The network of traits that predisposes a person to be helpful are all of the following EXCEPT

Practice Essentials

Sickle cell disease (SCD) and its variants are genetic disorders resulting from the presence of a mutated form of hemoglobin, hemoglobin S (HbS) [1, 2] (see the image below). The most common form of SCD found in North America is homozygous HbS disease (HbSS), an autosomal recessive disorder first described by Herrick in 1910. SCD causes significant morbidity and mortality, particularly in people of African and Mediterranean ancestry (see Pathophysiology). Morbidity, frequency of crisis, degree of anemia, and the organ systems involved vary considerably from individual to individual.

The network of traits that predisposes a person to be helpful are all of the following EXCEPT
Molecular and cellular changes of hemoglobin S.

Signs and symptoms

Screening for HbS at birth is currently mandatory in the United States. For the first 6 months of life, infants are protected largely by elevated levels of fetal hemoglobin (Hb F). SCD usually manifests early in childhood, in the following ways:

  • Acute and chronic pain: The most common clinical manifestation of SCD is vaso-occlusive crisis; pain crises are the most distinguishing clinical feature of SCD

  • Bone pain: Often seen in long bones of extremities, primarily due to bone marrow infarction

  • Anemia: Universally present, chronic, and hemolytic in nature

  • Aplastic crisis: Serious complication due to infection with parvovirus B19 (B19V)

  • Splenic sequestration: Characterized by the onset of life-threatening anemia with rapid enlargement of the spleen and high reticulocyte count

  • Infection: Organisms that pose the greatest danger include encapsulated respiratory bacteria, particularly Streptococcus pneumoniae; adult infections are predominantly with gram-negative organisms, especially Salmonella

  • Growth retardation, delayed sexual maturation, being underweight

  • Hand-foot syndrome: This is a dactylitis presenting as bilateral painful and swollen hands and/or feet in children

  • Acute chest syndrome: Young children present with chest pain, fever, cough, tachypnea, leukocytosis, and pulmonary infiltrates in the upper lobes; adults are usually afebrile, dyspneic with severe chest pain, with multilobar/lower lobe disease

  • Pulmonary hypertension: Increasingly recognized as a serious complication of SCD

  • Avascular necrosis of the femoral or humeral head: Due to vascular occlusion

  • Central nervous system (CNS) involvement: Most severe manifestation is stroke

  • Ophthalmologic involvement: Ptosis, retinal vascular changes, proliferative retinitis

  • Cardiac involvement: Dilation of both ventricles and the left atrium

  • Gastrointestinal involvement: Cholelithiasis is common in children; liver may become involved

  • Genitourinary involvement: Kidneys lose concentrating capacity; priapism is a well-recognized complication of SCD

  • Dermatologic involvement: Leg ulcers are a chronic painful problem

Approximately half the individuals with homozygous HbS disease experience vaso-occlusive crises. The frequency of crises is extremely variable. Some individuals have as many as 6 or more episodes annually, whereas others may have episodes only at great intervals or none at all. Each individual typically has a consistent pattern for crisis frequency. Triggers of vaso-occlusive crisis include the following:

  • Hypoxemia: May be due to acute chest syndrome or respiratory complications

  • Dehydration: Acidosis results in a shift of the oxygen dissociation curve

  • Changes in body temperature (eg, an increase due to fever or a decrease due to environmental temperature change)

Many individuals with HbSS experience chronic low-level pain, mainly in bones and joints. Intermittent vaso-occlusive crises may be superimposed, or chronic low-level pain may be the only expression of the disease.

See Presentation for more detail.

Diagnosis

SCD is suggested by the typical clinical picture of chronic hemolytic anemia and vaso-occlusive crisis. Electrophoresis confirms the diagnosis with the presence of homozygous HbS and can also document other hemoglobinopathies (eg, HbSC, HbS-beta+ thalassemia).

Laboratory tests used in patients with SCD include the following:

  • Mandatory screening for HbS at birth in the United States; prenatal testing can be obtained via chorionic villus sampling

  • Hemoglobin electrophoresis

  • CBC count with differential and reticulocyte count

  • Serum electrolytes

  • Hemoglobin solubility testing

  • Peripheral blood smear

  • Pulmonary function tests (transcutaneous O 2 saturation)

  • Kidney function (creatinine, BUN, urinalysis)

  • Hepatobiliary function tests, (ALT, fractionated bilirubin)

  • CSF examination: Consider LP in febrile children who appear toxic and in those with neurologic findings (eg, neck stiffness, positive Brudzinski/Kernig signs, focal deficits); consider CT scanning before performing lumbar puncture

  • Blood cultures

  • ABGs

  • Secretory phospholipase A2 (sPLA2)

In one study of 38 asymptomatic children with SCD, investigators found that hypertension and abnormal blood pressure patterns were prevalent in children with SCD. [3] They suggested using 24-hour ambulatory blood pressure monitoring (ABPM) to identify these conditions in young patients. [3]

In the study, 17 patients (43.6%) had ambulatory hypertension, whereas 4 (10.3%) had hypertension on the basis of their clinic blood pressure. Twenty-three patients (59%) had impaired systolic blood pressure dipping, 7 (18%) had impaired diastolic blood pressure dipping, and 5 (13%) had reversed dipping. [3]

Imaging studies

Imaging studies that aid in the diagnosis of sickle cell anemia in patients in whom the disease is suggested clinically include the following:

  • Radiography: Chest x-rays should be performed in patients with respiratory symptoms

  • MRI: Useful for early detection of bone marrow changes due to acute and chronic bone marrow infarction, marrow hyperplasia, osteomyelitis, and osteonecrosis

  • CT scanning: May demonstrate subtle regions of osteonecrosis not apparent on plain radiographs in patients who are unable to have an MRI [4] and to exclude renal medullary carcinoma in patients presenting with hematuria

  • Nuclear medicine scanning: 99mTc bone scanning detects early stages of osteonecrosis; 111In WBC scanning is used for diagnosing osteomyelitis

  • Transcranial Doppler ultrasonography: Can identify children with SCD at high risk for stroke

  • Abdominal ultrasonography: May be used to rule out cholecystitis, cholelithiasis, or an ectopic pregnancy and to measure spleen and liver size

  • Echocardiography: Identifies patients with pulmonary hypertension

  • Transcranial near-infrared spectroscopy or cerebral oximetry: Can be used as a screening tool for low cerebral venous oxygen saturation in children with SCD

See Workup for more detail.

Management

The goals of treatment in SCD are symptom control and management of disease complications. Treatment strategies include the following 7 goals:

  • Management of vaso-occlusive crisis

  • Management of chronic pain syndromes

  • Management of chronic hemolytic anemia

  • Prevention and treatment of infections

  • Management of the complications and the various organ damage syndromes associated with the disease

  • Prevention of stroke

  • Detection and treatment of pulmonary hypertension

Pharmacotherapy

SCD may be treated with the following medications:

  • Antimetabolites: Hydroxyurea

  • Hemoglobin oxygen-affinity modulators (eg, voxelotor)

  • P-selectin inhibitors (eg, crizanlizumab)

  • Opioid analgesics (eg, oxycodone/aspirin, methadone, morphine sulfate, oxycodone/acetaminophen, fentanyl, nalbuphine, codeine, acetaminophen/codeine)

  • Nonsteroidal analgesics (eg, ketorolac, aspirin, acetaminophen, ibuprofen)

  • Tricyclic antidepressants (eg, amitriptyline)

  • Antibiotics (eg, cefuroxime, amoxicillin/clavulanate, penicillin VK, ceftriaxone, azithromycin, cefaclor)

  • Vaccines (eg, pneumococcal, meningococcal, influenza, and recommended scheduled childhood/adult vaccinations)

  • Endothelin-1 receptor antagonists (eg, bosentan)

  • Phosphodiesterase inhibitors (eg, sildenafil, tadalafil)

  • Vitamins (eg, folic acid)

  • L-glutamine

  • Antiemetics (eg, promethazine)

Non-pharmacologic therapy

Other approaches to managing SCD include the following:

  • Stem cell transplantation: Can be curative

  • Transfusions: For sudden, severe anemia due to acute splenic sequestration, parvovirus B19 infection, or hyperhemolytic crises

  • Wound debridement

  • Physical therapy

  • Heat and cold application

  • Acupuncture and acupressure

  • Transcutaneous electric nerve stimulation (TENS)

Combination pharmacotherapy and non-pharmacotherapy

  • Vigorous hydration (plus analgesics): For vaso-occlusive crisis

  • Oxygen, antibiotics, analgesics, incentive spirometry, simple transfusion, and bronchodilators: For treatment of acute chest syndrome

See Treatment and Medication for more detail.

Background

Carriers of the sickle cell trait (ie, heterozygotes who carry one HbS allele and one normal adult hemoglobin [HbA] allele) have some resistance to the often-fatal malaria caused by Plasmodium falciparum. This property explains the distribution and persistence of this gene in the population in malaria-endemic areas. [5, 6, 7]

However, in areas such as the United States, where malaria is not a problem, the trait no longer provides a survival advantage. Instead, it poses the threat of SCD, which occurs in children of carriers who inherit the sickle cell gene from both parents (ie, HbSS).

Although carriers of sickle cell trait do not suffer from SCD, individuals with one copy of HbS and one copy of a gene that codes for another abnormal variant of hemoglobin, such as HbC or Hb beta-thalassemia, have a less severe form of the disease.

Genetics

SCD denotes all genotypes containing at least one sickle gene, in which HbS makes up at least half the hemoglobin present. Major sickle genotypes described so far include the following:

  • HbSS disease or sickle cell anemia (the most common form) - Homozygote for the S globin with usually a severe or moderately severe phenotype and with the shortest survival

  • HbS/b-0 thalassemia - Double heterozygote for HbS and b-0 thalassemia; clinically indistinguishable from sickle cell anemia (SCA)

  • HbS/b+ thalassemia - Mild-to-moderate severity with variability in different ethnicities

  • HbSC disease - Double heterozygote for HbS and HbC characterized by moderate clinical severity

  • HbS/hereditary persistence of fetal Hb (S/HPHP) - Very mild or asymptomatic phenotype

  • HbS/HbE syndrome - Very rare with a phenotype usually similar to HbS/b+ thalassemia

  • Rare combinations of HbS with other abnormal hemoglobins such as HbD Los Angeles, G-Philadelphia, HbO Arab, and others

Sickle cell trait or the carrier state is the heterozygous form characterized by the presence of around 40% HbS, absence of anemia, inability to concentrate urine (isosthenuria), and hematuria. Under conditions leading to hypoxia, it may become a pathologic risk factor.

SCD is the most severe and most common form. Affected individuals present with a wide range of clinical problems that result from vascular obstruction and ischemia. Although the disease can be diagnosed at birth, clinical abnormalities usually do not occur before age 6 months, when functional asplenia develops. Functional asplenia results in susceptibility to overwhelming infection with encapsulated bacteria. Subsequently, other organs are damaged. Typical manifestations include recurrent pain and progressive incremental infarction.

Newborn screening for sickle hemoglobinopathies is mandated in 50 states. Therefore, most patients presenting to the ED have a known diagnosis.

Pathophysiology

HbS arises from a mutation substituting thymine for adenine in the sixth codon of the beta-chain gene, GAG to GTG. This causes coding of valine instead of glutamate in position 6 of the Hb beta chain. The resulting Hb has the physical properties of forming polymers under deoxy conditions. It also exhibits changes in solubility and molecular stability. These properties are responsible for the profound clinical expressions of the sickling syndromes.

Under deoxy conditions, HbS undergoes marked decrease in solubility, increased viscosity, and polymer formation at concentrations exceeding 30 g/dL. It forms a gel-like substance containing Hb crystals called tactoids. The gel-like form of Hb is in equilibrium with its liquid-soluble form. A number of factors influence this equilibrium, including oxygen tension, concentration of Hb S, and the presence of other hemoglobins.

Oxygen tension is a factor in that polymer formation occurs only in the deoxy state. If oxygen is present, the liquid state prevails. Concentration of Hb S is a factor in that gelation of HbS occurs at concentrations greater than 20.8 g/dL (the normal cellular Hb concentration is 30 g/dL). The presence of other hemoglobins is a factor in that normal adult hemoglobin (HbA) and fetal hemoglobin (HbF) have an inhibitory effect on gelation.

These and other Hb interactions affect the severity of clinical syndromes. HbSS produces a more severe disease than sickle cell HbC (HbSC), HbSD, HbSO Arab, and Hb with one normal and one sickle allele (HbSA).

When red blood cells (RBCs) containing homozygous HbS are exposed to deoxy conditions, the sickling process begins. A slow and gradual polymer formation ensues. Electron microscopy reveals a parallel array of filaments. Repeated and prolonged sickling involves the membrane; the RBC assumes the characteristic sickled shape. (See image below.)

The network of traits that predisposes a person to be helpful are all of the following EXCEPT
Molecular and cellular changes of hemoglobin S.

After recurrent episodes of sickling, membrane damage occurs and the cells are no longer capable of resuming the biconcave shape upon reoxygenation. Thus, they become irreversibly sickled cells (ISCs). From 5-50% of RBCs permanently remain in the sickled shape.

When RBCs sickle, they gain Na+ and lose K+. Membrane permeability to Ca++ increases, possibly due, in part, to impairment in the Ca++ pump that depends on adenosine triphosphatase (ATPase). The intracellular Ca++ concentration rises to 4 times the reference level. The membrane becomes more rigid, possibly due to changes in cytoskeletal protein interactions; however, these changes are not found consistently. In addition, whether calcium is responsible for membrane rigidity is not clear.

Membrane vesicle formation occurs, and the lipid bilayer is perturbed. The outer leaflet has increased amounts of phosphatidyl ethanolamine and contains phosphatidylserine. The latter may play a role as a contributor to thrombosis, acting as a catalyst for plasma clotting factors. Membrane rigidity can be reversed in vitro by replacing HbS with HbA, suggesting that HbS interacts with the cell membrane.

Interactions with vascular endothelium

Complex multifactorial mechanisms involving endothelial dysfunction underlie the acute and chronic manifestations of SCD. [8] A current model proposes that vaso-occlusive crises in SCD result from adhesive interactions of sickle cell RBCs and leukocytes with the endothelium. [9]

In this model, the endothelium becomes activated by sickle cell RBCs, either directly, through adhesion molecules on the RBC surface, or indirectly through plasma proteins (eg, thrombospondin, von Willebrand factor) that act as a soluble bridge molecule. This leads, sequentiallly, to recruitment of adherent leukocytes, activation of recruited neutrophils and of other leukocytes (eg, monocytes or natural killer T cells), interactions of RBCs with adherent neutrophils, and clogging of the vessel by cell aggregates composed of RBCs, adherent leukocytes, and possibly platelets. [9]

Sickle cells express very late antigen–4 (VLA-4) on the surface. VLA-4 interacts with the endothelial cell adhesive molecule, vascular cell adhesive molecule–1 (VCAM-1). VCAM-1 is upregulated by hypoxia and inhibited by nitric oxide.

Hypoxia also decreases nitric oxide production, thereby adding to the adhesion of sickle cells to the vascular endothelium. Nitric oxide is a vasodilator. Free Hb is an avid scavenger of nitric oxide. Because of the continuing active hemolysis, there is free Hb in the plasma, and it scavenges nitric oxide, thus contributing to vasoconstriction.

In addition to leukocyte recruitment, inflammatory activation of endothelium may have an indispensable role in enhanced sickle RBC–endothelium interactions. Sickle RBC adhesion in postcapillary venules can cause increased microvascular transit times and initiate vaso-occlusion.

Several studies have shown involvement of an array of adhesion molecules expressed on sickle RBCs, including CD36, a-4-ß-1 integrin, intercellular cell adhesion molecule–4 (ICAM-4), and basal cell adhesion molecule (B-CAM). [10] Adhesion molecules (ie, P-selectin, VCAM-1, a-V-ß-3 integrin) are also expressed on activated endothelium. Finally, plasma factors and adhesive proteins (ie, thrombospondin [TSP], von Willebrand factor [vWf], laminin) play an important role in this interaction.

For example, the induction of VCAM-1 and P-selectin on activated endothelium is known to enhance sickle RBC interactions. In addition, a-V-ß-3 integrin is upregulated in activated endothelium in patients with sickle cell disease. a-V-ß-3 integrin binds to several adhesive proteins (TSP, vWf, red-cell ICAM-4, and, possibly, soluble laminin) involved in sickle RBC adhesion, and antibodies to this integrin dramatically inhibit sickle RBC adhesion.

In addition, under inflammatory conditions, increased leukocyte recruitment in combination with adhesion of sickle RBCs may further contribute to stasis.

Sickle RBCs adhere to endothelium because of increased stickiness. The endothelium participates in this process, as do neutrophils, which also express increased levels of adhesive molecules.

Deformable sickle cells express CD18 and adhere abnormally to endothelium up to 10 times more than normal cells, while ISCs do not. As paradoxical as it might seem, individuals who produce large numbers of ISCs have fewer vaso-occlusive crises than those with more deformable RBCs.

Other properties of sickle cells

Sickle RBCs also adhere to macrophages. This property may contribute to erythrophagocytosis and the hemolytic process.

The microvascular perfusion at the level of the pre-arterioles is influenced by RBCs containing Hb S polymers. This occurs at arterial oxygen saturation, before any morphologic change is apparent.

Hemolysis is a constant finding in sickle cell syndromes. Approximately one third of RBCs undergo intravascular hemolysis, possibly due to loss of membrane filaments during oxygenation and deoxygenation. The remainder hemolyze by erythrophagocytosis by macrophages. This process can be partially modified by Fc (crystallizable fragment) blockade, suggesting that the process can be mediated by immune mechanisms.

Sickle RBCs have increased immunoglobulin G (IgG) on the cell surface. Vaso-occlusive crisis is often triggered by infection. levels of fibrinogen, fibronectin, and D-dimer are elevated in these patients. Plasma clotting factors likely participate in the microthrombi in the pre-arterioles.

Development of clinical disease

Although hematologic changes indicative of SCD are evident as early as the age of 10 weeks, symptoms usually do not develop until the age of 6-12 months because of high levels of circulating fetal hemoglobin. After infancy, erythrocytes of patients with sickle cell anemia contain approximately 90% hemoglobin S (HbS), 2-10% hemoglobin F (HbF), and a normal amount of minor fraction of adult hemoglobin (HbA2). Adult hemoglobin (HbA), which usually gains prominence at the age of 3 months, is absent.

The physiological changes in RBCs result in a disease with the following cardinal signs:

  1. Hemolytic anemia
  2. Painful vaso-occlusive crisis
  3. Multiple organ damage from microinfarcts, including heart, skeleton, spleen, and central nervous system

Silent cerebral infarcts are associated with cognitive impairment in SCD. These infarcts tend to be located in the deep white matter where cerebral blood flow is low. [11]  However, cognitive impairment, particularly slower processing speed, may occur independent of the presence of infarction and may worsen with age. [12]

Musculoskeletal manifestations

The skeletal manifestations of sickle cell disease result from changes in bone and bone marrow caused by chronic tissue hypoxia, which is exacerbated by episodic occlusion of the microcirculation by the abnormal sickle cells. The main processes that lead to bone and joint destruction in sickle cell disease are as follows:

  • Infarction of bone and bone marrow

  • Compensatory bone marrow hyperplasia

  • Secondary growth defects

When the rigid erythrocytes jam in the arterial and venous sinusoids of skeletal tissue, the result is intravascular thrombosis, which leads to infarction of bone and bone marrow. Repeated episodes of these crises eventually lead to irreversible bone infarcts and osteonecrosis, especially in weight-bearing areas. These areas of osteonecrosis (avascular necrosis/aseptic necrosis) become radiographically visible as sclerosis of bone with secondary reparative reaction and eventually result in degenerative bone and joint destruction.

Infarction tends to occur in the diaphyses of small tubular bones in children and in the metaphyses and subchondrium of long bones in adults. Because of the anatomic distribution of the blood vessels supplying the vertebrae, infarction affecting the central part of the vertebrae (fed by a spinal artery branch) results in the characteristic H vertebrae of sickle cell disease. The outer portions of the plates are spared because of the numerous apophyseal arteries.

Osteonecrosis of the epiphysis of the femoral head is often bilateral and eventually progresses to collapse of the femoral heads. This same phenomenon is also seen in the humeral head, distal femur, and tibial condyles.

Infarction of bone and bone marrow in patients with sickle cell disease can lead to the following changes (see images below):

  • Osteolysis (in acute infarction)

  • Osteonecrosis (avascular necrosis/aseptic necrosis)

  • Articular disintegration

  • Myelosclerosis

  • Periosteal reaction (unusual in the adult)

  • H vertebrae (steplike endplate depression; also known as the Reynold sign or codfish vertebrae)

  • Dystrophic medullary calcification

  • Bone-within-bone appearance

  • The network of traits that predisposes a person to be helpful are all of the following EXCEPT
    Skeletal sickle cell anemia. H vertebrae. Lateral view of the spine shows angular depression of the central portion of each upper and lower endplate.

    The network of traits that predisposes a person to be helpful are all of the following EXCEPT
    Skeletal sickle cell anemia. Bone-within-bone appearance. Following multiple infarctions of the long bones, sclerosis may assume the appearance of a bone within a bone, reflecting the old cortex within the new cortex.

The shortened survival time of the erythrocytes in sickle cell anemia (10-20 days) leads to a compensatory marrow hyperplasia throughout the skeleton. The bone marrow hyperplasia has the resultant effect of weakening the skeletal tissue by widening the medullary cavities, replacing trabecular bone and thinning cortices.

Deossification due to marrow hyperplasia can bring about the following changes in bone:

  • Decreased density of the skull

  • Decreased thickness of outer tble of skull due to widening of diploe

  • Hair on-end striations of the calvaria

  • Osteoporosis sometimes leading to biconcave vertebrae, coarsening of trabeculae in long and flat bones, and pathologic fractures

Patients with sickle cell disease can have a variety of growth defects due to the abnormal maturation of bone. The following growth defects are often seen in sickle cell disease:

  • Bone shortening (premature epiphyseal fusion)

  • Epiphyseal deformity with cupped metaphysis

  • Peg-in-hole defect of distal femur

  • Decreased height of vertebrae (short stature and kyphoscoliosis)

Go to Skeletal Sickle Cell Anemia for complete information on this topic.

SCD can result in significant skeletal muscle remodeling and reduced muscle functional capacities, which contribute to exercise intolerance and poor quality of life. [13] In addition, changes in muscle and joints can result in altered posture and impaired balance control. [14]

Renal manifestations

Renal manifestations of SCD range from various functional abnormalities to gross anatomic alterations of the kidneys. See Nephrologic Manifestations of Sickle Cell Disease for more information on this topic.

Splenic manifestations

The spleen enlarges in the latter part of the first year of life in children with SCD. Occasionally, the spleen undergoes a sudden very painful enlargement due to pooling of large numbers of sickled cells. This phenomenon is known as splenic sequestration crisis.

The spleen undergoes repeated infarction, aided by low pH and low oxygen tension in the sinusoids and splenic cords. Despite being enlarged, its function is impaired, as evidenced by its failure to take up technetium during nuclear scanning.

Over time, the spleen becomes fibrotic and shrinks. This is, in fact, an autosplenectomy. The nonfunctional spleen is a major contributor to the immune deficiency that exists in these individuals. Failure of opsonization and an inability to deal with infective encapsulated microorganisms, particularly Streptococcus pneumoniae, ensue, leading to an increased risk of sepsis in the future.

Chronic hemolytic anemia

SCD is a form of hemolytic anemia, with red cell survival of around 10-20 days. Approximately one third of the hemolysis occurs intravascularly, releasing free hemoglobin (plasma free hemoglobin [PFH]) and arginase into plasma. PFH has been associated with endothelial injury including scavenging nitric oxide (NO), proinflammatory stress, and coagulopathy, resulting in vasomotor instability and proliferative vasculopathy.

A hallmark of this proliferative vasculopathy is the development of pulmonary hypertension in adulthood. Plasma arginase degrades arginine, the substrate for NO synthesis, thereby limiting the expected compensatory increase in NO production and resulting in generation of oxygen radicals. Plasma arginase is also associated with pulmonary hypertension and risk of early mortality.

Infection

Life-threatening bacterial infections are a major cause of morbidity and mortality in patients with SCD. Recurrent vaso-occlusion induces splenic infarctions and consequent autosplenectomy, predisposing to severe infections with encapsulated organisms (eg, Haemophilus influenzae, Streptococcus pneumoniae).

Lower serum immunoglobulin M (IgM) levels, impaired opsonization, and sluggish alternative complement pathway activation further increase susceptibility to other common infectious agents, including Mycoplasma pneumoniae, Salmonella typhimurium, Staphylococcus aureus, and Escherichia coli. Common infections include pneumonia, bronchitis, cholecystitis, pyelonephritis, cystitis, osteomyelitis, meningitis, and sepsis.

Pneumococcal sepsis continues to be a major cause of death in infants in some countries. Parvovirus B19 infection causes aplastic crises.

Etiology

SCD originated in West Africa, where it has the highest prevalence. It is also present to a lesser extent in India and the Mediterranean region. DNA polymorphism of the beta S gene suggests that it arose from five separate mutations: four in Africa and one in India and the Middle East. The most common of these is an allele found in Benin in West Africa. The other haplotypes are found in Senegal and Bantu, Africa, as well as in India and the Middle East.

The HbS gene, when present in homozygous form, is an undesirable mutation, so a selective advantage in the heterozygous form must account for its high prevalence and persistence. Malaria is possibly the selecting agent because a concordance exists between the prevalence of malaria and Hb S. Sickling might protect a person from malaria by either (1) accelerating sickling so that parasitized cells are removed or (2) making it more difficult for the parasite to metabolize or to enter the sickled cell. While children with sickle cell trait Hb SA seem to have a milder form of falciparum malaria, those with homozygous Hb S have a severe form that is associated with a very high mortality rate.

The sickling process that prompts a crisis may be precipitated by multiple factors. Local tissue hypoxia, dehydration secondary to a viral illness, or nausea and vomiting, all of which lead to hypertonicity of the plasma, may induce sickling. Any event that can lead to acidosis, such as infection or extreme dehydration, can cause sickling. More benign factors and environmental changes, such as fatigue, exposure to cold, and psychosocial stress, can elicit the sickling process. A specific cause is often not identified.

Vaso-occlusive crises are often precipitated by the following:

  • Cold weather (due to vasospasm)

  • Hypoxia (eg, flying in unpressurized aircraft)

  • Infection

  • Dehydration (especially from exertion or during warm weather)

  • Acidosis

  • Alcohol intoxication

  • Emotional stress

  • Pregnancy

Data also suggest a role for exertional stress, particularly when compounded with heat and hypovolemia.

Aplastic crises are often preceded by the following:

  • Infection with parvovirus B19

  • Folic acid deficiency

  • Ingestion of bone marrow toxins (eg, phenylbutazone)

Acute chest syndrome has been linked to the following:

  • Fat embolism

  • Infections

  • Pain episodes

  • Asthma. [15]

Epidemiology

SCD is present mostly in blacks. It also is found, with much less frequency, in eastern Mediterranean and Middle East populations. Individuals of Central African Republic descent are at an increased risk for overt renal failure.

United States statistics

The sickle gene is present in approximately 8% of black Americans. The expected prevalence of sickle cell anemia in the United States is 1 in 625 persons at birth. The actual prevalence is less because of early mortality. More than 2 million people in the United States, nearly all of them of African American ancestry, carry the sickle gene. More than 30,000 patients have homozygous HbS disease.

The following statistics are available from the Centers for Disease Control and Prevention and the National Institutes of Health [16, 17] :

  • Sickle cell anemia is the most common inherited blood disorder in the United States

  • In the United States, approximately 100,000 people have SCD

  • SCD occurs in about 1 of every 16,300 Hispanic-American births

  • Approximately 1 in 13 black or African Americans has sickle cell trait

In the United States, SCD accounts for less than 1% of all new cases of end-stage renal disease (ESRD). [18] The following factors are known to portend a greater likelihood of progression to overt renal failure: hypertension, nephrotic-range proteinuria, hematuria, severe anemia, and a Central African Republic heritage. [19, 20, 21] In patients with SCD, 5-18% develop renal failure. [22] In one study cohort, the median age at the time of renal failure in patients with SCD was 23.1 years.

International statistics

In several sections of Africa, the prevalence of sickle cell trait (heterozygosity) is as high as 30%. Although the disease is most frequently found in sub-Saharan Africa, it is also found in some parts of Sicily, Greece, southern Turkey, and India, all of which have areas in which malaria is endemic.

The mutation that results in HbS is believed to have originated in several locations in Africa and India. Its prevalence varies but is high in these countries because of the survival advantage to heterozygotes in regions of endemic malaria. As a result of migration, both forced and voluntary, it is now found worldwide.

Sex distribution

The male-to-female ratio is 1:1. No sex predilection exists, since sickle cell anemia is not an X-linked disease.

Although no particular gender predilection has been shown in most series, analysis of the data from the US Renal Data System demonstrated marked male predominance of sickle cell nephropathy in affected patients. [23]

Clinical characteristics at different ages

Although hematologic changes indicative of the disorder are evident as early as the age of 10 weeks, clinical characteristics of SCD generally do not appear until the second half of the first year of life, when fetal Hb levels decline sufficiently for abnormalities caused by HbS to manifest. SCD then persists for the entire lifespan. After age 10 years, rates of painful crises decrease, but rates of complications increase.

The median age at the time of renal failure in patients with SCD is 23.1 years, the median survival time after the diagnosis of ESRD is about 4 years, and the median age of death is 27 years, despite dialysis treatment. [24]

Prognosis

Because SCD is a lifelong disease, prognosis is guarded. The goal is to achieve a normal life span with minimal morbidity. As therapy improves, the prognosis also improves. Morbidity is highly variable in patients with SCD, partly depending on the level of HbF. Nearly all individuals with the condition are affected to some degree and experience multiple organ system involvement. Patients with Hb SA are heterozygous carriers and essentially are asymptomatic.

Vaso-occlusive crisis and chronic pain are associated with considerable economic loss and disability. Repeated infarction of joints, bones, and growth plates leads to aseptic necrosis, especially in weightbearing areas such as the femur. This complication is associated with chronic pain and disability and may require changes in employment and lifestyle.

Prognostic factors in SCD

The following prognostic factors have been identified as predictors of an adverse outcome [25] :

  • Hand-foot syndrome (dactylitis) in infants younger than 1 year

  • Hb level of less than 7 g/dL

  • Leukocytosis in the absence of infection

Hand-foot syndrome, which affects children younger than 5 years, has proved a strong predictor of overall severity (ie, death, risk of stroke, high pain rate, recurrent acute chest syndrome). Those that have an episode before age 1 year are at high risk of a severe clinical course. The risk is further increased if the child's baseline hemoglobin level is less than 7 g/dL or the baseline WBC count is elevated.

Pregnancy in SCD

Pregnancy represents a special area of concern. The high rate of fetal loss is due to spontaneous abortion. Placenta previa and abruption are common due to hypoxia and placental infarction. At birth, the infant often is premature or has low birth weight.

Mortality in SCD

Mortality is high, especially in the early childhood years. Since the introduction of widespread penicillin prophylaxis and pneumococcal vaccination, a marked reduction has been observed in childhood deaths. The leading cause of death is acute chest syndrome. Children have a higher incidence of acute chest syndrome but a lower mortality rate than adults; the overall death rate from acute chest syndrome is 1.8% and 4 times higher in adults than in children. Causes of death are pulmonary embolism and infection.

In the Dallas newborn cohort, estimated survival at 18 years was 94%. In a recent neonatal United Kingdom cohort followed in a hospital and community-based program including modern therapy with transcranial Doppler ultrasonography (TCD) screening, the estimated survival of HbSS children at 16 years was 99%. Data from the 1995 cooperative study of SCD (CSSCD) suggested that the median survival for individuals with SCD was 48 years for women and 42 years for men. [26] This life expectancy was considerably lower than that for African Americans who do not have SCD.

In Africa, available mortality data are sporadic and incomplete. Many children are not diagnosed, especially in rural areas, and death is often attributed to malaria or other comorbid conditions.

Data from Quinn et al in 2004 suggest that mortality from SCD has improved over the past 30 years. [27] In earlier reports, approximately 50% of patients did not survive beyond age 20 years, and most did not survive to age 50 years.

In one study, the median survival time in patients with SCD after the diagnosis of ESRD was about 4 years, and the median age of death after diagnosis was 27 years, despite dialysis treatment. [24]

The cooperative study of SCD (CSSCD) estimated that the median survival for individuals with SS was 48 years for women and 42 years for men. [26] In the Dallas newborn cohort, estimated survival at 18 years was 94%. In a recent neonatal United Kingdom cohort followed in a hospital and community-based program including modern therapy with TCD screening, the estimated survival of HbSS children at 16 years was 99%.

This significant increase in life expectancy and survival of patients with SCD has been achieved thanks to early detection and introduction of disease-modifying therapies. Neonatal screening, penicillin prophylaxis for children, pneumococcal immunization, red cell transfusion for selected patients and chelation therapy, hydroxyurea therapy, parental and patient education and, above all, treatment in comprehensive centers have all likely contributed to this effect on longevity.

However, as the population of patients with SCD grows older, new chronic complications are appearing. Pulmonary hypertension is emerging as a relatively common complication and is one of the leading causes of morbidity and mortality in adults with SCD. [28]

A study of 398 outpatients with SCD in France found that the prevalence of pulmonary hypertension confirmed by right heart catheterization was 6%; echocardiography alone had a low positive predictive value for pulmonary hypertension. [29]

Patient Education

Patients must be educated about the nature of their disease. They must be able to recognize the earliest signs of a vaso-occlusive crisis and seek help, treat all febrile illness promptly, and identify environmental hazards that may precipitate a crisis. Reinforcement should occur incrementally during the course of ongoing care.

Patients or parents should be instructed on how to palpate the abdomen to detect splenic enlargement, and the importance of observation for pallor, jaundice, and fever. Teach patients to seek medical care in certain situations, including the following:

  • Persistent fever (>38.3°C)

  • Chest pain, shortness of breath, nausea, and vomiting

  • Abdominal pain with nausea and vomiting

  • Persistent headache not experienced previously

Patients should avoid the following:

  • Alcohol

  • Nonprescribed prescription drugs

  • Cigarettes, marijuana, and cocaine

  • Seeking care in multiple institutions

Families should be educated on the importance of hydration, diet, outpatient medications, and immunization protocol. Emphasize the importance of prophylactic penicillin. Patients on hydroxyurea must be educated on the importance of regular follow-up with blood counts.

Patients (including asymptomatic heterozygous carriers) should understand the genetic basis of the disease, be educated about prenatal diagnosis, and know that genetic counseling is available. Genetic testing can identify parents at risk for having a child with sickle cell disease.

If both parents have the sickle cell trait, the chance that a child will have sickle cell disease is 25%. If one parent is carrying the trait and the other actually has disease, the odds increase to 50% that their child will inherit the disease. Screening and genetic counseling theoretically have the potential to drastically reduce the prevalence of SCD. This promise has not been realized. Some authors have recommended emergency department screening or referral for patients unaware of their status as a possible heterozygote. [30]

Families should be encouraged to contact community sickle cell agencies for follow-up information, new drug protocols, and psychosocial support. Families should also follow the advances of gene therapy, bone marrow transplantation, and the usage of cord blood stem cells.

For patient education information, see the Sickle Cell Disease Directory.

  1. Pecker LH, Lanzkron S. Sickle Cell Disease. Ann Intern Med. 2021 Jan. 174 (1):ITC1-ITC16. [QxMD MEDLINE Link].

  2. Sedrak A, Kondamudi NP. Sickle Cell Disease. 2021 Jan. [QxMD MEDLINE Link]. [Full Text].

  3. Shatat IF, Jakson SM, Blue AE, Johnson MA, Orak JK, Kalpatthi R. Masked hypertension is prevalent in children with sickle cell disease: a Midwest Pediatric Nephrology Consortium study. Pediatr Nephrol. 2013 Jan. 28(1):115-20. [QxMD MEDLINE Link].

  4. Linguraru MG, Orandi BJ, Van Uitert RL, Mukherjee N, Summers RM, Gladwin MT, et al. CT and image processing non-invasive indicators of sickle cell secondary pulmonary hypertension. Conf Proc IEEE Eng Med Biol Soc. 2008. 2008:859-62. [QxMD MEDLINE Link]. [Full Text].

  5. Olujohungbe A, Howard J. The clinical care of adult patients with sickle cell disease. Br J Hosp Med (Lond). 2008 Nov. 69(11):616-9. [QxMD MEDLINE Link].

  6. Johnson L, Carmona-Bayonas A, Tick L. Management of pain due to sickle cell disease. J Pain Palliat Care Pharmacother. 2008. 22(1):51-4. [QxMD MEDLINE Link].

  7. De D. Acute nursing care and management of patients with sickle cell. Br J Nurs. 2008 Jul 10-23. 17(13):818-23. [QxMD MEDLINE Link].

  8. Teixeira RS, Terse-Ramos R, Ferreira TA, Machado VR, Perdiz MI, Lyra IM, et al. Associations between endothelial dysfunction and clinical and laboratory parameters in children and adolescents with sickle cell anemia. PLoS One. 2017. 12 (9):e0184076. [QxMD MEDLINE Link]. [Full Text].

  9. Manwani D, Frenette PS. Vaso-occlusion in sickle cell disease: pathophysiology and novel targeted therapies. Blood. 2013 Dec 5. 122 (24):3892-8. [QxMD MEDLINE Link]. [Full Text].

  10. Zen Q, Batchvarova M, Twyman CA, Eyler CE, Qiu H, De Castro LM, et al. B-CAM/LU expression and the role of B-CAM/LU activation in binding of low- and high-density red cells to laminin in sickle cell disease. Am J Hematol. 2004 Feb. 75(2):63-72. [QxMD MEDLINE Link].

  11. Ford AL, Ragan DK, Fellah S, Binkley MM, Fields ME, Guilliams KP, et al. Silent infarcts in sickle cell anemia occur in the borderzone region and are associated with low cerebral blood flow. Blood. 2018 Jul 30. [QxMD MEDLINE Link].

  12. Stotesbury H, Kirkham FJ, Kölbel M, Balfour P, Clayden JD, Sahota S, et al. White matter integrity and processing speed in sickle cell anemia. Neurology. 2018 Jun 5. 90 (23):e2042-e2050. [QxMD MEDLINE Link]. [Full Text].

  13. Merlet AN, Chatel B, Hourdé C, Ravelojaona M, Bendahan D, Féasson L, et al. How Sickle Cell Disease Impairs Skeletal Muscle Function: Implications in Daily Life. Med Sci Sports Exerc. 2018 Aug 8. [QxMD MEDLINE Link].

  14. Silva PO, Ferreira AS, Lima CMA, Guimarães FS, Lopes AJ. Balance control is impaired in adults with sickle cell anaemia. Somatosens Mot Res. 2018 Jul 16. 1-10. [QxMD MEDLINE Link].

  15. Morris CR. Asthma management: reinventing the wheel in sickle cell disease. Am J Hematol. 2009 Apr. 84(4):234-41. [QxMD MEDLINE Link].

  16. National Institutes of Health. Introduction to Genes and Disease: Anemia, Sickle Cell. National Center for Biotechnology Information. Available at http://www.ncbi.nlm.nih.gov/books/NBK22238/. Accessed: October 14, 2015.

  17. Centers for Disease Control and Prevention. Sickle Cell Disease: Health Care Professionals: Data & Statistics. Centers for Disease Control and Prevention. Department of Health and Human Services. Available at http://www.cdc.gov/ncbddd/sicklecell/data.html. Accessed: September 28, 2016.

  18. Abbott KC, Hypolite IO, Agodoa LY. Sickle cell nephropathy at end-stage renal disease in the United States: patient characteristics and survival. Clin Nephrol. 2002 Jul. 58(1):9-15. [QxMD MEDLINE Link].

  19. Powars DR, Elliott-Mills DD, Chan L, Niland J, Hiti AL, Opas LM, et al. Chronic renal failure in sickle cell disease: risk factors, clinical course, and mortality. Ann Intern Med. 1991 Oct 15. 115(8):614-20. [QxMD MEDLINE Link].

  20. Derebail VK, Nachman PH, Key NS, Ansede H, Falk RJ, Kshirsagar AV. High prevalence of sickle cell trait in African Americans with ESRD. J Am Soc Nephrol. 2010 Mar. 21(3):413-7. [QxMD MEDLINE Link]. [Full Text].

  21. Audard V, Homs S, Habibi A, Galacteros F, Bartolucci P, Godeau B, et al. Acute kidney injury in sickle patients with painful crisis or acute chest syndrome and its relation to pulmonary hypertension. Nephrol Dial Transplant. 2010 Aug. 25(8):2524-9. [QxMD MEDLINE Link].

  22. Scheinman JI. In: Holliday M, Barratt TM, Avner ED (Eds.). Sickle cell nephropathy. Baltimore: Williams and Wilkins; 1994. Pediatric Nephrology: 908.

  23. Nissenson AR, Port FK. Outcome of end-stage renal disease in patients with rare causes of renal failure. I. Inherited and metabolic disorders. Q J Med. 1989 Nov. 73(271):1055-62. [QxMD MEDLINE Link].

  24. Saborio P, Scheinman JI. Disease of the month - Sickle cell nephropathy. J Amm Soc Nephrol. 1999. 10:187.

  25. Miller ST, Sleeper LA, Pegelow CH, Enos LE, Wang WC, Weiner SJ, et al. Prediction of adverse outcomes in children with sickle cell disease. N Engl J Med. 2000 Jan 13. 342(2):83-9. [QxMD MEDLINE Link].

  26. 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. N Engl J Med. 1994 Jun 9. 330(23):1639-44. [QxMD MEDLINE Link].

  27. Quinn CT, Rogers ZR, Buchanan GR. Survival of children with sickle cell disease. Blood. 2004 Jun 1. 103(11):4023-7. [QxMD MEDLINE Link]. [Full Text].

  28. Gladwin MT, Sachdev V, Jison ML, Shizukuda Y, Plehn JF, Minter K, et al. Pulmonary hypertension as a risk factor for death in patients with sickle cell disease. N Engl J Med. 2004 Feb 26. 350(9):886-95. [QxMD MEDLINE Link].

  29. Parent F, Bachir D, Inamo J, et al. A hemodynamic study of pulmonary hypertension in sickle cell disease. N Engl J Med. 2011 Jul 7. 365(1):44-53. [QxMD MEDLINE Link].

  30. Wright SW, Zeldin MH, Wrenn K, Miller O. Screening for sickle-cell trait in the emergency department. J Gen Intern Med. 1994 Aug. 9(8):421-4. [QxMD MEDLINE Link].

  31. Chiang EY, Frenette PS. Sickle cell vaso-occlusion. Hematol Oncol Clin North Am. 2005 Oct. 19(5):771-84, v. [QxMD MEDLINE Link].

  32. Rhodes M, Akohoue SA, Shankar SM, Fleming I, Qi An A, Yu C, et al. Growth patterns in children with sickle cell anemia during puberty. Pediatr Blood Cancer. 2009 Oct. 53(4):635-41. [QxMD MEDLINE Link]. [Full Text].

  33. Telfer P, Coen P, Chakravorty S, Wilkey O, Evans J, Newell H, et al. Clinical outcomes in children with sickle cell disease living in England: a neonatal cohort in East London. Haematologica. 2007 Jul. 92(7):905-12. [QxMD MEDLINE Link].

  34. Nicholson GT, Hsu DT, Colan SD, et al. Coronary artery dilation in sickle cell disease. J Pediatr. 2011 Nov. 159(5):789-794.e1-2. [QxMD MEDLINE Link].

  35. Dahoui HA, Hayek MN, Nietert PJ, et al. Pulmonary hypertension in children and young adults with sickle cell disease: evidence for familial clustering. Pediatr Blood Cancer. 2010. 54(3):398-402.

  36. Onyekwere OC, Campbell A, Teshome M, Onyeagoro S, Sylvan C, Akintilo A, et al. Pulmonary hypertension in children and adolescents with sickle cell disease. Pediatr Cardiol. 2008 Mar. 29(2):309-12. [QxMD MEDLINE Link].

  37. Parent F, Bachir D, Inamo J, et al. A hemodynamic study of pulmonary hypertension in sickle cell disease. N Engl J Med. 2011 Jul 7. 365(1):44-53. [QxMD MEDLINE Link].

  38. Akinsola FB, Kehinde MO. Ocular findings in sickle cell disease patients in Lagos. Niger Postgrad Med J. 2004 Sep. 11 (3):203-6. [QxMD MEDLINE Link].

  39. Hingorani M, Bentley CR, Jackson H, Betancourt F, Arya R, Aclimandos WA, et al. Retinopathy in haemoglobin C trait. Eye (Lond). 1996. 10 ( Pt 3):338-42. [QxMD MEDLINE Link].

  40. Sokol JA, Baron E, Lantos G, Kazim M. Orbital compression syndrome in sickle cell disease. Ophthalmic Plast Reconstr Surg. 2008 May-Jun. 24 (3):181-4. [QxMD MEDLINE Link].

  41. Wisotsky BJ, Tesser PM, Schultz JS. Trabecular meshwork hemorrhage in a patient with sickle cell trait. Arch Ophthalmol. 1995 Mar. 113 (3):381. [QxMD MEDLINE Link].

  42. Goldbaum MH, Jampol LM, Goldberg MF. The disc sign in sickling hemoglobinopathies. Arch Ophthalmol. 1978 Sep. 96 (9):1597-600. [QxMD MEDLINE Link].

  43. Babalola OE, Wambebe CO. When should children and young adults with sickle cell disease be referred for eye assessment?. Afr J Med Med Sci. 2001 Dec. 30 (4):261-3. [QxMD MEDLINE Link].

  44. Gill HS, Lam WC. A screening strategy for the detection of sickle cell retinopathy in pediatric patients. Can J Ophthalmol. 2008 Apr. 43 (2):188-91. [QxMD MEDLINE Link].

  45. [Guideline] Meschia JF, Bushnell C, Boden-Albala B, et al. Guidelines for the primary prevention of stroke: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2014 Dec. 45 (12):3754-832. [QxMD MEDLINE Link]. [Full Text].

  46. [Guideline] Centre for Clinical Practice at NICE (UK). Guidelines for the management of the acute painful crisis in sickle cell disease. Br J Haematol. 2012 Jun. 120(5):744-52. [QxMD MEDLINE Link]. [Full Text].

  47. [Guideline] US Preventive Services Task Force. Screening for sickle cell disease in newborns: U.S. Preventive Services Task Force recommendation statement. Agency for Healthcare Research and Quality (AHRQ). Sep 2007. [Full Text].

  48. Bernard AW, Venkat A, Lyons MS. Best evidence topic report. Full blood count and reticulocyte count in painful sickle crisis. Emerg Med J. 2006 Apr. 23(4):302-3. [QxMD MEDLINE Link]. [Full Text].

  49. Cerci SS, Suslu H, Cerci C, Yildiz M, Ozbek FM, Balci TA, et al. Different findings in Tc-99m MDP bone scintigraphy of patients with sickle cell disease: report of three cases. Ann Nucl Med. 2007 Jul. 21(5):311-4. [QxMD MEDLINE Link].

  50. [Guideline] Evidence-Based Management of Sickle Cell Disease: Expert Panel Report, 2014. National Heart, Lung and Blood Institute. Available at https://www.nhlbi.nih.gov/sites/default/files/media/docs/Evd-Bsd_SickleCellDis_Rep2014.pdf. September 8, 2014; Accessed: August 31, 2018.

  51. Lee MT, Piomelli S, Granger S, Miller ST, Harkness S, Brambilla DJ, et al. Stroke Prevention Trial in Sickle Cell Anemia (STOP): extended follow-up and final results. Blood. 2006 Aug 1. 108(3):847-52. [QxMD MEDLINE Link]. [Full Text].

  52. Adams RJ, Brambilla D. Discontinuing prophylactic transfusions used to prevent stroke in sickle cell disease. N Engl J Med. 2005 Dec 29. 353(26):2769-78. [QxMD MEDLINE Link]. [Full Text].

  53. Hammoudi N, Lionnet F, Redheuil A, Montalescot G. Cardiovascular manifestations of sickle cell disease. Eur Heart J. 2020 Apr 1. 41 (13):1365-1373. [QxMD MEDLINE Link].

  54. [Guideline] Brawley OW, Cornelius LJ, Edwards LR, Gamble VN, Green BL, Inturrisi C, et al. National Institutes of Health Consensus Development Conference statement: hydroxyurea treatment for sickle cell disease. Ann Intern Med. 2008 Jun 17. 148(12):932-8. [QxMD MEDLINE Link]. [Full Text].

  55. Payne J, Aban I, Hilliard LM, Madison J, Bemrich-Stolz C, Howard TH, et al. Impact of early analgesia on hospitalization outcomes for sickle cell pain crisis. Pediatr Blood Cancer. 2018 Aug 27. e27420. [QxMD MEDLINE Link].

  56. Hand L. Sickle Cell Treatment Guideline Released. Medscape Medical News. Available at http://www.medscape.com/viewarticle/831603. Accessed: September 14, 2014.

  57. Yawn BP, Buchanan GR, Afenyi-Annan AN, Ballas SK, Hassell KL, James AH, et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. JAMA. 2014 Sep 10. 312(10):1033-48. [QxMD MEDLINE Link].

  58. Brown T. FDA OKs First New Treatment for Sickle Cell in Almost 20 Years. Medscape Medical News. Available at http://www.medscape.com/viewarticle/882617. July 7, 2017; Accessed: July 10, 2017.

  59. FDA approves new treatment for sickle cell disease. U.S. Food & Drug Administration. Available at https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm566084.htm. July 7, 2017; Accessed: July 10, 2017.

  60. Niihara Y, et al; Investigators of the Phase 3 Trial of l-Glutamine in Sickle Cell Disease. A Phase 3 Trial of l-Glutamine in Sickle Cell Disease. N Engl J Med. 2018 Jul 19. 379 (3):226-235. [QxMD MEDLINE Link].

  61. 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. N Engl J Med. 2017 Feb 2. 376 (5):429-439. [QxMD MEDLINE Link]. [Full Text].

  62. Kutlar A, Kanter J, Liles DK, Alvarez OA, Cançado RD, Friedrisch JR, et al. Effect of crizanlizumab on pain crises in subgroups of patients with sickle cell disease: A SUSTAIN study analysis. Am J Hematol. 2019 Jan. 94 (1):55-61. [QxMD MEDLINE Link].

  63. Vichinsky E, Hoppe CC, Ataga KI, et al and the, HOPE Trial Investigators. A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease. N Engl J Med. 2019 Aug 8. 381 (6):509-519. [QxMD MEDLINE Link].

  64. Gluckman E, Cappelli B, Bernaudin F, et al, behalf of Eurocord, the Pediatric Working Party of the European Society for Blood and Marrow Transplantation, et al. Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood. 2017 Mar 16. 129 (11):1548-1556. [QxMD MEDLINE Link]. [Full Text].

  65. Curtis SA, Shah NC. Gene therapy in sickle cell disease: Possible utility and impact. Cleve Clin J Med. 2020 Jan. 87 (1):28-29. [QxMD MEDLINE Link]. [Full Text].

  66. Orkin SH, Bauer DE. Emerging Genetic Therapy for Sickle Cell Disease. Annu Rev Med. 2018 Oct 24. [QxMD MEDLINE Link].

  67. Ribeil JA, Hacein-Bey-Abina S, Payen E, et al. Gene Therapy in a Patient with Sickle Cell Disease. N Engl J Med. 2017 Mar 2. 376 (9):848-855. [QxMD MEDLINE Link]. [Full Text].

  68. bluebird bio Presents New Data for LentiGlobin Gene Therapy in Sickle Cell Disease at 60th Annual Meeting of the American Society of Hematology. bluebirdbio. Available at http://investor.bluebirdbio.com/news-releases/news-release-details/bluebird-bio-presents-new-data-lentiglobin-gene-therapy-sickle. December 3, 2018; Accessed: January 29. 2019.

  69. Esrick EB, Lehmann LE, Biffi A, et al. Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease. N Engl J Med. 2021 Jan 21. 384 (3):205-215. [QxMD MEDLINE Link].

  70. McGann PT, Ware RE. Hydroxyurea therapy for sickle cell anemia. Expert Opin Drug Saf. 2015 Sep 14. 1-10. [QxMD MEDLINE Link].

  71. Heeney MM, Ware RE. Hydroxyurea for children with sickle cell disease. Pediatr Clin North Am. 2008 Apr. 55(2):483-501, x. [QxMD MEDLINE Link].

  72. Strouse JJ, Lanzkron S, Beach MC, Haywood C, Park H, Witkop C, et al. Hydroxyurea for sickle cell disease: a systematic review for efficacy and toxicity in children. Pediatrics. 2008 Dec. 122(6):1332-42. [QxMD MEDLINE Link].

  73. Hankins JS, McCarville MB, Rankine-Mullings A, Reid ME, Lobo CL, et al. Prevention of conversion to abnormal TCD with hydroxyurea in sickle cell anemia: A phase III international randomized clinical trial. Am J Hematol. 2015 Sep 28. [QxMD MEDLINE Link].

  74. Siklos (hydroxyurea) Prescribing Information [package insert]. Bryn Mawr, Pennsylvania: Medunik USA, Inc. 12/2017. Available at [Full Text].

  75. Hilliard LM, Kulkarni V, Sen B, Caldwell C, Bemrich-Stolz C, Howard TH, et al. Red blood cell transfusion therapy for sickle cell patients with frequent painful events. Pediatr Blood Cancer. 2018 Aug 27. e27423. [QxMD MEDLINE Link].

  76. Firth PG, Head CA. Sickle cell disease and anesthesia. Anesthesiology. 2004 Sep. 101(3):766-85. [QxMD MEDLINE Link].

  77. Cappellini MD, Piga A. Current status in iron chelation in hemoglobinopathies. Curr Mol Med. 2008 Nov. 8(7):663-74. [QxMD MEDLINE Link].

  78. Rienhoff HY Jr, Viprakasit V, Tay L, et al. A phase 1 dose-escalation study: safety, tolerability, and pharmacokinetics of FBS0701, a novel oral iron chelator for the treatment of transfusional iron overload. Haematologica. 2011 Apr. 96(4):521-5. [QxMD MEDLINE Link]. [Full Text].

  79. Das BB, Sobczyk W, Bertolone S, Raj A. Cardiopulmonary stress testing in children with sickle cell disease who are on long-term erythrocytapheresis. J Pediatr Hematol Oncol. 2008 May. 30(5):373-7. [QxMD MEDLINE Link].

  80. Leen JS, Ratnakaram R, Del Priore LV, Bhagat N, Zarbin MA. Anterior segment ischemia after vitrectomy in sickle cell disease. Retina. 2002 Apr. 22 (2):216-9. [QxMD MEDLINE Link].

  81. Karim A, Laghmari M, Dahreddine M, Guedira K, Ibrahimy W, Essakali N, et al. [Hyphema with secondary hemorrhage: think about sickle cell disease]. J Fr Ophtalmol. 2004 Apr. 27 (4):397-400. [QxMD MEDLINE Link].

  82. Nasrullah A, Kerr NC. Sickle cell trait as a risk factor for secondary hemorrhage in children with traumatic hyphema. Am J Ophthalmol. 1997 Jun. 123 (6):783-90. [QxMD MEDLINE Link].

  83. Rogovik AL, Li Y, Kirby MA, Friedman JN, Goldman RD. Admission and length of stay due to painful vasoocclusive crisis in children. Am J Emerg Med. 2009 Sep. 27(7):797-801. [QxMD MEDLINE Link].

  84. Wang WC, Ware RE, Miller ST, et al. Hydroxycarbamide in very young children with sickle-cell anaemia: a multicentre, randomised, controlled trial (BABY HUG). Lancet. 2011 May 14. 377(9778):1663-72. [QxMD MEDLINE Link].

  85. Gladwin MT, Kato GJ, Weiner D, et al. Nitric oxide for inhalation in the acute treatment of sickle cell pain crisis: a randomized controlled trial. JAMA. 2011 Mar 2. 305(9):893-902. [QxMD MEDLINE Link].

  86. [Guideline] Howard J, Hart N, Roberts-Harewood M, Cummins M, Awogbade M, Davis B, et al. Guideline on the management of acute chest syndrome in sickle cell disease. Br J Haematol. 2015 May. 169 (4):492-505. [QxMD MEDLINE Link]. [Full Text].

  87. Styles L, Wager CG, Labotka RJ, Smith-Whitley K, Thompson AA, Lane PA, et al. Refining the value of secretory phospholipase A2 as a predictor of acute chest syndrome in sickle cell disease: results of a feasibility study (PROACTIVE). Br J Haematol. 2012 Jun. 157 (5):627-36. [QxMD MEDLINE Link]. [Full Text].

  88. Ogunlesi F, Heeney MM, Koumbourlis AC. Systemic corticosteroids in acute chest syndrome: friend or foe?. Paediatr Respir Rev. 2014 Mar. 15 (1):24-7. [QxMD MEDLINE Link].

  89. [Guideline] Brandow AM, Carroll CP, Creary S, Edwards-Elliott R, Glassberg J, Hurley RW, et al. American Society of Hematology 2020 guidelines for sickle cell disease: management of acute and chronic pain. Blood Adv. 2020 Jun 23. 4 (12):2656-2701. [QxMD MEDLINE Link]. [Full Text].

  90. Goodwin EF, Partain PI, Lebensburger JD, Fineberg NS, Howard TH. Elective cholecystectomy reduces morbidity of cholelithiasis in pediatric sickle cell disease. Pediatr Blood Cancer. 2016 Sep 19. [QxMD MEDLINE Link].

  91. Rogers ZR. Priapism in sickle cell disease. Hematol Oncol Clin North Am. 2005 Oct. 19(5):917-28, viii. [QxMD MEDLINE Link].

  92. Burnett AL, Bivalacqua TJ, Champion HC, Musicki B. Long-term oral phosphodiesterase 5 inhibitor therapy alleviates recurrent priapism. Urology. 2006 May. 67(5):1043-8. [QxMD MEDLINE Link].

  93. Burnett AL, Anele UA, Trueheart IN, Strouse JJ, Casella JF. Randomized controlled trial of sildenafil for preventing recurrent ischemic priapism in sickle cell disease. Am J Med. 2014 Jul. 127 (7):664-8. [QxMD MEDLINE Link]. [Full Text].

  94. Chinegwundoh FI, Smith S, Anie KA. Treatments for priapism in boys and men with sickle cell disease. Cochrane Database Syst Rev. 2020 Apr 6. 4:CD004198. [QxMD MEDLINE Link].

  95. [Guideline] Furie KL, Kasner SE, Adams RJ, Albers GW, Bush RL, Fagan SC, et al. Guidelines for the prevention of stroke in patients with stroke or transient ischemic attack: a guideline for healthcare professionals from the american heart association/american stroke association. Stroke. 2011 Jan. 42(1):227-76. [QxMD MEDLINE Link]. [Full Text].

  96. Adams RJ, McKie VC, Hsu L, Files B, Vichinsky E, Pegelow C, et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography. N Engl J Med. 1998 Jul 2. 339(1):5-11. [QxMD MEDLINE Link].

  97. DeBaun MR, Gordon M, McKinstry RC,et al. Controlled trial of transfusions for silent cerebral infarcts in sickle cell anemia. N Engl J Med. 2014 Aug 21. 371(8):699-710. [QxMD MEDLINE Link].

  98. [Guideline] Goldstein LB, Bushnell CD, Adams RJ, Appel LJ, Braun LT, Chaturvedi S, et al. Guidelines for the primary prevention of stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2011 Feb. 42(2):517-84. [QxMD MEDLINE Link]. [Full Text].

  99. Freed J, Talano J, Small T, Ricci A, Cairo MS. Allogeneic cellular and autologous stem cell therapy for sickle cell disease: 'whom, when and how'. Bone Marrow Transplant. 2012 Dec. 47 (12):1489-98. [QxMD MEDLINE Link]. [Full Text].

  100. Ware RE, Helms RW, SWiTCH Investigators. Stroke With Transfusions Changing to Hydroxyurea (SWiTCH). Blood. 2012 Apr 26. 119 (17):3925-32. [QxMD MEDLINE Link].

  101. Ware RE, Davis BR, Schultz WH, Brown RC, Aygun B, et al. Hydroxycarbamide versus chronic transfusion for maintenance of transcranial doppler flow velocities in children with sickle cell anaemia-TCD With Transfusions Changing to Hydroxyurea (TWiTCH): a multicentre, open-label, phase 3, non-inferiority trial. Lancet. 2016 Feb 13. 387 (10019):661-70. [QxMD MEDLINE Link].

  102. 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. Biol Blood Marrow Transplant. 2016 Feb. 22 (2):207-211. [QxMD MEDLINE Link]. [Full Text].

  103. Tanhehco YC, Bhatia M. Hematopoietic stem cell transplantation and cellular therapy in sickle cell disease: where are we now?. Curr Opin Hematol. 2019 Nov. 26 (6):448-452. [QxMD MEDLINE Link].

  104. Krishnamurti L, Neuberg DS, Sullivan KM, et al. Bone marrow transplantation for adolescents and young adults with sickle cell disease: Results of a prospective multicenter pilot study. Am J Hematol. 2019 Apr. 94 (4):446-454. [QxMD MEDLINE Link]. [Full Text].

  105. Kar BC. Splenectomy in sickle cell disease. J Assoc Physicians India. 1999 Sep. 47(9):890-3. [QxMD MEDLINE Link].

  106. Prevention of pneumococcal disease: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. 1997 Apr 4. 46:1-24. [QxMD MEDLINE Link].

  107. Kazancioglu R, Sever MS, Yüksel-Onel D, Eraksoy H, Yildiz A, Celik AV, et al. Immunization of renal transplant recipients with pneumococcal polysaccharide vaccine. Clin Transplant. 2000 Feb. 14(1):61-5. [QxMD MEDLINE Link].

  108. Fiore AE, Shay DK, Broder K, Iskander JK, Uyeki TM, Mootrey G, et al. Prevention and control of seasonal influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices (ACIP), 2009. MMWR Recomm Rep. 2009 Jul 31. 58:1-52. [QxMD MEDLINE Link].

  109. [Guideline] Liem RI, Lanzkron S, D Coates T, DeCastro L, Desai AA, Ataga KI, et al. American Society of Hematology 2019 guidelines for sickle cell disease: cardiopulmonary and kidney disease. Blood Adv. 2019 Dec 10. 3 (23):3867-3897. [QxMD MEDLINE Link]. [Full Text].

  110. [Guideline] DeBaun MR, Jordan LC, King AA, Schatz J, Vichinsky E, Fox CK, et al. American Society of Hematology 2020 guidelines for sickle cell disease: prevention, diagnosis, and treatment of cerebrovascular disease in children and adults. Blood Adv. 2020 Apr 28. 4 (8):1554-1588. [QxMD MEDLINE Link]. [Full Text].

  111. [Guideline] Chou ST, Alsawas M, Fasano RM, Field JJ, Hendrickson JE, Howard J, et al. American Society of Hematology 2020 guidelines for sickle cell disease: transfusion support. Blood Adv. 2020 Jan 28. 4 (2):327-355. [QxMD MEDLINE Link]. [Full Text].

  112. Kanter J, Liem RI, Bernaudin F, Bolaños-Meade J, Fitzhugh CD, Hankins JS, et al. American Society of Hematology 2021 guidelines for sickle cell disease: stem cell transplantation. Blood Adv. 2021 Sep 28. 5 (18):3668-3689. [QxMD MEDLINE Link]. [Full Text].

Author

Joseph E Maakaron, MD Research Fellow, Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Lebanon

Disclosure: Nothing to disclose.

Coauthor(s)

Ali T Taher, MD, PhD, FRCP Professor of Medicine, Associate Chair of Research, Department of Internal Medicine, Division of Hematology/Oncology, Director of Research, NK Basile Cancer Center, American University of Beirut Medical Center, Lebanon

Disclosure: Nothing to disclose.

Specialty Editor Board

Jeanne Yu, PharmD

Disclosure: Nothing to disclose.

Chief Editor

Emmanuel C Besa, MD Professor Emeritus, Department of Medicine, Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University

Emmanuel C Besa, MD is a member of the following medical societies: American Association for Cancer Education, American Society of Clinical Oncology, American College of Clinical Pharmacology, American Federation for Medical Research, American Society of Hematology, New York Academy of Sciences

Disclosure: Nothing to disclose.

Additional Contributors

Mark Ventocilla, OD, FAAO Chief Executive Officer, Elder Eye Care Group, PLC; Chief Executive Officer, Mark Ventocilla, OD, Inc; President, California Eye Wear, Oakwood Optical

Mark Ventocilla, OD, FAAO is a member of the following medical societies: American Academy of Optometry, American Optometric Association

Disclosure: Nothing to disclose.

Acknowledgements

Roy Alson, MD, PhD, FACEP, FAAEM Associate Professor, Department of Emergency Medicine, Wake Forest University School of Medicine; Medical Director, Forsyth County EMS; Deputy Medical Advisor, North Carolina Office of EMS; Associate Medical Director, North Carolina Baptist AirCare

Roy Alson, MD, PhD, FACEP, FAAEM is a member of the following medical societies: Air Medical Physician Association, American Academy of Emergency Medicine, American College of Emergency Physicians, American Medical Association, National Association of EMS Physicians, North Carolina Medical Society, Society for Academic Emergency Medicine, and World Association for Disaster and Emergency Medicine

Disclosure: Nothing to disclose.

Jeffrey L Arnold, MD, FACEP Chairman, Department of Emergency Medicine, Santa Clara Valley Medical Center

Jeffrey L Arnold, MD, FACEP is a member of the following medical societies: American Academy of Emergency Medicine and American College of Physicians

Disclosure: Nothing to disclose.

Robert J Arceci, MD, PhD King Fahd Professor of Pediatric Oncology, Professor of Pediatrics, Oncology and the Cellular and Molecular Medicine Graduate Program, Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine

Robert J Arceci, MD, PhD is a member of the following medical societies: American Association for Cancer Research, American Association for the Advancement of Science, American Pediatric Society, American Society of Hematology, and American Society of Pediatric Hematology/Oncology

Disclosure: Nothing to disclose.

Wadie F Bahou, MD Chief, Division of Hematology, Hematology/Oncology Fellowship Director, Professor, Department of Internal Medicine, State University of New York at Stony Brook

Wadie F Bahou, MD is a member of the following medical societies: American Society of Hematology

Disclosure: Nothing to disclose.

Dvorah Balsam, MD Chief, Division of Pediatric Radiology, Nassau University Medical Center; Professor, Department of Clinical Radiology, State University of New York at Stony Brook

Disclosure: Nothing to disclose.

Salvatore Bertolone, MD Director, Division of Pediatric Hematology/Oncology, Department of Pediatrics, Kosair Children's Hospital; Professor, University of Louisville School of Medicine

Salvatore Bertolone, MD is a member of the following medical societies: American Academy of Pediatrics, American Association for Cancer Education, American Association of Blood Banks, American Cancer Society, American Society of Hematology, American Society of Pediatric Hematology/Oncology, and Kentucky Medical Association

Disclosure: Nothing to disclose.

Barry E Brenner, MD, PhD, FACEP Professor of Emergency Medicine, Professor of Internal Medicine, Program Director, Emergency Medicine, Case Medical Center, University Hospitals, Case Western Reserve University School of Medicine

Barry E Brenner, MD, PhD, FACEP is a member of the following medical societies: Alpha Omega Alpha, American Academy of Emergency Medicine, American College of Chest Physicians, American College of Emergency Physicians, American College of Physicians, American Heart Association, American Thoracic Society, Arkansas Medical Society, New York Academy of Medicine, New York Academy of Sciences,and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Marcel E Conrad, MD Distinguished Professor of Medicine (Retired), University of South Alabama College of Medicine

Marcel E Conrad, MD is a member of the following medical societies: Alpha Omega Alpha, American Association for the Advancement of Science, American Association of Blood Banks, American Chemical Society, American College of Physicians, American Physiological Society, American Society for Clinical Investigation, American Society of Hematology, Association of American Physicians, Association of Military Surgeons of the US, International Society of Hematology, Society for Experimental Biology and Medicine, and Southwest Oncology Group

Disclosure: No financial interests None None

Nedra R Dodds, MD Medical Director, Opulence Aesthetic Medicine

Nedra R Dodds, MD is a member of the following medical societies: American Academy of Anti-Aging Medicine, American Academy of Cosmetic Surgery, American College of Emergency Physicians, American Medical Association, National Medical Association, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

James L Harper, MD Associate Professor, Department of Pediatrics, Division of Hematology/Oncology and Bone Marrow Transplantation, Associate Chairman for Education, Department of Pediatrics, University of Nebraska Medical Center; Assistant Clinical Professor, Department of Pediatrics, Creighton University School of Medicine; Director, Continuing Medical Education, Children's Memorial Hospital; Pediatric Director, Nebraska Regional Hemophilia Treatment Center

James L Harper, MD is a member of the following medical societies: American Academy of Pediatrics, American Association for Cancer Research, American Federation for Clinical Research, American Society of Hematology, American Society of Pediatric Hematology/Oncology, Council on Medical Student Education in Pediatrics, and Hemophilia and Thrombosis Research Society

Disclosure: Nothing to disclose.

Adlette Inati, MD Head, Division of Pediatric Hematology-Oncology, Medical Director, Children's Center for Cancer and Blood Diseases, Rafik Hariri University Hospital; Research Associate, Balamand University; Head of Post Bone Marrow Transplant Clinic and Consultant Hematologist, Chronic Care Center; Founding Faculty, Lebanese American University School of Medicine, Lebanon

Adlette Inati, MD is a member of the following medical societies: Alpha Omega Alpha, American Society of Hematology, European Hematology Association, and International Society of Hematology

Disclosure: Nothing to disclose.

Ziad N Kazzi, MD Assistant Professor, Department of Emergency Medicine, Emory University; Medical Toxicologist, Georgia Poison Center

Ziad N Kazzi, MD is a member of the following medical societies: American Academy of Clinical Toxicology, American Academy of Emergency Medicine, American College of Emergency Physicians, and American College of Medical Toxicology

Disclosure: Nothing to disclose.

Richard S Krause, MD Senior Clinical Faculty/Clinical Assistant Professor, Department of Emergency Medicine, University of Buffalo State University of New York School of Medicine and Biomedical Sciences

Richard S Krause, MD is a member of the following medical societies: Alpha Omega Alpha, American Academy of Emergency Medicine, American College of Emergency Physicians, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Ashok B Raj, MD Associate Professor, Section of Pediatric Hematology and Oncology, Department of Pediatrics, Kosair Children's Hospital, University of Louisville School of Medicine

Ashok B Raj, MD is a member of the following medical societies: American Academy of Pediatrics, American Society of Pediatric Hematology/Oncology, Children's Oncology Group, and Kentucky Medical Association

Disclosure: Nothing to disclose.

Sharada A Sarnaik, MBBS Professor of Pediatrics, Wayne State University School of Medicine; Director, Sickle Cell Center, Attending Hematologist/Oncologist, Children's Hospital of Michigan

Sharada A Sarnaik, MBBS is a member of the following medical societies: American Association of Blood Banks, American Association of University Professors, American Society of Hematology, American Society of Pediatric Hematology/Oncology, New York Academy of Sciences, and Society for Pediatric Research

Disclosure: Nothing to disclose.

Hosseinali Shahidi, MD, MPH Assistant Professor, Departments of Emergency Medicine and Pediatrics, State University of New York and Health Science Center at Brooklyn

Hosseinali Shahidi, MD, MPH is a member of the following medical societies: American Academy of Pediatrics, American College of Emergency Physicians, and American Public Health Association

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

Garry Wilkes MBBS, FACEM, Director of Emergency Medicine, Calvary Hospital, Canberra, ACT; Adjunct Associate Professor, Edith Cowan University; Clinical Associate Professor, Rural Clinical School, University of Western Australia

Disclosure: Nothing to disclose.

Mary L Windle, PharmD, Adjunct Associate Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Nothing to disclose.

Ulrich Josef Woermann, MD Consulting Staff, Division of Instructional Media, Institute for Medical Education, University of Bern, Switzerland

Disclosure: Nothing to disclose.

Grace M Young, MD Associate Professor, Department of Pediatrics, University of Maryland Medical Center

Grace M Young, MD is a member of the following medical societies: American Academy of Pediatrics and American College of Emergency Physicians

Disclosure: Nothing to disclose.

Which theory specifically predicts that we will be more helpful to our relatives than to others?

According to inclusive fitness theory, people are more willing to help those they are genetically related to because relatives share a kin altruism gene and are able to pass it along. We tested this theory by examining the effect of reproductive potential on altruism.

Which of the following theories specifically predicts that we will be more altruistic towards our relatives than towards close friends?

Hamilton's rule predicts that individuals should be more likely to altruistically help closer kin and likewise, be more likely to receive help from closer kin ( Hamilton 1964 ).

What norm states that we should help only when others deserve help?

The social responsibility norm tells us that we should try to help others who need assistance, even without any expectation of future paybacks. The social responsibility norm involves a sense of duty and obligation, in which people are expected to respond to others by giving help to those in need of assistance.

In which situation are people most likely to help out?

Researchers suggest that people are most likely to help others in certain circumstances:.
They have just seen others offering help..
They are not in a hurry..
They share some similarities with the person needing help..
They are in a small town or a rural setting..
They feel guilty..

Which of the following defines the reciprocity norm?

Reciprocity norm is the rule of human interaction that says people need to reciprocate the action of another person. Simply, this means that when a person is given a gift (which can take any number of forms) by another, the person must repay the gift.