocytopenia. This review aims to summarize the research advances and clinical guidelines on platelet transfusion, including platelet production methods, transfusion dosage, pathogen inactivation, leucocyte depletion, types of cancer-related thrombocytopenias, and platelet transfusion strategies and to discuss future research directions and perspectives.
Key Messages
While platelet transfusions remain indispensable for mitigating bleeding risk in immunotherapy and CAR-T recipients, the heterogeneity of patient responses underscores the need for prospective trials to evaluate the impact of transfusion practices on both hemostatic and immunologic outcomes.
Keywords: Platelet transfusion, Cancer, Thrombocytopenia
Introduction
Radiotherapy and chemotherapy remain fundamental to cancer therapy but frequently induce thrombocytopenia, predisposing patients to life-threatening hemorrhages such as intracranial bleeding. Previous studies reported that about 20% of solid tumor patients developed thrombocytopenia within 1 year of diagnosis, whereas rates exceed 60% in those with hematologic malignancies [1–3]. The incidence and severity of thrombocytopenia are influenced by chemotherapy regimen, dose intensity, and cancer type, manifesting clinically as fatigue, mucocutaneous bleeding, and petechiae, all of which markedly impair physical, emotional, and psychological well-being [4, 5]. Symptoms often include neurocognitive discomfort and profound weakness, correlating with declines in overall quality of life.
Prophylactic and therapeutic platelet transfusions are considered the cornerstone intervention to prevent and treat bleeding, with randomized trials demonstrating significant reductions in hemorrhagic mortality when protocol-driven transfusions are employed [6]. However, global platelet inventories are constrained, and shortages – especially in developing regions – affect patient care [7, 8]. Beyond hemostasis, platelets regulate innate and adaptive immunity and interact with tumor cells to promote immune evasion, angiogenesis, and metastatic seeding [9, 10]. Moreover, transfusion of activated platelets may inadvertently exacerbate tumor progression by supplying growth factors and shielding circulating tumor cells [11]. Despite the widespread adoption of transfusion protocols, there remain many contentious and unknown issues.
To comprehensively synthesize current advancements in platelet transfusion for cancer patients and discuss future research directions, a systematic literature search was conducted. The search was performed across major databases, including PubMed, Embase, and the Cochrane Library, from inception to May 1, 2025. The following search terms were used in various combinations: “platelet transfusion”, “cancer patients”, “thrombocytopenia”, “chemotherapy”, “radiotherapy”, “transfusion risks”, “platelet products”, and “transfusion guidelines”. Only original research articles, systematic reviews, and meta-analyses written in English were included. Conference abstracts, case reports, and editorials were excluded. Titles and abstracts were initially screened by two independent authors to identify potentially relevant studies, with full-text articles retrieved for further evaluation. Disagreements were resolved through discussion or by consulting a third reviewer. This rigorous selection process aimed to ensure that the review incorporated the most relevant and high-quality evidence, enabling a thorough and accurate discussion of the current state and future prospects of platelet transfusion in cancer patients.
Preparation and Quality Aspects of Platelets for Transfusion
Platelet concentrates (PCs) for transfusion can be produced using three methods: platelet-rich plasma, buffy coat, and apheresis-PC. Platelet-rich plasma platelet concentrate involves initial low-speed centrifugation to separate platelet-rich plasma, followed by a second centrifugation to concentrate platelets. Buffy coat platelet concentrate is derived from the buffy coat layer obtained from at least four donors after high-speed centrifugation, subsequently pooled and processed. This pooling step might influence various aspects related to transfusion risks, such as the potential for transmitting infectious agents or the likelihood of immune reactions due to exposure to multiple donors’ antigens. Apheresis-PC or single-donor platelets are collected directly from donors using apheresis machines, offering higher platelet yields and reduced leukocyte contamination compared to whole-blood-derived methods [12].
Storage and transportation conditions greatly affect platelet viability and function. Normally, PCs are stored at 20–24°C with continuous gentle agitation; their shelf life is limited to 5–7 days because of bacterial growth [13, 14]. Platelet storage temperature has been explored at 1–6°C, and the shelf life was extended with reduced bacterial growth; however, it may induce platelet function disturbance by lowering glycoprotein VI (GPVI) levels [15, 16]. Since GPVI is essential for platelet adhesion and aggregation, cold storage may not be an ideal option. Furthermore, platelet and their transportation may undergo mechanical stress, which leads to platelet fragmentation. This results in impaired platelet activation, increased GPVI receptor shedding, and mitochondria dysfunction and therefore compromises platelet function and transfusion safety [17].
To enhance transfusion safety, pathogen reduction treatments (PRTs) have been developed [18]. The INTERCEPT system inactivates a broad range of pathogens by using amotosalen and UVA light. Similarly, the MIRASOL system inactivates pathogens by riboflavin and UVB light without additional removal steps, preserving platelet functionality [19, 20]. PRTs reduce the risk for transfusion-transmitted infectious diseases effectively. Notably, in the context of global health challenges, their importance has become even more pronounced. For instance, the emergence of pathogens such as the West Nile virus in Europe and the growing relevance of hepatitis E in transfusion medicine, especially for immunocompromised cancer patients, underscore the critical role of PRTs [21]. The recent COVID-19 pandemic has further highlighted the potential impact of infectious threats on platelet transfusion practices. Concerns regarding the potential transmission of the virus through transfusion, as well as the need to maintain a safe and reliable blood supply amidst disruptions in the healthcare system, brought the importance of PRTs into sharp focus. Although there is currently limited evidence of COVID-19 transmission via blood products, the pandemic experience has emphasized the necessity of robust pathogen reduction strategies to prepare for future infectious disease outbreaks. Pathogen inactivation strategies such as THERAFLEX MB-Plasma and THERAFLEX UV-Platelets systems could reduce the risk of transmission of SARS-CoV-2 with an average ≥5.03 log10 [22]. However, it is important to note that PRTs are not without drawbacks. These treatments may slightly reduce platelet counts and post-transfusion recovery rates. Clinicians must carefully evaluate the benefits of enhanced infection prevention against the potential impact on platelet functionality to optimize transfusion outcomes for patients, especially for those immunocompromised patients. Future research should aim to further refine PRT technologies to minimize these drawbacks while maximizing their effectiveness in protecting patients from transfusion-transmitted infections.
Leukoreduction, removal of white blood cells from PCs, is an important quality assurance step. It reduces the risk of alloimmunization and transmission of leukocyte-associated pathogens [23, 24]. Leukoreduction can be achieved by filtration during or after preparation of components [25]. Additionally, virus inactivation is carried out by chemical drugs or ultraviolet light, and pathogen reduction is perfumed by filtration [26, 27]. Collectively, ensuring the platelet safety and efficacy necessitates the meticulous attention to collection methods, storage conditions, leukoreduction, and pathogen reduction protocols. These measures are critical to maintain platelet functionality and minimize transfusion-related risks.
Cancer-Related Thrombocytopenia
Cancer patients frequently develop thrombocytopenia, either from the malignancy itself or cancer treatment. In clinical practice, thrombocytopenia in cancer is categorized by etiology: direct effects of the tumor on hematopoiesis, cytotoxic chemotherapy, and immune-related mechanisms (notably checkpoint inhibitors). Each category has a distinct pathophysiology and implication. Thrombocytopenia poses various challenges to the cancer care. The normal level of platelet counts in adults ranges from 150 to 450 ×109/L. Thrombocytopenia is described as a platelet count that falls below the normal level. The 2020 revised version 5.0 of the US National Institute of Health Common Terminology Criteria for Adverse Events (CTCAE) classified thrombocytopenia associated with cancer treatment as follows: grade 1, 75–150 ×109/L; grade 2, 50–75 × 109/L; grade 3, 25–50 × 109/L; grade 4, <25 × 109/L [5].
Direct Cancer-Induced Thrombocytopenia
Tumor infiltration of bone marrow is a major direct cause of thrombocytopenia in cancer [28]. When malignant cells (e.g., solid tumor metastases) invade the marrow, they disrupt normal hematopoiesis. In advanced malignancies, disseminated tumor cells infiltrate the bone marrow via heterogeneous dissemination or direct invasion and destroy its architecture [29]. Clinically, this often presents with pancytopenia. Previous studies reported that bone marrow metastases typically cause moderate anemia and thrombocytopenia [29]. The cancers most likely to spread to bone marrow include gastric, breast, lung, and prostate cancers [30]. Metastatic disease can also induce extramedullary hematopoiesis, particularly in the spleen. Premature splenic release of hematopoietic cells can result in leukoerythroblastosis in peripheral blood [31]. A normal-sized spleen contains one-third of the total platelet. However, with splenomegaly, up to 50–90% of the platelet mass may be trapped in the spleen, causing peripheral thrombocytopenia [32]. The thrombocytopenia has significant implications for platelet transfusion. Due to the impaired platelet production caused by tumor infiltration, the efficacy of platelet transfusion may be compromised. Even with transfusions, the body’s ability to maintain an adequate platelet count is limited as the underlying bone marrow defect persists. Moreover, the presence of splenomegaly can lead to platelet sequestration in the enlarged spleen, which means that transfused platelets may be rapidly removed from the circulation, reducing their lifespan and effectiveness in preventing bleeding. As a result, higher transfusion thresholds or more frequent platelet transfusions may be required to achieve hemostasis, increasing the risk of transfusion-related complications. Clinicians must carefully balance the need for platelet transfusions against these potential risks, considering alternative strategies such as growth factor support to stimulate endogenous platelet production when possible. In addition, for patients with thrombocytopenia caused by splenomegaly, when considering platelet transfusion, it is also necessary to comprehensively evaluate the degree of splenomegaly and its impact on platelet sequestration to develop a more reasonable transfusion strategy.
Splenomegaly can occur as a reversible consequence of hepatic metastasis resection or due to hematologic malignancies, each with distinct bleeding risks. After hepatic metastasis resection, splenomegaly is frequently observed, primarily driven by growth factors and cytokines that induce liver regeneration and incidentally also promote spleen expansion. In one study, 78% of patients undergoing partial hepatectomy for colorectal metastases experienced an increase in spleen volume [33]. Six months after surgery, spleen volume had increased by 40–45% on average, before returning to baseline size at 9 months. An inverse correlation between spleen size and platelet count was observed following hepatectomy [34]. In this patient population, the bleeding risk may be relatively lower during the period of transient splenomegaly as the condition is self-limiting. However, close monitoring of platelet counts and bleeding signs is essential. Platelet transfusion was performed for patients with significant thrombocytopenia or those with active bleeding. Splenomegaly is also an important cause of thrombocytopenia in hematologic malignancies [35]. These patients typically face a higher bleeding risk due to the combination of reduced platelet production in the bone marrow and increased platelet sequestration in the enlarged spleen. Therefore, more aggressive transfusion thresholds may be considered, especially for these presenting additional risk factors for bleeding. Although splenectomy and partial splenic embolization are rarely performed in patients with solid or hematologic malignancies, they have been associated with sustained platelet count improvement and reduced transfusion requirements [36–38]. However, their use must be carefully evaluated, weighing the potential benefits against the surgical or procedural risks, particularly in patients with compromised health status due to their underlying malignancies.
Beyond physical marrow involvement, cancers can induce consumptive coagulopathies. Tumor-associated disseminated intravascular coagulation (DIC) consumes platelets and clotting factors. Systemic activation of coagulation in DIC leads to microvascular thrombosis and potentially life-threatening hemorrhage attributed to the consumption of platelets and coagulation factors [39]. In this context, an underlying malignancy (such as pancreatic or mucinous adenocarcinoma) often drives the DIC process. Similarly, cancer-associated thrombotic microangiopathy (TMA) can cause profound thrombocytopenia. TMA is characterized by widespread endothelial injury and microthrombus formation. In cancer patients, it is mediated by endothelial dysfunction and results in microangiopathic hemolytic anemia and platelet consumption. Cancer-related TMA tends to occur in advanced disease and carries a poor prognosis [40].
Chemotherapy-Induced Thrombocytopenia
Chemotherapy-induced thrombocytopenia (CIT) prevalence varies significantly by regimen and cancer type. In a US outpatient oncology study, gemcitabine-based regimens had the highest thrombocytopenia incidence at 64.2%, followed by platinum-based (55.2%), anthracycline-based (37.8%), and taxane-based regimens (21.9%) [41]. Another US claims study reported the highest association of thrombocytopenia with gemcitabine (14%), carboplatin (13%), and oxaliplatin (11%) [2]. Besides, the study by Hitron et al. [42] found the highest incidence of thrombocytopenia with the gemcitabine/cisplatin (57% for bladder cancer), gemcitabine/carboplatin (29% for lung cancer), and cisplatin/etoposide (18% for lung cancer) regimens. Regarding tumor types, a large US observational cohort found that 13% of solid tumor patients and 28% of hematologic malignancies had platelet counts <100 × 109/L within 3 months of starting chemotherapy [1]. Among common solid tumors, thrombocytopenia incidence was highest in lung cancer (14.3%), followed by colorectal (13.5%), pancreatic (12.9%), and breast (9.6%) cancer. Severe (grade ≥3) CIT occurred in about 6.1% of solid tumor patients versus 28.7% of those with hematologic cancers (Table 1) [1]. A recent guideline notes that CIT affects approximately one-third of patients with solid tumors and half of those with hematologic malignancies, underscoring its clinical significance [43].
Table 1.
Incidence of CIT (CTCAE: any grade) in typical chemotherapy regimens and cancer types [1]
| | Overall, % | Severe CIT (grade ≥ 3), % |
|---|---|---|
| Chemotherapy regimen | ||
| Gemcitabine-based | 15.8 | 8.0 |
| Platinum-based | 13.6 | 6.3 |
| Anthracycline | 16.4 | 10.7 |
| Taxane | 6.6 | 2.8 |
| Cancer type after chemotherapy | ||
| All solid tumors | 12.8 | 6.1 |
| Melanoma | 21.4 | 18.3 |
| Ovarian | 14.7 | 6.4 |
| Lung | 14.3 | 7.7 |
| Bladder | 14.2 | 9.5 |
| Colorectal | 13.5 | 6.0 |
| Uterine | 13.3 | 8.3 |
| Pancreatic | 12.9 | 5.4 |
| Breast | 9.6 | 3.6 |
| All hematologic malignancies | 28.2 | 28.7 |
| Multiple myeloma | 37.3 | 42.3 |
| Non-Hodgkin lymphoma | 24.4 | 22.3 |
| Hodgkin lymphoma | 4.7 | 6.4 |
CIT arises from myelosuppression; chemotherapeutic agents affect hematopoietic stem cells and bone marrow microenvironment, reducing megakaryocyte and platelet production [44]. Most CIT is transient (“nadir” counts mid-cycle and recover before the next cycle), but it may become persistent or recurrent with intensive regimens [5]. Clinically, CIT can compromise cancer treatment. It may lead to chemotherapy delays or dose reductions to prevent bleeding, which in turn can worsen oncologic outcomes. Bleeding risk rises as counts drop: in severe thrombocytopenia (e.g., <5 × 109/L), spontaneous hemorrhage is a leading fatal complication [45]. In practice, clinicians often reduce dose intensity when CIT occurs, as reflected by observed inferior survival with decreased dose intensity in various cancers.
Management of CIT is primarily supportive, aiming to reduce disease progression or achieve remission. There are currently no standardized guidelines for CIT, and published guidelines stress that only platelet transfusion reliably prevents bleeding in acute severe CIT. For example, transfusion is indicated for major invasive procedures at platelet counts of ≤50 × 109/L and prophylactic transfusions for <10 × 109/L [46]. Beyond transfusions, a few interventions are used: recombinant human IL-11 oprelvekin was historically approved to raise platelets, but its cardiac toxicity limits use [47]. Thrombopoietin receptor agonists (TPO-RAs, e.g., romiplostim/eltrombopag) have shown promise in stimulating platelet production. In a phase II trial, romiplostim prevented chemotherapy dose reductions in 70% of long-term users and maintained platelet counts with an acceptable safety profile [48]. While TPO-RAs can elevate platelet counts in CIT, their impact on reducing bleeding, transfusion needs, or chemotherapy delays remains uncertain [43]. Thus, current practice for CIT relies on dose modification, transfusion support, and in refractory cases consideration of off-label TPO-RA use.
Immune Thrombocytopenia
Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by low platelet counts due to both increased peripheral destruction and impaired production [49]. Autoantibodies target platelet glycoproteins, leading to their clearance and inducing megakaryocyte apoptosis. Additionally, cytotoxic T cells contribute to platelet destruction and suppress megakaryocyte maturation [50]. ITP may occur primarily or secondary to other conditions such as infection, malignancy, drug usage, and autoimmune disease concomitant. ITP associated with solid tumors has been described through case reports and patient series. Studies that link ITP to hematological neoplasms are mainly focused on non-Hodgkin and Hodgkin lymphoma [51–53]. For solid tumors, ITP is most common in lung cancer, Kaposi’s sarcoma, and breast cancer, followed by renal cell carcinoma and ovarian cancer, but is rare in prostate cancer [54, 55]. Additionally, thrombocytopenia affects 20% of patients with large granular lymphocyte lymphoma due to splenomegaly, ITP, and large granular lymphocyte-induced megakaryopoiesis [56].
Most ITP cases may be asymptomatic and require no emergent treatment, but rarely in patients with suspected ITP and critical hemorrhage, physicians should initiate urgent intervention to rapidly raise platelet counts and restore hemostasis. Current management of ITP is mainly following the 2019 clinical practice guidelines from the American Society of Hematology [57]. Acute therapies include platelet transfusion, high-dose corticosteroids, and intravenous immunoglobulin (IVIG), used alone or in combination with TPO-RAs [57, 58]. For the patients with upcoming procedures, platelet transfusions are previously used in a small fraction of the hospitalized ITP patients despite controversial benefits [59]. A machine learning model analyzing 34 variables found that procedural risk and patient factors (e.g., anticoagulant use, anemia, and age) are primary predictors of post-procedure bleeding in ITP [60]. Platelet transfusion volume correlated with bleeding risk but did not independently reduce it, underscoring the importance of individualized risk assessment over routine transfusion. Large randomized trials in ITP management are scarce, leading to practice variability. A systematic review identified 29 treatment combinations for critical bleeds in ITP [61]. The five most common regimens were corticosteroids plus platelet transfusion and splenectomy, corticosteroids with IVIG, and splenectomy alone. Reported mortality rates were 30.6% in adults and 19.7% in pediatric patients. The lower evidence quality limits the assessment of individual therapies’ impact on outcomes. These findings highlight the urgent need for consensus-based, standardized algorithms to optimize the treatment of ITP.
Clinical Guidelines and Practice for Platelet Transfusion in Cancer Patients
Prophylactic and Therapeutic Platelet Transfusions Use
At present, the main indication for platelet transfusion is bleeding and prevention of bleeding in thrombocytopenic patients with bone marrow failure, intensive chemotherapy, or hematopoietic stem cell transplantation [62, 63]. The Association for the Advancement of Blood & Biotherapies (AABB – formerly known as the American Association of Blood Banks) recommends prophylactic platelet transfusion when the platelet count falls below 10 × 109/L. Therapeutic platelet transfusions are indicated if the platelet count is lower or higher than 10 × 109/L and bleeding occurs [46, 64]. In patients with platelet counts >10 × 109/L, prophylactic platelet transfusion may be considered if additional bleeding risk factors are present or invasive interventions are planned.
Patients with hematologic malignancies who receive chemotherapy or hematopoietic stem cell transplantation often have long-term thrombocytopenia and need frequent platelet transfusions. Evidences from randomized clinical trials and meta-analyses support the use of prophylactic platelet transfusion strategy for patients with hematologic malignancies [62, 65, 66]. Wandt et al. [65] showed that prophylactic platelet transfusion could significantly decrease the incidence of grade 2–4 bleeding events compared with therapeutic transfusion only. In patients with hematological disorder, a transfusion threshold of 10 × 109/L does not increase bleeding risk compared to higher threshold (20 × 109/L or 30 × 109/L), though evidence quality remains low [67]. A systematic review identified seven randomized clinical trials with 1,642 hospitalized thrombocytopenic patients, and prophylactic platelet transfusion may reduce clinically important bleeding relative to no prophylaxis [68]. Notably, the impact on all-cause mortality remained uncertain, highlighting the need for further high-quality research.
In specific clinical scenarios, the optimal use of platelet transfusion remains undefined, such as critical illness, surgical and pediatric patients with thrombocytopenia [69]. Baron et al. [70] reported that, in critically ill cancer patients with thrombocytopenia, over 50% of transfusion episodes results in poor increments, defined as a corrected count increment <7 or a platelet transfusion recovery <0.2. Consistently, a multicenter observational study found 73.9% of prophylactic platelet transfusions in critically ill patients lead to suboptimal response, with 93.5% of hematologic patients exhibiting a corrected count increment ≤7 at 18–24 h after transfusion [71]. These results suggest that standard prophylactic strategies may be less efficient in this population.
Several papers also explored restrictive transfusion in intensive care. Retrospective study by Berenger et al. [72] have shown that a restrictive prophylactic strategy did not result in higher rate of grade ≥2 bleeding or increased 28-day mortality. However, an association between low platelet nadirs (<5 × 109/L) and mortality was found, suggesting that while restrictive strategies seem achievable in ICU, close monitoring is mandatory. Moreover, in procedural settings, omitting prophylaxis transfusion may be associated with increased bleeding risk. Van Baarle et al. [6] reported that thrombocytopenic cancer patients who did not receive prophylactic transfusions before central venous catheter placement experienced significantly more catheter-related bleeding.
Optimal Platelet Transfusion Dose
The optimal dose and threshold of prophylactic platelet transfusion is a major challenge in transfusion medicine. Several studies have compared the clinical outcomes of different doses of prophylactic platelet transfusion for patients with hematologic malignancies. Randomized controlled trials, including the PLADO study, have demonstrated that varying prophylactic platelet transfusion (low dose, 1.1 × 1011/m2; medium dose, 2.2 × 1011/m2; high dose, 4.4 × 1011/m2) in patients with hypoproliferative thrombocytopenia does not significantly influence bleeding incidence, duration of bleeding days, severe hemorrhage, and overall mortality rates [73]. Although higher doses increase the total platelet yield per transfusion, they also heighten the risk of transfusion-related adverse events, while lower doses simply require more frequent transfusions without compromising safety or efficacy. A comprehensive meta-analysis corroborated that low-dose prophylaxis (2.01–4.6 × 1011/L) does not raise bleeding risk compared to higher doses (3.35–7.7 × 1011/L) [74]. Based on these findings, the 2015 International Collaboration for Transfusion Medicine guidelines (ICTMG) recommended low-dose prophylactic platelet transfusion (e.g., 1.1 × 1011/m2 or 2.2 × 1011/m2) for hospitalized patients with chemotherapy- or transplant-induced thrombocytopenia, rather than high-dose strategies (4.4 × 1011/m2) [75]. The conclusions have been supported by subsequent systematic reviews and guideline updates [46, 76, 77]. These findings reported no increase in bleeding outcomes or transfusion frequency between low-dose (1.1 ×1011/m2 ± 25%), standard-dose (2.2 ×1011/m2 ± 25%), and high-dose (4.4 ×1011/m2 ± 25%) and recommended against routine use of high-dose prophylaxis.
Platelet Transfusion in Antibodies Treatment/CAR-T Therapy-Related Thrombocytopenia
Immunotherapy strategies like immune checkpoint inhibitors (ICIs, e.g., PD-1/PD-L1 and CTLA-4 antagonists) and CAR-T transfusion can rarely provoke an ITP-like syndrome in oncology patients. The incidence rate of any grade ≥3 thrombocytopenia is 8.6% in over 1,000 patients receiving ICIs, but only 1.73% was attributed to the ICI [78]. Those developing ICI-related severity thrombocytopenia experienced markedly worse overall survival compared to non-thrombocytopenia (4.17 vs. 13.31 months). Pharmacovigilance analyses reveal significant reporting signals for ICI‐induced ITP across most agents, with pembrolizumab and nivolumab demonstrating the highest odds ratios [79]; combination of PD-1/CTLA-4 appears to carry notable great risk, though no validated patient‐specific predictors have been established to date. Management of ICI‐associated thrombocytopenia follows established ITP protocols. Guidelines recommend continuation of ICIs for grade 1–2 thrombocytopenia, with close monitoring [57]; for grade 3–4 (platelets <50 × 109/L), ICIs should be withheld and high‐dose corticosteroids (±IVIG) initiated promptly. If platelet counts remain <50 × 109/L after 24–72 h of steroids, adjunctive therapies (such as infliximab or rituximab) and TPO-RAs like eltrombopag may be considered [80]; persistent, steroid‐refractory cases often necessitate permanent discontinuation of immunotherapy.
CAR-T therapy also induces significant hematotoxicity (immune effector cell-associated hematotoxicity), with grade ≥3 thrombocytopenia reported in 28–62% of recipients after CD19-directed products (axi-cel, tisa-cel, liso-cel) and contributing to profound and prolonged cytopenias [81, 82]. An international EHA/EBMT survey highlighted substantial heterogeneity in grading and managing immune effector cell-associated hematotoxicity post-CAR-T therapy [83]. These efforts promote best practice consensus recommendations that classify early (day 0–30) versus late (>day 30) cytopenias and outline diagnostic workups, risk stratification (e.g., CAR-HEMATOTOX score), and interventions including growth factors, antimicrobial prophylaxis, transfusions, and stem cell boosts.
Transfusion support is a key surrogate for severe cytopenias, with more than half of patients requiring red blood cells (RBCs) or platelet transfusion in the 6-month time surrounding infusion [81]. Multivariate analyses identify a high CAR-HEMATOTOX score (≥2) and prior transfusion history as consistent predictors of both early and late transfusion need for RBC and platelet, whereas factors like ICANS and tocilizumab use further influence late‐phase platelet requirements. Transfusions may also modulate immune responses that are linked to reduced CAR-T efficacy, with early and late platelet transfusions correlating with inferior PFS and overall survival [84]. Given the critical balance between preventing hemorrhage and avoiding immunosuppressive effects, current guidelines emphasize individualized risk assessment and judicious use of transfusion support. For the prolonged thrombocytopenic patients after CAR-T therapy, TPO-RAs show a possible benefit though data are limited to small case series [82, 85]. Overall, while platelet transfusions remain indispensable for mitigating bleeding risk in immunotherapy and CAR-T recipients, the heterogeneity of patient responses underscores the need for prospective trials to evaluate the impact of transfusion practices on both hemostatic and immunologic outcomes.
Future Development and Prospects of Platelet Transfusion in Cancer Patients
Current significant challenges of platelet transfusion include limited supply due to short shelf life, storage constraints, and transfusion refractoriness. Novel approaches are being investigated to improve platelet available, function, and transfusion efficacy. The ex vivo platelet generation from stem cells may be one promising approach, particularly induced pluripotent stem cells (iPSCs) [86]. This strategy provides a platelet source that is both immunologically compatible and sustainable. The creation of immortalized megakaryocyte progenitor cell lines and the VerMES bioreactor can increase platelet yield by using controlled turbulence [87]. These technologies have enabled to produce more than 100 billion iPSC-derived platelets. These platelets were verified in first-in-human clinical trial (iPLAT1) in a patient with alloimmune transfusion refractoriness [88]. Autologous platelet transfusion was shown to be safety in the trail, with doses increasing from 1 × 1010 to 1 × 1011 platelets [89]. However, platelet count exhibited no significant increase post-transfusion, potentially due to rapid clearance and abnormal platelet activation. Further researches are needed to improve function and circulation of iPSC-derived platelets.
New technologies (e.g., machine learning and artificial intelligence) are also applied to improve transfusion practice. Machine learning models can be used to predict the platelet transfusion need and optimize inventory management [90–92]. Using data from all patients admitted to a medical-multipurpose hospital, Engelke et al. [93] established a deep learning-based risk assessment system to predict platelet transfusion demand in the next 24 h. This model exhibited a high predictive accuracy in hematology-oncology departments (AUC-PR: 0.84; ROC-AUC: 0.98). Another study [94] developed five machine learning models to predict RBC and PLT transfusion requirement in acute myeloid leukemia. These models could also be employed to predict transfusion-related survival. In summary, the future of platelet transfusion in cancer care can be seen in new production methods, platelet biology understanding, and novel technological advancements.
Conflict of Interest Statement
The authors have no conflicts of interest to declare.
Funding Sources
This study was not supported by any sponsor or funder.
Author Contributions
Na Ma and Yufeng Wang wrote the manuscript. Yan Wang and Lvling Zhang reviewed the draft manuscript and made suggestions and modifications. All authors approved the final version for submission.
Funding Statement
This study was not supported by any sponsor or funder.
References
- 1. Shaw JL, Nielson CM, Park JK, Marongiu A, Soff GA. The incidence of thrombocytopenia in adult patients receiving chemotherapy for solid tumors or hematologic malignancies. Eur J Haematol. 2021;106(5):662–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Weycker D, Hatfield M, Grossman A, Hanau A, Lonshteyn A, Sharma A, et al. Risk and consequences of chemotherapy-induced thrombocytopenia in us clinical practice. BMC Cancer. 2019;19(1):151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Kantarjian H, Giles F, List A, Lyons R, Sekeres MA, Pierce S, et al. The incidence and impact of thrombocytopenia in myelodysplastic syndromes. Cancer. 2007;109(9):1705–14. [DOI] [PubMed] [Google Scholar]
- 4. Rodeghiero F. Introduction to a review series on the treatment of thrombocytopenic disorders: something old, something new. Haematologica. 2022;107(6):1239–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Gao A, Zhang L, Zhong D. Chemotherapy-induced thrombocytopenia: literature review. Discov Oncol. 2023;14(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. van Baarle FLF, van de Weerdt EK, van der Velden W, Ruiterkamp RA, Tuinman PR, Ypma PF, et al. Platelet transfusion before cvc placement in patients with thrombocytopenia. N Engl J Med. 2023;388(21):1956–65. [DOI] [PubMed] [Google Scholar]
- 7. Wandt H, Schäfer-Eckart K, Greinacher A. Platelet transfusion in hematology, oncology and surgery. Dtsch Arztebl Int. 2014;111(48):809–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Roberts N, James S, Delaney M, Fitzmaurice C. The global need and availability of blood products: a modelling study. Lancet Haematol. 2019;6(12):e606–15. [DOI] [PubMed] [Google Scholar]
- 9. Zhao J, Huang A, Zeller J, Peter K, McFadyen JD. Decoding the role of platelets in tumour metastasis: enigmatic accomplices and intricate targets for anticancer treatments. Front Immunol. 2023;14:1256129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Garcia-Leon MJ, Liboni C, Mittelheisser V, Bochler L, Follain G, Mouriaux C, et al. Platelets favor the outgrowth of established metastases. Nat Commun. 2024;15(1):3297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Li Y, Wang H, Zhao Z, Yang Y, Meng Z, Qin L. Effects of the interactions between platelets with other cells in tumor growth and progression. Front Immunol. 2023;14:1165989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Capraru A, Jalowiec KA, Medri C, Daskalakis M, Zeerleder SS, Mansouri Taleghani B. Platelet transfusion-insights from current practice to future development. J Clin Med. 2021;10(9):1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Nair P. Preservation of platelet function by cold storage for perfusion. The University of Texas at San Antonio; 2013. [Google Scholar]
- 14. Acker JP, Marks DC, Sheffield WP. Quality assessment of established and emerging blood components for transfusion. J Blood Transfus. 2016;2016:4860284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Miles J, Bailey SL, Obenaus AM, Mollica MY, Usaneerungrueng C, Byrne D, et al. Storage temperature determines platelet gpvi levels and function in mice and humans. Blood Adv. 2021;5(19):3839–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Trochanowska-Pauk N, Walski T, Bohara R, Mikolas J, Kubica K. Platelet storage-problems, improvements, and new perspectives. Int J Mol Sci. 2024;25(14):7779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Tynngård N, Alshamari A, Sandgren P, Kenny D, Vasilache AM, Abedi MR, et al. High fragmentation in platelet concentrates impacts the activation, procoagulant, and aggregatory capacity of platelets. Platelets. 2023;34(1):2159018. [DOI] [PubMed] [Google Scholar]
- 18. Lu W, Fung M. Platelets treated with pathogen reduction technology: current status and future direction. F1000Res. 2020;9:F1000 Faculty Rev-40. [Google Scholar]
- 19. Piccin A, Allameddine A, Spizzo G, Lappin KM, Prati D. Platelet pathogen reduction technology-should we stay or should we go…? J Clin Med. 2024;13(18):5359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Rebulla P, Prati D. Pathogen reduction for platelets-a review of recent implementation strategies. Pathogens. 2022;11(2):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Gravemann U, Boelke M, Könenkamp L, Söder L, Maurer M, Ziegler U, et al. West nile and usutu viruses are efficiently inactivated in platelet concentrates by uvc light using the theraflex uv-platelets system. Vox Sang. 2024;119(8):827–33. [DOI] [PubMed] [Google Scholar]
- 22. Hobson-Peters J, Amarilla AA, Rustanti L, Marks DC, Roulis E, Khromykh AA, et al. Inactivation of sars-cov-2 infectivity in platelet concentrates or plasma following treatment with ultraviolet c light or with methylene blue combined with visible light. Transfusion. 2023;63(2):288–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Salunkhe V, van der Meer PF, de Korte D, Seghatchian J, Gutiérrez L. Development of blood transfusion product pathogen reduction treatments: a review of methods, current applications and demands. Transfus Apher Sci. 2015;52(1):19–34. [DOI] [PubMed] [Google Scholar]
- 24. Schlenke P. Pathogen inactivation technologies for cellular blood components: an update. Transfus Med Hemother. 2014;41(4):309–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Di Minno G, Navarro D, Perno CF, Canaro M, Gürtler L, Ironside JW, et al. Pathogen reduction/inactivation of products for the treatment of bleeding disorders: what are the processes and what should we say to patients? Ann Hematol. 2017;96(8):1253–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Picker SM. Current methods for the reduction of blood-borne pathogens: a comprehensive literature review. Blood Transfus. 2013;11(3):343–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Solheim BG. Pathogen reduction of blood components. Transfus Apher Sci. 2008;39(1):75–82. [DOI] [PubMed] [Google Scholar]
- 28. Kuznetsov AI, Ivanov AL, Idelson LI, Mazurov AV. Mechanisms of thrombocytopenia in patients with lymphoproliferative diseases. Eur J Haematol. 1992;49(3):113–8. [DOI] [PubMed] [Google Scholar]
- 29. Yang H, He F, Yuan T, Xu W, Cao Z. Clinical features and treatment of bone marrow metastasis. Oncol Lett. 2023;26(2):332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Yang R, Jia L, Cui J. Mechanism and clinical progression of solid tumors bone marrow metastasis. Front Pharmacol. 2024;15:1390361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Piatek CI, Liebman HA. Thrombocytopenia in patients with solid tumors or hematologic malignancies. Platelets Thrombotic Non-Thrombotic Disord Pathophysiology. 2017:841–50. [Google Scholar]
- 32. Takamatsu Y, Jimi S, Sato T, Hara S, Suzumiya J, Tamura K. Thrombocytopenia in association with splenomegaly during granulocyte-colony-stimulating factor treatment in mice is not caused by hypersplenism and is resolved spontaneously. Transfusion. 2007;47(1):41–9. [DOI] [PubMed] [Google Scholar]
- 33. Jacobs KE, Visser BC, Gayer G. Changes in spleen volume after resection of hepatic colorectal metastases. Clin Radiol. 2012;67(10):982–7. [DOI] [PubMed] [Google Scholar]
- 34. Ando H, Nagino M, Arai T, Nishio H, Nimura Y. Changes in splenic volume during liver regeneration. World J Surg. 2004;28(10):977–81. [DOI] [PubMed] [Google Scholar]
- 35. Pozo AL, Godfrey EM, Bowles KM. Splenomegaly: investigation, diagnosis and management. Blood Rev. 2009;23(3):105–11. [DOI] [PubMed] [Google Scholar]
- 36. Thai LH, Mahévas M, Roudot-Thoraval F, Limal N, Languille L, Dumas G, et al. Long-term complications of splenectomy in adult immune thrombocytopenia. Medicine. 2016;95(48):e5098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Hill A, Elakkad A, Kuban J, Sabir S, Odisio B, Huang SY, et al. Durability of partial splenic artery embolization on platelet counts for cancer patients with hypersplenism-related thrombocytopenia. Abdom Radiol. 2020;45(9):2886–94. [Google Scholar]
- 38. El SDAEA, Allam HM, M AR. Can partial splenic embolization be a promising treatment for hypersplenism–related thrombocytopenia in oncological patients requiring systemic chemotherapy: a retrospective analysis. Zagazig Univ Med J. 2021;27:962–72. [Google Scholar]
- 39. Adelborg K, Larsen JB, Hvas AM. Disseminated intravascular coagulation: epidemiology, biomarkers, and management. Br J Haematol. 2021;192(5):803–18. [DOI] [PubMed] [Google Scholar]
- 40. Posado-Domínguez L, Chamorro AJ, Del Barco-Morillo E, Martín-Galache M, Bueno-Sacristán D, Fonseca-Sánchez E, et al. Cancer-associated thrombotic microangiopathy: literature review and report of five cases. Life. 2024;14(7):865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Wu Y, Aravind S, Ranganathan G, Martin A, Nalysnyk L. Anemia and thrombocytopenia in patients undergoing chemotherapy for solid tumors: a descriptive study of a large outpatient oncology practice database, 2000-2007. Clin Ther. 2009;31 Pt 2(Pt 2):2416–32. [DOI] [PubMed] [Google Scholar]
- 42. Hitron A, Steinke D, Sutphin S, Lawson A, Talbert J, Adams V. Incidence and risk factors of clinically significant chemotherapy-induced thrombocytopenia in patients with solid tumors. J Oncol Pharm Pract. 2011;17(4):312–9. [DOI] [PubMed] [Google Scholar]
- 43. Soff G, Leader A, Al-Samkari H, Falanga A, Maraveyas A, Sanfilippo K, et al. Management of chemotherapy-induced thrombocytopenia: guidance from the isth subcommittee on hemostasis and malignancy. J Thromb Haemost. 2024;22(1):53–60. [DOI] [PubMed] [Google Scholar]
- 44. De Silva E, Kim H. Drug-induced thrombocytopenia: focus on platelet apoptosis. Chem Biol Interact. 2018;284:1–11. [DOI] [PubMed] [Google Scholar]
- 45. Cines DB, Blanchette VS. Immune thrombocytopenic purpura. N Engl J Med. 2002;346(13):995–1008. [DOI] [PubMed] [Google Scholar]
- 46. Schiffer CA, Bohlke K, Delaney M, Hume H, Magdalinski AJ, McCullough JJ, et al. Platelet transfusion for patients with cancer: American society of clinical oncology clinical practice guideline update. J Clin Oncol. 2018;36(3):283–99. [DOI] [PubMed] [Google Scholar]
- 47. Al-Samkari H, Soff GA. Clinical challenges and promising therapies for chemotherapy-induced thrombocytopenia. Expert Rev Hematol. 2021;14(5):437–48. [DOI] [PubMed] [Google Scholar]
- 48. Wilkins CR, Ortiz J, Gilbert LJ, Yin S, Mones JV, Parameswaran R, et al. Romiplostim for chemotherapy-induced thrombocytopenia: efficacy and safety of extended use. Res Pract Thromb Haemost. 2022;6(3):e12701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. McMillan R, Wang L, Tomer A, Nichol J, Pistillo J. Suppression of in vitro megakaryocyte production by antiplatelet autoantibodies from adult patients with chronic itp. Blood. 2004;103(4):1364–9. [DOI] [PubMed] [Google Scholar]
- 50. Zufferey A, Kapur R, Semple JW. Pathogenesis and therapeutic mechanisms in immune thrombocytopenia (itp). J Clin Med. 2017;6(2):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Fallah M, Liu X, Ji J, Försti A, Sundquist K, Hemminki K. Autoimmune diseases associated with non-hodgkin lymphoma: a nationwide cohort study. Ann Oncol. 2014;25(10):2025–30. [DOI] [PubMed] [Google Scholar]
- 52. Fallah M, Liu X, Ji J, Försti A, Sundquist K, Hemminki K. Hodgkin lymphoma after autoimmune diseases by age at diagnosis and histological subtype. Ann Oncol. 2014;25(7):1397–404. [DOI] [PubMed] [Google Scholar]
- 53. Teixeira Mendes LS, Wotherspoon A. Marginal zone lymphoma: associated autoimmunity and auto-immune disorders. Best Pract Res Clin Haematol. 2017;30(1–2):65–76. [DOI] [PubMed] [Google Scholar]
- 54. Krauth MT, Puthenparambil J, Lechner K. Paraneoplastic autoimmune thrombocytopenia in solid tumors. Crit Rev Oncol Hematol. 2012;81(1):75–81. [DOI] [PubMed] [Google Scholar]
- 55. Zahidin MA, Mohd Noor NH, Johan MF, Abdullah AD, Zulkafli Z, Edinur HA. A review on secondary immune thrombocytopenia in Malaysia. Healthc. 2021;10(1):38. [Google Scholar]
- 56. Lai DW, Loughran TP Jr, Maciejewski JP, Sasu S, Song SX, Epling-Burnette PK, et al. Acquired amegakaryocytic thrombocytopenia and pure red cell aplasia associated with an occult large granular lymphocyte leukemia. Leuk Res. 2008;32(5):823–7. [DOI] [PubMed] [Google Scholar]
- 57. Neunert C, Terrell DR, Arnold DM, Buchanan G, Cines DB, Cooper N, et al. American society of hematology 2019 guidelines for immune thrombocytopenia. Blood Adv. 2019;3(23):3829–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Zitek T, Weber L, Pinzon D, Warren N. Assessment and management of immune thrombocytopenia (itp) in the emergency department: current perspectives. Open Access Emerg Med. 2022;14:25–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Goel R, Chopra S, Tobian AAR, Ness PM, Frank SM, Cushing M, et al. Platelet transfusion practices in immune thrombocytopenia related hospitalizations. Transfusion. 2019;59(1):169–76. [DOI] [PubMed] [Google Scholar]
- 60. Kang KW, Choi Y, Lim HJ, Kwak K, Choi YS, Park Y, et al. Impact of platelet transfusion and bleeding risk stratification in patients with immune thrombocytopenia before procedures. Sci Rep. 2025;15(1):5174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Chowdhury SR, Sirotich E, Guyatt G, Gill D, Modi D, Venier LM, et al. Treatment of critical bleeds in patients with immune thrombocytopenia: a systematic review. Eur J Haematol. 2025;114(3):458–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Estcourt L, Stanworth S, Doree C, Hopewell S, Murphy MF, Tinmouth A, et al. Prophylactic platelet transfusion for prevention of bleeding in patients with haematological disorders after chemotherapy and stem cell transplantation. Cochrane Database Syst Rev. 2012;2012(5):CD004269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Estcourt LJ, Stanworth SJ, Murphy MF. Platelet transfusions for patients with haematological malignancies: who needs them? Br J Haematol. 2011;154(4):425–40. [DOI] [PubMed] [Google Scholar]
- 64. Kaufman RM, Djulbegovic B, Gernsheimer T, Kleinman S, Tinmouth AT, Capocelli KE, et al. Platelet transfusion: a clinical practice guideline from the aabb. Ann Intern Med. 2015;162(3):205–13. [DOI] [PubMed] [Google Scholar]
- 65. Wandt H, Schaefer-Eckart K, Wendelin K, Pilz B, Wilhelm M, Thalheimer M, et al. Therapeutic platelet transfusion versus routine prophylactic transfusion in patients with haematological malignancies: an open-label, multicentre, randomised study. Lancet. 2012;380(9850):1309–16. [DOI] [PubMed] [Google Scholar]
- 66. Stanworth SJ, Estcourt LJ, Llewelyn CA, Murphy MF, Wood EM; TOPPS Study Investigators . Impact of prophylactic platelet transfusions on bleeding events in patients with hematologic malignancies: a subgroup analysis of a randomized trial. Transfusion. 2014;54(10):2385–93. [DOI] [PubMed] [Google Scholar]
- 67. Estcourt LJ, Stanworth SJ, Doree C, Hopewell S, Trivella M, Murphy MF. Comparison of different platelet count thresholds to guide administration of prophylactic platelet transfusion for preventing bleeding in people with haematological disorders after myelosuppressive chemotherapy or stem cell transplantation. Cochrane Database Syst Rev. 2015;2015(11):Cd010983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Anthon CT, Granholm A, Sivapalan P, Zellweger N, Pène F, Puxty K, et al. Prophylactic platelet transfusions versus no prophylaxis in hospitalized patients with thrombocytopenia: a systematic review with meta-analysis. Transfusion. 2022;62(10):2117–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Maier CL, Stanworth SJ, Sola-Visner M, Kor D, Mast AE, Fasano R, et al. Prophylactic platelet transfusion: is there evidence of benefit, harm, or no effect? Transfus Med Rev. 2023;37(4):150751. [DOI] [PubMed] [Google Scholar]
- 70. Baron E, Charpentier J, François A, Ben Hadj Amor H, Habr B, Cariou A, et al. Post-transfusion platelet responses in critically ill cancer patients with hypoproliferative thrombocytopenia. Transfusion. 2020;60(2):275–84. [DOI] [PubMed] [Google Scholar]
- 71. Reizine F, Le Marec S, Le Meur A, Consigny M, Berteau F, Bodenes L, et al. Prophylactic platelet transfusion response in critically ill patients: a prospective multicentre observational study. Crit Care. 2023;27(1):373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Berenger JB, Saillard C, Sannini A, Servan L, Gonzalez F, Faucher M, et al. Prophylactic versus restrictive platelet transfusion strategy in patients with haematological malignancies in the icu setting, a propensity-score analysis. J Crit Care. 2024;83:154817. [DOI] [PubMed] [Google Scholar]
- 73. Slichter SJ, Kaufman RM, Assmann SF, McCullough J, Triulzi DJ, Strauss RG, et al. Dose of prophylactic platelet transfusions and prevention of hemorrhage. N Engl J Med. 2010;362(7):600–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Cid J, Lozano M. Lower or higher doses for prophylactic platelet transfusions: results of a meta-analysis of randomized controlled trials. Transfusion. 2007;47(3):464–70. [DOI] [PubMed] [Google Scholar]
- 75. Nahirniak S, Slichter SJ, Tanael S, Rebulla P, Pavenski K, Vassallo R, et al. Guidance on platelet transfusion for patients with hypoproliferative thrombocytopenia. Transfus Med Rev. 2015;29(1):3–13. [DOI] [PubMed] [Google Scholar]
- 76. Estcourt LJ, Stanworth S, Doree C, Trivella M, Hopewell S, Blanco P, 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;2015(10):Cd010984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Estcourt LJ, Birchall J, Allard S, Bassey SJ, Hersey P, Kerr JP, et al. Guidelines for the use of platelet transfusions. Br J Haematol. 2017;176(3):365–94. [DOI] [PubMed] [Google Scholar]
- 78. Haddad TC, Zhao S, Li M, Patel SH, Johns A, Grogan M, et al. Immune checkpoint inhibitor-related thrombocytopenia: incidence, risk factors and effect on survival. Cancer Immunol Immunother. 2022;71(5):1157–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Liu G, Zhang S, Mo Z, Huang T, Yu Q, Lu X, et al. Association of thrombocytopenia with immune checkpoint inhibitors: a large-scale pharmacovigilance analysis based on the data from fda adverse event reporting system database. Front Pharmacol. 2024;15:1407894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Mingot-Castellano ME, Reguera-Ortega JL, Zafra Torres D, Hernani R, Lopez-Godino O, Guerreiro M, et al. Use of eltrombopag to improve thrombocytopenia and tranfusion requirement in anti-cd19 car-t cell-treated patients. J Clin Med. 2024;13(17):5117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Vic S, Thibert JB, Bachy E, Cartron G, Gastinne T, Morschhauser F, et al. Transfusion needs after car t-cell therapy for large b-cell lymphoma: predictive factors and outcome (a descar-t study). Blood Adv. 2024;8(6):1573–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Drillet G, Lhomme F, De Guibert S, Manson G, Houot R. Prolonged thrombocytopenia after car t-cell therapy: the role of thrombopoietin receptor agonists. Blood Adv. 2023;7(4):537–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Rejeski K, Subklewe M, Aljurf M, Bachy E, Balduzzi A, Barba P, et al. Immune effector cell-associated hematotoxicity: eha/ebmt consensus grading and best practice recommendations. Blood. 2023;142(10):865–77. [DOI] [PubMed] [Google Scholar]
- 84. Sut C, Tariket S, Aubron C, Aloui C, Hamzeh-Cognasse H, Berthelot P, et al. The non-hemostatic aspects of transfused platelets. Front Med. 2018;5:42. [Google Scholar]
- 85. Beyar-Katz O, Perry C, On YB, Amit O, Gutwein O, Wolach O, et al. Thrombopoietin receptor agonist for treating bone marrow aplasia following anti-cd19 car-t cells-single-center experience. Ann Hematol. 2022;101(8):1769–76. [DOI] [PubMed] [Google Scholar]
- 86. Yang J, Luan J, Shen Y, Chen B. Developments in the production of platelets from stem cells (review). Mol Med Rep. 2020;23(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Sugimoto N, Nakamura S, Shimizu S, Shigemasa A, Kanda J, Matsuyama N, et al. Production and nonclinical evaluation of an autologous ipsc-derived platelet product for the iplat1 clinical trial. Blood Adv. 2022;6(23):6056–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Kayama A, Eto K. Mass production of ipsc-derived platelets toward the clinical application. Regen Ther. 2024;25:213–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Sugimoto N, Kanda J, Nakamura S, Kitano T, Hishizawa M, Kondo T, et al. Iplat1: the first-in-human clinical trial of ipsc-derived platelets as a phase 1 autologous transfusion study. Blood. 2022;140(22):2398–402. [DOI] [PubMed] [Google Scholar]
- 90. Maynard S, Farrington J, Alimam S, Evans H, Li K, Wong WK, et al. Machine learning in transfusion medicine: a scoping review. Transfusion. 2024;64(1):162–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Schilling M, Rickmann L, Hutschenreuter G, Spreckelsen C. Reduction of platelet outdating and shortage by forecasting demand with statistical learning and deep neural networks: modeling study. JMIR Med Inform. 2022;10(2):e29978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Motamedi M, Dawson J, Li N, Down DG, Heddle NM. Demand forecasting for platelet usage: from univariate time series to multivariable models. PLoS One. 2024;19(4):e0297391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Engelke M, Schmidt CS, Baldini G, Parmar V, Hosch R, Borys K, et al. Optimizing platelet transfusion through a personalized deep learning risk assessment system for demand management. Blood. 2023;142(26):2315–26. [DOI] [PubMed] [Google Scholar]
- 94. Wei Y, Xia T, Ma J, Gao X, Ge Z. Machine learning applications for prediction of blood transfusion and survival in acute myeloid leukemia. Blood. 2022;140(Suppl 1):2831–2. [Google Scholar]
