THE LAST 3 decades have witnessed profound changes in the management of patients with thalassemia major. Regular red blood cell (RBC) transfusions eliminate the complications of anemia and compensatory bone marrow (BM) expansion, permit normal development throughout childhood, and extend survival.1 In parallel, transfusions result in a “second disease” while treating the first,2 that of the inexorable accumulation of tissue iron that, without treatment, is fatal in the second decade of life. Further altering the prognosis of thalassemia major over the last 20 years has been progress in the development of iron-chelating therapy for iron overload. Deferoxamine mesylate, first introduced in short-term studies in iron-loaded patients in the early 1960s, gained acceptance as standard therapy over a decade later in countries able to support the high costs of this therapy. Twenty years later, extended survival free of iron-induced complications, and dramatically improved quality of life, are observed in well-chelated patients. Indeed, over this period, iron-chelating therapy for thalassemia major has resulted in one of the most dramatic alterations in morbidity and mortality associated with a genetic disease. In this review the experience gained in the use of deferoxamine, the benefits of and problems associated with this agent in the treatment and prevention of iron overload, and recent progress in the development of orally effective iron-chelating drugs will be reviewed.

Adjuncts to the use of chelating therapy to reduce iron accumulation in patients with thalassemia major include the judicious use of transfusion to minimize iron loading while adequately suppressing endogenous erythropoiesis, the appropriate timing of splenectomy to minimize administration of transfusions, and the specific therapy of complications that may result from iron-induced organ damage. These will be briefly reviewed here.

Transfusion Regimens

Untransfused children with homozygous β-thalassemia usually exhibit some or all of the complications of anemia and ineffective erthropoiesis1; the prevention of these complications is the goal of regular transfusions.2-5 The transfusion regimen itself appears critical in the control of body iron loading. Maintenance of a regimen in which pretransfusion hemoglobin concentrations do not exceed 9.5 g/dL has been shown to result in a reduced transfusion requirement and improved control of body iron burden,6,7 compared with a transfusion schedule (termed “supertransfusion”) in which baseline hemoglobins are permitted to exceed 11 g/dL.8 

Type of RBC Concentrates

Early studies of the use of neocytes, or young RBCs, predicted that prolonged survival in vivo of these concentrates should reduce the RBC mass required to maintain appropriate baseline hemoglobin concentrations.8-11 Clinical investigations confirmed that an extension of transfusion interval of 13% to 25% in thalassemia could be achieved with the use of neocytes.12-16 A recent study found that a 15% extension of transfusion interval during administration of neocyte concentrates, expected to minimally reduce the requirement for iron chelation therapy, could be achieved but at the cost of an increased exposure to donated units and a fivefold increment in preparation expenses over those of standard concentrates.16 Hence, the use of neocytes should have a limited impact on the long-term management of most chronically transfused patients. In contrast, the use of automated RBC exchange transfusions in regularly transfused patients with sickle cell disease has been reported to substantially reduce transfusional iron accumulation, permitting reduction in the intensity of chelation therapy.17 Pilot studies in thalassemia major18 suggest that a similar approach may warrant careful evaluation in this disorder.

Splenectomy

In patients with thalassemia in whom yearly transfusion requirements exceed 200 mL packed cells per kilogram body weight, splenectomy should significantly diminish RBC requirements and iron accumulation.19,20 Hypersplenism may be avoided by early and regular transfusion; many patients reaching adolescence in this decade have not required splenectomy.21 Because of the risk of postsplenectomy infection, splenectomy should generally be delayed until the age of 5 years or later.22 

Treatment of Hepatitis

Liver disease is reported as a common cause of death after age 15 years in patients with thalassemia.23 Iron-induced hepatic damage is exacerbated by a second complication of transfusions, infection with hepatitis C virus,24,25 the most frequent cause of hepatitis in thalassemic children.26 The high incidence of liver failure and hepatocellular carcinoma in patients who have acquired the virus through transfusions27 supports the use of antiviral therapy for patients with thalassemia. The results of recent trials of interferon-α in hepatitis-C–infected patients with thalassemia28,29 suggest that the clinical and pathologic responses to this agent may be inversely related to body iron burden.29 The effectiveness of antiviral therapy in thalassemia may therefore depend on that of iron-chelating therapy; such therapy should be intensified in hepatitis-C–infected patients.

Iron Overload

The most important consequence of life-saving transfusions in thalassemia is the inexorable accumulation of iron within tissues, causing progressive organ dysfunction that is fatal without chelating therapy.2 The toxicity of iron has been thoroughly reviewed previously.30 Here, the sites and toxicity of chelatable iron important in patients with thalassemia will be briefly considered.

Nontransferrin-Bound Iron

The toxicity of iron is mediated, in part, by its catalysis of reactions which generate free hydroxyl radicals, propagators of oxygen-related damage.30-32 Hydroxyl radicals induce lipid peroxidation of cellular organelles including mitochondria, lysosomes, and sarcoplasmic membranes. Evidence of peroxidant damage has been demonstrated in vivo in the tissues of iron-loaded animals33 and of thalassemic patients.34 Iron unbound to storage or transport proteins is particularly toxic in this regard; in normal individuals, tight binding of plasma iron to the transport protein transferrin prevents the catalytic activity of iron in free radical production.35,36 In very heavily iron-loaded patients, transferrin becomes fully saturated and a nontransferrin-bound fraction of iron becomes detectable in plasma.37-43 Nontransferrin-bound iron may accelerate the formation of free hydroxyl radical41 and facilitate uptake of iron by tissues.35,44 The effectiveness of an iron-chelating agent depends in part on its ability to bind nontransferrin-bound iron over sustained periods of time, thereby decreasing tissue uptake and iron-catalyzed toxic reactions.

Chelatable Tissue Iron

On delivery to cells by transferrin, iron is immediately available for chelation from a low-molecular-weight iron pool through which the intracellular traffic of iron may pass.45 When this pool is large, it may be toxic to cells with a limited capacity to generate iron storage proteins.46,47 Excess iron is deposited in reticuloendothelial cells, where it appears to be relatively harmless, or in parenchymal tissues, where it may cause significant damage.30 

Deferoxamine

Iron overload may be prevented or treated with a chelating agent capable of complexing with iron and promoting its excretion. The only iron-chelating agent presently available for clinical use is deferoxamine B, a trihydroxamic acid produced by Streptomyces pilosus, with relative specificity for ferric iron.48 Deferoxamine is poorly absorbed orally49 and rapidly metabolized in plasma,50 conferring on the drug its principal drawback: the requirement for prolonged parenteral infusions during which plasma concentrations reach a plateau at 12 hours.30 The sources of iron chelatable by deferoxamine have been thoroughly reviewed.22,30,51,52 Iron bound by deferoxamine is rendered virtually inactive metabolically and deferoxamine can prevent or reverse effects of free radical formation and lipid peroxidation in many experimental systems.33,44,53-57 

Clinical Use of Deferoxamine

Substantial iron excretion was first reported after administration of intramuscular, intravenous (IV),58,59 and subcutaneous bolus injections60 of deferoxamine in the early 1960s. A decade later, chronic intramuscular administration was shown to slow iron accumulation and arrest hepatic fibrosis in transfused patients.61 Over the next 10 years, the effectiveness of 24-hour infusions of IV62,63 and subcutaneous deferoxamine,64,65 the efficacy and feasibility of 12-hour subcutaneous infusions,66 and the substantial fecal iron excretion induced by deferoxamine67 were reported. Together, these studies permitted the design of regimens of nightly subcutaneous deferoxamine using portable ambulatory pumps.64-66 Clinical studies important in our understanding of the use and benefits of deferoxamine are outlined in Table 1.

Table 1.

Important Studies of Deferoxamine Therapy in Thalassemia

FindingDateReference
IV and intramuscular administration promotes iron excretion 1962 58, 59 
Subcutaneous administration induces iron excretion 1964 60 
Intramuscular therapy stabilizes hepatic iron, arrests hepatic fibrosis in transfused patients 1974 61 
Supplemental ascorbic acid increases deferoxamine-induced urinary iron excretion 1974 62, 248 
24-h IV infusions calculated to achieve iron balance 1976 62, 63 
24-h subcutaneous infusions calculated to achieve iron balance 1976 64, 65 
Portable infusion pumps used to administer 24-h subcutaneous infusions 1976 64, 65 
12-h infusions calculated to achieve iron balance 1978 64 
Long-term subcutaneous therapy reduces hepatic iron concentration 1981 98, 99 
Significant fecal iron excretion induced by deferoxamine 1982 66 
Long-term subcutaneous therapy reduces incidence of cardiac disease in compliant patients 1985 83 
Early therapy extends survival in young cohorts of patients 1989 23 
IV and subcutaneous therapy normalizes hepatic iron concentration 1989 102 
Regular therapy started before age 10 years reduces incidence of gonadal dysfunction 1990 137 
Long-term therapy extends survival free of glucose intolerance and cardiac disease 1994 91, 92 
FindingDateReference
IV and intramuscular administration promotes iron excretion 1962 58, 59 
Subcutaneous administration induces iron excretion 1964 60 
Intramuscular therapy stabilizes hepatic iron, arrests hepatic fibrosis in transfused patients 1974 61 
Supplemental ascorbic acid increases deferoxamine-induced urinary iron excretion 1974 62, 248 
24-h IV infusions calculated to achieve iron balance 1976 62, 63 
24-h subcutaneous infusions calculated to achieve iron balance 1976 64, 65 
Portable infusion pumps used to administer 24-h subcutaneous infusions 1976 64, 65 
12-h infusions calculated to achieve iron balance 1978 64 
Long-term subcutaneous therapy reduces hepatic iron concentration 1981 98, 99 
Significant fecal iron excretion induced by deferoxamine 1982 66 
Long-term subcutaneous therapy reduces incidence of cardiac disease in compliant patients 1985 83 
Early therapy extends survival in young cohorts of patients 1989 23 
IV and subcutaneous therapy normalizes hepatic iron concentration 1989 102 
Regular therapy started before age 10 years reduces incidence of gonadal dysfunction 1990 137 
Long-term therapy extends survival free of glucose intolerance and cardiac disease 1994 91, 92 

The Heart

In the absence of chelating therapy, myocardial disease remains the life-limiting complication of transfusional iron overload. As detailed over 30 years ago, irregularly transfused, unchelated children frequently developed left ventricular hypertrophy and conduction disturbances by late childhood, and ventricular arrhythmias and refractory congestive failure by the mid-teens.68 Within the heart, even small amounts of unbound iron may generate reactive harmful oxygen metabolites and toxicity, while both chronic pulmonary hypertension69 and myocarditis70 may accelerate iron-induced cardiac failure in thalassemia. These observations may explain the variable correlation observed between the severity of myocardial iron deposition and that of cardiac fibrosis.71,72 

The Impact of Iron-Chelating Therapy on Cardiac Disease and Survival

The beneficial effects of deferoxamine therapy on survival and cardiac disease in patients with thalassemia were first reported in the early 1980s.23,73-80 Over the subsequent decade, several studies observing reduction in morbidity and mortality examined periods of follow-up too short to provide definitive conclusions regarding the long-term benefits of deferoxamine on cardiac disease.81-90 Only in the present decade did patients who started deferoxamine in early childhood reach an age at which long-term survival could be assessed with greater certainty. Two recent trials, both of over 10 years duration, have demonstrated unequivocally that effective long-term use of deferoxamine in thalassemia major is associated with long-term survival free of the complications of iron overload.91,92 Both studies identified the magnitude of the body iron burden as the principal determinant of clinical outcome. One trial used the serum ferritin to evaluate iron loading.91 Over the period of follow-up, patients with most serum ferritin concentrations less than 2,500 μg/L had an estimated cardiac disease free survival of 91% after 15 years, in contrast to patients in whom most serum ferritin concentrations had exceeded 2,500 μg/L, who had an estimated cardiac disease free survival after 15 years of less than 20% (Fig 1). The other trial quantitatively examined the relationship between the total amount of iron administered by transfusion, the cumulative use of deferoxamine and the magnitude of the body iron burden, as assessed by measurements of hepatic iron stores.92 Using a threshold for transfused iron and deferoxamine use that is equivalent to a hepatic storage iron of about 80 μmol iron per gram liver, wet weight (about 15 mg iron per gram liver, dry weight),93 patients were classified as having received ineffective or effective chelation therapy. Ineffective chelating-therapy was associated with the greatest risk of clinical complications and early death in patients with thalassemia major; the probability of survival to at least the age of 25 years was only 32% among patients above the threshold. By contrast, effective chelation helped protect against impaired glucose tolerance, diabetes mellitus, cardiac disease, and early death; no deaths had occurred among patients below the threshold.

Fig. 1.

Survival without cardiac disease according to the proportion of serum ferritin measurements greater than 2,500 μg/dL. The circles show cardiac disease free survival among patients in whom less than 33% of serum ferritin measurements exceeded 2,500 μg/L; squares show survival among patients in whom 33% to 67% of ferritin measurements exceeded 2,500 μg/L; and triangles show survival among patients in whom more than 67% of ferritin measurements exceeded 2,500 μg/L. (Reprinted by permission of The New England Journal of Medicine, Olivieri NF, Nathan DG, MacMillan JH, et al. Volume 331, pp 574-578, 1994. Copyright 1994. Massachusetts Medical Society. All rights reserved.)91 

Fig. 1.

Survival without cardiac disease according to the proportion of serum ferritin measurements greater than 2,500 μg/dL. The circles show cardiac disease free survival among patients in whom less than 33% of serum ferritin measurements exceeded 2,500 μg/L; squares show survival among patients in whom 33% to 67% of ferritin measurements exceeded 2,500 μg/L; and triangles show survival among patients in whom more than 67% of ferritin measurements exceeded 2,500 μg/L. (Reprinted by permission of The New England Journal of Medicine, Olivieri NF, Nathan DG, MacMillan JH, et al. Volume 331, pp 574-578, 1994. Copyright 1994. Massachusetts Medical Society. All rights reserved.)91 

Close modal

The Liver

The liver is a major repository of transfused iron. Hepatic parenchymal iron accumulation, demonstrated after only 2 years of transfusion therapy,2 may rapidly result in portal fibrosis in a significant percentage of patients: one center has observed portal fibrosis in a high percentage of biopsies in children under the age of 3 years.94 In young adults with thalassemia major, in whom liver disease remains a common cause of death,23 viral infection24,25 and alcohol ingestion95 may act synergistically with iron in accelerating the development of liver damage.

The Impact of Iron-Chelating Therapy on Liver Disease

Reports of reduction in liver iron concentration, improvement in laboratory abnormalities of liver function, and arrest of hepatic fibrosis provide evidence for the beneficial effects of subcutaneous deferoxamine on iron loading within the liver.61,96-101 High-dose IV deferoxamine has been reported to achieve the same benefits in patients with massively elevated hepatic iron concentrations.102 

Endocrine Function and Growth

The most common endocrine abnormalities in patients with thalassemia in the modern era include hypogonadotropic hypogonadism, growth hormone deficiency, and diabetes mellitus.103,104 Variable incidences of hypothyroidism,105 hypoparathyrodism,106 and low levels of adrenal androgen secretion with normal glucocorticoid reserve,107 have been less commonly reported. Although normal rates of prepubertal linear growth may be observed in patients maintained on regular transfusion programs,108-110 poor pubertal growth and impaired sexual maturation have been observed in well-transfused patients.107,110-114 Poor pubertal growth has been attributed to iron-induced selective central hypogonadism,105,110,115,116 interference of iron with the production of insulin-like growth factor (IGF-1),117-119 or both. The role of iron is supported by histologic findings of selective iron deposition in pituitary gonadotropes120 and by the reversibility of hypogonadism in primary hemochromatosis with intensive phlebotomy.121,122 Poor pubertal growth has also been attributed to several other causes, including impaired growth hormone responses to growth hormone-releasing hormone,123 abnormalities in growth hormone secretion124 or in the growth hormone-receptor itself125 in the presence of normal growth hormone reserves in most patients126-128; growth may improve with administration of exogenous growth hormone.129,130 Hyposecretion of adrenal androgen,106,107 delay in pubertal development itself,131,132 zinc deficiency,133-135 and free-hemoglobin–induced inhibition of cartilage growth136 have also been implicated in impairment of growth in patients with thalassemia major.

Impact of Iron-Chelating Therapy on Endocrine Function and Growth

The effectiveness of deferoxamine in the prevention of growth failure and gonadal dysfunction was first reported in a cohort of patients regularly treated since mid-childhood, 90% of whom reached normal puberty. In contrast, in a group of patients who had administered a relatively lower total dose of deferoxamine beginning in the early teens, only 38% achieved normal pubertal status. In both cohorts, final height did not differ significantly from mid-parental height.137 In parallel, a striking increase in fertility in men and women with thalassemia has been reported over the last decade.138 These findings contrast with older,107,110,113 and some recent,139 studies in which a high incidence of gonadal dysfunction in chelated patients has been reported. While insufficient length or intensity of therapy in some of these studies almost certainly explains the lack of reported benefit of deferoxamine in the preservation of pubertal function, it is disappointing to note that secondary ammenorrhea may eventually develop in many thalassemic women with previous evidence of normal pituitary function.139 Intensive deferoxamine administration itself may be associated with impaired linear growth.140-144 

Diabetes mellitus in thalassemia has been attributed to impaired secretion of insulin secondary to chronic pancreatic iron overload,106,145-150 and to insulin resistance151-154 as a consequence of iron deposition within liver152 or skeletal muscle.155 Diabetes has also been linked temporally to episodes of acute viral hepatitis in some patients.149,150 In most studies there exists a direct relationship between the development of diabetes and the severity and duration of iron overload.150,156 Iron-induced free hydroxyl radical-induced islet cell damage, shown to induce diabetes in animals,157 may also play a role in the development of this complication in iron-loaded patients.

Impact of Iron-Chelating Therapy on Diabetes

Reduction in the risk of diabetes mellitus and glucose intolerance has been reported in patients who used more deferoxamine in relationship to their transfusional iron load, compared to a group who had begun deferoxamine at a more advanced age and had administered therapy less intensively.92 

Reversal of Iron-Induced Organ Dysfunction

Evidence that established iron-induced dysfunction of the heart78,79,85,87 and liver96-101 may improve during intensive deferoxamine therapy has been presented in several reports. Even if administration of deferoxamine does not reverse iron-induced cardiac dysfunction altogether, the outlook for patients who develop cardiac disease in the modern era but who thereafter comply with chelating therapy is strikingly improved, compared with the prognosis reported 30 years ago in similar patients.68 A recent study reported that iron-induced cardiac disease was fatal in most patients in whom body iron burdens remained high, but that extended survivals were observed in patients who had reduced iron stores, as estimated by serum ferritin concentration, 2 years after the onset of this complication.158 In patients with true “end-stage” iron-related disease, both cardiac transplantation159 and combined cardiac and liver transplantation160 has been successful in extending survival in patients with thalassemia major.

Although pituitary growth hormone reserve has been reported to improve after deferoxamine therapy in adults with acquired transfusional iron overload,84 reversal of established pituitary failure has not been reported in thalassemia major. In contrast, improvement in both thyroid function75 and glucose intolerance22 has been reported following deferoxamine treatment in this disorder.

Management of Chelation Therapy

Several practical problems are associated with long-term chelation therapy. One of the most important of these is the accurate assessment of body iron burden, essential to the evaluation of the effectiveness of deferoxamine, as well as to that of new chelators entering clinical trials. As well, issues regarding the appropriate age for the initiation of deferoxamine treatment, the maintenance of balance between its effectiveness and toxicity, and the problems of compliance with deferoxamine arise frequently in the management of patients with thalassemia.

Both direct and indirect means for the assessment of body iron are available but no single indicator or combination of indicators is ideal for the evaluation of iron status in all clinical circumstances (Table 2). Measurement of hepatic iron stores provides the most quantitative means of assessing the body iron burden in patients with thalassemia major161 and may be considered the reference method for comparison with other techniques. Data that have accumulated over the past 10 years permit a quantitative approach to the management of iron overload, and provide guidelines for the control of body iron burden in individual patients treated with chelating therapy.

Table 2.

Assessment of Body Iron Burden in Thalassemia

TestCommentsReference
Indirect:  Most tests widely available  
Serum/plasma ferritin concentration  Noninvasive 162-169 
  Lacks sensitivity and specificity  
  Poorly correlated with hepatic iron concentration in individual patients  
Serum transferrin saturation  Lacks sensitivity 163, 164 
Tests of 24-h deferoxamine-induced urinary iron excretion  Less than half of outpatient aliquots collected correctly  
  Ratio of stool:urine iron variable; poorly correlated with hepatic iron concentration  
51, 52, 67, 163, 164 
Imaging of tissue iron 
Computed tomography: Liver Variable correlation with hepatic iron concentration reported 170-174 
Magnetic resonance: Liver Variable correlations with hepatic iron concentration reported 176-191 
  Treatment-induced changes confirmed by liver biopsy 192 
 Heart Only modality available to image cardiac iron stores; changes observed during chelating therapy are consistent with reduction in cardiac iron 192, 193 
 Anterior pituitary Only modality available to image pituitary iron; signal moderately well correlated with pituitary reserve 196-198 
Evaluation of organ function  Most tests lack sensitivity and specificity; may identify established organ dysfunction 199-207 
 
Direct:  Most tests not widely available  
Cardiac iron quantitation: Biopsy  Imprecise due to inhomogeneous distribution of cardiac iron 194, 195 
Hepatic iron quantitation: Biopsy  Reference method; provides direct assessment of body iron burden, severity of fibrosis and inflammation 161, 208-218 
  Safe when performed with ultrasound guidance 221 
Superconducting susceptometry (SQUID)  Noninvasive; excellent correlation with biopsy-determined hepatic iron 209-212 
TestCommentsReference
Indirect:  Most tests widely available  
Serum/plasma ferritin concentration  Noninvasive 162-169 
  Lacks sensitivity and specificity  
  Poorly correlated with hepatic iron concentration in individual patients  
Serum transferrin saturation  Lacks sensitivity 163, 164 
Tests of 24-h deferoxamine-induced urinary iron excretion  Less than half of outpatient aliquots collected correctly  
  Ratio of stool:urine iron variable; poorly correlated with hepatic iron concentration  
51, 52, 67, 163, 164 
Imaging of tissue iron 
Computed tomography: Liver Variable correlation with hepatic iron concentration reported 170-174 
Magnetic resonance: Liver Variable correlations with hepatic iron concentration reported 176-191 
  Treatment-induced changes confirmed by liver biopsy 192 
 Heart Only modality available to image cardiac iron stores; changes observed during chelating therapy are consistent with reduction in cardiac iron 192, 193 
 Anterior pituitary Only modality available to image pituitary iron; signal moderately well correlated with pituitary reserve 196-198 
Evaluation of organ function  Most tests lack sensitivity and specificity; may identify established organ dysfunction 199-207 
 
Direct:  Most tests not widely available  
Cardiac iron quantitation: Biopsy  Imprecise due to inhomogeneous distribution of cardiac iron 194, 195 
Hepatic iron quantitation: Biopsy  Reference method; provides direct assessment of body iron burden, severity of fibrosis and inflammation 161, 208-218 
  Safe when performed with ultrasound guidance 221 
Superconducting susceptometry (SQUID)  Noninvasive; excellent correlation with biopsy-determined hepatic iron 209-212 

Indirect Assessment

Serum or plasma estimates of body iron burden.The measurement of plasma or serum ferritin is the most commonly used indirect estimate of body iron stores.52,162-166 Normally, ferritin concentrations decrease with depletion of storage iron and increase with storage iron accumulation. A maximum glycosylated plasma ferritin concentration of about 4,000 μg/L may represent the upper physiologic limit of the rate of synthesis167; higher concentrations are thought to be caused by the release of intracellular ferritin from damaged cells. Interpretation of ferritin values may be complicated by a variety of conditions that alter concentrations independently of changes in body iron burden, including ascorbate deficiency, fever, acute infection, chronic inflammation, acute and chronic hepatic damage, hemolysis, and ineffective erythropoiesis,168,169 all of which are common in thalassemia major. In one study of patients with thalassemia major or sickle cell disease, the 95% prediction intervals for hepatic iron concentration, given the plasma ferritin, were so broad as to make determination of plasma ferritin a poor predictor of body stores. As a consequence, reliance on ferritin alone can lead to inaccurate assessment of body iron burden in individual patients (Fig 2). The serum iron, transferrin, transferrin saturation, and transferrin receptor concentration do not quantitatively reflect body iron stores.

Fig. 2.

Comparison of hepatic iron and serum ferritin concentrations in patients with thalassemia major. Indirect estimation of body iron load, based on serum ferritin concentration, is compared with the reference method, direct measurement of hepatic iron concentration (by chemical analysis or magnetic-susceptibility studies) in patients with thalassemia major treated with deferiprone. Open circles denote the values determined prior to deferiprone therapy and solid circles those at the time of final analysis after 1 to 5 years of treatment. The diagonal line denotes the simple linear least-squares regression between the two variables. (Reprinted by permission of The New England Journal of Medicine, Olivieri NF, Brittenham GM, Matsui D, et al. Volume 332, pp 918-922, 1995. Copyright 1995. Massachusetts Medical Society. All rights reserved.)93 

Fig. 2.

Comparison of hepatic iron and serum ferritin concentrations in patients with thalassemia major. Indirect estimation of body iron load, based on serum ferritin concentration, is compared with the reference method, direct measurement of hepatic iron concentration (by chemical analysis or magnetic-susceptibility studies) in patients with thalassemia major treated with deferiprone. Open circles denote the values determined prior to deferiprone therapy and solid circles those at the time of final analysis after 1 to 5 years of treatment. The diagonal line denotes the simple linear least-squares regression between the two variables. (Reprinted by permission of The New England Journal of Medicine, Olivieri NF, Brittenham GM, Matsui D, et al. Volume 332, pp 918-922, 1995. Copyright 1995. Massachusetts Medical Society. All rights reserved.)93 

Close modal

Twenty-four–hour deferoxamine-induced urinary iron excretion.The usefulness of measurement of the amount of chelated iron in the urine induced by a single intramuscular dose or prolonged subcutaneous infusion of deferoxamine112 has several limitations in the accurate assessment of body iron burden. Most important is the poor correlation between urinary iron excretion and hepatic iron concentration, in part because the relative amounts of iron excreted into stool and urine vary with the dose of deferoxamine administered, body iron burden, and erythroid activity.51 Chelator-induced urinary iron excretion is also vulnerable to extraneous influences by infection, inflammation, the activity and effectiveness of erythropoiesis, extramedullary hematopoiesis, liver disease, and ascorbic acid deficiency.

Imaging of tissue iron.Computed tomography,170-174 nuclear resonance scattering (NRS) from manganese-56,175 and the most widely used modality, magnetic resonance imaging,176-193 have all been used to evaluate tissue iron stores in vitro and in vivo, but none is clinically available for the measurement of hepatic iron concentrations. Biopsy-demonstrated reductions in hepatic iron have been reflected by magnetic resonance imaging (MRI) in individual patients192 (Fig 3), but correlations between hepatic iron concentrations determined by biopsy and those estimated by magnetic resonance have varied with differences in both equipment and method. Magnetic resonance represents the only imaging method in clinical use with the potential to detect iron within the heart189,192,193 (Fig 4). Although imprecision in the quantitation of cardiac iron obtained at biopsy194,195 prevents direct correlation with values of cardiac iron estimated by MRI in humans, good correlation between MRI-derived, and biopsy-determined, cardiac iron has been observed in a thalassemic mouse model.193 Furthermore, MRI changes consistent with the reduction of cardiac iron (Fig 5) that are paralleled by improvement in cardiac function have been reported in individual patients.192 Similarly, MRI studies of the iron-loaded anterior pituitary gland196,197 have reported variations in pituitary iron that are correlated with pituitary reserve in individual patients with thalassemia.198 In summary, although many studies show that MRI can reflect the presence of, and changes in, tissue iron in vivo, this method has not been validated as one that provides measurements of tissue iron that are quantitatively equivalent to those determined at tissue biopsy.

Fig. 3.

Prussian blue stain showing (top figures) hepatic iron in hepatocytes and portal macrophages, before (left) and after (right) 9 months of chelating therapy with the orally active chelating agent deferiprone in a patient with homozygous β thalassemia. Hepatic iron concentration in the sample on left was approximately 16 mg/g dry weight liver tissue; in that on the right hepatic iron concentration was less than 2 mg/g dry weight tissue. Coronal MRI (lower figures) of hepatic iron before (left) and after (right) therapy with the orally active iron chelating agent deferiprone in the same patient. Complete absence of liver signal in the MRI on the left is compatible with significant iron deposition, while improvement in signal intensity after 9 months of therapy (right) indicates that the liver iron content is reduced compared with that of the previous study. (Reprinted with permission.192 )

Fig. 3.

Prussian blue stain showing (top figures) hepatic iron in hepatocytes and portal macrophages, before (left) and after (right) 9 months of chelating therapy with the orally active chelating agent deferiprone in a patient with homozygous β thalassemia. Hepatic iron concentration in the sample on left was approximately 16 mg/g dry weight liver tissue; in that on the right hepatic iron concentration was less than 2 mg/g dry weight tissue. Coronal MRI (lower figures) of hepatic iron before (left) and after (right) therapy with the orally active iron chelating agent deferiprone in the same patient. Complete absence of liver signal in the MRI on the left is compatible with significant iron deposition, while improvement in signal intensity after 9 months of therapy (right) indicates that the liver iron content is reduced compared with that of the previous study. (Reprinted with permission.192 )

Close modal
Fig. 6.

Hepatic iron concentrations shown are those in normal individuals (approximately 0.6 to 1.2 mg iron per gram liver, dry weight)209; concentrations observed in heterozygotes for hereditary hemochromatosis associated with normal survival free of the complications of iron overload (approximately 3.2 to 7 mg iron per gram liver, dry weight),214 designated “optimal” (see text) and considered a goal for transfusion-dependent patients in whom phlebotomy cannot safely decrease body iron burden; concentrations associated with an increased risk of iron-induced complications including hepatic fibrosis and diabetes mellitus (exceeding 7 mg iron per gram liver, dry weight)215,217,218; and concentrations associated with a greatly increased risk for iron-induced cardiac disease and early death (at or exceeding 15 mg iron per gram liver, dry weight).91 Mean hepatic iron concentrations for patients with thalassemia major studied before the availability of iron-chelating therapy,61 and those observed in homozygotes and heterozygotes for hereditary hemochromatosis.214 

Fig. 6.

Hepatic iron concentrations shown are those in normal individuals (approximately 0.6 to 1.2 mg iron per gram liver, dry weight)209; concentrations observed in heterozygotes for hereditary hemochromatosis associated with normal survival free of the complications of iron overload (approximately 3.2 to 7 mg iron per gram liver, dry weight),214 designated “optimal” (see text) and considered a goal for transfusion-dependent patients in whom phlebotomy cannot safely decrease body iron burden; concentrations associated with an increased risk of iron-induced complications including hepatic fibrosis and diabetes mellitus (exceeding 7 mg iron per gram liver, dry weight)215,217,218; and concentrations associated with a greatly increased risk for iron-induced cardiac disease and early death (at or exceeding 15 mg iron per gram liver, dry weight).91 Mean hepatic iron concentrations for patients with thalassemia major studied before the availability of iron-chelating therapy,61 and those observed in homozygotes and heterozygotes for hereditary hemochromatosis.214 

Close modal
Fig. 4.

Sagittal MRI of the heart in three patients with homozygous β thalassemia and transfusional iron overload. (A, left) Normal signal from the septum (long arrow) and posterior wall of the heart, consistent with the presence of very mild cardiac iron loading, in a transfused patient regularly complianct with iron chelating therapy. The homogenous signal of the liver, consistent with very mild iron loading in this organ (short arrow), is also seen below the image of the heart. (B, middle): Imhomogenity of signal from the septum (long arrow) and posterior wall, consistent with moderate iron deposition in a transfused patient erratically compliant with iron chelating therapy. Loss of liver signal (short arrow) is consistent with heavier iron loading in this organ. (C, right): Absence of signal from the septum (arrow), posterior wall and liver (short arrow), compatible with heavy iron deposition in a transfused patient who has been noncompliant with iron chelating therapy over several years.

Fig. 4.

Sagittal MRI of the heart in three patients with homozygous β thalassemia and transfusional iron overload. (A, left) Normal signal from the septum (long arrow) and posterior wall of the heart, consistent with the presence of very mild cardiac iron loading, in a transfused patient regularly complianct with iron chelating therapy. The homogenous signal of the liver, consistent with very mild iron loading in this organ (short arrow), is also seen below the image of the heart. (B, middle): Imhomogenity of signal from the septum (long arrow) and posterior wall, consistent with moderate iron deposition in a transfused patient erratically compliant with iron chelating therapy. Loss of liver signal (short arrow) is consistent with heavier iron loading in this organ. (C, right): Absence of signal from the septum (arrow), posterior wall and liver (short arrow), compatible with heavy iron deposition in a transfused patient who has been noncompliant with iron chelating therapy over several years.

Close modal
Fig. 5.

Sagittal MRI of cardiac iron before (left) and after (right) therapy with the orally active iron chelating agent deferiprone in the same patient with homozygous β thalassemia whose liver histology and hepatic MRI are shown in Fig 3. Imhomogenity of cardiac signal in the MRI on the left is compatible with significant iron deposition, while improvement in signal intensity after nine months of chelating therapy indicates that the cardiac iron content is reduced compared with that of the previous study. (Reprinted with permission.192 )

Fig. 5.

Sagittal MRI of cardiac iron before (left) and after (right) therapy with the orally active iron chelating agent deferiprone in the same patient with homozygous β thalassemia whose liver histology and hepatic MRI are shown in Fig 3. Imhomogenity of cardiac signal in the MRI on the left is compatible with significant iron deposition, while improvement in signal intensity after nine months of chelating therapy indicates that the cardiac iron content is reduced compared with that of the previous study. (Reprinted with permission.192 )

Close modal

Assessment of Organ Function

Cardiac function.Electro- or resting echo-cardiograms may be normal late in the course of iron-induced cardiac disease, and therefore are not sufficiently sensitive for the early detection of iron-induced cardiac dysfunction.199-203 Decreased left ventricular contractile reserve in clinically asymptomatic patients can be demonstrated with multi-gated exercise cardiac radionuclide angiography203 or with low-dose dobutamine stimulation204; these modalities may be useful in the diagnosis of early iron-induced cardiac disease. Diastolic dysfunction in asymptomatic individuals204-207 has been shown to have prognostic significance for the development of symptomatic iron-induced cardiac disease in some,205,206 but not all,70 studies.

Anterior pituitary reserve.Measurement of peak serum lutenizing hormone following a bolus of gonadotropin releasing hormone may be useful in the evaluation of pituitary reserve.137 In one study, 72% of patients with absent or very mild pituitary iron loading had a normal increase of lutenizing hormone, while only 5% of those with moderate or severe pituitary iron loading had a normal response.198 

Direct Assessment

Measurement of hepatic iron concentration is the most quantitative, specific, and sensitive method for determining the body iron burden in patients with thalassemia major.208 Liver biopsy is the best direct means of assessing iron deposition, permitting chemical measurement of the nonheme (storage) iron concentration and histochemical examination of the pattern of iron accumulation in hepatocytes and Kupffer cells as well as evaluation of the extent of inflammation, fibrosis, and cirrhosis. Magnetic susceptometry using a superconducting quantum interference device (SQUID) magnetometer provides a direct measure of hepatic storage iron that is based on a fundamental physical property of ferritin and hemosiderin.209-212 Use of the magnetic susceptibility of a tissue to determine the storage iron is much simpler than the use of the resonance behavior produced by the application of the oscillating magnetic fields used in magnetic resonance studies. When body iron stores are increased, the results of noninvasive determinations of magnetic susceptibility and of the chemical analysis of hepatic tissue obtained by biopsy are quantitatively equivalent.209-211 Magnetic susceptometry has been useful in clinical investigation of iron overload but is not generally available, in part because only two sites, one in the United States209 and one in Germany,212 have the specialized equipment needed for measurements of hepatic magnetic susceptibility.

Because the magnitude of the body iron burden seems to be the principal determinant of clinical outcome,91-93 the prime goal of iron-chelating therapy in patients with thalassemia major is the control of body iron. The optimal body iron should minimize both the risk of adverse effects from the iron-chelating agent and the risk of complications from iron overload. With stable transfusion requirements and in the absence of other confounding factors, the lower the level of body iron desired, the higher the dose of iron chelator needed. As detailed below, with many of the adverse reactions encountered with deferoxamine, the higher the dose, the greater the risk of adverse reactions. As a consequence, therapy to maintain a normal body iron, corresponding to a hepatic iron of about 1 to 9 μmol iron per gram liver, wet weight (about 0.2 to 1.6 mg iron per gram liver, dry weight)209 might abate the likelihood of complications of iron overload but greatly increase the probability of dose-related drug toxicity. At the opposite extreme, with high body iron burdens corresponding to hepatic iron concentrations greater than 80 μmol iron per gram liver, wet weight (about 15 mg iron per gram liver, dry weight),92,93 deferoxamine toxicity is rare but the risk of cardiac disease and early death is greatly increased.

In the absence of prospective clinical trials in patients with thalassemia major adequate for the evaluation of life-long therapy, guidance about the risk of complications associated with lower levels of body iron may be derived from the clinical experience with hereditary hemochromatosis. In this autosomal recessive disorder, the iron overload is the result of an abnormality affecting the regulation of iron absorption that produces an inappropriate increase in iron uptake, with homozygotes developing a chronic progressive increase in body iron stores.52 A candidate gene for this disorder has been recently identified.213 Minor iron loading develops in about one quarter of those heterozygous for hereditary hemochromatosis, but body iron stores in these individuals do not seem to increase beyond about two to four times the upper limit of normal.214 Body iron loads of the magnitude found in heterozygotes for hereditary hemochromatosis have no apparent ill effects and are associated with a normal life expectancy.214 By contrast, homozygotes who develop greater iron burdens have an increased risk of cardiac disease, hepatic fibrosis, diabetes mellitus, endocrine abnormalities, and other complications of iron overload. Just as for transfusional iron overload, in the iron overload of hereditary hemochromatosis, the greater the body iron excess, the higher the risk of adverse consequences.215-218 The toxic manifestations of iron overload depend not only on the amount of excess iron but also on (1) the rate of iron accumulation, (2) the duration of exposure to increased iron, (3) the partition of the iron burden between relatively benign sites in the macrophage and more hazardous deposits in parenchymal cells, (4) ascorbate status, which helps determine the allocation of iron between macrophage and parenchymal cells, (5) the extent of internal redistribution of iron between macrophage and parenchymal sites, and (6) noniron-related factors, such as alcohol and viral hepatitis.52 Nonetheless, the considerations above would suggest that a conservative goal for iron chelation therapy in patients with thalassemia major is to maintain an “optimal” body iron corresponding to hepatic storage iron concentrations of about 18 to 38 μmol iron per gram liver, wet weight (about 3.2 to 7 mg iron per gram liver, dry weight), in the range found in heterozygotes for hereditary hemochromatosis. The risks of deferoxamine toxicity associated with regimens to maintain body iron within this range are likely minor (see below) but are almost certainly increased at lower body iron burdens. Patients with higher body iron burdens, up to about 80 μmol iron per gram liver, wet weight (about 15 mg iron per gram liver, dry weight) are considered to be at an increased risk of hepatic fibrosis, diabetes mellitus, and other complications and need more intensive iron chelation therapy. Patients with still higher body iron burdens are recognized as having a greatly increased risk of cardiac disease and early death and are candidates for continuous IV ambulatory deferoxamine219 or other special programs of management.102 These ranges are shown graphically in Fig 6 and suggestions for management are summarized in Table 3.

Table 3.

Management of Iron Chelating Therapy in Thalassemia

TimepointAssessmentCommentResultsTreatment Recommendations
At start of therapy Liver biopsy under U/S guidance with quantitative liver iron, histology, PCR for hepatitis C RNA Should be obtained after approximately 1 yr of regular transfusion HIC < 3.2 mg/g dry weight 
HIC ≥ 3.2 mg/g dry weight Defer chelation; reassess HIC in 6 mo 
Initiate DFO at 25 mg/kg/night × 5 nights/wk 
 Radiographs of cartilage in wrists, knees, thoracolumbosacral spine; bone age Should be reviewed by pediatric radiologist and endocrinologist with previous experience in toxicity of DFO 
 Standing and sitting heights  
 Serum ferritin, Fe, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 
 
Yearly, before age 5 yr Liver Bx under U/S guidance; assessments as above  HIC < 3.2 mg/g dry weight Discontinue DFO; reassess HIC in 6 mo 
   HIC ≥ 3.2 but <7 mg/g dry weight Continue DFO at 25 mg/kg/night × 5 nights/wk 
   HIC ≥ 7 mg/g dry weight Increase DFO to 35 mg/kg/night × 6-7 nights/wk 
 Radiographs as above Same as above If severe spinal or metaphyseal changes present, reduce DFO to 25 mg/kg/night × 4 nights/wk even if HIC ≥ 7 mg/g dry weight. Reassess in 6 mo  
 Standing and sitting heights    
 Serum ferritin, serum iron, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 
 
Q18 mo, from age 5-10 yr Liver Bx under U/S guidance with quantitative HIC, histology, PCR for hepatitis C RNA  HIC < 3.2 mg/g dry weight 
HIC ≥ 3.2 but < 7 mg/g dry weight Discontinue DFO; reassess HIC in 6 mo 
Maintain DFO at 40 mg/kg/night × 5 nights/wk 
   HIC ≥ 7 but < 15 mg/g dry weight Maintain DFO at 40 mg/kg/night × 6-7 nights/wk 
   HIC ≥ 15 mg/g dry weight Maintain DFO at 40-50 mg/kg/night × 7 nights/wk 
 Radiographs as above Same as above If abnormal, reassess HIC promptly Titrate DFO as above 
 Standing and sitting heights    
 Serum ferritin, Fe, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 
Timepoint Assessment Comment Results Treatment Recommendations 
TimepointAssessmentCommentResultsTreatment Recommendations
At start of therapy Liver biopsy under U/S guidance with quantitative liver iron, histology, PCR for hepatitis C RNA Should be obtained after approximately 1 yr of regular transfusion HIC < 3.2 mg/g dry weight 
HIC ≥ 3.2 mg/g dry weight Defer chelation; reassess HIC in 6 mo 
Initiate DFO at 25 mg/kg/night × 5 nights/wk 
 Radiographs of cartilage in wrists, knees, thoracolumbosacral spine; bone age Should be reviewed by pediatric radiologist and endocrinologist with previous experience in toxicity of DFO 
 Standing and sitting heights  
 Serum ferritin, Fe, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 
 
Yearly, before age 5 yr Liver Bx under U/S guidance; assessments as above  HIC < 3.2 mg/g dry weight Discontinue DFO; reassess HIC in 6 mo 
   HIC ≥ 3.2 but <7 mg/g dry weight Continue DFO at 25 mg/kg/night × 5 nights/wk 
   HIC ≥ 7 mg/g dry weight Increase DFO to 35 mg/kg/night × 6-7 nights/wk 
 Radiographs as above Same as above If severe spinal or metaphyseal changes present, reduce DFO to 25 mg/kg/night × 4 nights/wk even if HIC ≥ 7 mg/g dry weight. Reassess in 6 mo  
 Standing and sitting heights    
 Serum ferritin, serum iron, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 
 
Q18 mo, from age 5-10 yr Liver Bx under U/S guidance with quantitative HIC, histology, PCR for hepatitis C RNA  HIC < 3.2 mg/g dry weight 
HIC ≥ 3.2 but < 7 mg/g dry weight Discontinue DFO; reassess HIC in 6 mo 
Maintain DFO at 40 mg/kg/night × 5 nights/wk 
   HIC ≥ 7 but < 15 mg/g dry weight Maintain DFO at 40 mg/kg/night × 6-7 nights/wk 
   HIC ≥ 15 mg/g dry weight Maintain DFO at 40-50 mg/kg/night × 7 nights/wk 
 Radiographs as above Same as above If abnormal, reassess HIC promptly Titrate DFO as above 
 Standing and sitting heights    
 Serum ferritin, Fe, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 
Timepoint Assessment Comment Results Treatment Recommendations 
Q18 mo, after 10 yr Liver Bx under U/S guidance; assessments as above  HIC < 3.2 mg/g dry weight Discontinue DFO; reassess HIC in 6 mo 
   HIC ≥ 3.2 but < 7 mg/g dry weight Maintain DFO at 40 mg/kg/night × 5 nights/wk 
   HIC ≥ 7 but < 15 mg/g dry weight Maintain DFO at 40 mg/kg/night × 6-7 nights/wk 
   HIC ≥ 15 mg/g dry weight Maintain DFO at 50 mg/kg/night × 7 nights/wk 
 Radiographs as above Same as above If abnormal, reassess HIC promptly Titrate DFO as above 
 Standing and sitting heights    
 Serum ferritin, Fe, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 
Q18 mo, after 10 yr Liver Bx under U/S guidance; assessments as above  HIC < 3.2 mg/g dry weight Discontinue DFO; reassess HIC in 6 mo 
   HIC ≥ 3.2 but < 7 mg/g dry weight Maintain DFO at 40 mg/kg/night × 5 nights/wk 
   HIC ≥ 7 but < 15 mg/g dry weight Maintain DFO at 40 mg/kg/night × 6-7 nights/wk 
   HIC ≥ 15 mg/g dry weight Maintain DFO at 50 mg/kg/night × 7 nights/wk 
 Radiographs as above Same as above If abnormal, reassess HIC promptly Titrate DFO as above 
 Standing and sitting heights    
 Serum ferritin, Fe, and TIBC    
 Serum ALT    
 Hepatitis screen    
 WBC ascorbate concentration    
 
If WBC ascorbate low, administer vitamin C PO 100 mg/night during DFO infusion 

Abbreviations: PCR, polymerase chain reaction; WBC, white blood cell; PO, orally; Q, every; HIC, hepatic iron concentration; DFO, deferoxamine; U/S, ultrasound; TIBC, total iron binding capacity; ALT, alanine aminotransferase; BX, biopsy.

If measurement of the hepatic iron concentration is not feasible, serum ferritin concentrations provide an alternative but less reliable means of determining if the body iron is in a optimal range (Fig 2). As noted above, a serum ferritin concentration of about 2,500 μg/L may be used as a threshold value to identify patients at an increased risk of cardiac disease and early death.91 Patients with most serum ferritin concentrations in excess of 2,500 μg/L had an estimated cardiac disease free survival after 15 years of less than 20% (Fig 1). The serum ferritin concentrations corresponding to the optimal range for hepatic iron shown in Fig 6 are less clearly defined. Preliminary analysis of studies of a large number of adults with thalassemia major over more than 15 years of deferoxamine therapy found that very rigorous control of body iron burden — as estimated by maintenance of serum ferritin concentrations under 1,000 μg/L — was associated with a very low incidence of iron-induced complications. Iron-related morbidity increased strikingly with even slightly less effective iron-chelating therapy.220 

Uncertainties as to the optimal age for the start of chelation therapy continue to exist. Reports of abnormal linear growth and metaphyseal dysplasia observed in children treated with deferoxamine before the age of 3 years141-144 have prompted recommendations for later therapy.141 In parallel, ultrastructural observations of liver biopsy specimens in transfused patients with thalassemia, including a unique study of three infants whose biopsies at this early age, have revealed moderate to severe iron overload.94 Furthermore, elevated hepatic iron concentrations associated with hepatic fibrosis, not uniformly evident by determinations of serum ferritin or laboratory abnormalities of liver function, have been observed in transfused thalassemic children less than 3 years of age.221,222 These data suggest that that some modified program of chelating therapy is likely indicated before this age (below and Table 3).

How can one identify the patient for whom iron chelation therapy should be initiated? Because of the imprecision of indirect measurements, we recommend that initiation of therapy be based on hepatic iron concentration obtained after 1 year of regular transfusions. Liver biopsy under ultrasound guidance is a safe procedure in children, with a complication rate of 0 in patients aged less than 5 years in a series of 1,184 biopsies performed before marrow transplantation for thalassemia.221 Although this must be viewed as an estimate with certain confidence limits, a similar experience has been observed from two other centers, including our own, with large numbers of patients regularly undergoing liver biopsies under ultrasound guidance223 (and Olivieri N.F., unpublished data, November 1996).

Few guidelines exist with respect to the initiation of iron-chelating therapy. In practice, the approach of most clinicians is to determine the serum ferritin concentration after a period of regular transfusions and, based on the value of this parameter, to begin a regimen of nightly subcutaneous deferoxamine therapy. As emphasized above, reliance on serum ferritin measurements alone can lead to inaccurate assessment of body iron burden in individual patients.165 Therefore, we recommend that all children with thalassemia major undergo determination of liver iron concentration after 1 year of regular transfusions. The value of hepatic iron that should prompt therapy is in the range of the same concentrations which should be ideally maintained during chronic iron-chelating therapy, as discussed above.

If liver biopsy is not available at the start of therapy, therapy with subcutaneous deferoxamine, not exceeding 25 to 35 mg deferoxamine per kilogram body weight/24 hours in young children, should be initiated after approximately 1 year of regular transfusions. The basis for this recommendation, and a titration scheme that has provided ideal chelating efficacy while attempting to circumvent drug toxicity, is detailed below and in Table 4.

Table 4.

Monitoring of Deferoxamine-Related Toxicity

ToxicityInvestigationsFrequencyAlteration in Therapy
(1) High frequency sensorineural hearing loss Audiogram Yearly; if patient symptomatic, immediate reassessment Interrupt DFO immediately; directly assess body iron burden; discontinue DFO × 6 mo if HIC 3.2-7 mg/g dry weight tissue; repeat audiogram Q3 mo until normal or stable; adjust DFO to HIC as per Table 3  
 
(2) Retinal abnormalities Retinal examination Yearly; if patient symptomatic, immediate reassessment Interrupt DFO immediately; directly assess body iron burden; discontinue DFO × 6 mo if HIC 3.2-7 mg/g dry weight tissue; review Q3 mo until normal or stable; adjust DFO to HIC as per Table 3  
 
(3) Metaphyseal and spinal abnormalities X-rays of wrists, knees, thoraco-lumbar-sacral spine; bone age of wrist Yearly Reduce DFO to 25 mg/kg/d × 4/wk; directly assess body iron burden; discontinue DFO × 6 mo if HIC ≤3 mg/g dry weight tissue; Reassess HIC after 6 mo; adjust DFO to HIC as per Table 3  
 
(4) Decline in height velocity and/or sitting height Determination of sitting and standing heights Twice yearly As in (3) above; Regular (6-monthly) assessment by pediatric endocrinologist 
ToxicityInvestigationsFrequencyAlteration in Therapy
(1) High frequency sensorineural hearing loss Audiogram Yearly; if patient symptomatic, immediate reassessment Interrupt DFO immediately; directly assess body iron burden; discontinue DFO × 6 mo if HIC 3.2-7 mg/g dry weight tissue; repeat audiogram Q3 mo until normal or stable; adjust DFO to HIC as per Table 3  
 
(2) Retinal abnormalities Retinal examination Yearly; if patient symptomatic, immediate reassessment Interrupt DFO immediately; directly assess body iron burden; discontinue DFO × 6 mo if HIC 3.2-7 mg/g dry weight tissue; review Q3 mo until normal or stable; adjust DFO to HIC as per Table 3  
 
(3) Metaphyseal and spinal abnormalities X-rays of wrists, knees, thoraco-lumbar-sacral spine; bone age of wrist Yearly Reduce DFO to 25 mg/kg/d × 4/wk; directly assess body iron burden; discontinue DFO × 6 mo if HIC ≤3 mg/g dry weight tissue; Reassess HIC after 6 mo; adjust DFO to HIC as per Table 3  
 
(4) Decline in height velocity and/or sitting height Determination of sitting and standing heights Twice yearly As in (3) above; Regular (6-monthly) assessment by pediatric endocrinologist 

It has been recognized that most toxic effects of deferoxamine have been observed in patients during administration of doses exceeding 50 mg per kilogram body weight, or smaller doses in the presence of very modestly elevated body iron burdens.224 The observation that the toxicity of deferoxamine is enhanced as the serum ferritin concentration declines, and deferoxamine dose increases, is supported by most analyses of this complication.141,142,224-226 As emphasized above, attempts to maintain normal hepatic iron concentrations with deferoxamine in patients with thalassemia major may be associated with increased deferoxamine toxicity.

Adverse effects associated with deferoxamine include ocular and auditory abnormalities,225,227-235 sensorimotor neurotoxicity,236 changes in renal function,237,238 and pulmonary toxicity.239,240 A toxic manifestation of deferoxamine therapy of great concern in young children is failure of linear growth (Fig 7), associated with evidence of cartilagenous dysplasia of the long bones (Fig 8) and spine (Fig 9).141,142,241-247 Over the past 3 years, it has been recognized that short stature, primarily related to disproportionate truncal growth and loss of sitting height in thalassemic children,141,142 may be due to the effect of deferoxamine on spinal cartilage.244-246 At the same time, the findings of iron overload and hepatic damage in young transfused children outlined above have prompted our recommendations of the use of deferoxamine early in life, using reduced doses as a balance between risk and benefit. This practice is supported by studies of children who have received low-dose deferoxamine (15 to 35 mg/kg/night) since the age of 3 years, all of whom had normal sitting heights, standing heights, and normal spinal x-rays. By contrast, in a second cohort of children in which deferoxamine, administered at standard doses (50 mg/kg) from an equally early age had induced a comparable reduction in body iron burden, mean sitting height was markedly abnormal and significant x-ray abnormalities were observed.246 These data suggest that abnormal linear growth may be a direct toxic effect of prolonged administration of higher doses of deferoxamine, unrelated to changes in body iron. Because improvement in linear growth of patients with spinal abnormalities has not been observed even with reduction of deferoxamine dose, it would appear important to prevent this toxicity.

Fig. 7.

Decline in height percentile observed in a child with thalassemia major. The patient began therapy at age 4 years, 11 months (arrow) with nightly subcutaneous deferoxamine (initial dose, 11 mg deferoxamine per kilogram per day; mean dose over the first 3 years of therapy, 55 ± 17 mg/kg/d). This patient had normal radiographs before the start of deferoxamine (see Fig 8A) but subsequently developed marked growth failure with a dramatic decline in height percentile, from the 37th percentile for age 6 months before initiation of deferoxamine, to less than the 3rd percentile 36 months later. (Reprinted with permission.144 )

Fig. 7.

Decline in height percentile observed in a child with thalassemia major. The patient began therapy at age 4 years, 11 months (arrow) with nightly subcutaneous deferoxamine (initial dose, 11 mg deferoxamine per kilogram per day; mean dose over the first 3 years of therapy, 55 ± 17 mg/kg/d). This patient had normal radiographs before the start of deferoxamine (see Fig 8A) but subsequently developed marked growth failure with a dramatic decline in height percentile, from the 37th percentile for age 6 months before initiation of deferoxamine, to less than the 3rd percentile 36 months later. (Reprinted with permission.144 )

Close modal
Fig. 8.

Radiographs of the femoral and tibial metaphyses of a child treated with deferoxamine therapy. Shown are the metaphyses prior to initiation of nightly subcutaneous deferoxamine (Fig 7A); 3 years after initiation of deferoxamine therapy (Fig 7B); and 6 years after initiation of deferoxamine therapy (Fig 7C). Radiographs show evidence of progressive widening and irregularity of the unossified metaphyseal matrix, which has irregular sclerotic margins. Similar processes in the proximal tibial metaphyses produced both varus and valgus deformities requiring bracing and osteotomy. (Reprinted with permission.144 )

Fig. 8.

Radiographs of the femoral and tibial metaphyses of a child treated with deferoxamine therapy. Shown are the metaphyses prior to initiation of nightly subcutaneous deferoxamine (Fig 7A); 3 years after initiation of deferoxamine therapy (Fig 7B); and 6 years after initiation of deferoxamine therapy (Fig 7C). Radiographs show evidence of progressive widening and irregularity of the unossified metaphyseal matrix, which has irregular sclerotic margins. Similar processes in the proximal tibial metaphyses produced both varus and valgus deformities requiring bracing and osteotomy. (Reprinted with permission.144 )

Close modal
Fig. 9.

Lateral view of the thoracic spine in an 11-year, 9-month-old girl with thalassemia major treated with intensive deferoxamine throughout childhood. The spine shows decreased vertebral height with intervertebral disc calcification, flattening and lengthening and anterior tapering or the vertebrae, wedging and moderate kyphosis in this region. Detailed inset shows a bone-within-bone appearance, demarcating a zone of pronounced calicification. (Reprinted by permission from Pediatric Radiology, Spinal deformities in deferoxamine-treated beta-thalassemia major patients, Hartkamp MJ, Babyn PS, Olivieri NF, Volume 23, pp 525-528, Figure 2, 1993, Copyright Springer-Verlag GmbH & Co, KG. 1993.)244 

Fig. 9.

Lateral view of the thoracic spine in an 11-year, 9-month-old girl with thalassemia major treated with intensive deferoxamine throughout childhood. The spine shows decreased vertebral height with intervertebral disc calcification, flattening and lengthening and anterior tapering or the vertebrae, wedging and moderate kyphosis in this region. Detailed inset shows a bone-within-bone appearance, demarcating a zone of pronounced calicification. (Reprinted by permission from Pediatric Radiology, Spinal deformities in deferoxamine-treated beta-thalassemia major patients, Hartkamp MJ, Babyn PS, Olivieri NF, Volume 23, pp 525-528, Figure 2, 1993, Copyright Springer-Verlag GmbH & Co, KG. 1993.)244 

Close modal

In summary, deferoxamine-induced toxicity can be avoided by regular, direct assessment of body iron burden with regular evaluation of the hepatic iron concentration. If hepatic iron concentration is not regularly assessed, a “toxicity” index, defined as the mean daily dose of deferoxamine (mg/kg) divided by the serum ferritin concentration (μg/L) should be calculated for each patient every 6 months, and should not exceed 0.025.226 We recommend that doses of deferoxamine not exceed 50 mg/kg/d. Although higher doses have been used in an attempt to “rescue” patients with severe iron-related organ failure, such attempts have not infrequently been associated with deferoxamine toxicity, including permanent hearing loss and fatal pulmonary toxicity.225,240 Hence, it is difficult to justify the use of higher doses, especially because very few centers now administer deferoxamine to patients in whom the body iron burden has been determined precisely using the hepatic iron concentration, rather than estimated using the serum ferritin concentration. Regular evaluation of deferoxamine toxicity (Table 4) is strongly recommended in all patients maintained on any dose of deferoxamine.

Ascorbate supplementation.The dilemma of ascorbate supplementation has been thoroughly reviewed.30 Low ascorbic acid levels have been found in iron-loaded thalassemic patients248-250 in whom ascorbate supplementation results in a marked improvement in deferoxamine-induced iron excretion251 by expansion of the chelatable iron pool to which deferoxamine has access.248-253 In parallel, ascorbate-induced expansion of this pool may enhance free radical formation, and aggravate the toxicity of iron in vivo.254-257 Although routine ascorbate supplementation has been therefore discouraged in patients with thalassemia,22 observation of loss of sustained efficacy of deferoxamine in an unsupplemented patient should prompt determination of tissue ascorbate concentrations. If these are reduced, 100 mg ascorbic acid per day should be administered. If possible, patients should administer ascorbic acid approximately 30 minutes to 1 hour after the start of an infusion of deferoxamine, only on days during which deferoxamine is administered. The toxicity of ascorbate supplementation during therapy with other chelating agents is presently unknown.

The most common difficulty associated with long-term therapy with subcutaneous deferoxamine is erratic compliance with therapy, which may decline as supervision of this regimen becomes increasingly the responsibility of the patient; objectively monitored compliance with deferoxamine is less than 70% in many older patients.258 Compliance with deferoxamine may be improved with intensive social and psychological support.259,260 

IV deferoxamine.Regimens of IV ambulatory deferoxamine administered through implantable venous access ports reduce the local pain and irritation of subcutaneous infusions, and are associated with rapid reduction of body iron burden.102 Regimens of continuous IV ambulatory deferoxamine in which the infusion site is changed weekly by medical personnel require infusion site care and a weekly clinic visit, but remove the need for nightly self-administration and improve patient compliance.219 

Bolus injections of subcutaneous deferoxamine.Very recently, studies of iron-loaded nonthalassemic191 and thalassemic patients261 patients have reported that deferoxamine administered by twice daily subcutaneous injections may be as effective as the same dose administered by subcutaneous infusion. Although bolus injections were administered in early clinical studies of deferoxamine, these reports represent the first attempts to evaluate the response to subcutaneous bolus injections, rather than infusions, of deferoxamine. If these early observations are confirmed, such a regimen may provide an alternative to prolonged infusions and freedom from infusion pumps.

Other forms of deferoxamine: Hydroxyethyl starch deferoxamine (HES-deferoxamine).Chemical attachment of deferoxamine to a hydroxyethyl starch polymer creates a high-molecular-weight chelator with affinity for iron identical to, but a vascular half-life 10 to 30 times longer than, that of standard deferoxamine.262,263 During a 4-hour IV infusion of HES-deferoxamine at doses equivalent to approximately 80 mg deferoxamine per kilogram body weight, no serious adverse clinical effects were observed in normal subjects.264 The efficacy and safety of a single infusion of this compound has now been assessed in a rising single dose study of eight iron-loaded patients.265 In patients with thalassemia major, approximately 50 or 85 mg of HES-deferoxamine per kilogram body weight induced urinary iron excretion equal to that achieved during a mean of 3 days of subcutaneous deferoxamine, with one patient excreting as much urinary iron after a single infusion of HES-deferoxamine as was achieved during 7 days of subcutaneous deferoxamine. A single infusion of HES-deferoxamine reduced nontransferrin-bound iron to zero or very low concentrations for 12 to 96 hours after infusion; nontransferrin-bound iron increased at the time point at which circulating chelator concentration began to decrease below the total plasma iron concentration. In one patient, urticaria prompted drug discontinuation; subsequent skin testing showed no allergy to starch, deferoxamine, or HES-deferoxamine. The efficacy and lack of toxicity of HES-deferoxamine in this single dose study in iron-loaded patients suggest that, if efficacy can be modified so that iron excretion after one infusion achieves that during 1 week of subcutaneous deferoxamine, this new compound might play a useful role in long-term reduction of body iron burden in selected patients with iron overload.

Other Indications for Chelating Therapy

Thalassemia “intermedia.”Iron loading secondary to increased gastrointestinal iron absorption in patients with thalassemia “intermedia” is less accelerated than that of transfusional iron overload in thalassemia major.266,267 Striking elevations of hepatic iron concentration, in parallel with modestly elevated levels of serum ferritin, have been observed in adult patients with thalassemia intermedia268; therefore, direct determination of body iron burden is indicated in any patient with thalassemia intermedia and an elevated serum ferritin concentration. Chelating therapy should be initiated if the hepatic iron concentration exceeds 7 mg per gram dry weight liver tissue, and hepatic iron concentration should assessed at frequent intervals during therapy. As detailed below, the orally active iron-chelating agent deferiprone has been shown to be rapidly effective in reducing body iron stores in thalassemia intermedia.192 

Chelation therapy after BM transplantation (BMT) for thalassemia.Successful allogeneic BMT in thalassemia liberates patients from chronic transfusions269 but does not eliminate the necessity for iron-chelating therapy in all patients. Timely reduction of hepatic iron concentration is observed only in younger patients with low pretransplantation body iron burdens; parenchymal hepatic iron overload persists, up to 6 years after marrow transplantation, in most patients who do not receive posttransplant deferoxamine treatment.270 Short-term deferoxamine is safe and effective in the reduction of tissue iron in the “ex-thalassemic” patient,271 and should be initiated 1 year after successful marrow transplantation if the hepatic iron concentration exceeds 7 mg iron/gram liver tissue, dry weight, at that time.

Orally active iron chelators.The expense and inconvenience of deferoxamine has mandated a 20-year search for an orally active iron chelator, four of which have reached clinical trials in the past decade. The compounds N,N′-bis (2-hydroxybenzoyl) ethylenediamine N,N′-diacetic acid (HBED), the aryl hydrazone pyridoxal isonicotinoyl hydrazone (PIH), and the di-ethyl hydroxypyridinone CP94, have all been evaluated in short-term trials over the last 5 years,272-274 but are not under clinical development at this time. The orally active iron-chelating agent most extensively evaluated to date is 1,2-dimethyl-3-hydroxypyridin-4-one (deferiprone; L1), one of the 3-hydroxypyridin-4-one bidentate iron chelators patented in 1982 as an alternative to deferoxamine for the treatment of chronic iron overload.275 Deferiprone, a neutral molecule, forms a neutral 3:1 chelator:ferric iron complex at pH 7.4. The drug may mobilize iron from ferritin, hemosiderin, lactoferrin, and diferric transferrin.276,277 Animal studies have reported variable efficacy in rodents and rabbits,278-284 and efficiency of chelation apparently insufficient to maintain negative iron balance in iron-loaded primates.285 In transfused patients with thalassemia major, 75 mg of deferiprone per kilogram body weight induces urinary iron excretion approximately equivalent to that achieved with 30 to 40 mg of deferoxamine per kilogram,286-288 sufficient to induce net negative iron balance in many patients with thalassemia major. Because fecal iron excretion induced by deferiprone is much less than that by deferoxamine,286,289 the short-term efficacy of deferiprone is unquestionably inferior to that of deferoxamine.

Effectiveness of deferiprone in long-term trials of thalassemia.Although the earliest studies reported no sustained decrease in serum ferritin concentration over 1 to 15 months of deferiprone therapy,290-293 two trials subsequently reported statistically significant reductions in mean serum ferritin concentration in patients with thalassemia major, with the most substantial decreases observed in those whose prestudy ferritin concentrations exceeded 5,000 μg/L, and in whom treatment had been administered for at least 18 months.294,295 A recent study demonstrated that, over the short term, deferiprone may reduce or maintain the serum ferritin concentration to levels associated with cardiac disease free survival in deferoxamine-treated patients,93 using criteria derived from a prospective trial in deferoxamine-treated patients.91 As noted above, reliance on serum ferritin concentrations alone may lead to inaccurate assessment of body iron burden in individual patients, and direct assessment of changes in tissue iron is particularly crucial in the evaluation of any new chelator. These studies of serum ferritin supported, but did not establish, the efficacy of deferiprone in the reduction of body iron burden.

Reduction in hepatic iron stores during deferiprone therapy was first shown in a patient with thalassemia “intermedia,”192 followed by a study reporting deferiprone-induced reduction in hepatic iron concentration in patients with thalassemia major.93 This study demonstrated that deferiprone was able, over a mean of 3 years of therapy, to reduce or maintain hepatic storage iron at concentrations associated with prolonged survival free of the clinical complications of iron overload, using the criteria derived from a prospective trial in deferoxamine-treated patients of 80 μmol of iron per gram liver, wet weight (15 mg of iron per gram liver, dry weight).92 Overall, the mean hepatic iron concentration of this cohort declined over a mean period of 3 years. These patients constitute the only group worldwide to receive long-term deferiprone therapy in conjunction with repeated measurements of hepatic iron concentration. This trial was terminated by its sponsor, Apotex Pharmaceuticals (Weston, Ontario, Canada) in May 1996.

Relative effectiveness of deferiprone and deferoxamine.The relative effectiveness and safety of, and compliance with, deferiprone and deferoxamine were being compared in a prospective randomized trial begun in Canada in 1993.258 Patients stratified for hepatic iron concentration had been randomized to receive 75 mg deferiprone per kilogram per day, or 50 mg subcutaneous deferoxamine per kilogram per night. This trial was intended to provide information about the relative long-term effectiveness of deferiprone and deferoxamine, but was also terminated prematurely by Apotex Pharmaceuticals in May 1996.

Toxicity of Deferiprone

Animal studies.As detailed previously,288 deferiprone did not receive full formal toxicologic evaluation before being given to humans; permission to administer the drug in early studies in the United Kingdom, India, Europe, and Canada was granted on the basis of limited toxicity studies in rodents. Acute toxicity studies have estimated an LD50 of 1,000 mg/kg in mice,278 and one of approximately 650 mg/kg in rats.296 Subacute toxicity studies in non–iron-loaded animals reported anemia, leukopenia, and thrombocytopenia in mice,278,283 anemia and leukopenia in rats,297 and death in dogs298 at doses 2- and 10-fold times those administered to iron-loaded humans. Adrenal hypertrophy, gonadal and thymic atrophy, bone marrow atrophy and pancytopenia, growth retardation, and embryotoxicity have also been reported in animals.299 

Human trials.The most common adverse effect associated with administration of deferiprone has been arthralgias, primarily of the large joints,294,295,300 the etiology of which remains elusive. The most serious adverse effect associated with the administration of deferiprone has been severe neutropenia or agranulocytosis, first reported in 1989.301 To date, this complication has been reported in 13 patients, of whom 10 have thalassemia major,301-305 as early as 6 weeks and up to 21 months after the initiation of deferiprone. No deaths have been reported as a result of this adverse effect. In five patients in whom rechallenge with deferiprone has been attempted after white blood cell counts returned to normal, a second decrease in neutrophil count has been observed.305 The mechanism of deferiprone-induced neutropenia is unknown. Although studies in animals and early reports in humans suggested that this effect might be related to administration of high doses of deferiprone, at least 7 patients have developed agranulocytosis during administration of the standard daily dose of 75 mg deferiprone per kilogram body weight; this adverse effect thus appears not to be dose-dependent, but idiosyncratic and unpredictable. A large trial of deferiprone in Italy and the United States is expected to provide an estimate of the incidence of this serious adverse effect, which is likely to limit the widespread use of deferiprone therapy.

Concerns regarding the adverse effects of deferiprone on immunologic function were raised in a case report describing fatal “systemic lupus erythematosus” in a patient receiving deferiprone in India,306,307 in studies reporting inhibition of human lymphocyte proliferation by deferiprone in vitro,308 and in studies describing thymic atrophy in rats.299 The significance of these reports remains unclear. The data from the case reports do not indicate a definite causal relationship between the symptom complex and treatment with deferiprone. Although deaths related to infection in deferiprone-treated patients in India have been attributed to immune dysfunction,309 these deaths were considered by the physicians responsible for the long-term treatment of the patients to be no different from those of other Indian patients, in whom pyogenic meningitis is a relatively common cause of death.310 Other adverse effects reported with deferiprone administration include dermatologic changes associated with decreases in serum zinc concentration resolving with oral zinc supplementation,311 nausea, and transient or sustained liver enzyme abnormalities.312 

The licensing of deferiprone.Deferiprone was administered to humans before full animal toxicologic evaluation required by the United States Food and Drug Administration (FDA) had been obtained. The data from available animal toxicity studies and clinical trials were first reviewed by representatives of the FDA in 1991, at which time approval for an investigational new drug application for deferiprone was deferred. At a second review in 1993, representatives of the FDA judged that a prospective, randomized trial to compare therapy with deferiprone with deferoxamine, and a second prospective study to estimate the incidence of serious adverse effects of deferiprone in a large cohort of patients, would be required for the licensing of deferiprone in the United States. Both of these studies were supported in part by the Canadian pharmaceutical company Apotex Pharmaceuticals. The first has been terminated prematurely by Apotex, while the second was completed in September 1996. In 1995, deferiprone was licensed for sale in India.

Iron-chelating therapy with deferoxamine in patients with thalassemia major has dramatically altered the prognosis of this previously fatal disease. The successes achieved with deferoxamine, as well as the limitations of this treatment, have stimulated the design of alternative strategies of iron-chelating therapy, including orally active iron chelators. The development of the most promising of these, deferiprone, has progressed rapidly over the last 5 years; data from several trials have provided direct and supportive evidence for its short-term efficacy. At the same time, the toxicity of this agent mandates a careful evaluation of the balance between risk and benefit of deferiprone in patients with thalassemia, in most of whom long-term deferoxamine is safe and efficacious therapy.

Although support for both the long-term treatment cohort of deferiprone-treated patients93 and a randomized trial of deferiprone and deferoxamine258 was terminated prematurely by their corporate sponsor, APOTEX Pharmaceuticals (Weston, Canada) in 1996, follow-up of hepatic storage iron concentrations in both cohorts have provided information regarding the long-term effectiveness of deferiprone in thalassemia major. In the long-term treatment cohort of deferiprone-treated patients reported previously,93 hepatic iron concentrations are now above the threshold associated with increased risk of heart disease and early death in thalassemia major91 in one third of patients.313 In the randomized trial of deferiprone and deferoxamine,258 review of available hepatic iron concentrations in patients who had completed 2 years of study by August 1996 showed a mean increase in hepatic iron concentration of approximately 50% over baseline in patients treated with deferiprone, but no significant change in those treated with deferoxamine.314 These results, recently reported to the Canadian drug regulatory agency, Health Protection Branch, Ottawa, Canada, raise concerns that long-term therapy with deferiprone may not provide adequate control of body iron in a substantial proportion of patients with thalassemia major.

N.F.O. is a Scientist of the Medical Research Council of Canada.

Address reprint requests to Nancy F. Olivieri, MD, Director, Haemoglobinopathy Program, The Hospital for Sick Children, Room 9413, 555 University Ave, Toronto, Ontario, Canada M5G 1X8.

1
Weatherall DJ, Clegg JB: The Thalassemia Syndromes (ed 3). Oxford, UK, Blackwell Scientific Publications, 1981
2
Cohen AR: Management of iron overload in the pediatric patient. in Hematol Oncol Clin North Am 521, 1987
3
Wolman
IJ
Transfusion therapy in Cooley's anemia: Growth and health as related to long range hemoglobin levels. A progress report.
Ann NY Acad Sci
119
1964
736
4
Wolman
IJ
Ortolani
M
Some clinical features of Cooley's anemia patients as related to transfusion schedules.
Ann NY Acad Sci
165
1969
407
5
Piomelli
S
Danoff
S
Becker
M
Lipera
M
Travis
S
Prevention of bone malformations and cardiomegaly in Cooley's anemia by early hypertransfusion regimen.
Ann NY Acad Sci
165
1969
427
6
Cazzola
M
De Stefano
P
Ponchio
L
Locatelli
F
Dessi
C
Beguin
Y
Barella
S
Dessi
C
Cao
A
Galanello
R
Relationship between transfusion regimen and suppression of erythropoiesis in beta thalassemia major.
Br J Haematol
89
1995
473
7
Cazzola
M
Locatelli
F
De Stefano
P
Deferoxamine in thalassemia major [letter].
N Engl J Med
332
1995
271
8
Propper
RD
Button
LN
Nathan
DG
New approaches to the transfusion management of thalassemia.
Blood
55
1980
55
9
Piomelli
S
Seaman
C
Reibman
J
Tyrun
A
Graziano
J
Tabachnik
N
Separation of younger red cells with improved survival in vivo: An approach to chronic transfusion therapy.
Proc Natl Acad Sci USA
75
1978
3474
10
Corash
L
Klein
H
Deisseroth
A
Shafer
B
Rosen
S
Beman
J
Griffith
P
Neinhuis
A
Selective isolation of young erythrocytes for transfusion support of thalassemia major patients.
Blood
57
1981
599
11
Bracey
AW
Klein
HG
Chambers
S
Corash
L
Ex-vivo selective isolation of young red blood cells using the IBM-2991 cell washer.
Blood
61
1983
1068
12
Cohen
AR
Schmidt
JM
Martin
MB
Barnsley
W
Schwartz
E
Clinical trial of young red cell transfusions.
J Pediatr
104
1984
865
13
Marcus
RE
Wonke
B
Bantock
HM
Thomas
MJ
Parry
ES
Taite
H
Huehns
ER
A prospective trial of young red cells in 48 patients with transfusion-dependent thalassemia.
Br J Haematol
60
1985
153
14
Kevy
SV
Jacobson
MS
Fosburg
M
Renaud
M
Scanlon
A
Carmen
R
Nelson
E
A new approach to neocyte transfusion: Preliminary report.
J Clin Apheresis
4
1988
194
15
Simon
TL
Sohmer
P
Nelson
EF
Extended survival of neocytes produced by a new system.
Transfusion
29
1989
221
16
Collins
AF
Dias
GC
Haddad
S
Talbot
R
Herst
R
Tyler
BJ
Zuber
E
Blanchette
VS
Olivieri
NF
Evaluation of a new neocyte transfusion preparation vs. washed cell transfusion in patients with homozygous beta thalassemia.
Transfusion
34
1994
517
17
Cohen
AR
Martin
MB
Silber
JH
Kim
HC
Ohene-Frempong
K
Schwartz
E
A modified transfusion program for prevention of stroke in sickle cell disease.
Blood
79
1994
1657
18
Berdoukas
VA
Kwan
YL
Sansotta
ML
A study on the value of red cell exchange transfusion in transfusion dependent anemias.
Clin Lab Haematol
8
1986
209
19
Modell
B
Total management of thalassaemia major.
Arch Dis Child
52
1977
489
20
Cohen
A
Gayer
R
Mizanin
J
Longterm effect of splenectomy on transfusion requirements in thalassemia major.
Am J Hematol
30
1989
254
21
Olivieri NF: Unpublished observations, November 1996
22
Fosburg
M
Nathan
DG
Treatment of Cooley's anemia.
Blood
76
1990
435
23
Zurlo
MF
De Stefano
P
Borgna-Pignatti
C
Di Palma
A
Piga
A
Melevendi
C
Di Gregorio
F
Burattini
MG
Terzoli
S
Survival and causes of death in thalassaemia major.
Lancet
2
1989
27
24
Piperno
A
Fargion
S
D'Alba
R
Liver damage in Italian patients with hereditary hemochromatosis is highly influenced by hepatitis B and C virus infection.
J Hepatol
16
1992
364
25
Sher GD, Milone SD, Cameron R, Jamieson FB, Krajden M, Collins AF, Matsui D, Entsuah B, Berkovitch M, Hackman R, Francombe WH, Olivieri NF: Hepatitis C virus infection in transfused patients with β hemoglobinopathies accelerates iron-induced hepatic damage. Blood 82:360a, 1993 (abstr, suppl 1)
26
Lai
ME
De Virgilis
S
Argiolu
F
Farci
P
Mazzoleni
AP
Lisci
V
Rapicetta
M
Clemente
MG
Nurchis
P
Arnone
M
Balestrieri
A
Cao
A
Evaluation of antibodies to hepatitis C virus in a long-term prospective study of posttransfusion hepatitis among thalassemic children: Comparison between first- and second-generation assay.
J Pediatr Gastroenterol Nutr
16
1993
458
27
Tong
MT
El-Farra
NS
Reikes
AR
Co
RL
Clinical outcomes after transfusion-associated hepatitis C.
N Engl J Med
332
1995
1463
28
Donohue
SM
Wonke
B
Hoffbrand
AV
Reittie
J
Ganeshaguru
K
Scheuer
PJ
Brown
D
Dusheiko
G
Alpha interferon in the treatment of chronic hepatitis C infection in thalassaemia major.
Br J Haematol
83
1993
491
29
Clemente
MG
Congia
M
Lai
ME
Killiu
F
Lampis
R
Frau
F
Frau
MR
Faa
G
Diana
G
Dessi
C
Melis
A
Mazzoleni
AP
Cornnacchia
G
Cao
A
De Virgiliis
S
Effect of iron overload on the response to recombinant interferon-alfa treatment in transfusion-dependent patients with thalassemia major and chronic hepatitis C.
J Pediatr
125
1994
123
30
Hershko
C
Weatherall
DJ
Iron-chelating therapy.
CRC Crit Rev Clin Lab Sci
26
1988
303
31
Halliwell
B
Gutteridge
JMC
Oxygen toxicity, oxygen radicals, transition metals and disease.
Biochem J
219
1984
1
32
Slater
TF
Free radical mechanisms in tissue injury.
Biochem J
222
1984
1
33
Bacon
BR
Tavill
AS
Brittenham
GM
Park
CH
Recknagel
RO
Hepatic lipid peroxidation in vivo in rats with chronic iron overload.
J Clin Invest
71
1983
429
34
Heys
AD
Dormandy
TL
Lipid peroxidation in iron loaded spleens.
Clin Sci
60
1981
295
35
Hershko
C
Peto
TEA
Annotation: Non-transferrin plasma iron.
Br J Haematol
66
1987
149
36
Sutton
HC
Efficiency of chelated iron compounds as catalysts for the Haber-Weiss Reacion.
J Free Radical Biol Med
1
1985
195
37
Hershko
C
Graham
G
Bates
CW
Rachmilewitz
EA
Non-specific serum iron in thalassemia: An abnormal serum iron fraction of potential toxicity.
Br J Haematol
40
1978
255
38
Batey
RG
LaiChung
Fong P
Sherlock
S
The nature of serum iron in primary haemachromatosis.
Clin Sci
55
1978
24
39
Anuwatanakulchia
M
Pootrakul
P
Thuvasethakul
P
Wasi
P
Non-transferrin plasma iron in β-thalassaemia/HbE and haemoglobin H diseases.
Scand J Haematol
32
1984
153
40
Wagstaff
M
Peters
SW
Jones
BM
Jacobs
A
Free iron and iron toxicity in iron overload.
Br J Haematol
61
1985
566
41
Gutteridge
JMC
Rowley
DA
Griffiths
E
Halliwell
B
Low-molecular-weight iron complexes and oxygen radical reactions in idiopathic haemochromatosis.
Clin Sci
68
1985
463
42
Wang
WC
Ahmed
N
Hanna
M
Non-transferrin-bound iron in long-term transfusion in children with congenital anemias.
J Pediatr
108
1986
552
43
al-Refaie
FN
Wickens
DG
Wonke
B
Kontoghiorghes
GJ
Hoffbrand
AV
Serum non-transferrin-bound iron in beta-thalassaemia major patients treated with desferrioxamine and L1.
Br J Haematol
82
1992
431
44
Link
G
Pinson
A
Hershko
C
Heart cells in culture: A model of myocardial iron overload and chelation.
J Lab Clin Med
106
1985
147
45
White
GP
Jacobs
A
Grady
RW
Cerami
A
The use of chang cells cultured in vitro to evaluate potential iron chelating drugs.
Br J Haematol
33
1976
487
46
Jacobs A: in Fitzsimons DW (ed): Iron Metabolism: Ciba Foundation Symposium. Amsterdam, The Netherlands, Elsevier, 1977, p 91
47
Guterridge
JMC
Halliwell
B
Iron toxicity and oxygen radicals.
Bailliere's Clin Hematol
2
1989
195
48
Keberle
H
The biochemistry of desferrioxamine and its relation to iron metabolism.
Ann NY Acad Sci
119
1964
758
49
Callender
ST
Weatherall
DJ
Iron chelation with oral desferrioxamine.
Lancet
2
1980
689
50
Summers
MR
Jacobs
A
Tudway
D
Perera
P
Ricketts
C
Studies in desferrioxamine and ferrioxamine metabolism in normal and iron-loaded subjects.
Br J Haematol
42
1979
547
51
Pippard
M
Desferrioxamine induced iron excretion in humans.
Bailliere's Clin Hematol
2
1989
323
52
Brittenham GM: Disorders of iron metabolism: Deficiency and overload, in Hoffman R, Benz E, Shattil S, Furie B, Cohen H (eds): Hematology: Basic Principles and Practice. New York, NY, Churchill Livingstone, 1994, p 492
53
Willis
ED
Lipid peroxide formation in microsomes. The role of non-haem iron.
Biochem J
13
1969
325
54
Morehouse
LA
Thomas
CE
Aust
SD
Superoxide generation of NADPH-Cytochrome P-450 reductase: The effect of iron chelators and the role of superoxide in microsomal lipid peroxidation.
Arch Biochem Biophys
232
1984
366
55
O'Connell
MJ
Ward
RJ
Baum
H
Peters
TJ
The role of iron in ferritin- and haemosiderin-mediated lipid peroxidation in liposomes.
Biochem J
229
1985
135
56
Hershko
C
Link
G
Pinson
A
Modification of iron uptake and lipid peroxidation by hypoxia, ascorbic acid and α-tocopherol in iron-loaded rat myocardial cell cultures.
J Lab Clin Med
110
1987
355
57
Link
G
Athias
P
Grynberg
A
Pinson
A
Hershko
C
Effect of iron loading on transmembrane potential, contraction and automaticity of rat ventricular muscle cells in culture.
J Lab Clin Med
113
1989
103
58
Sephton-Smith
R
Iron excretion in thalassaemia major after administration of chelating agents.
Br Med J
2
1962
1577
59
Bannerman
RM
Callender
ST
Williams
DL
Effect of desferrioxamine and DTPA in iron overload.
Br Med J
2
1962
1573
60
Sephton-Smith
R
Chelating agents in the diagnosis and treatment of iron overload in thalassemia.
Ann NY Acad Sci
119
1964
776
61
Barry
M
Flynn
D
Letsky
E
Risdon
RA
Long term chelation therapy in thalassemia major: Effect on liver iron concentration, liver histology and clinical progress.
Br Med J
2
1974
16
62
Modell
CB
Beck
J
Long-term desferrioxamine therapy in thalassaemia.
Ann NY Acad Sci
232
1974
201
63
Propper
RD
Shurin
SB
Nathan
DG
Reassessment of the use of desferrioxamine B in iron overload.
N Engl J Med
294
1976
421
64
Hussain
MAM
Flynn
DM
Green
N
Hussein
S
Hoffbrand
AV
Subcutaneous infusion and intramuscular injection of desferrioxamine in patients with transfusional iron overload.
Lancet
2
1976
1278
65
Propper
RL
Cooper
B
Rufo
RR
Nienhuis
AW
Anderson
W
Bunn
HF
Rosenthal
A
Nathan
DG
Continuous subcutaneous administration of deferoxamine in patients with iron overload.
N Engl J Med
297
1977
418
66
Pippard
MJ
Callender
ST
Weatherall
DJ
Intensive iron-chelation therapy with desferrioxamine in iron-loading anaemias.
Clin Sci Mol Med
54
1978
99
67
Pippard
MJ
Callender
ST
Finch
CA
Ferrioxamine excretion in iron loaded man.
Blood
60
1982
288
68
Engle
MA
Erlandson
M
Smith
CH
Late cardiac complications of chronic, severe, refractory anemia with hemochromatosis.
Circulation
30
1964
689
69
Grisaru
D
Rachmilewitz
FA
Mosseri
M
Gotsman
Latair JS
Okon
E
Goldfarb
A
Hasin
Y
Cardiopulmonary assessment in β-thalassemia major.
Chest
98
1990
1138
70
Kremastinos
DTh
Tiniakos
G
Theodorakis
GN
Katritsis
DG
Toutouzas
PK
Myocarditis in β-thalassemia major: A cause of heart failure.
Circulation
91
1995
66
71
Buja
LM
Roberts
W
Iron in the heart: Etiology and clinical significance.
Am J Med
51
1971
209
72
MacDonald
RA
Mallory
GK
Haemochromatosis and haemosiderosis: Study of 21 autopsied cases.
Arch Int Med
105
1960
686
73
Graziano
JH
Piomelli
S
Hilgartner
M
Giardian
P
Karpatkin
M
Andrew
M
Lo
Iacomo N
Seaman
C
Chelation therapy in β-thalassemia major. III. The role of splenectomy in achieving iron balance.
J Pediatr
99
1981
695
74
Modell
B
Letsky
EA
Flynn
DM
Peto
R
Weatherall
DJ
Survival and desferrioxamine in thalassaemia major.
Br Med J
284
1982
1081
75
Flynn
DM
Hoffbrand
AV
Politis
D
Subcutaneous desferrioxamine: The effects of three years' treatment on liver iron, serum ferritin, and comments on echocardiography.
Birth Defects
18
1982
347
76
Weatherall
DJ
Pippard
MJ
Callender
ST
Iron loading in thalassemia — Five years with the pump.
N Engl J Med
308
1983
456
77
Pippard
MJ
Callender
ST
The management of iron chelation therapy.
Br J Haematol
54
1983
503
78
Freeman
AP
Giles
RW
Berdoukas
VA
Walsh
WF
Choy
D
Murray
PC
Early left ventricular dysfunction and chelation therapy in thalassemia major.
Ann Intern Med
99
1983
450
79
Marcus
RE
Davies
SC
Bantock
HM
Underwood
SR
Walton
S
Huehns
ER
Desferrioxamine to improve cardiac function in iron-overloaded patients with thalassaemia major.
Lancet
1
1984
392
80
Anon
High-dose chelation therapy in thalassaemia.
Lancet
1
1984
373
81
Hyman
CB
Agness
CL
Rodriguez-Funes
R
Zednikova
M
Combined subcutaneous and high-dose intravenous deferoxamine therapy of thalassemia.
Ann NY Acad Sci
445
1985
293
82
Giardina
PJV
Ehlers
KH
Engle
MA
Grady
RW
Hilgartner
MW
The effect of subcutaneous deferoxamine on the cardiac profile of thalassemia major: A five-year study.
Ann NY Acad Sci
445
1985
282
83
Wolfe
L
Olivieri
N
Sallan
D
Colan
S
Rose
V
Propper
R
Freedman
MH
Nathan
DG
Prevention of cardiac disease by subcutaneous deferoxamine in patients with thalassemia major.
N Engl J Med
312
1985
1600
84
Schafer
AI
Rabinowe
S
LeBoff
MS
Bridges
K
Cheron
RG
Dluhy
R
Long-term efficacy of deferoxamine iron chelation therapy in adults with acquired transfusional iron overload.
Arch Intern Med
145
1985
1217
85
Rahko
PS
Salerni
R
Uretsky
BF
Successful reversal by chelation therapy of congestive cardiomyopathy due to iron overload.
J Am Coll Cariol
8
1986
426
86
Brittenham G, Nienhuis A: Desferrioxamine use protects against heart disease and death from transfusional iron overload in thalassemia major. Blood 72:56a, 1988 (abstr, suppl 1)
87
Aldouri
MA WB
Hoffbrand
AV
Flynn
DM
Ward
SE
Agnew
JE
Hilson
AJW
High incidence of cardiomyopathy in beta-thalassemia patients receing transfusion and iron chelation: reversal by intensified chelation.
Acta Haematol
84
1990
113
88
Lerner
N
Blei
F
Bierman
F
Johnson
L
Piomelli
S
Chelation therapy and cardiac status in older patients with thalassemia major.
Am J Ped Hematol Oncol
12
1990
56
89
Olivieri
NF
McGee
A
Liu
P
Koren
G
Freedman
MH
Benson
LN
Cardiac disease-free survival in patients with thalassemia major treated with subcutaneous deferoxamine.
Ann NY Acad Sci
612
1990
584
90
Ehlers
KH
Giardina
PJ
Lesser
ML
Engle
MA
Hilgartner
MW
Prolonged survival in patients with beta-thalassemia major treated with deferoxamine.
J Pediatr
118
1991
549
91
Olivieri
NF
Nathan
DG
MacMillan
JH
Wayne
AD
Martin
M
McGee
A
Koren
G
Liu
PP
Cohen
AR
Survival of medically treated patients with homozygous β thalassemia.
N Engl J Med
331
1994
574
92
Brittenham
GM
Griffith
PM
Nienhuis
AW
McLaren
CE
Young
NS
Tucker
EE
Allen
CJ
Farrell
DE
Harris
JW
Efficacy of deferoxamine in preventing complications of iron overload in patients with thalassemia major.
N Engl J Med
331
1994
567
93
Olivieri
NF
Brittenham
GM
Matsui
D
Berkovitch
M
Blendis
LM
Cameron
RG
McClelland
RA
Liu
PP
Templeton
DM
Koren
G
Iron-chelation therapy with oral deferiprone in patients with thalassemia major.
N Engl J Med
332
1995
918
94
Iancu
TC
Neustein HB. Ferritin in human liver cells of homozygous β thalassemia
Ultrastructural observations.
Br J Haematol
37
1977
527
95
Tsukamoto
H
Horne
W
Kamimura
S
Niemelä
O
Parkkila
S
Yiä-Herttula
S
Brittenham
GM
Experimental liver cirrhosis induced by alcohol and iron.
J Clin Invest
96
1995
620
96
Hoffbrand
AV
Gorman
A
Laulicht
M
Garidi
M
Economikdou
J
Georgipoulou
P
Hussain
MAM
Flynn
DM
Improvement in iron status and liver function in patients with transfusional iron overload with long-term subcutaneous desferrioxamine.
Lancet
1
1979
946
97
Janka
GE
Mohring
P
Helmig
M
Haas
RJ
Betke
K
Intravenous and subcutaneous desferrioxamine therapy in children with severe iron overload.
Eur J Pediatr
137
1981
385
98
Cohen
A
Martin
M
Schwartz
E
Response to long-term deferoxamine therapy in thalassemia.
J Pediatr
99
1981
689
99
Cohen
A
Martin
M
Schwartz
E
Depletion of excessive liver iron stores with desferrioxamine.
Br J Haematol
58
1984
369
100
Cohen
A
Mizanin
J
Schwartz
E
Treatment of iron overload in Cooley's anemia.
Ann NY Acad Sci
445
1985
374
101
Aldouri
MA
Wonke
B
Hoffbrand
AV
Flynn
DM
Laulicht
M
Fenton
LA
Scheuer
PJ
Kibbler
CC
Allwood
CA
Brown
D
Thomas
HC
Iron state and hepatic disease in patients with thalassaemia major treated with long term subcutaneous desferrioxamine.
J Clin Pathol
40
1987
1352
102
Cohen
AR
Mizanin
J
Schwartz
E
Rapid removal of excessive iron with daily, high-dose intravenous chelation therapy.
J Pediatr
115
1989
151
103
Grundy
RG
Woods
RA
Savage
MO
Evans
JPM
Relationship of endocrinopathy to iron chelation status in young patients with thalassaemia major.
Arch Dis Child
71
1994
128
104
Kwan
EYW
Lee
ACW
Li
AMC
Tam
SCF
Chan
CF
Lau
YL
Low
LCK
A cross-sectional study of growth, puberty and endocrine function in patients with thalassaemia major in Hong Kong.
J Paediatr Child Health
31
1995
83
105
Landau
H
Matoth
I
Landau-Cordova
Z
Goldfarb
A
Rachmilewitz
EA
Glaser
B
Cross-sectional and longitudinal study of the pituitary-thyroid axis in patient with thalassaemia major.
Clin Endocrinol
38
1993
55
106
McIntosh
N
Endocrinopathy in thalassaemia major.
Arch Dis Child
51
1976
195
107
Sklar
CA
Lew
LQ
Yoon
DJ
David
R
Adrenal function in thalassemia major following long term treatment with multiple transfusions and chelation therapy. Evidence for dissociation of cortisol and adrenal androgen secretion.
Am J Dis Child
141
1987
327
108
Kattamis
C
Touliatos
N
Haidas
S
Matsaniotis
N
Growth of children with thalassaemia: Effect of different transfusional regimens.
Arch Dis Child
45
1970
502
109
Costin
G
Kogut
MD
Hyman
CB
Ortega
JA
Endocrine abnormalities in thalassemia major.
Am J Dis Child
133
1979
497
110
Maurer
HS
Lloyd-Still
JD
Ingrisano
C
Gonzalez-Crussi
F
Honig CR. A prospective evaluation of iron chelation therapy in children with severe β-thalassaemia
A six-year study.
Am J Dis Child
142
1988
287
111
Modell
B
Advances in the use of iron-chelating agents for the treatment of iron overload.
Prog Hematol
11
1979
267
112
Modell B, Berdoukas V: The Clinical Approach to Thalassaemia. London, UK, Grune and Stratton, 1984
113
Borgna-Pignatti
C
De Stefano
P
Zonta
L
Vullo
C
De Sanctis
V
Melevendi
C
Naselli
A
Masera
G
Terzoli
S
Gabutti
V
Piga
A
Growth and sexual maturation in thalassemia major.
J Pediatr
106
1985
150
114
Kattamis C, Liakopoulou T, Kattamis A: Growth and development in children with thalassaemia major. Acta Paediatr Scand 366:111, 1990 (suppl)
115
Kletzky
OA
Costin
G
Marrs
RP
Bernstein
G
March
CM
Mishell
DR Jr
Gonadotropin insufficiency in patients with thalassemia major.
J Clin Endocrinol Metab
48
1979
901
116
Wang
C
Tso
SC
Todd
D
Hypogonadotropic hypogonadism in severe β-thalassemia: Effect of chelation and pulsatile gonadotropin-releasing hormone therapy.
J Clin Endocrionol Metab
68
1989
511
117
Saenger
P
Schwartz
E
Markenson
AL
Graziano
JH
Levine
LS
New
AMI
Hilgartner
MW
Depressed serum somatomedin activity in beta-thalassemia.
J Pediatr
96
1980
214
118
Werther
GA
Matthews
RN
Burger
HG
Herington
AC
Lack of response of nonsuppressible insulin-like activity to short term administration of human growth hormone in thalassemia major.
J Clin Endocrionol Metab
53
1981
806
119
Herington
AC
Werthr
GA
Matthews
RN
Burger
HG
Studies on the possible mechanism for deficiency of nonsuppressible insulin-like activity in thalassemia major.
J Clin Endocrinol Metab
52
1981
293
120
Bergeron
C
Kovacs
K
Pituitary siderosis: A histologic, immunocytologic, and ultrastructural study.
Am J Pathol
9
1978
295
121
Kelly
TM
Edwards
CQ
Meikle
AW
Kushner
JP
Hypogonadism in hemochromatosis: Reversal with iron depletion.
Ann Intern Med
101
1984
629
122
Siemons
LJ
Mahler
CH
Hypogonadotropic hyogonadism in hemochromatosis: Recovery of reproductive function after iron depletion.
J Clin Endocrinol Metab
65
1987
585
123
Pintor
C
Cella
G
Manso
P
Corda
R
Dessi
C
Locatelli
V
Muller
EE
Impaired growth hormone (GH) response to GH-releasing hormone in thalassemia major.
J Clin Endocrinol Metab
62
1986
263
124
Shehadeh
N
Hazani
A
Rudolf
MCJ
Peleg
I
Benderly
A
Hochberg
Z
Neurosecretory dysfunction of growth hormone secretion in thalassaemia major.
Acta Paediatr Scand
79
1990
790
125
Postel-Vinay
MC
Girot
R
Leger
J
Hocquette
JF
McKelvie
P
Amar-Costesec
A
Rappaport
R
No evidence for a defect in growth hormone binding to liver membranes in thalassemia major.
J Clin Endocrinol Metab
68
1989
94
126
Masala
A
Melom
T
Gallisai
D
Alagna
S
Rovasio
PP
Rassu
S
Milia
AF
Endocrine functioning in multitransfused prepubertal patients with homogygous β-thalassemia.
J Clin Endocrinol Metab
58
1984
667
127
Tolis
G
Politis
C
Kontopoulou
I
Poulatzas
N
Rigas
G
Saridakis
C
Athanasiou
V
Mortoglou
A
Malachtari
Ling N
Pituitary somatotropic and corticotropic function in patients with β-thalassaemia on iron chelation therapy.
Birth Defects
23
1988
449
128
Leger
J
Girot
R
Crosnier
H
Postel-Vinay
MC
Rappaport
R
Normal growth hormone (GH) response to GH-releasing hormone in children with thalassemia major before puberty: A possible age-related effect.
J Clin Endocrinol Metab
69
1989
453
129
Scacchi
M
Danesi
L
De Martin
M
Dubini
A
Forni
L
Masala
A
Gallisai
D
Burrai
C
Terzoli
C
Marzano
C
Cavagnini
F
Treatment with biosynthetic growth hormone of short thalassaemic patients with impaired growth hormone secretion.
Clin Endocrinol
35
1991
335
130
Low
LCK
Kwan
EYW
Lim
YJ
Lee
ACW
Tam
CF
Lam
KSL
Growth hormone treatment of short Chinese children with β-thalassaemia major without growth hormone deficiency.
Clin Endocrinol
42
1995
359
131
Bozzola
M
Argente
J
Cristernino
M
Moretta
A
Valtorta
A
Biscaldi
I
Donnadieu
M
Evain-Brion
D
Severi
F
Effect of human chorionic gonadotropin on growth velocity and biological growth parameters in adolescents with thalassaemia major.
Eur J Pediatr
148
1989
300
132
Flynn
DM
Fairney
A
Jackson
D
Clayton
BE
Hormonal changes in thalassaemia major.
Arch Dis Child
51
1976
828
133
Arcasoy
A
Cavdar
A
Cin
S
Erten
J
Babacan
E
Gozdasoglu
S
Akar
N
Effects of zinc supplementation on linear growth in beta thalassemia (a new approach).
Am J Hematol
24
1987
127
134
Leek
JC
Vogler
JB
Gershwin
ME
Golub
MS
Hurley
LS
Hendrickx
AG
Studies of marginal zinc deprivation in rhesus monkeys. V. Fetal and infant skeletal effects.
Am J Clin Nutr
40
1984
1203
135
Nishi
Y
Hatano
S
Aihara
K
Fujie
A
Kihara
M
Transient partial growth hormone deficiency due to zinc deficiency.
J Am Coll Nutr
8
1989
93
136
Vassilopoulou-Sellin
R
Oyedeji
CO
Foster
PL
Thompson
MM
Saman
NA
Haemoglobin as a direct inhibitor of cartilage growth in vitro.
Horm Metab Res
21
1989
11
137
Bronspeigel-Weintrob
N
Olivieri
NF
Tyler
BJ
Andrews
D
Freedman
MH
Holland
FJ
Effect of age at the start of iron chelation therapy on gonadal function in β-thalassemia major.
N Engl J Med
323
1990
713
138
Jensen
CE
Tuck
SM
Wonke
B
Fertility in thalassaemia major: A report of 16 pregnancies, preconceptual evaluation and a review of the literature.
Br J Obstet Gynaecol
102
1995
625
139
Chatterjee
R
Katz
M
Cox
TF
Porter
JB
Prospective study of the hypothalamic-pituitary axis in thalassaemic patients who developed secondary amenorrhea.
Clin Endocrinol
39
1993
287
140
De Sanctis
V
Katz
M
Vullo
C
Bagni
B
Ughi
M
Wonke
B
Effect of different treatment regimes on linear growth and final height in β-thalassaemia major.
Clin Endocrinol
40
1994
91
141
Rodda
CP
Reid
ED
Johnson
S
Doery
J
Matthews
R
Bowden
DK
Short stature in homozygous β-thalassaemia is due to disproportionate truncal shortening.
Clin Endocrinol
42
1995
587
142
Piga
A
Luzzatto
L
Capalbo
P
Gambotto
S
Tricta
F
Gabutti
V
High-dose desferrioxamine as a cause of growth failure in thalassaemic patients.
Eur J Haematol
40
1988
380
143
DeVirgilis
S
Congia
M
Frau
F
Argiolu
F
Diana
G
Cucca
F
Varsi
A
Sanna
G
Podda
G
Fodde
M
Franco-Piratu
G
Cao
A
Deferoxamine-induced growth retardation in patients with thalassemia major.
J Pediatr
113
1988
661
144
Olivieri
NF
Koren
G
Harris
J
Khattak
S
Freedman
MH
Templeton
DM
Bailey
JD
Reilly
BJ
Growth failure and bony changes induced by deferoxamine.
Am J Ped Hematol Oncol
14
1992
48
145
Lassman
MN
Genel
M
Wise
JK
Hendler
R
Felig
P
Carbohydrate homeostasis and pancreatic islet cell function in thalassemia.
Ann Intern Med
80
1974
65
146
Costin
G
Kogut
MD
Hyman
C
Ortega
JA
Carbohydrate metablism and pancreatic islet-cell function in thalassemia major.
Diabetes
26
1977
230
147
Saudek
CD
Hemm
RM
Peterson
CM
Abnormal glucose tolerance in β-thalassemia major.
Metabolism
26
1977
43
148
Zuppinger
K
Molinari
B
Hirt
A
Imbach
P
Bugler
E
Tönz
O
Zurbrügg
RP
Increased risk of diabetes melliuts in beta-thalassaemia major.
Hel Paediat Acta
4
1979
197
149
De Sanctis
V
D'Ascola
G
Wonke
B
The development of diabetes mellitus and chronic liver disease in long term chelated β-thalassaemic patients.
Postgrad Med J
62
1986
831
150
De Sanctis
V
Zurlo
MG
Senesi
E
Boffa
C
Cavallo
L
Di Gregorio
F
Insulin dependent diabetes in thalassaemia.
Arch Dis Child
63
1988
58
151
Dandona
P
Hussain
MAM
Varghese
Z
Politis
D
Flynn
DM
Hoffbrand
AV
Insulin resistance and iron overload.
Ann Clin Biochem
20
1983
77
152
Merkel
PA
Simonson
DC
Amiel
SA
Plewe
G
Sherwin
RS
Pearson
HA
Tamborlane
WV
Insulin resistance and hyperinsulinemia in patients with thalassemia major treated by hypertransfusion.
N Engl J Med
318
1988
809
153
Dmochowski
K
Finegood
DT
Francombe
WH
Tyler
B
Zinman
B
Factors determining glucose tolerance in patients with thalassemia major.
J Clin Endocrinol Metab
77
1993
478
154
Cavello-Perin
P
Pacini
B
Cerutti
F
Bessone
A
Condo
C
Sacchetti
L
Piga
A
Pagano
G
Insulin resistance and hyperinsulinemia in homozygous β-thalassemia.
Metabolism
44
1995
281
155
Torrance JD, Charlton RW, Schmaman A, Lynch SR, Bothwell TH: Storage iron in ‘muscle.’ J Clin Path 21:495, 1968
156
Olivieri NF, Ramachandran S, Tyler B, Bril V, Moffatt K, Daneman D: Diabetes mellitus in older patients with thalassemia major: Relationship to severity of iron overload and presence of microvascular complications. Blood 76:72a, 1990 (abstr, suppl 1)
157
Awai M, Yamanoi Y, Kuwashima J, Seno S: Induction mechanism of diabetes by ferric nitriloacetate injection, in Saltman P, Hegenauer J (eds): The Biochemistry and Physiology of Iron. Amsterdam, The Netherlands, Elsevier/North, 1982, p 543
158
Olivieri NF, Snider MA, Nathan DG, Gee B, Muroff A, Martin M, Vichinsky EP, Cohen AR: Survival following the onset of iron-induced cardiac disease in thalassemia major. Blood 86:250a, 1995 (abstr, suppl 1)
159
Perrimond
H
Michel
G
Orsini
A
Kreitman
B
Metras
D
First report of a cardiac transplantation in a patient with thalassaemia major.
Br J Haematol
78
1991
467
160
Olivieri
NF
Liu
PP
Sher
GD
Collins
AF
McCusker
PJ
Levy
G
Grieg
P
Daley
P
Francombe
WH
Butany
J
Successful combined cardiac and liver transplantation in an adult with homozygous beta-thalassemia.
N Engl J Med
330
1994
1125
161
Pippard
MJ
Measurement of iron status.
Prog Clin Biol Res
309
1989
85
162
Finch
CA
Bellotti
V
Stray
SEA
Plasma ferritin determination as a diagnostic tool.
West J Med
145
1986
657
163
Brittenham
GM
Danish
EH
Harris
JW
Assessment of bone marrow and body iron stores.
Semin Hematol
18
1981
194
164
Borgna-Pignatti
C
Castriota-Scanderbeg
A
Methods of evaluating iron stores and efficacy of chelation in transfusional hemosiderosis.
Haematologica
76
1991
409
165
Brittenham
GM
Cohen
AR
McLaren
CE
Martin
MB
Griffith
PM
Niehuis
AW
Young
NS
Allen
CJ
Farrell
DE
Harris
JW
Hepatic iron stores and plasma ferritin concentration in patients with sickle cell anemia and thalassemia major.
Am J Hematol
42
1993
81
166
Finch
C
Regulators of iron balance in humans.
Blood
84
1994
1697
167
Worwood
M
Cragg
SJ
McLaren
C
Ricketts
C
Economidou
J
Binding of serum ferritin to concanavalin A: Patients with homozygous β thalassaemia and transfusional iron overload.
Br J Haematol
46
1980
409
168
Roeser
HP
Halliday
JW
Sizemore
DEA
Serum ferritin in ascorbic acid deficiency.
Br J Haematol
45
1980
457
169
Baynes
R
Bezwoda
W
Bothwell
T
Khan
Q
Mansoor
N
The non-immune inflammatory response: Serial changes in plasma iron, iron-binding capacity, lactoferrin, ferritin and C-reactive protein.
Scand J Clin Lab Invest
46
1986
695
170
Mitnick
JS
Basniak
MA
Megibow
AJ
Karpatkin
M
Feiner
HD
CT in beta-thalassemia: Iron deposition in the liver, spleen, and lymph nodes.
AJR
136
1981
1191
171
Long
JAJ
Doppman
JL
Nienbuis
AW
Mills
SR
Computed tomographic analysis of beta-thalassemic syndromes with hemochomatosis: Pathologic findings with clinical and laboratory correlations.
J Comput Assist Tomogr
4
1980
159
172
Guyader
D
Gandon
Y
Deugnier
Y
Jouanolle
H
Loreal
O
Simon
M
Bourel
M
Carsin
M
Brissol
P
Evaluation of computed tomography in the assessment of liver iron overload. A study of 46 cases of idiopathic hemochromatosis.
Gastroenterology
97
1989
737
173
Houang
MTW
Arozena
X
Skalicka
A
Huehns
ER
Shaw
DG
Correlation between computed tomographic values and liver iron content in thalassaemia major with iron overload.
Lancet
1
1979
1322
174
Olivieri
NF
Grisaru
D
Daneman
A
Martin
DJ
Rose
V
Freedman
MH
Computed tomography scanning of the liver to determine efficacy of iron chelation therapy in thalassemia major.
J Pediatr
114
1989
427
175
Wielopolski L, Zaino EC. Noninvasive in-vivo measurement of hepatic and cardiac iron. J Nucl Med 33:1278, 1992
176
Stark
DD
Moseley
ME
Bacon
BR
Moss
AA
Magnetic resonance imaging and spectroscopy of hepatic iron overload.
Radiology
154
1985
137
177
Krocker
RM
McVeigh
ER
Hardy
P
Bronskill
MJ
Henkelman
RM
In-vivo measurement of NMR relaxation times.
Magn Reson Med
2
1985
1
178
Kessing
P
Falke
T
Steiner
R
Bloem
H
Peters
A
Magnetic resonance imaging in hemosiderosis.
Diagn Imaging Clin Med
54
1985
7
179
Gomori
JM
Grossman
RI
Drott
HR
MR relaxation times and iron content of thalassemic spleens: An in vitro study.
AJR
150
1988
567
180
Hernandez
RJ
Sarnaik
SA
Lande
I
Aisen
AM
Glazer
GM
Chenevert
T
Martel
W
MR evaluation of liver iron overload.
J Comput Assist Tomogr
12
1988
91
181
Hardy
P
Henkelman
RM
Transverse relaxation rate enhancement caused by magnetic particles.
Magn Reson Imaging
7
1989
265
182
Johnston
DL
Rice
L
Vick
GW
Hedrick
TD
Rokey
R
Assessment of tissue iron overload by nuclear magnetic resonance imaging.
Am J Med
87
1989
40
183
Kaltwasser
JP
Gottschalk
R
Schalk
KP
Hartl
W
Non-invasive quantitation of llver iron-overload by magnetic resonance imaging.
Br J Haematol
74
1990
360
184
Bonkovsky
HL
Slaker
DP
Bills
EB
Wolf
DC
Usefulness and limitations of laboratory and hepatic imaging studies in iron-storage disease.
Gastroenterology
99
1990
1079
185
Chezmar
JL
Nelson
RC
Malko
JA
Bernadino
ME
Hepatic iron overload: Diagnosis and quantification by noninvasive imaging.
Gastrointestinal Radiology
15
1990
27
186
Gomori
JM
Horev
G
Tamary
H
Zandback
J
Korneich
L
Zaizov
R
Freud
E
Krief
O
Ben-Meir
J
Rotem
H
Kuspet
M
Rosen
P
Rachmilewitz
EA
Leewenthal
E
Gorodetskey
R
Hepatic iron overload: Quantitative MR imaging.
Radiology
179
1991
367
187
Chan
PCK
Lie
P
Cronin
C
Heathcote
J
Uldall
R
The use of nuclear magnetic resonance imaging in monitoring total body iron in hemadialysis patients with hemosiderosis treated with erythropoietin and phlebotomy.
Am J Kidney Dis
19
1992
484
188
Villari
N
Caramella
D
Lippi
A
Guazelli
C
Assessment of liver iron overload in thalassemic patients by MR imaging.
Acta Radiol
4
1992
347
189
Liu P, Olivieri N, Sullivan H, Henkelman M: Magnetic resonance imaging in beta-thalassemia: Detection of iron content and association with cardiac complications. J Am Coll Cardiol 21:491, 1993 (abstr)
190
Jenson
PD
Jensen
FT
Christensen
T
Ellegaard
J
Non-invasive assessment of tissue iron overload in the liver by magnetic resonance imaging.
Br J Haematol
87
1993
171
191
Jensen
PD
Jensen
FT
Christensen
T
Ellegaard
J
Evaluation of transfusional iron overload before and during iron chelation by magnetic resonance imaging of the liver and determination of serum ferritin in adult non-thalassaemic patients.
Br J Haematol
89
1995
880
192
Olivieri
NF
Koren
G
Matsui
D
Liu
PP
Blendis
L
Cameron
R
McClelland
RA
Templeton
DM
Reduction of tissue iron stores and normalization of serum ferritin during treatment with the oral iron chelator L1 in thalassemia intermedia.
Blood
79
1992
2741
193
Liu
P
Henkelman
M
Joshi
J
Hardy
P
Butany
J
Iwanochko
M
Clauberg
M
Dhar
M
Mai
D
Waien
S
Olivieri
NF
Quantitation of cardiac and tissue iron by nuclear magnetic resonance in a novel murine thalassemia-cardiac iron overload model.
Can J Cardiol
12
1996
155
194
Olson
LJ
Edwards
WD
McCall
JT
Ilstrup
DM
Gersh
BJ
Cardiac iron deposition in idiopathic hemochromatosis: Histologic and analytic assessment of 14 hearts from autopsy.
J Am Coll Cardiol
10
1987
1239
195
Fitchett
DH
Coltart
DJ
Littler
WA
Leyland
MJ
Trueman
T
Gozzard
DI
Peters
TJ
Cardiac involvement in secondary haemochromatosis: A catheter biopsy study and analysis of myocardium.
Cardiovasc Res
14
1980
719
196
Fujisawa
I
Asato
R
Nishimura
K
Togashi
K
Itoh
K
Nakano
Y
Itoh
H
Hashimoto
N
Takeuchi
J
Torizuka
K
Anterior and posterior lobes of the pituitary gland: assessment by 1.5 T MR imaging.
J Comput Assist Tomogr
11
1987
214
197
Fujisawa
I
Morikawa
M
Nakano
Y
Konishi
J
Hemochromatosis of the pituitary gland: MR imaging.
Radiology
168
1988
213
198
Berkovitch M, Milone S, Kucharzyk W, Liu P, Papadouris D, Collins AF, Olivieri NF: Differential iron deposition in the anterior pituitary and liver in homozygous beta-thalassemia: Prediction of gonadal failure by magnetic resonance imaging. Blood 82:359a, 1993 (abstr, suppl 1)
199
Cecchetti
G
Binda
A
Piperno
A
Nador
F
Fargion
S
Fiorelli
G
Cardiac alterations in 36 consecutive patients with idiopathic haemochromatosis: Polygraphic and echocardiographic evaluation.
Eur Heart J
12
1991
224
200
Valdes-Cruz
L
Reinenke
C
Rutkowski
M
Dudell
GG
Goldberg
SJ
Allen
HD
Sahn
DJ
Piomelli
S
Preclinical abnormal segmental cardiac manifestations of thalassemia major in children on transfusion-chelation therapy: Echographic alteration of left ventricular posterior wall contraction and relaxation patterns.
Am Heart J
103
1982
505
201
Olson
LJ
Baldus
WP
Tajik
AJ
Echocardiographic features of idiopathic hemochromatosis.
Am J Cardiol
60
1987
885
202
Benson L, Liu P, Olivieri N, Rose V, Freedom R: Left ventricular function in young adults with thalassemia. Circulation 80:274, 1989 (abstr)
203
Leon
MB
Borer
JS
Bacharach
SL
Green
MV
Benz
EJ Jr
Griffith
P
Nienhuis
AW
Detection of early cardiac dysfunction in patients with severe beta-thalassemia and chronic iron overload.
N Engl J Med
301
1979
1143
204
Spirito
P
Lupi
G
Melevendi
C
Vecchio
C
Restrictive diastolic abnormalities identified by Doppler echocardiography in patients with thalassemia major.
Circulation
82
1990
88
205
Liu
P
Olivieri
N
Iron overload cardiomyopathies: New insights into an old disease.
Cardiovasc Drugs Ther
8
1994
101
206
Hou
JW
Wu
MH
Lin
KH
Lue
HC
Prognostic significance of left ventricular diastolic Indexes in β-thalassaemia major.
Arch Pediatr Adolesc Med
148
1994
862
207
Lan
KC
Li
AMC
Hui
PW
Yeung
CY
Left ventricular function in β-thalassaemia major.
Arch Dis Child
64
1989
1046
208
Overmoyer
BA
McLaren
CE
Brittenham
GM
Uniformity of liver density and nonheme (storage) iron distribution.
Arch Pathol Lab Med
111
1987
549
209
Brittenham
GM
Farrell
DE
Harris
JW
Feldman
ES
Danish
EH
Magnetic-susceptibility measurement of human iron stores.
N Engl J Med
307
1982
1671
210
Brittenham
GM
Noninvasive methods for the early detection of hereditary hemochromatosis.
Ann NY Acad Sci
526
1988
199
211
Pootrakul
P
Kitcharoen
K
Yansukon
P
Wasi
P
Fucharoen
S
Charoenlarp
P
Brittenham
G
Pippard
MJ
Finch
CA
The effect of erythroid hyperplasia on iron balance.
Blood
71
1988
1124
212
Nielsen
P
Fischer
R
Engelhardt
R
Tondüry
P
Gabbe
EE
Janka
GE
Liver iron stores in patients with secondary haemosiderosis under iron chelation therapy with deferoxamine or deferiprone.
Br J Haematol
91
1995
827
213
Feder
JN
Gnirke
A
Thomas
W
Tsuchihashi
Z
Ruddy
DA
Basava
A
Dormishian
F
Domingo
Jr. R
Ellis
MC
Fullan
A
Hinton
LM
Jones
NL
Kimmel
BE
Kronmal
GS
Lauer
P
Lee
VK
Loeb
DB
Mapa
FA
McClelland
E
Meyer
NC
Mintier
GA
Moeller
N
Moore
T
Morikang
E
Prass
CE
Quintana
L
Starnes
SM
Schatzman
RC
Brunke
KJ
Drayna
DT
Risch
NJ
Bacon
BR
Wolff
R
A novel MHC class I-like gene is mutated in patients with hereditary hemochromatosis.
Nat Genet
13
1996
399
214
Cartwright
GE
Edwards
CQ
Kravitz
K
Skolnick
M
Amos
DB
Johnson
A
Buskjaer
L
Hereditary hemochromatosis: Phenotypic expression of the disease.
N Engl J Med
301
1979
175
215
Niederau
C
Fischer
R
Sonnenberg
A
Stremmel
W
Trampisch
HJ
Strohmeyer
G
Survival and causes of death in cirrhotic and in noncirrhotic patients with primary hemochromatosis.
N Engl J Med
313
1985
1256
216
Bassett
ML
Halliday
JW
Powell
LW
Value of hepatic iron measurements in early hemochromatosis and determination of the critical iron level associated with fibrosis.
Hepatology
6
1986
24
217
Niederau
C
Fischer
R
Purschel
A
Stremmel
W
Haussinger
D
Strohmeyer
G
Long-term survival in patients with hereditary hemochromatosis.
Gastroenterology
110
1996
1107
218
Loreal
O
Deugnier
Y
Moirand
R
Lauvin
L
Guyader
D
Jouanolle
H
Turlin
B
Lescoat
G
Brissot
P
Liver fibrosis in genetic hemochromatosis. Respective roles of iron and non-iron related factors in 127 homozygous patients.
J Hepatol
16
1992
122
219
Olivieri
NF
Berriman
AM
Davis
SA
Tyler
BJ
Ingram
J
Francombe
WH
Continuous intravenous administration of deferoxamine in adults with severe iron overload.
Am J Hematol
41
1992
61
220
Lai E, Belluzzo N, Muraca MF, Daneman R, Cao A, De Virgiliis S, Lisci V, Galanello R, Olivieri NF: The prognosis for adults with thalassemia major: Sardinia, 1995. Blood 86:251a, 1995 (abstr, suppl 1)
221
Angelucci
E
Baronciani
D
Lucarelli
G
Baldassarri
M
Galimberti
M
Giardini
C
Martinelli
F
Polchi
P
Posizzi
V
Ripalti
M
Nuretto
P
Needle liver biopsy in thalassaemia: Analyses of diagnostic accuracy and safety in 1184 consecutive biopsies.
Br J Haematol
89
1994
757
222
Berkovitch M, Collins AF, Papadouris D, Wesson D, Sirna JB, Brittenham GB, Olivieri NF: Need for early, low-dose chelation therapy in young children with transfused homozygous β thalassemia. Blood 82:359a, 1993 (abstr, suppl 1)
223
DeVirgiliis S: Personal communication. Sardinia, July 1995
224
Porter
J
Huehns
E
The toxic effects of desferrioxamine.
Balliere's Clin Hematol
2
1989
459
225
Olivieri
NF
Buncic
R
Chew
E
Gallant
T
Harrison
RV
Keenan
N
Logan
W
Mitchell
D
Ricci
G
Skarf
B
Taylor
M
Freedman
MH
Visual and auditory neurotoxicity in patients receiving subcutaneous deferoxamine infusions.
N Engl J Med
314
1986
869
226
Porter
JB
Jaswon
MS
Huehns
ER
East
CA
Hazell
JWP
Desferrioxamine ototoxicity: Evaluation of risk factors in thalassaemic patients and guidelines for safe dosage.
Br J Haematol
73
1989
403
227
Bloomfield
SE
Markenson
AI
Miller
DR
Peterson
CM
Lens opacities in thalassemia.
J Pediatr Ophtholmol Strab
15
1978
154
227a
Marsh M, Holbrook I, Clark C, Shaffer J: Tinnitus in a patient with beta thalassaemia intermedia on long term treatment with desferrioxamine. Postgrad Med J 57:582, 1981
228
Porter
J
Huehns
E
The toxic effects of desferrioxamine.
Bailliere's Clin Hematol
2
1989
459
229
Davies
SC
Hungerford
JL
Arden
GB
Marcus
RE
Miller
MH
Huehns
ER
Ocular toxicity of high-dose intravenous desferrioxamine.
Lancet
2
1983
181
230
Borgna-Pignatti
C
De Stefano
P
Broglia
AM
Visual loss in a patient on high-dose subcutaneous desferrioxamine.
Lancet
1
1984
681
231
Orton
R
Veber
L
Sulh
II
Ocular and auditory toxicity of high dose subcutaneous deferoxamine therapy.
Can J Ophthalmol
20
1985
153
232
Rahi
AHS
Hungerford
JL
Ahmed
A
Ocular toxicity of desferrioxamine: Light microscopic, histochemical and ultrastructural findings.
Br J Ophtholmol
70
1986
373
233
Dickerhoff
R
Acute aphasia and loss of vision with desferrioxamine overdose.
Am J Ped Hematol Oncol
9
1987
287
234
De Virgiliis
S
Turco
MP
Frau
F
Dessi
C
Argiolu
F
Sorcinelli
R
Sitzia
A
Cao
A
Depletion of trace elements and acute ocular toxicity induced by desferrioxamine in patients with thalassaemia.
Arch Dis Chil
63
1988
250
235
Pall
H
Blake
D
Winyard
P
Lunee
J
Williams
A
Good
P
Kritzinger
E
Lornish
A
Hider
R
Ocular toxicity of desferrioxamine: An example of copper promoted auto-oxidative damage.
Br J Ophthalmol
73
1989
42
236
Giardina PJ, Nealon N, McQueen M, Martin M, Schotland D, Cohen A: Sensorimotor neuropathy associated with high dose desferrioxamine. Blood 78:199a, 1993 (abstr, suppl 1)
237
Koren
G
Bentur
Y
Strong
D
Harvey
E
Klein
J
Baumal
R
Spielberg
SP
Freedman
MH
Acute changes in renal function associated with deferoxamine therapy.
Am J Dis Child
143
1989
1077
238
Koren
G
Kochavi-Atiya
Y
Bentur
Y
Olivieri
NF
The effects of subcutaneous deferoxamine administration on renal function in thalassemia major.
Int J Haematol
54
1992
371
239
Freedman
MH
Olivieri
NF
Grisaru
D
Mcluskey
I
Thorner
P
Pulmonary syndrome in patients receiving intravenous deferoxamine infusions.
Am J Dis Child
144
1990
565
240
Tenenbein
M
Kowalski
S
Sienko
A
Bowden
DH
Adamson
IYR
Pulmonary toxic effects of continuous desferrioxamine administration in acute iron poisoning.
Lancet
339
1992
699
241
Brill
PW
Winchester
P
Giardina
PJ
Cunningham-Rundles
S
Desferrioxamine-induced bone dysplasia in patients with thalassaemia major.
Am J Roentgenol
156
1991
561
242
Orxincolo
C
Scutellari
PN
Castaldi
G
Growth plate injury of the long bones in treated β-thalassemia.
Skeletal Radiol
21
1992
39
243
Sher GD, Belluzzo N, Babyn P, Collins AF, Bailey JD, Olivieri NF: Improvement in deferoxamine-induced bony abnormalities in transfusion-dependent patients following withdrawal or reduction of deferoxamine and initiation of the oral chelator L1. Blood 82:360a, 1993 (abstr, suppl 1)
244
Hartkamp
MJ
Babyn
PS
Olivieri
NF
Spinal deformities in deferoxamine-treated beta-thalassemia major patients.
Ped Radiol
23
1993
525
245
Hatori
M
Sparkman
J
Teixeira
CC
Grynpas
M
Nervina
J
Olivieri
N
Shapiro
IM
Effects of deferoxamine on chondrocyte alkaline phosphatase activity: pro-oxidant role of deferoxamine in thalassemia.
Calcif Tissue Int
57
1995
229
246
Olivieri NF, Basran RK, Talbot AL, Babyn P, Bailey JD: Abnormal growth in thalassemia major associated with deferoxamine-induced destruction of spinal cartilage and compromise of sitting height. Blood 86:482a, 1995 (abstr, suppl 1)
247
De Sanctis V, Pinamonti A, Di Palma A, Sprocati M, Atti G, Ganberini MR, Vullo C: Growth and development in thalassaemia major patients with severe bone lesions due to desferrioxamine. Eur J Pediatr 1996 (in press)
248
O'Brien
RT
Ascorbic acid enhancement of desferrioxamine-induced urinary iron excretion in thalassemia major.
Ann NY Acad Sci
232
1974
221
249
Cohen
A
Cohen
IJ
Schwartz
E
Scurvy and altered iron stores in thalassemia major.
N Engl J Med
304
1981
158
250
Chapman
RWG
Hussein
MAM
Gorman
A
Laulicht
M
Politis
D
Flynn
DM
Sherlock
S
Hoffbrand
AV
Effect of ascorbic acid deficiency on serum ferritin concentrations in patients with β-thalassaemia major and iron overload.
J Clin Pathol
35
1982
487
251
Hussain
MAM
Flynn
DM
Green
N
Hoffbrand
AV
Effect of dose, time, and ascorbate on iron excretion after subcutaneous desferrioxamine.
Lancet
1
1977
977
252
Bothwell
TH
Bradlow
BA
Jacobs
P
Keeley
K
Kramer
S
Seftel
H
Zail
S
Iron metabolism in scurvy with special reference to erythropoiesis.
Br J Haematol
10
1964
50
253
Bridges
KR
Hoffman
KE
The effects of ascorbic acid on the intracellular metabolism of iron and ferritin.
J Biol Chem
261
1986
14273
254
Henry
W
Echocardiographic evaluation of the heart in thalassemia major.
Ann Intern Med
91
1979
892
255
Nienhuis
AW
Vitamin C and iron.
N Engl J Med
304
1981
170
256
McClaren
CJ
Bett
JHN
Nye
JA
Halliday
JW
Congestive cardiomyopathy and hemochromatosis — Rapid progression possibly accelerated by excessive ingestion of ascorbic acid.
Aust NZ J Med
12
1982
187
257
Rowbotham
B
Roeser
HP
Iron overload associated with congenital pyruvate kinase deficiency and high dose ascorbic acid ingestion.
Aust NZ J Med
14
1984
667
258
Olivieri NF, Brittenham GM, Armstrong SAM, Basran RK, Daneman R, Daneman N, Iwanchko RM, Talbot AL, Koren G: First prospective randomized trial of the iron chelators deferiprone and deferoxamine. Blood 86:249a, 1995 (abstr, suppl 1)
259
Piga A, Magliano M, Bianco L, Capalbo P, Baccaccini R, Gabutti V: Compliance with chelation therapy in Torino, in Sirchia G, Zanella A (eds): Thalassaemia Today: Second Mediterranean Meeting on Thalassaemia: Milano, Italy, Policlinico di Milano, 1987, p 141
260
Zani
B
Di Palma
A
Vullo
C
Psychosocial aspects of chronic illness in adolescents with thalassaemia major.
J Adolescence
18
1995
387
261
Borgna-Pignatti C, Cohen AR: An alternative method of subcutaneous deferoxamine administration. Blood 86:483a, 1995 (abstr, suppl 1)
262
Hallaway
PE
Eaton
JW
Panter
SS
Hedlund
BE
Modulation of deferoxamine toxicity and clearance by covalent attachment to biocompatible polymers.
Proc Natl Acad Sci USA
86
1989
10108
263
Mahoney
JR
Hallaway
PE
Hedlund
BE
Eaton
JW
Acute iron poisoning. Rescue with macromolecular chelators.
J Clin Invest
84
1989
1362
264
Hedlund B: Personal communication, Seattle, WA, December 1995
265
Olivieri NF, Nisbet-Brown E, Srichairatanakool S, Dragsten P, Hallaway P, Hedlund B, Porter JB: Studies of iron excretion and non-transferrin-bound plasma iron following a single infusion of hydroxyethyl starch-deferoxamine: A new approach to iron chelation therapy. Blood 88:310a, 1996 (abstr, suppl 1)
266
Cossu
P
Toccafondi
C
Vardeu
F
Sanna
G
Frau
F
Lobrano
R
Cornacchia
G
Nucaro
A
Bertolino
F
Loi
A
DeVergillis
S
Cao
A
Iron overload and desferrioxamine chelation therapy in beta thalassemia intermedia.
Eur J Pediatr
137
1981
267
267
Pippard
MJ
Callender
ST
Warner
GT
Weatherall
DJ
Iron absorption and loading in beta-thalassaemia intermedia.
Lancet
2
1979
819
268
Galanello R: Personal communication, Cagliari, Sardinia, January 1996
269
Lucarelli
G
Galimberti
M
Polchi
P
Angelucci
E
Baronciani
D
Giardini
C
Andreani
M
Agostinelli
F
Albertini
F
Clift
RA
Marrow transplantation in patients with thalassemia responsive to iron chelation therapy.
N Engl J Med
329
1993
840
270
Muretto
P
Del Fiasco
S
Angelucci
E
De Rosa
F
Lucarelli
G
Bone marrow transplantation in thalassemia: Modifications of hepatic iron overload and associated lesions after long-term engrafting.
Liver
14
1994
14
271
Giardini
C
Galimberti
M
Lucarelli
G
Polchi
P
Angelucci
E
Baronciani
D
Gaziev
D
Erer
B
La Nasa
G
Barbanti
I
Muretto
P
Desferrioxamine therapy accelerates clearance of iron deposits after bone marrow transplantation for thalassaemia.
Br J Haematol
89
1995
868
272
Brittenham
GM
Pyridoxal isonicotinoyl hydrazone: Effective iron chelation after oral administration.
Ann NY Acad Sci
612
1990
315
273
Porter JB, Singh S, Epemolu RO, Ackerman R, Huehns ER, Hider RC: Oral efficacy and metabolism of 1,2-diethyl-3-hydroxypyridin-4-one in thalassemia major. Blood 78:207a, 1991 (abstr, suppl 1)
274
Grady RW, Giardina PJ, Salbe AD, Hilgartner MW: A clinical trial of HBED: An orally effective iron chelator. Blood 82:359a, 1993 (abstr, suppl 1)
275
Hider RC, Kontoghiorghes GJ, Silver J: U.K. Patent: GB-2118176, 1982
276
Kontoghiorghes
GJ
The study of iron mobilization from transferrin using α-ketohydroxy heteroaromatic chelators.
Biochem Biophys Acta
869
1986
141
277
Kontoghiorghes
GJ
Iron mobilization from ferritin using oxohydroxy heteroaromatic chelators.
Biochem J
233
1986
299
278
Porter
JB
Morgan
J
Hoyes
KP
Burke
LC
Huehns
ER
Hider
RC
Relative oral efficacy and acute toxicity of hydroxypyridin-4-one iron chelators in mice.
Blood
76
1990
2389
279
Kontoghiorghes
GJ
New orally active iron chelators.
Lancet
1
1985
817
280
Kontoghiorghes
GJ
Hoffbrand
AV
Orally active α-ketohydroxypyridine iron chelators intended for clinical use: In vivo studies in rabbits.
Br J Haematol
62
1986
607
281
Venkataram
S
Rahman
YE
Studies of an oral iron chelator: 1,2-dimethyl-3-hydroxypyrid-4-one. I. Iron excretion in rats: Development of a new rapid microwave method for iron analysis in faeces.
Br J Haematol
75
1990
274
282
Hershko
C
Link
G
Pinson
A
Avramovici-Grisaru
S
Sarel
S
Peter
HH
Hider
RC
Grady
RW
New orally effective iron chelators: animal studies.
Ann NY Acad Sci
612
1990
351
283
Porter
JB
Hoyes
KP
Abeysinghe
RD
Brooks
PN
Huehns
ER
Hider
RC
Comparison of the subacute toxicity and efficacy of 3-hydroxypyridin-4-one iron chelators in iron overloaded and nonoverloaded mice.
Blood
78
1991
2727
284
Zevin
S
Link
G
Grady
RW
Hider
RC
Peter
HH
Hershko
C
Origin and fate of iron mobilized by the 3-hydroxypyridin-4-one oral iron chelators: Studies in hypertransfused rats by selective radioiron probes of reticuloendothelial and hepatocellular iron stores.
Blood
79
1992
248
285
Bergeron
RJ
Streiff
RR
Weigand
J
Luchetta
G
Creary
EA
Peter
HH
A comparison of the iron-clearing properties of 1,2-dimethyl-3-hydroxypyrid-4-one, 1,2-diethyl-3-hydroxypyrid-4-one, and deferoxamine.
Blood
79
1992
1882
286
Olivieri
NF
Koren
G
Hermann
C
Bentur
Y
Chung
D
Klein
J
St Louis
P
Freedman
MH
McClelland
RA
Templeton
DM
Comparison of oral iron chelator L1 and desferrioxamine in iron-loaded patients.
Lancet
336
1990
1275
287
Kontoghiorghes
GJ
Aldouri
MA
Sheppard
LN
Hoffbrand
AV
1,2-dimethyl-3-hydroxypyrid-4-one, an orally active chelator for treatment of iron overload.
Lancet
1
1987
1294
288
Brittenham
GM
Development of iron-chelating agents for clinical use.
Blood
80
1992
569
289
Collins
AF
Fassos
FF
Stobie
SS
Lewis
N
Shaw
D
Fernandes
D
Fry
M
Templeton
DM
Koren
G
Olivieri
NF
Iron balance and dose response studies of the oral iron chelator 1,2-dimethyl-3-hydroxypyrid-4-one (L1) in iron-loaded patients with sickle cell disease.
Blood
83
1994
2329
290
Kontoghiorghes
GJ
Effective chelation of iron in β thalassaemia with the oral chelator 1,2-dimethyl-3-hydroxypyrid-4-one.
Br J Med
295
1987
1509
291
Tondury
P
Kontoghiorghes
GJ
Ridolfi-Luthy
AR
Hirt
A
Hoffbrand
AV
Lottenbach
AM
Sonderegger
T
Wagner
HP
L1(1,2-dimethyl-3-hydroxypyrid-4-one) for oral iron chelation in patients with beta-thalassaemia major.
Br J Haematol
76
1990
550
292
Agarwal
MB
Viswanathan
C
Ramanathan
J
Massil
DE
Shah
S
Supte
SS
Vasandani
D
Puniyani
RR
Oral iron chelation with L1.
Lancet
335
1990
601
293
Kontoghiorghes
GJ
Bartlett
AN
Hoffbrand
AV
Goddard
JG
Sheppard
L
Barr
J
Nortey
P
Long-term trial with the oral iron chelator 1,2-dimethyl-3-hydroxypyrid-4-one (L1).
Br J Haematol
76
1990
295
294
Al-Refaie
FN
Wonke
B
Hoffbrand
AV
Wickens
DG
Nortey
P
Kontoghiorghes
GJ
Efficacy and possible adverse effects of the oral iron chelator 1,2-dimethyl-3-hydroxypyrid-4-one (L1) in thalassemia major.
Blood
80
1992
592
295
Agarwal
MB
Gupte
SS
Viswanathan
C
Vasandani
D
Ramanathan
J
Desai
N
Puniyani
RR
Chhablani
AT
Long-term assessment of efficacy and safety of L1, an oral iron chelator, in transfusion-dependent thalassaemia: Indian trial.
Br J Haematol
82
1992
460
296
Kontoghiorghes
GJ
Design, properties, and effective use of the oral chelator L1 and other α-ketohydroxypyridines in the treatment of transfusional iron overload in thalassemia.
Ann NY Acad Sci
612
1990
339
297
Porter
JB
Hoyes
KP
Abeysinghe
R
Huehns
ER
Hider
RC
Animal toxicology of iron chelator L1 [letter].
Lancet
2
1989
156
298
Biesemeier JA, Laveglia J: 14-day oral toxicity study in dogs with 1,2-dimethyl-3-hydroxypyrid-4-one (DMHP, L1). Food and Drug Research Laboratories, Waverly, NY, Contract No NO1-DK-4-2255, NIDDK, NIH, USA, 1991
299
Berkoukas
VA
Bentley
P
Frost
H
Schnebli
HP
Toxicity of oral iron chelator L1 [letter].
Lancet
341
1993
1088
300
Berkovitch
M
Laxer
RM
Inman
R
Koren
G
Pritzker
KP
Fritzler
MJ
Olivieri
NF
Arthropathy in thalassemia patients receiving deferiprone.
Lancet
343
1994
1471
301
Hoffbrand
AV
Bartlett
AN
Veys
PP
O'Connor
NTJ
Kontoghiorghes
GJ
Agranulocytosis and thrombocytopenia in patient with Blackfan-Diamond anaemia during oral chelator trial [letter].
Lancet
2
1989
457
302
Goudsmit R, Kersten MJ: Long term treatment of transfusion hemosiderosis with the oral chelator L1. Drugs of Today 28:133, 1992 (suppl A)
303
Agarwal MB, Gupte SS, Viswanathan C, Vasandani D, Desai N, Chablani AT: Long term efficacy and toxicity of L1-oral chelator in transfusion dependent thalassaemics over the last three years. Abstracts of the Fifth International Conference on Thalassaemias and Haemoglobinopathies, Nicosia, Cyprus, 1993, p 192
304
al-Refaie
FN
Wonke
B
Hoffbrand
AV
Deferiprone-associated myelotoxicity.
Eur J Haematol
53
1994
298
305
Hoffbrand
AV
Oral iron chelators.
Semin Hematol
33
1996
1
306
Mehta
J
Singhal
S
Chablani
A
Revankar
R
Walvalkar
A
L1-induced systemic lupus erythematosus.
Indian J Hematol Blood Transf
9
1991
33
307
Mehta
J
Singhal
S
Revankar
R
Walvalkar
A
Chablani
A
Mehta
BC
Fatal systemic lupus erythematosus in patient taking oral iron chelator L1 [letter].
Lancet
337
1991
298
308
Pattanapanyasat
K
Webster
HK
Tongtawa
P
Kongcharoen
P
Hider
RC
Effect of orally active hydroxypyridinone iron chelators on human lymphocyte function.
Br J Haematol
82
1992
431
309
Mehta
J
Singhal
S
Mehta
BC
Deaths in patients receiving oral iron chelator L1 [letter].
Br J Haematol
85
1993
430
310
Agarwal
MB
Gupte
SS
Viswanathan
C
Vasandani
D
Ramanathan
J
Desai
N
Puniyani
RR
Chhablani
AT
Deaths in patients receiving oral iron chelator L1 [letter].
Br J Haematol
85
1993
430
311
al-Refaie
FN
Wonke
B
Wickens
DG
Aydinok
Y
Fielding
A
Hoffbrand
A
Zinc concentration in patients with iron overload receiving oral iron chelator 1,2-dimethyl-3-hydroxypyrid-4-one or desferrioxamine.
J Clin Pathol
47
1994
657
312
al-Refaie
FN
Hershko
C
Hoffbrand
AV
Kosaryan
M
Olivieri
NF
Tondury
P
Wonke
B
Results of long-term deferiprone (L1) therapy. A report by the International Study Group on Oral iron Chelators.
Br J Haematol
91
1995
224
313
Olivieri NF for the Toronto Iron Chelation Group: Long-term followup of body iron in patients with thalassemia major during therapy with the orally active iron chelator deferiprone (L1). Blood 88:310a, 1996 (abstr, suppl 1)
314
Olivieri NF for the Toronto Iron Chelation Group: Randomized trial of deferiprone (L1) and deferoxamine in thalassemia major. Blood 88:651a, 1996 (abstr, suppl 1)
Sign in via your Institution