U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • JNMA J Nepal Med Assoc
  • v.58(226); 2020 Jun

Logo of jnepalma

A Child Lost to Follow Up Carrying Beta Thalassemia Major: A Case Report

Prakash banjade.

1 Department of Internal Medicine, Gan Regional Hospital, Maavah Health Center, Republic of Maldives

Jeetendra Bhandari

2 Department of Emergency Medicine, Chitwan Medical College, Chitwan, Nepal

Thalassemia is inherited autosomal recessive disorders characterized by reduced rate of hemoglobin synthesis due to a defect in alpha or beta globin chain synthesis. Maldives has a beta thalassemia prevalence rate of 16-18%. Classical symptoms of beta thalassemia are common on those patients who present late for blood transfusion which is common among the south Asian countries due to resource poor situation. This case is a rare case report of commonly occurring phenomenon which has been reported less among south Asian region. Reporting this case will help health worker to manage cases accordingly. A five and half year prior diagnosed case of beta thalassemia at age of 2 years and lost to follow up presented with cough, Dyspnoea, Irritability, fatigue with classic symptom of beta thalassemia. She was managed with blood transfusion and kept on continuous follow up for transfusion and iron overload management.

INTRODUCTION

Thalassemia is inherited autosomal recessive disorders characterized by reduced rate of hemoglobin synthesis due to a defect in alpha or beta globin chain synthesis. 1 It has high cases in Mediterranean, Middle east, Central Asia, Indian Subcontinent and Far East. 2 Maldives has a beta thalassemia prevalence rate of 1618 %. 3 Major reason for high cases in these regions is due to high pressure of Plasmodium falciparum malaria infection. 4 Beta Thalassemia majorpresents as severe anaemia, Skeletal anomaly with typical clinical and radiological manifestations. 5 More classical symptom are observed among the non or under treated patients. 2

CASE REPORT

A five and half year-old girl, from Cozy corner, L Maavah, Maldives presented to Emergency unit. She presented with cough, Dyspnea, Irritability, and fatigue. She had no fever. She was diagnosed case of Beta Thalassemia. She was diagnosed at the age of 2 years. She was diagnosed and lost to follow up. Her family history was not significant for any blood related disorder or any genetic disease.

On physical examination patient was ill looking. Her vitals were stable. She was clinically anemic with brittle hair and nail. Patient’s finger nails and skin extremities exhibited whitish tinge and sclera showed pallor. Her skin was ashen grey in color. She appeared dehydrated and had a body weight of 13.11 kg. She was underbuilt, under-nourished with a short stature, with evident icterus, and yellow tinged fingernails. Decayed upper tooth, not associated with pain or swelling. Head and Neck examination showed maxillary expansion, retracted upper lip and saddle nose; all together depicting the classical “Chipmunk facies”. Also noted yellowish tinge at the junction of hard and soft palate. Intraoral examination, localized periodontitis and broken teeth in lower anterior aspect as shown ( Figure 1 ). Her abdominal examination didn’t show and sign of enlargement of spleen. Her ophthalmologic and audiologic examination were done and were within normal limits.

An external file that holds a picture, illustration, etc.
Object name is JNMA-58-226-436-g1.jpg

Haematological examination was performed. Her haemoglobin was 4.5 gm/dl. Hematologic investigation revealed microcytic hypochromic anaemia with anisocy-tosis, poikilocytosis, nucleated Red Blood Cells (RBC). The impression drawn from the peripheral smear study was that of haemolytic anaemia favouring Thalassemia going for haemolytic crisis. Later Haemoglobin (Hb) electrophoresis was done which too was in favor of Beta Thalassemia major. Her Human immunodeficiency Virus (HIV), Hepatitis B, and Hepatitis C was negative. Liver function test and Renal function test were within normal limit.

She was planned for blood transfusion. She was given 3 pints of packed cell. She was given each packed cell at the rate of 220 ml in every 4 hours. Her vitals were monitored regularly during the transfusion to see any transfusion related complications. No complication ware obtained during transfusion. Then she was investigated for Haemoglobin (Hb) and found out to be 9.5 gm/dl. She was then discharged and advised to follow up in 15 days. In follow up visit her ferritin was investigated and was found to be 3562.69 ng/ml. Then she was started on iron chelating agent. She was kept on Deferoxamine B(DFO) 2 gm per dose at the rate of 4 times in a week and oral Deferasirox 400 mg once in a day dose. Her Hb was 7.5 mg/dl, she was transfused with a pint of packed cell and discharged hose with a follow up in 20 days for transfusion. She is hospitalized every 20 days for transfusion of packed RBC. She is investigated for Liver function test and thyroid function testis every 20 days. Her growth and development is assessed in every OPD visit for follow up.

Individual with beta thalassemia usually present in early life within first 2 years of life which was similar in this case where she was presented at the age of 2 years. 6 Patient presenting with beta thalassemia need regular blood transfusion from the early age. If patient miss to have blood transfusion then the classic clinical picture appears on patient. Classical clinical picture includes growth retardation, pallor, jaundice, brown pigmentation of skin, poor musculature, genu varum, hepatosplenomegaly, leg ulcers, development of masses from extramedullary haematopoiesis and skeletal changes that is due to bone marrow expansion. The changes in skeleton including deformity of long bones of the legs and typical craniofacial changes including bossing of the skull, prominent malar eminence, depression or bridges of nose, tendency to a mongoloid slant of the eye and hypertrophy of maxillae, which tends to expose the teeth. 2 In the child that presented has some of the feature like these which might be due to the late starting of blood transfusion due to lost to follow up. The classic symptoms like these are only seen in developing countries, in which the resources for carrying out long term transfusion program are not available or patient tend to care less for the disease. 2 If regular transfusion of blood is started early and continued then growth and development occurs for first 10-12 years. 7 In this case the development has been hampered due to lost to transfuse the blood.

Patient with Thalassemia major have a severe microcytic and hypochromic anaemia. Peripheral blood smear shows anisocytosis, poikilocytosis (speculated tear drops and elongated cells) and nucleated RBC. 6 Most of features were seen in our case too suggesting the classic case of beta thalassemia. The treatment modality is always blood transfusion in a regular basis. Transfusion was performed to this patient too. Regular transfusion corrects anaemia, suppress erythropoiesis and inhibit increased gastrointestinal absorption of iron. 6 According to Thalassemia International Federation transfusion has to be done every 2-3 weeks 8 which was performed in our case too.

The most common secondary complication that is due to transfusion is iron overload. Which can be assessed through the level of ferritin level. Complication can be overcome by the use of chelating drugs like DFO and Deferasirox. 6 Despite of chelating agent one can have growth retardation and failure of sexual maturation. Complications of iron overload adults with Human homeostatic iron regulator protein (HFE)-associated hereditary hemochromatosis: involvement of heart (dilated cardiomyopathy and pericarditis), liver(chronic hepatitis, fibrosis, and cirrhosis) and endocrine glands( diabetes mellitus, hypoparathyroidism, hypothyroidism, hypopituitarism and low adrenal secretion). 6 For the complication of iron overload serum ferritin has to be investigated along with endocrine function, cardiac function. 6 Regular blood monitoring for the above condition that may appear has been performing in our patient too. Along with the blood work the semi-annual assessment of growth and development has to be done for every paediatric patient. 6

Although a common disease among south Asian Population, case reports are very less reported on different circumstance. We have tried to report case that has failed to follow up despite the diagnosis. Complication can occur in any patient who deny to follow up so proper counselling should be done so that the patient comes in regular follow up and save the patient that can be prevented with timely intervention. Along with this the community should take active participation to help the patients for regular follow up and proper supply of blood needed for the patient.

ACKNOWLEGDEMENTS

We would like to thank the parents of patient who provided us with information as well as consent for the study. We like to thank Mr.Adam Hussain (Manager and translator) and Maavah health centre, island of Maavah, Republic of Maldives for their support during writing of this case report.

Consent: JNMA Case Report Consent Form was signedby the patient and the original article is attached withthe patient’s chart.

Conflict of Interest:

brand logo

DREW C. BAIRD, MD, STUART H. BATTEN, MD, MS, AND STEVEN K. SPARKS, MD

Am Fam Physician. 2022;105(3):272-280

Author disclosure: No relevant financial relationships.

Thalassemia is a group of autosomal recessive hemoglobinopathies affecting the production of normal alpha- or beta-globin chains that comprise hemoglobin. Ineffective production of alpha- or beta-globin chains may result in ineffective erythropoiesis, premature red blood cell destruction, and anemia. Chronic, severe anemia in patients with thalassemia may result in bone marrow expansion and extramedullary hematopoiesis. Thalassemia should be suspected in patients with microcytic anemia and normal or elevated ferritin levels. Hemoglobin electrophoresis may reveal common characteristics of different thalassemia subtypes, but genetic testing is required to confirm the diagnosis. Thalassemia is generally asymptomatic in trait and carrier states. Alpha-thalassemia major results in hydrops fetalis and is often fatal at birth. Beta-thalassemia major requires lifelong transfusions starting in early childhood (often before two years of age). Alpha- and beta-thalassemia inter-media have variable presentations based on gene mutation or deletion, with mild forms requiring only monitoring but more severe forms leading to symptomatic anemia and requiring transfusion. Treatment of thalassemia includes transfusions, iron chelation therapy to correct iron overload (from hemolytic anemia, intestinal iron absorption, and repeated transfusions), hydroxyurea, hematopoietic stem cell transplantation, and luspatercept. Thalassemia complications arise from bone marrow expansion, extramedullary hematopoiesis, and iron deposition in peripheral tissues. These complications include morbidities affecting the skeletal system, endocrine organs, heart, and liver. Life expectancy of those with thalassemia has improved dramatically over the past 50 years with increased availability of blood transfusions and iron chelation therapy, and improved iron overload monitoring. Genetic counseling and screening in high-risk populations can assist in reducing the prevalence of thalassemia.

Thalassemia is a group of autosomal recessive hemoglobinopathies involving ineffective production of normal alpha- or beta-globin chains, which can lead to ineffective erythropoiesis, premature red blood cell destruction, and anemia. Other common hemoglobinopathies (e.g., sickle cell disease) arise from production of abnormal globin chains. This article summarizes key evidence regarding the diagnosis and treatment of thalassemia for primary care physicians.

Epidemiology

Thalassemia prevalence is highest in Africa, India, the Mediterranean, the Middle East, and Southeast Asia. Incidence in these regions may be decreasing because of prevention programs involving premarital and preconception counseling and testing. 1 – 4

Approximately 5% and 1.5% of the world population are carriers of alpha- and beta-thalassemia, respectively. 1 , 4

Thalassemia affects 6 per 100,000 conceptions in the Americas. 5 Data specific to the United States are lacking, but California has an estimated incidence of 1 in 10,000 and 1 in 55,000 for alpha- and beta-thalassemia, respectively. 4 , 6

Pathophysiology

Hemoglobin (Hb) comprises an iron-containing heme ring and four globin chains: two alpha and two nonalpha (beta, delta, gamma). 7 – 9 See diagram at https://www.aafp.org/afp/2009/0815/p339.html#afp20090815p339-f1 . HbF is the most common type in newborns. By six months of age, HbA is predominant. Clinically significant thalassemia presents with reduced HbA production. 7 , 8

Table 1 1 , 10 , 11 and Table 2 10 – 12 detail alpha- and beta-thalassemia subtypes.

Screening and Prevention

A complete blood count should be performed in all pregnant patients to screen for thalassemia. A low mean corpuscular volume warrants Hb electrophoresis. It is also reasonable to obtain Hb electrophoresis for those in high-risk ethnic groups (African, West Indian, Mediterranean, Middle Eastern, and Southeast Asian). 13

Preconception genetic counseling and testing should be discussed with patients who have risk factors (first-degree relative with thalassemia, history of stillbirth, high-risk ethnicity, and low mean corpuscular volume). 13

Prenatal diagnosis is performed via chorionic villus sampling at 10 to 12 weeks’ gestation or amniocentesis after 15 weeks’ gestation. 13

Newborn screening for thalassemia varies by state. HbSS, beta-thalassemia/HbS, and HbS/C are the only hemoglobinopathies considered to be core conditions on the U.S. Recommended Uniform Screening Panel. Other hemoglobinopathies are listed as secondary conditions, but neither alpha- nor beta-thalassemia is included on this list. 14

SIGNS AND SYMPTOMS

Thalassemia is generally asymptomatic in trait and carrier states. 10 , 11

Moderate to severe microcytic anemia can occur in intermediate and major types of thalassemia, presenting as fatigue, lightheadedness, and syncope, as well as poor growth in children. Active hemolysis may cause hyperbilirubinemia, jaundice, and gallstones. 10

Erythropoietin stimulation from chronic anemia may induce signs of bone marrow expansion (pseudotumors, frontal bossing, maxillary hypertrophy, malar prominence, depressed nasal bridge) and extramedullary hematopoiesis (hepatosplenomegaly). 10 , 11

Nonimmune hydrops fetalis occurs in alpha-thalassemia major/Hb Bart’s disease. 11

DIAGNOSTIC TESTING

Thalassemia should be considered in patients with microcytic anemia. 10

The differential diagnosis for microcytic anemia includes iron deficiency anemia, lead poisoning, sideroblastic anemia, anemia of chronic disease, copper deficiency, zinc toxicity, and hemoglobinopathies. 15

Initial evaluation includes a complete blood count and indices, peripheral smear, serum ferritin level, and blood lead level ( Table 3 ) . 10 , 11 , 16 – 18

Peripheral smear in patients with thalassemia will typically show microcytosis, hypochromia, poikilocytosis, and target cells. 10 , 11 , 17

Normal red blood cell distribution width with microcytosis may suggest thalassemia. However, it is not sensitive or specific enough to differentiate thalassemia from iron deficiency anemia and should be used only in the context of other laboratory findings. 16

Serum ferritin measurement is the best initial test to evaluate for iron deficiency. Thalassemia should be suspected in patients with microcytic anemia and normal or elevated ferritin levels ( Figure 1 ) . 10 , 15 , 19 Other iron-related studies can be useful when the ferritin level is intermediate to low or normal but suspicion for iron deficiency anemia is still present. 10 , 19

Decreased HbA on electrophoresis is characteristic of thalassemia. Other findings include elevated HbA 2 in beta-thalassemia, and Hb Bart’s disease in alpha-thalassemia major. 10 , 17 Hb electrophoresis findings are normal in alpha-thalassemia trait and carrier states ( Table 4 ) . 10 , 20 – 22

Genetic testing should be used to confirm the diagnosis and identify mutations that correlate with severity of disease. 10 , 17

Thalassemia can present with other hemoglobinopathies ( Table 5 ) . 9

Once thalassemia is diagnosed, the degree of iron overload can be investigated further with T2*-weighted cardiac or R2*-weighted liver magnetic resonance imaging, or rarely liver biopsy. 1 , 3 , 10 , 11

Given the varied clinical manifestations of thalassemia subtypes, the Thalassaemia International Federation distinguishes between transfusion-dependent thalassemia (TDT) and non–transfusion-dependent thalassemia (NTDT). Treatment decisions are more often based on this classification than on subtype classification (i.e., carrier, trait, minor, intermedia, major). 10 , 11

case study on thalassemia

Asymptomatic thalassemia subtypes do not require treatment.

Treatment goals include maintaining Hb levels at 9 to 10.5 g per dL (90 to 105 g per L), promoting normal growth, suppressing ineffective erythropoiesis and extramedullary hematopoiesis, and controlling iron overload. 3 , 10

TRANSFUSION THERAPY

Patients with TDT require regular transfusions, usually scheduled every two to five weeks. 10 Beta-thalassemia major, nondeletional HbH disease, survived Hb Bart’s disease, and severe HbE/beta-thalassemia are typically considered TDTs. 10

Those with NTDT may require occasional transfusions for symptomatic anemia, during pregnancy, before surgery, or during serious infections. They may ultimately need chronic transfusions as anemia, extramedullary hematopoiesis, and other complications worsen or to promote growth. 3 , 11

Table 6 specifies indications for transfusion. 1 , 3 , 10 , 11 , 23

Repeated transfusions increase the risk of alloimmunization, other transfusion-associated reactions, transfusion-related infections, and iron overload. 10 , 24

IRON CHELATION THERAPY

Iron chelation therapy corrects iron overload caused by hemolytic anemia, increased intestinal iron absorption, and repeated transfusions, and is recommended for TDT if ferritin levels exceed 1,000 ng per mL (1,000 mcg per L) and for NTDT when ferritin levels exceed 800 ng per mL (800 mcg per L). 3 , 10 Table 6 lists its indications. 1 , 3 , 10 , 11 , 23

Retrospective observational studies have reported decreased overall mortality and cardiac-related mortality with chelation therapy. Mortality appears to be reduced when the therapy is started earlier in life. 25 Small randomized controlled and observational studies have shown that chelation therapy effectively reduces liver and myocardial iron concentrations. 26 , 27

eTable A summarizes iron chelation therapy options.

OTHER THERAPIES

Hydroxyurea promotes HbF production, and small observational studies have shown an association between this therapy and decreased transfusion frequency in beta-thalassemia major and intermedia. 11 , 28 However, stronger evidence is lacking. 29

Hematopoietic stem cell transplantation is potentially curative for TDT. 10 , 30 Ideal candidates are younger than 12 years with a sibling donor who is a human leukocyte antigen match to the patient. 3 , 10 An unrelated matched donor could be considered. Prospective studies have shown that with transplantation, overall survival is greater than 90% and thalassemia-free survival is greater than 80%. 31

Genotyping and evaluation for transplantation (including human leukocyte antigen typing) should be ordered for all patients once, before 12 years of age if possible. In patients 12 years or older, gene therapy may be considered.

Luspatercept (Reblozyl) is an activin receptor ligand trap that promotes erythropoiesis and decreases overall transfusion burden in beta-thalassemia. In a phase 3 randomized trial, luspatercept had a number needed to treat of 6 for one adult with TDT to have a 33% reduction in transfusion burden. 32

Experimental gene therapies can induce normal beta-globin production or reactivate HbF production in patients with TDT and could dramatically reduce the need for long-term transfusions. 33 , 34

Complications of Thalassemia

Complications develop from uncontrolled chronic anemia, bone marrow expansion, extramedullary hematopoiesis, and iron overload. Iron deposition in peripheral tissues causes oxidative stress, free radical production, and organ dysfunction. 1 , 3

eTable B summarizes thalassemia-associated complications and recommended monitoring and treatment for each.

SKELETAL ABNORMALITIES

Stimulated expansion of bone marrow causes abnormal bone growth, including enlarged craniofacial bones and spinal deformities that can compress spinal nerves. 3 , 35 , 36

Disorders of skeletal growth, including delayed sexual maturation and endocrine disorders, are common in patients with thalassemia and result in shortened stature, increased bone turnover, and lowered bone mineral density. 3 , 35

Despite adequate transfusions and iron chelation therapy, 40% to 50% of patients with beta-thalassemia major have osteopenia or osteoporosis. 37 Physical activity, calcium and vitamin D supplementation, and hormone therapy for specific endocrine disorders are preventive strategies. 10

Bisphosphonates and zinc supplementation can improve bone mineral density in patients with thalassemia and osteoporosis, and calcium and vitamin D are also recommended. 10 , 35 , 37

ENDOCRINE DISORDERS

Diabetes mellitus arises from pancreatic iron deposition, impairing insulin production, and peripheral tissue iron deposition, impairing insulin utilization. 38

Nearly 44% of patients with beta-thalassemia major have nondiabetes endocrine disorders, most commonly hypogonadotropic hypogonadism, hypothyroidism, and hypoparathyroidism. 38

Growth hormone deficiency occurs in 8% to 14% of U.S. patients with TDT. Growth hormone treatment in children may mitigate the effects of short stature. 39

Primary or secondary amenorrhea can occur in patients with TDT. 24

Since the introduction of iron chelation therapies in 1975, the prevalence of hypogonadism, diabetes, and hypothyroidism has diminished in patients with thalassemia. 10

CARDIAC COMPLICATIONS

Worldwide, 25% of patients with beta-thalassemia major have cardiac iron overload, and 42% have cardiac complications such as electrocardiogram abnormalities, myocardial fibrosis, cardiomyopathy, pulmonary hypertension, heart failure, arrhythmias, heart valve disease, pericarditis, and myocarditis. 3 , 10 , 40

The adoption of T2*-weighted magnetic resonance imaging to detect cardiac iron overload in the early 2000s, along with effective iron chelation therapies, is estimated to have reduced cardiac mortality by 71% among patients with beta-thalassemia major in the United Kingdom. 41

Iron chelation therapy is the most widely accepted approach for preventing and treating cardiac iron overload. 10 Expert opinion, based on low-quality evidence, suggests that chelation therapy can improve asymptomatic and symptomatic heart disease and reverse early left ventricular dysfunction. 10 Additionally, lower ferritin levels are associated with prolonged survival and a lower probability of heart failure. 42

SPLENOMEGALY

An enlarged spleen results from splenic hyperfunctioning to remove defective red blood cells and erythropoietin stimulation to promote hematopoiesis. 43

Splenectomy can improve baseline Hb level by 1 to 2 g per dL (10 to 20 g per L) and reduce transfusion frequency. Splenectomy carries perioperative risks and higher long-term risk of venous thromboembolism, pulmonary hypertension, and encapsulated organism infections. 43

Splenectomy is reserved for select indications: iron overload from frequent transfusions that cannot be controlled with iron chelation therapy, hypersplenism with clinically significant cytopenias, and symptomatic splenomegaly. Splenectomy should be avoided in children younger than five years because of the high risk of sepsis postsplenectomy. 10

LIVER DISEASE

Up to 15% of U.S. patients with TDT will develop liver disease due to iron deposition, including cirrhosis (2% to 7%) and hepatocellular carcinoma (0.75% to 3.5%). 24

Liver iron concentration should be measured regularly by R2*-weighted magnetic resonance imaging starting at 10 years of age. 3 , 10

If liver iron concentration increases while on chelation therapy, patient compliance should be reviewed, and dosing, frequency, or the chelating agent may need to be changed. 10

Frequent transfusions increase the risk of hepatitis C and other bloodborne pathogens. 24

THROMBOTIC EVENTS

Observational studies have identified an increased risk of thromboembolism, particularly stroke, in patients with beta-thalassemia intermedia and major. 44

Currently, no guidelines recommend antiplatelet or anticoagulant therapy for primary prevention of thrombotic events in nonpregnant patients. The Thalassaemia International Federation does recommend thromboprophylaxis with low-molecular-weight heparin in pregnant patients with TDT or NTDT, and aspirin in pregnant patients who have had a splenectomy. 10 , 11

Patients with alpha-thalassemia intermedia (deletional HbH disease) typically have a normal lifespan and may have mild anemia and splenomegaly; a small proportion will require transfusions. Patients with nondeletional HbH disease more often require transfusions, may have TDT, and have more complications. 1

Most newborns with Hb Bart’s hydrops fetalis who did not receive intrauterine transfusion die shortly after birth. Intrauterine transfusion can increase survival. 45 One study demonstrated an increased survival rate from 40% between 1987 and 1992 to 100% between 2011 and 2016. 46 Current trials are investigating the use of intrauterine bone marrow transplantation. 47

Persons with beta-thalassemia major lived a mean of 17 years in 1970, most dying by 30 years of age. 48 Recent studies demonstrated mean survival ages of 50 and 57 for patients with beta-thalassemia major and minor, respectively. The increased survival is likely attributable to increasing availability of transfusion and iron chelation therapies, and improved iron overload monitoring. 25 , 49

This article updates a previous article on this topic by Muncie and Campbell . 9

Data Sources: This article was based on literature searches in Essential Evidence Plus, the Cochrane database, U.S. Preventive Services Task Force, PubMed, and Ovid using the term thalassemia. Search dates: October 2020 to October 2021.

The opinions and assertions contained herein are the private views of the authors and are not to be construed as official or as reflecting the views of the U.S. Army Medical Department, the U.S. Army at large, or the Defense Health Agency.

Piel FB, Weatherall DJ. The α-thalassemias. N Engl J Med. 2014;371(20):1908-1916.

Penman BS, Pybus OG, Weatherall DJ, et al.; Epistatic interactions between genetic disorders of hemoglobin can explain why the sickle-cell gene is uncommon in the Mediterranean. Proc Natl Acad Sci U S A. 2009;106(50):21242-21246.

Taher AT, Musallam KM, Cappellini MD. β-Thalassemias. N Engl J Med. 2021;384(8):727-743.

Kattamis A, Forni GL, Aydinok Y, et al.; Changing patterns in the epidemiology of β-thalassemia. Eur J Haematol. 2020;105(6):692-703.

Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ. 2008;86(6):480-487.

Bender MA, Yusuf C, Davis T, et al.; Newborn screening practices and alpha-thalassemia detection — United States, 2016. MMWR Morb Mortal Wkly Rep. 2020;69(36):1269-1272.

Chui DHK, Fucharoen S, Chan V. Hemoglobin H disease: not necessarily a benign disorder. Blood. 2003;101(3):791-800.

Chui DH, Waye JS. Hydrops fetalis caused by alpha-thalassemia: an emerging health care problem. Blood. 1998;91(7):2213-2222.

Muncie HL, Campbell J. Alpha and beta thalassemia. Am Fam Physician. 2009;80(4):339-344. Accessed August 30, 2021. https://www.aafp.org/afp/2009/0815/p339.html

Cappellini MD, Farmakis D, Porter J, et al., eds. 2021 guidelines for the management of transfusion dependent thalassaemia (4th edition). Thalassaemia International Federation. Accessed October 12, 2021. https://thalassaemia.org.cy/publications/tif-publications/guidelines-for-the-management-of-transfusion-dependent-thalassaemia-4th-edition-2021/

Taher A, Musallam K, Capellini MD, eds. Guidelines for the management of non-transfusion dependent thalassaemia, 2nd edition (2017). Thalassaemia International Federation. Accessed March 19, 2021. https://thalassaemia.org.cy/publications/tif-publications/guidelines-for-the-clinical-management-of-non-transfusion-dependent-thalassaemias-updated-version/

Viprakasit V, Ekwattanakit S. Clinical classification, screening and diagnosis for thalassemia. Hematol Oncol Clin North Am. 2018;32(2):193-211.

American College of Obstetricians and Gynecologists; Committee opinion no. 691: carrier screening for genetic conditions. Obstet Gynecol. 2017;129(3):e41-e55.

Advisory Committee on Heritable Disorders in Newborns and Children. U.S. Department of Health and Human Services. Recommended uniform screening panel. July 2018. Accessed May 21, 2021. https://www.hrsa.gov/advisory-committees/heritable-disorders/rusp/index.html

Short MW, Domagalski JE. Iron deficiency anemia: evaluation and management. Am Fam Physician. 2013;87(2):98-104. Accessed August 30, 2021. https://www.aafp.org/afp/2013/0115/p98.html

Hoffmann JJML, Urrechaga E, Aguirre U. Discriminant indices for distinguishing thalassemia and iron deficiency in patients with microcytic anemia: a meta-analysis. Clin Chem Lab Med. 2015;53(12):1883-1894.

Association of Public Health Laboratories. Hemoglobinopathies: current practices for screening, confirmation and follow-up. December 2015. Accessed May 21, 2021. https://www.cdc.gov/ncbddd/sicklecell/documents/nbs_hemoglobinopathy-testing_122015.pdf

Mentzer WC. Differentiation of iron deficiency from thalassaemia trait. Lancet. 1973;1(7808):882.

Van Vranken M. Evaluation of microcytosis [published correction appears in Am Fam Physician . 2011;83(7):792]. Am Fam Physician. 2010;82(9):1117-1122. Accessed August 30, 2021. https://www.aafp.org/afp/2010/1101/p1117.html

Munkongdee T, Chen P, Winichagoon P, et al.; Update in laboratory diagnosis of thalassemia. Front Mol Biosci. 2020;7:74.

Steinberg MH. Structure and function of normal hemoglobins. UpToDate. December 5, 2019. Accessed June 28, 2021. https://www.uptodate.com/contents/structure-and-function-of-normal-hemoglobins

Benz EJ Jr., Angelucci E. Clinical manifestations and diagnosis of the thalassemias. UpToDate. January 17, 2020. Accessed June 28, 2021. https://www.uptodate.com/contents/clinical-manifestations-and-diagnosis-of-the-thalassemias

Tubman VN, Fung EB, Vogiatzi M, et al; Thalassemia Clinical Research Network; Guidelines for the standard monitoring of patients with thalassemia: report of the Thalassemia Longitudinal Cohort. J Pediatr Hematol Oncol. 2015;37(3):e162-e169.

Betts M, Flight PA, Paramore LC, et al.; Systematic literature review of the burden of disease and treatment for transfusion-dependent β-thalassemia. Clin Ther. 2020;42(2):322-337.e2.

Ballas SK, Zeidan AM, Duong VH, et al.; The effect of iron chelation therapy on overall survival in sickle cell disease and β-thalassemia: a systematic review. Am J Hematol. 2018;93(7):943-952.

Chapchap EC, Silva MMA, de Assis RA, et al. Cardiac iron overload evaluation in thalassaemic patients using T2* magnetic resonance imaging following chelation therapy: a multicentre cross-sectional study [published online May 9, 2021]. Hematol Transfus Cell Ther . Accessed June 22, 2021. https://www.sciencedirect.com/science/article/pii/S2531137921000432

Taher AT, Porter JB, Viprakasit V, et al.; Deferasirox effectively reduces iron overload in non-transfusion-dependent thalassemia (NTDT) patients: 1-year extension results from the THALASSA study. Ann Hematol. 2013;92(11):1485-1493.

Banan M. Hydroxyurea treatment in β-thalassemia patients: to respond or not to respond?. Ann Hematol. 2013;92(3):289-299.

Ansari SH, Lassi ZS, Khowaja SM, et al.; Hydroxyurea (hydroxycarbamide) for transfusion-dependent β-thalassaemia. Cochrane Database Syst Rev. 2019;(3):CD012064.

Jagannath VA, Fedorowicz Z, Al Hajeri A, et al.; Hematopoietic stem cell transplantation for people with β-thalassaemia major. Cochrane Database Syst Rev. 2016;(11):CD008708.

Angelucci E. Hematopoietic stem cell transplantation in thalassemia. Hematology Am Soc Hematol Educ Program. 2010;2010(1):456-462.

Cappellini MD, Viprakasit V, Taher AT, et al; BELIEVE Investigators; A phase 3 trial of luspatercept in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2020;382(13):1219-1231.

Thompson AA, Walters MC, Kwiatkowski J, et al.; Gene therapy in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2018;378(16):1479-1493.

Frangoul H, Altshuler D, Cappellini MD, et al.; CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. N Engl J Med. 2021;384(3):252-260.

Tsartsalis AN, Lambrou GI, Tsartsalis D, et al.; The role of biphosphonates in the management of thalassemia-induced osteoporosis: a systematic review and meta-analysis. Hormones (Athens). 2018;17(2):153-166.

Einy S, Ben-Barak A, Kridin K, et al.; Craniofacial deformities in patients with beta-thalassemia: orthodontic versus surgical correction–a systematic review. J Pediatr Hematol Oncol. 2020;42(3):198-203.

Bhardwaj A, Swe KMM, Sinha NK, et al.; Treatment for osteoporosis in people with β-thalassaemia. Cochrane Database Syst Rev. 2016;(3):CD010429.

He LN, Chen W, Yang Y, et al.; Elevated prevalence of abnormal glucose metabolism and other endocrine disorders in patients with β-thalassemia major: a meta-analysis. Biomed Res Int. 2019;2019:6573497.

Ngim CF, Lai NM, Hong JY, et al.; Growth hormone therapy for people with thalassaemia. Cochrane Database Syst Rev. 2020;(5):CD012284.

Koohi F, Kazemi T, Miri-Moghaddam E. Cardiac complications and iron overload in beta thalassemia major patients—a systematic review and meta-analysis. Ann Hematol. 2019;98(6):1323-1331.

Modell B, Khan M, Darlison M, et al.; Improved survival of thalassemia major in the UK and relation to T2* cardiovascular magnetic resonance. J Cardiovasc Mag Reson. 2008;10(1):42.

Borgna-Pignatti C, Rugolotto S, De Stefano P, et al.; Survival and complications in patients with thalassemia major treated with transfusion and deferoxamine. Haematologica. 2004;89(10):1187-1193.

Sharma A, Easow Mathew M, Puri L. Splenectomy for people with thalassaemia major or intermedia. Cochrane Database Syst Rev. 2019;(9):CD010517.

Haghpanah S, Karimi M. Cerebral thrombosis in patients with β-thalassemia: a systematic review. Blood Coagul Fibrinolysis. 2012;23(3):212-217.

Weisz B, Rosenbaum O, Chayen B, et al.; Outcome of severely anaemic fetuses treated by intrauterine transfusions. Arch Dis Child Fetal Neonatal Ed. 2009;94(3):F201-F204.

Zwiers C, Oepkes D, Lopriore E, et al.; The near disappearance of fetal hydrops in relation to current state-of-the-art management of red cell alloimmunization. Prenat Diagn. 2018;38(12):943-950.

King AJ, Higgs DR. Potential new approaches to the management of the Hb Bart's hydrops fetalis syndrome: the most severe form of α-thalassemia. Hematology Am Soc Hematol Educ Program. 2018;2018(1):353-360.

Modell B, Khan M, Darlison M. Survival in beta-thalassaemia major in the UK: data from the UK Thalassaemia Register. Lancet. 2000;355(9220):2051-2052.

Ansari-Moghaddam A, Adineh HA, Zareban I, et al.; The survival rate of patients with beta-thalassemia major and intermedia and its trends in recent years in Iran. Epidemiol Health. 2018;40:e2018048.

Continue Reading

case study on thalassemia

More in AFP

More in pubmed.

Copyright © 2022 by the American Academy of Family Physicians.

This content is owned by the AAFP. A person viewing it online may make one printout of the material and may use that printout only for his or her personal, non-commercial reference. This material may not otherwise be downloaded, copied, printed, stored, transmitted or reproduced in any medium, whether now known or later invented, except as authorized in writing by the AAFP.  See permissions  for copyright questions and/or permission requests.

Copyright © 2024 American Academy of Family Physicians. All Rights Reserved.

  • Reference Manager
  • Simple TEXT file

People also looked at

Case report article, case report: clinical and hematological characteristics of ε γδβ thalassemia in an italian patient.

case study on thalassemia

  • 1 Department of Pediatric Hematology/Oncology and Hematopoietic Stem Cell Transplantation (HSCT), Meyer Children's University Hospital, Florence, Italy
  • 2 Department of Health Science, University of Florence, Florence, Italy
  • 3 Human Genetics Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy

Introduction: ε γδβ thalassemia is a rare form of β-thalassemia mostly described in children originating from Northern Europe. Only anecdotic cases from the Mediterranean area are reported. The diagnosis is challenging, considering the rarity of the disease and its heterogeneous clinical presentation. Most patients have neonatal microcytic anemia, sometimes requiring in utero and/or neonatal transfusions, and typically improving with age.

Case Description: We report on an Italian newborn presenting with severe neonatal anemia that required red blood cell transfusion. After the first months of life, hemoglobin levels improved with residual very low mean corpuscular volume. β and α thalassemia, IRIDA syndrome, and sideroblastic anemia were excluded. Finally, a diagnosis of ε γδβ thalassemia was made after microarray analysis of single nucleotide polymorphisms revealed a 26 kb single copy loss of chromosome 11p15.4, including the HBD, HBBP1, HBG1, and HBB genes.

Conclusions: Despite its rarity, the diagnosis of ε γδβ thalassemia should be considered in newborns with severe neonatal anemia requiring in utero and/or neonatal transfusions, but also in older infants with microcytic anemia, after excluding more prevalent red blood cell disorders.

Introduction

The ε γδβ thalassemia is an extremely rare heterozygous form of β-thalassemia, with around 40 reported cases in 2019 ( 1 ). In most cases, patients originated from ethnic backgrounds where β-thalassemia was not prevalent ( Table 1 ). Despite the extreme heterogeneity of the molecular bases of β-thalassemia in Italy, the first ε γδβ thalassemia deletion has been only identified in 2016 ( 23 ).

www.frontiersin.org

Table 1 . Origin and presentation of previously described patients with ε γδβ thalassemia.

ε γδβ thalassemias are caused by long deletions in the β-globin cluster and exist only in heterozygous form. Except for one case ( 8 , 27 ), the reported deletions are almost exclusively unique and in most cases de novo , explaining the phenotypic heterogeneity of the disease. Indeed, multiple clinical phenotypes of ε γδβ thalassemia have been reported, ranging from normal blood cell count to severe anemia requiring in utero and/or neonatal transfusions ( Table 1 ) ( 20 ). The underlying reasons for such a spectrum of clinical characteristics are unknown, but the type and length of the deletion are not responsible, as contrasting phenotypes have been reported in heterozygotes with identical deletions within the same family ( 8 ). At the molecular level ε γδβ thalassemias fall into two distinct categories: in group I all, or a greater part of the β-globin cluster, are removed, including the β-globin gene, whereas in group II extensive upstream regions are removed, leaving the β-globin gene itself intact although its expression is silenced because of inactivation of the upstream β-locus control region ( 23 ). Furthermore, co-existent α-globin gene triplication has been suggested to exacerbate the phenotype of ε γδβ thalassemia increasing the imbalance between the α and non-α globin chain ratio during fetal life ( 16 ).

Most patients with ε γδβ -thalassemia had neonatal erythroblastosis, reticulocytosis, hypochromia, and microcytosis ( Table 1 ), that later improved with age. Anemia usually remitted spontaneously during the first months of life, and the adult phenotype is similar to that of the β-thalassemia trait, but with more severe microcytosis ( 13 ).

Herein, we describe the clinical phenotype of a novel Italian ε γδβ deletion, the second patient from Italy described in the literature and the third from the Mediterranean Area, presenting with severe microcytic anemia in the neonatal period.

Case Description

A male, full-term infant of Tuscanian origin was born by induced vaginal delivery due to meconium-stained amniotic fluid. He presented with clinical and laboratory signs of sepsis (increased white blood cell count, C-reactive protein, and indirect bilirubin) and received wide spectrum antibiotics. Laboratory evaluations revealed microcytic anemia (hemoglobin, Hb, 10.8 g/dL, mean corpuscular volume, MCV, 65.4 fL). The clinical condition rapidly improved and hemoglobin rose to 12 g/dL, with persistent microcytemia. At the first follow-up visit at 1 month of age, hemoglobin had dropped to 6.4 g/dL, with a MCV of 53.8 fL, a mean cell hemoglobin concentration of 18.3 g/dL, a hematocrit of 19.9%, and an increased reticulocyte count (0.2 × 10 6 /L) ( Figure 1 ). No other signs of hemolysis were detected (normal bilirubin and lactate dehydrogenase levels). The peripheral blood smear revealed microcytic hypochromic erythrocytes with anisopoikilocytosis ( Figure 2 ). Hemoglobin electrophoresis showed a normal pattern with an unusually high proportion of HbA (HbF 47%, HbA2 0.8%, HbA 52.2%), no abnormal hemoglobin variants, nor evidence of β-thalassemia; abdominal ultrasound showed splenomegaly. The patient received a red blood cell transfusion and supplementation of iron and folic acid, which proved ineffective. Therefore, bone marrow aspiration was performed to exclude the presence of ring sideroblasts with Prussian blue staining ( Figure 2 ); normal plasmatic hepcidin values ruled out an Iron-Refractory Iron Deficiency Anemia (IRIDA) syndrome.

www.frontiersin.org

Figure 1 . Timeline graph showing the chronological evolution of the values of hemoglobin and mean corpuscular volume from birth to last follow-up. Hemoglobin electrophorese studies and transfusions are also reported. Hb, hemoglobin; EPh, electrophoresis; MCV, mean corpuscular volume.

www.frontiersin.org

Figure 2 . Peripheral blood smear (A) performed in the neonatal period, showing hypochromic erythrocytes with anisopoikilocytosis; isolated target cells, ovalocytes, ellissocytes, and dacrocytes are also visible (600x magnification, MGG). Bone marrow aspirate (B) performed at 2 months of age showing mild dyserythropoiesis (1000x magnification, MGG); no ring sideroblasts were found in the smear [ (C) 1000x magnification, Pearls coloration].

At 6 months of age, the blood cell count of the patient was consistent with a thalassemia trait (hemoglobin 9 g/dL, red blood cell 6.19 × 10 12 /L, MCV 52 fL, mean cell hemoglobin, MCH, 6.3 pg). The hemoglobin electrophoresis showed HbA2 value of 3.3%, and α gene deletions were excluded using Multiplex Ligation Probe Amplification (MLPA). Conversely, MLPA showed a heterozygous deletion in the short arm of chromosome 11 ( Figure 3 ). This was confirmed by microarray analysis of single nucleotide polymorphisms that revealed a 26 kb single-copy loss of a genomic region localized at 11p15.4. The lost genetic material included the HBD, HBBP1, HBG1 - and partially HBB - genes, a finding consistent with ε γδβ thalassemia. The family history was negative for similarly affected individuals and targeted parental testing via quantitative polymerase chain reaction confirmed the presence of a de novo deletion.

www.frontiersin.org

Figure 3 . Multiplex Ligation Probe Amplification (MLPA) showing a deletion of the OR51V1-1, HBB, HBD, HBBP1, HBG1 and HBG2 genes (red dots) on the short arm of chromosome 11. All deletion were detected in the heterozygous form. The first deleted probe was the 486 on the OR51V1-1 gene (hg18 loc.11–005,177842), while the last was the 373, after the end of the HBG2 gene (hg18 loc.11–005,233895).

At last follow-up (5 years of age), the patient had a hemoglobin of 11.2 g/dL, a MCV of 52.9 fL, and a MCH of 17 pg; hemoglobin electrophoresis revealed 0.1% of HbF and 3.3% of HbA2 ( Figure 1 ). The patient was in good clinical condition, with normal growth (72 nd centile of height and 80 th centile of weight, WHO curves) and cognitive development. No splenomegaly was found at the abdominal ultrasound, nor signs of iron overload/deficiency. Therefore, no specific follow-up plan nor specific interventions in case of minor ailments were deemed necessary, as for β-trait carriers.

Anemia during the neonatal period represents a challenge for the pediatrician, mainly for the multiplicity of conditions that are responsible for the condition during the first weeks of life. The etiology of neonatal anemia usually falls into three major categories: blood loss, decreased production, and increased destruction of erythrocytes ( 28 ). The differential diagnosis for hemolytic anemia in the newborn period includes alloimmunity, erythrocyte membrane defects, enzyme deficiencies, and hemoglobinopathies. The most frequent hemoglobinopathy associated with critically ill infants and hemolytic anemia is α thalassemia with deletion of three α globin genes ( 28 , 29 ).

ε γδβ thalassemia usually presents as severe neonatal hemolytic anemia that requires in utero and/or neonatal transfusions but this condition is rarely considered among the causes of neonatal anemia and therefore misdiagnosed, as in our case. A reduced MCV without abnormalities on hemoglobin electrophoresis in a newborn is not always detected in ε γδβ thalassemia ( Table 1 ), but when it is found, it can orient toward the diagnosis. Despite the high incidence of thalassemias in Italy, the significant microcytosis in our patient was initially deemed secondary to iron deficiency, as the intercurrent sepsis misdirected high indirect bilirubin values as a sign of hemolysis.

Although uncommon during the neonatal period, microcytosis can occur secondary to iron deficiency following feto-maternal hemorrhage. However, in most cases, it is associated with thalassemia, also depending on the α thalassemia allele frequency, which varies in different populations ( 30 ). After the neonatal period, the hematologic phenotype of microcytosis associated with normal hemoglobin electrophoresis, which is typical of ε γδβ thalassemia, can be associated to or confused with α thalassemia, but also, in presence of normal ferritin levels, with IRIDA. Unlike previously suggested, the severe phenotype of our patient was not justified by the presence of α triplication, which was excluded by MLPA analysis.

There is no established explanation for the phenotypic heterogeneity of the disease, but it is not dependent on the type and length of deletion ( 8 ). Although at the molecular level ε γδβ thalassemias fall into two distinct categories, the associated phenotypes of the two groups are similar. Therefore, the variable severity is likely to be influenced by other genetic and environmental factors.

The remission of anemia after the first months of life is a consequence of the increasing production of β-globin that reduces the imbalance between α/non-α globin chain synthesis. The residual adult phenotype is similar to that of the β-thalassemia trait but with normal, rather than increased, levels of hemoglobin A2 due to the loss of one δ locus, while the fetal hemoglobin is normal or minimally increased ( 13 ). The normal HbA2 levels make the hematologic phenotype also similar to that of carriers of α-thalassemia ( 23 ). However, data collected by Rooks et al. suggest that adult heterozygotes for ε γδβ -thalassemias tend to have more severe microcytosis and hypochromia even than β 0 -thalassemia carriers ( 13 ).

In conclusion, this case remarks the importance of considering the ε γδβ thalassemia in the differential diagnosis of hypochromic microcytic hemolytic anemias in the newborn period. In the post-natal period, microcytosis with normal ferritin values and without abnormalities on hemoglobin electrophoresis should also raise the suspicion for ε γδβ thalassemia.

Data Availability Statement

The original contributions presented in the study are included in the article/supplementary material, further inquiries can be directed to the corresponding author.

Ethics Statement

Ethical review and approval was not required for the study on human participants in accordance with the local legislation and institutional requirements. Written informed consent to participate in this study was provided by the participants' legal guardian/next of kin. Written informed consent was obtained from the minor(s)' legal guardian/next of kin for the publication of any potentially identifiable images or data included in this article.

Author Contributions

IF and FP wrote the manuscript and MV and CF critically reviewed it. IF, EC, and TC followed the patient. MM performed the genetic analysis. All authors contributed to the article and approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

1. Repnikova E, Roberts J, Mc Dermott S, Farooqi MS, Iqbal NT, Silvey M, et al. Clinical and molecular characterization of novel deletions causing epsilon gamma delta beta thalassemia: report of two cases. Pathol Res Pract. (2019) 215:152578. doi: 10.1016/j.prp.2019.152578

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Kan YW, Forget BG, Nathan DG. Gamma-beta thalassemia: a cause of hemolytic disease of the newborn. N Engl J Med. (1972) 286:129–34. doi: 10.1056/NEJM197201202860304

3. Oort M, Heerspink W, Roos D, Flavell RA, Bernini LF. Hemolytic disease of the newborn and chronic hypochromic microcytic anemia in one family: gamma-delta-beta thalassemia. Tijdschr Kindergeneeskd. (1981) 49:199–207.

PubMed Abstract | Google Scholar

4. Fearon ER, Kazazian HH, Waber PG, Lee JI, Antonarakis SE, Orkin SH, et al. The entire beta-globin gene cluster is deleted in a form of gamma delta beta-thalassemia. Blood. (1983) 61:1269–74. doi: 10.1182/blood.V61.6.1269.1269

5. Curtin P, Pirastu M, Kan YW, Gobert-Jones JA, Stephens AD, Lehmann H, et al. distant gene deletion affects beta-globin gene function in an atypical gamma delta beta-thalassemia. J Clin Invest. (1985) 76:1554–8. doi: 10.1172/JCI112136

6. Diaz-Chico JC, Huang HJ, Juricić D, Efremov GD, Wadsworth LD, Huisman TH. Two new large deletions resulting in epsilon gamma delta beta-thalassemia. Acta Haematol. (1988) 80:79–84. doi: 10.1159/000205607

7. Driscoll MC, Dobkin CS, Alter BP. Gamma delta beta-thalassemia due to a de novo mutation deleting the 5' beta-globin gene activation-region hypersensitive sites. Proc Natl Acad Sci U S A. (1989) 86:7470–4. doi: 10.1073/pnas.86.19.7470

8. Trent RJ, Williams BG, Kearney A, Wilkinson T, Harris PC. Molecular and hematologic characterization of Scottish-Irish type (epsilon gamma delta beta)zero thalassemia. Blood. (1990) 76:2132–8. doi: 10.1182/blood.V76.10.2132.2132

9. Fortina P, Delgrosso K, Werner E, Haines K, Rappaport E, Schwartz E, et al. A greater than 200 kb deletion removing the entire beta-like globin gene cluster in a family of Irish descent. Hemoglobin. (1991) 15:23–41. doi: 10.3109/03630269109072482

10. Abels J, Michiels JJ, Giordano PC, Bernini LF, Baysal E, Smetanina NS, et al. A de novo deletion causing epsilon gamma delta beta-thalassemia in a Dutch patient. Acta Haematol. (1996) 96:108–9. doi: 10.1159/000203726

11. Game L, Bergounioux J, Close JP, Marzouka BE, Thein SL. A novel deletion causing (epsilon gamma delta beta) degrees thalassaemia in a Chilean family. Br J Haematol. (2003) 123:154–9. doi: 10.1046/j.1365-2141.2003.04564.x

12. Harteveld CL, Osborne CS, Peters M, van der Werf S, Plug R, Fraser P, et al. Novel 112 kb (epsilonGgammaAgamma) deltabeta-thalassaemia deletion in a Dutch family. Br J Haematol. (2003) 122:855–8. doi: 10.1046/j.1365-2141.2003.04505.x

13. Rooks H, Bergounioux J, Game L, Close JP, Osborne C, Best S, et al. Heterogeneity of the epsilon gamma delta beta-thalassaemias: characterization of three novel English deletions. Br J Haematol. (2005) 128:722–9. doi: 10.1111/j.1365-2141.2005.05368.x

14. Furuya C, Yamashiro Y, Hattori Y, Hino M, Nishioka H, Shimizu Y, et al. A novel epsilon gamma delta beta thalassemia of 1. 4 Mb deletion found in a Japanese patient. Am J Hematol. (2008) 83:84–6. doi: 10.1002/ajh.21040

15. Brantberg A, Eik-Nes SH, Roberts N, Fisher C, Wood WG. Severe intrauterine anemia: a new form of epsilongammagammadeltabeta thalassemia presenting in utero in a Norwegian family. Haematologica. (2009) 94:1157–9. doi: 10.3324/haematol.2009.007534

16. Rose C, Rossignol J, Lambilliotte A, Depret S, Le Metayer N, Pissard S, et al. novel (epsilongammadeltabeta)(o)-thalassemia deletion associated with an alpha globin gene triplication leading to a severe transfusion dependent fetal thalassemic syndrome. Haematologica. (2009) 94:593–4. doi: 10.3324/haematol.2008.002675

17. Verhovsek M, Shah NR, Wilcox I, Koenig SC, Barros T, Thornburg CD, et al. Severe fetal and neonatal hemolytic anemia due to a 198 kb deletion removing the complete β-globin gene cluster. Pediatr Blood Cancer. (2012) 59:941–4. doi: 10.1002/pbc.24094

18. Rooks H, Clark B, Best S, Rushton P, Oakley M, Thein OS, et al. A novel 506kb deletion causing ε γδβ thalassemia. Blood Cells Mol Dis. (2012) 49:121–7. doi: 10.1016/j.bcmd.2012.05.010

19. Von Kanel T, Röthlisberger B, Schanz U, Dutly F, Huber AR, Saller E, et al. Swiss (ε γδβ )°-thalassemia patient with a novel 3-Mb deletion associated with mild mental impairment. Am J Hematol. (2013) 88:158–9. doi: 10.1002/ajh.23364

20. Shalev H, Landau D, Pissard S, Krasnov T, Kapelushnik J, Gilad O, et al. A novel epsilon gamma delta beta thalassemia presenting with pregnancy complications and severe neonatal anemia. Eur J Haematol. (2013) 90:127–33. doi: 10.1111/ejh.12047

21. Zebisch A, Schulz E, Grosso M, Lombardo B, Acierno G, Sill H, et al. Identification of a novel variant of epsilon-gamma-delta-beta thalassemia highlights limitations of next generation sequencing. Am J Hematol. (2015) 90:E52–4. doi: 10.1002/ajh.23913

22. Shooter C, Rooks H, Thein SL. Barnaby Clark. Next generation sequencing identifies a novel rearrangement in the HBB cluster permitting to-the-base characterization. Hum Mutat. (2015) 36:142–50. doi: 10.1002/humu.22707

23. Cardiero G, Prezioso R, Dembech S, Del Vecchio Blanco F, Scarano C, Lacerra G. Identification and molecular characterization of a novel 163 kb deletion: The Italian (ϵ γδβ )(0)-thalassemia. Hematology. (2016) 21:317–24. doi: 10.1080/10245332.2015.1133007

24. Goel R, Snow J, Pri-Paz SM, Cushing M, Vasovic LV. Intrauterine transfusions for severe fetal anemia and hydrops due to de novo ε γδβ -thalassemia. Transfusion. (2017) 57:876. doi: 10.1111/trf.13926

25. Hui ASY, Au PKC, Ting YH, Kan ASY, Cheng YKY, Leung AWK, et al. First Report of a Novel Deletion Due to ε γδβ -Thalassemia in a Chinese Family. Hemoglobin. (2017) 41:175–9. doi: 10.1080/03630269.2017.1366918

26. Muñoz Tormo-Figueres Á, Sanchis Calvo A, Guibert Zafra B. Epsilon gamma delta beta thalassemia: A rare cause of fetal and neonatal anemia. Med Clin (Barc). (2018) 150:368–9. doi: 10.1016/j.medcli.2017.10.011

27. Pirastu M, Kan YW, Lin CC, Baine RM, Holbrook CT. Hemolytic disease of the newborn caused by a new deletion of the entire beta-globin cluster. J Clin Invest. (1983) 72:602–9. doi: 10.1172/JCI111008

28. Allali S, Brousse V, Sacri AS, Chalumeau M, de Montalembert M. Anemia in children: prevalence, causes, diagnostic work-up, and long-term consequences. Expert Rev Hematol. (2017) 10:1023–8. doi: 10.1080/17474086.2017.1354696

29. Cappellini MD, Russo R, Andolfo I, Iolascon A. Inherited microcytic anemias. Hematology Am Soc Hematol Educ Program. (2020) 2020:465–70. doi: 10.1182/hematology.2020000158

30. Piel FB, Weatherall DJ. The α-thalassemias. N Engl J Med. (2014) 371:1908–16. doi: 10.1056/NEJMra1404415

Keywords: thalassemia, ε γδβ , children, newborn, anemia

Citation: Fotzi I, Pegoraro F, Chiocca E, Casini T, Mogni M, Veltroni M and Favre C (2022) Case Report: Clinical and Hematological Characteristics of ε γδβ Thalassemia in an Italian Patient. Front. Pediatr. 10:839775. doi: 10.3389/fped.2022.839775

Received: 20 December 2021; Accepted: 07 February 2022; Published: 17 March 2022.

Reviewed by:

Copyright © 2022 Fotzi, Pegoraro, Chiocca, Casini, Mogni, Veltroni and Favre. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) . The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Claudio Favre, claudio.favre@meyer.it

† These authors share first authorship

Heterogeneity of TDT

The role of regular transfusions in hbe β thalassemia, red cell alloimmunization in patients starting transfusions as adults, postnatal management of bart hydrops fetalis: atm, correspondence, challenges in chronic transfusion for patients with thalassemia.

  • Split-Screen
  • Request Permissions
  • Cite Icon Cite
  • Search Site
  • Open the PDF for in another window

Ashutosh Lal; Challenges in chronic transfusion for patients with thalassemia. Hematology Am Soc Hematol Educ Program 2020; 2020 (1): 160–166. doi: https://doi.org/10.1182/hematology.2020000102

Download citation file:

  • Ris (Zotero)
  • Reference Manager

Visual Abstract

graphic

The introduction of regular red cell transfusions 60 years ago transformed β-thalassemia major from a fatal childhood illness into a chronic disorder. Further advances in the prevention of transfusion-transmitted infections and management of iron overload have allowed survival and quality of life to approach normal. However, transfusion therapy for some other thalassemia syndromes continues to challenge clinical decision-making. Nearly one-half of the patients with E ß thalassemia are transfusion-dependent, yet the criteria for initiating transfusions or hemoglobin targets are not well defined. Patients with thalassemia intermedia who begin transfusions as adults are at very high risk for developing red cell alloimmunization and serious hemolytic transfusion reactions. In the growing number of survivors of Bart hydrops fetalis, the approach to transfusion therapy and iron chelation is rapidly evolving. A collaboration between hematology and transfusion medicine specialists will be essential to improving patient care and developing evidence-based guidelines.

Recognize the heterogeneity of transfusion-dependent thalassemia and its impact on management

Understand barriers to the effective use of red cell transfusions in thalassemia intermedia, hemoglobin E beta thalassemia, and alpha thalassemia major.

In the past decade, the classification of patients into transfusion-dependent thalassemia (TDT) and non-transfusion-dependent thalassemia (NTDT) was widely adopted. These terms were beneficial in planning the management of iron overload or choosing stem cell transplant or other curative therapy based upon a patient’s transfusion status. However, this approach can conceal the tremendous heterogeneity of TDT, a phenotypic group that encompasses β-thalassemia major, severe β-thalassemia intermedia, hemoglobin (Hb) E (HbE) β thalassemia, and certain α-thalassemia syndromes. Among these, β-thalassemia major is the largest category and is usually associated with the presence of 2 severe β-globin mutations. 1   These infants become symptomatic from anemia within the first year and regular transfusions are instituted before 2 years of age. 2   The natural history of β-thalassemia major has been the best characterized among various entities constituting TDT, and, consequently, the transfusion guidelines recommended by various groups for β-thalassemia major are largely similar. 1,3-8  

The guidelines developed for β-thalassemia major may not be appropriate in managing the other thalassemia syndromes that require regular transfusions. The severity of β thalassemia is determined by the imbalance between the α and non-α globin chains. A reduced or complete absence of β-globin synthesis leads to accumulation of excess α-globin chains that are toxic to the erythroid precursors. 9,10   The surplus of α-globin chains can be mitigated when 1 or both β-thalassemia alleles are mild (β + or β ++ ), there is concurrent deletion of α-globin genes, or elevated synthesis of γ-globin persists. 9   Elevated γ-globin synthesis sometimes arises from hereditary persistence of fetal Hb, but milder increases are more often from quantitative trait loci in Xmn1-HBG2 , HMIP , and BCL11A . 11   Various forms of β thalassemia may also differ in the total endogenous Hb (F, A, or E) or the oxygen-affinity characteristics (A and E compared with F) 12   that modify the adaptation to anemia. 13   Individuals with severe forms of α thalassemia, in contrast, produce nonfunctional Hb (Hb Bart or HbH), which causes underestimation of the true severity of anemia. 14   In β-thalassemia intermedia and HbE β thalassemia, the decision to commence regular transfusions is influenced not only by the severity of symptoms, but also on medical judgement. The latter is a subjective assessment of whether long-term prognosis would be better by accepting symptomatic anemia without transfusions instead of transfusion dependence and the associated potential complications. The recognition that patients with NTDT have worse quality of life than those with TDT and are at risk for severe complications has led to the extension of chronic transfusions to a larger proportion of patients than in the past. 15-18  

HbE β thalassemia is caused by compound heterozygosity for the E mutation (HBB:c.79G>A) and a β-thalassemia mutation. 19   The prevalence of HbE β thalassemia follows the distribution of the E mutation, which reaches very high frequencies in southeast Asia, southern China, and south Asia. Immigration from Asia to the west has increased the awareness of this syndrome and its distinctive natural history compared with β-thalassemia syndromes (caused by 2 β-thalassemia mutations). 20   The severity of HbE β thalassemia ranges from a mild, asymptomatic anemia to the development of transfusion dependence from early life. 19   The E mutation activates a cryptic splice site that reduces synthesis of β E messenger RNA. 21   The variable decrease in β E output is 1 of the factors underlying the variable disease phenotype, even though HbE is a functional Hb. The severity of β mutation (β + instead of β 0 ), coinheritance of α-thalassemia trait, and genetic traits that increase γ-globin synthesis reduce the severity of HbE β thalassemia. 19   Why patients may have dissimilar physiological response to nearly identical Hb levels, and why erythropoietin response to anemia declines with age, is incompletely understood. 22   One characteristic that differentiates HbE β thalassemia from β thalassemia (intermedia or major) is the different functional properties of HbE and HbF. Patients with HbE β thalassemia compensate by rightward shift in the oxygen affinity, which is not seen in β thalassemia where the HbF is the predominant Hb. 12   Although clinical symptoms increase progressively with severity of anemia, it may not be possible to predict the likelihood of transfusion dependence based on Hb concentration alone.

The patient was diagnosed with HbE β 0 thalassemia based on results of newborn screening. She was asymptomatic in early childhood with no limitation of physical activity, mild facial skeletal changes, and normal growth. Her Hb concentration was maintained between 6.7 and 7.1 g/dL without any blood transfusion. At 7 years of age, the spleen started to enlarge from 3 to 6.5 cm along with a decline in height velocity. She started regular blood transfusion at 10 years of age that led to resumption of normal growth and a decrease in spleen size to 2 cm ( Figure 1 ).

Case 1 depiction. In a patient with HbE β thalassemia (case 1), progressive increase in spleen size and reduced growth velocity led to start of regular transfusions (arrow). Transfusions led to reduction in splenomegaly over the following 2 years.

Case 1 depiction. In a patient with HbE β thalassemia (case 1), progressive increase in spleen size and reduced growth velocity led to start of regular transfusions (arrow). Transfusions led to reduction in splenomegaly over the following 2 years.

The patient was diagnosed with E β 0 thalassemia at 3 years and started regular transfusions with iron chelation. He was splenectomized at 12 years due to higher blood requirements and splenomegaly, following which Hb was spontaneously maintained between 6.9 to 7.2 g/dL. He was transfused intermittently 1 to 2 times per year when Hb dropped below 7 g/dL. In his early 20s, he developed dyspnea and continuous oxygen requirement from severe pulmonary arterial hypertension and heart failure. He had severe iron overload with liver iron concentration of 38 mg/g and cardiac magnetic resonance imaging T2* of 4.9 ms. Heart failure and pulmonary hypertension improved with supportive management, regular transfusions, and iron chelation ( Figure 2 ).

Case 2 depiction. In a transfusion-dependent patient with HbE β thalassemia (case 2), splenectomy was followed by discontinuation of regular transfusions. Fifteen years later, severe pulmonary arterial hypertension and congestive heart failure developed. These pathological changes were reversed by resumption of transfusions (arrow) and supportive care. LVEF%, left ventricular ejection fraction (percent); PASP, pulmonary artery systolic pressure.

Case 2 depiction. In a transfusion-dependent patient with HbE β thalassemia (case 2), splenectomy was followed by discontinuation of regular transfusions. Fifteen years later, severe pulmonary arterial hypertension and congestive heart failure developed. These pathological changes were reversed by resumption of transfusions (arrow) and supportive care. LVEF%, left ventricular ejection fraction (percent); PASP, pulmonary artery systolic pressure.

As expected for β-globin disorders, newborns with HbE β thalassemia do not develop anemia until the synthesis of HbF declines significantly over the first 6 months of life. Some infants display the classic symptoms observed in β-thalassemia major, including failure to thrive, hepatosplenomegaly, pallor, and fatigue. 23   More often, the symptoms are mild and escape attention until an incidental viral infection or a routine blood test reveals anemia. In California, where universal newborn screening is practiced for thalassemia, observations suggest that normal growth and development persist up to 1 year and beyond in many, but not all, infants with HbE β thalassemia. When the diagnosis is made later during life due to pallor, anemia, facial skeletal changes, growth failure, hepatosplenomegaly, or jaundice, the age at presentation is an important marker of the severity of disease. 24  

Decisions on appropriate timing to either initiate or discontinue chronic transfusion therapy, although difficult, are of principal importance to the management of HbE β thalassemia. 22   Patients with baseline Hb <6 g/dL should be placed on transfusions even when asymptomatic. Conversely, it is unlikely that patients with baseline Hb >8 g/dL would benefit from transfusions. Finally, those with Hb between 6 and 8 g/dL should be evaluated according to the proposed guidelines ( Table 1 ). Our practice is to evaluate children with HbE β thalassemia in the clinic every 3 months for careful assessment of growth, splenomegaly, facial skeletal changes, and Hb level. Electronic medical records are useful to review trends over time and to store photographs for assessment of bony changes.

Indications to begin chronic transfusion therapy in HbE β thalassemia

In our practice, splenectomy is discouraged as a strategy to avoid the need for transfusions because the effect on Hb may be short-term and it does not address the underlying severe pathophysiology. 22,25   On the contrary, splenectomy increases the risk of infections and a number of serious long-term complications such as thromboembolism, pulmonary hypertension, and iron-induced endocrinopathies. 25   Other avenues to improve anemia include hydroxyurea, which produces an ∼1 g/dL rise in total Hb in 50% of patients, 26-28   however, the response in HbE β thalassemia is variable and insufficient to eliminate the need for transfusions. 18   Luspatercept is an approved therapy to reduce the transfusion requirements in patients with TDT including HbE β thalassemia. 29   Results of a phase 2, open-label study of luspatercept in NTDT showed ≥1.5 g/dL improvement in Hb in 14 of 31 subjects, 30   but the pivotal trial has not yet been done. Mitapivat, a pyruvate kinase activator in the red cells, is also being evaluated in NTDT. 31  

Case 2 highlights the underrecognition of iron overload in patients with NTDT. 18,32   Patients who receive only intermittent transfusions can develop marked iron overload over time. Gastrointestinal absorption of iron is increased in nontransfused patients due to hepcidin insufficiency induced by ineffective erythropoiesis and elevated erythroferrone. 33-35   Screening for iron overload is recommended in all patients with NTDT irrespective of transfusion history. Although magnetic resonance imaging for assessment of liver iron concentration is more accurate, 36   we consider serum ferritin to be useful as a screening test for iron overload. Serum ferritin values underestimate the liver iron overload in NTDT compared with TDT, therefore, LIC should be measured when ferritin exceeds 300 ng/mL. Iron chelation is indicated if LIC is >5 mg/g liver weight 36   with deferasirox at a starting dose of 3.5 to 7 mg/kg per day (5-10 mg/kg if the dispersible tablet is used). 37   We evaluate liver iron concentration every 6 months during therapy and stop chelation therapy when LIC <3 mg/g is achieved.

The development of antibodies to red cell antigens is a significant threat to the long-term success of transfusion therapy. 38-43   In the United States, the proportion of patients with thalassemia with red cell alloimmunization is 17% to 22%, only slightly lower than sickle cell disease (19% to 31%). 38,44,45   Older age at initiation of transfusions and splenectomy are identified as major risk factors for developing alloimmunization in thalassemia. 38,41,46   The specificity of antibody and the risk of alloimmunization is influenced by the disparity in red cell antigens among different ethnic groups. 47   In countries where thalassemia predominantly affects immigrant communities, the greater degree of donor-recipient red cell antigen mismatch can elevate the risk of alloimmunization, as shown in Table 2 . 39,44,46,48-50   In particular, the low frequency of Kell and c antigens in patients of Asian background facilitates development of alloimmunization. 39,46   One-half of the patients with 1 alloantibody will develop further antibodies against 1 or more additional antigens. 44   Transfusions can become progressively more difficult and it may become nearly impossible to find matched units for certain patients. The most frequent antibodies are against Rh and Kell groups, 41,44   which implies that universal phenotypic matching for these antigens can be a cost-effective method to prevent development of most red cell antibodies in thalassemia. 41,46  

RBC antigen frequencies and prevalence of alloantibodies

RBC antigen frequencies among ethnic groups 48-50   and prevalence of significant alloantibodies among patients with TDT in the western region of the United States, 39,46   and North America and United Kingdom. 38  

pts, patients.

Fy a or Fy b .

†Jk a or Jk b .

A 50-year-old woman with β-thalassemia intermedia who had undergone splenectomy recently changed hospitals. She was diagnosed at the age of 30 years but was only transfused during pregnancy. Due to worsening fatigue, regular transfusions were recommended by her new hematology team. A red cell phenotype was checked, and an antibody screen was found to be negative. She was given 2 red cell units matched to c, E, and Kell antigens. Later that night, she developed chills and fever. Her Hb levels were 7.1 and 9.0 g/dL, respectively, before and after the transfusion. Laboratory testing showed that her lactate dehydrogenase was normal, bilirubin rose from 1.2 to 1.7 mg/dL, haptoglobin was low, and urine contained trace Hb. Two weeks later, the antibody screen was positive and anti-Fyb was identified. Historical antibody data obtained from the previous blood bank showed anti-c, anti-E, anti-Kell, and anti-Fyb antibodies had been previously identified 15 years ago. The patient was successfully maintained on regular transfusions with extended phenotypically matched red blood cells. Anti-Fyb could no longer be identified after 4 years, although her antibody screen was intermittently positive due to development of anti-Cw antibody ( Figure 3 ).

Case 3 depiction. In a patient with β-thalassemia intermedia with prior exposure to blood and negative antibody screen (case 3), the transfusion of red cell units matched only to Rh and Kell antigens led to the reemergence of anti-Fyb antibody and delayed hemolytic transfusion reaction. Regular transfusions were possible with extended phenotypic matching of red cell units. Anti-Fyb was no longer detectable after 4 years, though antibody screen remained positive intermittently due to other alloantibodies. IAT, indirect antiglobulin test.

Case 3 depiction. In a patient with β-thalassemia intermedia with prior exposure to blood and negative antibody screen (case 3), the transfusion of red cell units matched only to Rh and Kell antigens led to the reemergence of anti-Fyb antibody and delayed hemolytic transfusion reaction. Regular transfusions were possible with extended phenotypic matching of red cell units. Anti-Fyb was no longer detectable after 4 years, though antibody screen remained positive intermittently due to other alloantibodies. IAT, indirect antiglobulin test.

Although considered a much greater risk in sickle cell disease, 51   delayed hemolytic transfusion reactions (DHTRs) are also observed in thalassemia. 52   Hemolytic transfusion reactions have been reported due to anti-E, anti-Jk b , anti-Jk a , anti-c, anti-S, anti-Kell, and anti-f. 39   More than 25% of older children and adults with thalassemia will develop an alloantibody following 1 or more transfusions when red cell matching is limited to ABO/D only. 41,44,46   In the absence of further antigenic exposure, one-third of alloantibodies become undetectable within the first year of follow up. Anti-Jka antibodies are very evanescent, falling below the limit of detection within the first month of initial detection, whereas the anti-Kell and anti-E antibodies are undetectable in >50% at 6 months. 53,54   Between 20% and 25% of individuals of Chinese and Asian Indian ethnicity are Jka − , 60% to 80% are E − , and virtually all are Kell − . 48,55   An additional concern is c antigen which is negative in 40% to 50% of Asian patients, but the evanescence rate for anti-c antibody is lower (25%) compared with the other alloantibodies. 39  

Sensitized patients who receive a later transfusion, based on a negative antibody screen, rapidly increase antibody titer with reexposure that develops into a DHTR of variable severity. 51   DHTRs are less likely in regularly transfused patients where antibody screening is performed every few weeks and newly developed antibodies are unlikely to become undetectable before the next transfusion. 56   However, low-titer antibodies may be missed, or antibodies may develop while donor red cells are still circulating in significant amounts, leading to hemolysis. Intermittently transfused patients, on the other hand, are at high risk for antibody evanescence between transfusion episodes. The number of such patients with thalassemia, transfused 1 to 6 times per year, is small in the United States but significant in regions where blood availability is limited. Infrequent transfusions may be recommended during an infection, surgery, or pregnancy. 25   Transfusions during pregnancy are associated with a very high risk of alloimmunization, but these antibodies decrease in titer when transfusions are discontinued after childbirth. 57   Patient transfer between hospitals poses a persistent challenge to transfusion safety. 58   The lack of comprehensive antibody history can easily lead to the transfusion of red cells to which the patient is previously sensitized but now has a negative antibody screen. Although attempts have been made to provide patients with transfusion cards that list their phenotype and red cell antibodies, their use remains erratic. The absence of a centralized database for multiply transfused patients remains a serious shortcoming of the current practice of chronic transfusion therapy. 59  

The deletion of all 4 α-globin genes (--/--, homozygous α 0 thalassemia, or α-thalassemia major [ATM]) causes severe fetal anemia. 14,60   As fetal viability depends upon the preservation of embryonic ζ genes (ζ-/ζ-, or --/ζ-), the most frequent α 0 deletion associated with ATM is the southeast Asian deletion (-- SEA ). 14   The major Hb species in ATM is Hb Bart (γ 4 ), formed by self-association of γ chains into tetramers in the absence of α chains. Because Hb Bart is ineffective in transporting oxygen, most pregnancies end in fetal demise following a variable period of hydrops (Bart hydrops fetalis). 60   Intrauterine transfusions (IUTs) are essential for the fetus to reach viability with acceptable neonatal outcome. 61,62   All infants with ATM are transfusion-dependent from birth and require recognition of the nonfunctional Hb fractions for correct management. 62,63  

A child was born to parents who were carriers of the -- SEA deletion and had previously experienced a hydrops-associated stillbirth. During this pregnancy, fetal hydrops was detected at 20 weeks of gestation, and managed by IUT performed on 6 occasions. The baby, born at 37 weeks weighing 3.0 kg, was stable and underwent a red blood cell exchange transfusion at 48 hours. Following discharge, he started regular red cell transfusions every 4 weeks. The average pretransfusion Hb was 9.7 g/dL with Hb Bart and HbH accounting for 20% to 36% of the total value. Starting at 8 months, the transfusion regimen was changed to maintain HbH <20%, which corresponded to HbA >9.0 g/dL and total Hb of ∼11 g/dL in the pretransfusion blood sample. Growth proceeded at the normal pace, and developmental assessment at 3 years showed age-appropriate attainment of milestones ( Figure 4 ).

Case 4 depiction. A newborn with α-thalassemia major (−SEA/−SEA) was initially transfused with a goal of maintaining total Hb of 9 to 10 g/dL in the pretransfusion period (case 4). This was associated with effective functional Hb (total Hb − sum of HbH and Hb Bart) between 6 and 7 g/dL. Transfusion regimen was modified (arrow) to maintain HbA 9 to 10 g/dL, which was achieved with total Hb of ∼11 g/dL in the pretransfusion blood sample.

Case 4 depiction. A newborn with α-thalassemia major (− SEA /− SEA ) was initially transfused with a goal of maintaining total Hb of 9 to 10 g/dL in the pretransfusion period (case 4). This was associated with effective functional Hb (total Hb − sum of HbH and Hb Bart) between 6 and 7 g/dL. Transfusion regimen was modified (arrow) to maintain HbA 9 to 10 g/dL, which was achieved with total Hb of ∼11 g/dL in the pretransfusion blood sample.

The management of ATM is evolving with experience gained from data in international registries 61   and publication of case series. 62-65   In the absence of existing consensus guidelines, our institutional practices are provided in this discussion to fill gaps in published literature. Questions about the optimal management of pregnancies affected by ATM are being evaluated in an ongoing clinical trial (NCT02986698, fetus.ucsf.edu ).

The hematological management of ATM commences as soon as the diagnosis is suspected during pregnancy. In that small proportion of cases in which both parents are known carriers of the α 0 -thalassemia trait, the diagnosis should be established expeditiously with DNA testing from chorionic villus biopsy instead of ultrasound surveillance for fetal changes suggestive of hydrops. In most cases, however, the first indication is the detection of hydrops on ultrasound during the second trimester. In such cases, a presumptive diagnosis of ATM is appropriate when severe fetal anemia is observed by Doppler ultrasonography, 66   and the pregnant woman is not alloimmunized to RhD or other red cell antigens but has microcytosis and hypochromia. Hematologists have a critical role to play in confirmation of the diagnosis, nondirective counseling of the family, and prenatal management of the fetus.

When the decision is made to continue the pregnancy, the first IUT should be initiated as soon as possible. The strategy for conducting IUT in ATM is derived from consensus guidelines from the Society for Maternal-Fetal Medicine. 67   The decision to proceed with fetal blood sampling and IUT should be based on the detection of severe fetal anemia (defined as elevated peak systolic velocity in the fetal middle cerebral artery on Doppler ultrasonography 68   ) irrespective of the presence of hydrops. 67   Because Hb Bart, which constitutes nearly all the Hb in ATM, does not participate in oxygen transport, fetal hypoxia is disproportionate to any given Hb level. 69   The goals of complete correction of anemia or suppression of fetal erythropoiesis must be balanced against the risk of acute cardiovascular alterations and hyperviscosity with IUT. 67   Fetal anemia develops early in ATM with a mean Hb of 6.8 g/dL at 18 weeks, 69   which implies that there could be a role for intraperitoneal IUT prior to 18 weeks of gestation when intravascular IUT using the umbilical vein becomes feasible.

Resolution of hydrops is expected with an adequate IUT regimen. Specific to the diagnosis of ATM is the recommendation to measure Hb Bart in addition to total Hb in the pretransfusion sample. Following the initial gradual correction of anemia, the target hematocrit following transfusion after 24 weeks of gestation should be chosen at the higher end of the recommended range (40% to 50%). 67   An HbA level >10 g/dL and Hb Bart <20% in the pretransfusion fetal blood sample is expected with these goals. The delivery is planned at 37 to 38 weeks with the last IUT no later than 35 weeks of gestation. 67  

Perinatal events in ATM following suboptimal fetal management are characterized by a high incidence of preterm birth, intrauterine growth retardation, cesarean delivery, birth trauma, and difficult resuscitation. Newborns exhibit respiratory distress, pulmonary hypertension, organomegaly, effusions, and hyperbilirubinemia. 61,62   Some may have congenital anomalies affecting the genitourinary system. Anemia is profound and further distinguished by a high proportion of nonfunctional Hb Bart if >2 to 3 weeks have elapsed from the last intrauterine transfusion. An urgent simple transfusion with 5 to 10 mL/kg of high hematocrit red cell unit is usually given. When the proportion of Hb Bart is very high, an exchange transfusion will rapidly improve tissue oxygenation. The goal during the first few weeks is to maintain total Hb >12 g/dL and Hb Bart <20% while the need for critical care continues.

Following stabilization, infants are in a transition period up to 6 months with intensive transfusion support under close monitoring. This period is marked by the switch from Hb Bart to HbH, resolution of hepatosplenomegaly and cardiomegaly, improvement in thrombocytopenia and transaminitis, and the establishment of consistent weight gain. Transfusions aim to maintain nadir total Hb >12 g/dL with the total nonfunctional Hb (Hb Bart plus HbH) <20%. The interval between transfusions is initially 2 weeks, but gradually lengthened to 3 weeks. Red cell antigens should be determined by genetic testing to provide antigen-matched blood. At the end of 6 months, infants transition to a chronic transfusion protocol in which pretransfusion HbA is >9.0 g/dL and transfusion frequency is 3 to 4 weeks. Following the absolute HbA level instead of total Hb is important, otherwise children with ATM are at risk for undertransfusion. 63   Infusion centers lacking access to rapid Hb electrophoresis or high-performance liquid chromatography can aim to maintain the pretransfusion total Hb at 10.5 to 11 g/dL and reticulocyte count <500 000/μL. 63   Splenectomy is not recommended in the management of ATM. Transfusional iron overload is observed early within a few months after birth, but the assessment and management of iron in ATM is not well defined. Because of the concerns over hepatic inflammation and renal immaturity, chelation is postponed until 12 months of age.

The management of TDT should be adapted to the heterogeneity conferred by various genotypes. Although a universal transfusion protocol for TDT is unfeasible, common principles underlie the long-term goals of transfusion therapy for individuals with thalassemia. With improvement in life expectancy, decisions about initiation and intensity of transfusion support in TDT should be guided by long-term natural history studies that span the various life stages.

Ashutosh Lal, UCSF Benioff Children’s Hospital, 747 52nd St, Oakland, CA 94609; e-mail: [email protected] .

Competing Interests

Conflict-of interest disclosure: A.L. provided consultancy services to Chiesi USA and received research funding from Bluebird bio, Insight Magnetics, La Jolla Pharmaceutical Company, Novartis, Protagonist Therapeutics, and Terumo Corporation.

Author notes

Off-label drug use: None disclosed.

  • Previous Article
  • Next Article

Email alerts

Affiliations, american society of hematology.

  • 2021 L Street NW, Suite 900
  • Washington, DC 20036
  • TEL +1 202-776-0544
  • FAX +1 202-776-0545

ASH Publications

  • Blood Advances
  • Blood Neoplasia
  • Blood Vessels, Thrombosis & Hemostasis
  • Hematology, ASH Education Program
  • ASH Clinical News
  • The Hematologist
  • Publications
  • Privacy Policy
  • Cookie Policy
  • Terms of Use

This Feature Is Available To Subscribers Only

Sign In or Create an Account

Case Report: β-thalassemia major on the East African coast

Alexander W. Macharia Roles: Conceptualization, Data Curation, Formal Analysis, Investigation, Methodology, Visualization, Writing – Original Draft Preparation, Writing – Review & Editing George Mochamah Roles: Conceptualization, Formal Analysis, Investigation, Writing – Review & Editing Johnstone Makale Roles: Data Curation, Formal Analysis, Methodology, Project Administration Thad Howard Roles: Data Curation, Formal Analysis, Investigation, Methodology Neema Mturi Roles: Investigation, Methodology, Resources Peter Olupot-Olupot Roles: Validation, Writing – Review & Editing Anna Färnert Roles: Validation, Writing – Review & Editing Russell E. Ware Roles: Methodology, Visualization, Writing – Review & Editing Thomas N. Williams Roles: Funding Acquisition, Methodology, Supervision, Visualization, Writing – Review & Editing

β-thalassemia major, rs33941849, East Africa, HbA2, sequencing

Introduction

β-thalassemia is rare in most of Africa, with the exception of North Africa where the prevalence, causal pathogenic variants and disease outcomes have all been well described previously ( Hamamy & Al-Allawi, 2013 ). We recently reported elevated levels of HbA 2, suggestive of β-thalassemia, in a small proportion of children participating in a cohort study conducted in Kilifi county on the coast of Kenya ( Macharia et al ., 2019 ). We subsequently sequenced samples from the same children and found that 0.6% were carriers of one of four different β-thalassemia pathogenic variants: the β 0 -thalassemia variants CD22 (GAA➝TAA) (rs33959855), initiation codon (ATG➝ACG) (rs33941849) and IVS1-3ʹ end del 25bp (rs193922563) and the β + -thalassemia variant IVS-I-110 (G➝A) (rs35004220). Whereas the mutations observed in North Africa resemble those found in Middle Eastern countries, those identified in Kilifi were a mixture of mutations reported from Asia and the Middle East. To the best of our knowledge, no cases of β-thalassemia major – a condition in which both HBB genes are affected by a β 0 -thalassemia mutation to result in the complete loss of normal β 0 -globin production - have yet been reported from the East Africa region. Here, we describe what is, to the best of our knowledge, the first case of β 0 -thalassemia major to be recognised from within this region.

Written informed consent was provided by the parents of the study participant. Ethical approval for the study was granted by the Kenya Medical Research Institute Ethical Review Committee in Nairobi, Kenya (Number: SCC3891).

Patient report

The child, a two-and-a-half-year-old female, presented to Kilifi County Hospital in Kenya, with a one-week history of left sided abdominal swelling. No previous hospital admissions were reported. Clinical history suggested delayed developmental milestones; specifically, she was unable to walk without support. The child was the fourth born of five siblings, all of whom were alive and well as were both of her parents. Both her parents were of Mijikenda ethnolinguistic ancestry and no recent genetic admixture was apparent from the clinical history. On physical examination, the child was pale but had no signs of clinical jaundice. Her vital signs were essentially normal with the exception of a fever measured at 38.8 ° C per axilla. Fronto-maxillary skull bossing was apparent. Her abdomen was distended, soft and non-tender, massive splenomegaly being detected at 8cm below the costal margin. She was severely malnourished with a weight of 8.8 kg, a height of 78.5 cm, a height for age z-score (HAZ) of -3.79, a weight for age z-score (WAZ) of -3.20 and a weight for height z-score (WHZ) of -1.23. Further examination was essentially normal. The timeline of events is given in Table 1 .

Table 1. Timeline of events.

A full hemogram revealed marked anemia (Hb 6.6 g/dL), a low mean corpuscular volume (MCV) of 64 fL, a low mean corpuscular hemoglobin (MCH) of 19.4 pg, and a raised total white blood cell (WBC) count of 49.6 × 10 9 /µl which were predominantly lymphocytes. Her platelet count was normal at 321 × 10 6 /L and her creatinine mildly elevated at 32 μmol/l. Blood cultures and tests for malaria were negative. A peripheral blood film revealed nucleated red blood cells (RBCs), microcytes, dacrocytes, acanthocytes, giant platelets and a marked lymphocytosis ( Table 2 ).

Table 2. Complete blood count and peripheral blood film from the child with β-thalassemia.

Abbreviations: WBC, white blood cells; RBC, red blood cells; Hb, hemoglobin; HCT, hematocrit; MCV, mean cell volume; MCH, mean cell hemoglobin; PBF, peripheral blood film. ¢Age=2.5 years, ‡Age=3.5 years.

The child was admitted to the general pediatric ward with a working diagnosis of iron deficiency anemia, potentially complicated by bacterial sepsis, and with a differential diagnosis of sickle cell anemia. She was treated empirically with iron and folic acid supplementation for her anemia and with intravenous penicillin and gentamicin to cover sepsis. She was also prescribed malaria prophylaxis with proguanil pending analysis for sickle cell anemia by high-performance liquid chromatography (HPLC). Her fever subsided within two days of admission, at which point she was discharged home on oral amoxicillin, with follow-up planned for the following week.

The results of her HPLC analysis, received after discharge from hospital, revealed the absence of normal adult hemoglobin (HbA), normal levels of HbA 2 at 2.5% and elevated levels of fetal hemoglobin (HbF) (>80% of total Hb) that eluted in adjacent peaks A1b (16%) and LA1C/cHb1 (76.5%) ( Figure 1 ). The complete absence of HbA suggested a diagnosis of β 0 -thalassemia major. We therefore sequenced her HBB gene region as described in detail previously ( Clark & Thein, 2004 ), which revealed that the child was homozygous for the initiation codon (ATG➝ACG) mutation (rs33941849).

Figure 1. HPLC chromatograms from study participants with normal hemoglobin A individual (HbAA), homozygous hemoglobin S (HbSS) and homozygous β-thalassemia patient at first admission (age 2.5 years) and at second admission (age 3.5 years).

Initially lost to follow-up, the child re-presented at the age of three years 11 months with a one-week history of a cough and fever. On examination at that time, her spleen remained grossly enlarged at 10 cm, and she remained malnourished with a HAZ of -4.98, a WAZ of -4.01 and a WHZ of -0.99. Although hemodynamically stable, she was profoundly anemic (Hb 2.2 g/dL) and was therefore transfused and treated with folic acid supplementation and nutritional support. Repeat HPLC analysis revealed the continued absence of HbA together with elevated levels of HbF (>80%) and HbA 2 (at 5%) ( Figure 1 ). PCR for the α -3.7 deletional form of α-thalassemia was negative.

To the best of our knowledge, this is the first case of homozygous β-thalassemia to be reported from the East Africa region. The mutation responsible disrupts the transfer RNA binding site to result in a β 0 form of the disease. It appears to be rare in other populations: only 45 carriers have been reported in the literature to date, 20 of which were from our recently reported study ( Macharia et al ., 2020 ). Other reports of carriers have come from a wide range of countries including Switzerland ( Beris et al ., 1993 ), Belgium ( Wildmann et al ., 1993 ), Russia ( Molchanova et al ., 1998 ), India ( Gorakshakar et al ., 2018 ; Gupta et al ., 2002 ) and the former Yugoslavia ( Jankovic et al ., 1990 ). The only homozygous case described to date was a male child of Pakistani origin who presented at 10 months of age with a palpable liver and spleen at 7 cm and 3 cm below costal margin, respectively. His Hb was 9.2 g/dl, MCV of 73 fl and MCH of 33 pg. He was also found to be homozygous for the α -3.7 -thalassemia deletion and to have a Bantu β-globin gene cluster haplotype. He was managed with regular blood transfusions ( Khan et al ., 2000 ).

On comparing the current and previously described cases, all had anemia, a low MCV and massive splenomegaly. In our current patient, we also observed elevated levels of HbF and varying levels of HbA 2 at the two points of testing, an observation which is common in β-thalassemia major ( Steinberg & Rodgers, 2015 ). Options for the treatment of this condition in our context are limited. Throughout much of the world, first line management includes the provision of regular, leuco-depleted blood transfusions together with extended antigen typing of transfused blood to reduce the risk of alloimmunization. Iron-chelation is also used to mitigate the risk of iron overload ( Steinberg et al ., 2009 ) while more recently, allogeneic hematopoietic cell transplantation (HCT) is also being used as a potentially curative therapy. However, all these strategies are beyond the capacity of our local health-care system. Nevertheless, there is growing evidence to support the use of hydroxyurea, an HbF inducer, in the treatment of transfusion and non-transfusion dependent β-thalassemia ( Algiraigri et al ., 2017a ; Algiraigri et al ., 2017b ). We will investigate this strategy together with surgical splenectomy if the child returns for further follow-up in the hope that these will reduce the frequency at which transfusions will be required.

Conclusions

We have previously estimated the birth prevalence of β-thalassemia major in our local community at approximately 1 in 100,000 ( Macharia et al ., 2020 ). Nevertheless, low awareness of this condition among clinicians and the low availability of diagnostic facilities within the region mean that historically, individuals with β-thalassemia major have probably been misdiagnosed with other conditions such as sickle cell anemia or iron deficiency anemia as was the case with this child. As such, we hope that our case study will raise awareness about the existence and clinical importance of β-thalassemia major as a public health problem within the East Africa region and lead to the development of locally appropriate diagnostic and treatment guidelines.

Data availability

Underlying data.

All data underlying the results are available as part of the article and no additional source data are required.

Written informed consent for publication of their clinical details was obtained from the parents of the patient.

  •   Algiraigri AH, Wright NAM, Paolucci EO, et al. : Hydroxyurea for lifelong transfusion-dependent β-thalassemia: A meta-analysis. Pediatr Hematol Oncol. 2017a; 34 (8): 435–448. PubMed Abstract | Publisher Full Text
  •   Algiraigri AH, Wright NAM, Paolucci EO, et al. : Hydroxyurea for nontransfusion-dependent β-thalassemia: A systematic review and meta-analysis. Hematol Oncol Stem Cell Ther. 2017b; 10 (3): 116–125. PubMed Abstract | Publisher Full Text
  •   Beris P, Darbellay R, Speiser D, et al. : De novo initiation codon mutation (ATG-->ACG) of the beta-globin gene causing beta-thalassemia in a Swiss family. Am J Hematol. 1993; 42 (3): 248–253. PubMed Abstract | Publisher Full Text
  •   Clark BE, Thein SL: Molecular diagnosis of haemoglobin disorders. Clin Lab Haematol. 2004; 26 (3): 159–176. PubMed Abstract | Publisher Full Text
  •   Gorakshakar AC, Breganza PV, Colaco SP, et al. : Rare β- and δ-Globin Gene Mutations in the Pathare Prabhus: Original Inhabitants of Mumbai, India. Hemoglobin. 2018; 42 (5–6): 297–301. PubMed Abstract | Publisher Full Text
  •   Gupta A, Hattori Y, Agarwal S: Initiation codon mutation in an Asian Indian family. Am J Hematol. 2002; 71 (2): 134–136. PubMed Abstract | Publisher Full Text
  •   Hamamy HA, Al-Allawi NA: Epidemiological profile of common haemoglobinopathies in Arab countries. J Community Genet. 2013; 4 (2): 147–167. PubMed Abstract | Publisher Full Text | Free Full Text
  •   Jankovic L, Efremov GD, Josifovska O, et al. : An initiation codon mutation as a cause of a beta-thalassemia. Hemoglobin. 1990; 14 (2): 169–176. PubMed Abstract | Publisher Full Text
  •   Khan SN, Riazuddin S, Galanello R: Identification of three rare beta-thalassemia mutations in the Pakistani population. Hemoglobin. 2000; 24 (1): 15–22. PubMed Abstract | Publisher Full Text
  •   Macharia AW, Mochamah G, Uyoga S, et al. : β-Thalassemia pathogenic variants in a cohort of children from the East African coast. Mol Genet Genomic Med. 2020; 8 (7): e1294. PubMed Abstract | Publisher Full Text | Free Full Text
  •   Macharia AW, Uyoga S, Ndila C, et al. : The population dynamics of hemoglobins A, A 2 , F and S in the context of the hemoglobinopathies HbS and α-thalassemia in Kenyan infants. Haematologica. 2019; 104 (5): e184–e186. PubMed Abstract | Publisher Full Text | Free Full Text
  •   Molchanova TP, Postnikov Yu V, Gu LH, et al. : Historical note: the beta-thalassemia allele in the noble Russian family Lermontov is identified as the ATG-->ACG change in the initiation codon. Hemoglobin. 1998; 22 (3): 283–286. PubMed Abstract | Publisher Full Text
  •   Steinberg MH, Forget BG, Higgs DR, et al. : Disorders of Hemoglobin: Genetics Pathophysiology, and Clinical Management. 2nd edn. Cambridge University Press, Cambridge. 2009. Reference Source
  •   Steinberg MH, Rodgers GP: HbA 2 : biology, clinical relevance and a possible target for ameliorating sickle cell disease. Br J Haematol. 2015; 170 (6): 781–787. PubMed Abstract | Publisher Full Text
  •   Wildmann C, Larondelle Y, Vaerman JL, et al. : An initiation codon mutation as a cause of beta-thalassemia in a Belgian family. Hemoglobin. 1993; 17 (1): 19–30. PubMed Abstract | Publisher Full Text

Comments on this article Comments (0)

Open peer review.

Is the background of the case’s history and progression described in sufficient detail?

Are enough details provided of any physical examination and diagnostic tests, treatment given and outcomes?

Is sufficient discussion included of the importance of the findings and their relevance to future understanding of disease processes, diagnosis or treatment?

Is the case presented with sufficient detail to be useful for other practitioners?

Competing Interests: No competing interests were disclosed.

Reviewer Expertise: Hemoglobinopathies and thalassemia

  • Respond or Comment
  • COMMENT ON THIS REPORT

Reviewer Expertise: disorders of hemoglobin

Reviewer Status

Alongside their report, reviewers assign a status to the article:

Reviewer Reports

  • Martin H. Steinberg , Boston Medical Center, Boston, USA
  • Paloma Ropero , Hospital Clínico San Carlos, Madrid, Spain

Comments on this article

All Comments (0)

Competing Interests Policy

Provide sufficient details of any financial or non-financial competing interests to enable users to assess whether your comments might lead a reasonable person to question your impartiality. Consider the following examples, but note that this is not an exhaustive list:

  • Within the past 4 years, you have held joint grants, published or collaborated with any of the authors of the selected paper.
  • You have a close personal relationship (e.g. parent, spouse, sibling, or domestic partner) with any of the authors.
  • You are a close professional associate of any of the authors (e.g. scientific mentor, recent student).
  • You work at the same institute as any of the authors.
  • You hope/expect to benefit (e.g. favour or employment) as a result of your submission.
  • You are an Editor for the journal in which the article is published.
  • You expect to receive, or in the past 4 years have received, any of the following from any commercial organisation that may gain financially from your submission: a salary, fees, funding, reimbursements.
  • You expect to receive, or in the past 4 years have received, shared grant support or other funding with any of the authors.
  • You hold, or are currently applying for, any patents or significant stocks/shares relating to the subject matter of the paper you are commenting on.

Stay Updated

Sign up for content alerts and receive a weekly or monthly email with all newly published articles

Register with Wellcome Open Research

Already registered? Sign in

Not now, thanks

Are you a Wellcome-funded researcher?

If you are a previous or current Wellcome grant holder, sign up for information about developments, publishing and publications from Wellcome Open Research.

We'll keep you updated on any major new updates to Wellcome Open Research

The email address should be the one you originally registered with F1000.

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here .

If you still need help with your Google account password, please click here .

You registered with F1000 via Facebook, so we cannot reset your password.

If you still need help with your Facebook account password, please click here .

If your email address is registered with us, we will email you instructions to reset your password.

If you think you should have received this email but it has not arrived, please check your spam filters and/or contact for further assistance.

  • OSCE Slideshow
  • Observed OSCE
  • Experts speak
  • Preparation
  • Last minute
  • In the exam hall
  • Things to remember
  • B. OSCE Slideshow
  • C. Case Presentation
  • D. OSCE By Topics
  • Experts Speak

You are here:

  • Case Presentation

Case presentation : Thalassemia

9 September 2011, 20:38

Dr. Tushar Maniar

By Dr. ADITI   SHAH

This is a common case that is asked in the exam. We have put the relevent points to be covered while taking the case . Also, some of the Q &A are mentioned. Your feedback is welcome.

Case : THALASSEMIA

  • Consanguinity
  • Was admitted for receiving blood transfusion
  • Onset of noticing pallor —- months of age
  • Investigated and found to have an abnormal blood disorder
  • Time of 1 st transfusion
  • Frequency of transfusion
  • Any increase in frequency now
  • How many bags of blood at present… so calculate his trans requirement in ml/kg/year
  • Where receives trans?… charitable blood bank? Hospital? General ward bed ( to highlight financial constraints
  • h/o rash or fever during trans
  • when was chelation started
  • what drug… how is it taken, how much dose, how many times a week
  • any other treatment (ca, folic acid, antifailure drugs, hormonal supplements etc)
  • registered with thal society?
  • Advised for inv every 3 months
  • any special vaccines( if taken mention here or mention in immunization history)
  • any special inv done like MRI, bone scan
  • if advised surgery ( splenectomy), mention here

NEGATIVE HISTORY

  • H/O not gaining adequate ht, Secondary sexual characters, Recent increase in frequency of transfusion , with easy fatigability, easy bruisability, repeated infections and fever, Abdominal distension, Bone pain/ joint pain ( osteopenia, osteoporosis, AVN head of femur), Change in facial profile with prominent bones ( compli of disease itself)
  • h/o fatigue, swelling legs, palpitation ( cardiac iron overload)
  • h/o jaundice, right hypochondriac pain ( liver iron overload)
  • nausea , vomiting, pain at injection site, bone pain, joint pain,rash, jaundice, repeated inf ( compli of chelation)

FAMILY HISTORY:

Inv in parents, sibling… mention who are thal traits

NUTRITIONAL HISTORY

IMMUNIZATION – complete till date

  • mention cost of chelation and trans per month
  • concessional rate from thal society
  • earning members
  • mention about financial constraints if any

GENERAL EXAMINATION:

  • thal facies- elaborate
  • pallor present, no “ICCLE”, no platynychia. No koilonychia
  • Any hyperpigmentation

Abdomen: liver — cm below costal margin in mcline, nontender, firm, smooth surface, rounded borders, liver span—-, upper border felt in — intercostal space

Spleen: — cm, splenic notch, smooth surface, moves with respi

ALL SYSTEMS in detail

COMMON QUESTIONS:

  • 5 most common mutations in Indian population with β thalassemia are 619 bp deletion
  • IVS 1-5 (G-C)
  • IVS 1-1 (G-T)
  • FS 8/9 (+G)
  • FS 41/42(-CTTT) 9
  • Hb electrophoresis pattern in normal and in thal  variants
  • Which communities

Kutchis, Sindhis, Punjabis, Bhanushalis, Lohanas, Mahars, Neobuddhists, Gowdas

  • Which chromosome   -11
  • In 1 sentence describe blood trans in a thal pt

10- 15 ml/kg of pure red cell transfusion which are fresh, saline washed, leucodepleted , ABO and rh compatible at not more than 3-4 ml/kg/hour under supervision to maintain Hb above 10gm%

  • Reason for thal facies – bone marrow hyperplasia
  • Hypertransfusion regimen – maintain pretransfusion HB >10 gm%
  • Supertransfusion regimen – maintain pretransfusion HB >12 gm%
  • Moderate trans regimen – maintain pretransfusion HB between 9 and 10.5 gm%
  • No of normoblasts in adequately trans child – <5/100 WBCs
  • Causes of growth failure in thal – endocrine dysfunction + anemia+hypersplenism+ DFX+liver disease
  • Endocrine work up- when to do?

GTT, thyroid function test , ca, phosphorus every year from 5 yrs age.

  • MRI T2* values for cardiac iron load

>20ms- normal, 10-20 borderline iron overload, <10- severe iron overload

  • MRI T2* values for liver iron load Liver T2 * values of >6.3ms are considered normal, 6.3-2.7ms are mild, 2.7-1.4ms are moderate and <1.4ms are severe iron overload.
  • Aims of chelation : ferritin <2500 ng/ml
  • DFO- dose, duration, compli
  • DFP- details
  • DFX- details
  • Shuttle hypothesis   : use of 2 chelators together for better iron removal : eg DFP+DFO
  • Ind of splenectomy – >210 ml/kg/year of transfusion requirement
  • Why is this not a thal variant? 

Share this post:

Related posts

Click here for Case simulation on LN & TB by Dr. Tushar Maniar

Case simulation LN & TB

Click here for Case simulation on Thalassemia

Simulated case - Thalassemia

Case simulation by Dr Chokhani

Simulated cases by Dr Chokhani

Case simulation Cholestasis

Simulated case - Cholestasis

12 comments, leave a reply cancel reply.

Your email address will not be published. Required fields are marked *

Your comment

Your Name *

Your Email *

Save my name, email, and website in this browser for the next time I comment.

This site uses Akismet to reduce spam. Learn how your comment data is processed .

Recent Posts

  • Protected: 18th Feb 2024 Online Mock OSCE February 19, 2024
  • Protected: Online Mock Pediatric OSCE 10th Feb 2024 February 11, 2024
  • Protected: Online Mock Pediatric OSCE 15th Aug 2023 August 15, 2023
  • Protected: Online Mock Pediatric OSCE 6th Aug 2023 August 6, 2023
  • MCQ 49 Vomiting in children July 26, 2023
  • Member Area
  • NEWS & Updates
  • OSCE By Systems
  • PedsResident Writes
  • Theory Papers
  • Uncategorized

Ask The Experts

Core team & contributors.

  • 16th Jan Online OSCE registration
  • Know your exam
  • Members Area
  • Password Reset
  • Reading Material

Copyright © 2011 Cosmo Themes . All rights reserved

IMAGES

  1. Case study thalassemia

    case study on thalassemia

  2. (PDF) Thalassemia-Beta major-Case report

    case study on thalassemia

  3. Thalassemia symptoms, causes, types, diagnosis, test and treatment

    case study on thalassemia

  4. Thalassemia Case Study

    case study on thalassemia

  5. BIO310 CASE STUDY THALASSEMIA

    case study on thalassemia

  6. Thalassemia Case Presentation

    case study on thalassemia

VIDEO

  1. Thalassemia

  2. Long case: Thalassemia in Children- Prof Dr Mohsina Ibrahim- For FCPS/MCPS/CHILD HEALTH

  3. Children's Hospital Oakland

  4. #news World Thalassemia Day 2024 theme

  5. Discover Final Treatment for THALASSEMIA🩸🩸 #thalassemia #treatment

  6. What is Thalasemia in Pregnancy

COMMENTS

  1. A Child Lost to Follow Up Carrying Beta Thalassemia Major: A Case Report

    A five and half year prior diagnosed case of beta thalassemia at age of 2 years and lost to follow up presented with cough, Dyspnoea, Irritability, fatigue with classic symptom of beta thalassemia. ... with anisocy-tosis, poikilocytosis, nucleated Red Blood Cells (RBC). The impression drawn from the peripheral smear study was that of haemolytic ...

  2. Case Study on Thalassemia in Children

    Abstract. Thalassemia, also known as Cooley's anemia or Mediterranean anemia, is a group of hereditary hemolytic anemias characterized by a decrease in hemoglobin synthesis. The word thalassemia is derived from the Greek word thalassa, which means the great sea. Clinical finding: abdominal pain, fever (temperature - 101 degrees F), anemia (Hb 6 ...

  3. (PDF) HbE- THALASSEMIA: A CASE REPORT

    Abstract. BACKGROUND HbE is a variant haemoglobin with a mutation in beta-globin gene, which is the most common Hb variant in South-East Asia. Here, we present a case report of 17 yrs. old boy ...

  4. β-Thalassemias

    In a study involving patients with non-transfusion-dependent β-thalassemia, the severity of anemia (each decrease of 1 g per deciliter in the hemoglobin level) was correlated with the risk of ...

  5. Alpha- and Beta-thalassemia: Rapid Evidence Review

    Persons with beta-thalassemia major lived a mean of 17 years in 1970, most dying by 30 years of age. 48 Recent studies demonstrated mean survival ages of 50 and 57 for patients with beta ...

  6. How I treat non-transfusion-dependent β-thalassemia

    Since the previous How I Treat articles focusing on the thalassemias were published in Blood in 2011 and 2018, our armamentarium for the management of these disorders has expanded. 1, 2 In this article, we present 4 cases as examples of the most contemporary clinical approaches to the diagnosis and management of non-transfusion-dependent β-thalassemia (NTDT), specifically β-thalassemia ...

  7. Case Report: Clinical and Hematological Characteristics of ε

    Introductionεγδβ thalassemia is a rare form of β-thalassemia mostly described in children originating from Northern Europe. Only anecdotic cases from the Mediterranean area are reported. The diagnosis is challenging, considering the rarity of the disease and its heterogeneous clinical presentation. Most patients have neonatal microcytic anemia, sometimes requiring in utero and/or neonatal ...

  8. The α-Thalassemias

    DOI: 10.1056/NEJMra1404415. The thalassemias are the most common human monogenic diseases. 1 These inherited disorders of hemoglobin synthesis are characterized by a reduced production of globin ...

  9. (PDF) Thalassemia-Beta major-Case report

    Citation: Ambarkova V, Krmzova T, Nonkulovski Z (2021) Thalassemia-Beta major-Case report. Arch Hematol Case Rep Rev 6 (1): 021-025. Beta thalassemia ( thalassemia) is a group of inherited blood ...

  10. PDF Approach to Thalassemia

    Approach to Thalassemia: Part 2. These slides are not comprehensive and are meant to use as a visual aid for specific topics within these thalassemia podcasts. You meet Kal in the pediatric emergency room. ID: He is a 6 month old male who was born at 39 weeks via an elective C-section.

  11. Alpha‐thalassemia. Case report alpha‐thalassemia in a Costa Rican

    1 BACKGROUND. The ( - - SEA /) deletion is very common in southern China and in southeast Asia. We report a Costa Rican family of Chinese ethnic background (Guangdong province) with this mutation. This report emphasizes the importance of the alpha-thalassemia syndromes, their carrier prevalence in different populations, relevance in clinical care and family counseling, definitive diagnosis ...

  12. PDF Case Report Case Report on Thalassemia

    the families with thalassemia major child. The prevalence of beta thalassemia carrier is 3 to 4% which translate 35 to 45 million carriers. Thalassemia is an inherited blood disorder in which the body makes an abnormal form of haemoglobin. Haemoglobin is the protein molecule in red blood cells that carries oxygen.

  13. Beta Thalassemia

    Thalassemia (thal-uh-SEE-mee-uh) is a blood disorder that is inherited. This means it is passed down from one or both parents through their genes. When you have thalassemia, your body makes less hemoglobin than normal. Hemoglobin is an iron-rich protein in red blood cells. It carries oxygen to all parts of the body.

  14. β-Thalassemia: evolving treatment options beyond transfusion and iron

    Case 1 demonstrates several strengths and limitations of luspatercept. This medication is most likely to be useful in patients for whom a moderate increase in hemoglobin or a moderate reduction in transfusion volume would be impactful. ... including all 5 patients with α- thalassemia. 34 Phase 3 studies of mitapivat in TDT (NCT04770779) and ...

  15. Thalassemia: Types, Traits, Symptoms & Treatment

    Thalassemias. Thalassemia is an inherited blood disorder that affects your body's ability to produce hemoglobin and healthy red blood cells. Types include alpha and beta thalassemia. Thalassemia may cause you to experience anemia-like symptoms that range from mild to severe. Treatment can consist of blood transfusions and iron chelation therapy.

  16. RQ7KDODVVHPLDLQ&KLOGUHQ

    Case Study on Thalassemia in Children Yohana Sheikha, Ravi Christianb and Nimisha Shrivastavac aIntern, BSc MRIT , School of Allied health Sciences, Department Of Radiodiagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha; Email:[email protected]; Ph no:-9284850862,9561256158.

  17. Challenges in chronic transfusion for patients with thalassemia

    HbE β thalassemia is caused by compound heterozygosity for the E mutation (HBB:c.79G>A) and a β-thalassemia mutation. 19 The prevalence of HbE β thalassemia follows the distribution of the E mutation, which reaches very high frequencies in southeast Asia, southern China, and south Asia. Immigration from Asia to the west has increased the awareness of this syndrome and its distinctive ...

  18. Case Report: β-thalassemia major on...

    Background: β-thalassemia is rare in sub-Saharan Africa and to our knowledge there has been no case of homozygous β-thalassemia major reported from this region. In a recent cohort study, we identified four β-thalassemia mutations among 83 heterozygous carriers in Kilifi, Kenya. One of the mutations identified was a rare β-globin gene initiation codon mutation (ATG ACG) (rs33941849).

  19. Case presentation : Thalassemia

    Case : THALASSEMIA. HISTORY: Consanguinity. Community. Was admitted for receiving blood transfusion. Onset of noticing pallor —- months of age. Investigated and found to have an abnormal blood disorder. Time of 1 st transfusion. Frequency of transfusion.

  20. Thalassemia Case Study

    Thalassemia Case Study - Free download as Word Doc (.doc / .docx), PDF File (.pdf), Text File (.txt) or read online for free. The patient is a 20-year-old female diagnosed with severe anemia due to thalassemia. Lab results show low red blood cell count, hemoglobin, and hematocrit levels indicative of anemia. Thalassemia is a blood disorder where the body makes abnormal or inadequate amounts of ...

  21. Mitapivat Beneficial in Transfusion-Dependent Alpha- or Beta-Thalassemia

    Agios announces phase 3 ENERGIZE-T study of mitapivat met primary endpoint and all key secondary endpoints in adults with transfusion-dependent alpha- or beta-thalassemia. News release. Agios.

  22. Molecules

    β-Thalassemia is an inherited genetic disorder associated with β-globin chain synthesis, which ultimately becomes anemia. Adenosine-2,3-dialdehyde, by inhibiting arginine methyl transferase 5 (PRMT5), can induce fetal hemoglobin (HbF) levels. Hence, the materialization of PRMT5 inhibitors is considered a promising therapy in the management of β-thalassemia. This study conducted a virtual ...