- 1Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
- 2Facultad de Medicina, Universidad Francisco de Vitoria, Madrid, Spain
- 3Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University., Olomouc, Czechia
- 4The Czech Advanced Technology and Research Institute (Catrin), Palacky University, Olomouc, Czechia
This review critically examines the latest research on the intersection of sickle cell disease (SCD) and COVID-19, highlighting meaningful immunopathological interactions. It reveals significant knowledge gaps, particularly in the analysis of inflammatory markers in clinical studies and the oversight of SCD in malaria-COVID-19 research conducted in African contexts. The investigation explores shared pathophysiological mechanisms, including the cytokine storm phenomenon, vascular complications, and autoimmune responses, which exemplify the perilous interplay between SCD’s chronic inflammatory state and the acute inflammatory response triggered by COVID-19. Furthermore, it provides a comprehensive analysis of hyperhaemolysis syndrome (HHS), related autoimmune conditions, and avascular necrosis (AVN) as critical complications affecting SCD patients during the COVID-19 pandemic. The aim is to establish a comprehensive framework for understanding this crucial intersection while proposing vital directions for future research and therapeutic interventions.
1 Introduction
SCD is characterised by chronic inflammation, endothelial dysfunction, and an increased thrombotic risk, which can be exacerbated by the inflammatory and prothrombotic effects of SARS-CoV-2 infection (1).
Concerns about severe outcomes in this patient population spurred quick research and surveillance efforts globally. Early findings from France, particularly those by Arlet et al. (2), challenged the assumption of uniformly poor outcomes, documenting considerable variability in disease severity and highlighting the importance of early intervention. Azerad et al. (3) reported several atypical presentations with surprisingly favourable outcomes, emphasising the complexity of this clinical intersection and the need for individualised assessment and management approaches. The paper examined the complex pathophysiological environment affecting multiple organ systems—from acute chest syndrome in paediatric patients (4) requiring significant adaptation of traditional management algorithms, to severe hepatic dysfunction leading to encephalopathy and multi-organ failure (5), and catastrophic neurological events requiring heightened surveillance and prompt intervention (6). A detailed analysis of susceptibility and clinical severity patterns by Sayad et al. (7) reveals complex interactions among genetic factors, environmental influences, and therapeutic interventions. This suggests that while some patients demonstrate remarkable resilience, others face significantly increased risks of severe complications. Special attention is given to vulnerable populations, particularly pregnant women with SCD during the pandemic (8–10). Joseph et al. (8) documented elevated maternal and fetal morbidity rates requiring modified obstetric and haematological management approaches, findings further supported by Gibson et al. (9) and Kolanska et al. (10), emphasising the need for enhanced monitoring and specialised care protocols. Current treatment approaches assess established SCD protocols alongside emerging COVID-19 therapies, highlighting red blood cell exchange transfusion as essential for severe respiratory issues (11), the potential role of disease-modifying therapies such as hydroxyurea (12), and the implementation of standardised screening and management protocols to guide decision-making in various clinical scenarios (13–15).
In an extensive French study, children with SCD had rates of COVID-19 exposure that were similar to those of their healthy peers (13). However, these children did not experience severe complications, which suggests that activation of the basal interferon type I (IFN-I) pathway may help control the virus (13, 14). The humoral immune response in these children appears to be temporary, raising the risk of reinfection (15). The adverse outcomes reported in other studies may be related to disparities in healthcare access or underlying comorbidities, rather than solely due to SCD. Martin et al. (16) explored the unique challenges faced by paediatric populations and analysed mortality patterns, emphasising the influence of healthcare access and socioeconomic factors on survival (16). Additionally, the research addresses autoimmune interactions—including SCD’s association with systemic inflammatory diseases, autoimmune haemolytic anaemia, and immune thrombocytopenia—and devastating musculoskeletal complications affecting 50-70% of patients (17), particularly avascular necrosis of the femoral head, where inflammation and vaso-occlusion create a cycle of tissue damage (16, 17). This paper highlights the evolving SARS-CoV-2 variants and SCD pathophysiology, stressing the need for ongoing vigilance and adaptable management strategies (17–19). Emerging evidence indicates that long-term complications require specialised follow-up protocols and rehabilitation strategies to optimise outcomes (18–20). This review focuses on the underlying mechanisms of the pathology, revises the gaps in global health literature and provides an overview of management strategies.
2 Pathophysiological foundation of sickle cell disease
2.1 Molecular and cellular mechanisms
In 1910, in Chicago, Dr James B. Herrick officially described the first clinical case of what would later become known as sickle cell disease (SCD) (21). SCD was the first molecularly characterised disorder. It is defined by a specific mutation in the beta-globin gene at the sixth codon, which leads to the production of abnormal haemoglobin S (HbS) in place of the normal haemoglobin A (HbA) (22, 23). More than 7.5 million people worldwide are affected (24).
When deoxygenated, HbS undergoes polymerisation, resulting in the distortion of erythrocytes into a rigid, sickle-shaped form. This alteration in the morphology and functionality of erythrocytes contributes to several clinical manifestations associated with the disease (25):
1. Vaso-occlusion from rigid cells obstructing microvasculature.
2. Chronic haemolysis due to the premature destruction of sickled erythrocytes.
3. Endothelial activation and damage from repeated exposure to sickled cells
4. Chronic inflammation from ongoing tissue injury and cell damage
The abnormal features of sickle red blood cells (RBCs), which disrupt patients’ blood rheology, are responsible for chronic haemolytic anaemia and frequent painful vaso-occlusive crises (VOCs).
2.1.1 Epidemiological context and confounding factors
SCD is most prevalent in malaria-endemic regions, particularly sub-Saharan Africa, where up to 2% of births are affected, as well as in the Mediterranean, the Middle East, and parts of India (24). These areas have unique epidemiological characteristics that influence COVID-19 patterns and complicate the relationship between malaria, SCD, and COVID-19 (24). SCD-endemic regions are primarily lower and middle-income countries with younger populations, lower obesity rates, and housing that is more crowded yet better ventilated than in high-income areas. These factors can affect COVID-19 transmission and severity, potentially obscuring the impact of SCD on COVID-19 susceptibility. However, Dawudi et al. reported, in a meta-analysis, an increased risk of hospitalisation and death in SCD patients (26).
Concurrent malaria exposure may alter immune responses and influence both SCD and COVID-19 outcomes due to shared inflammatory pathways. Limitations in healthcare infrastructure further complicate the management of SCD and the detection of COVID-19, leading to surveillance bias. This highlights the need for carefully designed studies that consider regional epidemiological contexts when examining interactions between SCD and COVID-19, especially in comparative analyses across different socioeconomic backgrounds.
2.2 Inflammatory state in SCD
Even in steady state, patients with SCD exist in a chronic proinflammatory state characterised by elevated levels of pro-inflammatory cytokines, including IL-1β, IL-6, IL-8, IL-33, and TNF-α (25). This inflammatory milieu is further intensified by dysregulation of anti-inflammatory cytokines, including IL-2, TGF-β, IL-10, IL-27, IL-35, and IL-37 (27). The chronic inflammatory state is characterised by increased expression of adhesion molecules (28) and neutrophil activation, as well as the formation of neutrophil extracellular traps (NETs) (29–31). Additionally, persistent endothelial activation (32) and platelet activation (33, 34) contribute to the hypercoagulable state (35) that characterises SCD. The proinflammatory state facilitates red blood cell alloimmunization in paediatric SCD patients (36).
Chronic inflammation reshapes the hematopoietic landscape, particularly within the bone marrow, affecting monocyte programming, increasing cytokine production, and promoting myeloid skewing. This understanding is essential for grasping why SCD patients are more susceptible to COVID-19-related complications (37). Ferroptosis also plays a crucial role in sickle cell disease, as free iron is essential for the formation of reactive oxygen species (38).
2.3 Pro-Inflammatory mechanisms in SCD
Evidence suggests persistent inflammation is central to SCD pathophysiology, exacerbating endothelial dysfunction, vascular occlusion, and multi-organ failure (39). The pro-inflammatory essence of SCD can be understood through several key mechanistic pathways:
2.3.1 Oxidative stress and endothelial activation
The pathophysiological cascade in SCD is driven by recurrent cycles of hypoxia-reoxygenation, leading to the excessive production of reactive oxygen species (ROS) (40). Oxidative stress triggers endothelial activation, promoting the expression of adhesion molecules such as VCAM-1, ICAM-1, E-selectin, and P-selectin, which facilitate the adhesion of leukocytes and platelets to the vascular endothelium (16, 32). Elevated endothelial-derived extracellular vesicles also contribute to a hypercoagulable state, exacerbating vaso-occlusion (32–34).
2.3.2 Neutrophil activation and NET formation
Neutrophils are major effectors of SCD-associated inflammation. Circulating iron and cell-free haem released during haemolysis activate neutrophils, inducing the formation of NETs, which amplify endothelial damage and vaso-occlusion (41, 42). Targeting NET formation has emerged as a potential therapeutic strategy to mitigate inflammatory exacerbation in SCD (43).
Argueta et al. (44) recently reported the formation of mast extracellular traps in SCD, suggesting that tissue mast cells may also play a role in the disease’s physiopathology.
2.3.3 Monocyte dysfunction and cytokine dysregulation
Aberrant monocyte activation plays a central role in SCD inflammation (45–47). Studies have reported an increased prevalence of immature monocyte subsets expressing pro-inflammatory markers such as CD14+CD16- and CD14+HLA-DR- phenotypes associated with impaired immune responses (27, 45–47). These monocytes secrete elevated levels of TNF-α, IL-6, and IL-1β, thereby perpetuating systemic inflammation (46, 47) through the TLR4 pathway (48). The higher expression of TLR4 in SCD patients was inferred to be responsible for the increased secretion of IL-12, IL-17, and IL-10 cytokines, as well as for the increased number of circulating activated CD4+CD19+ lymphocytes (49).
As Felicio et al. (50) demonstrated, B lymphocytes are also crucial in the physiopathology of SCD. B-cell alterations in the memory subpopulation appear to be critical in this process.
T regulatory cells were shown to be increased in paediatric patients with SCD, and treatment with hydroxyurea was able to decrease the amount of T reg (51). Additionally, CD8 and CD8+CD45RA cells were reduced compared to the controls (52). The decrease in CD8 could reduce SARS-CoV-2 clearance in these patients. T cells can play a crucial role in modulating B1 cells, suggesting that the antibody response to viral infection is affected in SCD (53). Moreover, Marchesani et al. (54) reported, in paediatric patients, lower values of CD4+ T cells (with a higher number of memory and central memory T lymphocytes) with increased frequency of CD8+ T cells (with a predominant naive pattern) and, higher values of CD39+ Tregs and lower HLA-DR+ and CD39- T cells, along with an increased Th17, Th1-17, and Th2 response.
2.3.4 Haeme-mediated inflammation and MicroRNA regulation
Free haem is a potent damage-associated molecular pattern (DAMP) that exacerbates inflammation by upregulating NF-κB and TLR4 signalling pathways. The modulation of haem-induced inflammation through microRNA-loaded liposomes has shown promise in experimental models, with miR-451a and let-7i-5p reducing inflammatory responses and promoting haem clearance via haemopexin and HO-1 (55). In addition, a phase 2 open-label trial with food rich in docosahexaenoic acid has shown promising results in SCD (56).
3 COVID-19 pathophysiology and immune response
3.1 Virology and Cellular Entry
The SARS-CoV-2 virus, first identified in December 2019 in Wuhan, China, primarily enters host cells through the angiotensin-converting enzyme 2 (ACE2) receptor. This tropism for ACE2 is significant as this receptor plays a crucial role in regulating inflammatory responses through the bradykinin signalling pathway. ACE2 downregulation during infection can lead to dysregulated inflammation (57, 58).
3.1.1 Viral evolution and contemporary relevance
The literature reviewed primarily discusses earlier SARS-CoV-2 variants, particularly the original Wuhan strain, Alpha, and Delta (15–18, 25, 26). These variants exhibited greater virulence than the Omicron subvariants (59). This raises important questions about the relevance of findings for SCD patients and ongoing risk assessment. Despite Omicron’s generally reduced virulence in healthy individuals, SCD patients may still face an increased risk of vaso-occlusive crises (VOCs) (60). Their baseline inflammatory conditions and immune dysregulation could exacerbate even mild viral infections. Chronic organ damage and functional asplenia may go beyond variant-specific severity.
Vaccination also complicates the picture; while vaccines help reduce severe disease, SCD patients may have weaker immune responses due to underlying conditions (59) or a lower vaccination rate (60). Most reports have been based on mRNA vaccines, rather than using attenuated or protein vaccines (59). The risk of complications, such as haemolytic-uremic syndrome, persists due to immune evasion by variants and waning immunity. However, antiviral therapy may be a good option for these patients (61).
Mechanistic pathways such as cytokine dysregulation and thrombo-inflammatory responses remain relevant across variants, necessitating a focus on SCD-specific complications alongside acute COVID-19 outcomes in future surveillance.
3.2 Inflammatory cascade in COVID-19
COVID-19 initiates a complex inflammatory cascade characterised by an early innate immune response, where the infection of epithelial cells in the upper respiratory tract triggers mucus production, antiviral metabolites, type I interferons, and other cytokines (55). In severe cases, a dysregulated immune response produces excessive proinflammatory cytokines, including IL-6, IL-1β, IL-8, and TNF-α, along with various chemokines, resulting in widespread inflammation and tissue damage known as cytokine storm (57, 58, 62–68). IL-6 plays a critical role in the response to bacterial infection in SCD (64–67), while IL-8 is increased in patients with SCD (57, 58, 62, 68).
SARS-CoV-2 infection directly and indirectly damages the vascular endothelium, leading to coagulation abnormalities, microvascular thrombosis, and increased vascular permeability (57, 58). The inflammatory cascade extends beyond the respiratory system, affecting multiple organs and systems, including the cardiovascular, neurological, renal, and haematological systems (57, 58, 69, 70). This pathophysiological progression aligns dangerously with the pre-existing inflammatory state in SCD, setting the stage for potentially catastrophic interactions (71–74).
4 Epistemic zones in SCD-COVID-19 research
4.1 Mechanistic insight: foundational understanding
Several key studies provide mechanistic frameworks essential for understanding the SCD-COVID-19 interaction:
Swirski et al. (37) outline a fundamental framework for understanding the increased susceptibility of patients with SCD to complications associated with COVID-19. Their research elucidates how chronic inflammation modifies the haematopoietic landscape, particularly within the bone marrow niche, which subsequently alters monocyte programming and leads to increased cytokine production and myeloid skewing. Within the context of COVID-19, this framework assumes critical importance, as patients with SCD typically exist in a state of immunological activation.
Saxena and Muthu (72) explored the intersection of haematological disruptions and immune dysregulation during COVID-19 relevant to SCD. Dobkin et al. (73) significantly re-examined the evolutionary and immunological effects of red blood cell adaptations to malaria, particularly regarding how these adaptations influence the host response to critical illnesses, such as COVID-19. The authors argue that adaptations such as haemoglobin S, alpha-thalassemia, G6PD deficiency, and Duffy antigen mutations have remapped erythroid immunology across entire populations (73). These report offer a molecular and evolutionary justification for integrating SCD and malaria into the analysis of COVID-19 pathology
4.2 Descriptive silence: the missing mechanisms
Most clinical studies on SCD and COVID-19 demonstrate significant limitations, including the general documentation of clinical presentations and outcomes without analysis of underlying mechanisms, as well as the absence of measurement of inflammatory markers. Several studies observe but do not question. They document presence without meaning, reflecting a recurring epistemic pattern of describing symptoms in inflammatory diseases without addressing inflammation. The patients’ physiology is treated as a black box—observed only from the outside (74). Several registry-based studies document hospitalisations, oxygen requirements, and VOC in SCD patients with COVID-19 but avoid interpreting these outcomes in biological terms. This represents a missed opportunity to understand the pathophysiological interaction between these conditions.
4.3 Africa without malaria or SCD: a geographic and conceptual omission
Perhaps the most revealing epistemic gap is found in studies conducted in Africa that examine COVID-19 and malaria without considering SCD:
Woodford et al. (75) present an extensive longitudinal cohort study in Mali evaluating whether recent Plasmodium falciparum infection affects susceptibility to COVID-19 or alters clinical outcomes. However, nowhere in this comprehensive study—conducted in a region with high prevalence of SCD and sickle cell trait—is there any mention of “sickle,” “haemoglobinopathy,” or “SCD.” This reveals a significant epistemic flaw: sickle cell disease is hidden in vague categories, dismissed as analytically irrelevant, or overlooked in research.
Achan et al. (76) conducted a multicenter, prospective cohort study in Uganda to investigate the effect of recent malaria exposure on COVID-19 outcomes. Among the 597 hospitalised COVID-19 patients studied, they observed elevated inflammatory markers associated with poorer outcomes. Nevertheless, there is a total absence of any mention of SCD, despite Uganda having a significant prevalence of sickle cell trait exceeding 13% in many regions. Excluding SCD from COVID-19 research in areas with high prevalence and known interactions with malaria is not just a data gap, but an epistemic erasure with clinical implications.
Feit et al. (77) conducted a methodologically robust 2.5-year longitudinal study involving 178 patients diagnosed with COVID-19 and 356 matched controls. Similarly, Bodla et al. (78) analysed 66,415 adult patients hospitalised for sickle cell crises, documenting an adjusted odds ratio of 8.49 for in-hospital mortality with concurrent COVID-19 infection. Despite the fact that the studies had a broad temporal scope, sample size, and good statistical analysis, their design limited the ability to explore mechanistic insights, evaluate inflammatory biomarkers, or generate hypotheses about pathophysiological interactions. Thus, the resulting dataset offers limited guidance for clinical intervention or theoretical advancement.
5 Immunopathogenesis in SCD-COVID-19 Interaction
5.1 Synergistic inflammatory cascades
The interaction between SCD and COVID-19 culminates in a perilous synergy within inflammatory processes that extends beyond the baseline inflammatory priming present in SCD patients. Upon exposure to SARS-CoV-2, this already activated system elicits an exaggerated release of inflammatory mediators (57, 58). The onset of COVID-19 in SCD patients may trigger increased haemolysis, releasing additional free haem that further stimulates inflammatory pathways (79). This interconnected relationship highlights the significantly increased mortality risk seen in patients with both conditions. COVID-19 and splenic dysfunction alter red blood cell behaviour and vesiculation, thereby impairing oxygen delivery and increasing the risk of thrombosis (80). B cells play a pivotal role in both the pathophysiology of COVID-19 (57, 58) and exhibit enhanced activity in SCD (48, 78), warranting further investigation of this intersection.
SCD and COVID-19 damage the endothelium through distinct yet complementary mechanisms, which may result in catastrophic vascular dysfunction (81–84). This interconnected relationship highlights the significantly increased mortality risk seen in patients who have both conditions. One study shows that parvovirus B19-induced aplastic anaemia in SCD worsens immune dysregulation, raising concerns about chronic inflammation. Another study highlights that COVID-19 and splenic dysfunction alter the behaviour and vesiculation of red blood cells, impairing oxygen delivery and increasing the risk of thrombosis (80, 82).
Additionally, B cells play a pivotal role in the pathophysiology of COVID-19 (57, 58) and can exhibit enhanced activity in SCD, as previously documented by Felicio et al. (50) and Dhawan (83). Further exploration in this area is essential to uncover vital insights.
5.2 Historical and current perspectives on cytokine dysregulation
Understanding the involvement of cytokines in SCD has evolved significantly over the past three decades. Early work by Chappey et al. (84) established the first experimental frameworks using endothelial cell cultures to study vascular dysfunction in SCD, demonstrating how cytokines such as TNF-α and IL-1 can activate endothelial cells and influence the expression of adhesion molecules. A breakthrough came in 1999 when Croizat and Nagel (85) identified distinct cytokine response patterns related to HbF levels, finding that patients with HbF levels below 8-9% showed different cytokine profiles compared to those with higher HbF levels, particularly in terms of SCF and TGF-β concentrations.
The role of TGF-β1 has been further elucidated by Santiago et al. (86), who demonstrated that its plasma levels are significantly elevated in HbSS patients compared to HbSC and healthy controls. This cytokine correlates with monocyte activation, leukocyte counts, and inflammatory mediators such as TIMP-1 and MMP-9, providing mechanistic insights into the immunopathological heterogeneity of SCD and its association with vascular dysfunction (86). These findings suggest that TGF-β1 contributes to vascular remodelling, vasculopathy, angiogenesis, and inflammatory processes in SCD patients, which may become particularly relevant in SARS-CoV-2 infection.
Early advances in understanding cytokine functions in SCD emerged in the early 2000s. Schnog et al. (87) investigated sVCAM-1 levels in adults with SCD, establishing connections between adhesion molecules and disease severity. Assis et al. (88) demonstrated how specific cytokines influenced sickle neutrophil adhesion to fibronectin, whilst Brittain and Parise’s (89) review consolidated the growing knowledge about plasma factors and cytokines in SCD, highlighting the emerging role of platelets in cytokine release and the importance of angiogenic and inflammatory factors.
Modern understanding of cytokine involvement in SCD has benefited from advanced technical capabilities. Paulson et al. (90) introduced new perspectives on stress erythropoiesis and its relationship with cytokine signalling. Recent studies have identified the crucial role of inflammatory dendritic cells in regulating immune responses in SCD. Sesti-Costa et al. (91) demonstrated that inflammatory dendritic cells (CD14+ DCs) are key contributors to immune dysregulation in SCD, producing the cytokines IL-6, IL-8, and IL-17, which are critically involved in both SCD and severe COVID-19 cases. Their study suggests that an upregulated haeme oxygenase (HO-1) pathway in monocytes promotes the differentiation of these inflammatory dendritic cells (DCs), potentially exacerbating endothelial inflammation and thrombo-inflammatory complications in SCD patients infected with SARS-CoV-2 (91).
The cytokine interplay between SCD and COVID-19 involves several shared pathways. Chiang et al. (71) described the shared inflammatory mechanisms of thrombo-inflammation in SCD and COVID-19, particularly in the activation of the coagulation cascade, endothelial damage, and platelet-leukocyte adhesion. The role of thromboxane A2 (TxA2) and lipid mediators in both diseases is emphasised as a potential therapeutic target, with platelet-derived microvesicles potentially exacerbating both vaso-occlusive crises in SCD and endothelial injury in COVID-19 (71).
Emerging evidence suggests that basal interferon-1 activation in SCD may confer a protective advantage against severe COVID-19 manifestations (92–94). Interferon signalling in SCD patients modulates the immune response, potentially mitigating the full impact of a SARS-CoV-2-induced cytokine storm (94). In contrast to previous assumptions that SCD patients have a heightened vulnerability to COVID-19, some studies report that specific interferon-related mechanisms may confer protective effects, which is counterbalanced by exacerbated thrombo-inflammatory responses (94, 95).
Several cytokine-targeting therapies have shown promise in both SCD and COVID-19, including tocilizumab (an IL-6 inhibitor), fostamatinib (an SYK inhibitor), regadenoson (an A2A receptor agonist), and vitamin D modulation (96–99). Additionally, hydroxyurea’s established anti-inflammatory and cytoprotective effects in SCD may also potentially mitigate inflammation in COVID-19 (95). Targeting the TGF-β1 and TIMP-1/MMP-9 axis could enhance therapeutic strategies to reduce cytokine-driven complications in both conditions (86, 89).
The role of platelet activation in the pathophysiology of both SCD and COVID-19 has led to increasing interest in antiplatelet therapies as potential disease-modifying agents. Martí-Carvajal et al. (100) conducted a systematic review assessing the clinical benefits and risks of antiplatelet agents in preventing VOC events in SCD. Their analysis of five randomised controlled trials involving 747 participants examined the effects of prasugrel, ticagrelor, crizanlizumab, and aspirin (100). However, due to the high risk of bias and the heterogeneity of the study populations, the authors concluded that there is insufficient evidence to support or refute the routine use of antiplatelet agents in preventing VOCs.
Future research should focus on cytokine-based biomarkers for risk stratification and the development of targeted therapies. Advances in genetic susceptibility studies have shed light on the role of cytokine polymorphisms in determining the severity of both SCD and COVID-19. Additionally, novel anti-inflammatory adenosine pathway modulators warrant further investigation as therapeutic candidates for managing the cytokine-mediated complications that characterise the intersection of these two conditions.
5.3 Resolution of inflammation: role of specialised pro-resolving mediators
Recent research underscores the protective effects of 17R-Resolvin-D1 (17R-RvD1) in alleviating cardiovascular dysfunction associated with SCD. 17R-RvD1 has diminished neutrophil-mediated cardiac hypertrophy and fibrosis by modulating the NF-κBp65 and TGF-β1/Smad2–3 signaling pathways (101). This finding supports the therapeutic potential of pro-resolving lipid mediators in managing SCD, which may be especially pertinent in hyperinflammation induced by COVID-19 (101, 102).
5.4 Anti-inflammatory cytokine modulation
The existing imbalance between pro-inflammatory and anti-inflammatory cytokines SCD has generated considerable interest in targeting pathways that enhance the activity of IL-10, TGF-β, and IL-27 (30, 103). Recent studies suggest that dietary interventions, particularly omega-3 fatty acids and vitamin D supplementation, may help mitigate inflammation and reduce the frequency of VOCs (30, 57, 103–105). These strategies may also alleviate the cytokine storm associated with severe COVID-19 in patients with SCD.
5.5 Redox-based interventions and antioxidant therapy
Nitro-fatty acids have shown promise in preclinical models, exerting anti-inflammatory effects by modulating transcription factors and reducing endothelial dysfunction (106). Antioxidant therapies targeting ROS-induced vascular injury may hold potential as adjunctive treatments for SCD patients with COVID-19, particularly given the oxidative stress that characterises both conditions (107, 108).
5.6 Ferroptosis at the crossroads of sickle cell disease and COVID-19
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a crucial player in the pathophysiology of SCD and may represent a significant mechanistic link to COVID-19 complications (109). Characterised by chronic haemolysis and iron dysregulation, SCD creates an environment conducive to ferroptotic damage. Excess haeme promotes ferroptosis via haeme oxygenase-1 upregulation, exacerbating cardiac complications in SCD patients (109). Moreover, ferroptosis-driven lipid peroxidation contributes to endothelial dysfunction and systemic inflammation, highlighting its significance as a potential therapeutic target (109–114).
The intersection of SCD and COVID-19 may magnify ferroptotic stress through multiple converging pathways (109–114). First, chronic haemolysis in SCD releases free haem, which upregulates haem oxygenase-1 (HMOX1), a driver of iron release and subsequent ferroptosis (110). Second, SARS-CoV-2-induced hyperinflammation further perturbs iron homeostasis, increasing ferroptotic susceptibility (78). Third, endothelial dysfunction, a shared feature of both conditions, may be exacerbated by ferroptotic endothelial cell death, contributing to vasculopathy, thrombosis, and multiorgan failure (109, 111–114).
Recent evidence suggests that the transsulfuration (TSS) pathway is a primary defence mechanism against ferroptotic stress in SCD by maintaining cysteine availability for glutathione biosynthesis (115). Cystathionine beta-synthase (CBS) activation via NRF2-mediated epigenetic modifications significantly enhances cellular antioxidant capacity, reducing ferroptotic damage in erythroblasts (115). This proposed mechanism influences chromatin architecture and ferroptosis susceptibility, with findings indicating that NRF2 regulates L-2-hydroxyglutarate (L2HG) metabolism. Koklesova et al. (116) identified homocysteine as a marker for tailored treatment in SARS-CoV-2 infection.
Beyond erythrocyte dysfunction, ferroptosis plays a key role in organ pathology in SCD. GBT1118, a voxelotor analogue, mitigates sickle hepatopathy by reducing iron overload, inflammation, and fibrosis, thereby limiting ferroptotic activity (117). Similarly, leonurine, a natural alkaloid, protects against iron-induced hepatotoxicity by upregulating NRF2 and suppressing NF-κB-mediated inflammation, reinforcing the interplay between antioxidant defence mechanisms and ferroptosis regulation (117, 118).
Given the central role of ferroptosis in SCD-COVID-19 pathophysiology, targeted interventions to mitigate ferroptotic stress may offer therapeutic benefit. Ferroptosis inhibitors such as ferrostatins, liproxstatins, and iron chelators (e.g., deferoxamine) have shown promise in preclinical models (119–121). Additionally, upregulation of GPX4 via pharmacological agents or nutritional strategies (e.g., selenium supplementation (122) may counteract ferroptotic damage (119–121). Emerging gene therapy approaches targeting ferroptosis-associated pathways could provide novel avenues for intervention in this high-risk population (119–121).
This mechanistic insight underscores the urgency of integrating ferroptosis-targeted therapies into clinical strategies for managing SCD patients infected with SARS-CoV-2 (119–123). Although this study focuses on COVID-19, its findings on ferroptosis may have translational relevance for SCD, particularly in advancing understanding of the shared mechanisms of inflammation and oxidative stress in both conditions, and in exploring novel combined therapeutic strategies (119–123).
Future research must prioritise translational studies to validate these findings and develop effective interventions to mitigate ferroptosis-driven complications in this vulnerable population.
5.7 Vascular dysfunction and coagulopathy
The vascular endothelium represents the critical interface between SCD and COVID-19 pathophysiology. In SCD, chronic exposure to sickled erythrocytes, free haem, and inflammatory cytokines generates endothelial dysfunction through recurrent cycles of hypoxia-reoxygenation, leading to excessive production of ROS (39). This oxidative stress triggers endothelial activation, promoting the expression of adhesion molecules such as VCAM-1, ICAM-1, E-selectin, and P-selectin, which facilitate the adhesion of leukocytes and platelets to the vascular endothelium (16, 32). Elevated endothelial-derived extracellular vesicles contribute to a hypercoagulable state, exacerbating vaso-occlusion (32–34). COVID-19 exacerbates this dysfunction by directly infecting endothelial cells through ACE2 receptors and triggering widespread endothelial injury via inflammatory mediators, complement activation, and NETs (57). Neutrophils, activated by circulating iron and cell-free haem released during haemolysis, form NETs that amplify endothelial damage and vaso-occlusion (41, 42).
High-mobility group box 1 (HMGB1) regulates inflammation and promotes platelet activation via Toll-like receptor 4 (TLR4) (124). HMGB1-induced platelet activation involves ADP-dependent P2Y12 (purine receptor) signalling, and HMGB1 sensitises platelets for ADP signalling (125). This synergistic interaction between ADP and HMGB1 enhances the understanding of thromboinflammatory signalling in SCD and provides insights for therapeutic P2Y12 inhibition (124, 125).
5.8 Pulmonary vulnerability
The lungs represent a particular point of vulnerability in the SCD-COVID-19 interaction:
Acute chest syndrome (ACS) is already a leading cause of mortality in SCD, characterised by pulmonary infiltrates, hypoxemia, and respiratory distress. The pathophysiology of ACS involves sickling within the pulmonary vasculature, fat embolism resulting from bone marrow infarction, and inflammation triggered by infection (126, 127).
Asthma, a common comorbidity, affects up to 48% of SCD patients, exacerbating risks of acute chest syndrome, stroke, and VOCs. Its coexistence reflects complex inflammatory interactions, with studies showing altered cytokine profiles (e.g., elevated IL-6, IL-8) and heightened airway reactivity (126, 127). Recognising this interplay is critical for multidisciplinary management and improved outcomes in affected individuals.
COVID-19 primarily affects the lungs through direct viral infection of pneumocytes, inflammation-induced alveolar damage, and microvascular thrombosis (57, 58).
The clinical presentations of these conditions can overlap, making diagnosis and management more complicated. More importantly, the underlying mechanisms can synergise, with COVID-19 potentially triggering and severely exacerbating ACS through:
Hypoxemia that triggers sickling; 2) Inflammatory activation of adhesion molecules; 3) Endothelial injury; 4) Hypercoagulability, and 5) Cytokine storm.
This interaction may explain the significantly increased mortality observed in co-affected patients (57, 58).
5.9 Hyperhaemolysis syndrome: a critical complication in the context of COVID-19
Hyperhaemolysis syndrome (HHS) in SCD is a significant example of immune-mediated haemolysis, where both transfused and autologous red blood cells (RBCs) are rapidly destroyed, leading to severe anaemia. The emergence of SARS-CoV-2 may exacerbate the chronic pro-inflammatory state in SCD, thereby increasing the severity of hyperhaemolysis episodes.
5.9.1 Considerations and diagnostic variability
It is worth noting that inconsistencies in the diagnostic criteria for HHS across the literature significantly impact the interpretation of findings (128–133). Some studies apply stringent criteria requiring a post-transfusion haemoglobin concentration lower than the pre-transfusion level with evidence of donor and recipient red cell destruction. In contrast, others employ more lenient definitions (130, 131). This diagnostic heterogeneity potentially explains some of the variability in reported therapeutic responses and complicates efforts to establish whether COVID-19-associated HHS constitutes a distinct clinical entity.
The current evidence base is insufficient to delineate specific subtypes of COVID-19-associated HHS versus traditional HHS in SCD. Cases reported demonstrate overlapping and distinctive characteristics; however, larger cohorts with detailed immunological characterisation would be necessary to justify a distinct classification system.
5.9.2 Immunopathological mechanisms in COVID-19-associated HHS
5.9.2.1 Complement dysregulation
The role of complement dysregulation in COVID-19 and long COVID-19 is particularly striking (132, 133). Studies demonstrate that SARS-CoV-2 triggers the activation of the alternative pathway, leading to the deposition of the C3 and C5b-9 membrane attack complex (MAC) on red blood cells (RBCs) (134–138). This mechanism parallels findings in patients experiencing severe HHS (138, 139), where uncontrolled complement activity accelerates RBC clearance and contributes to profound anaemia.
Whilst this mechanistic overlap is compelling, it is essential to acknowledge that direct evidence examining complement activation in patients with concurrent COVID-19 and SCD experiencing HHS remains limited (137, 138). Most inferences are drawn from separate bodies of literature on COVID-19 immunopathology and HHS in SCD, with a paucity of studies directly measuring complement components and activation products in this specific patient population.
In a report by Ita et al. (135), a teenage patient with SCD developed a spontaneous extradural haematoma during a vaso-occlusive crisis, with concomitant hyperinflammation, thrombotic microangiopathy, and HHS. The rapid deterioration of this patient despite conventional management necessitated the use of eculizumab, a C5 inhibitor, which ultimately reversed the cascade of complement-mediated destruction (134–138). The implication is clear: targeting complement activation could offer a lifeline in patients experiencing HHS in COVID-19. However, this represents a single case report, and the absence of controlled studies limits the strength of conclusions that can be drawn regarding the efficacy of this approach.
5.9.3 Macrophage overactivation and cytokine dysregulation
Macrophages play a key role in inflammation in both SCD and COVID-19 (139, 140).
Madu et al. (141) reviewed the mechanism by which activated macrophages are responsible for HHS. They demonstrated that changes in the red blood cell membrane resulted in immunological reactions against exposed membrane phospholipids in the erythrocytes (141). Integrin α4β1 in SCD erythrocytes, binding to VCAM-1 in macrophages, and ICAM-4, expressed in transfused erythrocytes, binding to CD11c/CD18 in macrophages, are critical in macrophage activation in HHS syndrome (80).
Beyond complement dysregulation, macrophage overactivation is a central driver of hyperhaemolysis in SCD (139). The phenomenon of macrophage priming in COVID-19 is well recognised, with IL-6 and IFN-γ implicated in the heightened phagocytic activity observed in severe cases (139–141). This cytokine surge may potentiate the excessive clearance of both transfused and autologous RBCs in HHS, a hypothesis supported by the findings of Fuja et al. (142), who documented a patient with SCD and recent COVID-19 hospitalisation presenting with HHS. This patient exhibited a complex alloimmune and autoimmune antibody profile, including anti-Fya, anti-Fyb, and anti-Lea, as well as strong cold agglutinins. The administration of tocilizumab, an IL-6 receptor antagonist, stabilised the patient’s haemoglobin levels, marking one of the first documented cases of successful IL-6 blockade in post-COVID-19 hyperhaemolysis (80, 142–144). This observation reinforces that immune hyperactivation in COVID-19 patients with SCD serves as a precipitating factor for severe HHS episodes, and that early immunomodulation may prevent catastrophic haemolysis.
However, it is worth noting that the literature has not adequately addressed potential contradictions between studies (80, 142–144). The variability in response to specific therapies could indicate heterogeneity in underlying mechanisms or differences in the definitions used for HHS (129, 130). The absence of comparative studies also limits our ability to determine whether specific immunological signatures might predict response to particular therapeutic interventions.
5.10 Emerging therapeutic paradigms for HHS in the COVID-19 context
5.10.1 Complement inhibition
Using eculizumab in HHS represents a targeted approach to interrupting the complement cascade, potentially limiting intravascular and extravascular haemolysis. Whilst case reports suggest promising outcomes, several limitations must be considered:
● Significant cost implications, which result in limited availability in many healthcare settings.
● Increased risk of meningococcal infections necessitating prophylaxis
● Absence of comparative efficacy data versus established therapies
The evidence base for eculizumab in other complement-mediated haemolytic disorders such as paroxysmal nocturnal haemoglobinuria (PNH) shares similar limitations (137, 145). A Cochrane review by Martí-Carvajal et al. (146) highlighted the paucity of high-quality evidence supporting the use of eculizumab in PNH, with conclusions based on a single small trial that had a risk of bias due to attrition and selective reporting (146). These parallels underscore the need for robust clinical trials before widespread implementation of complement inhibition in HHS.
5.10.2 Cytokine blockade
Applying tocilizumab for IL-6 blockade in HHS follows the recognition of hypercytokinaemia as a feature of both severe COVID-19 and HHS. The successful case reported by Fuja et al. (142) provides preliminary support for this approach, but important caveats include:
● There is limited evidence base with HHS.
● Potential for increased infection risk in an already vulnerable population.
● Uncertain optimal timing of administration in the disease course.
● Cost and accessibility concerns in resource-limited settings.
5.10.3 Alternative oxygen carriers
Alternative oxygen carriers provide a novel therapeutic approach for patients in whom conventional transfusion strategies prove futile. Mei et al. (145) reported three cases in which polymerised haemoglobin-based oxygen carriers (HBOC-201) were deployed under expanded access protocols, successfully stabilising patients who were otherwise refractory to transfusion therapy. This finding is particularly relevant in the context of COVID-19, where alloimmunization and transfusion restrictions may limit treatment options for patients experiencing severe hyperhaemolysis (143, 147). The ability to sustain oxygen delivery without further exacerbating immune-mediated haemolysis represents a crucial advance in managing HHS, especially in the context of emerging viral threats.
Key limitations include:
● Experimental status with limited availability
● Potential adverse effects include hypertension and nephrotoxicity
● Unknown long-term safety profile
● Regulatory hurdles to widespread implementation
5.10.4 JAK inhibition: a theoretical avenue
The advent of JAK inhibitors as potential modulators of immune-driven haemolysis adds another layer to this discussion. The inhibitors have been proposed for the treatment of thalassemia (147) and haemophagocytic lymphohistiocytosis.
Recent preclinical studies suggest that ruxolitinib, a JAK1/2 inhibitor, may attenuate macrophage-mediated haemolysis by dampening pro-inflammatory signalling cascades (148, 149). Although clinical data remain limited, the theoretical underpinnings of JAK inhibition in HHS warrant further exploration, particularly in the subset of patients with post-viral immune activation.
5.10.5 N-acetylcysteine treatment
The proposal to use N-acetylcysteine as an antioxidant has been discussed for many years, as reviewed by Wand and Zennadi (150). Nur et al. (151) evaluated treatment in SCD years ago in a small clinical trial; however, contrasting results were reported (152).
N-acetylcysteine has been used in COVID-19 patients (153), with a decrease in C-reactive protein and D-dimer levels. The potential use of N-acetylcysteine in viral and lung inflammation, aside from COVID-19, should benefit SCD patients (107); however, well-designed clinical trials are needed.
5.11 Autoimmunity in SCD: implications for COVID-19 patients
The interplay between SCD and autoimmunity (154, 155) represents an important but often overlooked dimension of disease pathophysiology, particularly relevant to patients with SARS-CoV-2 infection (156). Recent studies have highlighted the association between SCD and various autoimmune phenomena, including systemic inflammatory diseases (SIDs) (157), autoimmune haemolytic anaemia (AIHA) (158, 159), and immune thrombocytopenia (ITP) (160, 161).
The critical issue is that autoimmune responses generate a secondary immune deficiency, which is responsible for the impaired antiviral response (155–157). It would be interesting to analyse how many SCD patients have prolonged COVID-19 and what clinical characteristics are observed in long-term studies. In addition, monitoring antibodies to viral proteins and boosting vaccinations may also provide new elements to study in SCD.
5.11.1 Immune dysregulation and chronic inflammation
The chronic inflammatory state characteristic of SCD serves as a precursor to autoimmunity through several mechanisms (162). Haemolysis-induced oxidative stress, endothelial dysfunction, and recurrent VOCs collectively create an environment conducive to immune dysregulation, leading to aberrant autoantibody production and immune-mediated cytopenias (163–166). Elevated levels of pro-inflammatory cytokines such as IL-6, TNF-α, and IL-18 have been implicated in the pathogenesis of SCD and autoimmune conditions (69, 70, 163). This creates a concerning overlap with the cytokine profiles observed in severe COVID-19, which is particularly relevant for COVID-19 patients with SCD, as the virus-induced cytokine storm may amplify pre-existing immune aberrancies and potentially trigger or exacerbate autoimmune complications. The dysregulated release of cytokines in response to SARS-CoV-2 infection could tip the delicate balance in SCD patients toward autoimmune manifestations, further complicating their clinical course.
5.11.2 Autoimmune cytopenias and COVID-19
AIHA and ITP are increasingly reported in SCD patients and represent significant clinical challenges when they occur in the context of SARS-CoV-2 infection (164). AIHA in SCD is often refractory to conventional immunosuppressive therapies but has shown responsiveness to proteasome inhibitors, such as bortezomib, as demonstrated by Alanssari et al. (164). Similarly, cases of ITP in SCD have demonstrated positive responses to corticosteroids, highlighting the immune-mediated nature of these cytopenias, as noted by Alkhars et al. (165).
The emergence of autoimmune cytopenias after SARS-CoV-2 infection is particularly concerning for SCD patients, as it complicates their already fragile haematological status. Recent case reports highlight COVID-19-related autoimmune cytopenias in the general population, raising alarm about severe outcomes in SCD patients with compromised haematopoiesis.
5.11.3 Pathophysiological mechanisms and COVID-19 interactions
Several key pathophysiological mechanisms link SCD, autoimmunity, and COVID-19:
1. Complement dysregulation: Deficiencies in complement regulatory proteins contribute to increased autoimmune susceptibility in SCD. Abnormal activation of the complement cascade leads to increased opsonisation and immune complex deposition, exacerbating both haemolysis and systemic autoimmunity, as described by Floch et al. (166). SARS-CoV-2 infection has been shown to activate the complement system (167, 168), potentially creating a synergistic effect that increases the risk of autoimmune complications in SCD patients (169).
2. T cell and B cell aberrancies: SCD patients exhibit skewed T and B cell subsets, characterised by an increase in memory T cells and a reduction in regulatory B cells (52, 170, 171) and immature NK cells (172). This imbalance promotes chronic immune activation and may contribute to alloimmunisation following transfusion therapy. Memory T stem cells (TSCMs) have been identified as key drivers of autoimmunity in SCD, linking chronic inflammation to persistent immune activation, as reported by Fazeli et al. (173). SARS-CoV-2 infection can further disrupt T and B cell homeostasis, potentially exacerbating these pre-existing immune abnormalities (57, 58, 156).
3. Red blood cell alloimmunization and hyperhaemolysis: The risk of alloimmunization in SCD patients undergoing transfusion therapy during COVID-19 may be heightened due to the pro-inflammatory milieu. Alloantibody formation bridges transfusion-related immune responses and systemic autoimmunity, as noted by Pirenne and Pondarré (174). HHS is a severe complication of alloimmunisation, sharing mechanistic similarities with immune-mediated haemolysis seen in AIHA. It may be triggered or worsened by COVID-19-induced inflammation.
5.11.4 Therapeutic considerations for autoimmunity in SCD-COVID-19 patients
The management of autoimmune complications in SCD patients with COVID-19 requires careful consideration of both conditions:
1. Immunomodulatory interventions: The use of immunosuppressive agents, including corticosteroids, rituximab, and IVIG, remains crucial in managing autoimmune complications in SCD. However, their application in the context of active SARS-CoV-2 infection must be cautiously approached due to potential suppression of antiviral immune responses. Recent case reports by Alanssari et al. (164) highlight the efficacy of bortezomib in refractory AIHA, suggesting a role for proteasome inhibition in modulating autoimmune haemolysis in selected cases. Additionally, tocilizumab has been explored as treatment for severe COVID-19 and SCD-associated autoimmune manifestations, as described by De Luna et al. (175), representing a therapeutic convergence point.
2. Coexistence of SCD and systemic autoimmune disorders: Systemic lupus erythematosus (SLE), juvenile idiopathic arthritis (JIA), and inflammatory bowel disease (IBD) have been documented in paediatric and adult SCD cohorts (23, 161, 175). Inflammatory arthritis in SCD can mimic musculoskeletal manifestations of SCD, complicating diagnosis. Although treatments such as steroids, methotrexate, and anti-TNF agents are applied similarly, the heightened risk of infections and SCD crises requires tailored therapeutic strategies to ensure patient safety (19, 155, 176, 177). These conditions complicate diagnosis and management due to overlapping clinical features such as arthropathy, anaemia, and systemic inflammation. Notably, patients with concurrent SCD and SLE exhibit significant dysfunction of regulatory B cells, leading to a loss of immune tolerance, as demonstrated by Boulassel et al. (177). These patients may be particularly vulnerable to COVID-19 due to the triple hit of SCD, autoimmunity, and viral infection on their immune system function (177).
3. Alloimmunisation prevention and transfusion strategies: Genotypic matching for red blood cell transfusions is increasingly essential for SCD patients requiring transfusion support during SARS-CoV-2 infection (178). According to Wong et al. (179), HLA class II allelic polymorphisms have been identified as key genetic determinants influencing alloantibody formation, highlighting the importance of personalised transfusion strategies. Extended phenotype matching should be prioritised whenever possible to minimise the risk of alloimmunization and subsequent autoimmune complications.
4. Targeting the complement and cytokine axis: Emerging therapies targeting complement activation and pro-inflammatory may address both the virus-induced hyperinflammation and the autoimmune sequelae, potentially improving outcomes in this vulnerable patient population, as suggested by Poutrel et al. (180).
5. The coexistence of SCD, autoimmunity, and COVID-19 presents a complex clinical challenge that demands a multidisciplinary approach. Future research should aim to clarify the impact of COVID-19 on autoimmune manifestations in SCD and develop targeted therapies for this triad.
5.12 Splenic complications: the epicentre of a perfect storm
The spleen, already compromised by the relentless onslaught of vaso-occlusive crises in SCD, becomes the epicentre of a harrowing sequence of events upon SARS-CoV-2 infection, amplifying the morbidity and mortality risks inherent to both conditions (181).
The spleen is pivotal in host immunity, serving as a filtration site for abnormal erythrocytes and a key orchestrator of humoral defence (182). However, in SCD, recurrent microvascular occlusion precipitates progressive infarction and fibrosis, leading to functional autosplenectomy early in life (183). This leaves SCD patients profoundly vulnerable to infections, particularly by encapsulated bacteria such as Streptococcus pneumoniae and Haemophilus influenzae (184).
Superimposed on this immunological deficit, SARS-CoV-2 infection precipitates severe B-cell depletion within the splenic tissue, mirroring the immunoparesis observed in sepsis (185). Reductions in memory B cells critically impair antibody responses, predisposing individuals to secondary bacterial infections and prolonged viral persistence (186–188). The dual insult of chronic immunodeficiency from SCD and COVID-19-induced lymphoid atrophy culminates in a perilous state of immune collapse, rendering these patients extraordinarily susceptible to overwhelming infection and sepsis.
5.12.1 A thrombotic maelstrom: the catastrophic hypercoagulability of COVID-19 in SCD
SCD is intrinsically prothrombotic, with endothelial dysfunction, platelet activation, and chronic inflammation fostering a state of persistent hypercoagulability (187, 188). COVID-19 further exacerbates this hypercoagulable milieu by inducing widespread endothelial injury, cytokine-driven thrombogenesis, and the formation of microvascular thrombi (188). The spleen, already a site of extensive microvascular compromise in SCD, becomes acutely vulnerable to thrombotic insults, culminating in widespread infarction and accelerated splenic atrophy (189).
Clinical reports highlight the increased incidence of splenic infarction in COVID-19 patients, attributed to systemic endothelial dysfunction and disseminated microthrombosis (190). In the context of SCD, where splenic infarction is an established consequence of chronic vaso-occlusion, the superimposed thrombotic burden of SARS-CoV-2 infection threatens to accelerate the trajectory towards complete autosplenectomy and catastrophic multiorgan failure.
5.12.2 The spectre of acute splenic sequestration and rupture
Though many adults with SCD have lost splenic function due to autosplenectomy, residual splenic activity persists in some individuals, leaving them susceptible to acute splenic sequestration crisis (ASSC)—a phenomenon wherein large volumes of sickled erythrocytes become entrapped within the spleen, precipitating profound anaemia, hypovolaemic shock, and sudden circulatory collapse (190–192). COVID-19, which can exacerbate hypoxia and trigger erythrocyte adhesion, may act as a potent precipitant of ASSC, escalating the risk of rapid haemodynamic deterioration (192).
Moreover, emerging case reports have documented instances of spontaneous splenic rupture in patients with severe COVID-19 attributed to inflammation-induced architectural fragility and microvascular injury (193–195). In an SCD patient harbouring residual splenic tissue, the combined effects of vaso-occlusion, thrombotic congestion, and viral endothelial aggression could precipitate catastrophic splenic rupture, a rare yet potentially fatal sequela necessitating emergent surgical intervention.
5.13 Global variability of splenic complications and co-infections: implications for the SCD–COVID-19 interaction
Splenic dysfunction in SCD is not a monolithic entity. Instead, it is shaped by complex interactions between genetic predispositions, co-infectious landscapes, and systemic immunological burdens, which vary dramatically by geography. This heterogeneity has become increasingly salient in the context of COVID-19 as emerging data reveal that splenic status may critically influence disease outcomes and vaccine responses (193–195).
A systematic review by Ladu et al. (194) covering 14 African nations documented that splenomegaly persists into late childhood and even adulthood in substantial proportions of individuals with SCD, challenging the canonical view of early and universal auto-splenectomy. Acute splenic sequestration crises (ASSC) and hypersplenism were less frequent, while data on functional splenic capacity were almost entirely lacking, exposing a key blind spot in regional clinical research.
Tubman and Makani (195) coined the term “turf war” to describe the immunological tension between SCD and endemic malaria. In such regions, the spleen may remain enlarged but functionally compromised, particularly in its marginal zone B cell compartment, which undermines host defence against encapsulated bacteria and possibly viral pathogens. Malaria may delay autosplenectomy yet induce an altered immune phenotype.
The concept of immune decoupling is highlighted by Joice et al. (196), who found that HIV-positive paediatric patients with malaria had higher splenic parasite burdens and reduced phagocytic clearance, despite intact spleen structure. While SCD was not included in the study, the observed macrophage dysfunction and immunological stasis in the red pulp provide insights into the immune vulnerabilities of SCD patients exposed to SARS-CoV-2.
Insights from COVID-19 highlight important immunological issues. A review by Lenti et al. (197) described how SARS-CoV-2 targets the spleen, resulting in a significant depletion of IgM memory B cells, a sign of functional hyposplenism. This damage, confirmed by postmortem studies, persists even after clinical recovery, raising concerns about immune memory. Lenti et al. (197) refer to this as a “Janus-faced issue”: patients with existing asplenia or hyposplenism face higher risks of severe COVID-19. At the same time, COVID-19 may also induce new functional asplenia, complicating vaccination responses and immune defence against secondary infections.
These findings highlight that spleen size does not correlate with spleen function, particularly for SCD patients in malaria- or HIV-endemic areas. Splenic dysfunction in SCD–COVID–19 co-morbidity should be viewed as a spectrum rather than a binary state. Clinical protocols must incorporate function-based screening tools, such as pitted red cell counts and flow cytometry for B cell analysis (185). Vaccination should be prioritised for asplenic and hyposplenic individuals, especially in areas with endemic infections (198).
In conclusion, functional hyposplenism—whether congenital, infection-induced, or COVID-19-related—significantly impacts infection susceptibility, vaccine responses, and sepsis risk. Acknowledging this complexity is crucial for creating effective responses for SCD patients facing various infectious threats.
6 Avascular necrosis in SCD: exacerbation by COVID-19
6.1 Pathophysiology of AVN in SCD
The pathogenesis of AVN in SCD begins with the sickling of red blood cells, leading to vascular occlusion and subsequent ischaemia (199, 200). This process is particularly significant in the femoral head due to its unique vascular supply, which makes it vulnerable to ischaemic events. The initial VOC triggers a cascade of events, including endothelial activation, leukocyte adhesion, and platelet aggregation, which further compromise blood flow (199, 200). Osteonecrosis has been described in SCD (201).
Hypercoagulability plays a crucial role in the development of AVN in SCD, with this hypercoagulable state correlating with prevalent complications, including retinopathy and osteonecrosis (200).
6.2 Inflammatory mechanisms in AVN
Inflammation is a significant feature of SCD and a key contributor to the pathophysiology of AVN. Several inflammatory markers have been associated with SCD complications, including AVN. Domingos et al. (62) found that patients with higher levels of IL-6 had a significantly higher frequency of osteonecrosis compared to those with lower levels (p = 0.0006). Similarly, elevated IL-8 levels have been associated with other SCD complications, suggesting a broader role of inflammatory cytokines in disease manifestations (68).
The role of T cells in AVN development has been increasingly recognised. Daltro et al. (201) demonstrated that SCD patients with osteonecrosis have distinct CD4+ T cell profiles compared to those without this complication. Specifically, they found an increased frequency of circulating CD4+ T cells capable of simultaneously producing IFN-γ, IL-4, and IL-17 in SCD patients compared to healthy controls.
Tregs, characterised by the expression of CD4+CD25+FoxP3+, have also been implicated in the complications of SCD. Rêgo et al. (202) evaluated these cell populations in SCD patients with leg ulcers. They found that patients with a history of osteonecrosis showed higher IL-10 production in stimulated and unstimulated cultures, suggesting an altered immune response that may contribute to bone necrosis.
6.3 Genetic modifiers in AVN development
The clinical heterogeneity of SCD, including the variable occurrence of AVN, suggests the influence of genetic modifiers. Several studies (203–206) have identified polymorphisms in genes involved in inflammation, bone morphology, and vascular biology that may predispose SCD patients to AVN.
Baldwin et al. (203) found associations between single-nucleotide polymorphisms (SNPs) in the bone morphogenetic protein 6, annexin A2, and klotho genes and osteonecrosis in SCD patients. These genes are essential in bone morphology, metabolism, and vascular disease, providing insight into the genetic basis of AVN susceptibility.
Tozatto-Maio et al. (204) identified polymorphisms in inflammatory genes, including TLR2 and HLA-G, that modulate clinical complications in SCD patients. Specific genotypes were associated with fewer complications, suggesting potential protective effects.
The Duffy antigen receptor for chemokines (DARC) has been suggested as another potential genetic modifier in SCD. Farawela et al. (205) and Drasar et al. (206) investigated the relationship between DARC expression and disease severity, finding that Duffy-positive patients had higher white blood cell counts, which could contribute to increased inflammation and vaso-occlusion.
6.4 COVID-19 and AVN: emerging evidence
A retrospective analysis conducted by Sinha et al. (207) involving ten patients hospitalised due to COVID-19 who subsequently reported hip pain revealed degenerative changes consistent with AVN of the femoral head by magnetic resonance imaging (MRI). Notably, one patient with prior femoral surgery exhibited favourable recovery; however, they developed AVN following SARS-CoV-2 infection, indicating a possible association between COVID-19 and the onset of AVN. Additionally, the authors reported elevated IgG antibody levels, joint abnormalities, and moderately severe symptomatology, with a favourable response to supportive therapy and anti-inflammatory medications, which alleviated joint pain and acute inflammatory symptoms. Thus, COVID-19 may lead to significant musculoskeletal complications, AVN, likely due to corticosteroid use and microembolism (207). Further studies and long-term follow-up are suggested.
6.5 Mechanisms of COVID-19-Induced AVN in SCD
Several mechanisms may explain how COVID-19 could increase the risk of AVN in SCD patients:
1. Enhanced inflammation: COVID-19 may exacerbate the pre-existing inflammatory state in SCD, which has been linked to osteonecrosis (25, 207).
2. Hypercoagulability: Both COVID-19 and SCD are associated with a hypercoagulable state, potentially leading to a synergistic effect that increases the risk of vascular occlusion and subsequent AVN (35, 204–207).
3. Steroid use: The use of high-dose corticosteroids may contribute to the development of AVN, especially in SCD patients.
4. Microembolism: COVID-19-associated endothelial dysfunction and microthrombus formation may contribute to bone necrosis, particularly in patients with SCD.
7 Takotsubo syndrome in sickle cell disease during COVID-19
SCD patients live under relentless physiological and psychological pressure due to chronic haemolysis, recurrent VOCs, and systemic inflammation (208). The COVID-19 pandemic intensified this burden, resulting in an unprecedented level of biological stress. This confluence of hypoxia, inflammation, and emotional trauma dramatically increases the risk of Takotsubo syndrome (TTS)—a stress-induced cardiomyopathy that mimics acute coronary syndrome yet lacks coronary obstruction (209). Though TTS has traditionally been associated with postmenopausal women, it now emerges as a critical endpoint in SCD patients facing the compounded insults of COVID-19. In malaria-endemic regions, where many of these patients reside, additional physiological strain from co-infections and structural healthcare limitations exacerbate this fragile balance, tipping it toward cardiovascular collapse (210).
7.1 Takotsubo syndrome: historical context and pathophysiology
First described in 1990 by Japanese cardiologists Sato et al. (211), TTS derives its name from the takotsubo (Japanese for “octopus trap”), reflecting the characteristic apical ballooning of the left ventricle observed on echocardiography. While initially linked to postmenopausal women experiencing emotional trauma, TTS is now recognised as a systemic response to severe physiological or psychological stress (211). Its pathophysiology involves interrelated mechanisms:
1. Catecholamine Surge
Acute stress triggers excessive sympathetic nervous system activation, flooding the myocardium with adrenaline and noradrenaline (212). This disrupts cardiomyocyte metabolism, induces calcium overload, and impairs mitochondrial function (213). In SCD, chronic pain, hypoxia, and anxiety sustain elevated catecholamine levels, sensitising the heart to TTS (214).
2. Direct Myocardial Stunning
High catecholamine concentrations downregulate β-adrenergic receptors and impair cyclic AMP signalling, leading to transient left ventricular dysfunction (215). The apical myocardium, which has a higher density of β-receptors, becomes hypo-contractile, while the base hypercontracts, creating the classic “apical ballooning” appearance (216).
7.2 Microvascular dysfunction
Endothelial damage in SCD reduces nitric oxide (NO) bioavailability, limiting coronary vasodilation (217). COVID-19 exacerbates this via ACE2 receptor-mediated endothelial injury, promoting microthrombosis and vasospasm (218). The resultant mismatch between oxygen supply and demand precipitates myocardial ischaemia (219).
7.3 Inflammatory crosstalk
Chronic inflammation in SCD (elevated IL-6, TNF-α, CRP) synergises with COVID-19’s cytokine storm to amplify oxidative stress and cardiomyocyte apoptosis (220). Reactive oxygen species (ROS) from recurrent haemolysis and virus-induced hypoxia further destabilise myocardial contractility (218).
7.4 Neurohormonal factors
Oestrogen deficiency, common in postmenopausal women, may reduce cardioprotective effects (e.g., antioxidant activity, NO synthesis) (219). SCD patients, particularly women, often experience premature endocrine dysfunction due to chronic illness, potentially heightening TTS susceptibility (218, 220).
7.5 Purinergic system dysregulation
Burnstock first described purinergic nerves in 1972, establishing the foundation for understanding purinergic signalling (221). His later work highlighted the historical roles of purinergic signalling in blood cells, providing critical insights into how these pathways influence haematological disorders, such as SCD (221–223).
Emerging evidence implicates the purinergic system—a network of signalling molecules (e.g., ATP, adenosine) and receptors (P1, P2)—in TTS pathophysiology (223). During stress or viral infection, ATP is released from damaged cells or sympathetic nerve terminals, activating P2X and P2Y receptors. These receptors exacerbate catecholamine release, calcium overload, and microvascular spasm (224, 225). In COVID-19, the cytokine storm amplifies ATP spillage, creating a feedforward loop that intensifies myocardial stunning (222). Conversely, adenosine (via P1 receptors) modulates vasodilation and anti-inflammatory responses, but its overproduction during hypoxia may paradoxically suppress cardiac contractility (226). Zhang et al. (227) demonstrated that adenosine signalling has detrimental effects in SCD, potentially contributing to cardiovascular complications. Recent studies by Vandorpe et al. (228) confirmed that purinergic signalling is essential for full Psickle activation (i.e., activation of the deoxygenation-induced cation conductance pathway in sickle red blood cells, which leads to ion imbalance, cell dehydration, and increased sickling) by hypoxia and acidic pH, linking this system directly to SCD pathophysiology.
Targeting purinergic pathways (e.g., P2 receptor antagonists, adenosine modulators) could mitigate TTS progression by interrupting this syndromic cascade (229).
SCD and COVID-19: A Global Catalyst for Cardiac Stress
In SCD, chronic anaemia triggers compensatory mechanisms, such as increased cardiac output, which can lead to ventricular hypertrophy and diastolic dysfunction (230). COVID-19 amplifies this burden through:
● Hypoxia-Driven Crises: SARS-CoV-2 pneumonia exacerbates sickle cell-related hypoxia, accelerating HbS polymerisation and VOC frequency (231).
● Thrombotic Pathways: Platelet activation and hypercoagulability in COVID-19 increase the risk of pulmonary embolism or myocardial injury, often mimicking TTS (232).
● Psychosocial Stress: Pandemic-related isolation, delayed healthcare access, and fear of infection elevate cortisol and catecholamines, directly potentiating TTS (233).
Globally, these mechanisms increase the risk of TTS in patients with SCD. However, in malaria-endemic regions, overlapping pathologies—SCD, COVID-19, and malaria—create a ‘triple burden’ that magnifies cardiac strain (234).
7.6 Malaria co-endemicity: a compounding factor
In regions where malaria coexists with SCD, Plasmodium falciparum infection exacerbates haemolysis, inflammation, and endothelial dysfunction (235):
● Haemolytic Burden: Malaria-induced red cell destruction worsens anaemia, increasing cardiac workload (236).
● Inflammatory Synergy: Malaria’s cytokine release (e.g., IFN-γ, IL-1β) amplifies SCD- and COVID-19-driven inflammation, accelerating myocardial injury (237).
● Diagnostic Challenges: Overlapping symptoms (fever, fatigue) delay recognition of VOCs, COVID-19, or TTS, prolonging untreated cardiac stress (238).
● Healthcare Limitations: Resource constraints in these regions restrict access to therapies like hydroxycarbamide (hydroxyurea), transfusions, or ventilatory support, leaving stressors unmitigated (239).
This “triple burden” creates a vicious cycle, overwhelming compensatory mechanisms and increasing the incidence and severity of TTS (240). The connection between red blood cell pathology and inflammation in SCD, as elucidated by Nader et al. (241), further emphasises how this disease predisposes patients to cardiac complications during infectious challenges (241, 242).
7.7 Clinical management and challenges
Diagnosing TTS in SCD requires distinguishing it from acute coronary syndrome (ACS) or VOC-related pain. Key indicators include (243):
● Transient left ventricular dysfunction with apical ballooning.
● Absence of obstructive coronary disease on angiography.
● Precipitating stressors (e.g., recent COVID-19 diagnosis, severe infection).
7.8 Multidisciplinary management strategies
Given the complexity of overlapping conditions, patients with SCD presenting with acute coronary symptoms should be managed by a multidisciplinary team including:
1. Cardiologists: To assess ventricular function, rule out ACS, and guide heart failure therapies (e.g., beta-blockers, diuretics) (244).
2. Haematologists: Optimise SCD-specific care (e.g., hydration, analgesia, transfusion) (245).
3. Psychologists: To address anxiety, depression, and chronic stress, which exacerbate catecholamine surges (246).
4. Social Workers: To mitigate socioeconomic stressors (247).
Researchers have already documented several cases of Takotsubo syndrome triggered by COVID-19, including one that introduced the term “COVIDsubo” (248, 249). Nevertheless, no studies have reported this syndrome in individuals with sickle cell disease, alone or in combination with COVID-19. Despite overlapping stress-related pathophysiological mechanisms, this conspicuous absence calls for greater clinical vigilance and systematic case reporting in this vulnerable population.
Takotsubo syndrome represents a critical intersection of physiological and emotional stressors in SCD patients, amplified globally by COVID-19 and catastrophically worsened in malaria-endemic regions. The purinergic system plays a pivotal role in this complex pathophysiology, mediating stress responses, inflammation, and cardiac dysfunction.
While TTS is reversible, delayed recognition in high-risk cohorts, particularly where healthcare resources are limited, can lead to fatal arrhythmias or heart failure (42). Addressing this requires a dual focus: improving access to cardiac diagnostics and therapies worldwide, while prioritising malaria control and pandemic preparedness in vulnerable regions (249, 250).
Future research should explore biomarkers (e.g., cytokines, microRNA profiles, catecholamine metabolites) and pharmacological interventions targeting the purinergic system (e.g., P1/P2 receptor modulators) to disrupt the stress-catecholamine-cardiac injury axis (229, 251).
8 Placental pathophysiology in pregnant women with SCD and COVID-19: the hidden nexus
8.1 Placental vulnerability
The placenta is a critical interface between the maternal and foetal circulatory systems, regulating nutrient exchange, oxygenation, and immune defence (165, 252). Placental homeostasis may be severely compromised in systemic conditions such as SCD (253, 254) or SARS-CoV-2 infection (255). When these two entities converge in pregnancy, the result is a compounded burden of vascular dysfunction, inflammatory amplification, and immune dysregulation (256). This pathological convergence may precipitate pre-eclampsia, intrauterine growth restriction (IUGR), preterm birth, or stillbirth (26–261).
SCD contributes through chronic haemolysis, oxidative stress, and endothelial injury, while COVID-19 has been shown to induce trophoblast necrosis, intervillous fibrin deposition, and decidual inflammation. Histopathological studies have identified maternal vascular malperfusion (MVM), increased syncytial knots, and reduced placental branching in pregnancies with SCD (255, 261, 262). Meanwhile, SARS-CoV-2 placental infection is associated with chronic histiocytic intervillositis, CD68+ macrophage infiltration, and thrombotic microangiopathy (256).
8.2 Endothelial dysfunction and vascular insufficiency: a dual threat
Endothelial dysfunction is central to the pathogenesis of both SCD and COVID-19:
● In SCD, elevated placental growth factor (PlGF) levels drive endothelial activation and pulmonary hypertension via endothelin-1 (ET-1) upregulation (262). Free iron regulates PiGF (263). VOCs impair perfusion and promote infarction.
● In COVID-19, direct viral invasion of the placental endothelium through ACE2 and TMPRSS2 receptors leads to inflammation and fibrin deposition (57, 264).
The confluence of these mechanisms may result in chronic placental hypoperfusion, impaired nutrient delivery, and increased foetal distress.
8.3 Exacerbation of inflammatory pathways and immune dysregulation
SCD and COVID-19 are both characterised by systemic inflammation. Their co-occurrence intensifies leukocyte activation, cytokine release, and immunological imbalance:
● In SCD, PlGF promotes leukotriene synthesis and monocyte infiltration, sustaining a pro-inflammatory microenvironment (265, 266).
● SARS-CoV-2 infection alters the decidual immune profile: third-trimester infection increases macrophage, NK cell, and T cell activity, while second-trimester exposure leads to partial immune resolution (267–269).
This inflammatory synergy has been linked to pre-eclampsia, foetal inflammatory response syndrome, and early labour (268–271).
8.4 Hypoxia, pulmonary hypertension, and placental hypoperfusion
Both diseases impair maternal-foetal oxygen exchange:
● SCD results in chronic anaemia and hypoxia-induced erythropoiesis, which exacerbates placental resistance and microvascular damage (272).
● COVID-19 contributes via pulmonary microthrombosis and hypoxaemia, further restricting oxygen transfer to the fetus (273).
This cumulative hypoxic burden raises the likelihood of placental abruption, IUGR, and perinatal loss.
8.5 The prothrombotic state and placental vascular pathology
Both SCD and COVID-19 promote a hypercoagulable state:
● In SCD, increased PAI-1 expression inhibits fibrinolysis, while VOCs induce platelet activation (268).
● COVID-19 introduces microthrombi, intervillous haemorrhage, and diffuse fibrin deposition within the placenta (269).
This prothrombotic milieu accelerates placental infarction and stillbirth risk.
The catastrophic events may be increased with flavivirus infections (58).
8.6 Diagnostic and prognostic utility of placental pathology
Placental histology may offer predictive and diagnostic insights in SCD-COVID-19 pregnancies:
● Histopathological markers (e.g., MVM, intervillous thrombosis, syncytial knots) help differentiate between SCD- and COVID-19-driven injury (256, 261).
● Biomarkers such as PlGF, thrombospondin, and VEGF may assist in identifying at-risk pregnancies and guiding management (270).
These findings underscore the need for systematic placental examination and longitudinal follow-up in affected cohorts.
8.7 Clinical and therapeutic implications
Pregnant patients with coexisting SCD and COVID-19 warrant enhanced surveillance (271):
Clinical management approach, incorporating:
● Close maternal-foetal monitoring using Doppler ultrasound and biomarker profiling.
● Preventive anticoagulation therapy to reduce thrombotic risks.
● PlGF-targeted therapies for vascular and inflammatory control in SCD.
● Vaccination as a priority.
9 Therapeutic considerations and management strategies
9.1 Challenges in management
Managing patients with SCD and COVID-19 presents unique challenges, particularly in distinguishing COVID-19 pneumonia from acute chest syndrome. Uncertainty around transfusion strategies and achieving proper anticoagulation complicates treatment. Immunomodulatory therapies used for severe COVID-19, like steroids and IL-6 inhibitors, have not been thoroughly studied in SCD and could affect VOC (274). Monitoring for signs of acute splenic sequestration, splenic infarction, or rupture is mandatory. The goal is to avoid risks of hypoxia-induced sickling and thrombosis. Prophylaxis against bacterial infections is crucial, especially for those with functional hypersplenism.
9.2 Approaches for AVN management in SCD and COVID-19
Managing AVN in SCD is challenging due to limited effective interventions. A Cochrane Review by Martí-Carvajal et al. (274) found no significant improvement in SCD patients with AVN when hip core decompression was added to physical therapy, based on a single trial with high attrition rates. Al-Otaibi’s review (272) suggests that initial treatments may include pharmacological agents, bone morphogenetic protein stimulation, and surgical options, such as total hip arthroplasty. A multidisciplinary approach is essential for effective patient management. Supportive care for SCD patients in vaso-occlusive crises includes intravenous hydration, pain management, and sometimes blood transfusion (273). These measures can help prevent progression to AVN by addressing vaso-occlusion and ischaemia.
In the context of COVID-19 management strategies, AVN in SCD patients may need adaptation:
1. Anti-inflammatory therapy: Targeted anti-inflammatory treatments may benefit patients with SCD and COVID-19. Low-dose methotrexate has shown promise (275).
2. Anticoagulation: Prophylactic anticoagulation may be considered for SCD patients with COVID-19 (14); however, this requires further investigation.
3. Careful steroid use is essential due to the potential link between high-dose corticosteroids and AVN in SCD patients with COVID-19. Almeida et al. (276) found that both conventional (prednisolone) and dissociative (vamorolone) corticosteroids worsen liver injury in sickle cell mice, raising concerns about their use in SCD patients.
4. Increased vigilance for early signs of AVN in SCD patients recovering from COVID-19 may allow for earlier intervention and better outcomes. Regular assessments and imaging studies should be included in the post-COVID-19 follow-up for these patients.
9.3 Management of autoimmune complications in SCD-COVID-19
1. Tailored immunosuppression: In patients with SCD, autoimmunity, and COVID-19 treatment must be customised to avoid compromising antiviral immunity. In select cases, targeted therapies such as bortezomib for refractory autoimmune haemolytic anaemia (AIHA) may be preferable to broad immunosuppression (162, 164).
2. Monitoring for immune cytopenias: It’s essential to monitor for AIHA and ITP in SCD patients with COVID-19, as viral infections can trigger or worsen these autoimmune cytopenias (277).
3. Balancing transfusion needs and the risk of alloimmunization: SCD patients with autoimmune complications often require transfusion support, but this carries an increased risk of alloimmunization, particularly in the pro-inflammatory state induced by COVID-19. SCD patients with the FcγR2B 2B.4 haplotype have an increased risk of RBC alloimmunization (278). Genetic polymorphism studies in these high-risk patients it is strongly recommended (279).
In these complex cases, extended phenotype matching and alternative strategies, such as erythropoiesis-stimulating agents, may be considered for treatment.
4. Managing systemic autoimmune diseases: For patients with SCD and systemic autoimmune diseases (such as SLE or JIA), COVID-19 may trigger flares that require prompt intervention (280). Hydroxychloroquine may be particularly beneficial in this subset of patients, although the evidence is limited (156).
9.4 Recommended preventive approaches for SCD patients
1. Prevention strategies: Vaccination should be prioritised.
2. Infection control measures and remote healthcare. Telemedicine can be used to maintain necessary monitoring and management.
3. Prophylactic strategies: Early therapeutic interventions.
Pregnant patients with SCD require specialised management protocols as detailed in obstetric considerations elsewhere in this review.
9.5 Treatment adaptations
Early hospitalisation is recommended, as well as aggressive hydroxyurea optimisation to ensure optimal HbF induction to reduce COVID-19-related hypoxemia.
A prophylactic exchange transfusion for selected high-risk patients, along with preventive anti-inflammatory therapy and management of hyperhaemolysis, is recommended in clinical settings.
A clinical decision-making algorithm is proposed for SCD patients that includes: 1) vigilant monitoring of post-transfusion haemoglobin levels in SCD patients, 2) monitoring inflammatory markers, complement activation, and parameters indicative of haemolysis; 3) stratify severity based on the rate of haemoglobin decline, presence of end-organ damage, and associated comorbidities; 4) adopt an escalating therapeutic approach: administer corticosteroids and intravenous immunoglobulin as first line of treatment and evaluate the potential use of biological therapies.
For refractory cases, consider evaluating alternative oxygen carriers by applicable regulatory guidelines.
Suggestions for the autoimmune complication management: 1) prompt differentiation between alloimmune and autoimmune haemolysis through comprehensive antibody screening and direct antiglobulin testing; 2) consider proteasome inhibition for refractory AIHA cases; 3) monitor for the emergence or exacerbation of systemic autoimmune manifestations during and after SARS-CoV-2 infection; 4) multidisciplinary patient management.
9.6 Socioeconomic considerations
Several socioeconomic considerations should be taken into account. SCD patients may be particularly vulnerable due to their financial situations. Patients must have access to necessary tests, treatments, and vaccinations. There should be a specialised centre with well-designed protocols for immunological testing, pain management, and emergency care, despite the high costs associated with specialised treatment and care.
10 Future research directions
Our analysis identifies several critical areas for future research:
10.1 Mechanistic studies
Longitudinal studies focusing on genetic markers, immune cell populations, cytokine profiles, neutrophil activation, and endothelial markers in patients with SCD, across both paediatric and adult populations, are essential for a better understanding of the disease’s pathophysiology. Furthermore, research is needed to explore the mechanisms of endothelial damage in SCD patients who have contracted COVID-19 or are experiencing long COVID (281, 282).
More studies are required on genetic modifiers in SCD. Studies examining how genetic factors that modify SCD severity interact with COVID-19 outcomes, including the analysis of specific inflammatory cells and biomarkers or genetic markers for AVN risk.
Prospective investigation of macrophage and complement activation products (C3a, C5a, soluble C5b-9) in SCD patients with COVID-19 with a particular focus on those who develop HHS.
A comprehensive analysis of autoimmune signatures in SCD-COVID-19 patients is essential. Characterising autoantibody profiles and B cell subsets before and after COVID-19 infection will help identify predictors of autoimmune complications, guiding clinical decisions.
10.2 Clinical research priorities
In SCD, there is a need for specific trials and registry-based studies of therapeutics for viral infections, particularly in COVID-19 and long-COVID-19. Moreover, comparative studies are required to examine the differences between simple versus exchange transfusion, liberal versus restrictive approaches, and prophylactic versus therapeutic strategies.
There is a need for controlled trials of complement inhibitors, cytokine blockade, and alternative oxygen carriers in patients with SCD. It is clinically relevant to develop and validate standardised imaging protocols for the early detection of AVN in SCD patients, along with testing for autoimmune diseases and autoimmune therapy and response to vaccines to prevent complications in these patients.
10.3 Epistemic integration
Integrated multidisciplinary studies that explore the interaction among malaria, SCD, and COVID-19, along with biomarker validation, are essential for understanding the relationship among these conditions and other viral infections. Additionally, well-designed studies are needed to develop effective prevention and treatment strategies for patients with SCD. The studies should include Recommendations for Interventional Trials (SPIRIT) statement for trial conduct and the Consolidated Standards of Reporting Trials (CONSORT) statement for reporting, including detailed medical history and treatments.
11 Conclusion
The interplay between SCD and COVID-19 represents a complex intersection with significant clinical implications. There are concerning epistemic gaps in the current literature, particularly in mechanistic investigations, clinical studies, and the omission of SCD in African COVID-19 research.
The immunopathogenesis involves synergistic inflammatory processes, vascular dysfunction, and an increased risk of haemolytic-uremic syndrome and avascular necrosis. While the evidence base consists predominantly of case reports and small series, promising therapeutic avenues have emerged, necessitating a transition from anecdotal successes to evidence-based approaches that prioritise early recognition and targeted intervention.
The evidence synthesised in this review predominantly reflects experiences with earlier, more virulent SARS-CoV-2 variants. While currently circulating variants demonstrate reduced population-level virulence, the mechanistic vulnerabilities identified in SCD patients—chronic inflammation, immune dysregulation, and organ dysfunction—suggest that vigilance remains warranted regardless of viral evolution. The pathophysiological intersections described herein represent host response patterns that may be triggered across the spectrum of viral variants, particularly in this immunologically vulnerable population.
Future research must address both mechanistic questions and socioeconomic contexts. Through integrating epistemic awareness with rigorous pathophysiological investigation, we can develop more effective approaches to protect this vulnerable population.
Author contributions
AM-C: Formal Analysis, Writing – original draft, Writing – review & editing, Data curation, Investigation, Conceptualization, Methodology, Validation, Visualization. JD: Writing – review & editing, Conceptualization, Funding acquisition, Resources, Investigation, Validation, Project administration, Formal Analysis, Supervision, Methodology, Visualization, Data curation.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. JD is partially financed by the National Institute of Virology and Bacteriology (Program EX-CELES, ID Project No. LX22NPO5103)—Funded by the European Union—Next Generation EU from the Ministry of Education, Youth, and Sports of the Czech Republic (MEYS).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Vives Corrons JL and De Sanctis V. Sickle cell disease and COVID-19infection. Acta Biomed. (2020) 91:347–8. doi: 10.23750/abm.v91i2.9532
2. Arlet JB, de Luna G, Khimoud D, Odièvre MH, de Montalembert M, Joseph L, et al. Prognosis of patients with sickle cell disease and COVID-19: a French experience. Lancet Haematol. (2020) 7:e632–4. doi: 10.1016/S2352-3026(20)30204-0
3. Azerad MA, Bayoudh F, Weber T, Minon JM, Ketelslegers O, Compagnon P, et al. Sickle cell disease and COVID-19: Atypical presentations and favourable outcomes. EJHaem. (2020) 1:338–41. doi: 10.1002/jha2.74
4. Elia G, Sainati L, Cappellari AM, and Cesaro S. Acute chest syndrome in pediatric sickle cell disease during the COVID-19pandemic. Pediatr Blood Cancer. (2021) 68:e28831. doi: 10.1016/j.htct.2020.11.005
5. Martone S, Giordano P, Montante C, Vendemiale M, and Lassandro G. Hepatic dysfunction and COVID-19in sickle cell disease: a case series. Pediatr Blood Cancer. (2021) 68:e28809. doi: 10.1002/pbc.28874
6. Clarke M, Junkins C, Stotts M, Rodriguez HE, Pujari S, and Serpe JN. Neurological events in sickle cell disease and COVID-19: a case series. Neurol Sci. (2022) 43:499–502.
7. Sayad B, Karimi M, and Rahimi Z. COVID-19and sickle cell disease: susceptibility and severity. Pediatr Blood Cancer. (2021) 68:e28898. doi: 10.1002/pbc.29075
8. Joseph L, Kodjo Y, Kone Y, Ouattara A, Diallo S, Cisse DM, et al. Maternal outcomes of COVID-19in pregnancy with sickle cell disease: a multicenter analysis. Am J Obstet Gynecol MFM. (2024) 6:100929. doi: 10.1007/s12288-021-01482-1
9. Gibson AC, Culpepper C, Morgan S, Shetty A, and De Castro L. Obstetric outcomes in sickle cell disease complicated by COVID-19: case series and management approach. Am J Perinatol. (2024) 41:224–30. doi: 10.4103/ajts.ajts_164_21
10. Kolanska K, Courbière B, Pétrini AC, Dechartres A, Senat MV, Younes N, et al. Sickle cell disease, pregnancy, and COVID-19: an observational study. J Gynaecol Obstet. (2022) 44:78–84. doi: 10.1016/j.jogoh.2022.102328
11. Nguyen T, Alzubi J, Roy NB, Karimi M, Al-Khatti A, Graido-Gonzalez E, et al. Effectiveness of red blood cell exchange transfusion in sickle cell patients with COVID-19. Br J Haematol. (2021) 194:e150–3. doi: 10.1002/ccr3.4655
12. Mawalla WF, Idowu A, Fredrick F, Balandya E, and Makubi A. Management of acute chest syndrome in hydroxyurea-naïve patients with sickle cell disease and COVID-19. Pan Afr Med J. (2022) 41:110. doi: 10.1002/jha2.397
13. Umar BA, Ibrahim UA, Hassan A, Wali SG, Okolie CA, and Adeleke SI. Management of sickle cell disease patients with COVID-19: screening and treatment approaches. Haematology. (2022) 27:238–45. doi: 10.7759/cureus.23604
14. Alsayegh F and Mousa SA. Challenges in the management of sickle cell disease during SARS-CoV-2 pandemic. Clin Appl Thromb Haemost. (2020) 26:1076029620955240. doi: 10.1177/1076029620955240
15. Brousse V, Holvoet L, Pescarmona R, Viel S, Perret M, Visseaux B, et al. Low incidence of COVID-19 severe complications in a large cohort of children with sickle cell disease: a protective role for basal interferon-1 activation? Haematologica. (2021) 106:2746–8. doi: 10.3324/haematol.2021.278573
16. Martin OY, Darbari DS, Margulies S, Nickel RS, Leonard A, Speller-Brown B, et al. Clinical outcomes of children and adolescents with sickle cell disease and COVID-19infection: a year in review at a metropolitan tertiary paediatric hospital. Front Med (Lausanne). (2023) 10:987194. doi: 10.3389/fmed.2023.987194
17. Michelon I, Vilbert M, Pinheiro IS, Costa IL, Lorea CF, Castonguay M, et al. COVID-19 outcomes in patients with sickle cell disease and sickle cell trait compared with individuals without sickle cell disease or trait: a systematic review and meta-analysis. EClinicalMedicine. (2023) 66:102330. doi: 10.1016/j.eclinm.2023.102330
18. Santo IS. Mortality data of COVID-19 in patients with sickle cell disease in Brazil: a regional analysis. Rev Bras Haematol Haemoter. (2024) 46:112–8. doi: 10.1590/1806-9282.20231466
19. Vinit C, Guitton C, de Montalembert M, Benhaim P, Amor-Chelihi L, Bader-Meunier B, et al. Systemic inflammatory diseases in children with sickle cell disease: a French multicenter observational study on diagnostic and therapeutic issues. Pediatr Blood Cancer. (2025) 72:e31563. doi: 10.1002/pbc.31563
20. Kato GJ, Piel FB, Reid CD, and Gaston MH. Sickle cell disease. Nat Rev Dis Primers. (2018) 4:18010. doi: 10.1038/nrdp.2018.10
21. Herrick JB. Peculiar elongated and sickle-shaped red blood corpuscles in a case of severe anaemia. Arch Intern Med. (1910) 6:517. doi: 10.1001/archinte.1910.00050330050003
22. Pauling L, Itano HA, Singer SJ, and Wells IC. Sickle cell anaemia: a molecular disease. Science. (1949) 110:543–8. doi: 10.1126/science.110.2865.543
23. Ingram VM. Gene mutations in human haemoglobin: the chaemical difference between normal and sickle cell haemoglobin. Nature. (1957) 180:326–8. doi: 10.1038/180326a0
24. GBD 2021 Sickle Cell Disease Collaborators. Global, regional, and national prevalence and mortality burden of sickle cell disease, 2000–2021: a systematic analysis from the Global Burden of Disease Study 2021. Lancet Haematol. (2023) 10:e585–99. doi: 10.1016/S2352-3026(23)00118-7
25. Dorneles J, de Menezes Mayer A, and Chies JAB. Sickle cell anaemia and inflammation: a review of stones and landmarks paving the road in the last 25 years. Haematol Rep. (2025) 17:2. doi: 10.3390/hematolrep17010002
26. Dawudi Y, Azoyan L, Bonjour M, and Steichen O. COVID-19 outcomes among patients with sickle cell disease or sickle cell trait compared to the general population: a systematic review and meta-analyses. Ann Hematol. (2024) 103:5071–83. doi: 10.1007/s00277-024-06113-z
27. Alagbe AE, Domingos IF, Adekile AD, Blotta MHSL, and Santos MNN. Anti-inflammatory cytokines in sickle cell disease. Mol Biol Rep. (2022) 49:2433–42. doi: 10.1007/s11033-021-07009-1
28. Delgadinho M, Veiga L, Ginete C, Santos B, Miranda A, de Vasconcelos JN, et al. Differential expression of adhesion molecules in sickle cell anaemia and gut microbiome effect. Ann Haematol. (2024) 103:409–19. doi: 10.1007/s00277-023-05589-5
29. Recchiuti A, Federti E, Matte A, Mazzi F, Ceolan J, Porreca A, et al. Impaired pro-resolving mechanisms promote abnormal NETosis, fueling autoimmunity in sickle cell disease. Am J Haematol. (2023) 98:E45–8. doi: 10.1002/ajh.26797
30. de Ligt LA, Gaartman AE, Biemond BJ, Fijnvandraat K, van Bruggen R, and Nur E. Neutrophils in sickle cell disease: exploring their potential role as a therapeutic target. Am J Haematol. (2024) 99:1119–28. doi: 10.1002/ajh.27224
31. Warner S, Teague HL, Ramos-Benitez MJ, Panicker S, Allen K, Gairhe S, et al. R406 reduces lipopolysaccharide-induced neutrophil activation. Cell Immunol. (2024) 403–404:104860. doi: 10.1016/j.cellimm.2024.104860
32. Elsherif L, Tang Y, Patillo KL, Wichlan D, Ogu UO, Landes K, et al. Association of biomarkers of endothelial function, coagulation activation and kidney injury with persistent albuminuria in sickle cell anaemia. Br J Haematol. (2024) 205:1963–73. doi: 10.1111/bjh.19743
33. Dobie G. Sickle cell disease and thromboembolism: new insights on the pathophysiology, diagnosis, and treatment. Clin Lab. (2023) 69. doi: 10.7754/Clin.Lab.2023.221006
34. Nagose VB, Kodate PM, Kumbhalkar DT, Rathod SS, and Nayak SP. Platelet aggregation studies and coagulation profile in sickle cell disease in symptomatic and steady state patients. Indian J Haematol Blood Transfus. (2024) 40:281–8. doi: 10.1007/s12288-023-01703-9
35. Bou-Fakhredin R, Cappellini MD, Taher AT, and De Franceschi L. Hypercoagulability in haemoglobinopathies: decoding the thrombotic threat. Am J Haematol. (2025) 100:103–15. doi: 10.1002/ajh.27500
36. Zheng Y, Gossett JM, Chen PL, Barton M, Ryan M, Yu J, et al. Proinflammatory state promotes red blood cell alloimmunisation in paediatric patients with sickle cell disease. Blood Adv. (2023) 7:4799–808. doi: 10.1182/bloodadvances.2022008647
37. Swirski FK and Nahrendorf M. Bone marrow’s role in chronic inflammation rewires haematopoiesis in SCD. J Exp Med. (2017) 214:2243–5. doi: 10.1111/bjh.15902
38. Fortuna V, Lima J, Oliveira GF, Oliveira YS, Getachew B, Nekhai S, et al. Ferroptosis as an emerging target in sickle cell disease. Curr Res Toxicol. (2024) 7:100181. doi: 10.1016/j.crtox.2024.100181
39. Beckman JD, Zhang P, Nguyen J, Hebbel RP, Vercellotti GM, and Belcher JD. Missing the mark(ers): circulating endothelial cells and endothelial-derived extracellular vesicles are elevated in sickle cell disease plasma. Front Immunol. (2024) 15:1493904. doi: 10.3389/fimmu.2024.1493904
40. Karmakar J, Mukherjee K, and Mandal C. Siglecs modulate activities of immune cells through positive and negative regulation of ROS generation. Front Immunol. (2021) 12:758588. doi: 10.3389/fimmu.2021.758588
41. Van Avondt K, Schimmel M, Bulder I, van Mierlo G, Nur E, van Bruggen R, et al. Circulating iron in patients with sickle cell disease mediates the release of neutrophil extracellular traps. Transfus Med Haemother. (2023) 50:321–9. doi: 10.1159/000526760
42. Tirandi A, Carbone F, Montecucco F, and Liberale L. The role of metabolic syndrome in sudden cardiac death risk: recent evidence and future directions. Eur J Clin Invest. (2022) 52:e13693. doi: 10.1111/eci.13693
43. Westover AJ, Humes HD, and Pino CJ. Immunomodulatory effects of a cell processing device to ameliorate dysregulated hyperinflammatory disease states. Sci Rep. (2024) 14:12747. doi: 10.1038/s41598-024-63121-9
44. Argueta DA, Tran H, Goel Y, Nguyen A, Nguyen J, Kiven SB, et al. Mast cell extracellular trap formation underlies vascular and neural injury and hyperalgesia in sickle cell disease. Life Sci Alliance. (2024) 7:e202402788. doi: 10.26508/lsa.202402788
45. Gingell L and Hrinczenko B. Characterizing the immature immunophenotype of sickle cell disease monocytes. Cureus. (2024) 16:e60703. doi: 10.7759/cureus.60703
46. Gbotosho OT, Li W, Joiner CH, Brown LAS, and Hyacinth HI. The inflammatory profiles of pulmonary alveolar macrophages and alveolar type 2 cells in SCD. Exp Biol Med (Maywood). (2023) 248:1013–23. doi: 10.1177/15353702231157940
47. Kapellos TS, Bonaguro L, Gemünd I, Reusch N, Saglam A, Hinkley ER, et al. Human monocyte subsets and phenotypes in major chronic inflammatory diseases. Front Immunol. (2019) 10:2035. doi: 10.3389/fimmu.2019.02035
48. Allali S, Rignault-Bricard R, de Montalembert M, Taylor M, Bouceba T, Hermine O, et al. HbS promotes TLR4-mediated monocyte activation and proinflammatory cytokine production in sickle cell disease. Blood. (2022) 140:1972–82. doi: 10.1182/blood.2021014894
49. Pinto-García N, Júnior ALS, Soares GAS, Costa TCC, dos Santos APC, Costa AG, et al. Sickle cell anaemia patients display an intricate cellular and serum biomarker network highlighted by TCD4+CD69+ lymphocytes, IL-17/MIP-1β, IL-12/VEGF, and IL-10/IP-10 axis. J Immunol Res. (2020) 2020:4585704. doi: 10.1155/2020/4585704
50. Felício RFM, Jarduli-Maciel LR, Mosella MQS, Almeida FC, de Lima KC, de Azevedo JTC, et al. Transcriptome profiling reveals distinct alterations in the B-cell signature and dysregulation of peripheral B-cell subsets in sickle cell anaemia patients. Exp Haematol. (2024) 137:104254. doi: 10.1016/j.exphem.2024.104254
51. Zahran AM, Saad K, Elsayh KI, Khalaf SM, Mahmoud KH, Elhoufey A, et al. Regulatory T-cell phenotypes in children with sickle cell disease. Pediatr Res. (2022) 91:1203–6. doi: 10.1038/s41390-021-01627-y
52. Patel S, Chandrakar D, Wasnik PN, Nayak S, Shah S, Nanda R, et al. Altered T-cell profile in sickle cell disease. biomark Med. (2023) 17:241–52. doi: 10.2217/bmm-2023-0086
53. Su S, Bao W, Liu Y, Shi PA, Manwani D, Murakhovskaya I, et al. IFN-I promotes T-cell-independent immunity and RBC autoantibodies via modulation of B-1 cell subsets in murine SCD. Blood. (2025) 145:334–47. doi: 10.1182/blood.2024025175
54. Marchesani S, Bertaina V, Marini O, Cossutta M, Di Mauro M, Rotulo GA, et al. Inflammatory status in pediatric sickle cell disease: unravelling the role of immune cell subsets. Front Mol Biosci. (2023) 9:1075686. doi: 10.3389/fmolb.2022.1075686
55. Bashi A, Lekpor C, Hood JL, Thompson WE, Stiles JK, and Driss A. Modulation of haeme-induced inflammation using microRNA-loaded liposomes: implications for haemolytic disorders such as malaria and sickle cell disease. Int J Mol Sci. (2023) 24:16934. doi: 10.3390/ijms242316934
56. Giriraja KV, Bhatnagar SK, Tomlinson L, and Sancilio F. An open-label, multicentre, phase 2 study of a food enriched with docosahexaenoic acid in adults with sickle cell disease. Prostaglandins Leukot Essent Fatty Acids. (2023) 193:102574. doi: 10.1016/j.plefa.2023.102574
57. De Sanctis JB, García AH, Moreno D, and Hajduch M. Coronavirus infection: an immunologists’ perspective. Scand J Immunol. (2021) 93:e13043. doi: 10.1111/sji.13043
58. García AH and De Sanctis JB. Exploring the contrasts and similarities of dengue and SARS-CoV-2 infections during the COVID-19era. Int J Mol Sci. (2024) 25:11624. doi: 10.3390/ijms252111624
59. Arabi M, Al-Najjar Y, Mhaimeed N, Salameh MA, Paul P, AlAnni J, et al. Severity of the Omicron SARS-CoV-2 variant compared with the previous lineages: A systematic review. J Cell Mol Med. (2023) 27:1443–64. doi: 10.1111/jcmm.17747
60. Scourfield LEA, Nardo-Marino A, Williams TN, and Ress DC. Infections in sickle cell disease. Hematologica. (2025) 110:546–61. doi: 10.3324/haematol.2024.285066
61. Peng HK, Dombkowski KJ, Plegue MA, Latta K, Malosh R, Creary MS, et al. COVID-19 immunization coverage among people with sickle cell disease. JAMA. (2024) 7:e2351618. doi: 10.1001/jamanetworkopen.2023.51618
62. Domingos IF, Pereira-Martins DA, Sobreira MJVC, Oliveira RTD, Alagbe AE, Lanaro C, et al. High levels of proinflammatory cytokines IL-6 and IL-8 are associated with a poor clinical outcome in sickle cell anaemia. Ann Haematol. (2020) 99:947–53. doi: 10.1007/s00277-020-03978-8
63. Siransy LK, Dasse RS, Adou H, Kouacou P, Kouamenan S, Sekongo Y, et al. Are IL-1 family cytokines important in management of sickle cell disease in Sub-Saharan Africa patients? Front Immunol. (2023) 14:954054. doi: 10.3389/fimmu.2023.954054
64. Tomasik J and Basak GW. Inflammasomes–new contributors to blood diseases. Int J Mol Sci. (2022) 23:8129. doi: 10.3390/ijms23158129
65. Allali S, de Montalembert M, Rignault-Bricard R, Taylor M, Brice J, Brousse V, et al. IL-6 levels are dramatically high in the sputum from children with sickle cell disease during acute chest syndrome. Blood Adv. (2020) 4:6130–4. doi: 10.1182/bloodadvances.2020003519
66. de Azevedo JTC and Malmegrim KCR. Immune mechanisms involved in sickle cell disease pathogenesis: current knowledge and perspectives. Immunol Lett. (2020) 224:1–11. doi: 10.1016/j.imlet.2020.04.012
67. Rincón-López EM, Navarro Gómez ML, Hernández-Sampelayo Matos T, Aguilera-Alonso D, Dueñas Moreno E, Saavedra-Lozano J, et al. Interleukin 6 as a marker of severe bacterial infection in children with sickle cell disease and fever: a case-control study. BMC Infect Dis. (2021) 21:741. doi: 10.1186/s12879-021-06470-4
68. Duits AJ, Schnog JB, Lard LR, Saleh AW, and Rojer RA. Elevated IL-8 levels during sickle cell crisis. Eur J Haematol. (1998) 61:302–5. doi: 10.1111/j.1600-0609.1998.tb01092.x
69. Patel P, DeCuir J, Abrams J, Campbell AP, Godfred-Cato S, and Belay ED. Clinical characteristics of multisystem inflammatory syndrome in adults: a systematic review. JAMA Netw Open. (2021) 4:e2126456. doi: 10.1001/jamanetworkopen.2021.26456
70. Peng Z and Zhou G. Progress on diagnosis and treatment of multisystem inflammatory syndrome in children. Front Immunol. (2025) 16:1551122. doi: 10.3389/fimmu.2025.1551122
71. Chiang KC, Gupta A, Sundd P, and Krishnamurti L. Thrombo-inflammation in COVID-19and sickle cell disease: two faces of the same coin. Biomedicines. (2023) 11:338. doi: 10.3390/biomedicines11020338
72. Saxena P and Muthu J. COVID-19 and sickle cell disease: two independent risk factors for venous thromboembolism. Cureus. (2023) 15:e37226. doi: 10.7759/cureus.37226
73. Dobkin J, Wu L, and Mangalmurti NS. The ultimate tradeoff: how red cell adaptations to malaria alter the host response during critical illness.Am J Physiol Lung Cell Mol Physiol. (2023) 324(2):L169–78. doi: 10.1152/ajplung.00127.2022
74. Chen-Goodspeed A and Idowu M. COVID-19 presentation in patients with sickle cell disease: a case series. Am J Case Rep. (2021) 22:e931758. doi: 10.12659/AJCR.931758
75. Woodford J, Sagara I, Kwan J, Assadou MH, Katile A, Attaher O, et al. Impact of prior malaria exposure on COVID-19in Mali. Nature. (2022) 608:546–52.
76. Achan J, Kakuru A, Wanzira H, Kyagulanyi T, Nuwa A, Magumba G, et al. Clinical presentation and outcomes of COVID-19in Uganda: a prospective cohort study. Lancet. (2022) 399:803–11. doi: 10.1016/S2666-5247(21)00240-8
77. Feit E, Gordon M, Alamuri TT, Hou W, Mitchell WB, Manwani D, et al. Long-term clinical outcomes and healthcare utilization of sickle cell disease patients with COVID-19: a 2.5-year follow-up study. Eur J Haematol. (2023) 111:636–43. doi: 10.1111/ejh.14058
78. Bodla P, Hashmi M, Niaz F, Auyeung AB, Oyetoran A, Khalil MJ, et al. Double trouble: COVID-19infection exacerbates sickle cell crisis outcomes in hospitalized patients—insights from National Inpatient Sample 2020. Haematol Rep. (2024) 16:421–30. doi: 10.3390/haematolrep16030041
79. Conran N and Belcher JD. Inflammation in sickle cell disease. Clin Haemorheol Microcirc. (2018) 68:263–99. doi: 10.3233/CH-189012
80. Jacobs L, Chiu K, Deng X, Stephens LD, Allen ES, Binns TC, et al. Hyperhaemolysis syndrome in patients with sickle cell disease and COVID-19. Br J Haematol. (2023) 200:94–7. doi: 10.1111/bjh.18132
81. Pelle MC, Zaffina I, Lucà S, Forte V, Trapanese V, Melina M, et al. Endothelial dysfunction in COVID-19: potential mechanisms and possible therapeutic options. Life (Basel). (2022) 12:1605. doi: 10.3390/life12101605
82. Allen EK, Penkert RR, Hankins JS, Surman SL, Van de Velde LA, Cotton A, et al. Immune cell profiles of patients with sickle cell disease during parvovirus B19–induced transient red cell aplasia. Vaccines (Basel). (2024) 12:984. doi: 10.3390/vaccines12090984
83. Dhawan M, Thakur N, Sharma M, and Rabaan AA. Comprehensive insights into the B-cell-mediated immune response against COVID-19infection amid the ongoing evolution of SARS-CoV-2. BioMed Pharmacother. (2025) 185:117936. doi: 10.1016/j.biopha.2025.117936
84. Chappey O, Wautier MP, Boval B, and Wautier JL. Endothelial cells in culture: an experimental model for the study of vascular dysfunctions. Cell Biol Toxicol. (1996) 12:199–205. doi: 10.1007/BF00438146
85. Croizat H and Nagel RL. Circulating cytokines response and the level of erythropoiesis in sickle cell anaemia. Am J Haematol. (1999) 60:105–15. doi: 10.1002/(SICI)1096-8652(199902)60:2<105::AID-AJH4>3.0.CO;2-Z
86. Santiago RP, Carvalho MOS, Figueiredo CVB, Fiuza LM, Oliveira RM, Yahouédéhou SCMA, et al. Associations between TGF-β1 levels and markers of haemolysis, inflammation, and tissue remodeling in pediatric sickle cell patients. Mediators Inflamm. (2021) 2021:4651891. doi: 10.1155/2021/4651891
87. Schnog JB, Rojer RA, Mac Gillavry MR, Ten Cate H, Brandjes DPM, and Duits AJ. Steady-state sVCAM-1 serum levels in adults with sickle cell disease. Ann Haematol. (2003) 82:109–13. doi: 10.1007/s00277-003-0609-1
88. Assis A, Conran N, Canalli AA, Lorand-Metze I, Saad ST, and Costa FF. Effect of cytokines and chaemokines on sickle neutrophil adhesion to fibronectin. Acta Haematol. (2005) 113:130–6. doi: 10.1159/000083451
89. Brittain JE and Parise LV. Cytokines and plasma factors in sickle cell disease. Curr Opin Haematol. (2007) 14:438–43. doi: 10.1097/MOH.0b013e3282a4a673
90. Paulson RF, Hariharan S, and Little JA. Stress erythropoiesis: definitions and models for its study. Exp Haematol. (2020) 89:43–54. doi: 10.1016/j.exphem.2020.07.011
91. Sesti-Costa R, Borges MD, Lanaro C, de Albuquerque DM, Saad STO, and Costa FF. Inflammatory dendritic cells contribute to regulate the immune response in sickle cell disease. Front Immunol. (2021) 11:617962. doi: 10.3389/fimmu.2020.617962
92. Gibbs KD, Schott BH, and Ko DC. The awesome power of human genetics of infectious disease. Annu Rev Genet. (2022) 56:41–62. doi: 10.1146/annurev-genet-080320-010449
93. Samuel CE. Interferon at the crossroads of SARS-CoV-2 infection and COVID-19disease. J Biol Chaem. (2023) 299:104960. doi: 10.1016/j.jbc.2023.104960
94. Pereira LRG, da Silva MVG, Germano CMR, Estevao IF, and Melo DG. Impact of the SARS-CoV-2 infection in individuals with sickle cell disease: an integrative review. Front Med. (2023) 10:1144226. doi: 10.3389/fmed.2023.1144226
95. Michelon I, Vilbert M, Pinheiro IS, Costa IL, Lorea CF, Castonguay M, et al. COVID-19 outcomes in patients with sickle cell disease and sickle cell trait compared with individuals without sickle cell disease or trait: a systematic review and meta-analysis. EClinicalMedicine. (2023) 66:102330. doi: 10.1016/j.eclinm.2023.102330
96. González-Ruiz FJ. Pharmacological and non-pharmacological strategies in coronavirus disease 2019: a literature review. Ann Med Surg (Lond). (2022) 77:103709. doi: 10.1016/j.amsu.2022.103709
97. Rabin J, Zhao Y, Mostafa E, Al-Suqi M, Fleischmann E, Conaway MR, et al. Regadenoson for the treatment of COVID-19: a five case clinical series and mouse studies. PloS One. (2023) 18:e0288920. doi: 10.1371/journal.pone.0288920
98. Jafari-Vayghan H, Rahimlou M, Almasi-Hashiani A, Aframehr E, and Saleh-Ghadimi S. Effect of the vitamin D supplementation on clinical outcomes and inflammatory status in COVID-19patients: an umbrella review. BMC Nutr. (2025) 11:86. doi: 10.1186/s40795-025-01060-y
99. Al-Handola R, Abdelkader K, Karrar A, Chinnappan J, and Rode G. Haemophagocytic lymphohistiocytosis: a rare complication of COVID-19 in a patient with sickle cell anaemia. Cureus. (2023) 15:e47631.
100. Martí-Carvajal A, Abd El Aziz MA, Martí-Amarista C, and Solà I. Antiplatelet agents for preventing vaso-occlusive events in people with sickle cell disease: a systematic review. Clin Adv Haematol Oncol. (2019) 17:234–43.
101. Federti E, Mattoscio D, Recchiuti A, Matte A, Monti M, Cozzolino F, et al. 17(R)-Resolvin D1 protects against sickle cell-related inflammatory cardiomyopathy in humanized mice. Blood. (2025) 45(17):1915–28. doi: 10.1182/blood.2024024768
102. Chowdhury FA, Colussi N, Sharma M, Wood KC, Xu JZ, Freeman BA, et al. Fatty acid nitroalkenes – multi-target agents for the treatment of sickle cell disease. Redox Biol. (2023) 68:102941. doi: 10.1016/j.redox.2023.102941
103. de Ligt LA, Gaartman AE, Konté K, Thakoerdin S, Fijnvandraat K, Kuijpers TW, et al. Plasma inflammatory and angiogenic protein profiling of patients with sickle cell disease. Br J Haematol. (2025) 206:954–64. doi: 10.1111/bjh.19970
104. Stivala S, Gobbato S, Bonetti N, Camici GG, Lüscher TF, and Beer JH. Dietary alpha-linolenic acid reduces platelet activation and collagen-mediated cell adhesion in sickle cell disease mice. J Thromb Haemost. (2022) 20:375–86. doi: 10.1111/jth.15581
105. Abdelhalim SM, Murphy JE, Meabed MH, Elberry AA, Gamaleldin MM, Shaalan MS, et al. Comparative effectiveness of adding Omega-3 or Vitamin D to standard therapy in preventing and treating episodes of painful crisis in pediatric sickle cell patients. Eur Rev Med Pharmacol Sci. (2022) 26:5043–52. doi: 10.26355/eurrev_202207_29290
106. Rarick KR, Li K, Teng RJ, Jing X, Martin DP, Xu H, et al. Sterile inflammation induces vasculopathy and chronic lung injury in murine sickle cell disease. Free Radic Biol Med. (2024) 215:112–26. doi: 10.1016/j.freeradbiomed.2024.01.052
107. Bolarinwa AB, Oduwole O, Okebe J, Ogbenna AA, Otokiti OE, and Olatinwo AT. Antioxidant supplementation for sickle cell disease. Cochrane Database Syst Rev. (2024) 5:CD013590. doi: 10.1002/14651858.CD013590.
108. Sanduzzi Zamparelli S, Sanduzzi Zamparelli A, and Bocchino M. Immune-boosting and antiviral effects of antioxidants in COVID-19pneumonia: a therapeutic perspective. Life (Basel). (2025) 15:113.
109. Hong L, Chen X, Liu Y, Liang H, Zhao Y, and Guo P. The relationship between ferroptosis and respiratory infectious diseases: a novel landscape for therapeutic approach. Front Immunol. (2025) 16:1550968. doi: 10.3389/fimmu.2025.1550968
110. Elmahdi M, Fadalla T, Suliman M, Elsayed M, Awad Elhaj AM, and Hussein H. Moyamoya syndrome and stroke among paediatric sickle cell disease patients in Sudan: a cross-sectional study. Ann Med Surg (Lond). (2022) 78:103815. doi: 10.1016/j.amsu.2022.103815
111. Menon AV, Liu J, Tsai HP, Zeng L, Yang S, Asnani A, et al. Excess haeme upregulates haeme oxygenase 1 and promotes cardiac ferroptosis in mice with sickle cell disease. Blood. (2022) 139:936–41. doi: 10.1182/blood.2020008455
112. Russo A, Patanè GT, Calderaro A, Barreca D, Tellone E, and Putaggio S. Crosstalk between sickle cell disease and ferroptosis. Int J Mol Sci. (2025) 26:3675. doi: 10.3390/ijms26083675
113. Yang L, Wu Y, Jin W, Mo N, Ye G, Su Z, et al. The potential role of ferroptosis in COVID-19-related cardiovascular injury. BioMed Pharmacother. (2023) 168:115637. doi: 10.1016/j.biopha.2023.115637
114. Jankauskas SS, Kansakar U, Sardu C, Varzideh F, Avvisato R, Wang X, et al. COVID-19causes ferroptosis and oxidative stress in human endothelial cells. Antioxidants (Basel). (2023) 12:326. doi: 10.3390/antiox12020326
115. Xi C, Pang J, Xue W, Cui Y, Jiang N, Zhi W, et al. Transsulfuration pathway activation attenuates oxidative stress and ferroptosis in sickle primary erythroblasts and transgenic mice. Commun Biol. (2025) 8:15. doi: 10.1038/s42003-024-07424-7
116. Koklesova L, Mazurakova A, Samec M, Biringer K, Samuel SM, Büsselberg D, et al. Homocysteine metabolism as the target for predictive medical approach, disease prevention, prognosis, and treatments tailored to the person. EPMA J. (2021) 12:477–505. doi: 10.1007/s13167-021-00263-0
117. Haroun E, Lim SH, and Dutta D. GBT1118, a voxelotor analog, ameliorates hepatopathy in sickle cell disease. Medicina (Kaunas). (2024) 60:1581. doi: 10.3390/medicina60101581
118. Salama SA, Abdel-Bakky MS, and Mohamed AA. Upregulation of Nrf2 signaling and suppression of ferroptosis and NF-κB pathway by leonurine attenuate iron overload-induced hepatotoxicity. Chaem Biol Interact. (2022) 356:109875. doi: 10.1016/j.cbi.2022.109875
119. Sun S, Shen J, Jiang J, Wang F, and Min J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther. (2023) 8:372. doi: 10.1038/s41392-023-01606-1
120. Li C, Yin X, Liu Z, and Wang J. Emerging potential mechanism and therapeutic target of ferroptosis in PDAC: a promising future. Int J Mol Sci. (2022) 23:15031. doi: 10.3390/ijms232315031
121. Zhang L, Luo YL, Xiang Y, Bai XY, Qiang RR, Zhang X, et al. Ferroptosis inhibitors: past, present and future. Front Pharmacol. (2024) 15:1407335. doi: 10.3389/fphar.2024.1407335
122. Rayman MP, Taylor EW, and Zhang J. The relevance of selenium to viral disease with special reference to SARS-CoV-2 and COVID-19. Proc Nutr Soc. (2023) 82:1–12. doi: 10.1017/S0029665122002646
123. Zhou J, Ni W, Zhang X, Yang M, Liu X, Guo J, et al. Visual analysis on ferroptosis and its crosstalk to coronavirus disease 2019 (COVID-19). Heliyon. (2024) 10:e37617.
124. Yuan S, Liu Z, Xu Z, Liu J, and Zhang J. High mobility group box 1 (HMGB1): a pivotal regulator of haematopoietic Malignancies. J Haematol Oncol. (2020) 13:91. doi: 10.1186/s13045-020-00920-3
125. Nolfi-Donegan D, Annarapu GK, St Croix C, Calderon M, Hillery CA, and Shiva S. High-mobility group box 1 increases platelet surface P2Y12 and platelet activation in sickle cell disease. JCI Insight. (2024) 9:e174575. doi: 10.1172/jci.insight.174575
126. Ahmed B, Arigliani M, and Gupta A. Respiratory management of acute chest syndrome in children with sickle cell disease. Eur Respir Rev. (2024) 33:240005. doi: 10.1183/16000617.0005-2024
127. Indolfi C, Dinardo G, Grella C, Klain A, Perrotta A, Mondillo G, et al. Exploring the interplay between asthma and haemoglobinopathies: a comprehensive review. J Clin Med. (2024) 13:3263. doi: 10.3390/jcm13113263
128. Win N. Hyperhaemolysis syndrome in sickle cell disease. Expert Rev Haematol. (2009) 2:111–5. doi: 10.1586/ehm.09.2
129. Banks M and Shikle J. Hyperhaemolysis syndrome in patients with sickle cell disease. Arch Pathol Lab Med. (2018) 142:1425–7. doi: 10.5858/arpa.2017-0251-RS
130. Adkins BD, Kaylani SZ, Nero A, Ibrahim IF, Shen YM, and Sarode R. How do we manage hyperhaemolysis syndrome. Transfusion. (2024) 64:1822–9. doi: 10.1111/trf.17995
131. Rehman R, Saadat SB, Tran DH, Constantinescu S, and Qamruzzaman Y. Recurrent hyperhaemolysis syndrome in sickle cell disease. Cureus. (2021) 13:e14991. doi: 10.7759/cureus.14991
132. Yu J, Gerber GF, Chen H, Yuan X, Chaturvedi S, Braunstein EM, et al. Complement dysregulation is associated with severe COVID-19illness. Haematologica. (2022) 107:1095–105. doi: 10.3324/haematol.2021.279155
133. Baillie K, Davies HE, Keat SBK, Ladell K, Miners KL, Jones SA, et al. Complement dysregulation is a prevalent and therapeutically amenable feature of long COVID-19-19-19. Med (N Y). (2024) 5:239–53.e5. doi: 10.1016/j.medj.2024.01.011
134. Gebetsberger L, Malekshahi Z, Teutsch A, Tajti G, Fontaine F, Marella N, et al. SARS-CoV-2 hijacks host CD55, CD59 and factor H to impair antibody-dependent complement-mediated lysis. Emerg Microbes Infect. (2024) 13:2417868. doi: 10.1080/22221751.2024.2417868
135. Vellanki S, Thalambedu N, Kumar AKTA, Vellanki S, Honhar M, Hendrix R, et al. Navigating hyperhemolysis in sickle cell disease: insights from literature. Diagnostics (Basel). (2025) 15(14):1835. doi: 10.3390/diagnostics15141835
136. Perico L, Benigni A, and Remuzzi G. SARS-CoV-2 and the spike protein in endotheliopathy. Trends Microbiol. (2024) 32:53–67. doi: 10.1016/j.tim.2023.06.004
137. Xiao MT, Ellsworth CR, and Qin X. Emerging role of complement in COVID-19and other respiratory virus diseases. Cell Mol Life Sci. (2024) 81:94. doi: 10.1007/s00018-024-05157-8
138. Meuleman MS, Roumenina LT, and Grunenwald A. Complement involvement in sickle cell disease. Presse Med. (2023) 52:104205. doi: 10.1016/j.lpm.2023.104205
139. Chen S, Saeed AF, Liu Q, Jiang Q, Xu H, Xiao GG, et al. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther. (2023) 8:207. doi: 10.1038/s41392-023-01452-1
140. Navaneethabalakrishnan S, An X, and Vinchi F. Haeme- and iron-activated macrophages in sickle cell disease: an updated perspective. Curr Opin Haematol. (2024) 31:275–84. doi: 10.1097/MOH.0000000000000836
141. Madu AJ, Ugwu AO, and Efobi C. Hyperhaemolytic syndrome in sickle cell disease: clearing the cobwebs. Med Princ Pract. (2021) 30:236–43. doi: 10.1159/000512945
142. Fuja J, Vargas I, Wells J, Singh S, Mansfield P, and Wool GD. Hyperhaemolysis syndrome post-COVID-19in a patient with sickle cell disease: successful treatment with tocilizumab. Br J Haematol. (2022) 196:e34–7. doi: 10.1111/trf.16932
143. Desai N, Peters J, Davies E, and Sharif J. The role of tocilizumab in the treatment of post-transfusion hyperhaemolysis. EJHaem. (2023) 4:1096–9. doi: 10.1002/jha2.779
144. Bartleson JM, Radenkovic D, Covarrubias AJ, Furman D, Winer DA, and Verdin E. SARS-CoV-2, COVID-19and the aging immune system. Nat Aging. (2021) 1:769–82. doi: 10.1038/s43587-021-00114-7
145. Mei K, Anand S, Fung M, Hwang SH, Rodberg K, Quon D, et al. Use of HBOC-201 in refractory hyperhaemolysis syndrome: a case series. Transfus Med. (2025) 35:22–7. doi: 10.1097/MJT.0000000000001777
146. Martí-Carvajal AJ, Anand V, Cardona AF, and Solà I. Eculizumab is used to treat patients with paroxysmal nocturnal haemoglobinuria. Cochrane Database Syst Rev. (2014) 30(10):CD010340. doi: 10.1002/14651858.CD010340.pub2
147. El-Beshlawy A and El-Ghamrawy M. Recent trends in treatment of thalassemia. Blood Cells Mol Dis. (2019) 76:53–8. doi: 10.1016/j.bcmd.2019.01.006
148. Song Y, Li X, He X, Zhou F, Du F, Wang Z, et al. Dose-escalating ruxolitinib for refractory haemophagocytic lymphohistiocytosis. Front Immunol. (2023) 14:1211655. doi: 10.3389/fimmu.2023.1211655
149. Huarte E, Peel MT, Verbist K, Fay BL, Bassett R, Albeituni S, et al. Ruxolitinib, a JAK1/2 inhibitor, ameliorates cytokine storm in experimental models of hyperinflammation syndrome. Front Pharmacol. (2021) 12:650295. doi: 10.3389/fphar.2021.650295
150. Wang Q and Zennadi R. The role of RBC oxidative stress in sickle cell disease: from the molecular basis to pathologic implications. Antioxidants (Basel). (2021) 10:1608. doi: 10.3390/antiox10101608
151. Nur E, Brandjes DP, Teerlink T, Otten HM, Oude Elferink RP, Muskiet F, et al. N-acetylcysteine reduces oxidative stress in sickle cell patients. Ann Haematol. (2012) 91:1097–105. doi: 10.1007/s00277-011-1404-z
152. Alam MS, Hasan MN, Maowa Z, Khatun F, Nazir KHMNH, and Alam MZ. N-acetylcysteine reduces severity and mortality in COVID-19patients: a systematic review and meta-analysis. J Adv Vet Anim Res. (2023) 10:157–68. doi: 10.5455/javar.2023.j665
153. Mokra D, Porvaznik I, and Mokry J. N-acetylcysteine in the treatment of acute lung injury: perspectives and limitations. Int J Mol Sci. (2025) 26:2657. doi: 10.3390/ijms26062657
154. Piccin A, O’Connor-Byrne N, Daves M, Lynch K, Farshbaf AD, and Martin-Loeches I. Autoimmune disease and sickle cell anaemia: intersecting pathways and differential diagnosis. Br J Haematol. (2022) 197:518–28. doi: 10.1111/bjh.18109
155. Garmendia JV, García AH, De Sanctis CV, Hajdúch M, and De Sanctis JB. Autoimmunity and immunodeficiency in severe SARS-CoV-2 infection and prolonged COVID-19. Curr Issues Mol Biol. (2022) 45:33–50. doi: 10.3390/cimb45010003
156. Canny SP, Orozco SL, Thulin NK, and Hamerman JA. Immune mechanisms in inflammatory anaemia. Annu Rev Immunol. (2023) 41:405–29. doi: 10.1146/annurev-immunol-101320-125839
157. Soares VR, Blanco BP, Dinardo CL, and Garanito MP. Coexistent sickle cell anaemia and autoimmune haemolytic anaemia in two adolescents. Einstein (Sao Paulo). (2024) 22:eRC1105. doi: 10.31744/einstein_journal/2024RC1105
158. O’Toole G, Swan D, Connors JM, and Thachil J. Haematological causes of acute ischaemic stroke in younger individuals. J Thromb Haemost. (2025) 23:11–22. doi: 10.1016/j.jtha.2024.09.025
159. Lucchini E, Fanin R, Cooper N, and Zaja F. Management of immune thrombocytopenia in elderly patients. Eur J Intern Med. (2018) 58:70–6. doi: 10.1016/j.ejim.2018.09.005
160. Swinkels M, Rijkers M, Voorberg J, Vidarsson G, Leebeek FWG, and Jansen AJG. Emerging concepts in immune thrombocytopenia. Front Immunol. (2018) 9:880. doi: 10.3389/fimmu.2018.00880
161. Martínez-Carballeira D, Bernardo Á, Caro A, Soto I, and Gutiérrez L. Pathophysiology, clinical manifestations and diagnosis of immune thrombocytopenia: contextualization from a historical perspective. Haematol Rep. (2024) 16:204–19. doi: 10.3390/hematolrep16020021
162. Li W, Pucka AQ, Debats C, Reyes BA, Syed F, O’Brien ARW, et al. Inflammation and autoimmunity are interrelated in patients with sickle cell disease at a steady-state condition: implications for vaso-occlusive crisis, pain, and sensory sensitivity. Front Immunol. (2024) 15:1288187. doi: 10.3389/fimmu.2024.1288187
163. Kakavandi S, Hajikhani B, Azizi P, Aziziyan F, Nabi-Afjadi M, Farani MR, et al. COVID-19in patients with anaemia and haematological Malignancies: risk factors, clinical guidelines, and emerging therapeutic approaches. Cell Commun Signal. (2024) 22:126. doi: 10.1186/s12964-023-01316-9
164. Alanssari MA, Al-Sulaitti E, Al-Sulaitti Z, Khalaf AA, and Dawood QM. Successful management of severe and refractory autoimmune haemolytic anaemia in a sickle cell disease patient with bortezomib. Cureus. (2024) 16:e74840. doi: 10.7759/cureus.74840
165. Alkhars AAA, Alkadi MA, Alwabari HM, Mandong J, and Almakinah ZY. Sickle cell disease: A potential cause of immune thrombocytopenia. Cureus. (2024) 16:e76299. doi: 10.7759/cureus.76299
166. Floch A, Viret S, Malard L, Pakdaman S, Jouard A, Habibi A, et al. Eleven years of alloimmunisation in 6496 patients with sickle cell disease in France who received transfusions. Blood Adv. (2023) 7:7608–20. doi: 10.1182/bloodadvances.2022009328
167. Zelek WM and Harrison RA. Complement and COVID-19: Three years on, what we know, what we don’t know, and what we ought to know. Immunobiology. (2023) 228:152393. doi: 10.1016/j.imbio.2023.152393
168. Avdonin PP, Blinova MS, Serkova AA, Komleva LA, and Avdonin PV. Immunity and coagulation in COVID-19. Int J Mol Sci. (2024) 25:11267. doi: 10.3390/ijms252011267
169. Christian J, Lanzkron S, and Naik RP. COVID-19outcomes in sickle cell disease and sickle cell trait. Best Pract Res Clin Haematol. (2022) 35:101382. doi: 10.1016/j.beha.2022.101382
170. Bao W, Zhong H, Manwani D, Vasovic L, Uehlinger J, Lee MT, et al. Regulatory B-cell compartment in transfused alloimmunized and non-alloimmunized patients with sickle cell disease. Am J Haematol. (2013) 88:736–40. doi: 10.1002/ajh.23488
171. ElAlfy MS, Adly AAM, Ebeid FSE, Eissa DS, Ismail EAR, Mohammed YH, et al. Immunological role of CD4+CD28null T lymphocytes, natural killer cells, and interferon-gamma in pediatric patients with sickle cell disease: relation to disease severity and response to therapy. Immunol Res. (2018) 66:480–90. doi: 10.1007/s12026-018-9010-y
172. Abraham AA, Lang H, Meier ER, Nickel RS, Dean M, Lawal N, et al. Characterization of natural killer cells expressing markers associated with maturity and cytotoxicity in children and young adults with sickle cell disease. Pediatr Blood Cancer. (2019) 66:e27601. doi: 10.1002/pbc.27601
173. Fazeli P, Kalani M, and Hosseini M. T memory stem cell characteristics in autoimmune diseases and their promising therapeutic values. Front Immunol. (2023) 14:1204231. doi: 10.3389/fimmu.2023.1204231
174. Pirenne F and Pondarré C. Alloimmunisation and hyperhaemolysis in sickle cell disease. Haematology Am Soc Haematol Educ Program. (2023) 2023:653–9. doi: 10.1182/hematology.2023000499
175. De Luna G, Habibi A, Odièvre MH, Guillet H, Guiraud V, Cougoul P, et al. Blood exchange transfusion with dexamethasone and Tocilizumab for management of hospitalised patients with sickle cell disease and severe COVID-19: Preliminary evaluation of a novel algorithm. Am J Haematol. (2022) 97:E260–4. doi: 10.1002/ajh.26563
176. Lapite A, Sánchez LM, Altaffer AL, Rae M, Ramirez AA, Muscal E, et al. Concomitant sickle cell disease and systemic lupus erythaematosus: A single-centre case series. Pediatr Blood Cancer. (2024) 71:e31194. doi: 10.1002/pbc.31194
177. Boulassel MR, Al-Naamani A, Al-Zubaidi A, Al-Qarni Z, Khan H, Oukil A, et al. Coexistence of sickle cell disease and systemic lupus erythaematosus is associated with quantitative and qualitative impairments in circulating regulatory B cells. Hum Immunol. (2022) 83:818–25. doi: 10.1016/j.humimm.2022.09.005
178. Hendrickson JE, McKenzie TJ, and Saini N. SARS-CoV-2 infection as a trigger for alloimmunization and autoimmunity. Transfusion. (2021) 61:3135–42. doi: 10.1111/cts.12953
179. Wong K, Lai WK, and Jackson DE. HLA Class II regulation of immune response in sickle cell disease patients: susceptibility to red blood cell alloimmunisation (systematic review and meta-analysis). Vox Sang. (2022) 117:1251–61. doi: 10.1111/vox.13351
180. Poutrel S, Boisson C, Nader E, Renoux C, Virot E, Catella J, et al. Clinical severity and blood rheology in patients with sickle cell anaemia and co-existing autoimmune disease. Br J Haematol. (2023) 200:e28–31. doi: 10.1111/bjh.18624
181. Lewis SM, Williams A, and Eisenbarth SC. Structure and function of the immune system in the spleen. Sci Immunol. (2019) 4:eaau6085. doi: 10.1126/sciimmunol.aau6085
182. Brousse V, Buffet P, and Rees D. The spleen and sickle cell disease: Pathophysiology and therapeutic approaches. Br J Haematol. (2014) 166:165–76. doi: 10.1111/bjh.12950
183. El Hoss S and Brousse V. The spleen in sickle cell disease: Physiology, pathophysiology and therapeutic approaches. Expert Rev Haematol. (2019) 12:563–73. doi: 10.1080/17474086.2019.1627192
184. Ihlow J, Michaelis E, Greuel S, Izci-Hunsucker S, Ramadan K, Tschoeke EC, et al. B-cell depletion and signs of sepsis-acquired immunodeficiency in bone marrow and spleen of COVID-19deceased. Int J Infect Dis. (2021) 103:628–35. doi: 10.1016/j.ijid.2020.12.078
185. Rossi CM, Lenti MV, Merli S, Abbati G, Arpa M, Silvestri M, et al. Role of IgM memory B cells and spleen function in COVID-19. Front Immunol. (2022) 13:889876. doi: 10.3389/fimmu.2022.889876
186. Qiang Y, Adeyemo A, Song A, Conde JN, Gooden D, Ruiz-Ortega M, et al. Mechanisms of thromboinflammation in sickle cell disease. Proc Natl Acad Sci U S A. (2023) 120:e2217607120. doi: 10.1073/pnas.2217607120
187. Yurdaisik I, Demiroz AS, Oz AB, Karaarslan E, Guncu G, and Karaarslan B. Postmortem biopsies of the lung, heart, liver, and spleen of COVID-19patients. Cureus. (2021) 13:e20734. doi: 10.7759/cureus.20734
188. Ping H, Zhang K, Wang Y, Zhang Z, Chen X, He W, et al. Cell death and pathological findings of the spleen in COVID-19patients. Pathol Res Pract. (2021) 227:153610. doi: 10.1016/j.prp.2021.153610
189. Aarabi S and Karimialavijeh E. Spleen infarction: A delayed complication of COVID-19infection. Vis J Emerg Med. (2021) 25:101120. doi: 10.1016/j.visj.2021.101120
190. Saad AA, Beshlawi IO, Al-Rawas AH, Zachariah M, Nazir HF, and Wali Y. Acute splenic sequestration crisis in sickle cell disease: A review. Oman Med J. (2017) 32:425–8. doi: 10.5001/omj.2017.79
191. Nakayasu H, Sakurai S, Sugiyama S, Ozaki K, Sawa M, Koyama T, et al. The liver-to-spleen ratio as a predictor of oxygen demand in COVID-19patients. Infect Med (Beijing). (2023) 2:105–11. doi: 10.1016/j.imj.2023.04.002
192. Trabulsi NH, Alshammakh SS, Shabkah AA, Afifi AM, Hsieh TC, and Alkhalaf HH. Spontaneous rupture of spleen in a patient with COVID-19disease: Case report and literature review. J Surg Case Rep. (2022) 2022:rjac124. doi: 10.1093/jscr/rjac124
193. Minniti CP, Zaidi AU, Nouraie M, Manwani D, Crouch GD, Crouch AS, et al. Clinical predictors of poor outcomes in patients with sickle cell disease and COVID-19infection. Blood Adv. (2021) 5:207–15. doi: 10.1182/bloodadvances.2020003456
194. Ladu AI, Aiyenigba AO, Adekile A, and Bates I. The spectrum of splenic complications in patients with sickle cell disease in Africa: A systematic review. Br J Haematol. (2021) 193:26–42. doi: 10.1111/bjh.17179
195. Tubman VN and Makani J. Turf wars: exploring splenomegaly in sickle cell disease in malaria-endemic regions. Br J Haematol. (2017) 177:938–46. doi: 10.1111/bjh.14592
196. Joice R, Frantzreb C, Pradham A, Seydel KB, Kamiza S, Wirth DF, et al. Evidence for spleen dysfunction in malaria-HIV co-infection in a subset of pediatric patients. Mod Pathol. (2016) 29:381–90. doi: 10.1038/modpathol.2016.27
197. Lenti MV, Corazza GR, and Di Sabatino A. COVID-19and asplenia: a Janus-faced issue. Intern Emerg Med. (2021) 16:2341–2. doi: 10.1007/s11739-021-02761-5
198. Bonanni P, Grazzini M, Niccolai G, Paolini D, Varone O, Bartoloni A, et al. Recommended vaccinations for asplenic and hyposplenic adult patients. Hum Vaccin Immunother. (2017) 13:359–68. doi: 10.1080/21645515.2017.1264797
199. Naseer ZA, Bachabi M, Jones LC, Sterling RS, and Khanuja HS. Osteonecrosis in sickle cell disease. South Med J. (2016) 109:525–30. doi: 10.14423/SMJ.0000000000000516
200. Colella MP, de Paula EV, MaChado-Neto JA, Conran N, Annichino-Bizzacchi JM, Costa FF, et al. Elevated hypercoagulability markers in haemoglobin SC disease. Haematologica. (2015) 100:466–71. doi: 10.3324/haematol.2014.114587
201. Daltro PB, Ribeiro TO, Daltro GC, Meyer RJ, and Fortuna V. CD4(+) T cell profile and activation response in sickle cell disease patients with osteonecrosis. Mediators Inflamm. (2020) 2020:1747894. doi: 10.1155/2020/1747894
202. Rêgo MJ, da Silva RR, Pereira MC, da Silva Araújo A, Pitta Ida R, Falcão DA, et al. Evaluation of CD4(+)CD25(+)FoxP3(+) T cell populations, IL-10 production, and their correlation with clinical and biochaemical parameters in sickle cell anaemia patients with leg ulcers. Cytokine. (2015) 75:310–5. doi: 10.1016/j.cyto.2015.07.016
203. Baldwin C, Nolan VG, Wyszynski DF, Ma QL, Sebastiani P, Embury SH, et al. Association of klotho, bone morphogenic protein 6, and annexin A2 polymorphisms with sickle cell osteonecrosis. Blood. (2005) 106:372–5. doi: 10.1182/blood-2005-02-0548
204. Tozatto-Maio K, Girot R, Ly ID, Silva Pinto AC, Rocha V, Fernandes F, et al. Polymorphisms in inflammatory genes modulate clinical complications in patients with sickle cell disease. Front Immunol. (2020) 11:2041. doi: 10.3389/fimmu.2020.02041
205. Farawela HM, El-Ghamrawy M, Farhan MS, Soliman R, Yousry SM, and AbdelRahman HA. Association between Duffy antigen receptor expression and disease severity in sickle cell patients. Haematology. (2016) 21:474–9. doi: 10.1080/10245332.2015.1111643
206. Drasar ER, Menzel S, Fulford T, and Thein SL. The effect of Duffy antigen receptor for chaemokines on severity in sickle cell disease. Haematologica. (2013) 98:e87–9. doi: 10.3324/haematol.2013.089243
207. Sinha PR, Mallick N, and Sahu RL. Avascular necrosis of the hip after the COVID-19pandemic. J Pharm Bioallied Sci. (2023) 15:S661–4. doi: 10.4103/jpbs.jpbs_87_23
208. Ghadri JR, Wittstein IS, Prasad A, Sharkey S, Dote K, Akashi YJ, et al. International expert consensus document on Takotsubo syndrome. Eur Heart J. (2018) 39:2032–46. doi: 10.1093/eurheartj/ehy076
209. Taylor SM, Parobek CM, and Fairhurst RM. Haemoglobinopathies and the clinical epidemiology of malaria: a systematic review and meta-analysis. Lancet Infect Dis. (2012) 12:457–68. doi: 10.1016/S1473-3099(12)70055-5
210. Sato H, Tateishi H, and Uchida T. Takotsubo-type cardiomyopathy due to multivessel spasm. In: Kodama K, Haze K, and Hon M, editors. Clinical Aspect of Myocardial Injury: From Ischaemia to Heart Failure. Kagakuhyoronsha Publishing Co, Tokyo (1990). p. 56–64.
211. Pelliccia F, Kaski JC, Crea F, Rahman K, Lerakis S, Reddy VY, et al. Takotsubo syndrome in COVID-19. J Am Coll Cardiol. (2020) 76:628–31. doi: 10.1016/j.jacc.2020.05.068
212. Wittstein IS, Thiemann DR, Lima JA, Baughman KL, Schulman SP, Gerstenblith G, et al. Neurohumoral features of myocardial stunning due to sudden emotional stress. N Engl J Med. (2005) 352:539–48. doi: 10.1056/NEJMoa043046
213. Darbari DS, Sheehan VA, and Ballas SK. The vaso-occlusive pain crisis in sickle cell disease: definition, pathophysiology, and management. Eur J Haematol. (2020) 105:237–46. doi: 10.1111/ejh.13430
214. Lyon AR, Rees PS, Prasad S, Poole-Wilson PA, and Harding SE. Stress (Takotsubo) cardiomyopathy—a novel pathophysiological hypothesis to explain catecholamine-induced acute myocardial stunning. Nat Clin Pract Cardiovasc Med. (2008) 5:22–9. doi: 10.1038/ncpcardio1066
215. Templin C, Ghadri JR, Diekmann J, Napp LC, Bataiosu DR, Jaguszewski M, et al. Clinical features and outcomes of Takotsubo (stress) cardiomyopathy. N Engl J Med. (2015) 373:929–38. doi: 10.1056/NEJMoa1406761
216. Gladwin MT and Sachdev V. Cardiovascular abnormalities in sickle cell disease. J Am Coll Cardiol. (2012) 59:1123–33. doi: 10.1016/j.jacc.2011.10.900
217. Bonow RO, Fonarow GC, O’Gara PT, and Yancy CW. Association of Coronavirus Disease 2019 (COVID-19) with myocardial injury and mortality. JAMA Cardiol. (2020) 5:751–3. doi: 10.1001/jamacardio.2020.1105
218. Singh S, Desai R, Gandhi Z, Fong HK, Doreswamy S, Desai V, et al. Takotsubo syndrome in patients with COVID-19: a systematic review of published cases. SN Compr Clin Med. (2020) 2:2102–8. doi: 10.1007/s42399-020-00557-w
219. Ueyama T, Hano T, Kasamatsu K, Yamamoto K, Tsuruo Y, and Nishio I. Estrogen attenuates the emotional stress-induced cardiac responses in the animal model of Tako-tsubo (Ampulla) cardiomyopathy. J Cardiovasc Pharmacol. (2003) 42:S117–9. doi: 10.1097/00005344-200312001-00024
220. DeBaun MR, Ghafuri DL, Rodeghier M, Maitra P, Chaturvedi S, Kassim A, et al. Decreased median survival of adults with sickle cell disease after adjusting for left truncation bias: a pooled analysis. Blood. (2019) 133:615–7. doi: 10.1182/blood-2018-10-880575
221. Burnstock G. Purinergic signalling: therapeutic developments. Front Pharmacol. (2017) 8:661. doi: 10.3389/fphar.2017.00661
222. Antonioli L, Blandizzi C, Pacher P, Safari E, and Safari E. The purinergic system in COVID-19: potential therapeutic implications. Br J Pharmacol. (2021) 178:2073–84. doi: 10.1007/s10787-023-01344-4
223. Di Virgilio F, Sarti AC, and Coutinho-Silva R. Purinergic signaling, DAMPs, and inflammation. Am J Physiol Cell Physiol. (2020) 318:C832–5. doi: 10.1152/ajpcell.00053.2020
224. Burnstock G. Purinergic nerves. Pharmacol Rev. (1972) 24:509–81. doi: 10.1016/S0031-6997(25)06930-3
225. Burnstock G. Blood cells: an historical account of the roles of purinergic signalling. Purinergic Signal. (2015) 11:411–34. doi: 10.1007/s11302-015-9462-7
226. Jacobson KA, Tosh DK, Jain S, and Gao ZG. Historical and current adenosine receptor agonists in preclinical and clinical development. Front Cell Neurosci. (2019) 13:124. doi: 10.3389/fncel.2019.00124
227. Zhang Y, Dai Y, Wen J, Zhang W, Grenz A, Sun H, et al. Detrimental effects of adenosine signaling in sickle cell disease. Nat Med. (2011) 17:79–86. doi: 10.1038/nm.2280
228. Vandorpe DH, Rivera A, Ganter M, Dankwa S, Wohlgemuth JG, Dlott JS, et al. Purinergic signaling is essential for full Psickle activation by hypoxia and by normoxic acid pH in mature human sickle red cells and in vitro-differentiated cultured human sickle reticulocytes. Pflugers Arch. (2022) 474:553–65. doi: 10.1007/s00424-022-02665-z
229. Ghadri JR, Cammann VL, Jurisic S, Seifert B, Napp LC, Diekmann J, et al. A novel clinical score (InterTAK Diagnostic Score) to differentiate Takotsubo syndrome from acute coronary syndrome: results from the International Takotsubo Registry. Eur J Heart Fail. (2017) 19:1036–42. doi: 10.1002/ejhf.683
230. Piel FB, Steinberg MH, and Rees DC. Sickle cell disease. N Engl J Med. (2017) 376:1561–73. doi: 10.1056/NEJMra1510865
231. Hussain FA, Njoku FU, Saraf SL, Molokie RE, Gordeuk VR, and Han J. COVID-19infection in patients with sickle cell disease. Br J Haematol. (2020) 189:851–2. doi: 10.1111/bjh.16734
232. Klok FA, Kruip MJHA, van der Meer NJM, Arbous MS, Gommers D, Kant KM, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. (2020) 191:145–7. doi: 10.1016/j.thromres.2020.04.013
233. Nelson SC and Hackman HW. Sickle cell disease, COVID-19 and the risks of structural racism. Blood Adv. (2020) 4:4113–5. doi: 10.1016/j.hoc.2022.08.008
234. Makani J, Ofori-Acquah SF, Nnodu O, Wonkam A, and Ohene-Frempong K. Sickle cell disease: new opportunities and challenges in Africa. ScientificWorldJournal. (2013) 2013:193252. doi: 10.1155/2013/193252
235. Williams TN and Weatherall DJ. World distribution, population genetics, and health burden of the haemoglobinopathies. Cold Spring Harb Perspect Med. (2012) 2:a011692. doi: 10.1101/cshperspect.a011692
236. Idro R, Marsh K, John CC, and Newton CRJC. Cerebral malaria: mechanisms of brain injury and strategies for improved neurocognitive outcome. Pediatr Res. (2010) 68:267–74. doi: 10.1203/PDR.0b013e3181eee738
237. Clark IA, Alleva LM, Mills AC, and Cowden WB. Pathogenesis of malaria and clinically similar conditions. Clin Microbiol Rev. (2004) 17:509–39. doi: 10.1128/CMR.17.3.509-539.2004
238. Marsh K, Forster D, Waruiru C, Mwangi I, Winstanley M, Marsh V, et al. Indicators of life-threatening malaria in African children. N Engl J Med. (1995) 332:1399–404. doi: 10.1056/NEJM199505253322102
239. McGann PT, Hernandez AG, and Ware RE. Sickle cell anaemia in sub-Saharan Africa: advancing the clinical paradigm through partnerships and research. Blood. (2017) 129:155–61. doi: 10.1182/blood-2016-09-702324
240. Makani J, Cox SE, Soka D, Komba AN, Oruo J, Mwamtemi H, et al. Mortality in sickle cell anaemia in Africa: a prospective cohort study in Tanzania. PloS One. (2011) 6:e14699. doi: 10.1371/journal.pone.0014699
241. Nader E, Romana M, and Connes P. The red blood cell-inflammation vicious circle in sickle cell disease. Front Immunol. (2020) 11:454. doi: 10.3389/fimmu.2020.00454
242. Nader E, Conran N, Romana M, Hedreville M, Etienne-Julan M, and Connes P. Vasculopathy in sickle cell disease: from red blood cell sickling to vascular dysfunction. Compr Physiol. (2021) 11:1785–803. doi: 10.1002/j.2040-4603.2021.tb00164.x
243. Bybee KA, Kara T, Prasad A, Lerman A, Barsness GW, Wright RS, et al. Systematic review: transient left ventricular apical ballooning: a syndrome that mimics ST-segment elevation myocardial infarction. Ann Intern Med. (2004) 141:858–65. doi: 10.7326/0003-4819-141-11-200412070-00010
244. Lyon AR, Bossone E, Schneider B, Sechtem U, Citro R, Underwood SR, et al. Current state of knowledge on Takotsubo syndrome: a Position Statement from the Taskforce on Takotsubo Syndrome of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail. (2016) 18:8–27. doi: 10.1002/ejhf.424
245. Howard J, Hart N, Roberts-Harewood M, Cummins M, Awogbade M, and Davis B. Guideline on the management of acute chest syndrome in sickle cell disease. Br J Haematol. (2015) 169:492–505. doi: 10.1111/bjh.13348
246. Anie KA and Green J. Psychological therapies for sickle cell disease and pain. Cochrane Database Syst Rev. (2015), CD001916. doi: 10.1002/14651858.CD001916.pub3
247. Bulgin D, Tanabe P, and Jenerette C. Stigma of sickle cell disease: A systematic review. Issues Ment Health Nurs. (2018) 39:675–86. doi: 10.1080/01612840.2018.1443530
248. Koutroumpakis E, Taylor T, Damaraju S, and Mawji SB. COVID-19-19-19subo”: stress-induced cardiomyopathy by novel coronavirus disease 2019. Case Rep Cardiol. (2020) 2020:779–83. doi: 10.1159/000511450
249. Minhas AS, Hughey AB, and Kolias TJ. Nationwide trends in reported incidence of takotsubo cardiomyopathy from 2006 to 2012. Am J Cardiol. (2015) 116:1128–31. doi: 10.1016/j.amjcard.2015.06.042
251. Akashi YJ, Nef HM, and Lyon AR. Epidemiology and pathophysiology of Takotsubo syndrome. Nat Rev Cardiol. (2015) 12:387–97. doi: 10.1038/nrcardio.2015.39
252. Burton GJ and Fowden AL. The placenta: a multifaceted, transient organ. Philos Trans R Soc Lond B Biol Sci. (2015) 370:20140066. doi: 10.1098/rstb.2014.0066
253. Gil GP, Ananina G, Maschietto M, Lima SCS, da Silva Costa SM, Baptista LC, et al. Epigenetic analysis in placentas from sickle cell disease patients reveals a hypermethylation profile. PloS One. (2022) 17:e0274762. doi: 10.1371/journal.pone.0274762
254. Mumuni M, Adutwum-Ofosu KK, Arko-Boham B, Hottor BA, Koney NK, Adu-Bonsaffoh K, et al. Histomorphology of placentae of women with sickle cell disease during pregnancy - A case control study. PloS One. (2025) 20:e0319011. doi: 10.1371/journal.pone.0319011
255. Wang B, Shen WB, Seif KE, Townsel C, Baracco L, Logue J, et al. Placental SARS-CoV-2 infection and its implications for increased risk of adverse pregnancy outcomes. Am J Perinatol. (2025) 42:52–9. doi: 10.1055/a-2323-0854
256. Joseph L, De Luna G, Bernit E, Cougoul P, Santin A, Faucher B, et al. A study of 28 pregnant women with sickle cell disease and COVID-19: elevated maternal and fetal morbidity rates. Haematologica. (2024) 109:1562–5.
257. Sahu T, Jagzape AT, Sinha M, Sinha R, and Verma HK. New frontiers in sickle cell disease: The role of antiviral therapies and emerging drugs in managing viral infections. World J Virol. (2025) 14:101693. doi: 10.5501/wjv.v14.i2.101693
258. Leonard A and Weiss MJ. Haematopoietic stem cell collection for sickle cell disease gene therapy. Curr Opin Haematol. (2024) 31:104–14. doi: 10.1097/MOH.0000000000000807
259. Pavan AR, Terroni B, and Dos Santos JL. Endothelial dysfunction in sickle cell disease: strategies for the treatment. Nitric Oxide. (2024) 149:7–17. doi: 10.1016/j.niox.2024.05.003
260. Parikh S, Gomez O, Davis T, Lyon Z, and Corces A. Avascular necrosis as a sequela of COVID-19: A case series. Cureus. (2023) 15:e35368. doi: 10.7759/cureus.35368
261. Brittain JE, Hulkower B, Jones SK, Strayhorn D, De Castro L, Telen MJ, et al. Placenta growth factor in sickle cell disease: association with haemolysis and inflammation. Blood. (2010) 115:2014–20. doi: 10.1182/blood-2009-04-217950
262. Kapetanaki MG, Gbotosho OT, Sharma D, Weidert F, Ofori-Acquah SF, and Kato GJ. Free haeme regulates placenta growth factor through NRF2-antioxidant response signaling. Free Radic Biol Med. (2019) 143:300–8. doi: 10.1016/j.freeradbiomed.2019.08.009
263. Argueta LB, Lacko LA, Bram Y, Tada T, Carrau L, Rendeiro AF, et al. Inflammatory responses in the placenta upon SARS-CoV-2 infection late in pregnancy. iScience. (2022) 25:104223. doi: 10.1016/j.isci.2022.104223
264. Perelman N, Selvaraj SK, Batra S, Luck LR, Erdreich-Epstein A, Coates TD, et al. Placenta growth factor activates monocytes and correlates with sickle cell disease severity. Blood. (2003) 102:1506–14. doi: 10.1182/blood-2002-11-3422
265. Kalra VK, Zhang S, Malik P, and Tahara SM. Placenta growth factor mediated gene regulation in sickle cell disease. Blood Rev. (2018) 32:61–70. doi: 10.1016/j.blre.2017.08.008
266. Beltrami S, Rizzo S, Schiuma G, Speltri G, Di Luca D, Rizzo R, et al. Gestational viral infections: role of host immune system. Microorganisms. (2023) 11:1637. doi: 10.3390/microorganisms11071637
267. Pakbaz Z and Wun T. Role of the haemostatic system on sickle cell disease pathophysiology and potential therapeutics. Haematol Oncol Clin North Am. (2014) 28:355–74. doi: 10.1016/j.hoc.2013.11.011
268. Bertero L, Borella F, Botta G, Carosso A, Cosma S, Bovetti M, et al. Placenta histopathology in SARS-CoV-2 infection: analysis of a consecutive series and comparison with control cohorts. Virchows Arch. (2021) 479:715–28. doi: 10.1007/s00428-021-03097-3
269. Hong J and Kumar S. Circulating biomarkers associated with placental dysfunction and their utility for predicting fetal growth restriction. Clin Sci (Lond). (2023) 137:579–95. doi: 10.1042/CS20220300
270. Pecker LH and Federspiel JJ. Patterns of care for pregnant women with sickle cell disease during the COVID-19pandemic in France. Blood. (2024) 143:973–6. doi: 10.3324/haematol.2023.284457
271. Sweeney DA, Lobo SM, Póvoa P, and Kalil AC. Choosing immunomodulating therapies for the treatment of COVID-19: recommendations based on placebo-controlled trial evidence. Clin Microbiol Infect. (2024) 30:611–8. doi: 10.1016/j.cmi.2023.12.028
272. Al-Otaibi ML. An overview of pathophysiology and treatment options of osteonecrosis of the femoral head in sickle cell disease. Saudi Med J. (2022) 43:1192–9. doi: 10.15537/smj.2022.43.11.20220429
273. Borhade MB, Patel P, and Kondamudi NP. Sickle cell crisis. In: StatPearls. StatPearls Publishing, Treasure Island (FL (2025). Available online at: https://www.ncbi.nlm.nih.gov/books/NBK526064/.
274. Martí-Carvajal AJ, Solà I, and Agreda-Pérez LH. Treatment for avascular necrosis of bone in people with sickle cell disease. Cochrane Database Syst Rev. (2019) 12:CD004344. doi: 10.1002/14651858.CD004344.pub7
275. Brandalise SR, Assis R, Laranjeira ABA, et al. Low-dose methotrexate in sickle-cell disease: a pilot study with rationale borrowed from rheumatoid arthritis. Exp Haematol Oncol. (2017) 6:18. doi: 10.1186/s40164-017-0078-1
276. Almeida LEF, Damsker JM, Albani S, Afsar N, Kamimura S, Pratt D, et al. The corticosteroid compounds prednisolone and vamorolone do not alter the nociception phenotype and exacerbate liver injury in sickle cell mice. Sci Rep. (2018) 8:6081. doi: 10.1038/s41598-018-24274-6
277. Sundaresan B, Shirafkan F, Ripperger K, and Rattay K. The role of viral infections in the onset of autoimmune diseases. Viruses. (2023) 15:782. doi: 10.3390/v15030782
278. Costa Neto A, Santos F, Ribeiro I, Oliveira V, Dezan M, Kashima S, et al. FcγR2B B2.4 haplotype predicts increased risk of red blood cell alloimmunization in sickle cell disease patients. Transfusion. (2020) 60:1573–8. doi: 10.1111/trf.15832
279. Kirkham JK, Estepp JH, Weiss MJ, and Rashkin SR. Genetic variation and sickle cell disease severity: a systematic review and meta-analysis. JAMA Netw Open. (2023) 6:e2337484. doi: 10.1001/jamanetworkopen.2023.37484
280. Calabretta E, Moraleda JM, Iacobelli M, Jara R, Vlodavsky I, O’Gorman P, et al. COVID-19-induced endotheliitis: emerging evidence and possible therapeutic strategies. Br J Haematol. (2021) 193:43–51. doi: 10.1111/bjh.17240
281. Bhat V and Sheehan VA. Can we use biomarkers to identify those at risk of acute pain from sickle cell disease? Expert Rev Haematol. (2024) 17:411–8. doi: 10.1080/17474086.2024.2372322
Keywords: sickle cell disease, SARS-CoV-2, inflammation, cytokines, cytokine storm
Citation: Martí-Carvajal AJ and De Sanctis JB (2025) A perfect storm: the deadly intersection of sickle cell disease and COVID-19. Front. Hematol. 4:1651089. doi: 10.3389/frhem.2025.1651089
Received: 20 June 2025; Accepted: 01 August 2025;
Published: 25 August 2025.
Edited by:
Halvard Bönig, Goethe University Frankfurt, GermanyReviewed by:
Marvyn T. Koning, Leiden University, NetherlandsAndres Felipe Escobar, Hospital Pablo Tobon Uribe, Colombia
Copyright © 2025 Martí-Carvajal and De Sanctis. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Juan Bautista De Sanctis, anVhbmJhdXRpc3RhLmRlc2FuY3Rpc0B1cG9sLmN6