Recent molecular studies of fetal hemoglobin (HbF) regulation have reinvigorated the field and shown promise for the development of clinical HbF inducers to be used in patients with β-thalassemia and sickle cell disease. However, while numerous promising inducers of HbF have been studied in the past in β-thalassemia patient populations, with limited success in some cases, no universally effective agents have been found. Here we examine the clinical studies of such inducers in an attempt to systematically review the field. We examine trials of agents, including 5-azacytidine, hydroxyurea, and short-chain fatty acids. This review highlights the heterogeneity of clinical studies done on these agents, including both the patient populations examined and the study end points. By examining the published studies of these agents, we hope to provide a resource that will be valuable for the design of future studies of HbF inducers in β-thalassemia patient populations.

Medscape EDUCATION Continuing Medical Education online

This activity has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education through the joint sponsorship of Medscape, LLC and the American Society of Hematology.

Medscape, LLC is accredited by the ACCME to provide continuing medical education for physicians.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1.0 AMA PRA Category 1 Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test with a 70% minimum passing score and complete the evaluation at http://www.medscape.org/journal/blood; and (4) view/print certificate. For CME questions, see page 2372.

Disclosures

The authors, Associate Editor Narla Mohandas, and CME questions author Charles P. Vega, Associate Professor and Residency Director, Department of Family Medicine, University of California-Irvine, declare no competing financial interests.

Learning objectives

Upon completion of this activity, participants will be able to:

  1. Assess the pathophysiology of β-thalassemia.

  2. Analyze the efficacy of hydroxyurea in the management of β-thalassemia.

  3. Analyze the safety of hydroxyurea in the management of β-thalassemia.

  4. Evaluate the use of other inducers of HbF among patients with β-thalassemia.

Release date: March 21, 2013; Expiration date: March 21, 2014

Increased production of fetal hemoglobin (HbF) can ameliorate the severity of both β-thalassemia and sickle cell disease (SCD), the major disorders of β-hemoglobin. The defective production of the β-globin molecule in patients with β-thalassemia can be compensated for by an increase in the production of the β-like globin molecule, γ-globin, which pairs together with α-globin chains to form HbF.1,2  The increased γ-globin production decreases the α/β-chain imbalance that is a hallmark of β-thalassemia. As a result, there is improvement in the ineffective erythropoiesis seen in the disease, decreased hemolysis, and increased total hemoglobin levels from the improved survival of red cells containing higher levels of HbF.3  The earliest clinical observations suggesting that this was the case came from patients with rare forms of β-thalassemia, particularly those with deletions that result in hereditary persistence of fetal hemoglobin, who express high levels of HbF and have a relatively benign clinical course.2  Additionally, infants with β-thalassemia only become symptomatic following the decrease in HbF production as the normal developmental fetal-to-adult hemoglobin switch occurs. More-recent clinical studies have substantiated the quantitative ameliorating effect of increased HbF production on the clinical course in a variety of patients with β-thalassemia.4-9 

These observations have prompted a 3-decade search for inducers of HbF that can therapeutically recapitulate what occurs in β-thalassemia patients who have naturally higher levels of HbF. These efforts started almost immediately after the cloning of the globin genes and began after the initial recognition that DNA methylation occurred at the silenced γ-globin genes. While these initial efforts have subsequently generated a large body of evidence in SCD that led to the successful use of HbF inducers, as exemplified by the widespread use of hydroxyurea, experience in patients with β-thalassemia remains much more limited.

Here we review all clinical studies evaluating HbF inducers that have been performed in β-thalassemia patients. We attempt to be simultaneously comprehensive and succinct in our analysis of the available evidence. Of note, there have been no large randomized clinical trials performed to date in this area of clinical investigation. This review serves a dual purpose. First, these studies have never been comprehensively reviewed, and therefore we have written this review to serve as a resource for those interested in examining the literature on clinical efforts to induce HbF in β-thalassemia. Second, recent basic science studies have identified extremely promising molecular regulators of HbF, including BCL11A, MYB, and KLF1.10-15  While no therapeutics currently exist to target these molecules or related pathways, it is likely that further work prompted by these molecular findings may lead to the discovery of promising candidates for clinical HbF inducers in the near future. Additionally, epigenetic modifier partners of these transcription factors, such as histone deacetylases (HDACs), already have inhibitors available in clinical use and thus may represent more-immediate targets for HbF induction trials.10,16  However, given the heterogeneous clinical studies of HbF inducers that have been performed to date, the best approaches to carry out such studies remains enigmatic. By carefully revisiting clinical studies that have been performed to date, we hope to provide a resource for all such future clinical studies in β-thalassemia patient populations.

Soon after the initial cloning of the globin genes in the late 1970s, it became apparent that DNA methylation was present at the silenced γ-globin genes in adult erythroid cells, but was absent from the active γ-globin genes in fetal erythroid cells.17  Around this time, the mechanism of action of the DNA methylation inhibitor 5-azacytidine was being elucidated.18,19  This led DeSimone et al20  to examine whether 5-azacytidine could induce HbF in phlebotomized primates. The results were stunning and led Ley et al21  to test 5-azacytidine in a patient with severe β-thalassemia. After 7 days of therapy, γ-globin synthesis increased 7-fold, normalizing the globin chain imbalance and leading to an increase in total hemoglobin level from 80 to 108 g/L. Due to concerns and debates about the safety of this agent (cytotoxicity, mutagenicity, immuno- and myelosupression, activation of latent viruses),22,23  subsequent use was limited to severe cases for whom conventional therapy was unfeasible. Dunbar et al24  reported a remarkable improvement in total hemoglobin from 62 to 92 g/L in an alloimmunized β-thalassemia patient after 5 days of 5-azacytidine therapy. Similar findings of increases in total hemoglobin level of approximately 30 g/L, transfusion independence, and improvement in cardiac function were described in a report on 3 patients with end-stage β-thalassemia.25  Myelotoxicities requiring dose modifications were the main concern. Similar reports from patients with SCD confirmed the rapid and favorable effects on HbF production and the hematological outcomes.26-29  Of note, a 2008 study suggested that 5-azacytidine induction of HbF was not the result of global DNA demethylation or changes in differentiation kinetics, but rather may be due to a localized demethylation of the γ-promoter, although other results suggest that posttranscriptional regulation could also play an important role.30 

Decitabine (5-aza-2’-deoxycytidine) was also shown to demethylate and reactivate the expression of the γ-globin gene. At low concentrations, it has a favorable safety profile without causing significant DNA damage or cytotoxicity. Small studies in SCD suggest that decitabine can substantially increase HbF and total hemoglobin in the majority of patients treated.31-34  A pilot study in 2011 showed that subcutaneous decitabine given at 0.2 mg/kg 2 times per week for 12 weeks increased total hemoglobin from 78.8 to 90.4 g/L (2 patients had elevations ≥15 g/L), and it increased absolute fetal hemoglobin from 36.4 to 42.9 g/L in 5 patients with β-thalassemia intermedia. Favorable changes in red blood cell indices were also noted. Treatment was well tolerated, with the main adverse event being an elevation in platelet counts.35 

Hydroxyurea (or hydroxycarbamide) is a cytotoxic, antimetabolic, and antineoplastic agent known for its use in the management of patients with myeloproliferative disorders36  and human immunodeficiency virus infection,37  where the drug acts as a potent inhibitor of ribonucleotide reductase, an enzyme required for DNA synthesis and repair. After being identified as a potent HbF inducer,38  hydroxyurea became 1 of the key therapeutic agents for the management of patients with SCD. However, the exact mechanisms by which hydroxyurea induces HbF production are not fully understood. A cytotoxic effect resulting in stress erythropoiesis, with increased HbF levels occurring as a result, is most commonly proposed.39  More-complex effects involving the production of nitric oxide and the soluble guanylyl cyclase and cyclic guanosine monophosphate–dependent protein kinase pathway gene have been proposed as being responsible for this activity.40-43  Hydroxyurea therapy exerts a 2- to 9-fold increase in γ-mRNA expression in β-thalassemia patients,44-48  leading to improvement in the α/non–α-chain imbalance and more-effective erythropoiesis.49  There is good correlation between in vitro γ-mRNA fold increase and the in vivo HbF fold increase;47,50  however, increases in HbF level did not always correlate with increases in total hemoglobin level in clinical studies, as described in subsequent sections. This may be best explained by findings from earlier studies showing increases in the α/β but not the α/γ biosynthetic ratio in β-thalassemia patients receiving hydroxyurea.51,52  Thus, in addition to its known effects in stimulating γ-globin production, hydroxyurea may have a more general role in augmenting globin synthesis, including β-globin in some patients who maintain the capacity to express normal β-globin chains.51 

Evidence from patients with SCD suggests that the benefits of hydroxyurea may not be limited to the increase in HbF levels, because improved erythrocyte morphology and deformability, a lowering of circulating leukocyte and reticulocyte counts, a reduction in hemolysis, and a potentially local release of nitric oxide also appear to contribute to improved clinical outcomes.53  Similar studies in patients with β-thalassemia are limited. Hydroxyurea therapy is not associated with considerable or steady effects on erythrocyte deformability in β-thalassemia,54  which may explain the reduced response to the drug in some patients.55  However, in splenectomized patients with hemoglobin E/β-thalassemia, there is evidence that hydroxyurea diminishes phosphatidylserine externalization on the red cell,54  an observation previously established in patients with SCD.56  Whether this is attributed to HbF induction and an associated decrease in α-globin aggregates remains to be elucidated.54  Regardless, phosphatidylserine membrane exposure is not only associated with reduced red cell survival but also with increased thrombin generation, leading to hypercoagulability and subsequent morbidity in patients with β-thalassemia.57 

Hematological outcomes

After early case reports documented hematological improvements in β-thalassemia patients treated with hydroxyurea,51,58-64  several studies evaluated the efficacy and safety of the drug in this patient population (Table 1).44,49,50,65-85  These primarily included small single-arm trials or retrospective cohort studies. Reported elevations in HbF level from baseline showed substantial variability, ranging between 1% and 90% and averaging 20%. Such increases are generally higher than those reported in SCD trials,43,53  although patients with β-thalassemia recruited in the available studies have higher baseline HbF levels. An association between the degree of HbF level increase and improved hematological outcomes was noted in some studies,70,73  while others failed to document such an association.49,65,82  In fact, some reports noted a reduction in HbF level, especially at low doses,86  despite observed hematological responses,51,72  further supporting the idea that the effects of hydroxyurea in β-thalassemia patients could extend beyond HbF induction.

Table 1

Clinical studies evaluating hydroxyurea therapy in patients with β-thalassemia

ReferenceSampleAge (y)*HU dose (mg/kg per d)*Duration (mo)Total Hb (g/L)*Fetal Hb*Comments
Karimi et al65  126 TM 22.3 10 96-156 Baseline: 85-100 — TI: NTD or TD [intermittent] 
Final: ≥90 (n = 51 [40%])  TM: 86 [68.3%] TD → NTD, 25 [19.8%] TD → 1 or 2 transfusions 
Final: ≥80 to <90 (n = 44 [35%])  
Final: ≥70 to <80 (n = 16 [13%])  
Final: <70 (n = 15 [12%])  
106 TI 18.1 10 96-156 Δ: +≥20 (n = 12 [11%]) — 
Δ: +10 to <20 (n = 44 [42%]) 
Δ: +<10 (n = 47 [44%]) 
Δ: no change (n = 3 [3%]) 
Amoozgar et al66  33 TI 20 10 12 Baseline: 98.4 Baseline: 79.3 g/L TI: NTD or TD [intermittent] 
Δ: +13.4 Δ: +8.0 g/L 
51 TI 17 None 12 Baseline: 92.8 — 
Δ: +4.7   
Ansari et al67  119 TM 2.6-25 16 24 -Baseline: 75 Baseline: 11.2% TI: NTD 
Δ: +1 (responders, n = 33 [27.7%]) Δ: +88.4% (responders) Responders: TD → NTD 
-Baseline: 82  Partial responders: >50% ↓ in transfusion need 
Δ: −14 (partial responders, n = 57 [47.9%])  
-Baseline: 79  Nonresponders: <50% ↓ in transfusion need 
Δ: −13 (nonresponders, n = 29 [24.4%])  
27 TI 3-13 16 24 Baseline: 67 Baseline: 98.5%  
Δ: +11 Δ: +1.4%  
Italia et al68  13 Hb E/β-thalassemia 8-34 15-20 17-23 -Baseline: 61 -Baseline: 22.3% Responders: TD → NTD 
(median 20) Δ: +11 (responders, n = 6 [46.2%]) Δ: +16% (responders) Partial responders: ↓ in transfusion need (30% to 50%) 
-Baseline: 63 F cells: +25.8% 
Δ: 0 (partial responders, n = 4 [30.8%]) -Baseline: 15.2% Nonresponders: no change in transfusion need 
-Baseline: 76 Δ: +16.6% (partial responders) 
Δ: −12 (nonresponders, n = 3 [23.1%]) F cells: +18.5% 
-Baseline: 2.9% 
Δ: +5.8% (nonresponders) 
F cells: +13.3% 
Karimi et al69  143 TI 21 10.7 18-120 Baseline: 74 — TI: NTD or TD 
   (median 68.4) Δ: +23   
Rigano et al50  24 TI 37 14.6 12 Baseline: 78 Baseline: 46.5% TI: NTD 
Δ: +15 Δ: +10.7% 17 patients with ΔHb > 10 g/L followed for >12 mo (mean: 68 mo); 9 maintained response, while 8 had reduction 
Ehsani et al70  16 TI 10.7 20 Δ: +16 Δ: +17 g/L TI: NTD 
Italia et al44  41 TM 12-28 15-20 20-24 -Baseline: 81 -Baseline: 5.4% TI: TD [intermittent] or TD 
Δ: −5 (partial responders, n = 13 [31.7%]) Δ: +32% (partial responders) Responders: TD → NTD 
-Baseline: 76 F cells: +40.9% Partial responders: >50% ↓ in transfusion need 
Δ: −5 (nonresponders, n = 28 [68.3%]) -Baseline: 3.7% 
Δ: +8.2% (nonresponders) Dose increases to 30 mg/kg per day in nonresponders had no benefit 
F cells: +13.8% 
38 TI 5-40 15-20 20-24 -Baseline: 76 -Baseline: 51.8%  
Δ: +15 (responders, n = 22 [57.9%]) Δ: +35.4% (responders) 
-Baseline: 79 F cells: +27.4% 
Δ: −2 (partial responders, n = 6 [15.8%]) -Baseline: 6.3% 
-Baseline: 63 Δ: +31.1% (partial responders) 
Δ: −2 (nonresponders, n = 10 [26.3%]) F cells: +33.3% 
-Baseline: 25.9% 
Δ: +20% (nonresponders) 
F cells: +32.8% 
Zamani et al71  49 TM 18.4 10 12 Baseline: 85.2 — Mean number of transfused units ↓ from 22.8 → 6.0 
Δ: −0.7 
12 [24.5%] became NTD 
 
Koren et al72  11 TM 18 10.9 5-60 Responders, n = 9 [81.8%] Baseline: 83% TI: NTD or TD [intermittent] 
(median 24) Δ: −1% (1 y) , −5% (2 y)  Responders: TD → NTD (maintaining an Hb of 70 g/L in TM and 60 g/L in TI) 
7 TI 18 10.9 36-96 -Baseline: 67 Baseline: 15%  
(median 60) Δ: +2 (5 TD, responders, n = 5 [100%]) Δ: +13% (1 y), +8% (2 y)  
-Baseline: 66 
Δ: +18 (2 NTD) 
Mtvarelidze et al73  6 TM 9.8 15 60 Baseline: 94 Baseline: 23% 3 [50%] patients: TD → NTD 
Δ: +1 (12 mo), +3 (60 mo) Δ: +8.3% (12 mo), +20.4 (60 mo) 2 [33.3%] patients: ↓ in transfusion need after 1 y 
Ansari et al74  23 TM 8.7 16 24 Baseline: 86 — 42.8% ↓ in blood volume transfused 
Δ: −14 68.7% ↑ in transfusion interval 
Bradai et al75  45 TM 10 17.4 12 Δ: +15 (good responders, n = 20 [44.4%]) — TI: TD 
Δ: +7 (partial responders, n = 9 [20%]) Good responders: >70% ↓ in transfusion need 
Δ: +2 (nonresponders, n = 16 [35.6%]) 
 Partial responders: 40% to 70% ↓ in transfusion need 
9 TI 12.5 17 12 Δ: +37 (8 good responders [88.9%], 1 nonresponder [11.1%]) — Nonresponders: <40% ↓ in transfusion need 
Mancuso et al76  18 TI 37 14.6 12 Δ: +15 — TI: NTD 
11 patients with ΔHb > 10 g/L were followed for >12 mo (mean: 66 mo); 7 maintained response, while 4 had reduction 
Taher and Sheikh-Taha84  7 TI 24 10-20 6-46 Baseline: 70 — TI: NTD 
(median 12) Δ: +5 
Singer et al85  45 Hb E/β-thalassemia 13 18-20 24 Baseline: 68 Baseline: 29.8% Patients weaned off transfusions before study 
Δ: +6 Δ: +6.7% 
  16 [38%] patients had ΔHb > 10 g/L 
Dixit et al77  37 TM + TI 10 10->20 -Baseline: 65 -Baseline: 67% -TI: NTD, TD [intermittent], or TD 
Δ: +26 (major responders, n = 17 [45.9%]) Δ: +9% (major responders) Major responders: TD → NTD (Hb > 80 g/L), or Hb ↑ >20 g/L 
-Baseline: 63 -Baseline: 70.2% 
Δ: +18 (minor responders, n = 9 [24.3%]) Δ: +8.5 (minor responders) Minor responders: TD → NTD (Hb < 80 g/L), >50% ↓ in transfusion need, or Hb ↑ 10 to 20 g/L 
-Baseline: 65 -Baseline: 40.9% 
Δ: +5 (nonresponders, n = 11 [29.7%]) Δ: +1% (nonresponders) 
 
  Nonresponders: <50% ↓ in transfusion need, or Hb ↑ <10 g/L 
Karimi et al78  163 TI 13.5 10 72 Baseline: 86.8 Nonsignificant changes TI: group 1 (n = 120) TD, group 2 (n = 16) TD [intermittent], and group 3 (n = 27) NTD 
Δ: +9.6 (group 2 + 3, 12 mo) 
 
 Group 1: 83 [69.2%] TD → NTD, 23 [19.2%] TD → 1 or 2 transfusions 
 Group 2: 14 [87.5%] TD → NTD 
Alebouyeh et al79  36 TM 16.3 20 >6 Baseline: 100 — TI: NTD 
Δ: +7 25 [69.4%] TM patients TD → NTD 
9 TI 14.7 20 34-58 Baseline: 93 —  
(median 40) Δ: +11 
Yavarian et al80  133 TM 17.1 10-15 24-60 Good responders: n = 81 [60.9%] — Good responders: TD → NTD (Hb > 95 g/L) 
(median 42) Moderate responders: n = 31 [23.3%] 
  Moderate responders: transfusion interval >6 mo (Hb 75 to 96 g/L) 
  Nonresponders: TD after 12 mo 
Bradai et al81  7 TM + TI 12 18.4 13-21 -Baseline: 45 Baseline: 90.9% TI: TD 
(median 19) Δ: +34 (TM) Δ: +6.8% 5 [71.4%] patients: TD → NTD 
-Baseline: 65 2 [28.6%] TD → 1 or 2 transfusions 
Δ: +40 (TI) 
de Paula et al82  4 TM 18 10->20 6-96 Δ: +30 in 1 patient after 3 mo, no change in 3 patients — TI: TD [intermittent] 
1 TM patient became NTD after 3 mo 
7 TI 35 10->20 Baseline: 77 Baseline: 44.9% ΔHb > 10 g/L noted in 3 TI patients at 6 mo, 1 patient showed further response at 12 mo, no additional responses noted after >12 mo 
Δ: +10 Δ: +12.3% 
Choudhry et al83  15 TM 3-6 50 3 5-d courses Δ: +8 2-fold rise in 8 [53.3%] patients Response continued for 6 mo in 9 [60%] patients 
Fucharoen et al49  13 Hb E/β-thalassemia 34 15 Baseline: 65 Baseline: 42% — 
Δ: +7.4 Δ: +32.5% 
F cells: ↑ by 1/3 
ReferenceSampleAge (y)*HU dose (mg/kg per d)*Duration (mo)Total Hb (g/L)*Fetal Hb*Comments
Karimi et al65  126 TM 22.3 10 96-156 Baseline: 85-100 — TI: NTD or TD [intermittent] 
Final: ≥90 (n = 51 [40%])  TM: 86 [68.3%] TD → NTD, 25 [19.8%] TD → 1 or 2 transfusions 
Final: ≥80 to <90 (n = 44 [35%])  
Final: ≥70 to <80 (n = 16 [13%])  
Final: <70 (n = 15 [12%])  
106 TI 18.1 10 96-156 Δ: +≥20 (n = 12 [11%]) — 
Δ: +10 to <20 (n = 44 [42%]) 
Δ: +<10 (n = 47 [44%]) 
Δ: no change (n = 3 [3%]) 
Amoozgar et al66  33 TI 20 10 12 Baseline: 98.4 Baseline: 79.3 g/L TI: NTD or TD [intermittent] 
Δ: +13.4 Δ: +8.0 g/L 
51 TI 17 None 12 Baseline: 92.8 — 
Δ: +4.7   
Ansari et al67  119 TM 2.6-25 16 24 -Baseline: 75 Baseline: 11.2% TI: NTD 
Δ: +1 (responders, n = 33 [27.7%]) Δ: +88.4% (responders) Responders: TD → NTD 
-Baseline: 82  Partial responders: >50% ↓ in transfusion need 
Δ: −14 (partial responders, n = 57 [47.9%])  
-Baseline: 79  Nonresponders: <50% ↓ in transfusion need 
Δ: −13 (nonresponders, n = 29 [24.4%])  
27 TI 3-13 16 24 Baseline: 67 Baseline: 98.5%  
Δ: +11 Δ: +1.4%  
Italia et al68  13 Hb E/β-thalassemia 8-34 15-20 17-23 -Baseline: 61 -Baseline: 22.3% Responders: TD → NTD 
(median 20) Δ: +11 (responders, n = 6 [46.2%]) Δ: +16% (responders) Partial responders: ↓ in transfusion need (30% to 50%) 
-Baseline: 63 F cells: +25.8% 
Δ: 0 (partial responders, n = 4 [30.8%]) -Baseline: 15.2% Nonresponders: no change in transfusion need 
-Baseline: 76 Δ: +16.6% (partial responders) 
Δ: −12 (nonresponders, n = 3 [23.1%]) F cells: +18.5% 
-Baseline: 2.9% 
Δ: +5.8% (nonresponders) 
F cells: +13.3% 
Karimi et al69  143 TI 21 10.7 18-120 Baseline: 74 — TI: NTD or TD 
   (median 68.4) Δ: +23   
Rigano et al50  24 TI 37 14.6 12 Baseline: 78 Baseline: 46.5% TI: NTD 
Δ: +15 Δ: +10.7% 17 patients with ΔHb > 10 g/L followed for >12 mo (mean: 68 mo); 9 maintained response, while 8 had reduction 
Ehsani et al70  16 TI 10.7 20 Δ: +16 Δ: +17 g/L TI: NTD 
Italia et al44  41 TM 12-28 15-20 20-24 -Baseline: 81 -Baseline: 5.4% TI: TD [intermittent] or TD 
Δ: −5 (partial responders, n = 13 [31.7%]) Δ: +32% (partial responders) Responders: TD → NTD 
-Baseline: 76 F cells: +40.9% Partial responders: >50% ↓ in transfusion need 
Δ: −5 (nonresponders, n = 28 [68.3%]) -Baseline: 3.7% 
Δ: +8.2% (nonresponders) Dose increases to 30 mg/kg per day in nonresponders had no benefit 
F cells: +13.8% 
38 TI 5-40 15-20 20-24 -Baseline: 76 -Baseline: 51.8%  
Δ: +15 (responders, n = 22 [57.9%]) Δ: +35.4% (responders) 
-Baseline: 79 F cells: +27.4% 
Δ: −2 (partial responders, n = 6 [15.8%]) -Baseline: 6.3% 
-Baseline: 63 Δ: +31.1% (partial responders) 
Δ: −2 (nonresponders, n = 10 [26.3%]) F cells: +33.3% 
-Baseline: 25.9% 
Δ: +20% (nonresponders) 
F cells: +32.8% 
Zamani et al71  49 TM 18.4 10 12 Baseline: 85.2 — Mean number of transfused units ↓ from 22.8 → 6.0 
Δ: −0.7 
12 [24.5%] became NTD 
 
Koren et al72  11 TM 18 10.9 5-60 Responders, n = 9 [81.8%] Baseline: 83% TI: NTD or TD [intermittent] 
(median 24) Δ: −1% (1 y) , −5% (2 y)  Responders: TD → NTD (maintaining an Hb of 70 g/L in TM and 60 g/L in TI) 
7 TI 18 10.9 36-96 -Baseline: 67 Baseline: 15%  
(median 60) Δ: +2 (5 TD, responders, n = 5 [100%]) Δ: +13% (1 y), +8% (2 y)  
-Baseline: 66 
Δ: +18 (2 NTD) 
Mtvarelidze et al73  6 TM 9.8 15 60 Baseline: 94 Baseline: 23% 3 [50%] patients: TD → NTD 
Δ: +1 (12 mo), +3 (60 mo) Δ: +8.3% (12 mo), +20.4 (60 mo) 2 [33.3%] patients: ↓ in transfusion need after 1 y 
Ansari et al74  23 TM 8.7 16 24 Baseline: 86 — 42.8% ↓ in blood volume transfused 
Δ: −14 68.7% ↑ in transfusion interval 
Bradai et al75  45 TM 10 17.4 12 Δ: +15 (good responders, n = 20 [44.4%]) — TI: TD 
Δ: +7 (partial responders, n = 9 [20%]) Good responders: >70% ↓ in transfusion need 
Δ: +2 (nonresponders, n = 16 [35.6%]) 
 Partial responders: 40% to 70% ↓ in transfusion need 
9 TI 12.5 17 12 Δ: +37 (8 good responders [88.9%], 1 nonresponder [11.1%]) — Nonresponders: <40% ↓ in transfusion need 
Mancuso et al76  18 TI 37 14.6 12 Δ: +15 — TI: NTD 
11 patients with ΔHb > 10 g/L were followed for >12 mo (mean: 66 mo); 7 maintained response, while 4 had reduction 
Taher and Sheikh-Taha84  7 TI 24 10-20 6-46 Baseline: 70 — TI: NTD 
(median 12) Δ: +5 
Singer et al85  45 Hb E/β-thalassemia 13 18-20 24 Baseline: 68 Baseline: 29.8% Patients weaned off transfusions before study 
Δ: +6 Δ: +6.7% 
  16 [38%] patients had ΔHb > 10 g/L 
Dixit et al77  37 TM + TI 10 10->20 -Baseline: 65 -Baseline: 67% -TI: NTD, TD [intermittent], or TD 
Δ: +26 (major responders, n = 17 [45.9%]) Δ: +9% (major responders) Major responders: TD → NTD (Hb > 80 g/L), or Hb ↑ >20 g/L 
-Baseline: 63 -Baseline: 70.2% 
Δ: +18 (minor responders, n = 9 [24.3%]) Δ: +8.5 (minor responders) Minor responders: TD → NTD (Hb < 80 g/L), >50% ↓ in transfusion need, or Hb ↑ 10 to 20 g/L 
-Baseline: 65 -Baseline: 40.9% 
Δ: +5 (nonresponders, n = 11 [29.7%]) Δ: +1% (nonresponders) 
 
  Nonresponders: <50% ↓ in transfusion need, or Hb ↑ <10 g/L 
Karimi et al78  163 TI 13.5 10 72 Baseline: 86.8 Nonsignificant changes TI: group 1 (n = 120) TD, group 2 (n = 16) TD [intermittent], and group 3 (n = 27) NTD 
Δ: +9.6 (group 2 + 3, 12 mo) 
 
 Group 1: 83 [69.2%] TD → NTD, 23 [19.2%] TD → 1 or 2 transfusions 
 Group 2: 14 [87.5%] TD → NTD 
Alebouyeh et al79  36 TM 16.3 20 >6 Baseline: 100 — TI: NTD 
Δ: +7 25 [69.4%] TM patients TD → NTD 
9 TI 14.7 20 34-58 Baseline: 93 —  
(median 40) Δ: +11 
Yavarian et al80  133 TM 17.1 10-15 24-60 Good responders: n = 81 [60.9%] — Good responders: TD → NTD (Hb > 95 g/L) 
(median 42) Moderate responders: n = 31 [23.3%] 
  Moderate responders: transfusion interval >6 mo (Hb 75 to 96 g/L) 
  Nonresponders: TD after 12 mo 
Bradai et al81  7 TM + TI 12 18.4 13-21 -Baseline: 45 Baseline: 90.9% TI: TD 
(median 19) Δ: +34 (TM) Δ: +6.8% 5 [71.4%] patients: TD → NTD 
-Baseline: 65 2 [28.6%] TD → 1 or 2 transfusions 
Δ: +40 (TI) 
de Paula et al82  4 TM 18 10->20 6-96 Δ: +30 in 1 patient after 3 mo, no change in 3 patients — TI: TD [intermittent] 
1 TM patient became NTD after 3 mo 
7 TI 35 10->20 Baseline: 77 Baseline: 44.9% ΔHb > 10 g/L noted in 3 TI patients at 6 mo, 1 patient showed further response at 12 mo, no additional responses noted after >12 mo 
Δ: +10 Δ: +12.3% 
Choudhry et al83  15 TM 3-6 50 3 5-d courses Δ: +8 2-fold rise in 8 [53.3%] patients Response continued for 6 mo in 9 [60%] patients 
Fucharoen et al49  13 Hb E/β-thalassemia 34 15 Baseline: 65 Baseline: 42% — 
Δ: +7.4 Δ: +32.5% 
F cells: ↑ by 1/3 

Hb, hemoglobin; HU, hydroxyurea; NTD, nontransfusion dependent; TD, transfusion dependent; TI, β-thalassemia intermedia; TM, β-thalassemia major.

*

All values represent the central tendency (mean or median) unless otherwise indicated. For HU dose, the fixed or actual mean/median maximal tolerated dose is indicated when reported; otherwise, the dosing scheme is described.

Statistically significant (P < .05).

Statistically insignificant (P > .05).

In the available studies, two main hematological outcomes were commonly evaluated, depending on patients’ transfusion status before hydroxyurea therapy. In transfusion-dependent phenotypes (β-thalassemia major, severe β-thalassemia intermedia, or severe hemoglobin E/β-thalassemia), patients were considered responders to therapy if they became transfusion independent. The approximate response rate ranged between 30% and 70% in β-thalassemia major patients and between 60% and 100% in β-thalassemia intermedia patients, although the latter group was less frequently transfused before hydroxyurea therapy. The response rate in patients with hemoglobin E/β-thalassemia was around 50%. The evaluation of reported elevations in total hemoglobin level in this transfusion-dependent group of patients is challenging considering the variable time of baseline measurement with respect to transfusion therapy. However, responding patients usually had higher increases in total hemoglobin level than nonresponding patients did. Partial response, usually defined as a decrease in transfusion requirements, ranged between 15% and 50% (Table 1). Although these findings remain promising, they should be interpreted with caution, especially in patients with β-thalassemia intermedia. The confidence intervals in studies reporting the highest response rates are expected to be wide considering the small sample sizes. Moreover, interpretation of the observed benefits may be challenging, as the indications for transfusion therapy with respect to target total hemoglobin levels vary considerably between studies and centers. The documented final total hemoglobin level achieved upon transfusion independence ranged between 60 and 100 g/L. Finally, recent evidence supports a benefit of transfusion therapy for the prevention or management of several morbidities in patients with β-thalassemia intermedia,87,88  and the role of hydroxyurea as an alternate therapy should be evaluated against similar end points, particularly given that analogous transitions in patients with SCD have not consistently met with success.89  However, it is noteworthy that improvements in transfusion requirements in most β-thalassemia patients were also associated with a favorable reduction in iron overload and hemolytic indices.

In studies including transfusion-independent patients with β-thalassemia, the primary hematological outcome was improvement in total hemoglobin level. Mean increases within studies ranged approximately between 5 and 25 g/L, with an average around 15 g/L (Table 1), which is comparable to findings in patients with SCD.43,53  However, a high variance is noted in total hemoglobin response in most studies, indicating that although some patients achieve considerable elevations, others have minimal or no change. The proportion of patients having total hemoglobin increases >10 g/L ranged between 40% and 70%.50,65,76,77,85  Improvement in anemia was usually associated with better exercise tolerance, appetite, and sense of well-being. Elevations in mean corpuscular volume and hemoglobin were consistently noted along with improvements in total hemoglobin level.

Predictors of response

Responses in β-thalassemia patients were observed at hydroxyurea doses ranging between 10 and 20 mg/kg per day, with most investigators opting to use a fixed low dose (10 mg/kg per day), while others escalated the dose according to toxicity (maximal tolerated dose) up to a maximum of 20 mg/kg per day (Table 1). These doses remain lower than those used in patients with SCD, which are often in excess of 20 mg/kg per day.43,53  Whether dose increments above 20 mg/kg per day could lead to more favorable responses warrants further study; however, a 2009 report suggests that a dose increase to 30 mg/kg per day in a small group of nonresponsive patients did not provide any additional benefit.44 

Experience from patients with SCD established the long-term efficacy of hydroxyurea.43,53  In β-thalassemia, most studies evaluated outcomes after 6, 12, or 24 months of therapy, although results from longer follow-up were also reported (Table 1). Response to hydroxyurea therapy was commonly noted in the first 3 to 6 months of therapy, with further improvements noted up to 12 months, and sustained responses observed over long-term follow-up.65,69,73,78-80,82,85  However, some studies noted a decline in hematological response beyond 12 months.50,76  Because of these observations, it has been theorized that long-term treatment with hydroxyurea may result in an impairment in the ability of certain hematopoietic stem cells to give rise effectively to erythroid lineage cells. Basic science studies suggest that peripheral blood hematopoietic stem and progenitor cells from β-thalassemia patients under long-term treatment with hydroxyurea lose the ability to undergo erythroid differentiation, which may lend some support to such theories.50 

Alongside dose and duration of therapy, several other factors were assessed for their association with hematological response in patients with β-thalassemia. Findings regarding the roles of age and HbF level at the start of treatment are conflicting.49,65,70,73,77,85  Moreover, although some studies found certain β-globin genotypes to be predictors of a favorable response,50,70,79  others failed to establish such an association.44,65,75,77,85  Similar discrepancies are noted for β-globin haplotypes.44,49  Patients with Lepore or δβ-thalassaemia genotypes usually showed a better response.50,61,90  Of note, several reports confirm that the effects of hydroxyurea in patients with hemoglobin S/β-thalassemia may be better than those even reported for homozygous hemoglobin S disease, because the synthesized γ-chains not only inhibit the sickling process, but they also neutralize the noxious effects of the excess α-chains and cut down on ineffective erythropoiesis.91-95  Co-inheritance of α-thalassemia was described as a predictor of good response in some studies,44,77,96  but it was found to have no effect in others.49,68,85  Homozygosity for the XmnI polymorphism (−158C>TGγ) was a strong predictor of favorable responses,44,72,75,79,80,97  although the case was different in some studies, especially those including patients with hemoglobin E/β-thalassemia.65,68,70,77  A 2012 study97  also showed that the rs766432 polymorphism at intron 2 of the BCL11A gene correlates strongly with a response to hydroxyurea therapy; further studies in this direction are encouraged. However, it is important to bear in mind that when genetic markers are studied, the markers themselves may not be causally linked to such effects, but rather variants in linkage disequilibrium with these markers may be the mediators of such effects.

There is some basic evidence that response to hydroxyurea may be affected by the degree of iron overload.98  However, clinical studies evaluating such an association are lacking.

Other clinical outcomes

Hydroxyurea therapy was also found to decrease the frequency of certain morbidities in patients with β-thalassemia. A beneficial role in patients with pulmonary hypertension was suggested, especially upon combination with the antioxidant L-carnitine, although these findings primarily relied on echocardiographic findings.66,99,100  In smaller studies, hydroxyurea therapy was also associated with improvements in endocrine function,80  leg ulcers,101  and extramedullary hemtopoietic tumors.102  These findings are further confirmed through a 2010 cross-sectional study of 584 β-thalassemia intermedia patients from the Middle East and Italy, where hydroxyurea therapy was associated with reduced adjusted odds of extramedullary hemtopoietic tumors (0.52, 95% CI, 0.30-0.91), pulmonary hypertension (0.42, 95% CI, 0.20-0.90), leg ulcers (0.10, 95% CI, 0.02-0.43), hypothyroidism (0.05, 95% CI, 0.01-0.45), and osteoporosis (0.02, 95% CI, 0.01-0.09).87  These effects were independent of total hemoglobin level or transfusion status, which further suggests that the benefit from hydroxyurea could extend beyond HbF induction and subsequent improvement of anemia. It should be noted that hydroxyurea’s effects, especially on phosphatidylserine exposure and hypercoagulability, are most notable in splenectomized β-thalassemia patients,54,81  which represent the subgroup of patients with a considerably high risk for morbidity from thrombotic events.87 

Safety of hydroxyurea in β-thalassemia patients

Hydroxyurea therapy was generally well tolerated at the doses used in β-thalassemia studies, with some studies reporting no adverse events at all, even with long-term therapy.66,73,76,81,83,84  The rate of myelotoxicity ranged between 2% and 30%,44,49,71,74,75,77,82,85  while some studies did not report any hematological toxicities.65,69,70,72  Myelotoxicity was usually dose dependent, especially when doses >20 mg/kg per day were used, and it could be reversed upon dose reduction. The bone marrow of β-thalassemia patients may be more sensitive to myelosuppression by hydroxyurea than occurs in other disorders, possibly due to medullary inflammation.86,103  There is only 1 report of leukemic transformation in a β-thalassemia intermedia patient following 3 years of hydroxyurea therapy at 19 mg/kg per day.75  The rate of gastrointestinal adverse events ranged between 1% and 30%.50,65,67,69,71,74,77,81  Some studies also reported dermatological (hyperpigmentation, alopecia, maculopapular rash, or facial erythema)65,69  and neurological (headache or dizziness)65,69  adverse events on long-term therapy, although others did not observe such symptoms or attributed them to other disease-related risk factors.67,74,104  No renal or hepatic side effects were reported with hydroxyurea therapy.67,69,71,74,75,82,85  Although some reports suggested that hydroxyurea may adversely affect gonadal function,87  others failed to document such an association even on long-term therapy.105  Interestingly, 2 patients became pregnant while on hydroxyurea and delivered normally, without any congenital malformations in the infants.72 

In the middle of the 1980s, as the initial clinical trials of 5-azacytidine and hydroxyurea were being reported, Perrine et al,106  as well as Bard et al,107  reported that the infants of diabetic mothers have a delayed fetal-to-adult hemoglobin switch. While the exact underlying mechanisms were not clear, hypotheses were put forth about potential mechanisms. Because it was known that hydroxybutyrate is elevated in mothers with diabetes, Perrine and colleagues108  tested the idea that butyrate or other similar short-chain fatty acids may be effective as inducers of fetal hemoglobin in sheep. An initial trial involving a 2- to 3-week infusion of arginine butyrate (at a dose of 500 mg/kg per day) in 3 SCD and 3 β-thalassemia patients (2 who were transfusion dependent) showed promise. Nearly all the patients in the trial showed an increase (2- to 6-fold) in γ-mRNA levels and in the synthesis of this globin chain, leading to improvement in the globin chain ratios.109,110  The 2 transfusion-dependent patients showed lower levels of plasma-free hemoglobin, which is an indicator of ineffective erythropoiesis. In addition, 1 of these patients showed a steady rise in total hemoglobin level from 45 to >100 g/L over the course of 7 weeks of therapy. No other markers of disease progression were measured, and no further assessment of ineffective erythropoiesis was done. A follow-up trial extended such therapy to 9 to 13 weeks, with doses of arginine butyrate escalating from 500 to 2000 mg/kg per day for 6 days per week, and it studied the responses in 5 SCD and 5 β-thalassemia patients.111  This trial had primary end points involving hematologic responses, as defined by an increase in total hemoglobin concentration of 20 g/L in patients with β-thalassemia. The 5 β-thalassemia patients had variable transfusion requirements, with some being transfusion independent (2 patients) and others requiring intermittent (2 patients) or regular transfusions (1 patient). Unfortunately, the primary hematologic end points were not achieved in this longer-term trial. It has been suggested that the loss of response to butyrates over long-term therapy may be a result of the antiproliferative effects on the bone marrow.112  This hypothesis is supported by data confirming that intermittent or pulse-butyrate therapy is associated with well-tolerated, marked, and sustained response in patients with SCD.112  A separate cohort study of oral sodium phenylbutyrate therapy at a dose of 20 g per day over the course of 41 to 460 days showed that 4 of 11 β-thalassemia patients had increased levels of total hemoglobin >10 g/L (mean increase: 21 g/L), along with an increased production of HbF.113  All these responders had higher average levels of erythropoietin at baseline (all >120 mU/mL) and were transfusion independent. Changes in the percent of HbF, absolute HbF levels, or α/non–α-globin ratios did not correlate with response to treatment, nor did the β-genotype. In addition, the reduction of markers of ineffective erythropoiesis and hemolysis, including lactate dehydrogenase and indirect bilirubin, was also noted in the patients who showed responses. Two cohort studies have examined the efficacy of the oral butyrate derivative isobutyramide to induce HbF. The first study was an open-label, phase 2 trial on a group of 12 patients with transfusion-independent β-thalassemia intermedia (mean age: 31 years) for a period of 28 days.114  There was some increase in the percent of HbF following treatment with 150 mg/kg per day of isobutyramide (P = .06, with wide variability among patients). No change in globin chain imbalance or in markers of ineffective erythropoiesis was noted in this study. Another study examined 8 patients with transfusion-dependent β-thalassemia and involved treatment of these patients with 350 mg/kg per day of oral isobutyramide for 126 to 384 days.115  HbF increased from 3% to 6%, while a drop in plasma-free hemoglobin was noted. Two of the 8 patients showed a decreased frequency of transfusion requirements in the setting of receiving this treatment, and a reduction in iron burden was also noted in some patients. Response to treatment was associated with high pretreatment HbF (>4.5%), high parental HbF, and increased erythropoietin levels. Treatment with short-chain fatty acids was generally tolerable in published trials, with side effects being minimal or limited to gastrointestinal disturbances. It has been suggested that the relatively poor response to butyrate therapy in patients with β-thalassemia compared with patients with SCD may be attributed to the effects of these agents on other globin genes. It was shown that butyrate exposure increases α-globin expression in progenitor-derived erythroid cells from patients with β-thalassemia, while it decreases α-globin mRNA levels in patients with SCD. Thus, the favorable effects of the butyrate-induced increase in γ-globin expression on α/β-chain imbalance in β-thalassemia may be reduced as a result of the associated increase in α-globin expression.116 

Trials of other derivatives of these agents, such as 2,2-dimethylbutyrate, were reported in 2012 in patients with SCD, and data from patients with β-thalassemia are awaited.117  These short-chain fatty acids are thought to work as inhibitors of HDACs, and specific inhibitors of these molecules have been produced, which may also be promising for future trials aimed at inducing HbF.10,16 

The use of recombinant human erythropoietin or the newer erythropoietic-stimulating agent darbepoetin alfa in patients with β-thalassemia is associated with increases in total hemoglobin level.118-123  A correlation between serum erythropoietin and HbF levels also exists.124  Earlier studies from primates and from patients with SCD suggested that erythropoietin augments the HbF response attributed to hydroxyurea therapy.125-127  More-prominent effects were also noted when hydroxyurea was combined with sodium phenylbutyrate.126  Consequently, several trials evaluated the value of combinations of these 3 agents for the management of β-thalassemia patients. Loukopoulos et al128  demonstrated that the combination of hydroxyurea and erythropoietin (50 000 U 3 times a week) is associated with higher increments in total hemoglobin level than hydroxyurea alone (17 vs 2 g/L after 6 months of therapy) in patients with β-thalassemia intermedia. Lower erythropoietin doses (10 000 U) did not achieve such effects. Long-term transfusion independence in a β-thalassemia major patient was also reported with this combination.129  In the late 1990s, Olivieri et al90  reported 2 β-thalassemia major siblings (homozygous for hemoglobin Lepore) who showed remarkable total hemoglobin responses and transfusion independence from HbF induction with the combination of sodium phenylbutyrate and hydroxyurea. However, further observations in this direction were not always promising. In 2 patients with β-thalassemia intermedia, the addition of sodium phenylbutyrate to hydroxyurea treatment failed to produce an increase in total hemoglobin, despite increasing HbF levels.64  Similarly, in another study on 45 patients with hemoglobin E/β-thalassemia treated with hydroxyurea, the addition of sodium phenylbutyrate had no benefit, while the addition of erythropoietin did benefit selected patients.85  In addition, there may exist some bias in the literature toward the reporting of positive results.

Thalidomide, a drug known for its immunomodulating and anti-angiogenic properties, has been suggested to induce γ-globin gene expression and to increase the proliferation of erythroid cells using in vitro culture models.130  Two case reports reported that thalidomide therapy at 75 to 100 mg/kg per day caused a progressive and rapid increase in total hemoglobin and HbF levels in β-thalassemia major patients.131,132  Promising roles of thalidomide derivatives (pomalidomide and lenalidomide) for the induction of HbF also were reported in 2008 and 2011 from in vitro and animal studies.133,134  These findings support the evaluation of such agents as a potential new therapy that may not have the cytotoxic side effects associated with other HbF inducers.

For HbF induction therapy to become part of the standard management for patients with β-thalassemia, there needs to be a great deal of work from both basic scientists and clinical researchers. Ideally, efforts to understand the exact mechanisms through which current and future agents exert their effects should go in parallel with the establishment of large randomized clinical trials and formal clinical development programs evaluating the efficacy and safety of these agents.

Several lessons can be learned from the pitfalls and successes of the available studies (Table 2), which should help the design of future trials in β-thalassemia cohorts. We herein provide some recommendations.

Table 2

Summary of findings and limitations of fetal hemoglobin inducer studies in patients with β-thalassemia

AgentMain positive findingsLimitations
DNA methylation inhibitors   
 5-azacytidine Marked hematological responses achieved. Few studies. 
 Small sample sizes. 
 Safety concerns. 
 Decitabine Hematological responses achieved. Few studies. 
Favorable effects on red cell indices noted. Small sample sizes. 
Treatment was well tolerated.  
Hydroxyurea Hematological responses achieved. Heterogonous phenotypes studied together. 
Favorable effects on red cell, hemolysis, and hypercoagulability indices noted. Heterogeneous study end points evaluated together. 
Favorable effects on clinical morbidities noted. Ideal dose and duration of therapy still controversial. 
Treatment was well tolerated. Lack of efficacy on long-term therapy. 
 Data on predictors of response still inconsistent. 
Short-chain fatty acids Hematological responses achieved. Small sample sizes. 
Favorable effects on red cell and hemolysis indices noted. Lack of efficacy on long-term therapy. 
Treatment was well tolerated.  
Erythropoietic-stimulating agents Hematological responses achieved. Few studies. 
Favorable effects on combination with hydroxyurea noted. Small sample sizes. 
Treatment was well tolerated. High doses required. 
 No additive effects with short-chain fatty acids. 
Thalidomide and derivatives Hematological responses achieved.Treatment was well tolerated. Few studies.Small sample sizes. 
AgentMain positive findingsLimitations
DNA methylation inhibitors   
 5-azacytidine Marked hematological responses achieved. Few studies. 
 Small sample sizes. 
 Safety concerns. 
 Decitabine Hematological responses achieved. Few studies. 
Favorable effects on red cell indices noted. Small sample sizes. 
Treatment was well tolerated.  
Hydroxyurea Hematological responses achieved. Heterogonous phenotypes studied together. 
Favorable effects on red cell, hemolysis, and hypercoagulability indices noted. Heterogeneous study end points evaluated together. 
Favorable effects on clinical morbidities noted. Ideal dose and duration of therapy still controversial. 
Treatment was well tolerated. Lack of efficacy on long-term therapy. 
 Data on predictors of response still inconsistent. 
Short-chain fatty acids Hematological responses achieved. Small sample sizes. 
Favorable effects on red cell and hemolysis indices noted. Lack of efficacy on long-term therapy. 
Treatment was well tolerated.  
Erythropoietic-stimulating agents Hematological responses achieved. Few studies. 
Favorable effects on combination with hydroxyurea noted. Small sample sizes. 
Treatment was well tolerated. High doses required. 
 No additive effects with short-chain fatty acids. 
Thalidomide and derivatives Hematological responses achieved.Treatment was well tolerated. Few studies.Small sample sizes. 

Study population

One of the main limitations in previous studies is the inclusion of a heterogeneous group of patients, especially with regard to transfusion dependence, which precludes the interpretation of study outcomes. Although there exists a variety of genotypes that can lead to a β-thalassemia syndrome, the distinction of various phenotypes is primarily attained through clinical parameters.1,135  As the hallmark of disease in these syndromes is ineffective erythropoiesis and subsequent anemia, transfusion dependence has classically been an essential factor in characterizing the various phenotypes and their severity. It should be noted, however, that commitment to transfusion therapy may be the consequence of physician, patient, or family preferences rather than a reflection of disease severity. For the purpose of selecting a patient cohort for inclusion in an HbF inducer trial, patients should essentially be divided into 2 distinct groups: (1) transfusion-dependent patients, and these include phenotypes where patients require regular-transfusion therapy for survival, such as β-thalassemia major or severe hemoglobin E/β-thalassemia, and (2) nontransfusion-dependent thalassemia (NTDT) patients, and these include patients not requiring regular-transfusion therapy for survival, such as β-thalassemia intermedia or mild/moderate hemoglobin E/β-thalassemia.136  Study end points for the 2 distinct groups may be dissimilar, as outlined below. One challenge, however, is that some patients with NTDT may still require occasional blood transfusions during infection or pregnancy or before surgery. They may also require more regular, yet temporary, transfusions in the case of poor growth or development during childhood or in the management of specific complications in adulthood where the benefit of transfusion therapy has been established.137-139  For this latter group, we recommend that patients should be off transfusions for at least 6 months before inclusion in an HbF inducer trial.

Study design

The ideal design would be that of a randomized, placebo-controlled trial, although data from well-conducted, single-arm, phase 2 studies would also be valuable. Once the benefit is established in an evidence-based manner, agents may be evaluated in comparative trials against each other or against other conventional therapies. HbF-inducing agents may also be trialed in combination, either together or with conventional therapies. Although, as outlined in “Primary study end points,” a reduction in the regular-transfusion requirement is a study end point in transfusion-dependent patients, the effect of a short-term treatment strategy that includes an HbF-inducing agent alongside transfusion therapy may still be worth evaluating for the management of certain morbidities in NTDT patients.87 

Primary study end points

Another challenge in the available studies is the definition of end points and response criteria, which does not allow for a systematic comparison of results. An essential question to answer is what response is expected from an HbF inducer in a β-thalassemia patient. In untreated patients with β-thalassemia, ineffective erythropoiesis and premature red cell death are the hallmarks of disease leading to chronic anemia and hypoxia, increased intestinal iron absorption, intra- and extravascular hemolysis, and a hypercoagulable state. These mechanisms collectively lead to a variety of clinical morbidities involving almost every organ system.3,140  The ineffective erythropoiesis in patients with β-thalassemia is estimated to be 10 to 20 times the normal basal erythropoietic level. Thus, the main objective of intervention would be to enable an effective supply of normal erythrocytes and partially suppress ineffective erythropoiesis and subsequent pathophysiologic mechanisms that lead to clinical morbidity. Erythroid activity decreases to 1 to 2 times normal levels with total hemoglobin values between 100 and 110 g/L, 1 to 4 times normal levels with values between 90 and 100 g/L, and 2 to 6 times normal levels with values between 86 and 90 g/L.141  Achieving a total hemoglobin level >90 g/L has been associated with significant reduction in morbidity in β-thalassemia patients,87,142  although lower levels (>70 g/L) may be equally beneficial in children with hemoglobin E/β-thalassemia who have a remarkable facility for adaptation to low hemoglobin levels.143,144  Moreover, total hemoglobin elevations of 10 to 20 g/L have been associated with improvement in disease severity in some studies.145 

In light of these observations, in patients with NTDT, response to HbF induction therapy may be defined as follows: Excellent Response, achieving an elevation in total hemoglobin level of 10 to 20 g/L and reaching a final total hemoglobin level >90 g/L; Good Response, achieving an elevation in total hemoglobin level of 10 to 20 g/L or reaching a final total hemoglobin level >90 g/L; Poor Response, achieving an elevation in total hemoglobin level <10 g/L; and No Response, no elevation in total hemoglobin level. For transfusion-dependent patients the following definitions may be used: Excellent Response, transfusion independence and reaching a pretransfusion total hemoglobin level >90 g/L; Good Response, ≥50% reduction in pretreatment transfusion requirement and reaching a pretransfusion total hemoglobin level >90 g/L; Poor Response, <50% reduction in pretreatment transfusion requirement and reaching a pretransfusion total hemoglobin level >90 g/L; and No Response, no change in transfusion requirement to reach a pretransfusion total hemoglobin level >90 g/L.

Elevations in HbF level should not be used as a primary study end point. As described earlier in this review, several studies reported discordance between changes in HbF and total hemoglobin levels. This may be a particular problem given that HbF inducers may exert their effects through pathways other than γ-chain expression.

Secondary study end points

Alongside changes in HbF level and safety measures, it would be worthwhile evaluating alterations in indices of pathophysiologic mechanisms (eg, globin chain ratios, hemolysis, iron overload, hypercoagulability), which could shed more light on the specific mechanisms of the action of HbF-inducing agents. Moreover, the evaluation of the effects of HbF inducers on the incidence of clinical morbidities (eg, pulmonary hypertension, leg ulcers, extramedullary hematopoietic pseudotumors) remains an area of extreme importance.

Dose and duration of therapy

Previous studies have mainly used 2 dosing strategies. For example, in the hydroxyurea studies reviewed in this review, most used a fixed starting dose <20 mg/kg per day, while some studies escalated the dose to the maximal tolerated dose. We recommend the latter approach for the following reasons: (1) There is a lack of pharmacokinetic or dynamic studies to identify the optimal dose in β-thalassemia patients. (2) Such studies were not always helpful in dosing hydroxyurea for patients with SCD. (3) Data on the efficacy or safety of high dosing in β-thalassemia patients is limited to single case reports or small case series. The duration of therapy should rely on the evaluated drug and the anticipated outcomes (end points). Total hemoglobin levels should generally be evaluated after 6 and 12 months of therapy. In light of the few reports, reviewed herein, that showed diminished response after 12 months of therapy, it may be necessary to reevaluate long-term response through cohort studies. This is also especially relevant in patients with β-thalassemia who show worsening of anemia and disease severity as they advance in age, suggesting that alterations in the dosing or course of management may be necessary.146  Secondary study end points can be evaluated pre- and posttherapy and, potentially, early on for markers of hemolysis and ineffective erythropoiesis. When the incidence of clinical morbidities is a study end point, it may be difficult to evaluate outcomes in short-term studies, except in cases when the trial is designed for the management of a specific morbidity.

Predictors of response

There is great controversy from the available studies on what factors predict good response to therapy. Conducting studies with large samples will allow for association analysis, and the role of the following factors should be evaluated: age, splenectomy status, β-globin genotype, α-globin genotype, molecular determinants of increased HbF production, and baseline hematologic profile (eg, total hemoglobin level, HbF level). Although through randomization the confounding effects of such risk factors are diminished, assessment through stratification remains essential.

Optimal design of future studies should assist with the improved allocation of therapy to those patients who demonstrate clear evidence of benefiting from treatment. While the efforts to induce HbF in β-thalassemia patients by using the available agents have been ongoing for over 3 decades, recent molecular studies suggest that there is great promise that more-effective targeted therapies to induce HbF can be developed. For clinical investigators interested in testing these potential therapies, many challenges lay ahead. We hope that the lessons from prior trials in this field can serve as a guide for these future efforts.

This work was not supported by a funding source.

Contribution: Conception and design: K.M.M. and V.G.S. Literature review and interpretation: K.M.M., A.T.T., M.D.C., and V.G.S. Manuscript drafting: K.M.M. and V.G.S. Manuscript review for important intellectual content: K.M.M., A.T.T., M.D.C., and V.G.S. All authors gave final approval of the manuscript for submission.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Vijay G. Sankaran, Division of Hematology/Oncology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115; e-mail: sankaran@broadinstitute.org.

1
Weatherall
 
DJ
Clegg
 
JB
The thalassaemia syndromes
2001
4th ed
Oxford
Blackwell Science
2
Sankaran
 
VG
Nathan
 
DG
Thalassemia: an overview of 50 years of clinical research.
Hematol Oncol Clin North Am
2010
, vol. 
24
 
6
(pg. 
1005
-
1020
)
3
Rund
 
D
Rachmilewitz
 
E
Beta-thalassemia.
N Engl J Med
2005
, vol. 
353
 
11
(pg. 
1135
-
1146
)
4
Musallam
 
KM
Sankaran
 
VG
Cappellini
 
MD
, et al. 
Fetal hemoglobin levels and morbidity in untransfused patients with β-thalassemia intermedia.
Blood
2012
, vol. 
119
 
2
(pg. 
364
-
367
)
5
Nuinoon
 
M
Makarasara
 
W
Mushiroda
 
T
, et al. 
A genome-wide association identified the common genetic variants influence disease severity in beta0-thalassemia/hemoglobin E.
Hum Genet
2010
, vol. 
127
 
3
(pg. 
303
-
314
)
6
Galanello
 
R
Sanna
 
S
Perseu
 
L
, et al. 
Amelioration of Sardinian beta0 thalassemia by genetic modifiers.
Blood
2009
, vol. 
114
 
18
(pg. 
3935
-
3937
)
7
Danjou
 
F
Anni
 
F
Perseu
 
L
, et al. 
Genetic modifiers of β-thalassemia and clinical severity as assessed by age at first transfusion.
Haematologica
2012
, vol. 
97
 
7
(pg. 
989
-
993
)
8
Badens
 
C
Joly
 
P
Agouti
 
I
, et al. 
Variants in genetic modifiers of β-thalassemia can help to predict the major or intermedia type of the disease.
Haematologica
2011
, vol. 
96
 
11
(pg. 
1712
-
1714
)
9
Uda
 
M
Galanello
 
R
Sanna
 
S
, et al. 
Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of beta-thalassemia.
Proc Natl Acad Sci USA
2008
, vol. 
105
 
5
(pg. 
1620
-
1625
)
10
Sankaran
 
VG
 
Targeted therapeutic strategies for fetal hemoglobin induction. Hematology Am Soc Hematol Educ Program. 2011;2011:459-465.
11
Sankaran
 
VG
Menne
 
TF
Šćepanović
 
D
, et al. 
MicroRNA-15a and -16-1 act via MYB to elevate fetal hemoglobin expression in human trisomy 13.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
4
(pg. 
1519
-
1524
)
12
Sankaran
 
VG
Menne
 
TF
Xu
 
J
, et al. 
Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A.
Science
2008
, vol. 
322
 
5909
(pg. 
1839
-
1842
)
13
Jiang
 
J
Best
 
S
Menzel
 
S
, et al. 
cMYB is involved in the regulation of fetal hemoglobin production in adults.
Blood
2006
, vol. 
108
 
3
(pg. 
1077
-
1083
)
14
Borg
 
J
Papadopoulos
 
P
Georgitsi
 
M
, et al. 
Haploinsufficiency for the erythroid transcription factor KLF1 causes hereditary persistence of fetal hemoglobin.
Nat Genet
2010
, vol. 
42
 
9
(pg. 
801
-
805
)
15
Zhou
 
D
Liu
 
K
Sun
 
CW
, et al. 
KLF1 regulates BCL11A expression and gamma- to beta-globin gene switching.
Nat Genet
2010
, vol. 
42
 
9
(pg. 
742
-
744
)
16
Bradner
 
JE
Mak
 
R
Tanguturi
 
SK
, et al. 
Chemical genetic strategy identifies histone deacetylase 1 (HDAC1) and HDAC2 as therapeutic targets in sickle cell disease.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
28
(pg. 
12617
-
12622
)
17
van der Ploeg
 
LH
Flavell
 
RA
DNA methylation in the human gamma delta beta-globin locus in erythroid and nonerythroid tissues.
Cell
1980
, vol. 
19
 
4
(pg. 
947
-
958
)
18
Groudine
 
M
Eisenman
 
R
Weintraub
 
H
Chromatin structure of endogenous retroviral genes and activation by an inhibitor of DNA methylation.
Nature
1981
, vol. 
292
 
5821
(pg. 
311
-
317
)
19
Jones
 
PA
Taylor
 
SM
Cellular differentiation, cytidine analogs and DNA methylation.
Cell
1980
, vol. 
20
 
1
(pg. 
85
-
93
)
20
DeSimone
 
J
Heller
 
P
Hall
 
L
, et al. 
5-Azacytidine stimulates fetal hemoglobin synthesis in anemic baboons.
Proc Natl Acad Sci USA
1982
, vol. 
79
 
14
(pg. 
4428
-
4431
)
21
Ley
 
TJ
DeSimone
 
J
Anagnou
 
NP
, et al. 
5-azacytidine selectively increases gamma-globin synthesis in a patient with beta+ thalassemia.
N Engl J Med
1982
, vol. 
307
 
24
(pg. 
1469
-
1475
)
22
Ley
 
TJ
Anagnou
 
NP
Young
 
NS
, et al. 
5-azacytidine for beta thalassaemia?
Lancet
1983
, vol. 
1
 
8322
pg. 
467
 
23
5-azacytidine for beta-thalassaemia? Lancet. 1983;1(8314-5):36-37.
24
Dunbar
 
C
Travis
 
W
Kan
 
YW
, et al. 
5-Azacytidine treatment in a beta (0)-thalassaemic patient unable to be transfused due to multiple alloantibodies.
Br J Haematol
1989
, vol. 
72
 
3
(pg. 
467
-
468
)
25
Lowrey
 
CH
Nienhuis
 
AW
Brief report: treatment with azacitidine of patients with end-stage beta-thalassemia.
N Engl J Med
1993
, vol. 
329
 
12
(pg. 
845
-
848
)
26
Dover
 
GJ
Charache
 
S
Boyer
 
SH
, et al. 
5-Azacytidine increases HbF production and reduces anemia in sickle cell disease: dose-response analysis of subcutaneous and oral dosage regimens.
Blood
1985
, vol. 
66
 
3
(pg. 
527
-
532
)
27
Charache
 
S
Dover
 
G
Smith
 
K
, et al. 
Treatment of sickle cell anemia with 5-azacytidine results in increased fetal hemoglobin production and is associated with nonrandom hypomethylation of DNA around the gamma-delta-beta-globin gene complex.
Proc Natl Acad Sci USA
1983
, vol. 
80
 
15
(pg. 
4842
-
4846
)
28
Dover
 
GJ
Charache
 
SH
Boyer
 
SH
, et al. 
 
5-Azacytidine increases fetal hemoglobin production in a patient with sickle cell disease. Prog Clin Biol Res. 1983;134:475-488.
29
Ley
 
TJ
DeSimone
 
J
Noguchi
 
CT
, et al. 
5-Azacytidine increases gamma-globin synthesis and reduces the proportion of dense cells in patients with sickle cell anemia.
Blood
1983
, vol. 
62
 
2
(pg. 
370
-
380
)
30
Mabaera
 
R
Greene
 
MR
Richardson
 
CA
, et al. 
Neither DNA hypomethylation nor changes in the kinetics of erythroid differentiation explain 5-azacytidine’s ability to induce human fetal hemoglobin.
Blood
2008
, vol. 
111
 
1
(pg. 
411
-
420
)
31
Saunthararajah
 
Y
Hillery
 
CA
Lavelle
 
D
, et al. 
Effects of 5-aza-2′-deoxycytidine on fetal hemoglobin levels, red cell adhesion, and hematopoietic differentiation in patients with sickle cell disease.
Blood
2003
, vol. 
102
 
12
(pg. 
3865
-
3870
)
32
Koshy
 
M
Dorn
 
L
Bressler
 
L
, et al. 
2-deoxy 5-azacytidine and fetal hemoglobin induction in sickle cell anemia.
Blood
2000
, vol. 
96
 
7
(pg. 
2379
-
2384
)
33
DeSimone
 
J
Koshy
 
M
Dorn
 
L
, et al. 
Maintenance of elevated fetal hemoglobin levels by decitabine during dose interval treatment of sickle cell anemia.
Blood
2002
, vol. 
99
 
11
(pg. 
3905
-
3908
)
34
Saunthararajah
 
Y
Molokie
 
R
Saraf
 
S
, et al. 
Clinical effectiveness of decitabine in severe sickle cell disease.
Br J Haematol
2008
, vol. 
141
 
1
(pg. 
126
-
129
)
35
Olivieri
 
NF
Saunthararajah
 
Y
Thayalasuthan
 
V
, et al. 
Thalassemia Clinical Research Network
A pilot study of subcutaneous decitabine in β-thalassemia intermedia.
Blood
2011
, vol. 
118
 
10
(pg. 
2708
-
2711
)
36
Campbell
 
PJ
Green
 
AR
The myeloproliferative disorders.
N Engl J Med
2006
, vol. 
355
 
23
(pg. 
2452
-
2466
)
37
Romanelli
 
F
Hoven
 
AD
Use of virostatics as a means of targeting human immunodeficiency virus infection.
Curr Pharm Des
2006
, vol. 
12
 
9
(pg. 
1121
-
1127
)
38
Platt
 
OS
Orkin
 
SH
Dover
 
G
, et al. 
Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia.
J Clin Invest
1984
, vol. 
74
 
2
(pg. 
652
-
656
)
39
Mabaera
 
R
West
 
RJ
Conine
 
SJ
, et al. 
A cell stress signaling model of fetal hemoglobin induction: what doesn’t kill red blood cells may make them stronger.
Exp Hematol
2008
, vol. 
36
 
9
(pg. 
1057
-
1072
)
40
Cokic
 
VP
Smith
 
RD
Beleslin-Cokic
 
BB
, et al. 
Hydroxyurea induces fetal hemoglobin by the nitric oxide-dependent activation of soluble guanylyl cyclase.
J Clin Invest
2003
, vol. 
111
 
2
(pg. 
231
-
239
)
41
Cokic
 
VP
Andric
 
SA
Stojilkovic
 
SS
, et al. 
Hydroxyurea nitrosylates and activates soluble guanylyl cyclase in human erythroid cells.
Blood
2008
, vol. 
111
 
3
(pg. 
1117
-
1123
)
42
Ikuta
 
T
Ausenda
 
S
Cappellini
 
MD
Mechanism for fetal globin gene expression: role of the soluble guanylate cyclase-cGMP-dependent protein kinase pathway.
Proc Natl Acad Sci USA
2001
, vol. 
98
 
4
(pg. 
1847
-
1852
)
43
McGann
 
PT
Ware
 
RE
Hydroxyurea for sickle cell anemia: what have we learned and what questions still remain?
Curr Opin Hematol
2011
, vol. 
18
 
3
(pg. 
158
-
165
)
44
Italia
 
KY
Jijina
 
FJ
Merchant
 
R
, et al. 
Response to hydroxyurea in beta thalassemia major and intermedia: experience in western India.
Clin Chim Acta
2009
, vol. 
407
 
1-2
(pg. 
10
-
15
)
45
Calzolari
 
R
Pecoraro
 
A
Borruso
 
V
, et al. 
Induction of gamma-globin gene transcription by hydroxycarbamide in primary erythroid cell cultures from Lepore patients.
Br J Haematol
2008
, vol. 
141
 
5
(pg. 
720
-
727
)
46
Watanapokasin
 
R
Sanmund
 
D
Winichagoon
 
P
, et al. 
Hydroxyurea responses and fetal hemoglobin induction in beta-thalassemia/HbE patients’ peripheral blood erythroid cell culture.
Ann Hematol
2006
, vol. 
85
 
3
(pg. 
164
-
169
)
47
Watanapokasin
 
Y
Chuncharunee
 
S
Sanmund
 
D
, et al. 
In vivo and in vitro studies of fetal hemoglobin induction by hydroxyurea in beta-thalassemia/hemoglobin E patients.
Exp Hematol
2005
, vol. 
33
 
12
(pg. 
1486
-
1492
)
48
Fibach
 
E
Burke
 
LP
Schechter
 
AN
, et al. 
Hydroxyurea increases fetal hemoglobin in cultured erythroid cells derived from normal individuals and patients with sickle cell anemia or beta-thalassemia.
Blood
1993
, vol. 
81
 
6
(pg. 
1630
-
1635
)
49
Fucharoen
 
S
Siritanaratkul
 
N
Winichagoon
 
P
, et al. 
Hydroxyurea increases hemoglobin F levels and improves the effectiveness of erythropoiesis in beta-thalassemia/hemoglobin E disease.
Blood
1996
, vol. 
87
 
3
(pg. 
887
-
892
)
50
Rigano
 
P
Pecoraro
 
A
Calzolari
 
R
, et al. 
Desensitization to hydroxycarbamide following long-term treatment of thalassaemia intermedia as observed in vivo and in primary erythroid cultures from treated patients.
Br J Haematol
2010
, vol. 
151
 
5
(pg. 
509
-
515
)
51
Zeng
 
YT
Huang
 
SZ
Ren
 
ZR
, et al. 
Hydroxyurea therapy in beta-thalassaemia intermedia: improvement in haematological parameters due to enhanced beta-globin synthesis.
Br J Haematol
1995
, vol. 
90
 
3
(pg. 
557
-
563
)
52
Sauvage
 
C
Rouyer-Fessard
 
P
Beuzard
 
Y
Improvement of mouse beta thalassaemia by hydroxyurea.
Br J Haematol
1993
, vol. 
84
 
3
(pg. 
492
-
496
)
53
Ware
 
RE
How I use hydroxyurea to treat young patients with sickle cell anemia.
Blood
2010
, vol. 
115
 
26
(pg. 
5300
-
5311
)
54
Singer
 
ST
Vichinsky
 
EP
Larkin
 
S
, et al. 
E/beta Thalassemia Study Group
Hydroxycarbamide-induced changes in E/beta thalassemia red blood cells.
Am J Hematol
2008
, vol. 
83
 
11
(pg. 
842
-
845
)
55
Orringer
 
EP
Blythe
 
DS
Johnson
 
AE
, et al. 
Effects of hydroxyurea on hemoglobin F and water content in the red blood cells of dogs and of patients with sickle cell anemia.
Blood
1991
, vol. 
78
 
1
(pg. 
212
-
216
)
56
Ataga
 
KI
Cappellini
 
MD
Rachmilewitz
 
EA
Beta-thalassaemia and sickle cell anaemia as paradigms of hypercoagulability.
Br J Haematol
2007
, vol. 
139
 
1
(pg. 
3
-
13
)
57
Musallam
 
KM
Taher
 
AT
Thrombosis in thalassemia: why are we so concerned?
Hemoglobin
2011
, vol. 
35
 
5-6
(pg. 
503
-
510
)
58
Saxon
 
BR
Rees
 
D
Olivieri
 
NF
Regression of extramedullary haemopoiesis and augmentation of fetal haemoglobin concentration during hydroxyurea therapy in beta thalassaemia.
Br J Haematol
1998
, vol. 
101
 
3
(pg. 
416
-
419
)
59
Hajjar
 
FM
Pearson
 
HA
Pharmacologic treatment of thalassemia intermedia with hydroxyurea.
J Pediatr
1994
, vol. 
125
 
3
(pg. 
490
-
492
)
60
Arruda
 
VR
Lima
 
CS
Saad
 
ST
, et al. 
Successful use of hydroxyurea in beta-thalassemia major.
N Engl J Med
1997
, vol. 
336
 
13
pg. 
964
 
61
Rigano
 
P
Manfré
 
L
La Galla
 
R
, et al. 
Clinical and hematological response to hydroxyurea in a patient with Hb Lepore/beta-thalassemia.
Hemoglobin
1997
, vol. 
21
 
3
(pg. 
219
-
226
)
62
Styles
 
L
Lewis
 
B
Foote
 
D
, et al. 
 
Preliminary report: hydroxyurea produces significant clinical response in thalassemia intermedia. Ann N Y Acad Sci. 1998;850:461-462.
63
Saxon
 
BR
Waye
 
JS
Olivieri
 
NF
 
Increase in hemoglobin concentration during therapy with hydroxyurea in Cooley’s anemia. Ann N Y Acad Sci. 1998;850:459-460.
64
Hoppe
 
C
Vichinsky
 
E
Lewis
 
B
, et al. 
Hydroxyurea and sodium phenylbutyrate therapy in thalassemia intermedia.
Am J Hematol
1999
, vol. 
62
 
4
(pg. 
221
-
227
)
65
Karimi
 
M
Haghpanah
 
S
Farhadi
 
A
, et al. 
Genotype-phenotype relationship of patients with β-thalassemia taking hydroxyurea: a 13-year experience in Iran.
Int J Hematol
2012
, vol. 
95
 
1
(pg. 
51
-
56
)
66
Amoozgar
 
H
Farhani
 
N
Khodadadi
 
N
, et al. 
Comparative study of pulmonary circulation and myocardial function in patients with β-thalassemia intermedia with and without hydroxyurea, a case-control study.
Eur J Haematol
2011
, vol. 
87
 
1
(pg. 
61
-
67
)
67
Ansari
 
SH
Shamsi
 
TS
Ashraf
 
M
, et al. 
Efficacy of hydroxyurea in providing transfusion independence in β-thalassemia.
J Pediatr Hematol Oncol
2011
, vol. 
33
 
5
(pg. 
339
-
343
)
68
Italia
 
KY
Jijina
 
FF
Merchant
 
R
, et al. 
Effect of hydroxyurea on the transfusion requirements in patients with severe HbE-beta-thalassaemia: a genotypic and phenotypic study.
J Clin Pathol
2010
, vol. 
63
 
2
(pg. 
147
-
150
)
69
Karimi
 
M
Cohan
 
N
Mousavizadeh
 
K
, et al. 
Adverse effects of hydroxyurea in beta-thalassemia intermedia patients: 10 years’ experience.
Pediatr Hematol Oncol
2010
, vol. 
27
 
3
(pg. 
205
-
211
)
70
Ehsani
 
MA
Hedayati-Asl
 
AA
Bagheri
 
A
, et al. 
Hydroxyurea-induced hematological response in transfusion-independent beta-thalassemia intermedia: case series and review of literature [published correction appears in Pediatr Hematol Oncol. 2011;28(6):538].
Pediatr Hematol Oncol
2009
, vol. 
26
 
8
(pg. 
560
-
565
)
71
Zamani
 
F
Shakeri
 
R
Eslami
 
SM
, et al. 
Hydroxyurea therapy in 49 patients with major beta-thalassemia.
Arch Iran Med
2009
, vol. 
12
 
3
(pg. 
295
-
297
)
72
Koren
 
A
Levin
 
C
Dgany
 
O
, et al. 
Response to hydroxyurea therapy in beta-thalassemia.
Am J Hematol
2008
, vol. 
83
 
5
(pg. 
366
-
370
)
73
Mtvarelidze Z, Kvezereli-Kopadze A, Kvezereli-Kopadze M, et al. Hematologic response to hydroxyurea therapy in children with beta-thalassemia major. Georgian Med News. 2008(156):91-94.
74
Ansari
 
SH
Shamsi
 
TS
Siddiqui
 
FJ
, et al. 
Efficacy of hydroxyurea (HU) in reduction of pack red cell (PRC) transfusion requirement among children having beta-thalassemia major: Karachi HU trial (KHUT).
J Pediatr Hematol Oncol
2007
, vol. 
29
 
11
(pg. 
743
-
746
)
75
Bradai
 
M
Pissard
 
S
Abad
 
MT
, et al. 
Decreased transfusion needs associated with hydroxyurea therapy in Algerian patients with thalassemia major or intermedia.
Transfusion
2007
, vol. 
47
 
10
(pg. 
1830
-
1836
)
76
Mancuso
 
A
Maggio
 
A
Renda
 
D
, et al. 
Treatment with hydroxycarbamide for intermedia thalassaemia: decrease of efficacy in some patients during long-term follow up.
Br J Haematol
2006
, vol. 
133
 
1
(pg. 
105
-
106
)
77
Dixit
 
A
Chatterjee
 
TC
Mishra
 
P
, et al. 
Hydroxyurea in thalassemia intermedia—a promising therapy.
Ann Hematol
2005
, vol. 
84
 
7
(pg. 
441
-
446
)
78
Karimi
 
M
Darzi
 
H
Yavarian
 
M
Hematologic and clinical responses of thalassemia intermedia patients to hydroxyurea during 6 years of therapy in Iran.
J Pediatr Hematol Oncol
2005
, vol. 
27
 
7
(pg. 
380
-
385
)
79
Alebouyeh
 
M
Moussavi
 
F
Haddad-Deylami
 
H
, et al. 
Hydroxyurea in the treatment of major beta-thalassemia and importance of genetic screening.
Ann Hematol
2004
, vol. 
83
 
7
(pg. 
430
-
433
)
80
Yavarian
 
M
Karimi
 
M
Bakker
 
E
, et al. 
Response to hydroxyurea treatment in Iranian transfusion-dependent beta-thalassemia patients.
Haematologica
2004
, vol. 
89
 
10
(pg. 
1172
-
1178
)
81
Bradai
 
M
Abad
 
MT
Pissard
 
S
, et al. 
Hydroxyurea can eliminate transfusion requirements in children with severe beta-thalassemia.
Blood
2003
, vol. 
102
 
4
(pg. 
1529
-
1530
)
82
de Paula
 
EV
Lima
 
CS
Arruda
 
VR
, et al. 
Long-term hydroxyurea therapy in beta-thalassaemia patients.
Eur J Haematol
2003
, vol. 
70
 
3
(pg. 
151
-
155
)
83
Choudhry
 
VP
Lal
 
A
Pati
 
HP
, et al. 
Hematological responses to hydroxyurea therapy in multitransfused thalassemic children.
Indian J Pediatr
1997
, vol. 
64
 
3
(pg. 
395
-
398
)
84
Taher
 
A
Sheikh-Taha
 
M
Hydroxyurea use in Lebanese patients with beta-thalassemia intermedia.
J Pediatr Hematol Oncol
2006
, vol. 
28
 
2
pg. 
107
 
85
Singer
 
ST
Kuypers
 
FA
Olivieri
 
NF
, et al. 
E/beta Thalassaemia Study Group
Fetal haemoglobin augmentation in E/beta(0) thalassaemia: clinical and haematological outcome.
Br J Haematol
2005
, vol. 
131
 
3
(pg. 
378
-
388
)
86
Amoyal
 
I
Goldfarb
 
A
Fibach
 
E
Flow cytometric analysis of hydroxyurea effects on fetal hemoglobin production in cultures of beta-thalassemia erythroid precursors.
Hemoglobin
2003
, vol. 
27
 
2
(pg. 
77
-
87
)
87
Taher
 
AT
Musallam
 
KM
Karimi
 
M
, et al. 
Overview on practices in thalassemia intermedia management aiming for lowering complication rates across a region of endemicity: the OPTIMAL CARE study.
Blood
2010
, vol. 
115
 
10
(pg. 
1886
-
1892
)
88
Musallam
 
KM
Taher
 
AT
Karimi
 
M
, et al. 
Cerebral infarction in β-thalassemia intermedia: breaking the silence.
Thromb Res
2012
, vol. 
130
 
5
(pg. 
695
-
702
)
89
Ware
 
RE
Helms
 
RW
SWiTCH Investigators
Stroke With Transfusions Changing to Hydroxyurea (SWiTCH).
Blood
2012
, vol. 
119
 
17
(pg. 
3925
-
3932
)
90
Olivieri
 
NF
Rees
 
DC
Ginder
 
GD
, et al. 
Treatment of thalassaemia major with phenylbutyrate and hydroxyurea.
Lancet
1997
, vol. 
350
 
9076
(pg. 
491
-
492
)
91
Voskaridou
 
E
Kalotychou
 
V
Loukopoulos
 
D
Clinical and laboratory effects of long-term administration of hydroxyurea to patients with sickle-cell/beta-thalassaemia.
Br J Haematol
1995
, vol. 
89
 
3
(pg. 
479
-
484
)
92
Loukopoulos
 
D
Voskaridou
 
E
Kalotychou
 
V
, et al. 
Reduction of the clinical severity of sickle cell/beta-thalassemia with hydroxyurea: the experience of a single center in Greece.
Blood Cells Mol Dis
2000
, vol. 
26
 
5
(pg. 
453
-
466
)
93
Bincoletto
 
C
Perlingeiro
 
RC
Saad
 
ST
, et al. 
Hydroxyurea promotes the reduction of spontaneous BFU-e to normal levels in SS and S/beta thalassemic patients.
Hemoglobin
2001
, vol. 
25
 
1
(pg. 
1
-
7
)
94
Rigano
 
P
Rodgers
 
GP
Renda
 
D
, et al. 
Clinical and hematological responses to hydroxyurea in Sicilian patients with Hb S/beta-thalassemia.
Hemoglobin
2001
, vol. 
25
 
1
(pg. 
9
-
17
)
95
Koren
 
A
Segal-Kupershmit
 
D
Zalman
 
L
, et al. 
Effect of hydroxyurea in sickle cell anemia: a clinical trial in children and teenagers with severe sickle cell anemia and sickle cell beta-thalassemia.
Pediatr Hematol Oncol
1999
, vol. 
16
 
3
(pg. 
221
-
232
)
96
Panigrahi
 
I
Dixit
 
A
Arora
 
S
, et al. 
Do alpha deletions influence hydroxyurea response in thalassemia intermedia?
Hematology
2005
, vol. 
10
 
1
(pg. 
61
-
63
)
97
Banan
 
M
Bayat
 
H
Azarkeivan
 
A
, et al. 
The XmnI and BCL11A single nucleotide polymorphisms may help predict hydroxyurea response in Iranian β-thalassemia patients.
Hemoglobin
2012
, vol. 
36
 
4
(pg. 
371
-
380
)
98
Konstantinou
 
E
Pashalidis
 
I
Kolnagou
 
A
, et al. 
Interactions of hydroxycarbamide (hydroxyurea) with iron and copper: implications on toxicity and therapeutic strategies.
Hemoglobin
2011
, vol. 
35
 
3
(pg. 
237
-
246
)
99
Karimi
 
M
Borzouee
 
M
Mehrabani
 
A
, et al. 
Echocardiographic finding in beta-thalassemia intermedia and major: absence of pulmonary hypertension following hydroxyurea treatment in beta-thalassemia intermedia.
Eur J Haematol
2009
, vol. 
82
 
3
(pg. 
213
-
218
)
100
Karimi
 
M
Mohammadi
 
F
Behmanesh
 
F
, et al. 
Effect of combination therapy of hydroxyurea with l-carnitine and magnesium chloride on hematologic parameters and cardiac function of patients with beta-thalassemia intermedia.
Eur J Haematol
2010
, vol. 
84
 
1
(pg. 
52
-
58
)
101
Gamberini
 
MR
Fortini
 
M
De Sanctis
 
V
 
Healing of leg ulcers with hydroxyurea in thalassaemia intermedia patients with associated endocrine complications. Pediatr Endocrinol Rev. 2004;2 (suppl 2):319-322.
102
Haidar
 
R
Mhaidli
 
H
Taher
 
AT
Paraspinal extramedullary hematopoiesis in patients with thalassemia intermedia.
Eur Spine J
2010
, vol. 
19
 
6
(pg. 
871
-
878
)
103
Felice
 
AE
Borg
 
J
Pizzuto
 
M
, et al. 
A review of cis-trans interplay between DNA sequences 5′ to the (G)gamma- and beta-globin genes among Hb F-Malta-I heterozygotes/homozygotes and beta-thalassemia homozygotes/compound heterozygotes, and the effects of hydroxyurea on the Hb F/F-erythrocyte; the need for large multicenter trials.
Hemoglobin
2007
, vol. 
31
 
2
(pg. 
279
-
288
)
104
Zargari
 
O
Kimyai-Asadi
 
A
Jafroodi
 
M
Cutaneous adverse reactions to hydroxyurea in patients with intermediate thalassemia.
Pediatr Dermatol
2004
, vol. 
21
 
6
(pg. 
633
-
635
)
105
Karimi
 
M
Zekavat
 
OR
Haghpanah
 
S
, et al. 
Comparative study of hypogonadism in beta-thalassemia intermedia patients with and without hydroxyurea.
Hematology
2012
, vol. 
17
 
2
(pg. 
122
-
124
)
106
Perrine
 
SP
Greene
 
MF
Faller
 
DV
Delay in the fetal globin switch in infants of diabetic mothers.
N Engl J Med
1985
, vol. 
312
 
6
(pg. 
334
-
338
)
107
Bard
 
H
Prosmanne
 
J
Relative rates of fetal hemoglobin and adult hemoglobin synthesis in cord blood of infants of insulin-dependent diabetic mothers.
Pediatrics
1985
, vol. 
75
 
6
(pg. 
1143
-
1147
)
108
Perrine
 
SP
Rudolph
 
A
Faller
 
DV
, et al. 
Butyrate infusions in the ovine fetus delay the biologic clock for globin gene switching.
Proc Natl Acad Sci USA
1988
, vol. 
85
 
22
(pg. 
8540
-
8542
)
109
Perrine
 
SP
Ginder
 
GD
Faller
 
DV
, et al. 
A short-term trial of butyrate to stimulate fetal-globin-gene expression in the beta-globin disorders.
N Engl J Med
1993
, vol. 
328
 
2
(pg. 
81
-
86
)
110
Perrine
 
SP
Olivieri
 
NF
Faller
 
DV
, et al. 
Butyrate derivatives. New agents for stimulating fetal globin production in the beta-globin disorders.
Am J Pediatr Hematol Oncol
1994
, vol. 
16
 
1
(pg. 
67
-
71
)
111
Sher
 
GD
Ginder
 
GD
Little
 
J
, et al. 
Extended therapy with intravenous arginine butyrate in patients with beta-hemoglobinopathies.
N Engl J Med
1995
, vol. 
332
 
24
(pg. 
1606
-
1610
)
112
Atweh
 
GF
Sutton
 
M
Nassif
 
I
, et al. 
Sustained induction of fetal hemoglobin by pulse butyrate therapy in sickle cell disease.
Blood
1999
, vol. 
93
 
6
(pg. 
1790
-
1797
)
113
Collins
 
AF
Pearson
 
HA
Giardina
 
P
, et al. 
Oral sodium phenylbutyrate therapy in homozygous beta thalassemia: a clinical trial.
Blood
1995
, vol. 
85
 
1
(pg. 
43
-
49
)
114
Domenica Cappellini
 
M
Graziadei
 
G
Ciceri
 
L
, et al. 
Oral isobutyramide therapy in patients with thalassemia intermedia: results of a phase II open study.
Blood Cells Mol Dis
2000
, vol. 
26
 
1
(pg. 
105
-
111
)
115
Reich
 
S
Bührer
 
C
Henze
 
G
, et al. 
Oral isobutyramide reduces transfusion requirements in some patients with homozygous beta-thalassemia.
Blood
2000
, vol. 
96
 
10
(pg. 
3357
-
3363
)
116
Fathallah
 
H
Taher
 
A
Bazarbachi
 
A
, et al. 
Differences in response to fetal hemoglobin induction therapy in beta-thalassemia and sickle cell disease.
Blood Cells Mol Dis
2009
, vol. 
43
 
1
(pg. 
58
-
62
)
117
Kutlar
 
A
Ataga
 
K
Reid
 
M
, et al. 
A phase 1/2 trial of HQK-1001, an oral fetal globin inducer, in sickle cell disease.
Am J Hematol
2012
, vol. 
87
 
11
(pg. 
1017
-
1021
)
118
Singer
 
ST
Vichinsky
 
EP
Sweeters
 
N
, et al. 
Darbepoetin alfa for the treatment of anaemia in alpha- or beta- thalassaemia intermedia syndromes.
Br J Haematol
2011
, vol. 
154
 
2
(pg. 
281
-
284
)
119
Rachmilewitz
 
EA
Aker
 
M
Perry
 
D
, et al. 
Sustained increase in haemoglobin and RBC following long-term administration of recombinant human erythropoietin to patients with homozygous beta-thalassaemia.
Br J Haematol
1995
, vol. 
90
 
2
(pg. 
341
-
345
)
120
Bourantas
 
K
Economou
 
G
Georgiou
 
J
Administration of high doses of recombinant human erythropoietin to patients with beta-thalassemia intermedia: a preliminary trial.
Eur J Haematol
1997
, vol. 
58
 
1
(pg. 
22
-
25
)
121
Nişli
 
G
Kavakli
 
K
Vergin
 
C
, et al. 
Recombinant human erythropoietin trial in thalassemia intermedia.
J Trop Pediatr
1996
, vol. 
42
 
6
(pg. 
330
-
334
)
122
Nisli
 
G
Kavakli
 
K
Aydinok
 
Y
, et al. 
Recombinant erythropoietin trial in children with transfusion-dependent homozygous beta-thalassemia.
Acta Haematol
1997
, vol. 
98
 
4
(pg. 
199
-
203
)
123
Chaidos
 
A
Makis
 
A
Hatzimichael
 
E
, et al. 
Treatment of beta-thalassemia patients with recombinant human erythropoietin: effect on transfusion requirements and soluble adhesion molecules.
Acta Haematol
2004
, vol. 
111
 
4
(pg. 
189
-
195
)
124
Galanello
 
R
Barella
 
S
Turco
 
MP
, et al. 
Serum erythropoietin and erythropoiesis in high- and low-fetal hemoglobin beta-thalassemia intermedia patients.
Blood
1994
, vol. 
83
 
2
(pg. 
561
-
565
)
125
Rodgers
 
GP
Dover
 
GJ
Uyesaka
 
N
, et al. 
Augmentation by erythropoietin of the fetal-hemoglobin response to hydroxyurea in sickle cell disease.
N Engl J Med
1993
, vol. 
328
 
2
(pg. 
73
-
80
)
126
McDonagh
 
KT
Dover
 
GJ
Donahue
 
RE
, et al. 
Hydroxyurea-induced HbF production in anemic primates: augmentation by erythropoietin, hematopoietic growth factors, and sodium butyrate.
Exp Hematol
1992
, vol. 
20
 
10
(pg. 
1156
-
1164
)
127
Lavelle
 
D
Molokie
 
R
Ducksworth
 
J
, et al. 
Effects of hydroxurea, stem cell factor, and erythropoietin in combination on fetal hemoglobin in the baboon.
Exp Hematol
2001
, vol. 
29
 
2
(pg. 
156
-
162
)
128
Loukopoulos
 
D
Voskaridou
 
E
Stamoulakatou
 
A
, et al. 
 
Hydroxyurea therapy in thalassemia. Ann N Y Acad Sci. 1998;850:120-128.
129
Kumar
 
M
Fallon
 
RJ
Hill
 
JS
, et al. 
Esthesioneuroblastoma in children.
J Pediatr Hematol Oncol
2002
, vol. 
24
 
6
(pg. 
482
-
487
)
130
Aerbajinai
 
W
Zhu
 
J
Gao
 
Z
, et al. 
Thalidomide induces gamma-globin gene expression through increased reactive oxygen species-mediated p38 MAPK signaling and histone H4 acetylation in adult erythropoiesis.
Blood
2007
, vol. 
110
 
8
(pg. 
2864
-
2871
)
131
Aguilar-Lopez
 
LB
Delgado-Lamas
 
JL
Rubio-Jurado
 
B
, et al. 
Thalidomide therapy in a patient with thalassemia major.
Blood Cells Mol Dis
2008
, vol. 
41
 
1
(pg. 
136
-
137
)
132
Masera
 
N
Tavecchia
 
L
Capra
 
M
, et al. 
Optimal response to thalidomide in a patient with thalassaemia major resistant to conventional therapy.
Blood Transfus
2010
, vol. 
8
 
1
(pg. 
63
-
65
)
133
Moutouh-de Parseval
 
LA
Verhelle
 
D
Glezer
 
E
, et al. 
Pomalidomide and lenalidomide regulate erythropoiesis and fetal hemoglobin production in human CD34+ cells.
J Clin Invest
2008
, vol. 
118
 
1
(pg. 
248
-
258
)
134
Meiler
 
SE
Wade
 
M
Kutlar
 
F
, et al. 
Pomalidomide augments fetal hemoglobin production without the myelosuppressive effects of hydroxyurea in transgenic sickle cell mice.
Blood
2011
, vol. 
118
 
4
(pg. 
1109
-
1112
)
135
Steinberg
 
MH
Forget
 
BG
Higgs
 
DR
, et al. 
Disorders of hemoglobin: genetics, pathophysiology, and clinical management
2009
2nd ed
New York
Cambridge University Press
136
Weatherall
 
DJ
 
The definition and epidemiology of non-transfusion-dependent thalassemia. Blood Rev. 2012;26 (suppl 1):S3-S6.
137
Taher
 
AT
Musallam
 
KM
Karimi
 
M
, et al. 
 
Contemporary approaches to treatment of beta-thalassemia intermedia. Blood Rev. 2012;26 (suppl 1):S24-S27.
138
Vichinsky
 
E
 
Advances in the treatment of alpha-thalassemia. Blood Rev. 2012;26 (suppl 1):S31-S34.
139
Olivieri
 
NF
 
Treatment strategies for hemoglobin E beta-thalassemia. Blood Rev. 2012;26 (suppl 1):S28-S30.
140
Musallam
 
KM
Taher
 
AT
Rachmilewitz
 
EA
β-Thalassemia Intermedia: A Clinical Perspective.
Cold Spring Harb Perspect Med
2012
, vol. 
2
 
7
pg. 
a013482
 
141
Cazzola
 
M
De Stefano
 
P
Ponchio
 
L
, et al. 
Relationship between transfusion regimen and suppression of erythropoiesis in beta-thalassaemia major.
Br J Haematol
1995
, vol. 
89
 
3
(pg. 
473
-
478
)
142
Cappellini
 
MD
Cohen
 
A
Eleftheriou
 
A
, et al. 
Guidelines for the clinical management of thalassemia
2009
ed 2nd rev
Nicosia, Cyprus
Thalassaemia International Federation
143
O’Donnell
 
A
Premawardhena
 
A
Arambepola
 
M
, et al. 
Age-related changes in adaptation to severe anemia in childhood in developing countries.
Proc Natl Acad Sci USA
2007
, vol. 
104
 
22
(pg. 
9440
-
9444
)
144
Allen
 
A
Fisher
 
C
Premawardhena
 
A
, et al. 
Adaptation to anemia in hemoglobin E-ß thalassemia.
Blood
2010
, vol. 
116
 
24
(pg. 
5368
-
5370
)
145
Sripichai
 
O
Makarasara
 
W
Munkongdee
 
T
, et al. 
A scoring system for the classification of beta-thalassemia/Hb E disease severity.
Am J Hematol
2008
, vol. 
83
 
6
(pg. 
482
-
484
)
146
Taher
 
AT
Musallam
 
KM
El-Beshlawy
 
A
, et al. 
Age-related complications in treatment-naïve patients with thalassaemia intermedia.
Br J Haematol
2010
, vol. 
150
 
4
(pg. 
486
-
489
)
Sign in via your Institution