• Non ci sono risultati.

Intravenous iron administration in iron deficiency anemia: a multicentre retrospective observational study comparing the effects of ferric carboxymaltose and sodium ferric gluconate

N/A
N/A
Protected

Academic year: 2021

Condividi "Intravenous iron administration in iron deficiency anemia: a multicentre retrospective observational study comparing the effects of ferric carboxymaltose and sodium ferric gluconate"

Copied!
26
0
0

Testo completo

(1)

UNIVERSITÀ DEGLI STUDI DI PISA

Scuola di Specializzazione in Patologia Clinica e Biochimica Clinica

TESI DI SPECIALIZZAZIONE

Intravenous iron administration in iron deficiency anemia: a

multicentre retrospective observational study comparing the

effects of ferric carboxymaltose and sodium ferric gluconate

RELATORE

Dott. Alessandro Mazzoni

CANDIDATA

Dott.ssa Luisa Coluccia

(2)
(3)

3

I

NDEX

A

BSTRACT

p. 4

B

ACKGROUND

p. 6

M

ATERIALS AND METHODS

Study overview

p. 8

Study arms and outcome measures

p. 8

Statistical analysis

p. 9

R

ESULTS

Baseline data

p. 10

Iron exposure

p. 11

Primary endpoint

p. 11

Secondary endpoints

p. 13

Adverse events and transfusion need

p. 15

D

ISCUSSION

p. 18

(4)

4

ABSTRACT

BACKGROUND. Appropriate therapeutic management of iron deficiency anemia

(IDA) is essential to prevent serious health risks and indiscriminate exposure to allogeneic blood transfusions. Iron therapy is administered intravenously in patients failing to respond to oral iron treatment or needing rapid iron repletion. Several parenteral iron carbohydrate complex formulations are now available, classified as stable, such as ferric carboxymaltose, or labile, such as sodium ferric gluconate, according to the release rate of iron, directly taken up by transferrin and other proteins. The first one preparations allow clinically well-tolerated administration of a single much higher dose of intravenous iron by more rapid infusion than the second one and various therapeutic areas can take advantages of their use, as appropriate therapy for IDA correction.

STUDY DESIGN AND METHODS. In this retrospective study, we considered 346

patients with IDA, eligible for the intravenous treatment. 222 patients (group A) received an average of three intravenous injections of ferric carboxymaltose (500 mg / visit), other 124 patients (group B) received an average of ten infusions of sodium ferric gluconate (62,5 mg / visit). Patients were assessed at first visit and at least after 2, 5 and 7 weeks from the beginning of the treatment. The primary endpoint was the difference in the mean Hb, ferritin and transferrin saturation values detected throughout the whole observation period between the two treatment groups. Drug-related adverse events and transfusion need were also evaluated.

(5)

5

RESULTS. Significantly lower baseline levels of Hb, ferritin and transferrin

saturation (TSAT) were detected in group A, than in group B. Nevertheless, we found a faster and greater Hb, ferritin and TSAT increase with ferric carboxymaltose dosing regimen compared with sodium ferric gluconate. Throughout the whole observational period, group A showed a mean Hb level of 11,3 g/dL (95% CI 11,1 – 11,6), a mean ferritin level of 221,1 ng/mL (95% CI 184,4 – 257,7) and a mean TSAT of 20,7% (95% CI 18,9 – 22,5); group B showed a mean Hb level of 10,6 g/dL (95% CI 10,4 – 10,8), a mean ferritin level of 85,4 ng/mL (95% CI 50,9 – 119,9) and a mean TSAT of 14,1% (95% CI 11,8 – 16,4). After drug administration, statistical analysis showed a significant difference between the two groups in terms of mean Hb level (0,74 g/dL, 95% CI 0,42 – 1,06, p<0,0001), mean ferritin level (135,7 ng/mL, 95% CI 85,3 – 186,1, p<0,0001) and mean TSAT (6,6%, 95% CI 3,7 – 9,6, p<0,0001). The occurrence of intravenous iron-related adverse drug effects was very low and the difference among the two groups was not statistically significant (p=0,8). No serious adverse events were witnessed. Patients requiring transfusions after the beginning of the treatment decreased and were 1.4% in group A and 4.8% in group B (p=0,05). Even patients with baseline Hb ≤ 8 g/dL showed a greater increase in Hb concentration in group A compared to group B and a very low proportion of them needed transfusions.

CONCLUSION. The results of this study suggest that ferric carboxymaltose can be

used safely and more effectively than sodium ferric gluconate to rapidly improve and maintain raised Hb concentration and TSAT values after drug withdrawal in patients with iron-restricted erythropoiesis, avoiding their exposure to the potential risks of untreated anemia, unnecessary transfusions or both.

(6)

6

Background

Several burgeoning data, including analysis of benchmark studies (1-2), suggest that there is a sleeping giant of undertreated patients with an unsafe condition of iron deficiency anemia (IDA) (3), for which intravenous iron supplementation should be warranted. Eligibility criteria for this therapy are: oral iron non-compliance / intolerance or ineffectiveness (4); severe IDA in pregnancy (beyond the first trimester) (5-6) and postpartum (7-8); excessive bleeding (9-10); surgeries that decrease iron absorption (gastrectomy, duodenal bypass) (11); gastrointestinal tract disorders (active inflammatory bowel diseases; H. pylori colonization; impaired gastric acid secretion; coeliac disease) (12-16); erythropoietic-stimulating agents (ESA) therapy in chronic kidney disease (CKD) (17-18) and in cancer (19-21);

CKD (irrespective of ESA) (22); genetic iron refractory IDA (IRIDA) (23); unaccepted blood transfusions for religious issues; a clinical need for a rapid iron supply, closely related to functional capacity, e.g. in the perioperative setting (24); and acute event-related anaemia (AERA), in critical illness and tissue damage for the changes in iron metabolism induced by inflammatory effect (25).

For all these clinical and surgical settings, different formulations of parenteral iron are now available, consisting of a mineral core of iron oxohydroxide, surrounded by a carbohydrate shell, that determine their pharmacological and biological differences. Following injection, stable complexes, with high molecular weight and structural homogeneity, such as ferric carboxymaltose (FCM) (molecular mass of 150,000 Daltons), slowly deliver iron to transferrin, only via macrophages

(7)

7

endocytosis and consequent controlled export (26). These preparations permit rapid and larger dose (up to 1000 mg / visit) infusions to be administered over short therapeutic course (27).

In less stable preparations, such as sodium ferric gluconate (SFG) (molecular mass of 37,500 Daltons), instead, iron is rapidly released from carbohydrate complex and only if they are given slowly in small doses (up to 62,5 - 125 mg / visit ) is the iron taken up, at first, via endocytosis by the macrophages and not directly by transferrin. If transferrin was quickly fully saturated, there would be large amounts of toxic non-transferrin bound iron in the serum, responsible for lipid peroxidation, reactive oxygen species formation (28) and, as a consequence, systemic side effects (29). The typical therapeutic course of such labile preparations requires up to 5 injections and so administration

times are markedly longer (30) than for stable complexes.

The primary objective of this study was to evaluate the efficacy and safety of two different forms of parenteral iron, FCM compared with SFG, in patients with confirmed IDA of various etiologies.

(8)

8

Materials and methods

Study overview

In order to evaluate the effects of the two intravenous iron preparations, we performed a retrospective observational study, analyzing 346 patients consecutively admitted at 3 different centers for intravenous iron support between January 2009 and December 2015.

Study arms and outcome measures

Subjects who received FCM, administered in an average of three infusions of 500 mg, were allocated to the first arm (Group A); subjects who received SFG, administered in an average of ten infusions of 62,5 mg, were allocated to the second arm (Group B).

All patients assigned to group A and B were clustered depending on various etiologies of IDA (pregnancy, postpartum,

heavy uterine bleeding, gastrointestinal disorders, chronic heart failure, hematological disease, chronic disease or unexplained) and baseline hemoglobin (Hb) (≤8; 8,1-10; >10 g/dL).

Laboratory tests, showing hematopoietic response and iron repletion, were obtained at the beginning of iron therapy and 2, 5 and 7 weeks later.

The primary endpoint was the difference in the mean Hb, ferritin and transferrin saturation (TSAT) values detected throughout the whole observation period from baseline to Day 49 between the two treatment groups.

Other secondary endpoints included for each group, mean changes in Hb, ferritin and TSAT from baseline to Day 14, 35 and 49; the difference in the percentage of patients achieving an increase in Hb value of at least 1 g/dL any time between baseline and Day 14; the highest Hb value observed any time from baseline to Day

(9)

9

35 and 49; mean change from baseline to the highest observed Hb value from baseline to Day 14 and 49, and proportion of patients requiring transfusion, in the cluster of patients with Hb≤8 g/dL, allocated to both treatment groups.

Treatment-emergent adverse events and transfusion requirements in both groups, throughout the whole observation period of 7 weeks from the initiation of therapy, were also analyzed.

Statistical analysis

Variables with normal distribution are expressed as mean and SD, while categorical data as frequency and percentage. Between groups, comparisons of data were performed by t-test for unpaired data or the Mann-Whitney U-test for continuous variables, and by the chi-square test for categorical variables. The independent relationship between the two forms of parenteral iron (FCM and

SFG) and laboratory parameters over time (dependent variable) was investigated by a multiple linear mixed model and data were expressed as regression coefficient, 95% confidence interval and p value. The significance level was set at p≤0,05.

(10)

10

Results

Baseline data

A total of 346 patients, affected by IDA, were allocated to the two different groups: 222 supported with FCM to group A, and 124 supported with SFG to group B.

Baseline characteristics of these patients are listed in Table 1. Statistically significant differences were detected in laboratory data between the two groups, group A exhibiting lower mean values for Hb, ferritin and TSAT than group B.

Table 1. Demographic characteristics*

Group A: FCM (n=222) Group B: SFG (n=124) p value Age (years) 58.4±17.7 55.8±18.4 0.29 Sex Female Male 171 (77.0) 51 (23.0) 98 (79.0) 26 (21.0) 0.67 Etiology of IDA

Pregnancy, post-partum, HUB Gastrointestinal disorders Chronic disease Chronic heart failure Hematological disease Unexplained 69 (32.4) 70 (32.9) 35 (16.4) 4 (1.9) 6 (2.8) 29 (13.1) 23 (18.5) 31 (25.0) 22 (17.7) 1 (0.8) 10 (8.1) 37 (29.8) <0.0001 Baseline Hb (g/dL) 8.7±1.2 9.4±1.4 <0.0001 Hb category (g/dL) ≤8 8.1-10 >10 66 (30.4) 124 (57.1) 27 (12.4) 26 (21.0) 55 (44.4) 43 (34.7) <0.0001 Baseline ferritin (ng/mL) 11.1±29.3 14.2±26.6 0.0003 Baseline TSAT (%) 4.5±3.5 6.2±5.2 0.0006 Previous ABT requirement 14 (6.3) 25 (20.2) <0.0001 Previous iron therapy 96 (43.2) 56 (45.2) 0.7303 Active bleeding 41 (18.6) 28 (22.6) 0.37 *Data are expressed as means ± SD or as number (%)

ABT=allogeneic blood transfusion; FCM=ferric carboxymaltose; HUB=heavy uterine bleeding; IDA=iron deficiency anemia; SFG=sodium ferric gluconate; TSAT=transferrin saturation.

(11)

11

Iron exposure

Although the Ganzoni formula (31) is the best way to calculate patient’s total body iron deficit and to fully replete the iron stores, in clinical practice we utilized approved product labels (13), stating specific dosing regimens, to estimate intravenous iron needs.

The mean cumulative dose of intravenous iron received from subjects was significantly higher in group A than group B: 1439,4 (±536,9) mg versus 632,4 (±344) mg (p<0,0001); while the proportion of patients treated with iron therapy before baseline was similar between groups: 43,2% in group A and 45,2% in group B (p=0,7303).

Primary endpoint

Group A was associated with a faster and greater increase in Hb, ferritin and TSAT level than group B, and the difference between the two groups reached the

maximum from baseline to Day 28 (Figure 1) for Hb level (2,47 g/dL; 95% CI 1,68-3,27); to Day 7 (Figure 2) for ferritin level (322,4 ng/ml; 95% CI 187,7-457,2) and to Day 14 (Figure 3) for TSAT value (21,9%; 95% CI 13,5-30,4).

Mean difference in mean Hb, ferritin and TSAT values detected throughout the whole observation period between Group A and Group B was statistically significant and respectively: 0,74 g/dL (95% CI 0,42 -1,06), p<0,0001; 135,7 ng/mL (95% CI 85,3-186,1), p<0,0001; and 6,6% (95% CI 3,7 -9,6), p<0,0001; being mean Hb, ferritin and TSAT value observed in this period higher in group A than group B: respectively 11,3 g/dL (95% CI 11,1 - 11,6), 221,1 ng/ml (95% CI 184,4 - 257,7) and 20,7% (95% CI 18,9 - 22,5) versus 10,6 g/dL (95% CI 10,4 - 10,8), 85,4 ng/mL (95% CI 50,9 - 119,9) and 14,1% (95% CI 11,8 - 16,4).

(12)

12

Figure 1. Effects of ferric carboxymaltose (Group A, red line) and sodium ferric gluconate

(Group B, blue line) on hemoglobin levels throughout the observation period of the study.

Figure 2. Effects of ferric carboxymaltose (Group A, red line) and sodium ferric gluconate

(Group B, blue line) on ferritin levels throughout the observation period of the study.

Hemoglobin (g/dL)

(13)

13

Figure 3. Effects of ferric carboxymaltose (Group A, red line) and sodium ferric gluconate

(Group B, blue line) on transferrin saturation throughout the observation period of the study.

Secondary endpoints

Mean increase in Hb, ferritin, and TSAT from baseline to highest value by Day 14 was significantly greater in group A than group B: respectively, 2 (±1,2) g/dL versus 1,1(±0,9) g/dL, p=0,0057; 253,4 (±165,5) ng/ml versus 49,3 (±46,8) ng/ml, p=0,019 and 22,8 (±20,6)% versus 2,5 (±3,3)%, p=0,0398 (Table 2).

Interestingly, while disappeared at Day 35 (Table 3), such statistically significant differences between the FCM group and the SFG group, were confirmed at Day 49, when the average of Hb, ferritin and TSAT improvement from baseline was respectively 3,8 (±1,7) g/dL versus 1,2 (±1,3) g/dL, p<0,0001; 124,7 (±124,4) ng/ml versus 55,8 (±83,5) ng/ml,

(14)

14

p=0,0351 and 16,3 (±7,6) % versus 2,2 (±1,3) %, p=0,0009 (Table 4). Of note, at the end of the observational period, in

SFG group TSAT level returned close to baseline, while in FCM group TSAT remained stable, over 16%.

Table 2. Mean increase from baseline ferritin, Hb and TSAT to Day 14*

Group A Group B p value Ferritin (ng/mL) 253.4 ± 165.5 49.3 ± 46.8 0.019 Hb (g/dL) 2.0 ± 1.2 1.1 ± 0.9 0.0057 TSAT (%) 22.8 ± 20.6 2.5 ± 3.3 0.0398 *Data are expressed as mean ± SD. Hb=haemoglobin; TSAT=transferrin saturation.

Table 3. Mean increase from baseline ferritin, Hb and TSAT to Day 35*

Group A Group B p value Ferritin (ng/mL) 115.2 ± 97.9 49.5 ± 45.4 0.4939 Hb (g/dL) 2.2 ± 2.3 1.8 ± 1.8 0.5269 TSAT (%) 14.9 ± 9.5 11.9 ± 11.8 1 *Data are expressed as mean ± SD. Hb=haemoglobin; TSAT=transferrin saturation.

Table 4. Mean increase from baseline ferritin, Hb and TSAT to Day 49*

Group A Group B p value Ferritin (ng/mL) 124.7 ± 124.4 55.8 ± 83.5 0.0351 Hb (g/dL) 3.8 ± 1.7 1.2 ± 1.3 < 0.0001 TSAT (%) 16.3 ± 7.6 2.2 ± 1.3 0.0009 *Data are expressed as mean ± SD. Hb=hemoglobin; TSAT=transferrin saturation.

(15)

15

The proportion of patients who achieved an increase in Hb level of at least 1 g/dL from baseline to Day 14 was significantly greater in group A than group B: 82,1% (95% CI 66,9 - 91,2) versus 45% (95% CI 25,3 - 66,4), p=0,005.

Likewise, the highest Hb value reached from baseline to Day 35 was significantly greater in group A than group B: 11 (±1,4) g/dL versus 10,6 (±1,4) g/dL, p= 0,015; such as the highest Hb value recorded from baseline to Day 49: 11,5 (±1,7) g/dL versus 10,9 (±1,3) g/dL, p=0,0006.

In the subgroup of 92 patients with Hb level ≤8 g/dL (66 supported with FCM and 26 with SFG), we found a mean increase in Hb from baseline to Day 14 that did not significantly differ, between group A and group B: 2,4 (±1,4) g/dL versus 1,4 (±0,8) g/dL, p=0,1837. The difference between the groups became statistically significant to Day 49, when mean increase in Hb level from baseline

was 4,6 (±2,1) g/dL in the FCM group versus 2,7 (±0,8) g/dL in the SFG group (p=0,0041).

In both subgroups of severely anemic subjects, we found a very low proportion of patients requiring transfusion after the beginning of parenteral iron therapy: 3,03% in group A and 3,85% in group B (p=1). Fisher exact test was used for statistical analysis and the lack of a statistically significant difference between the two subgroups may be due to sample size that didn’t provide the power to detect a treatment difference.

Adverse events and transfusion need

There weren’t any serious adverse events in both groups and there wasn’t a statistically significant difference in the proportion of patients who experienced a possibly drug-related side effect: 5.4 % in group A versus 7.2 % in group B, p=0,0821. The most common adverse

(16)

16

events in the group A were mild hypersensitivity reactions, reversible even without therapy (2,3%) and dizziness (1,8%); conversely, the most common

adverse events in the group B were nausea/vomit (1,6%), and epigastric pain (1,6%) (Table 5).

Table 5. Adverse events*

Adverse event Group A Group B Headache 0 (0.0) 1 (0.8) Dizziness 4 (1.8) 0 (0.0) Nausea/vomit 1 (0.5) 2 (1.6) Epigastric pain 0 (0.0) 2 (1.6) Back pain 0 (0.0) 1 (0.8) Mild hypersensitivity reactions 5 (2.3) 1 (0.8) Abdominal pain 0 (0.0) 1 (0.8) Dysphagia 1 (0.5) 0 (0.0) *Data are expressed as number (%).

The proportion of patients receiving transfusions before starting intravenous therapy was significantly higher in group B than group A (20,2% versus 6,3%, p<0,0001), such us after the beginning of treatment, when subjects requiring

transfusion decreased and were 4,8% in group B versus 1,4% in group A (p=0,05) (Table 6), with 19 packed red blood cells (PRBC) units transfused in group B versus 6 in group A.

(17)

17

Table 6. Transfusion need before and after intravenous iron injection*

Group A Group B P value Patients transfused before iron injection 14 (6.3) 25 (20.2) < 0.0001 Patients transfused after iron injection 3 (1.4) 6 (4.8) 0.05 *Data are expressed as number (%).

(18)

18

Discussion

Our aim was to compare the effects of two forms of parenteral iron in patients with iron-restricted erythropoiesis.

We analyzed patients with every type of iron deficiency (ID): absolute ID, functional ID (for following treatment with ESA), and ID associated with chronic disease (ACD/IDA).

The ACD/IDA diagnosis wasn’t always straightforward, because of the lack of a widely available assay for determination of hepcidin-25 isoform (32), that, best of all, reflects iron need for erythropoiesis (33-34), without being an invasive method, like bone marrow aspiration stained with Prussian blue.

Subjects labeled “unexplained”, unresponsive to oral iron therapy, mostly “baby boomers” with a negative gastrointestinal workup, represented a sharp proportion in both groups of our

study and, after hematologic evaluation, were examined by serology for H. pylori IgG, celiac disease, serum gastrin and antiparietal/intrinsic factor antibodies, even if their noncompliance, for frequent side effects and multiple daily doses concerning to oral iron therapy (35), often affected this refractoriness.

In clinical practice, FCM stands out by the rate at which a maximum single dose can be infused safely, with fewer visits need, facilitating a rapid haematological response and iron replacement.

Our findings show that this newer formulation produces significantly greater and faster increases in Hb, ferritin and TSAT levels than SFG, maintaining raised their values even after drug withdrawal (Figure 1, 2, 3).

The low rate of adverse events of FCM was comparable to that of SFG, and supports the evidence that these preparations are safe (36), provided that

(19)

19

published European Medicines Agency’s Committee for Medicinal Products for Human Use advice (37) is followed. Infusion reactions (itching, flushing, dizziness, nausea), described in our patients and common with newer formulations (38), usually did not require any therapy and resolved without residua (39-40). Misinterpretation of the clinical nature of these mild reactions, deemed as serious adverse events and treated over-aggressively, often with vasopressors and antihistamines, fueled the historical perception of peril attributed to the intravenous iron, that novel comprehensive reviews reply (22,36). Furthermore, in terms of safety, our experience confirmes that intravenous iron administration, by readily increasing hematopoietic response rate, decreases allogeneic blood transfusion need, being proportion of subjects requiring transfusion, even among severely anemic

patients, lower in the FCM group than SFG group.

Properly administered, FCM, with an estimated incidence of serious adverse events of <1:200.000, is much safer than allogeneic blood transfusion (41), associated with events responsible for major morbidity in 1:21.413 components issued (according to the Serious Hazards of Transfusion 2012 data) (42).

In respect to infectious complications, transfusion of even a single PRBC unit may limit the immune response, induce inflammatory mediators (43) and delivers on average 200 mg of Heme Fe, which has growth-promoting effect on microorganisms (44), in contrast to commercial intravenous iron complex, that has been demonstrated in various surgical settings not to increase infection rate (45-46).

Additionally, FCM, although its higher acquisition cost than SFG, may be

(20)

20

considered a less expensive alternative (47-48). Factors to be taken into consideration include the lower frequency of infusion need and the resulting advantages both for the patient, with lower working days lost, and for the health system, with lower overhead costs: both direct (e.g., medical and nursing staff, infusion material, laboratory tests , with materials needed for sampling) and indirect (e.g., utilities, property and nonprofessional personnel), being the second ones the highest proportion of the total cost of the treatment.

Nowadays, the central purpose of health care organizations, including transfusion medicine services, is to maintain the greatest benefit for patients at lower overall cost, and for patients with iron deficiency, without hemodynamic instability, it means to actively treat underlying disease and replete iron stores for erythropoiesis, with oral and/or

intravenous iron supplementation, without adopting transfusion as front line therapy if clinically unnecessary (49).

In conclusion, our data suggest that, in appropriately selected individuals needing replenishment of iron stores, administration of FCM is an accurate approach, as safe and more effective than SFG, rapidly increasing medullary response and iron store, maintaining raised Hb and TSAT values even after drug withdrawal and decreasing transfusion need, with costs and complications related.

(21)

21

References

1. Gombotz H, Rehak PH, Shander A, Hofmann A. Blood use in elective surgery: the Austrian benchmark study. Transfusion 2007; 47: 1468-80

2. Gombotz H, Rehak PH, Shander A, Hofmann A. The second Austrian benchmark study for blood use in elective surgery: results and practice change. Transfusion 2014 ; 54: 3178-85

3. Kassebaum NJ, Jasrasaria R, Naghavi M, et al. A systematic analysis of global anemia burden from 1990 to 2010. Blood 2014; 123: 615-24

4. Bregman DB, Morris D, Koch TA, He A, Goodnough LT. Hepcidin levels predict non-responsiveness to oral iron therapy in patients with iron deficiency anemia. Am J Hematol 2013; 88:97-101 5. Khalafallah AA, Dennis AE. Iron deficiency anaemia in pregnancy and postpartum: pathophysiology and effect of

oral versus intravenous iron therapy. Journal of Pregnancy, 2012; article ID 630519

6. Pavord S, Myers B, Robinson S, Allard S, Strong J, Oppenheimer C, British Committee for Standards in Haematology. UK guidelines on the management of iron deficiency in pregnancy. Br J Haematol. 2012;156(5):588–600.

7. Breymann C, Gliga F, Bejenariu C, Strizhova N. Comparative efficacy and safety of intravenous ferric carboxymalthose in the treatment of postpartum iron deficiency anemia. Int J Gynaecol Obstet 2008; 101:67-73

8. Seid MH, Derman RJ, Baker JB, Banach W, Goldberg C, Rogers R. Ferric carboxymaltose injection in the treatment of postpartum iron deficiency anemia: a randomized controlled clinical trial. Am J Obstet Gynecol 2008; 199:435

9. Van Wyck DB, Mangione A, Morrison J, Hadley PE, Jehle JA, Goodnough LT.

(22)

22

Large-dose intravenous ferric carboxymaltose injection for iron deficiency anemia in heavy uterine bleeding: a randomized, controlled trial. Transfusion 2009; 49:2719-28.

10. Kozek-Langenecker SA, Afshari A, Albaladejo P, Santullano CA, De Robertis E, Filipescu DC, et al. Management of severe perioperative bleeding: guidelines from the European Society of Anaesthesiology. Eur J Anaesthesiol 2013 Jun; 30(6):270-382

11. Malone M, Barish C, He A, Bregman D. Comparative review of the safety and efficacy of ferric carboxymaltose versus standard medical care for the treatment of iron deficiency anemia in bariatric and gastric surgery patients. Obesity Surgery, 2013; 23(9):1413-1420

12. Gasche C, Berstad A, Befrits R et al. Guidelines on the diagnosis and management of iron deficiency and anemia in inflammatory bowel disease.

Inflammatory Bowel Disease, 2007; 13(12):1545-1553

13. Kulnigg S, Stoinov S, Simanenkov V, et al. A novel intravenous iron formulation for treatment of anemia in inflammatory bowel disease: the ferric carboxymaltose (FERINJECT) randomized controlled trial. Am J Gastroenterol 2008;103:1182-92

14. Monzon H, Fornè M, Esteve M, et al. Helicobacter pylori infection as a cause of iron deficiency anaemia of unknown origin. World J Gastroenterol. 2013; 19(26):4166-4171

15. Schade SG, Cohen RJ, Conrad ME. Effect of hydrochloric acid on iron absorption. N Engl J Med. 1968;279(13):672-674

16. Harper JW, Holleran SF, Ramakrishnan R, Bhagat G, Green PH. Anemia in celiac disease is multifactorial in etiology. Am J Hematol.2007;82(11):996-1000

(23)

23

17. Fishbane S, Frei GL, Maesaka J. Reduction in recombinant human erythropoietin doses by the use of chronic intravenous iron supplementation. Am J Kidney Dis 1995;26:41-46

18. Sunder-Plassmann G, Horl WH. Importance of iron supply for the erythropoietin therapy. Nephrol Dial Transplant 1995;10:2070-2076

19. Hedenus M, Birgegard G, Nasman P et al. Addition of intravenous iron to epoetin beta increases hemoglobin response and decreases epoetin dose requirement in anemic patients with lymphoproliferative malignancies: a randomized multicenter study. Leukemia 2007, 21(4):627-632

20. Petrelli F, Borgonovo K, Cabiddu M, et al. Addition of iron to erythropoiesis-stimulating agents in cancer patients: a meta-analysis of randomized trials. J Cancer Res Clin Oncol 2012;138:179-87

21. Auerbach M, Liang AS, Glaspy J. Intravenous iron in chemotherapy and cancer-related anemia. Community Oncology 2012;9(9):289-295

22. Lyseng Williamson KA, Keating GM. Ferric carboxymaltose: a review of its use in iron-deficiency anemia. Drugs 2009;69:739-756

23. Melis MA, Cau M, Congiu R, et al. A mutation in the TMPRSS6 gene, encoding a trans membrane serine protease that suppresses hepcidin production, in familial iron deficiency anemia refractory to oral iron. Haematologica, 2008;93(10):1473-1479

24. Leal-Noval SR, Munoz M, Asuero M, Contreras E, Garcia-Erce JA, Llau JV, Moral V, et al. Spanish Expert Panel on Alternatives to Allogeneic Blood Transfusion. Spanish Consensus Statement on alternatives to allogeneic blood transfusion: the 2013 update of the

(24)

24

“Seville Document”. Blood Transfus 2013;11(4):585-610

25. Iperen CE, Wiel van de CE, Mark JJM. Acute event related anaemia. Br J Haematol 2002;115, 739-743

26. Beshara S, Lundqvist H, Sundin J, Lubberink M, Tolmachev V et al. Pharmacokinetics and red cell utilization of iron(III)-hydroxide-sucrose complex in anaemic patients: a study using positron emission tomography. Br J Haematol 1999;104:296-302

27. Geisser P, Baer M, Schaub E. Structure/histotoxicity relationship of parenteral iron preparations. Drug Res. 1992;42:1439-1452

28. Evans RW, Rafique R, Zarea A, Rapisarda C et al. Nature of non-transferrin-bound-iron: studies on iron citrate complexes and the thalassemic sera. J Biol Inorg Chem 2008; 13:57-74 29. Chandler G, Harchowal J, Macdougall IC. Intravenous iron sucrose: establishing

a safe dose. Am J Kidney Dis.2001;38:988-991

30. Geisser P, Burckhardt. The pharmacokinetics and pharmacodynamics of iron preparations. Pharmaceutics 2011; 3(1):12-33

31. Ganzoni AM. Intravenous iron-dextran: therapeutic and experimental possibilities. Schweizerische Medizinische Wochenschrift 1970;100(7):301-303

32. Kroot JJC, Kemna EHJM, Bansal SS et al. Results of the first international round robin for the quantification of urinary and plasma hepcidin assays: need for standardization. Haematologica 2009;94(12):1748-1752

33. Ganz T. Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation. Blood 2003;102(3):783-8

34. Nemeth E, Tuttle MS, Powelson J et al. Hepcidin regulates cellular iron efflux

(25)

25

by binding to ferroportin and inducing its internalization. Science 2004;306(5704):2090-3

35. Cancelo-Hidalgo MJ, Castelo-Branco C, Palacios S et al. Tolerability of different oral iron supplements: a systematic review. Curr Med Res Opin 2013;29:291-303

36. Qunibi WY. The efficacy and safety of current intravenous iron preparations for the management of iron-deficiency anaemia: a review. Drug Res.2010; 60:399-412

37. Rampton D, Folkersen J, Fishbane S, Hedenus M, Howaldt S, Locatelli F, Patni S, Szebeni J, Weiss G. Hypersensitivity reactions to intravenous iron: guidance for risk minimization and management. Haematologica 2014; 99: 1671-1676 38. Szebeni J. Complement activation-related pseudoallergy: a new class of drug-induced acute immune toxicity. Toxicology. 2005; 216(2–3):106–21.

39. Auerbach M, Ballard H, Glaspy J. Clinical update: intravenous iron for anaemia. Lancet. 2007;369(9572):1502–4. 40. Fishbane S, Ungureanu VD, Maesaka JK, Kaupke CJ, Lim V, Wish J. The safety of intravenous iron dextran in hemodialysis patients. Am J Kidney Dis. 1996;28(4):529–34.

41. Chertow GM, Mason PD, Vaage-Nilsen O, Ahlmen J. Update on adverse drug events associated with parenteral iron. Nephrol Dial Transplant. 2006;21(2):378–82.

42. Bolton-Maggs PH, Cohen H. Serious Hazards of Transfusion (SHOT) haemovigilance and progress is improving transfusion safety. Br J Haematol. 2013;163(3):303-14

43. Ferraris VA, Davenport DL, Saha SP, Austin PC, Zwischenberger JB. Surgical outcomes and transfusion of minimal amounts of blood in the operating room. Arch Surg 2012;147:49-55

(26)

26

44. Andrews SC, Robinson AK, Rodriguez-Quinones F. Bacterial iron homeostasis. FEMS Microbiol Rev 2003;27:215-37

45. Munoz M, Gomez-Ramirez S, Cuenca J, et al. Very-short-term perioperative intravenous iron administration and postoperative outcome in major orthopedic surgery: a pooled analysis of observational data from 2547 patients. Transfusion 2014;54:289-99 46. Torres S, Kuo YH, Morris K, Neibart R, Holtz JB, Davis JM. Intravenous iron following cardiac surgery does not increase the infection rate. Surg Infect (Larchmt)2006;7:361-6

47. Calvet X, Ruiz MA, Dosal A et al. Cost-minimization analysis favours intravenous ferric carboxymaltose over ferric sucrose for the ambulatory treatment of severe iron deficiency. PLoS One 2012;7:e45604

48. Bhandari S. Update of a comparative analysis of cost minimization following the introduction of newly available intravenous iron therapies in hospital practice. Ther Clin Risk Manag 2011;7:501-9

49. Hare GMT, Tsui AKY, Ozawa S, Shander A. Anaemia: Can we define haemoglobin thresholds for impaired oxygen homeostasis and suggest new strategies for treatment? Best Practice & Research Clinical Anaesthesiology 2013;27:85-98

Riferimenti

Documenti correlati

Figure 3.18: Time resolution measured at the test beam with a small scale prototype as a function of the energy deposited in the crystals of the calorimeter prototype....

18: Novara G, Ficarra V, Mocellin S, Ahlering TE, Carroll PR, Graefen M, Guazzoni G, Menon M, Patel VR, Shariat SF, Tewari AK, Van Poppel H, Zattoni F, Montorsi F, Mottrie A, Rosen

For example, as we are going to see in the next chapter, Loop Quantum Gravity predicts some sort of intrinsic discretization of spacetime and, even thought the Minkowski limit of

implication by negation. Lambek and P.J. Introduction to higher order categorical logic. In Graduate Texts in Mathematics. Notions of computations and monads. A semantic view

Il confronto tra i dati medi di densità della rinnovazione rispetto a quelli presenti nello strato arboreo, evidenziano come da un N° iniziale di semi-frutti,

This paper thereby aims to contribute to proposed models of mindfulness for research and theory building by proposing a specific model for the unique psychological and neural

Died of leukemia relapse (2) 10 GvHD drug toxicity and occult HBV infection none expired for acute liver failure two days after biopsy 11 drug toxicity GvHD introduce steroids and