Hypereosinophilic Syndrome

Back

Background

Hypereosinophilic syndrome (HES) is a myeloproliferative disorder (MPD) characterized by persistent eosinophilia that is associated with damage to multiple organs.[1, 2, 3, 4, 5, 6, 7] Peripheral eosinophilia with tissue damage has been noted for approximately 80 years, but Hardy and Anderson first described the specific syndrome in 1968.[8] In 1975, Chusid et al defined the three features required for a diagnosis of hypereosinophilic syndrome[4] :

However, due to advances in the diagnostic techniques, secondary causes of eosinophilia can be identified in a proportion of cases that would have otherwise been classified as idiopathic hypereosinophilic syndrome.

The differential diagnosis (see DDx) of hypereosinophilic syndrome includes other causes of eosinophilia,[1, 9, 10, 11] which may be classified as familial or acquired. Familial eosinophilia is an autosomal dominant disorder with a stable eosinophil count and a benign clinical course. Acquired eosinophilia is further divided into secondary, clonal, and idiopathic eosinophilia.[12]

Secondary eosinophilia

Secondary eosinophilia is a cytokine-derived (interleukin-5 [IL-5]) reactive phenomenon. Worldwide, parasitic diseases are the most common cause, whereas in developed countries, allergic diseases are the most common cause.[1] Other causes include the following:

Clonal eosinophilia

Clonal eosinophilia is diagnosed by bone marrow histology, cytogenetics, and molecular genetics. Causes include the following:

Molecularly defined disorders include the following:

Clinicopathologically assigned disorders include the following:

Idiopathic eosinophilia

Idiopathic eosinophilia is a diagnosis of exclusion when secondary and clonal causes of eosinophilia are excluded. Hypereosinophilic syndrome is a subset of idiopathic eosinophilia characterized by persistent eosinophilia (AEC >1500/µL) of longer than 6 months' duration associated with organ damage. However, long-term follow-up and X-linked clonality studies indicate that at least some patients with hypereosinophilic syndrome have an underlying clonal myeloid malignancy or a clonal or phenotypically abnormal T-cell population, suggesting a true secondary process.

The literature now favors the view that cases of idiopathic hypereosinophilic syndrome with FIP1L1 indeed represent chronic eosinophilic leukemia, because these patients have a molecular genetic abnormality, specifically an FIP1L1–PDGFRA fusion gene.[15] In addition, there are documented cases of acute transformation to either AML or granulocytic sarcoma in some cases of hypereosinophilic syndrome after an interval as long as 24 years. In such cases, a diagnosis of chronic eosinophilic leukemia is made in retrospect when acute transformation provides indirect evidence that the condition was likely to have been a clonal, neoplastic, MPD from the beginning.

In addition, some patients with hypereosinophilic syndrome present with features typical of MPDs, such as hepatosplenomegaly, the presence of leukocyte precursors in the peripheral blood, increased alkaline phosphatase level, chromosomal abnormalities, and reticulin fibrosis. Cytogenetic studies in such cases may be normal, but molecular genetic studies may show aberrations.

The best-described aberration is the interstitial deletion on chromosome 4q12, resulting in fusion of the 5’ portion of the FIP1L1 gene to the 3’ portion of the PDGFRA gene. This fusion gene encodes for the FIP1L1–PDGFR alpha protein, the constitutively activated tyrosine kinase activity that induces eosinophilia. The prevalence of such a mutation is 0.4% in unselected cases of eosinophilia, but it can be as high as 12–88% in cohorts that meet the World Health Organization (WHO) criteria for idiopathic hypereosinophilic syndrome, particularly those with features of MPD (increased levels of tryptase and mast cells in the bone marrow).

Patients with hypereosinophilic syndrome with the PDGFRA mutation have a very high incidence of cardiac involvement and carry a poor prognosis without therapy. Fortunately, the results of imatinib therapy in such cases of hypereosinophilic syndrome are very encouraging.

The other subset of idiopathic eosinophilia, hypereosinophilic syndrome with clonal or immunophenotypically aberrant T-cells, is associated with increased secretion of IL-5 and cutaneous manifestations. Simon et al reported immunophenotypic abnormality in 16 of 60 patients with hypereosinophilic syndrome.[16] Moreover, nine patients had CD3+CD4+CD8- T cells, three had CD3+CD4-CD8+ cells, three had CD3+CD4-CD8- cells, and two had CD3-CD4+ cells (one patient had two distinct populations). Progression to T-cell lymphoma was observed in this subset of patients with hypereosinophilic syndrome, particularly those with the CD3-CD4+ phenotypes.[16, 17]

Chronic eosinophilic leukemia

Chronic eosinophilic leukemia is caused by autonomous proliferation of clonal eosinophilic precursors. Simplified criteria for the diagnosis of chronic eosinophilic leukemia include the following:

Some of the cytogenetic abnormalities that have been described in chronic eosinophilic leukemia include t(5:12) and t(8:13), and molecular genetic abnormalities include the FIP1L1-PDGFRA fusion gene and ETV6-PDGFRβ.

Other resources

Other sources of information are as follows:

Pathophysiology

Eosinophil production is governed by several cytokines, including IL-3, IL-5, and granulocyte-macrophage colony-stimulating factor (GM-CSF). IL-5 appears to be the most important cytokine that is responsible for differentiation of the eosinophil line.[2, 9]

Unlike neutrophils, eosinophils can survive in the tissues for weeks. Their survival in tissues depends on the sustained presence of cytokines. Only eosinophils and basophils and their precursors have receptors for IL-3, IL-5, and GM-CSF. In vitro, eosinophils survive less than 48 hours in the absence of cytokines.

Eosinophil granules contain toxic cationic proteins, which are the primary mediators of tissue damage. These toxins include major basic protein, eosinophil peroxidase, eosinophil-derived neurotoxin, and eosinophil cationic protein. The latter two are ribonucleases. Free radicals produced by the eosinophilic peroxidase and the respiratory burst oxidative pathway of the infiltrating eosinophils further enhance the damage.

Eosinophils amplify the inflammatory cascade by secreting chemoattractants that recruit more eosinophils. Such chemoattractants include the following:

Several mechanisms have been proposed for the pathogenesis of hypereosinophilic syndrome, including overproduction of eosinophilopoietic cytokines, their enhanced activity, and defects in the normal suppressive regulation of eosinophilopoiesis. Organ damage induced by hypereosinophilic syndrome is due to the eosinophilic infiltration of the tissues accompanied by the mediator release from the eosinophil granules. Hence, the level of eosinophilia is not a true reflection of organ damage.

The most serious complication of hypereosinophilic syndrome is cardiac involvement, which can result in myocardial fibrosis, chronic heart failure (CHF), and death. The mechanisms of cardiac damage are not entirely understood, but the damage is marked by severe endocardial fibrotic thickening of either or both ventricles, resulting in restrictive cardiomyopathy due to inflow obstruction.

Epidemiology

Frequency

Various sources indicate that true hypereosinophilic syndrome is rare. The most common cause of eosinophilia in the United States is an allergic reaction or allergic disease. Worldwide, the most common cause of eosinophilia is parasitosis.[18]

Mortality/Morbidity

Hypereosinophilic syndrome is a chronic and progressive disorder that is potentially fatal. Blast transformation could occur after many years. True idiopathic hypereosinophilic syndrome is generally indolent, but patients with characteristics suggestive of a myeloproliferative/neoplastic disorder and those who develop chronic heart failure have a worse prognosis.

An older review of 57 patients with advanced hypereosinophilic syndrome reported a mean survival of 9 months and a 3-year survival rate of 12%.[4] A later analysis from France noted an 80% survival at 5 years and a 42% survival at 15 years among 40 patients with hypereosinophilic syndrome.[19]

Race-, Sex- and Age-related Demographics

No racial predilection is reported for hypereosinophilic syndrome. There is a male predominance in hypereosinophilic syndrome, with a male-to-female ratio of 9:1.

Hypereosinophilic syndrome is most commonly diagnosed in patients aged 20-50 years, with a peak incidence in the fourth decade. Hypereosinophilic syndrome is rare in children. The incidence of hypereosinophilic syndrome seems to decrease in the elderly population.

History

Hypereosinophilic syndrome is a heterogeneous disease process; thus, it has multiple manifestations, which may occur simultaneously or individually. The presentation can be sudden and dramatic, with cardiac, neurologic, or thrombotic complications, but, more often, the onset is insidious.[20] In one case series, hypereosinophilic syndrome was discovered as an incidental finding in 12% of patients.

Virtually any organ system may be involved in hypereosinophilic syndrome, but the heart, central nervous system (CNS), skin, and respiratory tract are commonly involved. Thromboembolic disease is not infrequent. Major symptoms of hypereosinophilic syndrome include the following:

The cardiac system is one of the most frequently involved systems, and cardiac complications are a leading cause of mortality. Damage typically occurs in three stages: (1) initial acute necrosis early in the disease process that typically has no clinical manifestations but may occasionally be severe enough to cause symptoms; (2) thrombotic phase; and (3) endomyocardial fibrosis. Common symptoms in these phases include chest pain, dyspnea, or orthopnea.

Hematologic manifestations are largely nonspecific and may include fatigue, which may be due to the anemia that is occasionally observed with hypereosinophilic syndrome. Left upper quadrant pain may indicate splenomegaly, which occurs in about 40% of patients. Thrombotic episodes occur frequently and often present as neurologic symptoms. The thrombotic events may occur solely due to cardiac disease, or they may be caused by hypercoagulability. The mechanism of hypercoagulability is unknown.

Neurologic symptoms are as follows:

Respiratory manifestations are as follows:

Rheumatologic manifestations are as follows:

Dermatologic manifestations are as follows:

Gastrointestinal manifestations are as follows[22] :

Constitutional signs and symptoms are as follows:

Physical

The physical findings of hypereosinophilic syndrome are varied and parallel the clinical history.

Cardiac findings are as follows:

Hematologic findings include splenomegaly in approximately 40% of patients.

Neurologic findings are as follows:

Pulmonary findings are as follows:

Rheumatologic findings are as follows:

The skin is among the most common organ systems involved in hypereosinophilic syndrome; more than half of all patients have cutaneous involvement. In a minority of reports, skin involvement is the only manifestation of hypereosinophilic syndrome.

Most skin eruptions fall into two patterns. One pattern is angioedematous or urticarial and associated with a benign prognosis. The other pattern is erythematous, pruritic papules, plaques, and nodules, with or without ulceration. A special form of urticaria is dermatographism, which occurs in up to 75% of affected patients.

Less common cutaneous manifestations include the following:

Note the images below.



View Image

Indurated edematous plaques of hypereosinophilic syndrome on a patient's legs.



View Image

Erythroderma in a patient with hypereosinophilic syndrome.

Gastrointestinal findings are as follows:

Approach Considerations

After a thorough workup has excluded causes for secondary eosinophilia, a diagnosis of hypereosinophilic syndrome is suspected in cases of persistent eosinophilia. Any such patients with a documented absolute eosinophil count (AEC) greater than 1500/µL on at least two occasions should be evaluated for hypereosinophilic syndrome, regardless of the presence of symptoms.

A complete blood cell (CBC) count and peripheral smear is warranted.[24]

Serum tryptase levels are elevated in FIP1LI-PDGFRA –positive hypereosinophilic syndrome, as well as systemic mastocytosis with chronic eosinophilic leukemia (SM-CEL). Hence, in such cases, the workup should include the following:

Bone marrow biopsy should be evaluated for the following :

Human leukocyte antigen (HLA) typing should be done early in the course of hypereosinophilic syndrome for patients with aggressive disease, cytogenetic aberration, or the FIPL1/PDGFRA fusion gene.

Other studies include the following:

Initial evaluation of suspected hypereosinophilic syndrome should include tests to look for any evidence of end-organ damage, as follows:

Wang has proposed an algorithm for the workup of patients with hypereosinophilia.[25]

Imaging Studies

Echocardiography is helpful in the initial evaluation and monitoring of cardiac disease in patients suspected with hypereosinophilic syndrome.[26] Intracardiac thrombi may be detected, as well as the fibrosis that appears not only as areas of increased echogenicity but often as posterior mitral valve leaflet thickening. Because the papillary muscles are often involved in hypereosinophilic syndrome, mitral and tricuspid dysfunction may also be detected by echocardiography.

CT scanning of the chest, abdomen, and pelvis is done to look for lymphadenopathy and splenomegaly.

Histologic Findings

Eosinophil infiltrates are present in affected tissues in patients with hypereosinophilic syndrome. Cutaneous histologic features vary with the pattern of presentation. In patients with papular or nodular lesions, perivascular mixed cellular infiltrates (eosinophils and other cell types) are present. However, vasculitis is not present.

Blood Studies

Results of hematologic studies in patients with hypereosinophilic syndrome are as follows:

Other blood study results are as follows:

Biopsy

An endocardial biopsy may be performed via cardiac catheterization if any question about the diagnosis of hypereosinophilic syndrome exists.[28]

Perform bone marrow aspiration and biopsy, and submit samples for cytogenetic studies. Occasionally, the findings may suggest an atypical presentation of chronic myelogenous leukemia. Cytogenetic abnormalities or the presence of a myeloproliferative picture in the bone marrow may be indicative of more aggressive disease.

If cutaneous involvement is present, skin biopsies may be performed to rule out other diagnoses that have similar skin presentations, such as the following:

Approach Considerations

Whether and how to treat symptomatic hypereosinophilic syndrome depends on the clinical presentation, laboratory findings, and mutational analysis.[29] Currently, there are no recommendations for treating asymptomatic patients with hypereosinophilic syndrome, as treatment itself is not without risks. Such patients are closely monitored with serum troponin level measurements every 3-6 months, and echocardiography and pulmonary function tests every 6-12 months.

In contrast, cases of hypereosinophilic syndrome with myeloproliferative features, particularly those with FIP1L1/PDGFRA mutation, should be treated aggressively. These patients carry a worse prognosis without treatment.

In all patients withoutFIP1L1/PDGFRA mutation, glucocorticoids are the first-line therapy.[3] About one third of these cases do not respond to steroids. In such patients, interferon alpha and hydroxyurea are the second-line drugs of choice.[30] For those individuals whose conditions do not respond to first- and second-line therapy, high-dose (400 mg/d) imatinib is the treatment of choice.

For patients withFIP1L1/PDGFRA mutation, imatinib is the drug of choice. The response rate in these cases approaches 100% in various studies.

For hypereosinophilic syndrome that is refractory to the usual treatments, chemotherapeutic agents that have been used with some success include chlorambucil, etoposide, vincristine, and 2-cda (2-chlorodeoxyadenosine) and cytarabine. However, alkylating agents are usually avoided in view of their potential to induce leukemias.

Alternatively, in refractory cases, particularly those resistant to imatinib therapy, hematopoietic stem cell transplantation (HSCT) has been shown to reverse the organ dysfunction. However, because of the limited experience and complications associated with HSCT, its routine use is not justified at the present time.

Recurrent thromboembolic complications occur despite anticoagulant therapy in hypereosinophilic syndrome. Currently, there are no recommendations for prophylactic use of aspirin or warfarin in the absence of documented thrombi in hypereosinophilic syndrome.

Leukapheresis is indicated as an emergency therapy in hypereosinophilic syndrome to control symptoms due to hyperleukocytosis.

Consult a hematologist to assist with the diagnosis, management, and follow-up care of patients with unexplained eosinophilia.

Surgical Care

Management of cardiovascular disease

Valve replacement with bioprosthetic valves may be required in patients with hypereosinophilic syndrome and regurgitant lesions. Risk of thrombosis with mechanical valves is very high in patients with hypereosinophilic syndrome despite therapeutic anticoagulation.

Endocardiectomy may be required for patients with endomyocardial fibrosis, and thrombectomy may be required for individuals with thrombosis.

Splenectomy

Evidence of hypersplenism and pain due to splenic infarction are indications for splenectomy.

Glucocorticoids

Due to the rapidity and reliability of its effect, a 5-day course of prednisone (1 mg/kg/d or 60 mg/d) is the initial treatment of choice for all FIP1L1/PGDFRA– negative patients. Eosinopenia occurs within hours of steroid administration. Subsequently, prednisone is tapered from a daily dose to the lowest dose required on alternate days to maintain disease control.

Glucocorticoids decrease eosinophilopoiesis by suppressing the transcription of genes for interleukin-3 (IL-3), IL-5, and granulocyte macrophage colony-stimulating factor (GM-CSF). These agents also inhibit cytokine-dependent survival of eosinophils, resulting in their increased apoptosis. Steroids are also believed to increase rapid sequestration of eosinophils.

Almost 70% of patients with hypereosinophilic syndrome respond well to steroid therapy, especially those who present with urticaria and high IgE levels. Response to steroid therapy indicates a better prognosis.

A course of steroid therapy is also given to asymptomatic patients to establish hypereosinophilic syndrome responsiveness to steroids, in case rapidly progressive organ involvement develops in the future.

Steroids are also used in the management of imatinib-induced cardiogenic shock. In such circumstances, elevation of the serum troponin level or an abnormal echocardiographic study is an indication for starting steroids.

Imatinib

Imatinib mesylate (Gleevec) is the drug of choice for hypereosinophilic syndrome with FIP1L1/PDGFRA. A tyrosine kinase inhibitor, imatinib is also a potent inhibitor of other mutations, such as BCR-ABL, C-KIT, and PDGFRβ.

In patients with hypereosinophilic syndrome with FIP1L1/PDGFRA, imatinib induces clinical hematologic and molecular remission in the majority of cases. Resolution of symptoms and normalization of eosinophil count occur within 1-2 weeks. Bone marrow abnormalities, including myelofibrosis, resolve within 1–2 months.

In contrast, structural abnormalities in the cardiovascular system and fixed neurologic deficit may not improve with imatinib therapy. However, imatinib is shown to arrest progression of endomyocardial fibrosis if therapy is initiated before the onset of structural abnormalities.

However, in true idiopathic hypereosinophilic syndrome (FIP1L1/PDGFRA– negative), low-dose imatinib (100 mg/d) may not produce a durable remission. Response rates vary from 20% to 80%. This is thought to be due to alternate PDGFRA fusion partners. A higher dose (400 mg/d) is likely to produce partial to complete remission.

In addition, experience with imatinib in chronic myelogenous leukemia (CML) shows that it is not effective in eliminating the early progenitor cells in CML. Extrapolating these results to hypereosinophilic syndrome, lifelong therapy with imatinib would be required in majority of patients. Because FIP1L1/PDGFRA– positive hypereosinophilic syndrome is predominantly a disease of young men and oligospermia is a complication of imatinib, sperm banking before initiation of therapy should be considered.

Other complications of imatinib include the following:

A few cases of hypereosinophilic syndrome with acquired resistance to imatinib have been reported in the literature. These cases have been associated with single-base (T6741) substitution. A newer agent, PKC-412 (N-benzoyl-staurosporine; midostaurin) has been shown to have efficacy against T6741 mutation in animal models and in vitro. It competes for binding to the adenosine triphosphate (ATP) site on the protein kinase C (PKC) family of serine-threonine kinases. Bone marrow transplantation is an alternative in imatinib-resistant cases.[31]

Molecular responsiveness to imatinib is assessed by screening for the PDGFRA mutation in the peripheral blood by fluorescent in situ hybridization (FISH) or reverse transcriptase–polymerase chain reaction (RT-PCR) at 3-6 month intervals in the first year and at 6-12 months intervals thereafter.

Khoury et al reported on features that predict responsiveness to treatment with imatinib, as indicated by eosinophil count < 1.5 x 109 L at 1 month and improvement in clinical symptoms.[32] Patient groups and response rates in their study were as follows:

After patients who responded had remained in complete remission for at least 18 months, imatinib was tapered and discontinued in eight of the 16 FP patients and in one of the 13 with MHES. Six of the eight FP patients   and the patient with MHES remained in remission off therapy for a median of 29 months (range 14-36 months).

Second-line Agents

Interferon alpha is a second-line drug of choice for patients whose condition does not respond to glucocorticoids.[33, 34, 35] Hydroxyurea has also demonstrated efficacy for steroid-refractory cases.[31]

Alemtuzumab (CamPath), an anti-CD52 monoclonal antibody, has been shown to control symptoms as well as eosinophilia in patients with refractory hypereosinophilic syndrome.[36, 37] Strati et al reported complete hematologic response (CHR) for a median duration of 66 weeks in 10 of 12 patients and for a median duration of 123 weeks in five of six patients retreated after relapse; time to progression was significantly longer in patients who received alemtuzumab maintenance therapy than in those who were only observed.[38]

Mepolizumab, a humanized anti–interleukin-5 monoclonal antibody, demonstrated corticosteroid-sparing effects in a double-blind, placebo-controlled study of FIP1L1/PDGFRA-negative, corticosteroid-responsive subjects with hypereosinophilic syndrome. A study of long-term use (median exposure of 251 weeks) found that mepolizumab was well tolerated and effective for this purpose.[39]

Medication Summary

The goals of pharmacotherapy are to reduce morbidity and to prevent complications in patients with hypereosinophilic syndrome. Glucocorticoids are the first-line therapy in patients without the FIP1L1/PDGFRA mutation, while imatinib is the first-line choice in patients with the FIP1L1/PDGFRA mutation. A variety of second-line agents may be used for refractory cases.

Prednisone (Deltasone, Rayos)

Clinical Context:  Initial DOC. Once eosinophils are suppressed, the dose may be slowly tapered. Patients whose condition responds to steroids tend to have a better prognosis.

Class Summary

Corticosteroids often cause a rapid reduction in level of the eosinophilia. The mechanisms for this are not entirely clear.

Hydroxyurea (Hydrea, Droxia)

Clinical Context:  Second line of treatment. Goal is to reduce total white blood cell (WBC) count to < 10,000 cells/µL. One week of therapy may be required before a reduction of the eosinophil count is observed. Anemia and thrombocytopenia are common complications associated with this drug.

Vincristine (Vincasar PFS)

Clinical Context:  May be instituted in patients whose condition fails or is only partially responsive to hydroxyurea. A response is often observed within 1-3 d. Marrow suppression is less common than with hydroxyurea, but occurrence of neurologic toxicity may limit treatment and closely resemble the neurologic symptoms of hypereosinophilic syndrome.

Chlorambucil (Leukeran)

Clinical Context:  Primary alkylating agent used in cases in which prednisone fails and in those patients who cannot tolerate hydroxyurea or vincristine. A reasonable alternative for long-term treatment. Bone marrow suppression may be a problem.

Etoposide (Toposar)

Clinical Context:  Etoposide is a glycosidic derivative of podophyllotoxin that exerts a cytotoxic effect by stabilizing the normally transient covalent intermediates formed between the DNA substrate and topoisomerase II. The drug leads to single-stranded and double-stranded DNA breaks that arrest cellular proliferation in the late S or early G2 phase of cell cycle.

Cytarabine

Clinical Context:  An antimetabolite antineoplastic agent, cytarabine is converted intracellularly to the active compound cytarabine-5'-triphosphate, which inhibits DNA polymerase. It is metabolized in the liver, with a half-life of 1-3 h. The agent is widely distributed, including in the central nervous system and tears, after IV administration.

Imatinib (Gleevec)

Clinical Context:  Imatinib is specifically designed to inhibit the tyrosine kinase activity of bcr-abl kinase in Ph1-positive leukemic CML cell lines. It is well absorbed after oral administration, with maximum concentrations achieved within 2-4 hours. Elimination is primarily in feces in the form of metabolites.

Class Summary

Chemotherapeutic agents may be used in patients whose conditions are refractory to steroid treatment. As a group, antineoplastic agents interfere with the production of eosinophils, but they may also cause toxicity to normal tissues, especially the bone marrow.

Peginterferon alfa 2a (Pegasys, Pegasys ProClick)

Clinical Context:  Has been reported to effectively suppress eosinophilia in several different patients using several different doses. Some patients have had progression of disease despite therapy.

Interferon alpha-2b (Intron A)

Clinical Context:  Has been reported to effectively suppress eosinophilia in several different patients using several different doses. Some patients have had progression of disease despite therapy.

Class Summary

Immunomodulators are naturally produced proteins with antiviral, antitumor, and immunomodulatory actions. Alpha, beta, and gamma interferons may be given topically, systemically, and intralesionally. These agents have demonstrated efficacy in small trials.

Complications

Hypereosinophilic syndrome has many and varied complications. Their development depends entirely on which organ systems are involved in the disease process (see Presentation and Pathophysiology). The most serious complication of hypereosinophilic syndrome is cardiac involvement, which can lead to myocardial fibrosis, chronic heart failure, and death (see Pathophysiology).

Prognosis

Hypereosinophilic syndrome carries a variable prognosis. It is a chronic and progressive disorder that is potentially fatal if left untreated. Blast transformation may occur after many years.

Although initial studies of hypereosinophilic syndrome showed a very poor prognosis (a 3-y survival rate of 12%),[4] management of cardiovascular disease by early echocardiographic monitoring and advances in medical and surgical therapies have improved the overall survival. A study of 40 cases by Lefebcve et al showed a 5-year survival of 80% and a 15-year survival of 42%.[19]

The availability of tyrosine kinase inhibitors such as imatinib, which prevent progression of cardiac disease and other organ damage—particularly in FIP1L1/PGDFRA–positive cases—will likely further improve the prognosis of hypereosinophilic syndrome. However, FIP1L1/PGDFRA– negative cases of hypereosinophilic syndrome that are resistant to corticosteroids have not been shown to have a durable response to imatinib.

Lastly, additional insight into the molecular pathogenesis of such cases of hypereosinophilic syndrome is required to develop effective targeted therapies.

Features that indicate a favorable prognosis in hypereosinophilic syndrome include the following:

The presence of features that are suggestive of myeloproliferative disorder (MPD) and leukocytosis greater than 90,000/μ L carry a worse prognosis in hypereosinophilic syndrome.

Long-term Monitoring

Periodically observe patients with hypereosinophilic syndrome to confirm that the eosinophilia is controlled and that no evidence of new or worsening organ involvement occurs. Patients should be advised to report any new or worsening symptoms.

Follow up patients with hypereosinophilic syndrome with serum troponin levels every 3 months, as well as with echocardiograms and pulmonary function tests every 6 months.

What are the diagnostic criteria for chronic eosinophilic leukemia?What is hypereosinophilic syndrome?Which conditions are included in the differential diagnoses of hypereosinophilic syndrome?What causes secondary eosinophilia?What causes of clonal eosinophilia?What causes of idiopathic eosinophilia?What is the pathophysiology of hypereosinophilic syndrome?What is the role of chemoattractants in the pathophysiology of hypereosinophilic syndrome?What are the mechanisms for the pathogenesis of hypereosinophilic syndrome?What is the prevalence of hypereosinophilic syndrome?What are the mortality rates of hypereosinophilic syndrome?Which patient groups have the highest prevalence of hypereosinophilic syndrome?Which clinical history findings are characteristic of hypereosinophilic syndrome?What are the neurologic symptoms of hypereosinophilic syndrome?What are the respiratory symptoms of hypereosinophilic syndrome?What are the rheumatologic symptoms of hypereosinophilic syndrome?What are the dermatologic symptoms of hypereosinophilic syndrome?What are the GI symptoms of hypereosinophilic syndrome?What are the constitutional signs and symptoms of hypereosinophilic syndrome?Which cardiac findings are characteristic of hypereosinophilic syndrome?Which hematologic findings are characteristic hypereosinophilic syndrome?Which neurologic findings are characteristic of hypereosinophilic syndrome?Which pulmonary findings are characteristic of hypereosinophilic syndrome?Which rheumatologic findings are characteristic of hypereosinophilic syndrome?Which dermatologic findings are characteristic of hypereosinophilic syndrome?Which cutaneous findings suggest hypereosinophilic syndrome?Which GI findings are characteristic of hypereosinophilic syndrome?Which conditions should be included in the differential diagnoses of hypereosinophilic syndrome?What should be the focus of history in the evaluation of hypereosinophilic syndrome?Which medication-induced eosinophilic reactions should be included in the differential diagnoses for hypereosinophilic syndrome?What are the differential diagnoses for Hypereosinophilic Syndrome?How is hypereosinophilic syndrome diagnosed?Which tests should be performed in the evaluation of hypereosinophilic syndrome in patients with elevated serum tryptase levels?What is the role of bone marrow biopsy in the diagnosis of hypereosinophilic syndrome?When is human leukocyte antigen (HLA) typing indicated in the workup of hypereosinophilic syndrome?Which studies are helpful in the workup of hypereosinophilic syndrome?Which studies are performed in the initial evaluation of suspected hypereosinophilic syndrome?What is the role of imaging studies in the workup of hypereosinophilic syndrome?Which histologic findings are characteristic of hypereosinophilic syndrome?Which blood study findings are characteristic of hypereosinophilic syndrome?What is the role of biopsy in the evaluation of hypereosinophilic syndrome?How is hypereosinophilic syndrome treated?How is refractory hypereosinophilic syndrome treated?How are thromboembolic complications treated in hypereosinophilic syndrome?How is hyperleukocytosis treated in hypereosinophilic syndrome?When is consultation with a hematologist indicated for the treatment of hypereosinophilic syndrome?How is vascular disease treated in hypereosinophilic syndrome?When is splenectomy indicated in the treatment of hypereosinophilic syndrome?What is the role of glucocorticoids in the treatment of hypereosinophilic syndrome?What is the role of imatinib in the treatment of hypereosinophilic syndrome?What are the second-line medications used in the treatment of hypereosinophilic syndrome?What are goals of drug treatment for hypereosinophilic syndrome?Which medications in the drug class Immunomodulators are used in the treatment of Hypereosinophilic Syndrome?Which medications in the drug class Antineoplastic Agents are used in the treatment of Hypereosinophilic Syndrome?Which medications in the drug class Corticosteroids are used in the treatment of Hypereosinophilic Syndrome?What are the possible complications of hypereosinophilic syndrome?What is the prognosis of hypereosinophilic syndrome?What are favorable prognostic factors for hypereosinophilic syndrome?What is included in the long-term monitoring of patients with hypereosinophilic syndrome?

Author

Venkata Anuradha Samavedi, MBBS, MD, Internist in Houston, TX

Disclosure: Nothing to disclose.

Coauthor(s)

Joe C Files, MD, Director, Division of Hematology, Associate Chairman, Professor, Department of Internal Medicine, University of Mississippi Medical Center

Disclosure: Nothing to disclose.

Ronald A Sacher, MBBCh, FRCPC, DTM&H, Professor of Internal Medicine and Pathology, Director, Hoxworth Blood Center, University of Cincinnati Academic Health Center

Disclosure: Nothing to disclose.

Vincent E Herrin, MD, FACP, Professor of Medicine, Division of Hematology and Medical Oncology, Director, Medicine Residency Program, University of Mississippi School of Medicine

Disclosure: Nothing to disclose.

Youwen Zhou, MD, PhD, FRCPC, Associate Professor, Department of Dermatology and Skin Science, University of British Columbia Faculty of Medicine; Director, Hyperhidrosis Specialty Clinic, Co-Director, Psoriasis and Phototherapy Centre, Consulting Physician, Department of Dermatology, Vancouver General Hospital; Co-Director, Vitiligo and Pigmentation Clinic, Oncologist Consultant, Skin Tumor Program, BC Cancer Agency

Disclosure: Nothing to disclose.

Specialty Editors

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

Disclosure: Received salary from Medscape for employment. for: Medscape.

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

Disclosure: Nothing to disclose.

Additional Contributors

Antoni Ribas, MD, Assistant Professor of Medicine, Division of Hematology-Oncology, University of California at Los Angeles Medical Center

Disclosure: Nothing to disclose.

Paul Schick, MD, Emeritus Professor, Department of Internal Medicine, Jefferson Medical College of Thomas Jefferson University; Research Professor, Department of Internal Medicine, Drexel University College of Medicine; Adjunct Professor of Medicine, Lankenau Hospital

Disclosure: Nothing to disclose.

References

  1. Curtis C, Ogbogu P. Hypereosinophilic Syndrome. Clin Rev Allergy Immunol. 2016 Apr. 50 (2):240-51. [View Abstract]
  2. Galli SJ, Goetzl EJ. Eosinophils, basophils, and mast cells. Handin RI, Stossel TP, Lux SE, Stossel TP, eds. Blood: Principles and Practice of Hematology. Baltimore, Md: Lippincott Williams & Wilkins; 1995. 621-40.
  3. Klion AD, Bochner BS, Gleich GJ, et al, and The Hypereosinophilic Syndromes Working Group. Approaches to the treatment of hypereosinophilic syndromes: a workshop summary report. J Allergy Clin Immunol. 2006 Jun. 117(6):1292-302. [View Abstract]
  4. Chusid MJ, Dale DC, West BC, Wolff SM. The hypereosinophilic syndrome: analysis of fourteen cases with review of the literature. Medicine (Baltimore). 1975 Jan. 54(1):1-27. [View Abstract]
  5. Simon HU, Rothenberg ME, Bochner BS, Weller PF, Wardlaw AJ, Wechsler ME, et al. Refining the definition of hypereosinophilic syndrome. J Allergy Clin Immunol. 2010 Jul. 126(1):45-9. [View Abstract]
  6. Klion AD. Eosinophilic myeloproliferative disorders. Hematology Am Soc Hematol Educ Program. 2011. 2011:257-63. [View Abstract]
  7. Liao W, Long H, Chang CC, Lu Q. The Eosinophil in Health and Disease: from Bench to Bedside and Back. Clin Rev Allergy Immunol. 2015 Sep 26. [View Abstract]
  8. Hardy WR, Anderson RE. The hypereosinophilic syndromes. Ann Intern Med. 1968 Jun. 68(6):1220-9. [View Abstract]
  9. Wardlaw AJ. Eosinophils and Related Disorders. Kaushansky K, Lichtman MA, Prchal JT, Levi MM, Press OW, Burns LJ, Caligiuri MA, eds. Williams Hematology. 9th ed. New York, NY: McGraw-Hill Education; 2016. 947-64.
  10. Tefferi A, Patnaik MM, Pardanani A. Eosinophilia: secondary, clonal and idiopathic. Br J Haematol. 2006 Jun. 133(5):468-92. [View Abstract]
  11. Rothenberg ME. Eosinophilia. N Engl J Med. 1998 May 28. 338(22):1592-600. [View Abstract]
  12. Gleich GJ, Leiferman KM. The hypereosinophilic syndromes: current concepts and treatments. Br J Haematol. 2009 May. 145(3):271-85. [View Abstract]
  13. Helbig G, Wieczorkiewicz A, Dziaczkowska-Suszek J, Majewski M, Kyrcz-Krzemien S. T-cell abnormalities are present at high frequencies in patients with hypereosinophilic syndrome. Haematologica. 2009 Sep. 94(9):1236-41. [View Abstract]
  14. Cincin AA, Ozben B, Tanrikulu MA, Baskan O, Agirbasli M. Large apical thrombus in a patient with persistent heart failure and hypereosinophilia: Löffler endocarditis. J Gen Intern Med. 2008 Oct. 23(10):1713-8. [View Abstract]
  15. Yamada Y, Sanchez-Aguilera A, Brandt EB, et al. FIP1L1/PDGFRalpha synergizes with SCF to induce systemic mastocytosis in a murine model of chronic eosinophilic leukemia/hypereosinophilic syndrome. Blood. 2008 Sep 15. 112(6):2500-7. [View Abstract]
  16. Simon HU, Plötz SG, Dummer R, Blaser K. Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia. N Engl J Med. 1999 Oct 7. 341(15):1112-20. [View Abstract]
  17. Brugnoni D, Airó P, Rossi G, et al. A case of hypereosinophilic syndrome is associated with the expansion of a CD3-CD4+ T-cell population able to secrete large amounts of interleukin-5. Blood. 1996 Feb 15. 87(4):1416-22. [View Abstract]
  18. O'Connell EM, Nutman TB. Eosinophilia in Infectious Diseases. Immunol Allergy Clin North Am. 2015 Aug. 35 (3):493-522. [View Abstract]
  19. Lefebvre C, Bletry O, Degoulet P, et al. [Prognostic factors of hypereosinophilic syndrome. Study of 40 cases] [French]. Ann Med Interne (Paris). 1989. 140(4):253-7. [View Abstract]
  20. Klion A. Hypereosinophilic syndrome: current approach to diagnosis and treatment. Annu Rev Med. 2009. 60:293-306. [View Abstract]
  21. Schreiber A, LeBlanc M, Thibeault MM, Mathieu S. Generalized Recalcitrant Pruritus as the Presenting Manifestation of Hypereosinophilic Syndrome. J Cutan Med Surg. 2016 Jan 29. [View Abstract]
  22. Nanagas VC, Kovalszki A. Gastrointestinal Manifestations of Hypereosinophilic Syndromes and Mast Cell Disorders: a Comprehensive Review. Clin Rev Allergy Immunol. 2018 Jul 12. [View Abstract]
  23. Roufosse F, Weller PF. Practical approach to the patient with hypereosinophilia. J Allergy Clin Immunol. 2010 Jul. 126 (1):39-44. [View Abstract]
  24. Curtis C, Ogbogu PU. Evaluation and Differential Diagnosis of Persistent Marked Eosinophilia. Immunol Allergy Clin North Am. 2015 Aug. 35 (3):387-402. [View Abstract]
  25. Wang SA. The Diagnostic Work-Up of Hypereosinophilia. Pathobiology. 2018 Jun 29. 1-14. [View Abstract]
  26. Adams JC, Dal-Bianco JP, Kumar G, Callahan MJ. Hypereosinophilic syndrome with characteristic left ventricular thrombus demonstrated by contrast echocardiography. Neth Heart J. 2009 Apr. 17(4):169-70. [View Abstract]
  27. Fauci AS, Harley JB, Roberts WC, et al. NIH conference. The idiopathic hypereosinophilic syndrome. Clinical, pathophysiologic, and therapeutic considerations. Ann Intern Med. 1982 Jul. 97(1):78-92. [View Abstract]
  28. Kuchynka P, Palecek T, Masek M, Cerny V, Lambert L, Vitkova I, et al. Current Diagnostic and Therapeutic Aspects of Eosinophilic Myocarditis. Biomed Res Int. 2016. 2016:2829583. [View Abstract]
  29. Roufosse F. Management of Hypereosinophilic Syndromes. Immunol Allergy Clin North Am. 2015 Aug. 35 (3):561-75. [View Abstract]
  30. Parrillo JE, Fauci AS, Wolff SM. Therapy of the hypereosinophilic syndrome. Ann Intern Med. 1978 Aug. 89(2):167-72. [View Abstract]
  31. Gotlib J. World Health Organization-defined eosinophilic disorders: 2014 update on diagnosis, risk stratification, and management. Am J Hematol. 2014 Mar. 89(3):325-37. [View Abstract]
  32. Khoury P, Desmond R, Pabon A, Holland-Thomas N, Ware JM, Arthur DC, et al. Clinical features predict responsiveness to imatinib in platelet derived growth factor receptor alpha-negative hypereosinophilic syndrome. Allergy. 2016 Jan 21. [View Abstract]
  33. Luciano L, Catalano L, Sarrantonio C, et al. AlphaIFN-induced hematologic and cytogenetic remission in chronic eosinophilic leukemia with t(1;5). Haematologica. 1999 Jul. 84(7):651-3. [View Abstract]
  34. Yamada O, Kitahara K, Imamura K, et al. Clinical and cytogenetic remission induced by interferon-alpha in a patient with chronic eosinophilic leukemia associated with a unique t(3;9;5) translocation. Am J Hematol. 1998 Jun. 58(2):137-41. [View Abstract]
  35. Malbrain ML, Van den Bergh H, Zachée P. Further evidence for the clonal nature of the idiopathic hypereosinophilic syndrome: complete haematological and cytogenetic remission induced by interferon-alpha in a case with a unique chromosomal abnormality. Br J Haematol. 1996 Jan. 92(1):176-83. [View Abstract]
  36. Verstovsek S, Tefferi A, Kantarjian H, Manshouri T, Luthra R, Pardanani A, et al. Alemtuzumab therapy for hypereosinophilic syndrome and chronic eosinophilic leukemia. Clin Cancer Res. 2009 Jan 1. 15(1):368-73. [View Abstract]
  37. Schwartz LB, Sheikh J, Singh A. Current strategies in the management of hypereosinophilic syndrome, including mepolizumab. Curr Med Res Opin. 2010 Aug. 26(8):1933-46. [View Abstract]
  38. Strati P, Cortes J, Faderl S, Kantarjian H, Verstovsek S. Long-term follow-up of patients with hypereosinophilic syndrome treated with Alemtuzumab, an anti-CD52 antibody. Clin Lymphoma Myeloma Leuk. 2013 Jun. 13(3):287-91. [View Abstract]
  39. Roufosse FE, Kahn JE, Gleich GJ, Schwartz LB, Singh AD, Rosenwasser LJ, et al. Long-term safety of mepolizumab for the treatment of hypereosinophilic syndromes. J Allergy Clin Immunol. 2013 Feb. 131(2):461-7.e1-5. [View Abstract]
  40. Bain B. The idiopathic hypereosinophilic syndrome and eosinophilic leukemias. Haematologica. 2004 Feb. 89(2):133-7. [View Abstract]
  41. Bain BJ. Eosinophilia--idiopathic or not?. N Engl J Med. 1999 Oct 7. 341(15):1141-3. [View Abstract]
  42. Oliver JW, Deol I, Morgan DL, Tonk VS. Chronic eosinophilic leukemia and hypereosinophilic syndromes. Proposal for classification, literature review, and report of a case with a unique chromosomal abnormality. Cancer Genet Cytogenet. 1998 Dec. 107(2):111-7. [View Abstract]
  43. Weller PF. The idiopathic hypereosinophilic syndrome. Arch Dermatol. 1996 May. 132(5):583-5. [View Abstract]
  44. Weller PF, Bubley GJ. The idiopathic hypereosinophilic syndrome. Blood. 1994 May 15. 83(10):2759-79. [View Abstract]
  45. Bain BJ. Relationship between idiopathic hypereosinophilic syndrome, eosinophilic leukemia, and systemic mastocytosis. Am J Hematol. 2004 Sep. 77(1):82-5. [View Abstract]
  46. Hsieh FH. Hypereosinophilic syndrome. Ann Allergy Asthma Immunol. 2014 Jun. 112(6):484-8. [View Abstract]
  47. Schwartz RS. The hypereosinophilic syndrome and the biology of cancer. N Engl J Med. 2003 Mar 27. 348(13):1199-200. [View Abstract]

Indurated edematous plaques of hypereosinophilic syndrome on a patient's legs.

Erythroderma in a patient with hypereosinophilic syndrome.

Indurated edematous plaques of hypereosinophilic syndrome on a patient's legs.

Erythroderma in a patient with hypereosinophilic syndrome.