Open access peer-reviewed chapter - ONLINE FIRST

Innovations in Sickle Cell Care: Navigating the Dynamic Treatment Landscape

Written By

Oluwafemi Ajoyemi Ala

Submitted: 23 February 2024 Reviewed: 22 April 2024 Published: 26 July 2024

DOI: 10.5772/intechopen.1005752

Current Practices in Sickle Cell Disease IntechOpen
Current Practices in Sickle Cell Disease Edited by Marwa Zakaria

From the Edited Volume

Current Practices in Sickle Cell Disease [Working Title]

Marwa Zakaria

Chapter metrics overview

38 Chapter Downloads

View Full Metrics

Abstract

Sickle cell anemia (SCA) is a genetic blood disorder characterized by the presence of abnormal hemoglobin, leading to the formation of sickle-shaped red blood cells. This causes vaso-occlusive crises, chronic anemia, and organ damage. Recent advancements in SCA treatment, including genetic therapies like CRISPR-Cas9, stem cell transplantation, disease-modifying drugs such as hydroxyurea, and telemedicine, offer hope for improved patient outcomes. However, challenges such as access to care and high treatment costs persist. This review discusses recent advances in SCA treatment, highlighting the potential of these therapies to transform patient care and improve quality of life. SCA is a hereditary blood disorder caused by a mutation in the gene that encodes hemoglobin, a protein responsible for carrying oxygen in red blood cells. This leads to production of abnormal hemoglobin, hemoglobin S (HbS). When oxygen levels are low, HbS molecules can polymerize and cause red blood cells to become rigid and assume a sickle shape. These sickle-shaped cells can block blood flow, leading to vaso-occlusive crises, chronic anemia, and organ damage. Recent advancements in the treatment of sickle cell anemia have offered new hope for patients. However, on-going research activities offer hope for continued improvements in the management of this complex disease.

Keywords

  • sickle cell anemia
  • genetic therapies
  • hematopoietic stem cell
  • transplantation
  • disease-modifying drugs
  • telemedicine
  • sickle cell disease
  • gene

1. Introduction

Sickle cell anemia (SCA) is a hereditary blood disorder characterized by the presence of abnormal hemoglobin, which causes red blood cells to become rigid and assume a sickle shape. This leads to various complications, such as pain, organ damage, and increased risk of infections. SCA is caused by a mutation in the gene that encodes the beta-globin subunit of hemoglobin, resulting in the production of abnormal hemoglobin known as hemoglobin S (HbS) [1, 2, 3]. When oxygen levels are low, HbS molecules can polymerize and cause red blood cells to become stiff and sickle-shaped. These sickle cells are prone to breaking down prematurely, leading to anemia, and can also block blood flow, causing vaso-occlusive crises.

Historically, SCA was associated with high mortality rates, particularly in childhood [4, 5]. However, advances in the understanding of the disease and its management have led to significant improvements in outcomes for patients. The discovery of the genetic basis of SCA in the 1950s paved the way for the development of new treatment strategies. The introduction of penicillin prophylaxis in the 1980s significantly reduced the risk of infections, a major cause of morbidity and mortality in SCA patients.

In recent years, there has been a growing recognition of the need for comprehensive care for SCA patients, including preventive measures, early intervention, and on-going monitoring. This approach, known as comprehensive care, aims to prevent complications, manage symptoms, and improve the overall quality of life for SCA patients. Comprehensive care includes regular monitoring of growth and development, screening for complications such as stroke and kidney disease, and providing access to multidisciplinary care teams.

Advances in the management of SCA have also been driven by improvements in supportive care, such as blood transfusions, pain management, and management of acute chest syndrome, a serious complication of SCA characterized by inflammation and blockage of blood vessels in the lungs. The development of disease-modifying therapies, such as hydroxyurea, has also significantly improved outcomes for SCA patients by reducing the frequency of vaso-occlusive crises and the need for hospitalization.

Despite these advancements, challenges remain in the management of SCA. Access to care, particularly for patients in low-resource settings, remains a major barrier to optimal treatment. Additionally, the high cost of treatment can be prohibitive for many patients, particularly in countries without universal healthcare coverage. Furthermore, there is a need for more targeted therapies that address the underlying genetic and molecular mechanisms of SCA, as current treatments mainly focus on symptom management.

Thus, significant advancements have been made in the management of sickle cell anemia, leading to improved outcomes and quality of life for patients. However, there is still much work to be done to address the remaining challenges and develop more effective therapies for this complex and debilitating disease. Continued research, advocacy, and investment in healthcare infrastructure are crucial to improving care and outcomes for SCA patients worldwide.

Advertisement

2. Genetic therapies

Genetic therapies have emerged as a promising avenue for the treatment of sickle cell anemia (SCA). These therapies aim to address the underlying genetic mutation responsible for the disease, offering the potential for a cure or long-term management of symptoms. Recent advancements in genetic engineering techniques, such as CRISPR-Cas9, have sparked significant interest in the field of SCA treatment (Table 1) [7].

TherapyDescriptionCurrent Status
CRISPR-Cas9Gene editing tool that allows for precise modification of the DNA sequence. Can be used to target and correct the specific mutation in the beta-globin gene in SCA patients.Early stages of clinical trials; promising results so far
Gene TherapyUses lentiviral vectors to introduce a functional copy of the hemoglobin gene into hematopoietic stem cells. These modified stem cells can produce normal hemoglobin.Early stages of development; promising results in trials

Table 1.

Overview of genetic therapies for sickle cell anemia [6].

CRISPR-Cas9 is a revolutionary gene editing tool that allows for precise modification of the DNA sequence. In the context of SCA, CRISPR-Cas9 can be used to target and correct the specific mutation in the beta-globin gene that leads to the production of abnormal hemoglobin. Early studies using CRISPR-Cas9 in animal models of SCA have shown promising results, with a significant reduction in sickle cell-related complications (Figure 1).

Figure 1.

Schematic Representation of CRISPR-Cas9 Gene Editing in Sickle Cell Anemia [6, 7]. [CRISPR-Cas9 Gene Editing in Sickle Cell Anemia] (https://example.com/crispr-cas9.jpg).

Early trials of gene therapy for SCA have shown promising results, with some patients achieving a significant reduction in sickle cell-related complications. However, like CRISPR-Cas9 therapy, gene therapy is still in the early stages of development, and more research is needed to determine its long-term safety and efficacy.

Figure 2 illustrates the process of CRISPR-Cas9 gene editing in sickle cell anemia. The CRISPR-Cas9 complex targets the specific mutation in the beta-globin gene, leading to a correction of the genetic defect and the production of normal hemoglobin.

Figure 2.

Pictorial Analysis of Gene Editing in Sickle Cell Anemia Disease [7, 8].

Clinical trials using CRISPR-Cas9 in human patients with SCA are still in the early stages, but the results so far are encouraging. Some patients have experienced a complete remission of symptoms, including a reduction in the frequency of vaso-occlusive crises and an improvement in overall quality of life. However, more research is needed to fully understand the long-term effects and safety profile of CRISPR-Cas9 therapy in SCA patients.

In addition to CRISPR-Cas9, other genetic approaches are also being explored for the treatment of SCA. One such approach is gene therapy using lentiviral vectors to introduce a functional copy of the hemoglobin gene into hematopoietic stem cells [79]. These modified stem cells can then be transplanted back into the patient, where they can produce normal hemoglobin and replace the defective red blood cells.

Genetic therapies hold great promise for the treatment of sickle cell anemia. Advances in gene editing techniques such as CRISPR-Cas9 and gene therapy using lentiviral vectors offer new hope for patients with this debilitating disease [6]. While more research is needed to fully understand the long-term effects and safety of these therapies, the early results are promising, and genetic therapies may soon revolutionize the treatment of sickle cell anemia.

Thus, genetic therapies are at the forefront of sickle cell anemia treatment research, offering the potential for a cure or long-term management of symptoms. While still in the early stages of development, therapies such as CRISPR-Cas9 and gene therapy show promise in correcting the genetic defect underlying sickle cell anemia. Continued research and clinical trials are needed to fully understand the safety and efficacy of these therapies and to bring them to the patients who need them.

2.1 Stem cell transplantation

Hematopoietic stem cell transplantation (HSCT), also known as bone marrow transplantation, is a potentially curative treatment for sickle cell anemia (SCA), with the greatest cure rates attributed to this treatment plan. However, the use of HSCT is limited by the lack of suitable human leukocyte antigen (HLA)-matched donors and decreased application in older patients with significant morbidity (Table 2) [10, 11].

TechniqueDescriptionAdvantagesChallenges
Reduced-intensityUses less toxic conditioning regimens, reducing the risk of complications such as graft rejection and GVHD.Expanded pool of eligible donors, increased success rate of transplantation.Limited availability in some regions, high cost of transplantation.
Gene-edited stem cellsInvolves genetically modifying stem cells to correct the mutation that causes SCA.Potential for a more precise and targeted treatment.Limited availability, long-term effects unknown.
Haploidentical transplantationUses stem cells from a partially matched donor, typically a family member.More readily available than fully matched donors.Increased risk of GVHD, long-term effects unknown.

Table 2.

Comparison of stem cell transplantation techniques for sickle cell anemia [10, 11].

Myeloablative, HLA-identical sibling transplantation in children with sickle cell disease (SCD) offers excellent long-term survival, with overall and event-free survival rates of 95 and 92%, respectively. However, the risk of graft-versus-host-disease, infections, infertility, and other long-term transplant complications further limits its widespread use [12, 13, 14].

HSCT procedure involves replacing the patient’s diseased bone marrow with healthy donor marrow that can produce normal hemoglobin. Stem cell transplantation offers the possibility of a permanent cure for SCA by providing a new source of healthy red blood cells.

Historically, stem cell transplantation was associated with significant risks, including graft rejection, graft-versus-host disease (GVHD), and transplant-related mortality. However, recent advancements in transplant techniques have significantly improved outcomes and reduced these risks. One such advancement is the use of reduced-intensity conditioning (RIC) regimens [15], which are less toxic than traditional conditioning regimens, allowing the extension of this modality to children and adults with significant morbidity [15, 16, 17, 18]. This has expanded the pool of eligible donors and increased the success rate of transplantation. However, these approaches are also associated with an increased risk of graft failure. Thus, the optimal RIC regimen that strikes the optimal balance between maximizing the rate of stable engraftment while minimizing transplant-related morbidity and mortality is not fixed and thereby relatively unknown [5, 8, 16, 17, 18, 19, 20, 21]. Alternative donor transplants, most prominently, partial HLA-mismatched related transplants (haploidentical), are being investigated with promising initial results [22].

In addition to reduced-intensity conditioning regimens, other advancements in stem cell transplantation for SCA include the use of gene-edited stem cells and haploidentical transplantation. Gene-edited stem cells are stem cells that have been genetically modified to correct the mutation that causes SCA. This approach offers the potential for a more precise and targeted treatment for SCA patients (Figure 3). Haploidentical transplantation involves using stem cells from a partially matched donor, typically a family member [5, 19, 20, 21, 22, 23, 24, 25]. This approach has the advantage of being more readily available than fully matched donors, making it a potentially more accessible treatment option for SCA patients.

Figure 3.

Suggested Approaches for HSCT for patients with Sickle Cell Anemia Disease [5, 8, 18, 19, 20, 21, 22, 23, 24].

Allogeneic transplant is curative for sickle cell anemia disease but has mainly been performed in children with severe disease, usually for overt strokes, recurrent acute chest syndrome, or recurrent vaso-occlusion pain episodes despite adherence to hydroxyurea therapy [21, 22, 23, 24, 25, 26, 27].

Also, for most of the cases seen globally, bone marrow-derived stem cells from HLA-matched sibling donors are primarily used, following myeloablative conditioning. Recent approaches are exploring the use of stem cells derived from blood, peripheral blood stem cells (PBSCT), and umbilical cord stem cells (UCBT) from a related newborn baby. Updates on the results of HLA-matched sibling HSCT for sickle cell disease performed worldwide between 1986 and 2013, reported to the European Blood and Marrow Transplant, Eurocord, and the Center for International Blood and Marrow Transplant Research (CIBMTR), were recently published [5, 8, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26]. More so, another benefit of HSCT includes abating progressive organ dysfunction [21, 22, 23, 24, 25, 26], which is associated with early death in older patients with sickle cell anemia disease (Table 3).

Myeloablative matched sibling donor transplant and related cord blood transplantNon-Myeloablative matched sibling donor transplantPartially mismatched related donor (haploidentical) BMT
Advantages
  • Potential for cure

  • Large cumulative experience

  • Good rates of engraftment

  • Attenuates progressive

  • vasculopathy/end-organ damage

  • Disease-free survival: 85%

  • Overall survival: 97%

  • (Long follow up: 10–15 years)

  • Does not exclude recipients with chronic organ damage

  • More applicable to adults with severe SCD

  • Disease-free survival: 79%

  • Overall survival: 95%

  • (Long follow up: 5–10 years)

  • Does not exclude recipients with end-organ damage

  • Available to most patients (more than 50% of patients with SCD will have a suitable donor)

  • Disease-free survival: 58%

  • Overall survival: 100%

  • (Follow up: < 5 years)

Disadvantages
  • Limited availability (∼10% of SCD patients with suitable donor)

  • Patient with end-organ damage usually excluded

  • Not recommended in adults with severe SCD

  • Graft rejection

  • Transplant-related mortality

  • Complications such as acute lung injury, infertility, endocrine and metabolic issues, and chronic GVHD

  • Limited availability (∼10% of SCD patients with suitable donor)

  • Higher risk of graft failure

  • Limited experience

  • Graft rejection

  • Transplant-related mortality

  • Late effects such as endocrine and metabolic issues and chronic GVHD

  • Limited experience

  • Higher risk of graft rejection

  • Late effects

Table 3.

Advantages and disadvantages of different transplant approaches in patients with severe sickle cell disease [5, 12, 13, 14, 28, 29].

GVHD: Graft-versus-host disease; SCD: Sickle cell disease.

It is worthy of note that for all genotypes, disease-related morbidity is the driving factor in pursuing an HSCT. Also, HSCT for adults with sickle cell disease is better tolerated with a low-intensity regimen but may require prolonged immune suppression to maintain stable mixed-donor chimerism (recipient and donor cells) (Table 4).

Matched sibling donor transplantMatched unrelated donor transplant (MUD)Partially mismatched related donor (haploidentical); unrelated cord blood transplant
Overt stroke or central nervous system event lasting >24 hOvert stroke or any neurologic deficit lasting >24 hRecurrent stroke despite adequate chronic blood transfusion therapy or progressive central nervous system changes
Impaired neuropsychological function with abnormal cerebral magnetic resonance imaging and angiographyElevated TCD velocity unresponsive to hydroxyurea or chronic blood transfusion therapySevere SCD symptoms unresponsive to hydroxyurea therapy
Elevated TCD velocity unresponsive to hydroxyurea or chronic blood transfusion therapyRecurrent acute chest syndrome despite hydroxyurea therapy
Recurrent acute chest syndrome despite hydroxyurea therapyRecurrent severe pain episodes despite hydroxyurea therapy
Recurrent severe pain episodes despite hydroxyurea therapyRed cell alloimmunization (transfusion support) plus established indication for chronic blood transfusion therapy
Red cell alloimmunization plus established indication for chronic blood transfusion therapyPulmonary hypertension or an echocardiographic finding of tricuspid valve regurgitant jet velocity ≥ 2.7 m/s
Pulmonary hypertension or an echocardiographic finding of tricuspid valve regurgitant jet velocity ≥ 2.7 m/sBone and joint involvement
Recurrent priapismRecurrent priapism
Sickle nephropathySickle nephropathy
Bone and joint involvement
Sickle retinopathy
Stage I or II sickle cell lung disease

Table 4.

Current indications for HSCT in patients with severe sickle cell disease unresponsive to hydroxyurea therapy [22, 26, 30].

AVN = avascular necrosis; TCD = transcranial Doppler; VOE = vaso-occlusive pain episodes; SCD: Sickle cell disease.

Clinical trials using these advanced stem cell transplantation techniques have shown promising results. Patients who have undergone stem cell transplantation have experienced a significant reduction in sickle cell-related complications and an improvement in overall quality of life. Some patients have even achieved a complete cure, with normal hemoglobin levels and no further need for transfusions.

Therefore, successful HSCT offers long-term protection from clinical and sub-clinical vaso-occlusion associated with sickle cell anemia disease, regardless of donor source, and minimizing late effects following HSCT is now a major therapeutic goal being constantly aimed. With a successful HSCT, end-organ complications that commonly develop in patients with sickle cell anemia disease, including stroke, pulmonary hypertension, acute chest syndrome, proteinuria, and haematuria, are usually not observed in patients after such transplant process, as defined by the stable engraftment of donor cells [23, 31, 32, 33, 34, 35, 36, 37, 38].

Another major concern that has to be ruled out in order to measure the success of HSCT is gonadal insufficiency and infertility; specifically ovarian failure in females and low testosterone levels in males or hypogonadotropic hypogonadism. Thus, to ascertain the success of HSCT in this situation, health-related quality of life (QOL), which is determined by measures of physical, psychological, and social functioning, must have significantly improved after a certain period, starting from say 1 year, following successful HSCT.

The successes recorded so far with HSCT concerning improved transplant outcomes for patients with sickle cell disease have also increased the demand for HSCT for patients with SCD from low-income countries, despite the huge cost implications. The HSCT demand is partly driven by the lack of standard care (hydroxyurea, chronic blood transfusion therapy, and comprehensive care programs) in their countries of origin, and is largely dependent on family preference and socio-economic status. However, facilities for both acute and late-effects management post-transplant are usually lacking in these low-income countries. Thus, following HSCT in high-income countries, patients living in low-income countries usually need long-term follow-up care abroad at significant costs. Medical facilities in low-income countries should address local challenges such as parasitaemia, malnutrition, availability of blood products, and other laboratory support services while developing a long-term strategy for managing patients who have undergone the procedure [33, 34, 35, 36, 37, 39, 40]. For patients residing in low-income countries seeking HSCT abroad, extensive discussion with the patient and family regarding the risk-benefit ratio, as well as a contingency plan to manage transplant-related complications for at least 2 years after the procedure at a hospital with adequate transplant expertise, are warranted.

Lastly in the section, there are still on-going trials that will help determine the true impact of HSCT on safety, progressive vasculopathy, and chronic organ dysfunction; these will also help put more clarity and equally clear doubts as to whether patients with sickle cell anemia disease are at greater risk for declining organ function or whether RIC regimens offer better outcomes following HSCT. Thus, more research is needed to further improve the safety and efficacy of stem cell transplantation and to make it a more accessible treatment option for all sickle cell anemia patients.

Advertisement

3. Disease-modifying therapies

Disease-modifying therapies play a crucial role in the management of sickle cell anemia (SCA), aiming to reduce the frequency and severity of painful crises, improve quality of life, and prevent long-term complications. These therapies target various aspects of the underlying pathophysiology of SCA, including the production of abnormal hemoglobin and the adhesion of sickle cells to blood vessel walls, which worsen vaso-occlusion (Table 5) [23, 26, 31, 32, 33, 34, 35, 36, 37, 39, 42].

TherapyMechanism of ActionClinical Status
HydroxyureaIncreases production of foetal hemoglobin, which can substitute for abnormal hemoglobin S.Widely used; proven efficacy in clinical trials
VoxelotorIncreases the affinity of hemoglobin for oxygen, preventing the polymerization of hemoglobin S.FDA-approved for the treatment of SCA
CrizanlizumabTargets P-selectin, a cell adhesion molecule involved in the adhesion of sickle cells to blood vessel walls.FDA-approved for the prevention of vaso-occlusive crises in SCA

Table 5.

Overview of few disease-modifying therapies for sickle cell anemia [26, 37, 41].

Hydroxyurea is one of the oldest and most widely used disease-modifying therapies for SCA. It works by increasing the production of foetal hemoglobin, which can substitute for the abnormal hemoglobin S and reduce sickle cell-related complications. Clinical studies have shown that hydroxyurea can reduce the frequency of painful crises, hospitalizations, and the need for blood transfusions in SCA patients. It is also associated with improvements in overall survival and quality of life.

Newer drugs, such as voxelotor (formerly known as GBT440) and crizanlizumab, offer more targeted approaches to treating SCA. Voxelotor is a hemoglobin oxygen affinity modulator that works by increasing the affinity of hemoglobin for oxygen, preventing the polymerization of hemoglobin S and the formation of sickle cells. Clinical trials have shown that voxelotor can significantly reduce the percentage of sickle cells in the blood and improve hemoglobin levels in SCA patients.

Crizanlizumab is a monoclonal antibody that targets P-selectin, a cell adhesion molecule involved in the adhesion of sickle cells to blood vessel walls. By inhibiting P-selectin, crizanlizumab reduces the adhesion of sickle cells and the formation of vaso-occlusive events. Clinical trials have demonstrated that crizanlizumab can reduce the frequency of painful crises and the need for hospitalizations in SCA patients [23, 26, 31, 32, 33, 34, 35, 36, 37, 39].

In addition to these pharmacological therapies, on-going research is exploring the use of gene therapy to boost the production of foetal hemoglobin and prevent sickle cell complications. Gene therapy approaches aim to introduce a functional copy of the hemoglobin gene into stem cells, allowing them to produce normal hemoglobin and reduce the production of abnormal hemoglobin S. Early studies using gene therapy for SCA have shown promising results, with some patients achieving sustained increases in foetal hemoglobin levels and improvements in clinical outcomes.

Table 6 illustrates the mechanism of action of disease-modifying therapies in sickle cell anemia. Hydroxyurea increases the production of foetal hemoglobin; voxelotor increases the affinity of hemoglobin for oxygen, and crizanlizumab targets P-selectin to reduce the adhesion of sickle cells to blood vessel walls.

Table 6.

Mechanism of action of various disease-modifying therapies in sickle cell anemia [26, 43, 44].

Before concluding this section, it would be unfair not to mention the impact of pain initiatives and management in the assessment of sickle cell anemia disease. It is, therefore, imperative to understand the synergy of the drugs and therapies used in the management of pain with the other disease-modifying therapies; this synergy helps to get the best clinical, socio-economic, and humanistic outcomes from the management of sickle cell disease, and also equally promote the lives of the patients while seeking permanent answers in terms of therapies. Here below are some of the illustrations of the efforts that have been made to standardize the management of pain globally and also discuss the processes involved in the management of severe pain, especially in an emergency room (Figures 4 and 5).

Figure 4.

Evidenced-Based Standard Practice of Care Algorithm for Vaso-Occlusive Pain Episodes [26, 45, 46, 47].

Figure 5.

Chart of Severe Acute Pain Management for Sickle Cell Disease in the Emergency Room [45, 46, 48, 49].

Therefore, disease-modifying therapies play a crucial role in the management of sickle cell anemia, offering targeted approaches to reduce the frequency and severity of complications. Hydroxyurea, voxelotor, and crizanlizumab, are examples of therapies that have shown promise in improving outcomes for SCA patients. On-going research into gene therapy holds the potential to further improve treatment options and ultimately find a cure for this complex and debilitating disease.

Advertisement

4. Telemedicine and remote monitoring

Telemedicine and remote monitoring have transformed the landscape of sickle cell anemia (SCA) management, offering new avenues for patient care, particularly in rural and underserved areas. These technologies encompass a wide range of applications, including virtual consultations, remote monitoring of vital signs, and electronic health record (EHR) systems, all of which contribute to improved patient outcomes and quality of life.

One of the key advantages of telemedicine in SCA management is its ability to bridge the gap between patients and healthcare providers, allowing for more frequent and convenient access to care. This is especially important for SCA patients, who often require regular monitoring and may experience sudden severe symptoms that necessitate immediate attention. Telemedicine enables patients to consult with healthcare providers remotely, reducing the need for frequent hospital visits and improving overall adherence to treatment regimens.

Remote monitoring technologies also play a crucial role in SCA management, allowing for the continuous monitoring of vital signs and disease progression. For example, wearable devices can track parameters such as heart rate, oxygen saturation, and activity levels, providing valuable insights into a patient’s health status. These devices can alert healthcare providers to potential complications, enabling early intervention and preventing serious health issues.

In addition to improving patient care, telemedicine and remote monitoring technologies have facilitated the participation of SCA patients in clinical trials and research studies (Table 7). By allowing patients to participate remotely, these technologies have expanded the pool of eligible participants and increased the diversity of study populations, leading to a better understanding of the disease and its treatment.

BenefitDescription
Increased access to careAllows patients in rural and underserved areas to consult with healthcare providers remotely.
Improved adherence to treatmentEnables patients to monitor their condition and adhere to treatment regimens more effectively.
Enhanced patient engagementEncourages patients to take a more active role in managing their health and well-being.
Early detection of complicationsAllows for the early detection of potential complications, enabling timely intervention.
Facilitates participation in clinical trialsEnables patients to participate in clinical trials and research studies remotely.

Table 7.

Benefits of telemedicine and remote monitoring in sickle cell anemia management [48, 50].

With the use of telemedicine and remote monitoring technologies in the management of sickle cell anemia, patients can consult with healthcare providers, access medical records, and monitor their condition remotely, improving overall health outcomes and quality of life.

Looking ahead, future advancements in telemedicine hold the potential to further enhance patient care and support for those living with SCA. For example, artificial intelligence (AI) algorithms can analyze large datasets of patient information to identify patterns and predict disease progression, enabling more personalized treatment approaches. Additionally, telemedicine platforms can be integrated with EHR systems to provide a seamless and comprehensive view of a patient’s health history, enabling more informed decision-making by healthcare providers.

Thus, telemedicine and remote monitoring technologies have revolutionized the management of sickle cell anemia, offering new opportunities for patient care and support. These technologies have improved access to care, adherence to treatment, and patient engagement, leading to better outcomes for SCA patients. Continued advancements in telemedicine hold the potential to further enhance patient care and support for those living with this complex and challenging disease.

Advertisement

5. Patient education and support programs

Patient education and support programs play a critical role in the management of sickle cell anemia (SCA), empowering patients to take control of their health and improve their quality of life. These programs provide a range of services, including disease education, lifestyle modification guidance, and psychosocial support, all aimed at helping patients cope with the challenges of living with a chronic illness.

One of the key components of patient education programs is disease management education. Patients with SCA often require specialized care to manage their condition effectively. Education programs provide patients with information about their disease, including its causes, symptoms, and treatment options. This information helps patients make informed decisions about their care and empowers them to communicate effectively with their healthcare providers.

Lifestyle modification guidance is another important aspect of patient education programs for SCA. Patients with SCA can benefit from making changes to their lifestyle, such as adopting a healthy diet, staying hydrated and avoiding activities that can trigger a sickle cell crisis. Education programs provide patients with practical tips and strategies for managing their lifestyle to minimize the impact of SCA on their daily lives.

Psychosocial support is also a crucial component of patient education programs for SCA. Living with a chronic illness can be challenging, and patients may experience feelings of isolation, anxiety, or depression. Education programs provide patients with the tools and resources they need to cope with these challenges, including access to mental health services, support groups, and counseling.

Patient engagement in their care is a key goal of patient education programs for SCA. Studies have shown that patients who are actively involved in their care are more likely to adhere to their treatment regimens and achieve better health outcomes. Patient education programs help patients develop the skills and confidence they need to actively participate in their care, leading to improved overall health and well-being.

Peer support networks and advocacy groups also play a vital role in supporting patients with SCA. These groups provide a platform for patients to connect with others who are living with the same condition, share experiences, and provide mutual support. Peer support networks and advocacy groups also work to raise awareness about SCA and advocate for better access to care and treatment for patients.

Table 8 illustrates the components of patient education and support programs for sickle cell anemia, including disease management education, lifestyle modification guidance, psychosocial support, and patient engagement.

ComponentDescription
Disease management educationProvides information about the causes, symptoms, and treatment options for sickle cell anemia.
Lifestyle modification guidanceOffers practical tips and strategies for managing lifestyle factors that can impact SCA.
Psychosocial supportProvides access to mental health services, support groups, and counseling for emotional support.
Patient engagementEmpowers patients to take an active role in their care and make informed decisions about treatment.

Table 8.

Components of patient education and support programs for sickle cell anemia [24, 31, 35, 51].

It would be unfair to conclude this section without mentioning the influence of the attitude of patients, the level of knowledge of sickle cell anemia disease by healthcare providers who are crucial stakeholders, and the overall social determinants of treatment-seeking traits among the disease sufferers, as they all impact on the health outcomes and education of patients in terms of symptoms prevention and/or alleviation, as well as the reduction in rates of incessant hospitalization.

Figures 6 and 7 are meant to further explain some issues surrounding the knowledge of health workers, attitude of patients, and some social determinants as highlighted.

Figure 6.

Theory of Self-Care Management for Sickle Cell Disease [36, 39].

Figure 7.

Social Determinants of Sickle Cell Disorders – The Sickle Cell, Individual, Community Knowledge and Environment (SICKLE) Model [36, 39, 52].

Therefore, patient education and support programs are essential for empowering patients with sickle cell anemia, in order to manage their condition effectively. These programs provide patients with the information, tools, and support they need to cope with the challenges of living with a chronic illness. By educating and supporting patients, these programs help to improve treatment adherence, health outcomes, and overall quality of life for individuals with sickle cell anemia.

Advertisement

6. Public health initiatives

Public health initiatives are crucial in addressing the challenges of sickle cell anemia (SCA) by raising awareness, improving access to care, and supporting affected individuals and communities. These initiatives encompass a range of strategies, including education, screening, and advocacy, aimed at reducing the burden of SCA and improving outcomes for patients [23, 31, 32, 33, 34, 35, 36, 37, 39].

One of the key components of public health initiatives for SCA is raising awareness about the disease among healthcare providers, policymakers, and the general public. Many people are unaware of the impact of SCA and the importance of early diagnosis and treatment. Public health campaigns can help educate the public about SCA, its symptoms, and the available treatment options, leading to earlier diagnosis and better outcomes for patients.

Efforts to educate healthcare providers about SCA are also critical. Many healthcare providers may not be familiar with the unique challenges faced by patients with SCA or the latest treatment guidelines [36, 37]. Public health initiatives can provide training and resources to healthcare providers to improve their knowledge and understanding of SCA, leading to better care for patients.

Screening programs are another important component of public health initiatives for SCA. Screening can help identify individuals with SCA or sickle cell trait carriers, allowing for early intervention and treatment [36]. Screening programs can also provide valuable data for public health researchers to better understand the prevalence of SCA and its impact on communities [36, 37, 39].

Family planning and genetic counseling are also key aspects of public health initiatives for SCA. Individuals who are carriers of the sickle cell trait have a 25% chance of passing the disease on to their children if their partner is also a carrier. Genetic counseling can help individuals understand their risk and make informed decisions about family planning, reducing the prevalence of SCA in future generations.

Such other information as seen above will also help to elaborate the importance of the various components of public health initiatives for sickle cell anemia, including raising awareness, educating healthcare providers, screening programs, family planning, and genetic counseling.

In likewise manner, challenges that hamper the effects of public health initiatives can also have a lasting impact on the outcomes of health, both either expected or unintended as the case may be, once policies and initiatives are distorted (Table 9).

Challenges/Burden of SCD in NigeriaGovernment policies and programmes to address the challenges
  • The 2018 National Demographic Health Survey (NDHS) report for the country put the prevalence of SCD to be highest in the South West (2%), lowest in the South (0.3%) and 21% HbAS and 5% HbAC for southwest and overall prevalence of 1% among the children below 5 years

  • Establishment of six zonal Centres of Excellence for SCD which are equipped with HPLC for early detection and comprehensive care of diagnosed babies

  • Creation of a National Desk for SCD at the FMoH

  • Review of the existing National Guideline for the Management and Control of SCD

  • SCD is among the top 10 non-communicable diseases (NCDs) causing significant disability, morbidity and mortality impacting negatively on the attainment of Sustainable Development Goals (SDGs) 1, 3, 4 and 10

  • Protocol for the Universal Newborn Screening for SCD

  • Poor integration of SCD prevention and control services with other health and nonhealth services especially maternal and child health services

  • Expansion of the national immunization schedule to include pneumococcal and influenza vaccination for children with SCD

  • Cultural beliefs and ignorance (myths) about SCD across the country

  • Launching of the National Multisectoral Action Plan for SCD and other Prioritized NCDs

  • Too many uncoordinated activities of NGOs in SCD community in Nigeria

  • Streamlining and coordinating activities of NGO/CSOs in the SCD community

  • Non-prioritization of SCD due to poor understanding of the contribution and linkage of the disease to poverty and mortality indices in Nigeria by the major development partners

  • High level advocacy resulting in legislative frameworks that ensure SCD is accorded priority attention required considering the high burden of the disease

  • Inability of government to mobilize the much-needed resources for SCD interventions

  • Multilateral collaboration and partnerships with international organization such as WHO and local industries and NGOs

  • Scaling up high level advocacy and dialog for SCD

Table 9.

Example of feedback from policymakers (hospital management staff and government representatives from Nigeria’s Federal Ministry of Health) to assess the challenges and resolutions [19, 35, 45].

Furthermore, assessment and analysis of data on the causes of death as well as the path to death of sickle cell disease sufferers are very important in public health research, as these will help to give futuristic projections to causes of mortalities and possible ways of avoiding such future occurrences (Figure 8) (Table 10).

Figure 8.

Flowchart showing prevalence, cause-specific mortality, and total sickle cell disease mortality estimation process [35, 45, 48, 53].

Experience/ChallengesStrategies/Expectations
Access
  • Provision of more specialized sickle cell centres

  • Capacity building, enhancement, and job protection for health workers to enable them put in their best

  • Delay before accessing care. Quantified to be about 2 h or more before talking to a doctor. Turnaround time for investigations can take up to 3 days for one consultation

  • Difficulty in accessing a preferred healthcare worker

  • Healthcare workers are unfriendly

  • Specialist SCD centres are few and difficult to access

  • Unorthodox, and uncertified care is very much available and easy to access

Contextual knowledge (knowledge of healthcare providers on needs of patients)
  • Every clinic day should be a unique journey with exciting new things to look forward to

  • Family support is poor

  • Clinic days are too routine and rigid and often difficult to fit into individual patient’s schedules

Communication
  • Provision of more specialized sickle cell centres and employment of more healthcare providers especially social workers and haemoglobinopathy counselors

  • Healthcare workers are too much in a hurry to write drugs and dismiss a patient without listening to the problems of the patient

  • The guinea pig mentality. Sometimes patients believe that everything done for the patient is just to make him/her give blood for research

Comprehensiveness
  • Consultations not holistic as social and spiritual issues are seldom attended to

  • Multidisciplinary team management

  • Lack of counseling that may lead to the philosophy “once I am fine, no need to go to the clinic”, self-medication and patronage of quacks may then follow

  • Health education and public enlightenment for all

  • Improvement of emergency care and blood transfusion services

  • Stigma

Coordination
Emergency care and blood transfusion are inefficient
  • The National Health Insurance Service (NHIS) is unfriendly to patients. Many times, routine drugs for SCD are not available in the scheme or out of stock

  • Government should make NHIS more friendly and effectively accommodate sickle cell patient

High cost of services
  • Increase health insurance coverage and inclusion of hydroxyurea in NHIS drugs list

  • Cost implications, an average of 10,000 naira ($20) on drugs monthly, excluding transport, investigations, consultation fee and others

  • Important drugs for care such as hydroxyurea are expensive and difficult to access

Table 10.

Quality of healthcare services (examples of feedback from patients’ care givers, patients, and support non-governmental organizations in some hospitals in Nigeria) [19, 35, 45, 54].

Thus, public health initiatives play a critical role in addressing the challenges of sickle cell anemia by raising awareness, improving access to care, and supporting affected individuals and communities. Continued investment in these initiatives is essential to reduce the burden of SCA globally and improve outcomes for patients.

Advertisement

7. Collaborative research efforts

Collaborative research efforts have been instrumental in advancing our understanding of sickle cell anemia (SCA) and developing new treatment options for this complex disease. These collaborative efforts involve partnerships between academia, industry, and patient advocacy groups, bringing together diverse expertise and resources to address the challenges of SCA comprehensively.

One of the key outcomes of collaborative research efforts in SCA is the identification of novel drug targets. Researchers have identified specific pathways and molecules involved in the pathogenesis of SCA, leading to the development of targeted therapies that can modify the course of the disease. For example, drugs targeting the adhesion of sickle cells to blood vessel walls, such as crizanlizumab, have shown promise in reducing the frequency of vaso-occlusive crises in SCA patients (Figures 9 and 10).

Figure 9.

Translating Clinical Care of Sickle Cell Disease to low-resource Countries through International Research Collaborations (A) [45, 48].

Figure 10.

Translating Clinical Care of Sickle Cell Disease to low-resource Countries through International Research Collaborations (B) [45, 48].

List of some of the therapies and drugs developed through collaborative efforts is shown in Table 11.

Drug compoundDrug action
Target: Erythrocyte rheology. Reducing polymerization, and improving cellular hydration
Hydroxyurea*Increases HbF. reduces HbS polymerization
Senicapoc*Improves RBC dehydration
Metformin*Increases HbF
Aes-103*Prevents HbS polymerization
Voxelotor*Prevents HbS polymerization
Target: Reducing cellular adhesion and vaso-occlusion
Hydroxyurea*Improves red cell rheology
Crizanlizumab*P-selectin inhibitor
Rivipansel*Pan-selectin inhibitor
Intravenous lg*Neutrophil integrin Mac-1 inhibition
Tinzaparin*Reduces RBC adhesion
Dalteparin*Reduces RBC adhesion
Sevuparin*Reduces RBC adhesion
Eptifibatide*Inhibits platelet aggregation
NKTT120*iNKT-blocking monoclonal antibody
Ticagrelor*Inhibits platelet aggregation
Prasugrel*Inhibits platelet aggregation
Target: improving endothelial dysfunction
Hydroxyurea*Nitric oxide donor
L-glutamine*Nitric oxide donor
Haptoglobin*Hemoglobin scavenger
Oral or intravenous nitrite*Nitric oxide donor
Arginine*Nitric oxide donor
Inhaled nitric oxide*Nitric oxide donor
Antioxidants*Reduce production of ROS
Target: Improving sterile inflammation
Haemopexin/haptoglobin*Haem/Hb-binding proteins, antioxidant
MP4CO*Modulates HO-1 and inflammation
Various anti oxidants*Reduce crisis and inflammation
TLR4-inhibition**Mediates haem-induced inflammation
DNAse-1**Dissolves NET produced by neutrophils
Canakinumab*IL-1β inhibitor
Montelukast*Cysteinyl leukotrienes receptor antagonist
Simvastatin*Statin therapy to protect vascular endothelium
Anakinra**IL-1 inhibitor

Table 11.

Short list of currently approved drugs and study trials of potential future treatments for sickle cell disease through various collaborative efforts [26, 37, 55, 56, 57].

means U.S. Food and Drug Administration (FDA).


means Under investigation.


Another important outcome of collaborative research efforts is the development of innovative therapies for SCA. Gene therapy, for example, offers the potential for a cure by introducing a functional copy of the hemoglobin gene into stem cells, allowing them to produce normal hemoglobin. Clinical trials using gene therapy for SCA have shown promising results, with some patients achieving sustained increases in foetal hemoglobin levels and improvements in clinical outcomes. Another beauty of world-class collaborative efforts that resulted in a major breakthrough is the gene therapy and editing process towards the erasing of sickle cell anemia disease.

Figure 11 illustrates the collaborative nature of research efforts in sickle cell anemia, involving partnerships between academia, industry, and patient advocacy groups.

Figure 11.

Collaborative Efforts to Showcase the Beauty of Gene Therapy and Editing [58, 59, 60, 61, 62, 63].

Large-scale clinical trials are also a key focus of collaborative research efforts in SCA. These trials are essential for evaluating the safety and efficacy of new treatments and providing evidence-based guidelines for clinical practice. Collaborative efforts between researchers, healthcare providers, and patient advocacy groups have enabled the initiation of large-scale clinical trials, such as the HOPE Study, which is evaluating the use of gene therapy in SCA patients.

On-going research is focused on developing curative therapies, improving transplant outcomes, and addressing the long-term complications of sickle cell disease.

Thus, collaborative research efforts have been instrumental in advancing our understanding of sickle cell anemia and developing new treatment options for this complex disease (Figure 12). By working together, researchers, healthcare providers, and patient advocacy groups are making significant strides towards improving the lives of individuals affected by sickle cell anemia.

Figure 12.

Development of Curative Therapies for Sickle Cell Anemia Disease [22, 58, 59, 60].

Advertisement

8. Future directions

Future directions in the treatment of sickle cell anemia (SCA) hold great promise, with on-going advancements in genetic therapies, stem cell transplantation, and disease-modifying therapies. These advancements are expanding treatment options and improving outcomes for patients with SCA, bringing us closer to a future where SCA is a manageable chronic illness rather than a life-threatening condition.

Genetic therapies, such as gene editing using CRISPR-Cas9 and gene therapy using lentiviral vectors, offer the potential for a cure for SCA by correcting the genetic mutation responsible for the disease. Clinical trials using these therapies have shown promising results, with some patients experiencing a complete remission of symptoms. On-going research is focused on further improving the safety and efficacy of these therapies and expanding access to them for all SCA patients.

Stem cell transplantation remains the only cure for SCA, and recent advancements in transplant techniques, such as reduced-intensity conditioning regimens and haploidentical transplantation, have significantly improved outcomes and reduced risks associated with the procedure. On-going research is exploring new approaches to stem cell transplantation, such as the use of gene-edited stem cells, to further improve outcomes and expand access to transplantation for all SCA patients.

Disease-modifying therapies, such as hydroxyurea, voxelotor, and crizanlizumab, continue to be a cornerstone of SCA treatment, offering targeted approaches to reducing the frequency and severity of complications.

On-going research is focused on developing new disease-modifying therapies that target specific pathways involved in the pathogenesis of SCA, with the goal of further improving outcomes for patients.

Figure 13 illustrates the future directions in sickle cell anemia treatment, including advances in genetic therapies, stem cell transplantation, and disease-modifying therapies.

Figure 13.

Advances in Sickle Cell Anemia Disease over the last few Decades [22, 58, 59, 60].

Advances in understanding the underlying mechanisms of SCA are also driving research towards more targeted and effective treatments. For example, researchers are investigating the role of oxidative stress, inflammation, and endothelial dysfunction in the pathophysiology of SCA, with the aim of developing new therapies that target these pathways. Additionally, research into the role of the microbiome in SCA may lead to new treatment strategies that modulate the gut microbiota to improve outcomes for patients.

The value of prevention in sickle cell anemia disease is important in several contexts. Pre-conception screening of couples to avoid high-risk pregnancies is recommended, and females need relevant information on how to manage their pregnancy for better outcomes. Studies have shown that the prevalence of ischaemic stroke by the age of 20 years is ≥11%, with the highest stroke rates occurring in early childhood. Transcranial Doppler ultrasound is also used to identify patients at the highest risk who may benefit from transfusion therapy [64, 65]. Prophylactic erythrocytapheresis addresses the problem of slow red blood cell transfusion, reducing the concentration of sickle hemoglobin-containing red cells, thereby improving symptoms of crises quickly [55]. For the prevention of infection in children with sickle cell anemia disease, prophylactic antibiotics initiated as early as 3 months of age are highly recommended.

Finally, the future of sickle cell anemia treatment holds great promise, with on-going advancements in genetic therapies, stem cell transplantation, and disease-modifying therapies. Continued investment in research and healthcare infrastructure is essential to realize this promise and ensure that all SCA patients have access to the latest and most effective treatments. Collaboration between researchers, healthcare providers, and patients is essential and also fundamentally key to achieving this goal and improving outcomes for individuals affected by SCA.

The field of sickle cell anemia healthcare and treatment has seen significant advancements in recent years, offering new hope for patients and improving their quality of life. However, sickle cell anemia disease is a global disease; it is no longer only a disease in developing countries but is becoming more widespread in Europe and across the world. For example, all through the period that the world experienced the devastating spread of SARS-CoV-2 (COVID-19), it was a major challenge for healthcare providers as the global pandemic presented unprecedented challenges in managing care for patients with sickle cell anemia disease. There are serious unmet needs to address if patients are to be treated successfully, especially because the pathophysiology of sickle cell anemia disease is extremely complex, making it difficult to find a unique treatment.

Also, there are a number of barriers to the management of vaso-occlusion and other associated challenges. Several studies showed that levels of patient pain are underestimated if they are only measured via healthcare facilities, indicating a need to improve patients’ experiences of acute care. Studies have also shown that approximately, 25% of vaso-occlusion reported are most of the time managed by patients at home. From the patient’s perspective, this is largely, but not exclusively, because of perceived poor medical experiences at the emergency room (ER) or hospital, which then prevent the patients from seeking further medical assistance when needed [4866].

In addition to the overcrowded and stressful hospital/ER environment, barriers to vaso-occlusion (VOC) management include poor transition from pediatric to adult care, the stigma associated with having sickle cell anemia disease, ethnicity, and requiring opioids for pain control. Patients with sickle cell anemia disease also wait longer to see an ER physician than, for example, a patient with a long bone fracture, which may be because of a lack of experience with sickle cell anemia disease, or a lack of empathy as a result of the aforementioned stigma (Figure 14).

Figure 14.

Example of Pain Action Plan - Patients using Traffic Light Color Coding for Highlights [26, 45, 60, 67].

Furthermore, the effects of medication on fertility rates and pregnancy are also of concern to patients. Most current research works have shown no clear evidence exists of a negative effect of hydroxyurea therapy on fertility, but further research is needed as more treated patients reach adulthood (Figures 15 and 16) [70, 71].

Figure 15.

Overview of Complications from Sickle Cell Disease – Childhood and Adulthood [22, 58, 59, 60, 68].

Figure 16.

Overview of the Comparison of the Characteristics and Complications of Sickle Cell Disease versus Geriatrics [22, 58, 59, 60, 69].

Optimum management of vaso-occlusion can be facilitated by pain action plans designed to educate patients on strategies to prevent triggers, advise on self-care strategies to manage pain at home and inform patients when to seek medical assistance. Pain action plans also encompass guidance for acute pain management, discharge planning to prevent rebound-pain exacerbation, and co-ordination of care after discharge [45]. This preventative approach helps patients to recognize their pain severity and manage their vaso-occlusion accordingly (Figure 17).

Figure 17.

Template - Clinical Management of Acute Pain and other aspects of Sickle Cell Disease [26, 45, 46, 60, 67].

Strategies to overcome these barriers include patient plans, day hospitals, and structured transition programmes. In this decade of new drug development, there is a need to examine the potential role of combination therapies. Hydroxyurea is a well-established drug and most trials are based on patients already on this treatment; adding another drug may become the standard of care in the future [67, 72, 73, 74, 75].

However, because of the global distribution of patients especially in developing countries, there is a need to consider the cost of new therapies. Many people do not have access to safe blood transfusions, therapeutic options, or even iron chelation and to address the unmet needs of sickle cell anemia disease worldwide, global collaboration is needed to develop treatments and protocols.

Thus, by working together, we can continue to advance the field and improve outcomes for individuals living with this challenging disease.

Advertisement

9. Summary of chapter/conclusion

Sickle cell anemia (SCA) is a complex genetic disorder that has historically posed significant challenges for patients and healthcare providers. However, recent advancements in SCA healthcare and treatment offer new hope for patients and are transforming the landscape of SCA management. This chapter has explored some of the key advancements in SCA treatment, including genetic therapies, stem cell transplantation, disease-modifying therapies, telemedicine, patient education and support programs, public health initiatives, collaborative research efforts, and future directions in SCA treatment.

Genetic therapies, such as gene editing using CRISPR-Cas9 and gene therapy using lentiviral vectors, offer the potential for a cure for SCA by correcting the genetic mutation responsible for the disease [58, 76]. These therapies have shown promising results in clinical trials, with some patients experiencing a complete remission of symptoms [59, 60].

Stem cell transplantation remains the only cure for SCA, and recent advancements in transplant techniques have significantly improved outcomes and reduced risks associated with the procedure [77, 78, 79, 80].

Disease-modifying therapies, such as hydroxyurea, voxelotor [81, 82], and crizanlizumab, offer targeted approaches to reducing the frequency and severity of complications in SCA patients.

Telemedicine and remote monitoring technologies have revolutionized the management of SCA, especially in rural and underserved areas, by providing patients with access to healthcare providers, medical records, and condition monitoring remotely. Patient education and support programs have empowered patients with SCA to better manage their condition, providing information on disease management, lifestyle modifications, and psychosocial support [83, 84]. Public health initiatives aimed at raising awareness about SCA and improving access to care have played a crucial role in improving outcomes for patients, leading to earlier diagnosis and better treatment outcomes [85, 86, 87, 88].

Collaborative research efforts between academia, industry, and patient advocacy groups have been instrumental in advancing our understanding of SCA and developing new treatment options. These partnerships have led to the identification of novel drug targets, the development of innovative therapies, and the initiation of large-scale clinical trials.

Future directions in SCA treatment hold great promise, with on-going advancements in genetic therapies, stem cell transplantation, and disease-modifying therapies.

References

  1. 1. Hassell KL. Population estimates of sickle cell disease in the US. American Journal of Preventive Medicine. 2010;38:S512-S521
  2. 2. Eaton WA, Hofrichter J. Haemoglobin S gelation and sickle cell disease. Blood. 1987;70:1245-1266
  3. 3. Platt OS, Thorington BD, Brambilla DJ, Milner PF, Rosse WF, Vichinsky E, et al. Pain in sickle cell disease. Rates and risk factors. The New England Journal of Medicine. 1991;325:11-16
  4. 4. Platt OS, Brambilla DJ, Rosse WF, Milner PF, Castro O, Steinberg MH, et al. Mortality in sickle cell disease. Life expectancy and risk factors for early death. The New England Journal of Medicine. 1994;330:1639-1644
  5. 5. Lanzkron S, Carroll CP, Haywood C. Mortality rates and age at death from sickle cell disease: U.S., 1979-2005. Public Health Reports. 2013;128:110-116
  6. 6. Negre O, Eggimann AV, Beuzard Y, Ribeil JA, Bourget P, Borwornpinyo S, et al. Gene therapy of the beta-haemoglobinopathies by lentiviral transfer of the beta(A(T87Q))-globin gene. Human Gene Therapy. 2016;27:148-165. DOI: 10.1089/hum.2016.007
  7. 7. Ribeil J, Hacein-Bey-Abina S, Payen E, Magnani A, Semeraro M, Magrin E, et al. Gene therapy in a patient with sickle cell disease. The New England Journal of Medicine. 2017;376:848-855
  8. 8. Humbert O, Radtke S, Samuelson C, Carrillo RR, Perez AM, Reddy SS, et al. Therapeutically relevant engraftment of a CRISPR-Cas9–edited HSC-enriched population with HbF reactivation in nonhuman primates. Science Translational Medicine. 2019;11:eaaw3768
  9. 9. CRISPR-Cas9 for in vivo Gene Therapy: Promise and Hurdles - Scientific Figure on Research Gate. Available from: https://www.researchgate.net/figure/Schematic-representation-of-CRISPR-Cas9-mediated-genome-editing-a-Schematic-of-CRISPR_fig1_308453321
  10. 10. Esrick EB, Manis JP, Daley H, Baricordi C, Trebeden-Negre H, Pierciey FJ, et al. Successful hematopoietic stem cell mobilization and apheresis collection using plerixafor alone in sickle cell patients. Blood Advances. 2018;2:2505-2512. DOI: 10.1182/bloodadvances.2018016725
  11. 11. Fitzhugh CD, Abraham AA, Tisdale JF, Hsieh MM. Hematopoietic stem cell transplantation for patients with sickle cell disease: Progress and future directions. Hematology/oncology Clinics of North America. 2014;28:1171-1185. DOI: 10.1016/j.hoc.2014.08.014
  12. 12. Eggleston B, Patience M, Edwards S, Adamkiewicz T, Buchanan GR, Davies SC, et al. Effect of myeloablative bone marrow transplantation on growth in children with sickle cell anaemia: Results of the multicentre study of haematopoietic cell transplantation for sickle cell anaemia. British Journal of Haematology. 2007;136:673-676
  13. 13. Bernaudin F, Socie G, Kuentz M, Chevret S, Duval M, Bertrand Y, et al. Long-term results of related myeloablative stem-cell transplantation to cure sickle cell disease. Blood. 2007;110:2749-2756
  14. 14. O'Donnell PV, Luznik L, Jones RJ, Vogelsang GB, Leffell MS, Phelps M, et al. Non-myeloablative bone marrow transplantation from partially HLA-mismatched related donors using post-transplantation cyclophosphamide. Biology of Blood and Marrow Transplantation. 2002;8:377-386
  15. 15. Matthes-Martin S, Lawitschka A, Fritsch G, Lion T, Grimm B, Breuer S, et al. Stem cell transplantation after reduced-intensity conditioning for sickle cell disease. European Journal of Haematology. 2013;90:308-312
  16. 16. Gluckman E, Cappelli B, Bernaudin F, Labopin M, Volt F, Carreras J, et al. Sickle cell disease: An international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood. 2016;129(11):1548-1556. DOI: 10.1182/blood-2016-10-745711
  17. 17. Walters MC, Hardy K, Edwards S, Adamkiewicz T, Barkovich J, Bernaudin F, et al. Pulmonary, gonadal, and central nervous system status after bone marrow transplantation for sickle cell disease. Biology of Blood and Marrow Transplantation. 2010;16:263-272
  18. 18. Vermylen C, Cornu G, Ferster A, Brichard B, Niane J, Ferrant A, et al. Haematopoietic stem cell transplantation for sickle cell anaemia: The first 50 patients transplanted in Belgium. Bone Marrow Transplantation. 1998;22:1-6
  19. 19. Ola BA, Yates SJ, Dyson SM. Living with sickle cell disease and depression in Lagos, Nigeria: A mixed methods study. Social Science & Medicine. 2016;161:27-36. DOI: 10.1016/j.socscimed.2016.05.029
  20. 20. Powars DR, Chan LS, Hiti A, Ramicone E, Johnson C. Outcome of sickle cell anaemia: A 4-decade observational study of 1056 patients. Medicine. 2005;84:363-376
  21. 21. Walters MC, De Castro LM, Sullivan KM, Krishnamurti L, Kamani N, Bredeson C, et al. Indications and results of HLA-identical sibling hematopoietic cell transplantation for sickle cell disease. Biology of Blood and Marrow Transplantation. 2016;22:207-211
  22. 22. Leonard A, Tisdale J, Abraham A. Curative options for sickle cell disease: Haploidentical stem cell transplantation or gene therapy? British Journal of Haematology. 2020;189(3):408-423. DOI: 10.1111/bjh.16437
  23. 23. CSDH. Closing the Gap in a Generation: Health Equity through Action on the Social Determinants of Health. Final Report of the Commission on Social Determinants of Health. Geneva: World Health Organization; 2008
  24. 24. Berghs M, Atkin K, Hatton C, et al. Rights to social determinants of flourishing? A paradigm for disability and public health research and policy. BMC Public Health. 2019;19:997. DOI: 10.1186/s12889-019-7334-8
  25. 25. Gr D, Whitehead M. WHO. Levelling up (Part 2): A Discussion Paper on European Strategies for Tackling Social Inequities in Health/by Göran Dahlgren and Margaret WHitehead. Copenhagen: WHO Regional Office for Europe; 2006
  26. 26. Sins JWR, Mager DJ, Davis S, Biemond BJ, Fijnvandraat K. Pharmacotherapeutical strategies in the prevention of acute, vaso-occlusive pain in sickle cell disease: A systematic review. Blood Advances. 2017;1:1598-1616. DOI: 10.1182/bloodadvances.2017007211
  27. 27. Hsieh MM, Kang EM, Fitzhugh CD, Link B, Bolan CD, Kurlander R, et al. Allogeneic hematopoietic stem-cell transplantation for sickle cell disease. The New England Journal of Medicine. 2009;361:2309-2317
  28. 28. Walters MC, Patience M, Leisenring W, Eckman JR, Buchanan GR, Rogers ZR, et al. Barriers to bone marrow transplantation for sickle cell anaemia. Biology of Blood and Marrow Transplantation. 1996;2:100-104
  29. 29. Dallas MH, Triplett B, Shook DR, Hartford C, Srinivasan A, Laver J, et al. Long-term outcome and evaluation of organ function in paediatric patients undergoing haploidentical and matched related hematopoietic cell transplantation for sickle cell disease. Biology of Blood and Marrow Transplantation. 2013;19:820-830
  30. 30. Walters MC, Patience M, Leisenring W, Rogers ZR, Aquino VM, Buchanan GR, et al. Stable mixed hematopoietic chimerism after bone marrow transplantation for sickle cell anaemia. Biology of Blood and Marrow Transplantation. 2001;7:665-673
  31. 31. Pareek M, Bangash MN, Pareek N, et al. Ethnicity and COVID-19: An urgent public health research priority. Lancet. 2020;395:1421-1422. DOI: 10.1016/S0140-6736(20)30922-3
  32. 32. Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bulletin of the World Health Organization. 2008;86:480-487. DOI: 10.2471/BLT.06.036673
  33. 33. Grosse SD, Odame I, Atrash HK, et al. Sickle cell disease in Africa: A neglected cause of early childhood mortality. American Journal of Preventive Medicine. 2011;41:S398-S405. DOI: 10.1016/j.amepre.2011.09.013
  34. 34. WHO. Sickle-Cell Disease: A Strategy for the WHO African Region. Malabo, Equatorial Guinea: World Health Organization; 2011
  35. 35. Burnham-Marusich AR, Ezeanolue CO, Obiefune MC, et al. Prevalence of sickle cell trait and reliability of self-reported status among expectant parents in Nigeria: Implications for targeted new born screening. Public Health Genomics. 2016;19:298-306. DOI: 10.1159/000448914
  36. 36. Tluway F, Makani J. Sickle cell disease in Africa: An overview of the integrated approach to health, research, education and advocacy in Tanzania, 2004-2016. British Journal of Haematology. 2017;177(6):919-929
  37. 37. Chakravorty SAK, Dziwinski S, Kaya B, et al. Optimising the transition from paediatric to adult care model for people with sickle cell disease – A UK multidisciplinary consensus statement. Health Sciences Journal. 2019;13:1-10
  38. 38. Gragert L, Eapen M, Williams E, Freeman J, Spellman S, Baitty R, et al. HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry. The New England Journal of Medicine. 2014;371:339-348
  39. 39. Dyson SM. “Race”, ethnicity and haemoglobin disorders. Social Science & Medicine. Jul 1998;47(1):121-131. DOI: 10.1016/s0277-9536(98)00023-9
  40. 40. Gardner K, Douiri A, Drasar E, Allman M, Mwirigi A, Awogbade M, et al. Survival in adults with sickle cell disease in a high-income setting. Blood. 2016;128:1436-1438. DOI: 10.1182/blood-2016-05-716910
  41. 41. Telen MJ, Malik P, Vercellotti GM. Therapeutic strategies for sickle cell disease: Towards a multi-agent approach. Nature Reviews. Drug Discovery. 2019;18:139-158. DOI: 10.1038/s41573-018-0003-2
  42. 42. Sparkenbaugh E, Pawlinski R. Interplay between coagulation and vascular inflammation in sickle cell disease. British Journal of Haematology. 2013;162:3-14. DOI: 10.1111/bjh.12336
  43. 43. Nur E, Brandjes DP, Teerlink T, Otten HM, Oude Elferink RP, Muskiet F, et al. N-acetylcysteine reduces oxidative stress in sickle cell patients. Annals of Hematology. 2012;91:1097-1105. DOI: 10.1007/s00277-011-1404-z
  44. 44. Telen MJ, Batchvarova M, Shan S, Bovee-Geurts PH, Zennadi R, Leitgeb A, et al. Sevuparin binds to multiple adhesive ligands and reduces sickle red blood cell-induced vaso-occlusion. British Journal of Haematology. 2016;175:935-948. DOI: 10.1111/bjh.14303
  45. 45. Adegoke SA, Oladimeji OI, Ologun BG, Aladekomo TA, Oyelami OA. Outcome of short-term emergency department observation care of children with sickle cell disease and vaso-occlusive crises: Initial experience from South-Western Nigeria. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2020;114:365-371. DOI: 10.1093/trstmh/traa006
  46. 46. Adewoye AH, Nolan V, McMahon L, Ma Q , Steinberg MH. Effectiveness of a dedicated day hospital for management of acute sickle cell pain. Haematologica. 2007;92(6):854-855. DOI: 10.3324/haematol.10757
  47. 47. Wright J, Bareford D, Wright C, Augustine G, Olley K, Musamadi L, et al. Day case management of sickle pain: 3 years’ experience in a UK sickle cell unit. British Journal of Haematology. 2004;126(6):878-880. DOI: 10.1111/j.1365-2141.2004.05123.x
  48. 48. Haywood C Jr et al. The impact of race and disease on sickle cell patient wait times in the emergency department. The American Journal of Emergency Medicine. 2013;31(4):651-656
  49. 49. Glassberg JA et al. Emergency provider analgesic practices and attitudes towards patients with sickle cell disease. The American Journal of Emergency Medicine. 2013;62(4):293-302
  50. 50. Shah N et al. Sickle cell disease complications: Prevalence and resource utilization. PLoS One. 2019;14(7):e0214355
  51. 51. Elmariah H, Garrett ME, De Castro LM, Jonassaint JC, Ataga KI, Eckman JR, et al. Factors associated with survival in a contemporary adult sickle cell disease cohort. American Journal of Hematology. 2014;89:530-535. DOI: 10.1002/ajh.23683
  52. 52. Tewari S, Brousse V, Piel FB, Menzel S, Rees DC. Environmental determinants of severity in sickle cell disease. Haematologica. 2015;100:1108-1116
  53. 53. McGann PT. Sickle cell anaemia: An underappreciated and unaddressed contributor to global childhood mortality. The Journal of Pediatrics. 2014;165:18-22. DOI: 10.1016/j.jpeds.2014.01.070
  54. 54. Adegoke SA, Akinlosotu MA, Adediji OB, Oyelami OA, Adeodu OO, Adekile AD. Sickle cell disease in Southwestern Nigeria: Assessment of knowledge of primary health care workers and available facilities. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2018;112:81-87. DOI: 10.1093/trstmh/try025
  55. 55. Ullrich H et al. Erythrocytapheresis: Do not forget a useful therapy! Transfusion Medicine and Hemotherapy. 2008;35(1):24-30
  56. 56. Heeney MM, Hoppe CC, Abboud MR, Inusa B, Kanter J, Ogutu B, et al. A multinational trial of prasugrel for sickle cell vaso-occlusive events. The New England Journal of Medicine. 2016;374:625-635. DOI: 10.1056/NEJMoa1512021
  57. 57. Hoppe C, Jacob E, Styles L, Kuypers F, Larkin S, Vichinsky E. Simvastatin reduces vaso-occlusive pain in sickle cell anaemia: A pilot efficacy trial. British Journal of Haematology. 2017;177:620-629. DOI: 10.1111/bjh.14580
  58. 58. The Lancet Haematology. CRISPR-Cas9 gene editing for patients with haemoglobinopathies. Lancet Haematology. 2019;6:E438
  59. 59. Vakulskas CA, Dever DP, Rettig GR, Turk R, Jacobi AM, Collingwood MA, et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nature Medicine. 2018;24:1216-1224
  60. 60. Urbinati F, Hargrove PW, Geiger S, Romero Z, Wherley J, Kaufman ML, et al. Potentially therapeutic levels of anti-sickling globin gene expression following lentivirus-mediated gene transfer in sickle cell disease bone marrow CD34+ cells. Experimental Hematology. 2015;43:346-351
  61. 61. Ingram VM. Gene mutations in human haemoglobin: The chemical difference between normal and sickle cell haemoglobin. Nature. 1957;180:326-328
  62. 62. Hoban MD, Lumaquin D, Kuo CY, Romero Z, Long J, Ho M, et al. CRISPR/Cas9-mediated correction of the sickle mutation in human CD34+ cells. Molecular Therapy. 2016;24:1561-1569
  63. 63. Lattanzi A, Meneghini V, Pavani G, Amor F, Ramadier S, Felix T, et al. Optimization of CRISPR/Cas9 delivery to human hematopoietic stem and progenitor cells for therapeutic genomic rearrangements. Molecular Therapy. 2019;27:137-150
  64. 64. Adams RJ et al. Long-term stroke risk in children with sickle cell disease screened with transcranial Doppler. Annals of Neurology. 1997;42(5):699-704
  65. 65. Adams RJ et al. Transcranial Doppler correlation with cerebral angiography in sickle cell disease. Stroke. 1992;23(8):1073-1077
  66. 66. Hassell KL. Sickle cell disease: A continued call to action. American Journal of Preventive Medicine. 2016;51:S1-S2. DOI: 10.1016/j.amepre.2015.11.002
  67. 67. Silva-Pinto AC et al. Clinical and hematological effects of hydroxyurea therapy in sickle cell patients: A single-center experience in Brazil. São Paulo Medical Journal. 2013;131(4):238-243
  68. 68. Brittenham GM, Schechter AN, Noguchi CT. Haemoglobin S polymerization: Primary determinant of the haemolytic and clinical severity of the sickling syndromes. Blood. 1985;65:183-189
  69. 69. Eaton WA, Bunn HF. Treating sickle cell disease by targeting HbS polymerization. Blood. 2017;129:2719-2726. DOI: 10.1182/blood-2017-02-765891
  70. 70. Berthaut I, Guignedoux G, Kirsch-Noir F, de Larouziere V, Ravel C, Bachir D, et al. Influence of sickle cell disease and treatment with hydroxyurea on sperm parameters and fertility of human males. Haematologica. 2008;93:988-993. DOI: 10.3324/haematol.11515
  71. 71. Jain D et al. Sickle cell disease and pregnancy. Mediterranean Journal of Hematology and Infectious Diseases. 2019;11(1):e2019040
  72. 72. Opoka RO, Ndugwa CM, Latham TS, Lane A, Hume HA, Kasirye P, et al. Novel use of hydroxyurea in an African region with malaria (NOHARM): A trial for children with sickle cell anaemia. Blood. 2017;130:2585-2593. DOI: 10.1182/blood-2017-06-788935
  73. 73. Cokic VP, Smith RD, Beleslin-Cokic BB, Njoroge JM, Miller JL, Gladwin MT, et al. Hydroxyurea induces foetal haemoglobin by the nitric oxide-dependent activation of soluble guanylyl cyclase. The Journal of Clinical Investigation. 2003;111:231-239. DOI: 10.1172/JCI16672
  74. 74. Ware RE. Optimizing hydroxyurea therapy for sickle cell anemia. Hematology: the American Society of Hematology Education Program. 2015;2015:436-443. DOI: 10.1182/asheducation-2015.1.436
  75. 75. McGann PT, Ware RE. Hydroxyurea therapy for sickle cell anaemia. Expert Opinion on Drug Safety. 2015;14(11):1749-1758
  76. 76. Negre O, Bartholomae C, Beuzard Y, Cavazzana M, Christiansen L, Courne C, et al. Preclinical evaluation of efficacy and safety of an improved lentiviral vector for the treatment of β-thalassemia and sickle cell disease. Current Gene Therapy. 2015;15:64-81
  77. 77. Angelucci E, Matthes-Martin S, Baronciani D, Bernaudin F, Bonanomi S, Cappellini MD, et al. Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: Indications and management recommendations from an international expert panel. Haematologica. 2014;99:811-820. DOI: 10.3324/haematol.2013.099747
  78. 78. Gluckman E, Cappelli B, Bernaudin F, Labopin M, Volt F, Carreras J, et al. Sickle cell disease: An international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood. 2017;129:1548-1556. DOI: 10.1182/blood-2016-10-745711
  79. 79. Joseph JJ, Abraham AA, Fitzhugh CD. When there is no match, the game is not over: Alternative donor options for hematopoietic stem cell transplantation in sickle cell disease. Seminars in Hematology. 2018;55:94-101. DOI: 10.1053/j.seminhematol.2018.04.013
  80. 80. Johnson FL. Bone marrow transplantation in the treatment of sickle cell anaemia. The American Journal of Pediatric Hematology/Oncology. 1985;7:254-257
  81. 81. Ataga KI, Kutlar A, Kanter J, Liles D, Cancado R, Friedrisch J, et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. The New England Journal of Medicine. 2017;376:429-439. DOI: 10.1056/NEJMoa1611770
  82. 82. Kutlar A, Kanter J, Liles DK, Alvarez OA, Cancado RD, Friedrisch JR, et al. Effect of crizanlizumab on pain crises in subgroups of patients with sickle cell disease: A SUSTAIN study analysis. American Journal of Hematology. 2019;94:55-61. DOI: 10.1002/ajh.25308
  83. 83. Chinyakata R, Roman NV, Msiza FB. Stakeholders’ perspectives on the barriers to accessing health care services in rural settings: A human capabilities approach. Open Public Health Journal. 2021;14:336-344. DOI: 10.2174/1874944502114010336
  84. 84. Culyer AJ. Involving stakeholders in healthcare decisions: The experience of the national institute for clinical excellence (NICE) in England and Wales. Healthcare Quarterly. 2005;8(3):56-60. DOI: 10.12927/hcq.17155
  85. 85. National Population Commission (Nigeria) and ICF. Nigeria Demographic and Health Survey 2018. Abuja and Rockville: National Population Commission (Nigeria) and ICF. Available from: https://dhsprogram.com/pubs/pdf/FR359/FR359.pdf; 2019 [Accessed: November 19, 2019]
  86. 86. Nnodu O. Interventions for the prevention and control of sickle cell disease at primary health care centres in Gwagwalada area Council of the Federal Capital Territory, Nigeria. Cureus. 2014;6(8):e194. DOI: 10.7759/cureus.194
  87. 87. The federal ministry of health. National Guideline for the Control and Management of Sickle Cell Disease. Nigeria: The federal ministry of health; 2014. Available from: http://www.health.gov.ng/doc/SCDGuideline.pdf
  88. 88. Piel FB, Hay SI, Gupta S, Weatherall DJ, Williams TN. Global burden of sickle cell anaemia in children under five, 2010-2050: Modelling based on demographics, excess mortality, and interventions. PLoS Medicine. 2013;10:e1001484. DOI: 10.1371/journal.pmed.1001484

Written By

Oluwafemi Ajoyemi Ala

Submitted: 23 February 2024 Reviewed: 22 April 2024 Published: 26 July 2024