Transfusion therapy in sickle cell disease
Review Article

Transfusion therapy in sickle cell disease

Yvette C. Tanhehco1, Patricia A. Shi2,3,4, Joseph Schwartz5

1Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; 2New York Blood Center, New York, NY, USA; 3Department of Hematology-Oncology, Albert Einstein College of Medicine, Bronx, NY, USA; 4Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Long Island, NY, USA; 5Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA

Contributions: (I) Conception and design: All authors; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: None; (V) Data analysis and interpretation: None; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Patricia A. Shi, MD. 310 E 67th Street, New York, NY 10065, USA. Email: pshi@nybc.org; Yvette C. Tanhehco, PhD, MD, MS. Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, 622 W 168th Street, Harkness Pavilion 4-418A, New York, NY, USA. Email: yct2103@cumc.columbia.edu.

Abstract: Sickle cell disease (SCD), the most common hemoglobinopathy worldwide after thalassemia, is characterized by a mutation in the β-globin gene that results in the production of hemoglobin S (HbS). HbS polymerizes under deoxygenated conditions, causing red blood cell (RBC) rigidity that leads to vaso-occlusion, hemolysis and endothelial damage. In addition to hydration, pain medication, and disease-modifying drug therapy, RBC transfusion is a mainstay of treatment. Simple or exchange (manual or automated) transfusions may be performed. RBC transfusions improve blood oxygen content, decrease HbS-containing RBCs, increase HbAA RBCs with normal oxygen affinity, and may suppress endogenous hematopoiesis. The goal of RBC transfusions is to decrease HbS levels to less than 30% and maintain the patient’s hematocrit at less than 30%. The choice of performing a simple or exchange transfusion is partly dependent on the indication. Acute indications include acute stroke, acute chest syndrome (ACS), acute multisystem organ failure, intrahepatic cholestasis, hepatic/splenic sequestration, and priapism. Chronic indications include stroke prophylaxis, silent infarcts, recurrent ACS, recurrent painful episodes and complicated pregnancy. The frequency of RBC transfusions and quantity of RBC units transfused should be weighed against the risks of transfusion reactions. Aside from being at risk for the common transfusion reactions such as febrile nonhemolytic transfusion reactions and allergic reactions, patients with SCD are at an increased risk for alloimmunization, autoimmunization, iron overload and delayed hemolytic reactions with hyperhemolysis. This review discusses various aspects of transfusion therapy for SCD.

Keywords: Red blood cell transfusion; sickle cell disease (SCD); hemoglobin S


Received: 21 September 2021; Accepted: 05 November 2021; Published: 31 March 2022.

doi: 10.21037/aob-21-67


Introduction

Sickle cell disease (SCD) is an inherited monogenic disease characterized by the β-globin gene mutation (1) hemoglobin S (HbS, β6 glutamic acid-valine). SCD is an important hemoglobinopathy worldwide in terms of frequency and social impact, recently recognized as a global public health problem by the World Health Organization (2). In sub-Saharan Africa, up to one third of adults are carriers of the defective sickle cell gene, and 1% to 2% of babies are born with the disease (3). Globally, approximately 300,000 infants are born per year with SCD (4). The most common types of SCD are homozygous hemoglobin S (HbSS disease), hemoglobin SC disease, and sickle β thalassemia. HbSS disease and sickle β0 thalassemia often are referred to as sickle cell anemia because they have similar severity (5).

SCD is a multisystem disorder with high morbidity and mortality. Hemoglobin polymerization, leading to erythrocyte rigidity and vaso-occlusion, is central to its pathophysiology, and chronic anemia, hemolysis, and vasculopathy are hallmarks of the disease (6). These physiologic derangements often lead to multiorgan damage throughout the lifespan. Acute, chronic, and acute-on-chronic complications contribute to end-organ damage and adversely affect life expectancy as well as quality of life (1).

Because polymerization of sickle hemoglobin (HbS) is the root cause of SCD complications, SCD therapies are focused on preventing HbS polymerization and reducing or suppressing the circulating amount of HbS (7). This review focuses on transfusion therapy as a supportive and preventive measure. Indications for transfusion, transfusion-related complications relatively specific to SCD, and methods for transfusion, i.e., simple and red blood cell (RBC) exchange are discussed.


Rationale and methods of transfusion in SCD

In patients without SCD, the typical purpose of red cell transfusion is to increase hemoglobin (Hb) to improve blood oxygen content according to the equation: CaO2=(SaO2×Hb×1.34)+0.003(PaO2) where CaO2 is the arterial oxygen content, SaO2 is the arterial oxygen saturation and PaO2 is the arterial oxygen tension. In SCD, however, there are two additional benefits of transfusion. One is lowering of the percentage of HbS-containing red cells with the capacity to undergo Hb polymerization under deoxygenated conditions; this is critical, because Hb polymerization is the root cause of all the downstream damage in SCD (8). For example, the resulting impaired rheology of deoxygenated red cells is sufficient to increase blood viscosity and slow blood flow velocity at arterial oxygen tension even without additional contributions from inflammation, adhesion, and endothelial and leukocyte activation (9,10), and there is improvement in sickle red cell rheology with transfusion (9,11). A second benefit is increasing the percentage of red cells with normal oxygen affinity, as HbSS red cells have lower oxygen affinity (12), probably due to high intracellular HbS concentration rather than high 2,3-DPG. This increases oxygen saturation at a given PaO2, thus increasing blood oxygen content and decreasing the probability that HbS-containing red cells will reach the point of sickling with tissue oxygen extraction (13). A third theoretical benefit is that increasing Hb, given the longer life span of HbAA red cells, may decrease endogenous reticulocytosis and production of HbS-containing red cells (14,15), although studies have not demonstrated this to date (16,17). Transfusion can be administered through three methods, depending on clinical considerations and feasibility: simple transfusion (ST) [transfusion of allogeneic packed red blood cells (PRBC) without autologous red cell removal], manual red cell exchange (RCE) [autologous whole blood phlebotomy alternating with allogeneic PRBC and fluid infusion (18)], or automated RCE (removal of autologous RBC with allogeneic PRBC replacement using an apheresis device).


Simple transfusion

ST is the technically easiest method of transfusion, especially in acute clinical scenarios, due to the typical use of peripheral venous access and standard nursing. As is true in the general non-SCD population, ST is indicated with isolated symptomatic anemia, such as that resulting from parvovirus B19 aplastic crisis. It is important to point out, however, that SCD patients are anemic at baseline, with patients of SS and SB0 genotype typically having chronic Hbs between 7–9 gm/dL (19-21). Furthermore, with vaso-occlusive episodes, SCD patients should receive hypotonic saline or D5W hydration to decrease MCHC and thus reduce HbS polymerization (22,23), hydration, however, will cause a lowering of the Hb without an actual decrease in blood oxygen content. Therefore, the standard restrictive transfusion thresholds (24,25) (typically 7–8 gm/dL) for transfusion established in randomized clinical trials may not be applicable to a SCD patient chronically tolerized to anemia. It is especially relevant to avoid unnecessary transfusion in uncomplicated vaso-occlusive episodes because the acute inflammatory state may increase the risk of red cell alloimmunization (26). However, even a clinically asymptomatic Hb <5 gm/dL is likely an indication for transfusion (in the absence of known hyperhemolysis), because acute anemia around 4 gm/dL or lower in SCD has been associated with acute silent cerebral ischemia and infarction (27). Others recommend ST with a fall in Hb ≥2 gm/dL below the baseline Hb, or in children, a Hb <6 gm/dL (28). Standard blood management practices such as minimizing phlebotomy for blood samples should be practiced.

ST has also been recommended by expert consensus for splenic sequestration (29,30). Limited case reports indicate potential safety of RCE for sequestration without hypotension (31), however, and RCE has the theoretical advantage of potentially minimizing hyperviscosity issues that can occur when sequestered red cells re-enter the circulation. Even when RCE is preferable due to its ability to more rapidly lower HbS%, such as with acute stroke (32), transient ischemic attack (33), or rapidly progressive or severe acute chest syndrome (ACS) (34), ST may be indicated acutely as a temporizing measure until RCE can be performed. Finally, per the randomized controlled TAPS trial (35), ST to a Hb target of 10 gm/dL is indicated pre-operatively in SS or Sβ0 patients with a baseline Hb <9 gm/dL undergoing surgeries requiring general or regional anesthesia; only 13 of 66 patients assessed in the trial had surgeries defined as low-risk, but one of those in the no-transfusion group had the serious adverse event of ACS after an umbilical hernia repair. Furthermore, a randomized trial (36) showed no significant differences in the peri-operative complications with ST versus RCE, including the most common serious complication of ACS, and the RCE group had a significantly higher rate of new red cell alloantibody development as well as delayed hemolytic transfusion reactions (DHTR).

ST, although requiring the least number of red cell units, has inevitable iron loading because no autologous red cells are removed (37,38). In a planned secondary analysis of transfusion data from the Silent Cerebral Infarct Multi-Center Clinical Trial, the median (interquartile range) ferritin level after 1 year of transfusions was 1,800 ng/mL (1,426–2,204 ng/mL) with ST, 1,530 ng/mL (1,205–1,805 ng/mL) with manual exchange, and 355 ng/mL (179–579 ng/mL) with automated exchange (P<0.001) (38).

In adults, ST of 1 red cell unit will typically increase the Hb in an average size adult by 1 gm/dL (and the Hct by 3%); children must be dosed by weight, and typically 10–15 mL/kg will increase the Hb by 2–3 gm/dL (39). In chronic ST, no more than about 3 units have been transfused per transfusion episode (37). Ideally, the volume of red cells needed to achieve the desired Hct should be precisely calculated as: [(desired Hctstarting Hct)×patient total blood volume]Hct of red cell unit  due to the risk of hyperviscosity or transfusion-associated circulatory overload (TACO). This is especially true in patients with pre-existing positive fluid balance or cardiovascular or renal disease, and rate of transfusion should be slow if clinically appropriate (40,41). The risk of hyperviscosity and TACO with ST typically precludes achievement of a HbS reduction to ≤30–50% in the acute setting; furthermore, increasing Hct to >30% without lowering the HbS ≤30% is not recommended, because the resulting increase in blood viscosity (42) may contribute to further vaso-occlusive complications, including posterior reversible encephalopathy syndrome and even death (43-45).


Exchange transfusion

Although automated and manual RCE require more red cell units than ST, their advantage is the ability to more rapidly achieve a low HbS level without an increase in hematocrit or total blood volume, if desired; iron loading (37,38,46), hyperviscosity, and TACO can thus be minimized. Compared to ST, RCE has an improved hematocrit:viscosity ratio, which is a mechanical estimate of oxygen delivery potential in the microcirculation (47). RCE has also been shown to result in an increase in arterial oxygen pressure, thus contributing to a further increase in Hb oxygen saturation (13,48); this is posited to be due to improvements in pulmonary microcirculation which may reduce ventilation-perfusion mismatching and intrapulmonary shunting. Importantly, the increased number of units used with chronic RCE has not been associated with increased red cell alloimmunization rates (49,50).

Both manual and automated methods require specialized expertise and supplies and may require central line placement; automated RCE of course requires the apheresis device. In manual RCE, whole blood removal alternates sequentially with replacement, whereas with automated RCE red cells selectively are removed and can be replaced concurrently. Therefore, manual RCE takes more time than automated RCE and has the additional risk of adverse events from volume shifts. Automated RCE also has the advantage of precise programming of the target HbS %, hematocrit, and overall fluid balance. It is recommended that a pre- and post-CBC and Hb fractionation be obtained to precisely tailor the programmed parameters (34). In patients whose extracorporeal volume with apheresis exceeds 10–15% of their total blood volume, an albumin prime is recommended.

Precise programming of the target fraction of cells remaining and increasing the target end Hct may be able to reduce red cell usage, as circumstances require (15). A modified automated RCE, in which RBC depletion by isovolemic hemodilution with saline or 5% albumin occurs prior to RCE, may also decrease red cell usage or increase the post-procedure Hct, associated with increased pre-procedure Hct (14,51). The increased pre-procedure Hct may be beneficial given that anemia is associated with silent brain infarcts in SCD (52). Isovolemic hemodilution should be avoided in clinical scenarios where induction of further anemia may be detrimental, such as recent cerebral ischemic events; to reduce acute silent cerebral ischemic events (53), hemodilution should probably not decrease the hematocrit to less than 21% and/or more than 20% from baseline (34).

Regardless of method of transfusion, to minimize increased blood viscosity, the end hematocrit (Hct) goal of transfusion, for acute indications, is usually targeted no higher than 30% and the HbS target is <30% (54). Acute indications for RCE include acute stroke (32); rapidly progressive or severe ACS (34); multi-organ failure (55); fat embolism syndrome (56); acute sickle cell hepatic crisis, sickle cell intra-hepatic cholestasis, or acute hepatic sequestration (57,58); and pre-operatively in patients with SC and Sβ+ disease and Hb ≥10 gm/dL undergoing moderate-risk surgery (59) or patients undergoing very high risk surgery (34). For chronic indications, the end Hct is usually targeted no higher than 33-36% and the HbS target ranges between 30–50% (54), depending on the indication. Chronic indications for RCE or ST include prophylaxis against stroke (60,61), silent infarcts (62), recurrent ACS (62,63), recurrent painful episodes (62,63), and complicated pregnancy (34,54,64). After at least 1 year of chronic transfusion, hydroxyurea may be non-inferior to transfusion for primary stroke prophylaxis in patients with no Magnetic Resonance Angiography (MRA)-defined severe vasculopathy (65), but not for secondary stroke prophylaxis (66).


Indications for RBC transfusion

The indications for RBC transfusion in patients with SCD may be categorized as acute and chronic (67). Acute indications for RCE include acute stroke (32); rapidly progressive or severe ACS (34); multi-organ failure (55); fat embolism syndrome (56); acute sickle cell hepatic crisis, sickle cell intra-hepatic cholestasis, or acute hepatic sequestration (57,58); and pre-operatively in patients with SC and Sβ+ disease and Hb ≥10 gm/dL undergoing moderate-risk surgery (59) or patients undergoing very high risk surgery (34). Chronic indications for RCE include prophylaxis against stroke (60,61), silent infarcts (62), recurrent ACS (62,63), recurrent painful episodes (62,63), and complicated pregnancy (34,54,64). As noted above, after at least 1 year of chronic transfusion, hydroxyurea may be non-inferior to transfusion for primary stroke prophylaxis in patients with no MRA-defined severe vasculopathy (65), but not for secondary stroke prophylaxis (66).

In the absence of transfusion, stroke occurs in approximately 11% of patients with sickle cell anemia by the age of 20 (68,69). The Stroke Prevention Trial in Sickle Cell Anemia (STOP), where 130 children with SS or Sβ0 were randomized to red cell transfusions to maintain HbS ≤30% or not, demonstrated that red cell transfusions significantly reduces the risk of a first stroke in children with SCD who have abnormal results on transcranial Doppler (TCD) ultrasonography (60). The mean interval between transfusions was 25±8 days. Only 1 child in the transfusion group had a stroke (cerebral infarction) as opposed to 11 children (10 cerebral infarctions, 1 intracerebral hematoma) in the standard of care group, which represents a 92% difference in the risk of stroke (P<0.001). Due to this compelling evidence, the trial was terminated early (60). The STOP trial led to the recommendations for TCD screening and prophylactic transfusion for children with abnormal velocities on ultrasonography (70).

Due to concerns of adverse effects with transfusing red cells indefinitely, the Optimizing Primary Stroke Prevention in Sickle Cell Anemia (STOP II) trial was undertaken, where patients after ≥30 months of chronic transfusion were randomized to continue transfusion or not. The trial was halted after only 79 of the planned 100 patients were enrolled because almost half of the patients in the transfusion-halted group reached the primary composite end-point (2 strokes, 14 reversions to abnormal velocity on TCD) within 10 months after randomization, whereas none of the 38 children in the transfusion-continued group had an end-point event (P<0.001). Overall, this trial demonstrated that when red cell transfusions are discontinued in primary stroke prevention patients whose TCD velocities have normalized, there is a high rate of reversion to abnormal blood-flow velocities or stroke (61). In primary stroke prevention, the TWITCH trial showed that after 12 months of transfusion therapy, changing to hydroxyurea is non-inferior to transfusion in SCD children without severe vasculopathy (65), but in secondary stroke prevention, the SWITCH trial suggests that hydroxyurea is inferior to transfusion (66); there were no (0/66) strokes on transfusions/chelation but 10% (7/67) on hydroxyurea/phlebotomy when the trial was stopped for futility for lack of a difference in iron removal between the two arms.

ACS, characterized by fever, respiratory symptoms and a new pulmonary infiltrate, is the second most common cause of hospitalization and the leading cause of death in patients with SCD (71-73). Red cell transfusion early in the course of ACS has been found to hasten resolution (74), improve oxygenation (75,76), and reduce recurrence (63,77). Several studies have implicated elevated levels of secretory phospholipase A2 (sPLA2) to be correlated with clinical severity of ACS (78-83). The feasibility study Preventing Acute Chest Syndrome with Transfusion Trial (PROACTIVE, ClinicalTrials.gov NCT00951808) was thus conducted to determine if elevation of serum levels of sPLA2 >100 ng/mL and fever could predict the development of ACS and whether ACS could be prevented by a single early transfusion of 7–13 mL/kg (79). Analysis of 203 subjects showed the sPLA2 threshold of >100 ng/mL to have a specificity of 82%, but sensitivity of only 41% and positive predictive value (PPV) of only 21%. sPLA2 ≥48 ng/mL provided the optimal trade-off between sensitivity (73%) and specificity (71%), but still only provided a PPV of 24%.

Patients with SCD frequently require surgery due to obstructive sleep apnea, adenotonsillar hypertrophy, cholelithiasis, splenic sequestration, and avascular necrosis. Perioperative complications related and not related to SCD are common. The Transfusion Alternatives Preoperatively in Sickle Cell Disease (TAPS) study was a multicenter, randomized clinical trial that assessed in pediatric and adult patients with HbSS or HbSβ0 undergoing low- or medium-risk elective surgery whether perioperative complication rates would be altered by preoperative transfusion within 10 days of surgery to a target Hb of 10 gm/dL (35). Thirteen of 33 (39%) in the no-preoperative-transfusion group had clinically significant complications as opposed to 5/34 (15%) in the preoperative-transfusion group (P=0.023). Serious adverse events were observed in 10/33 (30%) subjects in the no-preoperative group and 1/34 (3%) subjects in the preoperative-transfusion group (95% CI: 1.2–12.2, P=0.027). Ten of the 11 serious adverse events were ACS (9 non-preoperative-transfusion group, 1 preoperative transfusion group). Hospital length of stay and readmission rates were similar between the two groups. Additionally, the study found that without preoperative transfusion, the need for perioperative transfusion was increased. Preoperative transfusion, therefore, is beneficial for patients undergoing low-risk and medium-risk surgeries (35). The type of RBC transfusion for pre-operative management depends on the patient’s baseline Hb level (34). For patients with a Hb level less than 9 gm/dL, ST is recommended while for patients with a Hb level equal to greater than 9 gm/dL, RBC exchange transfusion to a HbS target <60% is recommended. The target Hb level in both cases should not exceed 11 gm/dL. For patients undergoing very high risk surgery such as neurosurgery or cardiac surgery, RBC exchange transfusion is recommended (34).

Pregnancy in women with SCD is associated with increased risks of sickle-related complications such as pain crises, pulmonary complications and infection; maternal and perinatal mortality, pregnancy-related complications; and fetal complications (84,85). Studies of the role of prophylactic red cell transfusions in pregnant women are limited (64,86-88). The only prospective randomized controlled study so far randomized 72 pregnant women with SCD to prophylactic transfusions versus on-demand transfusions for medical or obstetric emergencies (87). The incidence of painful crises was significantly reduced (P<0.01) and other SCD-related complications substantially reduced (P<0.07), but there was no significant difference in perinatal outcomes (87). However, a large meta-analysis did find a benefit for both maternal and neonatal outcomes (64). A study protocol has been designed to determine the feasibility and acceptability of a future phase 3 randomized controlled trial (89) to establish the clinical and cost effectiveness of serial prophylactic exchange blood transfusion in pregnant women with SCD (TAPS-2). Pregnant women will be randomized to receive red cell transfusion only when clinically indicated or automated RCE to maintain HbS-containing red cells <30% starting at or before 18 weeks of gestation until the end of pregnancy. In the meantime, current guidelines recommend considering prophylactic transfusion at regular intervals at the onset of pregnancy for women with: a history of severe SCD-related complications prior to current pregnancy; additional features of high-risk pregnancy (e.g., comorbidities or multiparity), or the development of SCD-related complications during the current pregnancy. A target Hb level of at least 7 gm/dL and HbS level of <50% is recommended (34).

Contraindications to transfusion in SCD include acute vaso-occlusive pain episodes and asymptomatic anemia (90). Vaso-occlusive pain episodes are primarily managed with supportive treatment, hypotonic fluid hydration, and pharmacologic drugs such as opioids but psychological, social and behavioral interventions may be incorporated for a more holistic approach (91). The treatment of asymptomatic anemia should be directed at the underlying cause (e.g., parvovirus B19 infection or renal insufficiency).


Adverse reactions to RBC transfusion

Red cell transfusions can lead to severe discomfort, morbidity or even mortality and increases healthcare costs (92). Patients with SCD may be at a greater risk for transfusion reactions due to the higher number of red cell units transfused at one time. Febrile non-hemolytic transfusion reactions and allergic transfusion reactions are the most common types of transfusion reactions experienced by any patient receiving red cell transfusions. They have a prevalence of 1,000–3,000 per 100,000 units transfused and 112.2 per 100,000 units transfused, respectively (92). The less common but more serious transfusion reactions that could lead to significant morbidity and mortality such as acute hemolytic transfusion reactions (prevalence: 2.5–7.9/100,000 units transfused), DHTR (prevalence: 40/100,000 units transfused), transfusion related acute lung injury (prevalence: 0.4–1/100,000 units transfused), transfusion associated circulatory overload (prevalence: 10.9/100,000 units transfused), and septic transfusion reactions (prevalence: 0.03–3.3/100,000 units transfused) may also occur in the frequently transfused SCD patient population (92). This review will focus on three types of transfusion adverse events that are more specific for patients with SCD who are chronically transfused; namely, red cell antibody formation, iron overload and hyperhemolytic transfusion reaction.

RBC antibody formation

Alloantibody formation against RBC blood group antigens is a major complication for patients with SCD. Our understanding of the pathophysiological mechanisms that contribute to this phenomenon is limited. One factor that increases alloantibody induction in patients with SCD is their exposure to red cell antigens present in the blood donor population (mostly Caucasian) but lacking on their own red cells. Others factors are immune dysregulation in white blood cell subsets and their response to heme, platelets, RBCs and complement (93-96). The inflammatory status of the recipient at the time of RBC exposure may also influence whether a patient becomes alloimmunized or not; red cell transfusion in the setting of vaso-occlusive crisis, ACS, a viral illness, or other inflammatory disorder may increase the risk for alloimmunization (26,97,98).

In order to mitigate alloantibody formation, it is a common practice to match the RBC antigens for Rh D, C, and E, and K in accordance with the National Institutes of Health (NIH) Expert Panel, American Society of Hematology, and British Society for Haematology guidelines (30,34,43,99). The RBC alloimmunization rate is reported to be as high as 50% without any prophylactic matching but decreases significantly with prophylactic matching (100-102). With limited prophylactic matching (i.e., C, E, K), the RBC alloimmunization rate ranges from 5–24% (103) and with extended prophylactic matching that includes matching for Duffy, Kidd and S antigens, the RBC alloimmunization rate can be as low as 7% (104).

Even with transfusion of red cell units from African American donors matched for Rh D, C, and E, and K, however, 7 of 59 (12%) episodic and 55 of 123 (45%) chronically transfused patients with SCD were nevertheless Rh alloimmunized; 25 patients (40%) had greater than 1 Rh antibody (105). Rh alloimmunization was due to variant RH genes in either the transfused patients or their African-American donors, with both populations having 29% of RHD and 53% of RHCE alleles altered.

Patients with SCD may also develop autoantibodies. SCD disease-related inflammation and RBC membrane changes with neoantigen exposure may contribute to the predisposition for autoantibody formation. One retrospective study found that 29% (23/78) of pediatric patients developed clinically significant alloantibodies and 8% (6/78) also developed autoantibodies. Four of these 6 patients (66%) had previously identified IgG alloantibodies (106). Autoantibodies may cause clinically significant hemolysis and complicate the immunohematology workup as well as subsequent pRBC crossmatching.

Iron overload

Repeated STs, as is required for patients with SCD, may lead to iron overload. Each pRBC unit contains approximately 200–250 mg of iron, about the amount normally absorbed in one year (107,108). Iron is retained and recycled by the reticuloendothelial system in the body and there is no physiologic regulatory mechanism for excreting excess iron. Senescent RBCs are phagocytized by macrophages (34) in the liver and spleen, where iron is scavenged into endosomes and delivered to the vascular space via ferroportin (107,109). Once in the plasma, the iron is rapidly bound by serum transferrin and transported to the bone marrow. Excess transferrin-bound iron is taken up by the liver, which is where iron is predominantly stored in the body (107). When transferrin becomes completely saturated, non-transferrin bound iron may be deposited in the liver, heart, pancreas, endocrine glands, and joints (107,110). Studies have shown that iron may be transported by zinc (111) or calcium (112) transporters as well.

Iron overload may occur after approximately 10–20 units of pRBC are transfused. In general, a 15 mL/kg transfusion will raise hepatic iron, normally less than 1.5 mg/g dry weight (113), by approximately 1 mg/mL dry weight (114). Among 12 pediatric patients with an average of 15.4 transfusions over 21 months, the mean liver iron concentration (LIC) was 9.4 mg/g dry weight without chelation therapy (115), a level at which one starts to see portal fibrosis. Therefore, patients with a history of at least 10 transfusions and a serum ferritin >1,000 mcg/L should undergo magnetic resonance imaging (MRI) of the liver to detect iron overload. MRI is the gold standard for monitoring iron levels in various organs of the body due to its accuracy, reproducibility and tolerability by patients (107); the same method (R2, T2* or R2*) should be used over time. Given that cardiac iron loading is uncommon in SCD, cardiac MRI should be considered only with high iron burden (LIC >15 mg/g) for 2 years or more, evidence of end organ damage due to transfusional iron overload, or evidence of cardiac dysfunction (34).

Iron chelation, by which the three FDA-approved agents differ by route of administration, half-life, and toxicities, should be started once the LIC is >3 mg/g, and definitely once the LIC is >7 mg/g (113). Over 1 year, a similar dose-dependent decrease in liver iron content was observed with oral deferasirox (−3.0±6.2 mg Fe/g dry weight) and intravenous/subcutaneous deferoxamine (−2.8±10.4 mg Fe/g dry weight) (116). Deferiprone may be the most effective agent with cardiac iron overload (113).

Hyperhemolytic transfusion reaction

A small percentage of transfused patients with SCD (1–4%) experience a hyperhemolytic transfusion reaction (HHTR), defined by a rapid Hb decline to below the pre-transfusion level accompanied by a rapid decline of post-transfusion HbA level about 7–10 days after RBC transfusion (34,117,118). Autologous as well as transfused red cells are hemolyzed in a phenomenon called “bystander hemolysis” (119,120). Most patients also have a negative antibody screen and direct antiglobulin test, signifying the absence of any detectable antibodies (118), although occasionally, patients may exhibit a new alloantibody or autoantibody (121). Possible mechanisms of bystander hemolysis include increased red cell exposure of phosphatidylserine (119), antibodies to antigens other than red cell antigens (120), macrophage activation by the inflammatory milieu (117,122-124), and alternative complement activation by heme (125). Such mechanisms are not mutually exclusive—for example, exposure of phosphatidylserine on the outer membrane of HbS-containing RBCs increases macrophage adherence compared to HbA-containing RBCs (126).

Continued RBC transfusion may lead to death in patients with HHTR, as it may further exacerbate hemolysis (117,127). The type of treatment to administer depends on the severity of anemia and briskness of hemolysis. Supportive care includes erythropoietin with or without IV iron, oxygen as needed, and limiting activity to minimize oxygen consumption. Mild HHTR may be treated with steroids, while more severe hemolysis should also be treated with intravenous immunoglobulin (IVIG) (117,128). Second-line treatments include eculizumab, a monoclonal antibody that inhibits the complement pathway by blocking cleavage of C5, in order to decrease hemolysis; and/or rituximab, an anti-CD20 monoclonal antibody, in order to prevent red cell alloantibody production if further transfusion is necessary (129-132). Tocilizumab, an anti-interleukin-6 receptor monoclonal antibody, was used successfully in an HHTR case with hyperferritinemia under the premise of activated macrophage inhibition (133).


Conclusions

Despite advances in therapeutic options for patients with SCD, transfusion therapy remains a mainstay. There are immediate benefits to transfusion, such as lowering of the percentage of HbS-containing red cells with the capacity to undergo Hb polymerization under deoxygenated conditions. In addition, transfusion increases the percentage of RBC with normal oxygen affinity, thus increasing oxygen saturation and blood oxygen content and decreasing the probability that HbS-containing red cells will reach the point of sickling. Transfusion of RBCs can be delivered via three methods including ST, manual RCE, or automated RCE. Each of these methods carry advantages and disadvantages as described above. The choice of delivery method for a specific patient will depend on clinical considerations and feasibility.

Regardless of method of transfusion, the indications for RBC transfusion in patients with SCD may be categorized as acute and chronic. As general rule of thumb, the end hematocrit (Hct) goal of transfusion, for acute indications, is usually targeted no higher than 30% and the HbS target is <30%; Similarly, for chronic indications, the end Hct is usually targeted no higher than 33–36% and the HbS target ranges between 30–50%, depending on the indication. There are also contraindications to transfusion in SCD, such as acute vaso-occlusive pain episodes and asymptomatic anemia. Last but not least, one has to consider the risks associated with transfusion therapy: transfusion reactions, increased risk for alloantibody and autoantibody formation, development of iron overload, and hyperhemolysis.


Acknowledgments

Funding: Dr. Shi was supported by NHLBI PO1 HL149626.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editor (Paul D. Mintz) for the series “Transfusion Therapy: Principles and Practices” published in Annals of Blood. The article has undergone external peer review.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://aob.amegroups.com/article/view/10.21037/aob-21-67/coif). The series “Transfusion Therapy: Principles and Practices” was commissioned by the editorial office without any funding or sponsorship. YCT is owner and consultant of Leukocytes LLC, and she is on the Board of Directors for BBANYS and Committee Chair of BBANYS, AABB and ASFA, but it does not have any relevant financial interests to this manuscript. JS reports that he is board member of FACT and committee membership of ASFA and AABB, but none of these are related to the current manuscript. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Azar S, Wong TE. Sickle Cell Disease: A Brief Update. Med Clin North Am 2017;101:375-93. [Crossref] [PubMed]
  2. Pinto VM, Balocco M, Quintino S, et al. Sickle cell disease: a review for the internist. Intern Emerg Med 2019;14:1051-64. [Crossref] [PubMed]
  3. Meremikwu MM, Okomo U. Sickle cell disease. BMJ Clin Evid 2016.
  4. Sickle Cell Disease. World Heatlh Organization; 2021 August 30. Available online: https://www.afro.who.int/health-topics/sickle-cell-disease.
  5. Noronha SA, Sadreameli SC, Strouse JJ. Management of Sickle Cell Disease in Children. South Med J 2016;109:495-502. [Crossref] [PubMed]
  6. Rees DC, Williams TN, Gladwin MT. Sickle-cell disease. Lancet 2010;376:2018-31. [Crossref] [PubMed]
  7. Meier ER. Treatment Options for Sickle Cell Disease. Pediatr Clin North Am 2018;65:427-43. [Crossref] [PubMed]
  8. Eaton WA, Bunn HF. Treating sickle cell disease by targeting HbS polymerization. Blood 2017;129:2719-26. [Crossref] [PubMed]
  9. Lu X, Wood DK, Higgins JM. Deoxygenation Reduces Sickle Cell Blood Flow at Arterial Oxygen Tension. Biophys J 2016;110:2751-8. [Crossref] [PubMed]
  10. Chien S, Usami S, Bertles JF. Abnormal rheology of oxygenated blood in sickle cell anemia. J Clin Invest 1970;49:623-34. [Crossref] [PubMed]
  11. Rab MAE, Kanne CK, Bos J, et al. Oxygen gradient ektacytometry-derived biomarkers are associated with vaso-occlusive crises and correlate with treatment response in sickle cell disease. Am J Hematol 2021;96:E29-32. [Crossref] [PubMed]
  12. Seakins M, Gibbs WN, Milner PF, et al. Erythrocyte Hb-S concentration. An important factor in the low oxygen affinity of blood in sickle cell anemia. J Clin Invest 1973;52:422-32. [Crossref] [PubMed]
  13. Uchida K, Rackoff WR, Ohene-Frempong K, et al. Effect of erythrocytapheresis on arterial oxygen saturation and hemoglobin oxygen affinity in patients with sickle cell disease. Am J Hematol 1998;59:5-8. [Crossref] [PubMed]
  14. Ziemba Y, Xu C, Fomani KM, et al. Safety and benefits of automated red cell depletion-exchange compared to standard exchange in patients with sickle cell disease undergoing chronic transfusion. Transfusion 2021;61:526-36. [Crossref] [PubMed]
  15. Uter S, An HH, Linder GE, et al. Measures to reduce red cell use in patients with sickle cell disease requiring red cell exchange during a blood shortage. Blood Adv 2021;5:2586-92. [Crossref] [PubMed]
  16. Zinkham WH, Seidler AJ, Kickler TS. Variable degrees of suppression of hemoglobin S synthesis in subjects with hemoglobin SS disease on a long-term transfusion regimen. J Pediatr 1994;124:215-9. [Crossref] [PubMed]
  17. Kaushal M, Byrnes C, Khademian Z, et al. Examination of Reticulocytosis among Chronically Transfused Children with Sickle Cell Anemia. PLoS One 2016;11:e0153244. [Crossref] [PubMed]
  18. Swerdlow PS. Red cell exchange in sickle cell disease. Hematology Am Soc Hematol Educ Program 2006;48-53. [Crossref] [PubMed]
  19. Day ME, Rodeghier M, DeBaun MR. Children with HbSβ0 thalassemia have higher hemoglobin levels and lower incidence rate of acute chest syndrome compared to children with HbSS. Pediatr Blood Cancer 2018;65:e27352. [Crossref] [PubMed]
  20. Hayes RJ, Beckford M, Grandison Y, et al. The haematology of steady state homozygous sickle cell disease: frequency distributions, variation with age and sex, longitudinal observations. Br J Haematol 1985;59:369-82. [Crossref] [PubMed]
  21. West MS, Wethers D, Smith J, et al. Laboratory profile of sickle cell disease: a cross-sectional analysis. The Cooperative Study of Sickle Cell Disease. J Clin Epidemiol 1992;45:893-909. [Crossref] [PubMed]
  22. Carden MA, Fay M, Sakurai Y, et al. Normal saline is associated with increased sickle red cell stiffness and prolonged transit times in a microfluidic model of the capillary system. Microcirculation 2017; [Crossref] [PubMed]
  23. Carden MA, Fay ME, Lu X, et al. Extracellular fluid tonicity impacts sickle red blood cell deformability and adhesion. Blood 2017;130:2654-63. [Crossref] [PubMed]
  24. Trentino KM, Farmer SL, Leahy MF, et al. Systematic reviews and meta-analyses comparing mortality in restrictive and liberal haemoglobin thresholds for red cell transfusion: an overview of systematic reviews. BMC Med 2020;18:154. [Crossref] [PubMed]
  25. Carson JL, Stanworth SJ, Roubinian N, et al. Transfusion thresholds and other strategies for guiding allogeneic red blood cell transfusion. Cochrane Database Syst Rev 2016;10:CD002042. [Crossref] [PubMed]
  26. Fasano RM, Booth GS, Miles M, et al. Red blood cell alloimmunization is influenced by recipient inflammatory state at time of transfusion in patients with sickle cell disease. Br J Haematol 2015;168:291-300. [Crossref] [PubMed]
  27. Dowling MM, Quinn CT, Plumb P, et al. Acute silent cerebral ischemia and infarction during acute anemia in children with and without sickle cell disease. Blood 2012;120:3891-7. [Crossref] [PubMed]
  28. DeBaun MRaC, S.T. Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques 2021 [07/11/2021]. Available online: https://www.uptodate.com/contents/red-blood-cell-transfusion-in-sickle-cell-disease-indications-and-transfusion-techniques
  29. Linder GE, Chou ST. Red cell transfusion and alloimmunization in sickle cell disease. Haematologica 2021;106:1805-15. [Crossref] [PubMed]
  30. Yawn BP, Buchanan GR, Afenyi-Annan AN, et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. JAMA 2014;312:1033-48. [Crossref] [PubMed]
  31. Vijayanarayanan A, Omosule AJ, Saad H, et al. Acute Splenic Sequestration Crisis in Hemoglobin SC Disease: Efficiency of Red Cell Exchange. Cureus 2020;12:e12224. [Crossref] [PubMed]
  32. Hulbert ML, Scothorn DJ, Panepinto JA, et al. Exchange blood transfusion compared with simple transfusion for first overt stroke is associated with a lower risk of subsequent stroke: a retrospective cohort study of 137 children with sickle cell anemia. J Pediatr 2006;149:710-2. [Crossref] [PubMed]
  33. DeBaun MR, Jordan LC, King AA, et al. American Society of Hematology 2020 guidelines for sickle cell disease: prevention, diagnosis, and treatment of cerebrovascular disease in children and adults. Blood Adv 2020;4:1554-88. [Crossref] [PubMed]
  34. Chou ST, Alsawas M, Fasano RM, et al. American Society of Hematology 2020 guidelines for sickle cell disease: transfusion support. Blood Adv 2020;4:327-55. [Crossref] [PubMed]
  35. Howard J, Malfroy M, Llewelyn C, et al. The Transfusion Alternatives Preoperatively in Sickle Cell Disease (TAPS) study: a randomised, controlled, multicentre clinical trial. Lancet 2013;381:930-8. [Crossref] [PubMed]
  36. Vichinsky EP, Haberkern CM, Neumayr L, et al. A comparison of conservative and aggressive transfusion regimens in the perioperative management of sickle cell disease. The Preoperative Transfusion in Sickle Cell Disease Study Group. N Engl J Med 1995;333:206-13. [Crossref] [PubMed]
  37. Fasano RM, Leong T, Kaushal M, et al. Effectiveness of red blood cell exchange, partial manual exchange, and simple transfusion concurrently with iron chelation therapy in reducing iron overload in chronically transfused sickle cell anemia patients. Transfusion 2016;56:1707-15. [Crossref] [PubMed]
  38. Kelly S, Rodeghier M, DeBaun MR. Automated exchange compared to manual and simple blood transfusion attenuates rise in ferritin level after 1 year of regular blood transfusion therapy in chronically transfused children with sickle cell disease. Transfusion 2020;60:2508-16. [Crossref] [PubMed]
  39. AABB ARC, America's Blood Centers, Armed Services Blood Program. Circular of Information for the Use of Human Blood and Blood Components. https://www.aabb.org/news-resources/resources/circular-of-information2017
  40. Murphy EL, Kwaan N, Looney MR, et al. Risk factors and outcomes in transfusion-associated circulatory overload. Am J Med 2013;126:357.e29-38. [Crossref] [PubMed]
  41. De Cloedt L, Savy N, Gauvin F, et al. Transfusion-Associated Circulatory Overload in ICUs: A Scoping Review of Incidence, Risk Factors, and Outcomes. Crit Care Med 2019;47:849-56. [Crossref] [PubMed]
  42. Detterich J, Alexy T, Rabai M, et al. Low-shear red blood cell oxygen transport effectiveness is adversely affected by transfusion and further worsened by deoxygenation in sickle cell disease patients on chronic transfusion therapy. Transfusion 2013;53:297-305. [Crossref] [PubMed]
  43. Davis BA, Allard S, Qureshi A, et al. Guidelines on red cell transfusion in sickle cell disease. Part I: principles and laboratory aspects. Br J Haematol 2017;176:179-91. [Crossref] [PubMed]
  44. Di Vincenzo A, Marson P, Puato M. Acute Splenic Sequestration Crisis After Red Blood Cell Exchange for Acute Chest Syndrome in an Adult With Sickle β-Thalassemia: What Went Wrong? Ther Apher Dial 2018;22:207-8. [Crossref] [PubMed]
  45. Royal JE, Seeler RA. Hypertension, convulsions, and cerebral haemorrhage in sickle-cell anaemia patients after blood-transfusions. Lancet 1978;2:1207. [Crossref] [PubMed]
  46. Kim HC, Dugan NP, Silber JH, et al. Erythrocytapheresis therapy to reduce iron overload in chronically transfused patients with sickle cell disease. Blood 1994;83:1136-42. [Crossref] [PubMed]
  47. Detterich JA. Simple chronic transfusion therapy, a crucial therapeutic option for sickle cell disease, improves but does not normalize blood rheology: What should be our goals for transfusion therapy? Clin Hemorheol Microcirc 2018;68:173-86. [Crossref] [PubMed]
  48. Lanzkowsky P, Shende A, Karayalcin G, et al. Partial exchange transfusion in sickle cell anemia. Use in children with serious complications. Am J Dis Child 1978;132:1206-8. [Crossref] [PubMed]
  49. Wahl SK, Garcia A, Hagar W, et al. Lower alloimmunization rates in pediatric sickle cell patients on chronic erythrocytapheresis compared to chronic simple transfusions. Transfusion 2012;52:2671-6. [Crossref] [PubMed]
  50. Michot JM, Driss F, Guitton C, et al. Immunohematologic tolerance of chronic transfusion exchanges with erythrocytapheresis in sickle cell disease. Transfusion 2015;55:357-63. [Crossref] [PubMed]
  51. Hequet O, Poutrel S, Connes P, et al. Automatic depletion with Spectra Optia allows a safe 16% reduction of red blood cell pack consumption in exchanged sickle cell anemia patients. Transfusion 2019;59:1692-7. [Crossref] [PubMed]
  52. Ford AL, Ragan DK, Fellah S, et al. Silent infarcts in sickle cell disease occur in the border zone region and are associated with low cerebral blood flow. Blood 2018;132:1714-23. [Crossref] [PubMed]
  53. Quinn CT, McKinstry RC, Dowling MM, et al. Acute silent cerebral ischemic events in children with sickle cell anemia. JAMA Neurol 2013;70:58-65. [Crossref] [PubMed]
  54. Karafin MS, Hendrickson JE, Kim HC, et al. Red cell exchange for patients with sickle cell disease: an international survey of current practices. Transfusion 2020;60:1424-33. [Crossref] [PubMed]
  55. Hassell KL, Eckman JR, Lane PA. Acute multiorgan failure syndrome: a potentially catastrophic complication of severe sickle cell pain episodes. Am J Med 1994;96:155-62. [Crossref] [PubMed]
  56. Greaves P, Mathew V, Peters C, et al. Successful outcome of three patients with sickle-cell disease and fat embolism syndrome treated with intensive exchange transfusion. Clin Case Rep 2016;5:39-43. [Crossref] [PubMed]
  57. Allali S, de Montalembert M, Brousse V, et al. Hepatobiliary Complications in Children with Sickle Cell Disease: A Retrospective Review of Medical Records from 616 Patients. J Clin Med 2019;8:1481. [Crossref] [PubMed]
  58. Gardner K, Suddle A, Kane P, et al. How we treat sickle hepatopathy and liver transplantation in adults. Blood 2014;123:2302-7. [Crossref] [PubMed]
  59. Neumayr L, Koshy M, Haberkern C, et al. Surgery in patients with hemoglobin SC disease. Preoperative Transfusion in Sickle Cell Disease Study Group. Am J Hematol 1998;57:101-8. [Crossref] [PubMed]
  60. Adams RJ, McKie VC, Hsu L, et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography. N Engl J Med 1998;339:5-11. [Crossref] [PubMed]
  61. Adams RJ, Brambilla DOptimizing Primary Stroke Prevention in Sickle Cell Anemia. (STOP 2) Trial Investigators. Discontinuing prophylactic transfusions used to prevent stroke in sickle cell disease. N Engl J Med 2005;353:2769-78. [Crossref] [PubMed]
  62. DeBaun MR, Gordon M, McKinstry RC, et al. Controlled trial of transfusions for silent cerebral infarcts in sickle cell anemia. N Engl J Med 2014;371:699-710. [Crossref] [PubMed]
  63. Miller ST, Wright E, Abboud M, et al. Impact of chronic transfusion on incidence of pain and acute chest syndrome during the Stroke Prevention Trial (STOP) in sickle-cell anemia. J Pediatr 2001;139:785-9. [Crossref] [PubMed]
  64. Malinowski AK, Shehata N, D'Souza R, et al. Prophylactic transfusion for pregnant women with sickle cell disease: a systematic review and meta-analysis. Blood 2015;126:2424-35; quiz 2437. [Crossref] [PubMed]
  65. Ware RE, Davis BR, Schultz WH, et al. Hydroxycarbamide versus chronic transfusion for maintenance of transcranial doppler flow velocities in children with sickle cell anaemia-TCD With Transfusions Changing to Hydroxyurea (TWiTCH): a multicentre, open-label, phase 3, non-inferiority trial. Lancet 2016;387:661-70. [Crossref] [PubMed]
  66. Ware RE, Helms RW. SWiTCH Investigators. Stroke With Transfusions Changing to Hydroxyurea (SWiTCH). Blood 2012;119:3925-32. [Crossref] [PubMed]
  67. Kozanoglu I, Ozdogu H. Use of red blood cell exchange for treating acute complications of sickle cell disease. Transfus Apher Sci 2018;57:23-6. [Crossref] [PubMed]
  68. Ohene-Frempong K, Weiner SJ, Sleeper LA, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood 1998;91:288-94. [PubMed]
  69. Powars D, Wilson B, Imbus C, et al. The natural history of stroke in sickle cell disease. Am J Med 1978;65:461-71. [Crossref] [PubMed]
  70. Goldstein LB, Adams R, Becker K, et al. Primary prevention of ischemic stroke: A statement for healthcare professionals from the Stroke Council of the American Heart Association. Stroke 2001;32:280-99. [Crossref] [PubMed]
  71. Platt OS, Brambilla DJ, Rosse WF, et al. Mortality in sickle cell disease. Life expectancy and risk factors for early death. N Engl J Med 1994;330:1639-44. [Crossref] [PubMed]
  72. Sprinkle RH, Cole T, Smith S, et al. Acute chest syndrome in children with sickle cell disease. A retrospective analysis of 100 hospitalized cases. Am J Pediatr Hematol Oncol 1986;8:105-10. [PubMed]
  73. Vichinsky EP. Comprehensive care in sickle cell disease: its impact on morbidity and mortality. Semin Hematol 1991;28:220-6. [PubMed]
  74. Mallouh AA, Asha M. Beneficial effect of blood transfusion in children with sickle cell chest syndrome. Am J Dis Child 1988;142:178-82. [PubMed]
  75. Emre U, Miller ST, Gutierez M, et al. Effect of transfusion in acute chest syndrome of sickle cell disease. J Pediatr 1995;127:901-4. [Crossref] [PubMed]
  76. Vichinsky EP, Neumayr LD, Earles AN, et al. Causes and outcomes of the acute chest syndrome in sickle cell disease. National Acute Chest Syndrome Study Group. N Engl J Med 2000;342:1855-65. [Crossref] [PubMed]
  77. Hankins J, Jeng M, Harris S, et al. Chronic transfusion therapy for children with sickle cell disease and recurrent acute chest syndrome. J Pediatr Hematol Oncol 2005;27:158-61. [Crossref] [PubMed]
  78. Ballas SK, Files B, Luchtman-Jones L, et al. Secretory phospholipase A2 levels in patients with sickle cell disease and acute chest syndrome. Hemoglobin 2006;30:165-70. [Crossref] [PubMed]
  79. Styles L, Wager CG, Labotka RJ, et al. Refining the value of secretory phospholipase A2 as a predictor of acute chest syndrome in sickle cell disease: results of a feasibility study (PROACTIVE). Br J Haematol 2012;157:627-36. [Crossref] [PubMed]
  80. Styles LA, Aarsman AJ, Vichinsky EP, et al. Secretory phospholipase A(2) predicts impending acute chest syndrome in sickle cell disease. Blood 2000;96:3276-8. [Crossref] [PubMed]
  81. Styles LA, Abboud M, Larkin S, et al. Transfusion prevents acute chest syndrome predicted by elevated secretory phospholipase A2. Br J Haematol 2007;136:343-4. [Crossref] [PubMed]
  82. Styles LA, Schalkwijk CG, Aarsman AJ, et al. Phospholipase A2 levels in acute chest syndrome of sickle cell disease. Blood 1996;87:2573-8. [Crossref] [PubMed]
  83. Naprawa JT, Bonsu BK, Goodman DG, et al. Serum biomarkers for identifying acute chest syndrome among patients who have sickle cell disease and present to the emergency department. Pediatrics 2005;116:e420-5. [Crossref] [PubMed]
  84. Oteng-Ntim E, Ayensah B, Knight M, et al. Pregnancy outcome in patients with sickle cell disease in the UK--a national cohort study comparing sickle cell anaemia (HbSS) with HbSC disease. Br J Haematol 2015;169:129-37. [Crossref] [PubMed]
  85. Oteng-Ntim E, Meeks D, Seed PT, et al. Adverse maternal and perinatal outcomes in pregnant women with sickle cell disease: systematic review and meta-analysis. Blood 2015;125:3316-25. [Crossref] [PubMed]
  86. Benites BD, Benevides TC, Valente IS, et al. The effects of exchange transfusion for prevention of complications during pregnancy of sickle hemoglobin C disease patients. Transfusion 2016;56:119-24. [Crossref] [PubMed]
  87. Koshy M, Burd L, Wallace D, et al. Prophylactic red-cell transfusions in pregnant patients with sickle cell disease. A randomized cooperative study. N Engl J Med 1988;319:1447-52. [Crossref] [PubMed]
  88. Okusanya BO, Oladapo OT. Prophylactic versus selective blood transfusion for sickle cell disease in pregnancy. Cochrane Database Syst Rev 2016;12:CD010378. [Crossref] [PubMed]
  89. Oakley LL, Awogbade M, Brien S, et al. Serial prophylactic exchange blood transfusion in pregnant women with sickle cell disease (TAPS-2): study protocol for a randomised controlled feasibility trial. Trials 2020;21:347. [Crossref] [PubMed]
  90. Sharma D, Ogbenna AA, Kassim A, et al. Transfusion support in patients with sickle cell disease. Semin Hematol 2020;57:39-50. [Crossref] [PubMed]
  91. Uwaezuoke SN, Ayuk AC, Ndu IK, et al. Vaso-occlusive crisis in sickle cell disease: current paradigm on pain management. J Pain Res 2018;11:3141-50. [Crossref] [PubMed]
  92. Delaney M, Wendel S, Bercovitz RS, et al. Transfusion reactions: prevention, diagnosis, and treatment. Lancet 2016;388:2825-36. [Crossref] [PubMed]
  93. Balandya E, Reynolds T, Obaro S, et al. Alteration of lymphocyte phenotype and function in sickle cell anemia: Implications for vaccine responses. Am J Hematol 2016;91:938-46. [Crossref] [PubMed]
  94. Lard LR, Mul FP, de Haas M, et al. Neutrophil activation in sickle cell disease. J Leukoc Biol 1999;66:411-5. [Crossref] [PubMed]
  95. Zhang D, Xu C, Manwani D, et al. Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood 2016;127:801-9. [Crossref] [PubMed]
  96. Yazdanbakhsh K, Shaz BH, Hillyer CD. Immune Regulation of sickle Cell Alloimmunization. ISBT Sci Ser 2017;12:248-53. [Crossref] [PubMed]
  97. Evers D, van der Bom JG, Tijmensen J, et al. Red cell alloimmunisation in patients with different types of infections. Br J Haematol 2016;175:956-66. [Crossref] [PubMed]
  98. Ryder AB, Hendrickson JE, Tormey CA. Chronic inflammatory autoimmune disorders are a risk factor for red blood cell alloimmunization. Br J Haematol 2016;174:483-5. [Crossref] [PubMed]
  99. Davis BA, Allard S, Qureshi A, et al. Guidelines on red cell transfusion in sickle cell disease Part II: indications for transfusion. Br J Haematol 2017;176:192-209. [Crossref] [PubMed]
  100. Venkateswaran L, Teruya J, Bustillos C, et al. Red cell exchange does not appear to increase the rate of allo- and auto-immunization in chronically transfused children with sickle cell disease. Pediatr Blood Cancer 2011;57:294-6. [Crossref] [PubMed]
  101. Vichinsky EP, Luban NL, Wright E, et al. Prospective RBC phenotype matching in a stroke-prevention trial in sickle cell anemia: a multicenter transfusion trial. Transfusion 2001;41:1086-92. [Crossref] [PubMed]
  102. O'Suoji C, Liem RI, Mack AK, et al. Alloimmunization in sickle cell anemia in the era of extended red cell typing. Pediatr Blood Cancer 2013;60:1487-91. [Crossref] [PubMed]
  103. Fasano RM, Meyer EK, Branscomb J, et al. Impact of Red Blood Cell Antigen Matching on Alloimmunization and Transfusion Complications in Patients with Sickle Cell Disease: A Systematic Review. Transfus Med Rev 2019;33:12-23. [Crossref] [PubMed]
  104. Lasalle-Williams M, Nuss R, Le T, et al. Extended red blood cell antigen matching for transfusions in sickle cell disease: a review of a 14-year experience from a single center (CME). Transfusion 2011;51:1732-9. [Crossref] [PubMed]
  105. Chou ST, Evans P, Vege S, et al. RH genotype matching for transfusion support in sickle cell disease. Blood 2018;132:1198-207. [Crossref] [PubMed]
  106. Aygun B, Padmanabhan S, Paley C, et al. Clinical significance of RBC alloantibodies and autoantibodies in sickle cell patients who received transfusions. Transfusion 2002;42:37-43. [Crossref] [PubMed]
  107. Wood JC. Use of magnetic resonance imaging to monitor iron overload. Hematol Oncol Clin North Am 2014;28:747-64. vii. [Crossref] [PubMed]
  108. Wood JC. The use of MRI to monitor iron overload in SCD. Blood Cells Mol Dis 2017;67:120-5. [Crossref] [PubMed]
  109. Ganz T. Systemic iron homeostasis. Physiol Rev 2013;93:1721-41. [Crossref] [PubMed]
  110. Hankins JS, Smeltzer MP, McCarville MB, et al. Patterns of liver iron accumulation in patients with sickle cell disease and thalassemia with iron overload. Eur J Haematol 2010;85:51-7. [PubMed]
  111. Wang CY, Jenkitkasemwong S, Duarte S, et al. ZIP8 is an iron and zinc transporter whose cell-surface expression is up-regulated by cellular iron loading. J Biol Chem 2012;287:34032-43. [Crossref] [PubMed]
  112. Xu YY, Wan WP, Zhao S, et al. L-type Calcium Channels are Involved in Iron-induced Neurotoxicity in Primary Cultured Ventral Mesencephalon Neurons of Rats. Neurosci Bull 2020;36:165-73. [Crossref] [PubMed]
  113. Coates TD, Wood JC. How we manage iron overload in sickle cell patients. Br J Haematol 2017;177:703-16. [Crossref] [PubMed]
  114. Poggiali E, Cassinerio E, Zanaboni L, et al. An update on iron chelation therapy. Blood Transfus 2012;10:411-22. [PubMed]
  115. Olivieri NF. Progression of iron overload in sickle cell disease. Semin Hematol 2001;38:57-62. [Crossref] [PubMed]
  116. Vichinsky E, Onyekwere O, Porter J, et al. A randomised comparison of deferasirox versus deferoxamine for the treatment of transfusional iron overload in sickle cell disease. Br J Haematol 2007;136:501-8. [Crossref] [PubMed]
  117. Win N. Hyperhemolysis syndrome in sickle cell disease. Expert Rev Hematol 2009;2:111-5. [Crossref] [PubMed]
  118. Banks M, Shikle J. Hyperhemolysis Syndrome in Patients With Sickle Cell Disease. Arch Pathol Lab Med 2018;142:1425-7. [Crossref] [PubMed]
  119. King KE, Shirey RS, Lankiewicz MW, et al. Delayed hemolytic transfusion reactions in sickle cell disease: simultaneous destruction of recipients' red cells. Transfusion 1997;37:376-81. [Crossref] [PubMed]
  120. Petz LD. Bystander immune cytolysis. Transfus Med Rev 2006;20:110-40. [Crossref] [PubMed]
  121. Friedman DF, Kim HC, Manno CS. Hyperhaemolysis associated with red cell transfusion in sickle cell disease. Transfusion 1993;33:148.
  122. Scheunemann LP, Ataga KI. Delayed hemolytic transfusion reaction in sickle cell disease. Am J Med Sci 2010;339:266-9. [Crossref] [PubMed]
  123. Liu Y, Pal M, Bao W, et al. Type I interferon is induced by hemolysis and drives antibody-mediated erythrophagocytosis in sickle cell disease. Blood 2021;138:1162-71. [Crossref] [PubMed]
  124. Belcher JD, Marker PH, Weber JP, et al. Activated monocytes in sickle cell disease: potential role in the activation of vascular endothelium and vaso-occlusion. Blood 2000;96:2451-9. [Crossref] [PubMed]
  125. Chonat S, Mener A, Verkerke H, et al. Role of complement in alloimmunization and hyperhemolysis. Curr Opin Hematol 2020;27:406-14. [Crossref] [PubMed]
  126. Schwartz RS, Tanaka Y, Fidler IJ, et al. Increased adherence of sickled and phosphatidylserine-enriched human erythrocytes to cultured human peripheral blood monocytes. J Clin Invest 1985;75:1965-72. [Crossref] [PubMed]
  127. Win N, Doughty H, Telfer P, et al. Hyperhemolytic transfusion reaction in sickle cell disease. Transfusion 2001;41:323-8. [Crossref] [PubMed]
  128. Petz LD, Calhoun L, Shulman IA, et al. The sickle cell hemolytic transfusion reaction syndrome. Transfusion 1997;37:382-92. [Crossref] [PubMed]
  129. Boonyasampant M, Weitz IC, Kay B, et al. Life-threatening delayed hyperhemolytic transfusion reaction in a patient with sickle cell disease: effective treatment with eculizumab followed by rituximab. Transfusion 2015;55:2398-403. [Crossref] [PubMed]
  130. Pirenne F, Bartolucci P, Habibi A. Management of delayed hemolytic transfusion reaction in sickle cell disease: Prevention, diagnosis, treatment. Transfus Clin Biol 2017;24:227-31. [Crossref] [PubMed]
  131. Unnikrishnan A, Pelletier JPR, Bari S, et al. Anti-N and anti-Doa immunoglobulin G alloantibody-mediated delayed hemolytic transfusion reaction with hyperhemolysis in sickle cell disease treated with eculizumab and HBOC-201: case report and review of the literature. Transfusion 2019;59:1907-10. [Crossref] [PubMed]
  132. Vlachaki E, Gavriilaki E, Kafantari K, et al. Successful Outcome of Hyperhemolysis in Sickle Cell Disease following Multiple Lines of Treatment: The Role of Complement Inhibition. Hemoglobin 2018;42:339-41. [Crossref] [PubMed]
  133. Lee LE, Beeler BW, Graham BC, et al. Posttransfusion hyperhemolysis is arrested by targeting macrophage activation with novel use of Tocilizumab. Transfusion 2020;60:30-5. [Crossref] [PubMed]
doi: 10.21037/aob-21-67
Cite this article as: Tanhehco YC, Shi PA, Schwartz J. Transfusion therapy in sickle cell disease. Ann Blood 2022;7:9.

Download Citation