Skip to main content

On the cutting edge of sickle cell disease: a snapshot narrative review

Abstract

Background

Sickle cell disease (SCD) is the most common hemoglobinopathy worldwide, characterized by vaso-occlusive crises and ischemia that affects patients on a multi-system level. Despite being a genetically simple disease due to a single base mutation, SCD poses many therapeutic challenges. Additionally, its impact on patients’ life remains significant. This narrative review aims to provide a snapshot of recent highlights of the significant progress in SCD therapy, and the impact of SCD on patients’ life, including  the complications, morbidity, and mortality factors of the disease.

Methodology

Google Scholar and PubMed were searched for “sickle cell disease”. Only full-text English language original research articles were included in this review. In total, 600 articles were screened, 300 from each database, which were published from 2020 to 2024-06-01. A total of 139 studies were included in this review, after screening for inclusion.

Conclusions

The increasing global incidence of sickle cell disease underscores the urgency for healthcare interventions to address the health challenges of an aging population living with this chronic condition. While treatment options for sickle cell disease have broadened, their availability is still limited. Among these options, stem cell transplant stands out as the definitive treatment, with ongoing efforts to enhance the donor pool. The disease significantly affects patients' quality of life and overall health, with emerging neurological and psychiatric issues. Additionally, the impact of sickle cell disease on reproductive health in both men and women presents a pressing need for further research to meet reproductive challenges.

Background

Sickle cell disease (SCD) is the most common hemoglobinopathy globally, which is typically prevalent in sub-Saharan African, Indian, and Middle Eastern regions [1]. Migration and population expansion have led to its presence in non-malarial endemic regions. Each year, around 300,000 babies are born with SCD [2]. SCD’s pathophysiology involves the sickling and hemolysis of red blood cells, which obstructs blood flow in small blood vessels. This results in vaso-occlusion and ischemia episodes, which can significantly impact the physical and psychosocial health of patients with SCD [3]. Other complications of the disease include pain, infections, anemia, acute chest syndrome, kidney disease [4], pancreatitis [5], delayed growth and development, stroke, and organ damage [3]. Complications and manifestations of SCD are further compounded by the presence of other concurrent conditions like thalassemia [6, 7], G6PD deficiency [8, 9], and hemophilia [10, 11].

SCD was first described more than 110 years ago, and was only officially recognized as a disease of major public health issue by the World Health Organization in May 2006 [12]. Definitive treatment options for SCD remain limited and often require a multi-disciplinary approach [3, 13]. There are only four FDA-approved drugs available for the therapy of SCD—hydroxyurea, L-glutamine, crizanlizumab, and voxelotor [14]. However, some of these medications are largely unavailable in SCD endemic areas. Meanwhile, gene therapy such as CRISPR-Cas9 has emerged as a controversial therapy, but may be limited by off-target effects, safety concerns, issues related to the delivery of the system to the appropriate cells of the body, efficacy [15], and extremely high costs which is a major health equity concern [16]. Very recent advancements include the degradation of an HbF repressor, widely interspaced zinc finger—WIZ, which was tolerated in murine studies and induced fetal hemoglobin production; these findings could be the foundations for the development of another oral treatment for SCD [17, 18].

Hematopoietic stem cell transplant (HSCT) from a matched donor is the only treatment cure for SCD, but it is not always a viable option due to the lack of matched sibling donors resulting in a limited donor pool [19, 20], and it carries significant risks. Its use has been generally limited to those who have experienced severe complications, such as stroke [21]. Sustainable and equitable care for SCD is needed to improve the global care and health of patients with SCD [16].

Sickle cell disease continues to be a global concern. Updates in sickle cell disease (SCD) trends show a rising annual death rate among adult SCD patients in the USA, with the median age of death increasing from 28 to 43 years [22]. Deaths primarily stem from chronic conditions, emphasizing the need for improved management strategies as this population ages. Additionally, surveillance studies seem to identify more patients with SCD [23]. Globally, the incidence of SCD showed a 13.7% rise in newborns with SCD which is attributed to population expansion people ethnically from sub-Saharan Africa and the Caribbean. The clinical burdens of SCD are expected to progressively increase with patients requiring procedures such as dialysis or organ transplantation [24]. Similar trends are observed in high-income countries [24,25,26].

Given the critical impact of sickle cell disease (SCD) on patients and the escalating global need for comprehensive insights into advancements and trends in SCD therapy, and its continued acute and chronic impact on patients physical and psychosocial health, this snapshot narrative review aims to provide a highlight of recent research on SCD, focusing on therapeutic care progress, morbidity and associated complications, and the current impacts on patients with SCD.

Methodology

Google Scholar and PubMed was searched for (“sickle cell disease,” “SCD”) query on 2024-06-01 with a temporal limit since 2020. The first 300 articles were screened by title and abstract from each of the databases. We included only full-text English language original research articles (see Fig. 1).

Fig. 1
figure 1

Flowchart depicting the search strategy for this review. Only original full-text research published from 2020 to 2024 was included in this study

We excluded non-original research articles such as reviews, short reports, letters to the editor, abstracts, conference meetings, policy documents, guidelines, commentaries, case reports, perspectives, dispatches, correspondence, drug profiles, research protocols, editorials, books, book chapters, and comments. Animal studies were also excluded. We excluded original research studies with primary outcomes related to financial, insurance, or economic aspects, as well as those examining health utilization. Studies not directly involving SCD patient outcomes and their data were excluded. Studies focusing on parents, caretakers, stakeholders, screening tools, online medical information, artificial intelligence/machine learning tools, policy or politics, public health efforts, COVID-19, current practice surveys, management of surveillance programs or clinics, and development of treatment programs were excluded. Preprint results, retracted articles, and articles with expressions of concern were part of the exclusion criteria.

For the purposes of a comprehensive narrative review of clinically relevant original research, we included full-text studies that addressed treatment and therapeutic research, disease impact on SCD patients, reported complications and factors influencing disease morbidity and mortality, as well as the impact on women’s and men’s health.

A total of 600 articles were screened. A final total of 139 original research articles were referenced in this narrative review. The original research studies included were conducted across 26 countries worldwide. The majority of studies were overwhelmingly published by authors from the USA.

Figure 2 presents a mind map highlighting some of the key findings in this study.

Fig. 2
figure 2

Mind map depicting the key findings relevant to this review

Medications and therapy

Although new adjunctive therapies have emerged for the therapy of SCD, none of them are considered cures for SCD[27]. Hematopoietic cell transplantation (HCST) is currently the only known cure, while advancements in gene editing technology offer promising prospects for the future. Recent original research on therapeutics reveals interesting clinical outcomes.

Gene and antibody therapy and CRISPR-Cas9

One study examined the efficacy of LentiGlobin, a type of gene therapy for SCD that involves autologous transplantation of hematopoietic stem cells transduced with a lentiviral vector which encodes for a modified B-globin gene. This results in the production of anti-sickling hemoglobin, designed to inhibit polymerization. In the examined patients, hemolysis markers were reduced and had resolution of severe vaso-occlusive crisis (VOC). No patients developed hematological malignancy at follow-up [28]. Meanwhile, in another study, autologous cells transduced with lentiviral vector were assessed for safety and clinical response. All patients were found to have stable HbF induction; the clinical manifestations of SCD were reduced or totally absent by the end of the follow-up period [29]. Exagamglogene autotemcel (exa-cel), a non-viral cell therapy that works by reactivating fetal hemoglobin synthesis, was found to decrease occlusive crisis in 97% of patients [30]. However, long-term risks of genotoxicity are a concern. GHVD is a possible complication in autologous gene therapy, especially in scenarios with a degree of mismatching in African-American patients [31].

Gene editing for SCD has emerged as an interesting and controversial topic. The long-term response and safety of using gene editing technology remains to be determined. One study using CRISPR-Cas9 was used to disrupt HBG1 and HBG2 gene promoters, to induce the development of fetal hemoglobin. The infusion of OTQ923 in three individuals suffering from severe sickle cell disease led to a sustained increase in the production of fetal hemoglobin in their red blood cells [32].

Few studies have been published on crizanlizumab, a monoclonal antibody, for the treatment of sickle cell disease (SCD) [33]. One study using crizanlizumab at 5mg/kg body weight observed a decrease in acute care visits in SCD patients, but revealed a high discontinuation rate [33]. One German retrospective study found that crizanlizumab showed no difference than placebo, as well as significant severe events and admissions [34].

Hematopoietic stem cell transplant

Hematopoietic stem cell transplant (HSCT) from a matched donor remains as the ultimate cure for SCD [19, 20], but it is not a commonly utilized therapy (due to limited availability of matched donors), and is associated with complications like graft-versus-host disease (GVHD). HCST from a matched sibling donor consistently showed event-free survival and low incidence of GVHD [35]. One study reported the use of non-myeloablative HSCT. They found no clonal evolution or myeloid malignancies; graft failure was seen in some patients which appears to be dependent on ABO matching and non-ABO antibodies [36]. Plerixafor mobilization and apheresis in patients with sickle cell may allow the collection of sufficient hematopoietic stem cell collection, the quantities of which vary among SCD patients. Factors affecting a higher yield included age < 30, a higher baseline and pre-apheresis CD34+ count, a high baseline white blood cell and platelet count, minimal hospitalizations, and no chronic pain [37]. HLA-matched unrelated donors were surprisingly found to have 100% overall survival and no severe GHVD. Unrelated donor transplantations may yield excellent outcomes, although patient risk factors such as advanced disease stage and comorbidities should be considered [38].

Hydroxyurea

Hydroxyurea (HU) remains a staple in the care of SCD. One study observed that HU treatment modulates inflammation and oxidative stress in both HbSS and HbSC genotypes [39]. Patients without the use of HU showed increased levels of plasma advanced oxidation protein product and interleukin-8 [39]. Some studies report on the efficacy of voxelotor, a once daily tablet that inhibits the polymerization of HbS [40]. Treatment was associated with improved hemoglobin levels, decreased reticulocyte percentage, and decreased total bilirubin. Concomitant use with hydroxyurea (HU) resulted in a more robust and complementary effect [41].

Pain

With regards to pain, a recent study showed a ninefold increase in the odds of discharge in children with SCD who were given intranasal fentanyl, and they were less likely to be admitted. Meanwhile, children who received oral opioids had a threefold greater odds of discharge than those who did not [42]. Additionally, the use of sublingual buprenorphine to transition from opioid care produced few medical adverse events, including recurrent or protracted withdrawal and worsened pain. Its use was associated with a marked reduction in acute care utilization by 72.5% on average [43]. Meanwhile, a randomized control trial found a significant response in pain management using digital cognitive behavioral therapy for pain intervention, among adolescents with SCD. Significant reductions in average pain intensity and the number of pain days were observed, while improvements were seen with coping attempts, mood, and fatigue [44].

Transfusions

Red blood cell exchange therapy was found to beneficial for pediatric patients requiring it for chronic transfusion, but its use in the prevention of stroke remains to be elucidated. Using a femoral double‐lumen central venous catheter and 500 units in each lumen has been recommend due to limited venous access in this patient group [45]. Hemoglobin levels post-exchange therapy were significantly increased, and a total exchange volume of > 35 mL/kg was found to be the best way to reduce HbS to < 30% [45]. Blood transfusion may be pivotal in preoperative care in SCD patients, where one study reported increased risk of complications in the post-op period in those who did not receive transfusion. Prolonged hospitalization was observed in those who did not receive pre-op blood transfusion [46]. Meanwhile, preoperative blood transfusion did not show improved outcomes for semi-elective abdominal surgery (cholecystectomy, splenectomy, or appendectomy) in children with SCD. No statistically significant differences were found with regards to 30-day readmission or 30-day surgical complications, and this encourages investigations in order to minimize unnecessary transfusions [47].

With regard to hemodilution, one study confirmed the safety of isovolumic hemodilution in SCD patients with a history of stroke compared with the standard red cell exchange. Lower RBC usage and higher post-procedure hematocrit were observed, and this is clinically relevant, while no infarct progression was seen on MRI/MRA. Statistically significant drops in blood pressure were observed, but this was not clinically significant [48].

Impact of SCD on patients

Physical and psychosocial impact

Sickle Cell disease continues to have a significant impact on the quality of life of patients. Pain especially bears a significant impact [49]. One multi-country survey revealed that SCD significantly continued to affect aspects of patient life across areas including the performance of household daily activities, hygiene, family and social life, and relationships with spouse and partner [50]. Patients also reported significant impact on their emotional well-being, and a worry that their disease will worsen. SCD impaired patients total working hours, while some were dismissed from work as a result of their condition, and reported lower household income [51, 52]. Likewise, SCD affected students’ attendance and adversely affected their school performance[50]. Meanwhile, academic performance in SCD patients may be linked to other social determinants such as household socioeconomic status [53]. SCD patients report a general experience of stigma, and this invites policy reform, programs, and health initiatives to overcome this [54].

Breakthrough solutions

There appears to be associations between adolescent health literacy and individual traits such as age, annual household income, and caregiver education level. The findings suggest that future research should explore additional factors, including media use, in order to better understand health literacy in adolescents with SCD, and inform future evaluation and intervention projects [55]. Social support networks for patients with SCD are pivotal for their well-being, in order to provide emotional support and support during health crisis events [56]. Furthermore, improving disease knowledge using a web-based technological interface appears to have positive outcomes, especially in the transition of youth with SCD into adult care (which should be done ideally within the first 6 months of the transition of care) [57,58,59]. Promoting executive functioning [60], self-efficacy and self-management skills allowed SCD patients to take a more active role in managing their condition [61].

Complications and morbidity and mortality

Sickle cell disease (SCD) poses significant challenges in terms of mortality and morbidity, with a range of complications that can adversely impact patients' well-being and life expectancy, as well as the complications of therapy.

Systemic complications

Cardiac, acute chest or respiratory failure, and sudden unexplained death were the most common causes of death in one study [62]. Iron overload, depression, and pulmonary hypertension were leading predictors of death [62]. Meanwhile, elevated iron stores were found in patients with at least 3 transfusion per year [63]. An echocardiogram may be warranted in patients transitioning from adolescent to adult care as this population is at risk for cardiovascular disease [64].

Acute kidney injury, decreased kidney function, chronic kidney disease [65, 66], stroke [67, 68], osteomyelitis [69], cholelithiasis [70], avascular necrosis [71], non-rheumatic valvular heart disease [72], and arrhythmias are other complications that were reported on in the literature [73,74,75]. Meanwhile, adverse childhood experiences were linked to poor outcomes and increased morbidity [76]. Suicide risk was observed in one study [77]. Patients with SCD are at an increased risk of GIT bleeding events, especially upper GI bleeds. Any bleeding event was found to place a patient at a twofold increase in risk of death [78]. Infection [79, 80], acute chest syndrome, asthma [81, 82], pulmonary embolism, and death during a pain episode are causes of morbidity [83,84,85,86]. Meanwhile, pain and fever were among the commonest reasons for seeking acute care [87]. Tonisillar hypertrophy, up to obstructive grades, has been reported on in SCD patients [88]. Consultations with pulmonary care specialists are pivotal in the care of SCD patients [81].

Neuropsychological and special senses issues

Patients report significant impact on sleep [89], opioid use, depression [90], obstructive sleep apnea [91], pain [92,93,94] including gastrointestinal pain [95], hearing loss [96], and neurological/cognitive and behavioral challenges [97,98,99,100,101,102,103,104,105,106,107,108,109,110,111]. Proliferative sickle cell retinopathy has been reported among other ocular manifestations [112,113,114], which may be associated with elevated E-selectin levels [115].

A diagnosis of depression was found to significantly increased risk of readmission among the patients aged 18–29 years [116]. Meanwhile, hydroxyurea may play a role in mitigating neurocognitive decline in SCD patients [111, 117]. Interestingly, the neurocognitive dysfunction seen in SCD patients may not entirely be explained by iron overload effects on the brain [118]. Impaired cerebrovascular hemodynamics are evident in SCD patients [119, 120]. Meanwhile, early neuropsychological surveillance and assessment may protect from further neurocognitive decline [121].

Treatment-related issues

Complications associated with therapy are also prevalent. An association with inappropriate use of intravenous fluid use in the therapy of VOC’s was found with complications such as acute kidney injury, volume overload, and acute chest syndrome, prompting the need for controlled trials to clarify the role of fluids in the treatment of VOCs [122]. Due to the need for blood transfusion, and increased hospital contact, SCD patients remain at an increased risk of infections like HCV [123].

No observed increase in mortality was found for SCD patients after cardiothoracic surgical procedures [124].

Chronic health concerns

In the context of an aging SCD population, more chronic conditions are a health concern. Liver complications, such as cirrhosis, are a significant cause of morbidity in SCD patients [125]. SCD hepatopathy is a condition that can arise from sickling into the hepatic sinusoids. Liver transplant may be warranted in patients with SCD in the case of acute liver failure or end stage chronic liver disease. One study observed the 1- and 5-year survival to be 75% and 65% respectively, while high morbidity is a concern in the early postoperative period [126]. A potential association may exist between SCD and autoimmune liver disease [127]

Risk factors for increased complications

Risk factors and exposures that can trigger SCD events or complications include short-term air pollution [128]. Also, vitamin deficiencies (A and D) in SCD patients can compound the risk of infection [129], while high-dose bolus of vitamin D plus 1000 IU daily was found effective to treat hypovitaminosis D in this patient group [130]. Meanwhile, lifetime risk of obesity and diabetes appears to be low among SCD patients [131]. The presence of Moyamoya syndrome (a disease defined as the chronic and progressive narrowing of the arteries in the brain) conferred a 20× risk of stroke in patients with SCD [132]. Finally, it is important to differentiate between sickle cell genotypes as each may be associated with its own laboratory findings and clinical complications, with sickle cell anemia being the most severe form [133, 134]. SCD may be compounded by the coexistence of other hemoglobinopathies [135]. Interestingly, sickle cell anemia with co-inheritance with a-thalassemia appears to have a protective role against many disease-related complications [136]

Women’s health and family planning

Pregnant individuals affected by sickle cell disease (SCD) encounter increased risk of pregnancy complications such as eclampsia, sepsis, venous thromboembolism, and intrauterine growth restriction compared to the general population [137]. The underlying causes of these unfavorable pregnancy outcomes can be attributed to the pathophysiology of the disease, compounded by socioeconomic disparities in care [138, 139]. Pregnant women with SCD were significantly associated with maternal anemia, maternal lower blood pressure, thromboembolism [140], inpatient mortality, preeclampsia/eclampsia [141] hospital readmissions, need for blood transfusion, opioid-related disorder [142], intrauterine fetal demise, low birth weight, neonatal asphyxia, transfer to the ICU, early neonatal death, and severe postpartum anemia [143,144,145]. Other studies confirm increased severe maternal morbidity among women with SCD [145,146,147]. There is a need for enhanced scientific and political effort to improve the outcomes for pregnant patients with SCD.

One study examined the impact of hydroxyurea use during conception, pregnancy, and infant outcomes. Hydroxyurea use during pregnancy was found to increase the risk of still birth or miscarriage, and it was also associated with decreased birth weight in infants born full term [148]. Caution for its use during pregnancy is still advised. Meanwhile, its use was associated with decreased ovarian reserve in women with SCD [149]. Therapy for pregnant women with SCD remains an underdeveloped area that needs more comprehensive research and therapy development to meet its challenge [150]. Overall, the risk of maternal and fetal adverse events may be determined by risk profiles including the presence of low first-trimester hemoglobin, admission in the year before pregnancy, multiple transfusions before pregnancy, a history of maternal cardiac complications, and the presence of HbSS/HbSβ0-thalassemia genotype [151].

For parents wishing to conceive, only half knew about their hemoglobinopathy status [152] or their partners. Meanwhile, having SCD was a reason why some patients did not wish to conceive [133]. Knowledge about other reproductive options was limited, and choices such as IVF were limited due to personal, cultural, or financial factors [133]. Meanwhile, preimplantation genetic diagnosis may be a solution for the prevention of SCD [153]. Young adults shifting to adult care bring forth unique reproductive needs. Multi-disciplinary and intersectional care may meet these challenges [154], as well as fertility and genetic counseling [155]

Men’s health

Priapism prevalence is higher in men with SCD than those without. Men report using exercise as a coping mechanism, as well as concerns for their self-image and feelings of sadness, embarrassment, and fear. Erectile dysfunction was found twofold more in men with SCD [156]. Compensated hypogonadism was also reported on in men with SCD [157]

Conclusions

SCD significantly impacts the lives of patients, affecting a broad range of physical and psychosocial aspects. Additionally, there is new hope as more therapeutic methods and findings will continue to shape the care for SCD patients. From this review, we can derive valuable insights into the significant impact of SCD on patients, which underscores the growing need for a unified global comprehensive approach to managing this disease, especially in the context of a chronically aging world population which will bring with it unique and complex healthcare challenges.

This review also highlights key findings in the literature that can point to new research directions, such as the rising concern of neurological and psychological complications among SCD patients that remains to be addressed. Another implication is the limited therapy options for pregnant women with SCD, which may inspire more research and targeted interventions and health policy, as this group is at significant risk of morbidity and mortality.

Our review findings can also delineate areas for policy and guideline development which highlight the necessity for targeted interventions that can focus on improving access to quality care, such as among adolescents transitioning into adult care, men's and women's health, and psychiatric and mental health support for SCD patients. Furthermore, exploring the intersectionality of genetics, personalized medicine, and social determinants of health in SCD care could pave the way for more effective and individualized treatment strategies for SCD patients. In summary, the practical implications and future research directions derived from this review underscore the importance of a comprehensive and proactive approach to SCD management, while also serving as a concise summary of vital trends in therapy, care, and complications that may be valuable to physicians, students, educators, and patients alike.

This review has some limitations, as it only included the original research identified from two major databases. Therefore, there is a potential publication bias represented in the methods, which may not have included findings from research not identified by these databases, potentially leaving behind unique findings in studies from SCD endemic areas. To mitigate this, we screened the first 300 articles in each database in the order in which they appeared, and the majority of the studies included in this narrative were published in the USA and other Western countries, with limited findings from the African/SCD endemic area research narrative, yet this can be the focus of future studies. A publication date bias may also be present which may overlook older but relevant studies. Nonetheless, this review comprises a snapshot summary of the significant recent findings and outcomes from an exhaustive amount of literature, which should inspire more research and continued reading into the discourse on sickle cell disease.

Availability of data and materials

Not applicable.

Abbreviations

CRISPR-Cas9:

Clustered regularly interspaced palindromic repeats

G6PD:

Glucose-6-phosphate dehydrogenase

GIT:

Gastrointestinal

GVHD:

Graft-versus-host Disease

HBG1/2:

Hemoglobin subunit gamma 1/2

HbS:

Hemoglobin S

HCV:

Hepatitis C virus

HLA:

Human leukocyte antigen

HSCT:

Hematopoietic stem cell transplantation

HU:

Hydroxyurea

IU:

International units

IVF:

In vitro fertilization

SCD:

Sickle cell disease

VOC:

Vaso-occlusive crisis

WIZ:

Widely interspaced zinc finger

References

  1. Darbari DS, Sheehan VA, Ballas SK (2020) The vaso-occlusive pain crisis in sickle cell disease: Definition, pathophysiology, and management. Eur J Haematol 105:237–246. https://doi.org/10.1111/ejh.13430

    Article  PubMed  Google Scholar 

  2. Thein M, Thein S (2016) World Sickle Cell Day 2016: a time for appraisal. Indian J Med Res 143:678. https://doi.org/10.4103/0971-5916.191917

    Article  PubMed  PubMed Central  Google Scholar 

  3. Elendu C, Amaechi DC, Alakwe-Ojimba CE, Elendu TC, Elendu RC, Ayabazu CP et al (2023) Understanding Sickle cell disease: causes, symptoms, and treatment options. Medicine 102:e35237. https://doi.org/10.1097/MD.0000000000035237

    Article  PubMed  PubMed Central  Google Scholar 

  4. Amarapurkar P, Roberts L, Navarrete J, El Rassi F (2022) Sickle cell disease and kidney. Adv Chronic Kidney Dis 29:141-148.e1. https://doi.org/10.1053/j.ackd.2022.03.004

    Article  PubMed  Google Scholar 

  5. Sharma A, Khadka B, Sharma A, Shah KB, Shrestha AN (2023) Recurrent acute pancreatitis in an adult female with sickle cell disease: a case report. Ann Med Surg 85:37–40. https://doi.org/10.1097/MS9.0000000000000178

    Article  Google Scholar 

  6. Vincent O, Oluwaseyi B, James B, Saidat L (2016) Coinheritance of Β-thalassemia and sickle cell anaemia in southwestern Nigeria. Ethiop J Health Sci 26:517. https://doi.org/10.4314/ejhs.v26i6.3

    Article  PubMed  PubMed Central  Google Scholar 

  7. Santos B, Delgadinho M, Ferreira J, Germano I, Miranda A, Arez AP et al (2020) Co-Inheritance of alpha-thalassemia and sickle cell disease in a cohort of Angolan pediatric patients. Mol Biol Rep 47:5397–5402. https://doi.org/10.1007/s11033-020-05628-8

    Article  PubMed  Google Scholar 

  8. Nnamezie Igwilo H, Salawu L, Adeoye Adedeji T (2021) The impact of glucose-6-phosphate dehydrogenase deficiency on the frequency of vasoocclusive crisis in patients with sickle cell anemia. Plasmatology. https://doi.org/10.1177/263485352110405285

    Article  Google Scholar 

  9. Karafin MS, Fu X, D’Alessandro A, Thomas T, Hod EA, Zimring JC et al (2018) The clinical impact of glucose-6-phosphate dehydrogenase deficiency in patients with sickle cell disease. Curr Opin Hematol 25:494–499. https://doi.org/10.1097/MOH.0000000000000455

    Article  PubMed  Google Scholar 

  10. Dhiman P, Chaudhary R, Sudha K (2015) Sickle cell-β thalassemia with concomitant hemophilia A: a rare presentation. Blood Res 50:264. https://doi.org/10.5045/br.2015.50.4.264

    Article  PubMed  PubMed Central  Google Scholar 

  11. Patel S, Krishnan VP, Kanvinde P, Mudaliar S, Shah N, Swami A et al (2020) Clinical course of patients with sickle cell anemia and co-inherited hematological disorders: experience at a tertiary hematological centre. Ind J Hematol Blood Transf 36:754–757. https://doi.org/10.1007/s12288-020-01303-x

    Article  Google Scholar 

  12. Esoh K, Wonkam-Tingang E, Wonkam A (2021) Sickle cell disease in sub-Saharan Africa: transferable strategies for prevention and care. Lancet Haematol 8:e744–e755. https://doi.org/10.1016/S2352-3026(21)00191-5

    Article  PubMed  Google Scholar 

  13. Salinas Cisneros G, Thein SL (2020) Recent advances in the treatment of sickle cell disease. Front Physiol. https://doi.org/10.3389/fphys.2020.00435

    Article  PubMed  PubMed Central  Google Scholar 

  14. Leibovitch JN, Tambe AV, Cimpeanu E, Poplawska M, Jafri F, Dutta D et al (2022) l-glutamine, crizanlizumab, voxelotor, and cell-based therapy for adult sickle cell disease: Hype or hope? Blood Rev 53:100925. https://doi.org/10.1016/j.blre.2021.100925

    Article  PubMed  Google Scholar 

  15. Ma L, Yang S, Peng Q, Zhang J, Zhang J (2023) CRISPR/Cas9-based gene-editing technology for sickle cell disease. Gene 874:147480. https://doi.org/10.1016/j.gene.2023.147480

    Article  PubMed  Google Scholar 

  16. Wonkam A, Munung NS, Guindo A, Nnodu O (2024) Priorities for sickle cell disease global research and implementation. The Lancet. https://doi.org/10.1016/S0140-6736(24)01400-4

    Article  PubMed  Google Scholar 

  17. Higgs D, Kassouf M (1979) Developing a pill to treat sickle cell disease. Science 2024(385):27–28. https://doi.org/10.1126/science.adq3757

    Article  Google Scholar 

  18. Ting PY, Borikar S, Kerrigan JR, Thomsen NM, Aghania E, Hinman AE et al (1979) A molecular glue degrader of the WIZ transcription factor for fetal hemoglobin induction. Science 2024(385):91–99. https://doi.org/10.1126/science.adk6129

    Article  Google Scholar 

  19. Leonard A, Furstenau D, Abraham A, Darbari DS, Nickel RS, Limerick E et al (2023) Reduction in vaso-occlusive events following stem cell transplantation in patients with sickle cell disease. Blood Adv 7:227–234. https://doi.org/10.1182/bloodadvances.2022008137

    Article  PubMed  Google Scholar 

  20. Mahesri M, Schneeweiss S, Globe D, Mutebi A, Bohn R, Achebe M et al (2021) Clinical outcomes following bone marrow transplantation in patients with sickle cell disease: a cohort study of US Medicaid enrollees. Eur J Haematol 106:273–280. https://doi.org/10.1111/ejh.13546

    Article  PubMed  Google Scholar 

  21. Walters MC, De Castro LM, Sullivan KM, Krishnamurti L, Kamani N, Bredeson C et al (2016) Indications and results of HLA-Identical sibling hematopoietic cell transplantation for sickle cell disease. Biol Blood Marrow Transplant 22:207–211. https://doi.org/10.1016/j.bbmt.2015.10.017

    Article  PubMed  Google Scholar 

  22. Payne AB, Mehal JM, Chapman C, Haberling DL, Richardson LC, Bean CJ et al (2020) Trends in sickle cell disease-related mortality in the United States, 1979 to 2017. Ann Emerg Med 76:S28-36. https://doi.org/10.1016/j.annemergmed.2020.08.009

    Article  PubMed  Google Scholar 

  23. Reeves SL, Horiuchi S, Zhou M, Paulukonis S, Snyder A, Wilson-Frederick S et al (2024) Case ascertainment of sickle cell disease using surveillance or single administrative database case definitions. Public Health Rep 139:187–194. https://doi.org/10.1177/00333549231166465

    Article  PubMed  Google Scholar 

  24. Beillat M, Durand-Zaleski I, Pirenne F, Bénard S, Chillotti L, Galacteros F (2023) Prevalence and cost of sickle cell disease in France: real-world analysis using data from the Echantillon Généraliste des Bénéficiaires. Front Public Health. https://doi.org/10.3389/fpubh.2023.1215605

    Article  PubMed  PubMed Central  Google Scholar 

  25. Nelson A, Ho PJ, Haysom H, Waters N, Wellard C, Chee M et al (2024) Sickle cell disease in Australia: a snapshot from the Australian Haemoglobinopathy Registry. Intern Med J 54:764–772. https://doi.org/10.1111/imj.16297

    Article  PubMed  Google Scholar 

  26. Brousse V, Bernaudin F, Melaine A, Goguillot M, Gallagher M, Benard S et al (2023) Severity and burden of sickle cell disease in France: a nationwide realworld study. Haematologica. https://doi.org/10.3324/haematol.2022.282098

    Article  PubMed  PubMed Central  Google Scholar 

  27. Abdel-Hadi L, Ventura Carmenate Y, Castillo-Aleman YM, Sheikh S, Zakaria A, Phillips J (2023) Treatment of sickle cell disease - options and perspective. Am J Blood Res 13:61–70

    PubMed  PubMed Central  Google Scholar 

  28. Kanter J, Walters MC, Krishnamurti L, Mapara MY, Kwiatkowski JL, Rifkin-Zenenberg S et al (2022) Biologic and clinical efficacy of lentiglobin for sickle cell disease. N Engl J Med 386:617–628. https://doi.org/10.1056/NEJMoa2117175

    Article  PubMed  Google Scholar 

  29. Esrick EB, Lehmann LE, Biffi A, Achebe M, Brendel C, Ciuculescu MF et al (2021) Post-transcriptional genetic silencing of BCL11A to treat sickle cell disease. N Engl J Med 384:205–215. https://doi.org/10.1056/NEJMoa2029392

    Article  PubMed  Google Scholar 

  30. Frangoul H, Locatelli F, Sharma A, Bhatia M, Mapara M, Molinari L et al (2024) Exagamglogene autotemcel for severe sickle cell disease. N Engl J Med 390:1649–1662. https://doi.org/10.1056/NEJMoa2309676

    Article  PubMed  Google Scholar 

  31. Parikh S, Brochstein JA, Galamidi E, Schwarzbach A, Kurtzberg J (2021) Allogeneic stem cell transplantation with omidubicel in sickle cell disease. Blood Adv 5:843–852. https://doi.org/10.1182/bloodadvances.2020003248

    Article  PubMed  PubMed Central  Google Scholar 

  32. Sharma A, Boelens J-J, Cancio M, Hankins JS, Bhad P, Azizy M et al (2023) CRISPR-Cas9 editing of the HBG1 and HBG2 promoters to treat sickle cell disease. N Engl J Med 389:820–832. https://doi.org/10.1056/NEJMoa2215643

    Article  PubMed  PubMed Central  Google Scholar 

  33. Cheplowitz H, Block S, Groesbeck J, Sacknoff S, Nguyen AL, Gopal S (2023) Real-world data of crizanlizumab in sickle cell disease: a single-center analysis. J Hematol 12:105–108. https://doi.org/10.14740/jh1127

    Article  PubMed  PubMed Central  Google Scholar 

  34. Poppenborg F, Röth A, Yamamoto R, Reinhardt HC, Alashkar F (2024) Crizanlizumab in adult patients with sickle cell disease: a retrospective German analysis. Front Hematol. https://doi.org/10.3389/frhem.2024.1374181

    Article  Google Scholar 

  35. Benítez-Carabante MI, Beléndez C, González-Vicent M, Alonso L, Uría-Oficialdegui ML, Torrent M et al (2021) Matched sibling donor stem cell transplantation for sickle cell disease: results from the Spanish group for bone marrow transplantation in children. Eur J Haematol 106:408–416. https://doi.org/10.1111/ejh.13566

    Article  PubMed  Google Scholar 

  36. Damlaj M, Alahmari B, Alaskar A, Alhejazi A, Alsadi H, Ahmed M et al (2024) Favorable outcome of non-myeloablative allogeneic <scp>transplantation</scp> in adult patients with severe sickle cell disease: a single center experience of 200 patients. Am J Hematol 99:1023–1030. https://doi.org/10.1002/ajh.27295

    Article  PubMed  Google Scholar 

  37. Leonard A, Sharma A, Uchida N, Stroncek D, Panch SR, West K et al (2021) Disease severity impacts plerixafor-mobilized stem cell collection in patients with sickle cell disease. Blood Adv 5:2403–2411. https://doi.org/10.1182/bloodadvances.2021004232

    Article  PubMed  PubMed Central  Google Scholar 

  38. Kogel F, Hakimeh D, Sodani P, Lang P, Kühl J, Hundsdoerfer P et al (2021) Allogeneic hematopoietic stem cell transplantation from sibling and unrelated donors in pediatric patients with sickle cell disease—A single center experience. Pediatr Transplant. https://doi.org/10.1111/petr.13892

    Article  PubMed  Google Scholar 

  39. Connes P, Möckesch B, Tudor Ngo Sock E, Hardy-Dessources M, Reminy K, Skinner S et al (2021) Oxidative stress, inflammation, blood rheology, and microcirculation in adults with sickle cell disease: effects of hydroxyurea treatment and impact of sickle cell syndrome. Eur J Haematol 106:800–807. https://doi.org/10.1111/ejh.13607

    Article  PubMed  Google Scholar 

  40. Estepp JH, Kalpatthi R, Woods G, Trompeter S, Liem RI, Sims K et al (2022) Safety and efficacy of voxelotor in pediatric patients with sickle cell disease aged 4 to 11 years. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.29716

    Article  PubMed  Google Scholar 

  41. Muschick K, Fuqua T, Stoker-Postier C, Anderson AR (2022) Real-world data on voxelotor to treat patients with sickle cell disease. Eur J Haematol 109:154–161. https://doi.org/10.1111/ejh.13782

    Article  PubMed  Google Scholar 

  42. Rees CA, Brousseau DC, Ahmad FA, Bennett J, Bhatt S, Bogie A et al (2023) Intranasal fentanyl and discharge from the emergency department among children with sickle cell disease and vaso-occlusive pain: a multicenter pediatric emergency medicine perspective. Am J Hematol 98:620–627. https://doi.org/10.1002/ajh.26837

    Article  PubMed  PubMed Central  Google Scholar 

  43. David MS, Jones J, Lauriello A, Nnake I, Plazas Montana M, Lasko K et al (2022) Converting adults with sickle cell disease from full agonist opioids to buprenorphine: a reliable method with safety and early evidence of reduced acute care utilization. Am J Hematol 97:1435–1442. https://doi.org/10.1002/ajh.26699

    Article  PubMed  Google Scholar 

  44. Palermo TM, Lalloo C, Zhou C, Dampier C, Zempsky W, Badawy SM et al (2024) A cognitive–behavioral digital health intervention for sickle cell disease pain in adolescents: a randomized, controlled, multicenter trial. Pain 165:164–176. https://doi.org/10.1097/j.pain.0000000000003009

    Article  PubMed  Google Scholar 

  45. Elenga N, Vantilcke V, Martin E, Cuadro E, Selles P, Basset T (2022) Red blood cell exchange in children with sickle cell disease. Int J Hematol 115:107–113. https://doi.org/10.1007/s12185-021-03221-8

    Article  PubMed  Google Scholar 

  46. Alwaheed AJ, Alqatari SG, AlKhafaji DM, Al Argan RJ, Al Sultan OA, AlSulaiman RS et al (2022) Clinical outcome of pre-operative blood transfusion for sickle cell disease patients in post-operative complications. Hosp Pract 50:361–367. https://doi.org/10.1080/21548331.2022.2121574

    Article  Google Scholar 

  47. Salvi PS, Solomon DG, Cowles RA (2022) Preoperative transfusion and surgical outcomes for children with sickle cell disease. J Am Coll Surg 235:530–538. https://doi.org/10.1097/XCS.0000000000000267

    Article  PubMed  Google Scholar 

  48. Ziemba Y, Xu C, Fomani KM, Nandi V, Yuan T, Rehmani S et al (2021) Safety and benefits of automated red cell depletion-exchange compared to standard exchange in patients with sickle cell disease undergoing chronic transfusion. Transfusion (Paris) 61:526–536. https://doi.org/10.1111/trf.16225

    Article  Google Scholar 

  49. Moody KL (2022) Association of sickle cell pain & symptoms on health-related quality of life among pediatric patients. J Pain Sympt Manage 64:304–309. https://doi.org/10.1016/j.jpainsymman.2022.05.003

    Article  Google Scholar 

  50. Osunkwo I, Andemariam B, Minniti CP, Inusa BPD, El Rassi F, Francis-Gibson B et al (2021) Impact of sickle cell disease on patientsʼ daily lives, symptoms reported, and disease management strategies: results from the international <scp>Sickle Cell World Assessment Survey</scp> ( <scp>SWAY</scp> ). Am J Hematol 96:404–417. https://doi.org/10.1002/ajh.26063

    Article  PubMed  PubMed Central  Google Scholar 

  51. Shafrin J, Thom HHZ, Keeney E, Gaunt DM, Zhao LM, Bhor M et al (2021) The impact of vaso-occlusive crises and disease severity on quality of life and productivity among patients with sickle cell disease in the US. Curr Med Res Opin 37:761–768. https://doi.org/10.1080/03007995.2021.1897556

    Article  PubMed  Google Scholar 

  52. Osunkwo I, James J, El-Rassi F, Nero A, Minniti CP, Trimnell C et al (2022) Burden of disease, treatment utilization, and the impact on education and employment in patients with sickle cell disease: a comparative analysis of high- and low- to middle-income countries for the international Sickle Cell World Assessment Survey. Am J Hematol 97:1055–1064. https://doi.org/10.1002/ajh.26576

    Article  PubMed  PubMed Central  Google Scholar 

  53. Heitzer AM, Okhomina VI, Trpchevska A, MacArthur E, Longoria J, Potter B et al (2023) Social determinants of neurocognitive and academic performance in sickle cell disease. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.30259

    Article  PubMed  PubMed Central  Google Scholar 

  54. Bhat D, Babu BV, Surti SB, Ranjit M, Sarmah J, Sridevi P et al (2023) Stigma of sickle cell disease among Indian tribal population: a multi-centric qualitative study. J Natl Med Assoc 115:556–565. https://doi.org/10.1016/j.jnma.2023.09.006

    Article  PubMed  Google Scholar 

  55. Caldwell EP (2020) Health literacy in adolescents with sickle cell disease: the influence of caregiver health literacy. J Special Pediatr Nurs. https://doi.org/10.1111/jspn.12284

    Article  Google Scholar 

  56. Desine S, Eskin L, Bonham VL, Koehly LM (2021) Social support networks of adults with sickle cell disease. J Genet Couns 30:1418–1427. https://doi.org/10.1002/jgc4.1410

    Article  PubMed  Google Scholar 

  57. Travis K, Wood A, Yeh P, Allahabadi S, Chien L-C, Curtis S et al (2020) Pediatric to adult transition in sickle cell disease: survey results from young adult patients. Acta Haematol 143:163–175. https://doi.org/10.1159/000500258

    Article  PubMed  Google Scholar 

  58. Saulsberry AC, Hodges JR, Cole A, Porter JS, Hankins J (2020) Web-based technology to improve disease knowledge among adolescents with sickle cell disease: pilot study. JMIR Pediatr Parent 3:e15093. https://doi.org/10.2196/15093

    Article  PubMed  PubMed Central  Google Scholar 

  59. Sisler I, McClish DK, Valrie C, Villella A, Smith WR (2022) Satisfaction and access to care for adults and adolescents with sickle cell disease: ASCQ-Me quality of care and the SHIP-HU study. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.29948

    Article  PubMed  Google Scholar 

  60. Jones KE, Nyman TM, Daly BP, Jacobson LA, Tarazi RA (2022) Executive functioning predicts adaptive functioning and self-care independence in pediatric sickle cell disease. J Pediatr Psychol 47:206–214. https://doi.org/10.1093/jpepsy/jsab094

    Article  PubMed  Google Scholar 

  61. Crosby LE, Hood A, Kidwell K, Nwankwo C, Peugh J, Strong H et al (2020) Improving self-management in adolescents with sickle cell disease. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.28492

    Article  PubMed  PubMed Central  Google Scholar 

  62. Njoku F, Pugh N, Brambilla D, Kroner B, Shah N, Treadwell M et al (2024) Mortality in adults with sickle cell disease: results from the sickle cell disease implementation consortium (SCDIC) registry. Am J Hematol 99:900–909. https://doi.org/10.1002/ajh.27279

    Article  PubMed  Google Scholar 

  63. Amanor E, Kwarteng A, Larbi A, Fordjour FA, Koranteng KK, Sackey DS et al (2022) Iron stores in steady-state sickle cell disease children accessing care at a sickle cell disease clinic in Kumasi, Ghana: a cross-sectional study. Health Sci Rep. https://doi.org/10.1002/hsr2.934

    Article  PubMed  PubMed Central  Google Scholar 

  64. Loar RW, George A, Varghese NP, Liu AM, Colquitt JL, Pignatelli RH (2021) Assessing the atria in pediatric sickle cell disease: beyond the dilation. Echocardiography 38:189–196. https://doi.org/10.1111/echo.14956

    Article  PubMed  Google Scholar 

  65. Ajibawo T, Okunowo O (2024) Chronic kidney disease and outcomes in hospitalized sickle cell disease patients: a National Inpatient Sample analysis. Eur J Haematol 113:24–31. https://doi.org/10.1111/ejh.14201

    Article  PubMed  Google Scholar 

  66. Zahr RS, Ataga KI, Lebensburger JD, Winer JC (2024) Kidney failure outcomes in children and young adults with sickle cell disease in the United States Renal Data System. Pediatr Nephrol 39:619–623. https://doi.org/10.1007/s00467-023-06136-0

    Article  PubMed  Google Scholar 

  67. Srisuwananukorn A, Raslan R, Zhang X, Shah BN, Han J, Gowhari M et al (2020) Clinical, laboratory, and genetic risk factors for thrombosis in sickle cell disease. Blood Adv 4:1978–1986. https://doi.org/10.1182/bloodadvances.2019001384

    Article  PubMed  PubMed Central  Google Scholar 

  68. Wang Y, Fellah S, Fields ME, Guilliams KP, Binkley MM, Eldeniz C et al (2021) Cerebral oxygen metabolic stress, microstructural injury, and infarction in adults with sickle cell disease. Neurology. https://doi.org/10.1212/WNL.0000000000012404

    Article  PubMed  PubMed Central  Google Scholar 

  69. Weisman JK, Nickel RS, Darbari DS, Hanisch BR, Diab YA (2020) Characteristics and outcomes of osteomyelitis in children with sickle cell disease: a 10-year single-center experience. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.28225

    Article  PubMed  Google Scholar 

  70. Adeniyi O, Akinsete A, Odeghe E, Olowoyeye O, Okeke O, Seyi-Olajide J et al (2022) Cholelithiasis in children and adolescents with sickle cell disease: experience in a resource-limited setting. Ann Afr Med 21:58. https://doi.org/10.4103/aam.aam_81_20

    Article  PubMed  PubMed Central  Google Scholar 

  71. Alshurafa A, Soliman AT, De Sanctis V, Ismail O, Abu-Tineh M, Hemadneh MK et al (2023) Clinical and epidemiological features and therapeutic options of avascular necrosis in patients with sickle cell disease (SCD): a cross-sectional study. Acta Biomed. https://doi.org/10.23750/abm.v94i5.14603

    Article  PubMed  PubMed Central  Google Scholar 

  72. Quelal K, Torres A, Shahi A, Almani MU, Yadav N (2023) Prevalence and predictors of non-rheumatic valvular heart disease in patients with sickle cell disease: insights from the National In-Patient Database in 2016 and 2017. J Investig Med 71:489–494. https://doi.org/10.1177/10815589231162525

    Article  PubMed  Google Scholar 

  73. McCormick M, Richardson T, Warady BA, Novelli EM, Kalpatthi R (2020) Acute kidney injury in paediatric patients with sickle cell disease is associated with increased morbidity and resource utilization. Br J Haematol 189:559–565. https://doi.org/10.1111/bjh.16384

    Article  PubMed  Google Scholar 

  74. Patel U, Desai R, Hanna B, Patel D, Akbar S, Zubair M et al (2020) Sickle cell disease-associated arrhythmias and in-hospital outcomes: insights from the National Inpatient Sample. J Arrhythm 36:1068–1073. https://doi.org/10.1002/joa3.12418

    Article  PubMed  PubMed Central  Google Scholar 

  75. Olaniran KO, Allegretti AS, Zhao SH, Achebe MM, Eneanya ND, Thadhani RI et al (2020) Kidney function decline among black patients with sickle cell trait and sickle cell disease: an observational cohort study. J Am Soc Nephrol 31:393–404. https://doi.org/10.1681/ASN.2019050502

    Article  PubMed  Google Scholar 

  76. Pernell B, Nagalapuram V, Lebensburger J, Lin CP, Baskin ML, Pachter LM (2022) Adverse childhood experiences in children and adolescents with sickle cell disease: a retrospective cohort study. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.29494

    Article  PubMed  Google Scholar 

  77. Sheshadri V, Shabeer P, Santhirapala V, Jayaram A, Krishnamurti L, Menon N (2021) Mortality in sickle cell disease: a population-based study in an aboriginal community in the Gudalur Valley, Nilgiris, Tamil Nadu, India. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.28875

    Article  PubMed  Google Scholar 

  78. Hariharan N, Brunson A, Mahajan A, Keegan THM, Wun T (2020) Bleeding in patients with sickle cell disease: a population-based study. Blood Adv 4:793–802. https://doi.org/10.1182/bloodadvances.2019000940

    Article  PubMed  PubMed Central  Google Scholar 

  79. Ssenyondwa J, George PE, Carlos Bazo-Alvarez J, Mercedes R, Kanywa JB, Naturinda E et al (2020) Impact of sickle cell disease on presentation and progression of paediatric HIV: a retrospective cohort study. Tropical Med Int Health 25:897–904. https://doi.org/10.1111/tmi.13408

    Article  Google Scholar 

  80. Akpa O, Babalola O, Odetunde A, Fagbamigbe A, Fasola F, Brown B et al (2022) Correlates of transfusion transmissible infections among patients with sickle cell disease in Nigeria: case-control study. J Immunoassay Immunochem 43:435–451. https://doi.org/10.1080/15321819.2022.2046601

    Article  PubMed  Google Scholar 

  81. Saxena S, Afolabi-Brown O, Ballester L, Schmucker N, Smith-Whitley K, Allen J et al (2022) Benefit of pulmonary subspecialty care for children with sickle cell disease and asthma. Pediatr Pulmonol 57:885–893. https://doi.org/10.1002/ppul.25845

    Article  PubMed  Google Scholar 

  82. Chan KH, Stark JM, Mosquera RA, Brown DL, Menon N, Nguyen TT et al (2023) Screening for asthma in preschool children with sickle cell disease. J Asthma 60:1787–1792. https://doi.org/10.1080/02770903.2023.2187305

    Article  PubMed  Google Scholar 

  83. Rettenbacher E, Zaal J, Heijboer H, van der Plas EM, Hof M, Biemond BJ et al (2021) Mortality and causes of death from sickle cell disease in the Netherlands, 1985–2017. J Pediatr Hematol Oncol 43:258–265. https://doi.org/10.1097/MPH.0000000000002193

    Article  PubMed  Google Scholar 

  84. Seck M, Ba O, Faye BF, Touré SA, Guèye YB, Dieng N et al (2021) Homozygous sickle cell disease related mortality in Senegal (2011–2020). EJHaem 2:711–715. https://doi.org/10.1002/jha2.273

    Article  PubMed  PubMed Central  Google Scholar 

  85. Jones RS, Ford AL, Donahue MJ, Fellah S, Davis LT, Pruthi S et al (2024) Distribution of silent cerebral infarcts in adults with sickle cell disease. Neurology. https://doi.org/10.1212/WNL.0000000000209247

    Article  PubMed  PubMed Central  Google Scholar 

  86. Ilerhunmwuwa NP, Adeniran O, Inyang L, Wasifuddin M, Hakobyan N, Perry JC et al (2023) Prevalence and outcomes of pulmonary embolism with sickle cell disease: analysis of the Nationwide Inpatient Sample, 2016–2020. Eur J Haematol 111:441–448. https://doi.org/10.1111/ejh.14025

    Article  PubMed  Google Scholar 

  87. Jacob SA, Mueller EL, Cochrane AR, Carroll AE, Bennett WE (2020) Variation in hospital admission of sickle cell patients from the emergency department using the Pediatric Health Information System. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.28067

    Article  PubMed  PubMed Central  Google Scholar 

  88. Adekanye A, Akaba K, Umana A, Mgbe R, Francis P, Okoi-Obuli J (2023) Demographics of tonsillar hypertrophy among sickle cell disease patients in Calabar. Niger J Clin Pract 26:1023–1028. https://doi.org/10.4103/njcp.njcp_59_23

    Article  PubMed  Google Scholar 

  89. Rhodes A, Martin S, Wolters P, Rodriguez Y, Toledo-Tamula MA, Struemph K et al (2020) Sleep disturbance in adults with sickle cell disease: relationships with executive and psychological functioning. Ann Hematol 99:2057–2064. https://doi.org/10.1007/s00277-020-04058-7

    Article  PubMed  PubMed Central  Google Scholar 

  90. Alsalman M, Alhabrati A, Alkuwaiti A, Alramadhan N, AlMurayhil N, Althafar A et al (2022) Depression among patient with sickle cell disease: prevalence and prediction. Niger J Clin Pract 25:1274. https://doi.org/10.4103/njcp.njcp_50_22

    Article  PubMed  Google Scholar 

  91. Abijay CA, Kemper WC, Pham A, Johnson RF, Mitchell RB (2023) Pediatric obstructive sleep apnea and sickle cell disease: demographic and polysomnographic features. Laryngoscope 133:1766–1772. https://doi.org/10.1002/lary.30638

    Article  PubMed  Google Scholar 

  92. Román ME, Highland J, Retherford D, Pan AY, Panepinto JA, Brandow AM (2020) Neuropathic pain is associated with poor health-related quality of life in adolescents with sickle cell disease: a preliminary report. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.28698

    Article  PubMed  Google Scholar 

  93. Harris KM, Preiss L, Varughese T, Bauer A, Calhoun CL, Treadwell M et al (2023) Examining mental health, education, employment, and pain in sickle cell disease. JAMA Netw Open 6:e2314070. https://doi.org/10.1001/jamanetworkopen.2023.14070

    Article  PubMed  PubMed Central  Google Scholar 

  94. Wilson JD, Lanzkron S, Pecker LH, Bediako SM, Han D, Beach MC (2020) Psychosocial and clinical risk factors associated with substance use in observational cohort of patients with sickle cell disease. Subst Use Misuse 55:2205–2212. https://doi.org/10.1080/10826084.2020.1797807

    Article  PubMed  PubMed Central  Google Scholar 

  95. Dike CR, Fittro S, Oster RA, Morrow CD, Brandow A, Demark-Wahnefried W et al (2023) Gastrointestinal symptoms, diagnostic evaluations, and abdominal pathology in children with sickle cell disease. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.30699

    Article  PubMed  Google Scholar 

  96. Raiza Fontes Barros Bomfim J, Orge Anunciação Bacelar C, Marques da Silva Neto M, Salles C, Marice Teixeira Ladeia A, Renata Rissatto-Lago M (2022) Association between hearing impairment, school performance and cognitive function in children and adolescents with sickle cell disease. Trop Med Int Health 27:244–250. https://doi.org/10.1111/tmi.13722

    Article  PubMed  Google Scholar 

  97. Knisely MR, Pugh N, Kroner B, Masese R, Gordeuk V, King AA et al (2020) Patient-reported outcomes in sickle cell disease and association with clinical and psychosocial factors: report from the sickle cell disease implementation consortium. Am J Hematol 95:1066–1074. https://doi.org/10.1002/ajh.25880

    Article  PubMed  PubMed Central  Google Scholar 

  98. Bills SE, Schatz J, Hardy SJ, Reinman L (2020) Social-environmental factors and cognitive and behavioral functioning in pediatric sickle cell disease. Child Neuropsychol 26:83–99. https://doi.org/10.1080/09297049.2019.1577371

    Article  PubMed  Google Scholar 

  99. Heitzer AM, Longoria J, Okhomina V, Wang WC, Raches D, Potter B et al (2021) Hydroxyurea treatment and neurocognitive functioning in sickle cell disease from school age to young adulthood. Br J Haematol 195:256–266. https://doi.org/10.1111/bjh.17687

    Article  PubMed  PubMed Central  Google Scholar 

  100. Fields ME, Mirro AE, Guilliams KP, Binkley MM, Gil Diaz L, Tan J et al (2020) Functional connectivity decreases with metabolic stress in sickle cell disease. Ann Neurol 88:995–1008. https://doi.org/10.1002/ana.25891

    Article  PubMed  PubMed Central  Google Scholar 

  101. Connolly ME, Bills SE, Hardy SJ (2021) Cognitive functioning and educational support plans in youth with sickle cell disease. J Pediatr Hematol Oncol 43:e666–e676. https://doi.org/10.1097/MPH.0000000000002092

    Article  PubMed  Google Scholar 

  102. Saulsberry-Abate AC, Partanen M, Porter JS, Podila PSB, Hodges JR, King AA et al (2021) Cognitive performance as a predictor of healthcare transition in sickle cell disease. Br J Haematol 192:1082–1091. https://doi.org/10.1111/bjh.17351

    Article  PubMed  PubMed Central  Google Scholar 

  103. Robbins MA, Carroll CP, North CS (2020) Psychological symptom screening in an adult sickle cell disease clinic and predictors of treatment follow up. Psychol Health Med 25:1192–1200. https://doi.org/10.1080/13548506.2020.1736313

    Article  PubMed  Google Scholar 

  104. Nawaiseh MB, Yassin AM, Al-Sabbagh MQ, AlNawaiseh A, Zureigat H, Aljbour AlMajali D et al (2024) Abnormal neurologic findings in patients with sickle cell disease without a history of major neurologic events. Neurol Clin Pract. https://doi.org/10.1212/CPJ.0000000000200215

    Article  PubMed  Google Scholar 

  105. Martin S, Roderick MC, Abel C, Wolters P, Toledo-Tamula MA, Fitzhugh C et al (2020) Neurocognitive functioning in symptomatic adults with sickle cell disease: a description and comparison with unaffected siblings. Neuropsychol Rehabil 30:1666–1681. https://doi.org/10.1080/09602011.2019.1598876

    Article  PubMed  Google Scholar 

  106. Heitzer AM, Cohen DL, Okhomina VI, Trpchevska A, Potter B, Longoria J et al (2022) Neurocognitive functioning in preschool children with sickle cell disease. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.29531

    Article  PubMed  PubMed Central  Google Scholar 

  107. Longoria JN, Pugh NL, Gordeuk V, Hsu LL, Treadwell M, King AA et al (2021) Patient-reported neurocognitive symptoms influence instrumental activities of daily living in sickle cell disease. Am J Hematol 96:1396–1406. https://doi.org/10.1002/ajh.26315

    Article  PubMed  PubMed Central  Google Scholar 

  108. Lance EI, Cannon AD, Shapiro BK, Lee L-C, Johnston MV, Casella JF (2021) Co-occurrence of neurodevelopmental disorders in pediatric sickle cell disease. J Dev Behav Pediatr 42:463–471. https://doi.org/10.1097/DBP.0000000000000914

    Article  PubMed  PubMed Central  Google Scholar 

  109. Heitzer AM, Longoria J, Porter JS, MacArthur E, Potter B, Ding J et al (2023) Internalizing symptoms in adolescents with sickle cell disease. J Pediatr Psychol 48:91–103. https://doi.org/10.1093/jpepsy/jsac068

    Article  PubMed  Google Scholar 

  110. Wang W, Freeman M, Hamilton L, Carroll Y, Kang G, Moen J et al (2021) Developmental screening of three-year-old children with sickle cell disease compared to controls. Br J Haematol 195:621–628. https://doi.org/10.1111/bjh.17780

    Article  PubMed  Google Scholar 

  111. Heitzer AM, Rashkin SR, Trpchevska A, Longoria JN, Rampersaud E, Olufadi Y et al (2024) Catechol-O-methyltransferase gene (COMT) is associated with neurocognitive functioning in patients with sickle cell disease. Curr Res Transl Med 72:103433. https://doi.org/10.1016/j.retram.2023.103433

    Article  PubMed  Google Scholar 

  112. AlRyalat SA, Jaber BA-DM, Alzarea AA, Alzarea AA, alosaimi WA, Al Saad M. (2021) Ocular manifestations of sickle cell disease in different genotypes. Ophthalmic Epidemiol 28:185–190. https://doi.org/10.1080/09286586.2020.1801762

    Article  PubMed  Google Scholar 

  113. Oladimeji OI, Adeodu OO, Onakpoya OH, Adegoke SA (2021) Prevalence of ocular abnormalities in relation to sickle cell disease severity among children in South-western, Nigeria. Eur J Ophthalmol 31:2659–2665. https://doi.org/10.1177/1120672120957615

    Article  PubMed  Google Scholar 

  114. Hassan T, Badr M, Hanna D, Arafa M, Elhewala A, Dabour S et al (2021) Retinopathy in Egyptian patients with sickle cell disease. Medicine 100:e28355. https://doi.org/10.1097/MD.0000000000028355

    Article  PubMed  PubMed Central  Google Scholar 

  115. Agouti I, Masson E, Loundou A, Jean E, Arnaud L, Abdili E, et al (2023) Plasma levels of E‐selectin are associated with retinopathy in sickle cell disease. Eur J Haematol 2023;110:271–279. https://doi.org/10.1111/ejh.13902

    Article  PubMed  Google Scholar 

  116. Chen M, Ataga KI, Hankins JS, Zhang M, Gatwood JD, Wan JY et al (2023) Age-related differences in risks and outcomes of 30-day readmission in adults with sickle cell disease. Ann Hematol 102:2329–2342. https://doi.org/10.1007/s00277-023-05365-5

    Article  PubMed  Google Scholar 

  117. Partanen M, Kang G, Wang WC, Krull K, King AA, Schreiber JE et al (2020) Association between hydroxycarbamide exposure and neurocognitive function in adolescents with sickle cell disease. Br J Haematol 189:1192–1203. https://doi.org/10.1111/bjh.16519

    Article  PubMed  Google Scholar 

  118. Elalfy MS, Ibrahim AS, Ibrahim GS, Hussein HMAG, Mohammed HGE, Ebeid FSE (2021) Hidden brain iron content in sickle cell disease: impact on neurocognitive functions. Eur J Pediatr 180:2677–2686. https://doi.org/10.1007/s00431-021-04189-7

    Article  PubMed  Google Scholar 

  119. Afzali-Hashemi L, Baas KPA, Schrantee A, Coolen BF, van Osch MJP, Spann SM et al (2021) Impairment of cerebrovascular hemodynamics in patients with severe and milder forms of sickle cell disease. Front Physiol. https://doi.org/10.3389/fphys.2021.645205

    Article  PubMed  PubMed Central  Google Scholar 

  120. Sayin ES, Sobczyk O, Poublanc J, Mikulis DJ, Fisher JA, Kuo KHM et al (2022) Assessment of cerebrovascular function in patients with sickle cell disease using transfer function analysis. Physiol Rep. https://doi.org/10.14814/phy2.15472

    Article  PubMed  PubMed Central  Google Scholar 

  121. Ampomah MA, Drake JA, Anum A, Amponsah B, Dei-Adomakoh Y, Anie K et al (2022) A case-control and seven-year longitudinal neurocognitive study of adults with sickle cell disease in Ghana. Br J Haematol 199:411–426. https://doi.org/10.1111/bjh.18386

    Article  PubMed  Google Scholar 

  122. Gaartman AE, Sayedi AK, Gerritsma JJ, de Back TR, van Tuijn CF, Tang MW et al (2021) Fluid overload due to intravenous fluid therapy for vaso-occlusive crisis in sickle cell disease: incidence and risk factors. Br J Haematol 194:899–907. https://doi.org/10.1111/bjh.17696

    Article  PubMed  PubMed Central  Google Scholar 

  123. Mawuli G, Dzudzor B, Tachi K, Kuma AAB-A, Odame-Aboagye J, Obeng BM et al (2022) Hepatitis C virus (HCV) infection among patients with sickle cell disease at the Korle-Bu teaching hospital. Virol J 19:73. https://doi.org/10.1186/s12985-022-01797-z

    Article  PubMed  PubMed Central  Google Scholar 

  124. Misra A, Halas R, Kobayashi D, Walters HL, Bondarenko I, Thomas R et al (2023) Outcomes of patients with sickle cell disease and trait after congenital heart disease surgery. Ann Thorac Surg 115:1494–1502. https://doi.org/10.1016/j.athoracsur.2022.04.021

    Article  PubMed  Google Scholar 

  125. Manganas K, Delicou S, Xydaki A, Kourakli A, Evliati L, Vlachaki E et al (2023) Predisposing factors for advanced liver fibrosis in patients with sickle cell disease. Br J Haematol 202:1192–1198. https://doi.org/10.1111/bjh.18970

    Article  PubMed  Google Scholar 

  126. Levesque E, Lim C, Feray C, Salloum C, Quere A, Robin B et al (2020) Liver transplantation in patients with sickle cell disease: possible but challenging—a cohort study. Transpl Int 33:1220–1229. https://doi.org/10.1111/tri.13669

    Article  PubMed  Google Scholar 

  127. Waisbourd-Zinman O, Frenklak R, Hakakian O, Hilmara D, Lin H (2021) Autoimmune liver disease in patients with sickle cell disease. J Pediatr Hematol Oncol 43:254–257. https://doi.org/10.1097/MPH.0000000000001985

    Article  PubMed  Google Scholar 

  128. George PE, Maillis A, Zhu Y, Liu Y, Lane PA, Lam W et al (2023) Are children with sickle cell disease at particular risk from the harmful effects of air pollution? Evidence from a large, urban/peri-urban cohort. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.30453

    Article  PubMed  Google Scholar 

  129. Penkert RR, Azul M, Sealy RE, Jones BG, Dowdy J, Hayden RT et al (2022) Hypothesis: low vitamin A and d levels worsen clinical outcomes when children with sickle cell disease encounter parvovirus B19. Nutrients 14:3415. https://doi.org/10.3390/nu14163415

    Article  PubMed  PubMed Central  Google Scholar 

  130. Grégoire-Pelchat P, Pastore Y, Robitaille N, LeMay S, Khamessan A, Kleiber N et al (2021) Comparison of two vitamin D supplementation strategies in children with sickle cell disease: a randomized controlled trial. Br J Haematol 192:385–394. https://doi.org/10.1111/bjh.17119

    Article  PubMed  Google Scholar 

  131. Jang T, Mo G, Stewart C, Khoury L, Ferguson N, Egini O et al (2021) Obesity and diabetes mellitus in patients with sickle cell disease. Ann Hematol 100:2203–2205. https://doi.org/10.1007/s00277-021-04578-w

    Article  PubMed  Google Scholar 

  132. Rallo MS, Akel O, Kalakoti P, Sun H (2022) Characteristics and outcomes of stroke hospitalizations in patients with sickle cell disease and moyamoya syndrome. J Stroke Cerebrovasc Dis 31:106705. https://doi.org/10.1016/j.jstrokecerebrovasdis.2022.106705

    Article  PubMed  Google Scholar 

  133. Schultz CL, Tchume-Johnson T, Jackson T, Enninful-Eghan H, Schapira MM, Smith-Whitley K (2020) Reproductive intentions in mothers of young children with sickle cell disease. Pediatr Blood Cancer. https://doi.org/10.1002/pbc.28227

    Article  PubMed  PubMed Central  Google Scholar 

  134. da Guarda CC, Yahouédéhou SCMA, Santiago RP, dos Neres JS S, de Fernandes CF L, Aleluia MM et al (2020) Sickle cell disease: a distinction of two most frequent genotypes (HbSS and HbSC). PLoS One 15:e0228399. https://doi.org/10.1371/journal.pone.0228399

    Article  PubMed  PubMed Central  Google Scholar 

  135. Dave K, Desai S, Italia Y, Mukherjee MB, Mehta P, Desai G (2022) Newborn screening and clinical profile of children with sickle cell disease in a tribal area of Gujarat. Indian Pediatr 59:230–233

    Article  PubMed  Google Scholar 

  136. Ali Al-Barazanchi Z, Abdulateef S, Hassan M (2021) Co-Inheritance of α-thalassemia gene mutation in patients with sickle cell Disease: Impact on clinical and hematological variables. Niger J Clin Pract 24:874. https://doi.org/10.4103/njcp.njcp_11_20

    Article  PubMed  Google Scholar 

  137. Adesina OO, Brunson A, Fisch SC, Yu B, Mahajan A, Willen SM et al (2023) Pregnancy outcomes in women with sickle cell disease in California. Am J Hematol 98:440–448. https://doi.org/10.1002/ajh.26818

    Article  PubMed  PubMed Central  Google Scholar 

  138. Cortright L, Buckman C, Tumin D, Holder D, Leonard S (2020) Social determinants of health and emergency department use among children with sickle cell disease. J Pediatr Hematol Oncol 42:e42–e45. https://doi.org/10.1097/MPH.0000000000001669

    Article  PubMed  Google Scholar 

  139. Iliyasu Z, Borodo AM, Jibir BW, Nass NS, Aliyu MH (2021) A child with sickle cell disease can’t live with just anyone” A mixed methods study of socio-behavioral influences and severity of sickle cell disease in northern Nigeria. Health Sci Rep 2021:4. https://doi.org/10.1002/hsr2.222

    Article  Google Scholar 

  140. Agarwal S, Stanek JR, Vesely SK, Creary SE, Cronin RM, Roe AH et al (2023) Pregnancy-related thromboembolism in women with sickle cell disease: an analysis of <scp>National Medicaid Data</scp>. Am J Hematol 98:1677–1684. https://doi.org/10.1002/ajh.27045

    Article  PubMed  Google Scholar 

  141. Oakley LL, Mitchell S, von Rege I, Hadebe R, Howard J, Robinson SE et al (2022) Perinatal outcomes in women with sickle cell disease: a matched cohort study from London. UK Br J Haematol 196:1069–1075. https://doi.org/10.1111/bjh.17983

    Article  PubMed  Google Scholar 

  142. Darlington F, Acha BM, Roshan T, Ikeanyionwu C, Kutse S, Abajue U et al (2020) Opioid-related disorders among pregnant women with sickle cell disease and adverse pregnancy outcomes. Pain Med 21:3087–3093. https://doi.org/10.1093/pm/pnaa188

    Article  PubMed  Google Scholar 

  143. Nkwabong E, Ngoundjou Dongmo P, Tayou C, Nana NT (2022) Outcome of pregnancies among women with sickle cell disease. J Matern Fetal Neonatal Med 35:1108–1112. https://doi.org/10.1080/14767058.2020.1743657

    Article  PubMed  Google Scholar 

  144. Early ML, Luo A, Solow M, Matusiak K, Eke AC, Shehata N et al (2024) Natural history of blood pressure in sickle cell disease pregnancy. Br J Haematol 204:658–667. https://doi.org/10.1111/bjh.19133

    Article  PubMed  Google Scholar 

  145. Early ML, Eke AC, Gemmill A, Lanzkron S, Pecker LH (2023) Severe maternal morbidity and mortality in sickle cell disease in the national inpatient sample, 2012–2018. JAMA Netw Open 6:e2254552. https://doi.org/10.1001/jamanetworkopen.2022.54552

    Article  PubMed  PubMed Central  Google Scholar 

  146. Ha TK, Boulet SL, Cotsonis G, Geary F, Jamieson DJ, Lindsay M (2023) Association of sickle cell disease with severe maternal morbidity. Obstet Gynecol 141:163–169. https://doi.org/10.1097/AOG.0000000000004986

    Article  PubMed  Google Scholar 

  147. Mortara M, Turay MS, Boyle S, Caracciolo C, Bah S, Kargbo H et al (2023) Impact and burden of sickle cell disease in critically ill obstetric patients in a high dependency unit in Sierra Leone—a registry based evaluation. BMC Pregn Childbirth 23:580. https://doi.org/10.1186/s12884-023-05888-9

    Article  Google Scholar 

  148. Kroner BL, Hankins JS, Pugh N, Kutlar A, King AA, Shah NR et al (2022) Pregnancy outcomes with hydroxyurea use in women with sickle cell disease. Am J Hematol 97:603–612. https://doi.org/10.1002/ajh.26495

    Article  PubMed  PubMed Central  Google Scholar 

  149. Pecker LH, Hussain S, Christianson MS, Lanzkron S (2020) Hydroxycarbamide exposure and ovarian reserve in women with sickle cell disease in the Multicenter Study of Hydroxycarbamide. Br J Haematol 191:880–887. https://doi.org/10.1111/bjh.16976

    Article  PubMed  PubMed Central  Google Scholar 

  150. Montironi R, Cupaiolo R, Kadji C, Badr DA, Deleers M, Charles V et al (2022) Management of sickle cell disease during pregnancy: experience in a third-level hospital and future recommendations. J Matern Fetal Neonatal Med 35:2345–2354. https://doi.org/10.1080/14767058.2020.1786054

    Article  PubMed  Google Scholar 

  151. Malinowski AK, Kuo KHM, Tomlinson GA, Palcu P, Ward R, Shehata N (2021) Distinct maternal and fetal pregnancy outcomes in women with sickle cell disease can be predicted using routine clinical and laboratory data. Br J Haematol 194:1063–1073. https://doi.org/10.1111/bjh.17607

    Article  PubMed  PubMed Central  Google Scholar 

  152. Oluwole EO, Okoye CD, Ogunyemi AO, Olowoselu OF, Oyedeji OA (2022) Knowledge, attitude and premarital screening practices for sickle cell disease among young unmarried adults in an urban community in Lagos Nigeria. Pan Afr Med J. https://doi.org/10.11604/pamj.2022.42.8.27705

    Article  PubMed  PubMed Central  Google Scholar 

  153. Vali S, Mukhtar S, Nandi A, Wilson K, Oakley L, El-Toukhy T et al (2020) Cumulative outcome of pre-implantation genetic diagnosis for sickle cell disease: a 5-year review. Br J Haematol 191:875–879. https://doi.org/10.1111/bjh.16930

    Article  PubMed  Google Scholar 

  154. Linton EA, Williams EC, Early ML, Prince E, Stewart RW, Lanzkron S et al (2023) Family planning needs of young adults with sickle cell disease. EJHaem 4:587–594. https://doi.org/10.1002/jha2.711

    Article  PubMed  PubMed Central  Google Scholar 

  155. Stanek CJ, Reich J, Theroux CI, Creary SE, Quinn GP, Nahata L (2023) Reproductive health counseling among youth with sickle cell disease. J Pediatr Adolesc Gynecol 36:393–398. https://doi.org/10.1016/j.jpag.2023.03.002

    Article  PubMed  Google Scholar 

  156. Idris IM, Abba A, Galadanci JA, Mashi SA, Hussaini N, Gumel SA et al (2020) Men with sickle cell disease experience greater sexual dysfunction when compared with men without sickle cell disease. Blood Adv 4:3277–3283. https://doi.org/10.1182/bloodadvances.2020002062

    Article  PubMed  PubMed Central  Google Scholar 

  157. Ribeiro APMR, Silva CS, Zambrano JCC, de Miranda J OF, Molina CAF, Gomes CM et al (2021) 2021 Compensated hypogonadism in men with sickle cell disease. Clin Endocrinol (Oxf) 94:968–972. https://doi.org/10.1111/cen.14428

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

No funding was received in the creation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

RM conceived the idea. RM and EM completed screened the relevant studies for inclusion and wrote the manuscript in its final form. All authors contributed in this work according to the ICJME criteria for authorship.

Corresponding author

Correspondence to Rahma Menshawey.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Menshawey, R., Menshawey, E. On the cutting edge of sickle cell disease: a snapshot narrative review. Egypt J Med Hum Genet 25, 115 (2024). https://doi.org/10.1186/s43042-024-00573-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43042-024-00573-8

Keywords