/
Monitoring and treatment iron overload in Monitoring and treatment iron overload in

Monitoring and treatment iron overload in - PowerPoint Presentation

giovanna-bartolotta
giovanna-bartolotta . @giovanna-bartolotta
Follow
429 views
Uploaded On 2015-09-25

Monitoring and treatment iron overload in - PPT Presentation

thalassaemia Professor John Porter Red Cell Disorders Unit University College London Hospitals and UCL jporteruclacuk Professor John Porter Red Cell Disorders Unit University College London ID: 139663

patients iron ferritin cardiac iron patients cardiac ferritin dfo deferasirox serum blood chelation lic day overload thalassaemia year liver years 001 patient

Share:

Link:

Embed:

Download Presentation from below link

Download Presentation The PPT/PDF document "Monitoring and treatment iron overload i..." is the property of its rightful owner. Permission is granted to download and print the materials on this web site for personal, non-commercial use only, and to display it on your personal computer provided you do not modify the materials and that you retain all copyright notices contained in the materials. By downloading content from our website, you accept the terms of this agreement.


Presentation Transcript

Slide1

Monitoring and treatment iron overload in thalassaemia

Professor John Porter

Red Cell Disorders Unit

University College London Hospitals and UCL

j.porter@ucl.ac.ukSlide2

Professor John Porter

Red Cell Disorders Unit

University College London

Hospitals

and UCL j.porter@ucl.ac.uk

M

onitoring and treatment

iron

overload

in

thalassaemiaSlide3

OutlineWhat are the treatment and monitoring options available for iron overload in Thalassaemia MajorOn what are guidelines about ferritin targets based and should we be more ambitious?What are the goals of chelation treatment ?How can monitoring help to achieve these goals?

What can be achieved ?- a personal perspectiveSlide4

Monitoring options

Iron loading rateSerum ferritinLiver Iron concentrationCardiac evaluation – function & T2* Endocrine evaluation – growth, function & MRIAdherence and quality of lifeSlide5

Highly variable iron

excretion is required to balance

transfusional iron loading in Thalassaemia Major

Iron accumulation from transfusion in TM

(n = 586)

233mls/kg/y blood (if

Hct

0.6)

about 40 units/year for a 70 kg person

0.4 ± 0.11 mg/kg/day (mean) of iron

< 0.3mg /kg day 19% of patients

0.3-0.5 mg/kg/day 61%

> 0.5 mg/kg/day

20

%

Cohen,Glimm and Porter. Blood 2008;111:583-7Slide6

Dosing to balance iron transfusional rate

Deferasirox

Deferasirox

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0

5

10

15

20

25

30

Mean total body iron excretion

± SD (mg Fe/kg/day)

Actual doses (mg/kg/day)

0

10

20

30

40

50

60

D

eferoxamine

Studies 107 and 108

Deferoxamine (5 days/week)

Average transfusion iron intake SCD

Average transfusion iron intake

thalassaemia

Cohen AR, et al. Blood. 2008;111:583-7.Slide7

Change in LIC at low defarasirox doses in NTDT

mean loading rate 0.01 mg/kg/day (primarily from increased GI absorption)Taher

, Porter,. et al Blood (2012) , 120, 970-7,

But at 10mg/kg/day, the mean LIC increased at 1y in TM with mean loading rate 0.4mg/kg/day

Cappellini

et al,

Blood

. 2006;107:3455-

3462

LIC change

(mg/g dry wt) from baseline

Ferritin change

(

ng

/ml) from baselineSlide8

Use serum ferritin measures to achieve harmless body iron levels?Clear evidence linking long-term ferritin control to outcome

Convenience and low cost Permit frequent repeated measurements Allows early trend recognition Ferritin trend is increasing; focus on adherenceconsider dose increase

chelator regime changeFerritin trend decreasing If rapid, dose adjust to minimise risks of over chelation for ‘soft landing’If levels already low- dose reduction to allow maintenance of current level

Slide9

Limitations of just using serum ferritin ? Variability in LIC accounts for only 57% of variability in serum ferritin

1 Raised by inflammation or tissue damage Lowered by vitamin C deficiency 2 Origin of serum ferritin differs above values of 4K 3 Relationship of ferritin to body iron (LIC) varies in

different diseases Low relative to LIC in Thal Intermedia 4 (hepatocellular > macrophages)

Higher and variable in SCD 5 Relationship of ferritin to LIC differs with different chelators,6,7

Brittenham et al, Am J Hematol 1993;42:81-5

Chapman et al, J

Clin

Pathol

1982;35:487-91.

Worwood

, M. 1980 Br J

Haematol

46,409-16

Origa

,

Hamatologica 2007, 92 5835. Porter & Huehns, Acta

Haematologica

Fischer et al. Brit J

Haem

2003, 121 938-948

Ai

Leen

Ang

, et al, Blood, 201, 116, Abstract 4246.

Slide10

Why monitor & control liver iron ?

Ferritin alone may not reflect true body iron and chelation trendsLIC predicts total body storage iron in TM1Absence of pathology heterozygotes

of HH where liver levels < 7 mg/g dry weightLiver pathology abnormal ALT if LIC > 17 mg/g dry weight2liver fibrosis progression if LIC > 16 mg/

g dry weight3Cardiac pathology at high levels

Increased LIC linked to risk of cardiac iron in unchelated patients 2,6LIC >15 mg/g dry weight association with cardiac deathall of 15/53 TM patients who died4improvement of subclinical cardiac dysfunction with venesection alone post-BMT

5

Angelucci

E, et al. N

Engl

J Med. 2000;343:327-

31.

Jensen

PD, et al. Blood. 2003;101:91-6.

Angelucci

E, et al. Blood. 2002;100:17-21

.

Brittenham

GM, et al. N

Engl

J Med. 1994;331:

567-73.

Mariotti

E, et al.

Br J Haematol

. 1998;103:916-21.

Buja LM, Roberts WC. Am J Med. 1971;51:209-21

ALT = alanine aminotransferase;

BMT = bone marrow transplantation. Slide11

Low Heart

T2* inreases risk of low LVEF

LVEF = left ventricular ejection fraction.

Anderson et al.

Eur Heart J

. 2001;22:2171

.

Heart T2* (ms)

LVEF (%)

90

80

70

60

50

40

30

20

10

0

0

20

40

60

80

Severe cardiac iron

Minimal liver iron

Severe liver iron

Minimal cardiac ironSlide12

Relationship between cardiac T2* and cardiac failure

Kirk P, et al. Circulation. 2009;120:1961-8.

0

0.1

0.2

0.3

0.4

0.5

0.6

0

30

60

90

120

150

180

210

240

270

300

330

360

Proportion of patients developing cardiac failure

Follow-up time (days)

< 6 ms

6–8 ms

8–10 ms

> 10 msSlide13

Other Approaches to assessing Iron overload

Effects on specific organsOther Organs

Endocrine screening- assessment of functionGrowth monitoring, bone ageRole of MRI screening of pancreas 1, 2 ?Measurement of NTBI/

LPIPredictive value of response 3

Au WY, et al. Haematologica. 2008;93:785. Noetzli

LJ, et al. Blood. 2009;114:4021-6

.

3.

Aydinok

Y, et al.

.

Haematologica

, 2012, 97,6, 835-

41Slide14

MRI and assessment of endocrine complications in Thalassaemia Major

Au WY, et al. Haematologica

. 2008;93:785.

Cardiac

MRI T2*

correlates with endocrine dysfunction

Pancreatic T2* poor correlation with diabetes

Pituitary T2 correlates with multiple endocrine dysfunctions

Age

Ferritin

Cardiac T2*

Hepatic T2*

Pan T2*

Pit T2

Pit SIR

Pit T2*

Heart failure (n = 34)

NS

NS

< 0.001

NS

0.001

0.013

0.017

0.009

Diabetes

(

n = 44)

< 0.001

(0.001)

NS

< 0.001

(0.004)

NS

NS

0.015

0.001

0.055

Hypogonadism

(n = 84)

< 0.001 (0.001)

NS

< 0.001 (0.049)

NS

0.057

< 0.001

< 0.001 (0.05)

NS

Hypothyroid

(n=36)

0.061

<0.001

< 0.001

NS

NS

< 0.001

< 0.001 (0.023)

NS

Hypoparathyroid

(n = 16)

0.001 (0.008)

NS

< 0.001 (0.006)

NS

NS

0.062

0.003

0.058

= not analysed; EF = ejection fraction; NS = not significant; Pan = pancreatic; Pit = pituitary; SIR = signal intensity ratio of pituitary to muscle.

*p < 0.05; **p < 0.01; ***p < 0.001

. n=180Slide15

Assessment – when?

Iron intake rate Each transfusionChelation dose & frequency 3

monthlyGrowth & sexual development 6 monthly children

Liver function 3 monthly

Sequential ferritin 3 monthlyGTT, thyroid, Ca metab Yearly in adults

Liver

iron Yearly from age 8-10

Heart

function Yearly from age 8-10

Heart iron (T2*

) Yearly from age 8-10

Observation Frequency

ExpenseSlide16

Prevention of iron mediated damageBalance input and output - iron balanceAchieve harmless levels of body iron safelyRescue

patients with high levels of body ironpatients with high levels of cardiac ironpatients with heart dysfunction

Goals of chelation therapySlide17

Licensed iron chelators

Property

DFO

Deferiprone

Deferasirox

Route

Sc

, iv

(8–12 hours,

5 days/week)

Oral

3 times daily

Oral

Once daily

Half-life

20–30 minutes

3–4 hours

8–16 hours

Excretion

Urinary,

fecal

Urinary

Fecal

Main adverse effects

in PI

Local reactions, ophthalmologic, auditory, growth retardation, allergic

Gastrointestinal disturbances, agranulocytosis/ neutropenia, arthralgia, elevated liver enzymes

Gastrointestinal disturbances, rash, mild non-progressive

creatinine

increase, elevated liver enzymes, ophthalmologic, auditory

Usual dose (mg/kg/day)

25–60

75–100

20–30Slide18

Chelation regimes

DFO monotherapySc 8-12hcontinuous (sc or iv)Deferiprone monotherapypo 3 x dailyCombined Deferiprone and DFO

Deferiprone daily with DFO nocte n x weekDeferiprone daily + DFO at same timeDeferasirox monotherapyNew combinations and drugsSlide19

‘Harmless body iron levels’ ?what are guidelines based on ?

Experience with thalassaemia major

Experience with DFOControl of ferritin and LIC links to risk of cardiac diseaserisk of under and over chelationSlide20

Guidelines with DFO therapy

Begin after 10–20 blood transfusionsor when serum ferritin > 1,000 µg/LDose adults 40-60mg/kg 8-12h nocte

minimum 5x/wkMaintainserum ferritin < 2,500 µg/L (1,000 µg/L recommended)LIC < 7 mg/g dry weight

Intensify dose or frequency ifif severe iron overloadHigh ferritin values persistently > 2,500 µg/L

High liver iron > 15 mg/g dry weight or significant cardiac diseaseSignificant cardiac dysrhythmias Evidence of failing ventricular functionEvidence of severe cardiac iron loading Reduce dose ifFerritin <1000µg/L

Ratio of mean daily dose (mg/kg) / ferritin >0.025Slide21

Guidelines based on

Risks of over-chelation with DFO

Risks of starting too early

effects on growtheffects on bones, especially < 3 years of age1,2

Risks of too high a dosegrowth affected: > 70 mg/kg/day, normalized ≤ 40 mg/kg/day3skeletal/bones: > 70 mg/kg in children1eyes: visual symptoms > 80 mg/kg/day

4

otoxicity

4,5

Risks at low iron loads

effects on growth: patients had mean ferritin of 1,300 µg/L

3

otoxicity

: with serum ferritin < 2,000 µg/L or when ratio dose/ferritin

too high

5

neurotoxicity in non-iron-overloaded RA patients at low doses

6ocular toxicity in dialysis patient7

1.

Olivieri

NF, et al. Am J

Pediatr

Hematol

Oncol

. 1992;14:48-56.

2. Brill PW, et al.

Am.J.Roentgenol

. 1991;156:561-5. 3.

Piga

A, et al.

Eur

J

Haematol

.

1998;40:380-1.

4.

Olivieri

NF, et al. N

Engl

J Med

.

1986;314:869-73. 5. Porter JB, et al. Br J Haematol

. 1989;73:403-9.6. Blake DR, et al. Q J Med. 1985;56:345-55. 7. Rubinstein M, et al. Lancet. 1985;325:817-8.Slide22

Survival probability

Borgna-Pignatti C, et al. Haematologica. 2004;89:1187-93.

(

p < 0.00005)

0

1.00

0.75

0.50

0.25

0

5

10

15

20

25

30

Age (years)

Birth cohort

1960–1964

1965–1969

1970–1974

1975–1979

1980–1984

1985–1997

DFO Chelation

therapy

has improved

patient

survival in TMSlide23

Decline in complications with

iron chelation

Birth 1970–1974*

Birth 1980–1984

Death at 20 years

6.3%

1%

Hypogonadism

64.5%

14.3%

Diabetes

15.5%

0.8%

Hypothyroidism

16.7%

4.9%

Patients with

β

-thalassaemia major

born after 1960 (N = 977)

*DFO i.m., 1975;

DFO s.c., 1980.

In 1995, 121 patients switched to deferiprone (censored at this time)

Borgna-Pignatti C, et al.

Haematologica. 2004;89:1187-93.Slide24

Is

there a risk of over-chelation with other chelation regimes

?How low can we go?

How is risk of chelator toxicity related toAbsolute chelator dose

Dose in relation toBody iron load Transfusional iron loading rateRate of decrease of load with chelationSlide25

Do DFP doses >75mg/kg/d

affect tolerability?

Unwanted Effect Dose dependence?

GI distrurbances 3-24% at 75mg/kg (1-3)

66% at 100mg/kg

(n=29)

(4)

Neutropaenia insufficient human numbers

Agranulocytosis insufficient human numbers

Thrombocyopenia age <6y (7/44) ? dose effect

(5)

Arthropathy ? improved arthropathy at 50mg/kg

(6)

Neurotoxicity Yes with unintended large doses

1. Al Rafae Brit J Haematol, 1995;

91:224-9.2. Ceci A, et al.

Br J Haematol. 2002;118: 330-6.

3. Cohen AR, et al. Br J Haematol.

2000;108:305-12.

4. Pennell DJ, et al. Blood. 2006;107:3738-44

5. Naithani et al, Eur J Haematol. 2005 ;74:217-20

6. Lucal et a, Ceylon Med, 45, 71-4.J 2000

.Slide26

Low serum ferritin without toxicity with long-term combined therapy

53 patients 5-7y on DFO 20-60mg/kg/day and deferiprone 75mg/kg/day ‘individually tailored’Ferritin

bl 3421µg/L - 87 µg/L at 5-7y T2* bl 28ms

- 38 ms at 5-7yLIC

bl 12.7 - 0.8mg/g dry wt at 5-7yGTT normal bl 23% - 64% at 5-7y

Thyroxine

replacement

bl

34% - 20% at 5-7y

Secondary amen

bl

19/26

- 3/19 spontaneous ovulation No toxicity

Farmaki et al presentation at ITC 2008 FC07 Pg. 92

Farmaki et al Br J Haematol, 466-75 (2010)Slide27

Porter JB, et al. Blood. 2008;112:[abstract 5423].

Experience

with serum ferritin

< 1,000

μg/L

174 adult and paediatric patients (out of 474) were chelated to serum ferritin levels < 1,000

μ

g/L

% of patients achieving serum ferritin < 1,000 µg/L

Year 1

Year 2

Year 3

Year 4

Year 5

The incidence of drug-related AEs did not appear to increase during the periods after serum ferritin levels first decreased < 1,000 μg/L

YearsSlide28

Safety profile of serum ferritin <1000

μg/L

Investigator-assessed d

rug-related adverse events (n  5%)

, n (%)

Serum ferritin

< 1,000

μ

g/L

(n = 174)

≥ 1,000

μ

g/L

(n = 300)

Nausea

26 (14.9)

38 (12.7)

Diarrhoea

17 (9.8)

42 (14.0)

Vomiting

14 (8.0)

25 (8.3)

Abdominal pain

12 (6.9)

32 (10.7)

Rash

9 (5.2)

16 (5.3)

Upper abdominal pain

6 (3.4)

20 (6.7)

The incidence of drug-related adverse events did not appear to increase during the periods after serum ferritin levels first decreased < 1,000

μ

g/L

Safety profile

was similar to patients with serum ferritin levels > 1,000

μ

g/L

No increase in the proportion of patients with creatinine increases > 33% above baseline and ULN or with ALTs > 10 x ULN

Porter JB, et al. Blood. 2008;112:[abstract 5423].

ALT = alanine transaminase.Slide29

Duration of therapy: 52 weeks

Deferasirox 20–30 mg/kg/day

DFO 35–50 mg/kg/infusion infused 3–7 days/week

Combined chelation therapy with DFX and DFO in transfusion-dependent thalassaemia

Lal

, Porter,

et al. Blood. 2010;116:[abstract 4269].

Aim: to explore safety and efficacy of combined

deferasirox

and DFO in patients with transfusion-dependent

thalassaemia

who had failed standard chelation therapy with single drug (US24T)

15 patients enrolled and randomized into 3 equally sized groups

Group A

Adults

LIC <15 mg/g dry

wt

Group B

Adults

LIC >15 mg/g dry

wt

Group C

8–18 years

LIC >5 mg/g drywtSlide30

DFX + DFO:improvements in iron overload

Cardiac improvements (in three patients who had T2* < 20 ms at baseline)T2* < 20 ms at baseline (6.5–19.5 ms): improved +2.43 ms (8.8–21.3 ms) (p = 0.027)

LVEF < 60% at baseline (47.4–58.1%): improved to 60.6–64.4%Median LPI decreased: 0.87 µM to 0.05 µM (p = 0.004)

Median LIC (mg/g)

Median plasma NTBI (µM)

DFO

DFO + deferasirox

p < 0.001

Median serum ferritn (

μ

g/L)

BL 1 year

48%

(p = 0.003)

43%

(p = 0.008)

BL 1 year

LIC

Serum ferritin

NTBI

0

500

1,000

1,500

2,000

0

5

10

15

0

1

2

3

Lal A, et al. Blood. 2010;116:[abstract 4269].

LPI = labile plasma iron.Slide31

DFX + DFO:improvements in iron overload

Lal

, Porter et al Blood.Cells Mol Dis. 2012 in press.Slide32

DFP + DFX?A patient case34-year-old female with TM, 2 units of packed red blood cells, every 20 days

Deferoxamine – failed to comply T2* liver 1.1 ms, cardiac T2* 9.4 msserum ferritin > 2,800 µg/LDeferasirox, 20 mg/kg for 12 months

liver T2* 3.33 ms, cardiac T2* 10.6 ms Deferasirox 30 mg/kg for 24 months liver 7.

81 ms, cardiac T2* 13.8 ms serum ferritin 2,080 µg/LDeferasirox 30 mg/kg/day + deferiprone 75 mg/kg/day

for 12 months serum ferritin 397 μg/Lliver T2* 15.3 ms, cardiac T2* 21.1 ms

Voskaridou E, et al. Br J Haematol. 2011;154:654-6.

Combination

Cardiac

Liver

Year

MRI T2* (ms)

2005

2006

2007

2008

2009

2010

25

15

5

0

20

10

Year

Serum ferritin (µg/L)

2005

2006

2007

2008

2009

2010

3,000

1,500

500

0

2,000

1,000

2,500

Serum ferritinSlide33

DFP + DFX

Farmaki, et al. Blood Cells Mol Dis. 2011;47;33-40.

GTT = glucose tolerance test.

* p < 0.001

Patient selection

16 TM > 20 years old

Either intolerance to DFO or

‘inconvenience to DFO’

Serum ferritin > 500 µg/L

> 1 iron overload complication

(clinical or laboratory)

Treatment:

up to 2 years of

DFX (20–25 mg/kg/day)

+ DFP (75–100 mg/kg/day)

Outcome

Reversal of cardiac dysfunction in 2/4

Mean LVEF increased significantly

GTT improved in 2/8 with impaired GTT

Improvement in gonadal function

Tolerability

No serum

creatinine

> ULN

No

agranulocytosis

, neutropenia, thrombocytopenia

3/15 (20%) minor GI disturbance

Baseline

After

Serum ferritin (µg/L)

581±346

103±60

LIC (mg/g dry wt)

1.6±1.1

1.0±0.2

Cardiac T2* (ms)

34.1±5.8

36.9±5.6*

LVEF (%)

61±6.0

65±7.6*

2-hour GTT (mg/dL)

150±87

111±24

Creatinine (mg/dL)

0.9

1.0Slide34

Approaches to LIC measurementBiopsyPatient safety and acceptanceSample size (4mg wet wt)

Inhomogeneity of distributionMeasurements not standardisedSQUIDOnly 4 machinesVery expensive to run and maintainNot

standardisedMRIT2* calibration underaseimates LIC by two foldR2* (= 1/T2*) R2 (ferriscan) standardised

, easy to introduceSlide35

How has chelation therapy and monitoring impacted on outcome in transfusion dependent thalassaemia

- a local perspective in UKSlide36

Treatment of Thalassaemia

Major in the UK

1960 → 1970 → 1980 → 1990 → 2005

1980 – SC desferoxamine standard of care

1964 – IM desferoxamine

1984 – Bone marrow transplant initiated

1987 – Deferiprone

1999 – CMR

Deferasirox

Cardiac failure secondary to cardiac iron overload is reported as the leading cause of death amongst patients with

TM

Survival substantially improved with introduction of iron chelation therapy but despite this by 2000, 50% UK patients died before the age of 35 in 2000

1

.

CMR introduced in London 1999 – what impact has this had

Cohort of 121 patients monitored and treated at UCLH/

Whittinton

since 1999Slide37

DFO

DFP

DFP + DFO

DFO

DFP

DFP + DFO

DFX

2010

Chelation regimes

DFP + DFO

DFP

DFXSlide38

Impact of a decade of cardiac MRI assessment on cardiac T2*

Cohort of 132 patients from UCLH/Whittington hospitals

Thomas AS, et al. Blood. 2010;116:[abstract 1011].

Baseline

Median 9 years follow-up

Proportion of patients (%)

70

50

30

10

0

60

40

20

T2* ≤ 20 ms

T2* < 10 ms

60

23

17

7

p < 0.001

p < 0.001Slide39

MortalityTotal of 8 deaths amongst 132 patients:

2 female, 6 malemedian age at death 35.6 years (range 27.3-48.4)None directly related to myocardial ironMortality rate 1.65 / 1000 patient y (95% CI 0.71-3.24

)Previous reports from UK thalassaemia registry:1980-1999: 12.7 deaths / 1000 patient y 2

2000-2003: 4.3 deaths / 1000 patient y 3

1. Thomas

AS, et al. Blood. 2010;116:[abstract 1011

]

2 Modell

et al , Lancet 355:2051-2, 2000

3. Modell

et al , J.

Cardiovas

Magnetic Resonance, 2008Slide40

Causes of Death and cardiac MRI

Cardiac MRI at death, n = 8

T2* > 20ms 3 pt with hepatitis C complications 1 sudden deathT2* 10-20ms

1 pt with meningitis 1

pt with cancerT2* < 10ms 2 pt with sepsisSlide41

Chelator regime at death

DFO (n= 4)DFP (n= 2)Combination DFP + DFO ( n= 1)DFX ( n= 1)Slide42

0

10

20

30

40

50

60

70

80

90

100

Causes of death in

β

-

thalassaemia

major in the UK

Adapted from UK Thalassaemia Registry data from Modell B, et al. J Cardiovasc Magn Reson. 2008;10:42.

Thomas AS, et al. Blood. 2010;116:[abstract 1011].

Use of modern iron chelation therapy and regular CMR monitoring has dramatically reduced the iron overload-related mortality in the Red-cell Disorders Unit

Mortality rates per cohort

Patients (%)

Hepatitis C

complications

Other/unknown

Malignancy

Infection

BMT complication

Anaemia

Iron overload

1950–1959

1960–1969

1970–1979

1980–1989

1990–1999

2000–2003

This cohort

BMT = bone marrow transplantation;

CMR = cardiac magnetic resonance imagingSlide43

Optimal Outcome - What else

do we need ?Recognition that chronic diseases pose special challenges which require targeted resourcesRapid

access to free careStaff with expert knowledge & experience Continuity of care (especially staff)

Systems organized to allow best care with minimum disruption

to ordinary life Identity (a ‘unit’) for patients allowing a focus for care but not isolated from hospital Multidisciplinary team with integrated clinics & investigations

Optimal

monitoring and intensification for high risk

patients

ANDSlide44

ConclusionsWith modern chelation regimes, used alone or in combination and when applied with modern monitoring techniques, excellent survival can be obtainedThe challenge over the next decade will to be to improve quality of life in an ageining population by;

Further decreasing morbidities associated with thalassaemia and iron overloadFurther improving infrastructure and delivery of care to thalassaemia patients inside and outside treatment ‘centres’Slide45

Then

and

Now

What can be achieved with transfusion, chelation and optimal monitoringSlide46

ThankyouSlide47

A decade of cardiac monitoring with modern

chelation therapies for TM, UCLH/Whittington

Cohort of 132 patients received 1

st CMR 1999-2000109 of these available for long term CMR FU

Follow up median 9.2 years (range 7.0-10.6)Minimum CMR follow up of 7 yMedian age at 1st CMR 27.9 years (range 7.7-49.5)

58 female, 51 maleSlide48

Variables studied

% Patients with evidence of myocardial ironAt 1st CMRAt latest CMRSurvival in cohort with baseline CMR 1999-2000

Cause of deathT2* at deathModes of chelation

At baselineAt latest follow upAt death

Number of switches in chelatorSlide49

Causes of Death by cardiac MRI

Cardiac MRI at death, n = 8

T2* > 20ms 3 pt with hepatitis C complications 1 sudden deathT2* 10-20ms

1 pt with meningitis

1 pt with cancerT2* < 10ms 2 pt with sepsisSlide50

Changes in Chelation

69% changed chelator at least once based on:Iron assessmentFerritin trend

LIC trendm T2* trendSide effects/tolerabilityAdherence or patient preference

Availability of new chelators: trials/funding decisionsSlide51

Impact of monitoring and comprehensive support on outcomeSlide52

Survival (%)

0

60

80

50

40

30

20

10

70

90

100

0

28

26

24

22

20

18

16

14

12

10

8

6

4

2

30

32

34

36

38

40

Years

300

3

65

225

3

00

150

2

25

75

1

50

0

7

5

Frequency of

DFO chelation

and survival in

T

halassaemia

M

ajor

Gabutti V, Piga A. Acta Haematol. 1996;95:26-36.

Infusions/yearSlide53

Survival in UK as a whole and UCLH

- a question of optimal care?Modell et al1

200050% of thalassaemia major patients in

UK die < the age 35y Modell et al., Lancet 2000; 9220:2051-2

Davis et al, 2001, UCLH experience N=103, 78% survival at 40yrsNo death in cohorts after 1971

Porter & Davis Best

Pract

Res

Clin

Haematol

, 15, 328-68 2002Slide54

Crude mortality rates in

thalassaemia major

 

Birth cohort

 

UK as a whole

 

UCLH

1955-1964

 

1964-1974

 

1975-1984

 

1985-1994

 

1995-2000

56%

 

34%

 

14%

 

3%

 

2%

29%

 

15%

 

0%

 

0%

 

0%

Overall

24%

Porter & Davis Best Pract Res Clin Haematol, 15, 328-68 2002

11.7%

Slide55

Reasons for better outcome?

Experience of clinicians - large clinic ?Different patient group ?Different Chelation regimen ?Better monitoring and intervention ?

Better Patient support of compliance adherence ?Slide56

DELIVERY OF THALASSAEMIA CARE IN UK as a

WHOLE807 patients cared for by 164 physicians nationwide71 physicians 1 patient77 physicians 2-9 patients

12 physicians 10-30 patients4 physicians 50 or more

Modell et al., Lancet 2000; 9220:2051-2Slide57

UK National Guidelines for Thalassaemia

Transfusion and other care locally butAt least yearly review in specialist centre

UK Thalassaaemia SocietySlide58

Thalassaemia

Major management in the UK

1960 → 1970 → 1980 → 1990 →

2005

1980 – SC desferoxamine standard of care

1964 – IM desferoxamine

1984 – Bone marrow transplant initiated

1987 – Deferiprone

1999

– CMR

Deferasirox

Cardiac failure secondary to cardiac iron overload is reported as the leading cause of death amongst patients with TM

Survival substantially improved with introduction of iron chelation therapy but despite this by 2000, 50% UK patients died before the age of 35 in 2000

1

.

1 Modell et al, Lancet 2000 355:2051-2Slide59

A decade of cardiac monitoring with modern

chelation therapies for TM, UCLH/Whittington

Cohort of 132

patients received 1st CMR 1999-2000

109 of these available for long term CMR FUFollow up median 9.2 years (range 7.0-10.6)Minimum

CMR

follow up

of 7 y

Median age at 1

st

CMR 27.9 years (range 7.7-49.5)

58 female, 51 maleSlide60

Impact of a decade of cardiac MRI assessment on cardiac T2*

Cohort of 132 patients from UCLH/Whittington hospitals

Thomas AS, et al. Blood. 2010;116:[abstract 1011].

Baseline

Median 9 years follow-up

Proportion of patients (%)

70

50

30

10

0

60

40

20

T2* ≤ 20 ms

T2* < 10 ms

60

23

17

7

p < 0.001

p < 0.001Slide61

MortalityTotal of 8 deaths amongst 132 patients:

2 female, 6 malemedian age at death 35.6 years (range 27.3-48.4)Mortality rate 1.65 / 1000 patient y (95% CI 0.71-3.24)

Previous reports from UK thalassaemia registry:

1980-1999: 12.7 deaths / 1000 patient y 12000-2003: 4.3 deaths / 1000 patient y 2

1 Modell et al , Lancet 355:2051-2, 2000

2

Modell et al , J.

Cardiovas

Magnetic Resonance, 2008Slide62

Causes of Death and cardiac MRI

Cardiac MRI at death, n = 8

T2* > 20ms 3 pt with hepatitis C complications 1 sudden deathT2* 10-20ms

1 pt with meningitis 1

pt with cancerT2* < 10ms 2 pt with sepsisSlide63

Chelator regime at death

DFO (n= 4)DFP (n= 2)Combination DFP + DFO ( n= 1)DFX ( n= 1)Slide64

Changing Causes of death in TMSlide65

Optimal Care for chronic anaemias - What do we need ?

Optimal monitoring technigues - yes but also need…….Setting to optimise treatment adherence

Recognition that chronic diseases pose special challenges which require targeted resourcesRapid access to free careStaff with expert knowledge & experience

Continuity of care (especially staff)Systems organized to allow best care with minimum disruption to ordinary life

Identity (a ‘unit’) for patients allowing a focus for care but not isolated from hospital Multidisciplinary team with integrated clinics & investigationsSlide66

SummaryIron toxicity occurs in tissues where

excess storage iron accumulates though NTBI uptakeDistribution of excess iron differs in transfusional and non transfusional iron overloadDistribution also differs depending on underling disorder (e.g. Thal vs sickle)

Heart and endocrine tissues more sensitive to excess iron than liverNo single measure assesses risk of iron overload equally in all clinical conditionsAssessment of iron overload needs to estimate both;

The degree of body iron overloadThe distribution of iron excess (extra-hepatic vs hepatic)Transferrin saturation- good screening tool – less useful for monitoring

Ferritin is a useful marker of iron overload with prognostic significanceLIC assessment can estimate total body iron storesMyocardial T2* by MRI, validated with prognostic significance MRI other tissuesPlasma iron speciation and quantitationSlide67

1960

1970

1980

1990

200020102020

1980 – SC desferoxamine standard of care

1964 – IM desferoxamine

1984 – Bone marrow transplant initiated

1987 – Deferiprone trials

1999 - CMR

1980-1999: 12.7 deaths per 1000 patient years

2000-2003: 4.3 deaths per 1000 patient years

1999-2010: 1.65 deaths per 1000 patient years

??Exjade Trails

The future for thalassaemia care……..Slide68

Special challenges in treating haemoglobin disorders?

Challengers for the carersLife-long conditions do not fit comfortably with a hospital environmentProviding sustained support throughout life is rarely possible for one doctor

Challenges of knowledge and/or experience of rare conditionsChallenges of minimising the problems of transitioning from childhood to adolescence department or hospital

Challenges of operating within an ocology domiated environmentSlide69

Summary - MonitoringIron toxicity occurs in tissues where

excess storage iron accumulates though NTBI uptakeDistribution of excess iron differs in transfusional and non transfusional iron overloadDistribution also differs depending on underling disorder (e.g. Thal vs sickle)

Heart and endocrine tissues more sensitive to excess iron than liverNo single measure assesses risk of iron overload equally in all clinical conditionsAssessment of iron overload needs to estimate both;

The degree of body iron overloadThe distribution of iron excess (extra-hepatic vs hepatic)

Transferrin saturation- good screening tool – less useful for monitoringFerritin is a useful marker of iron overload with prognostic significanceLIC assessment can estimate total body iron storesMyocardial T2* by MRI, validated with prognostic significance MRI other tissuesPlasma iron speciation and quantitationSlide70

Long term use of deferoxamine is associated with falling cardiac mortality

Age (years)

0

10

20

30

40

0

0.25

0.50

0.75

1.00

1955-64 (n=21)

1965-74 (n=39)

1975-97 (n=42)

Davis and Porter.

Adv Exp Med Biol

. 2002;509:91.

Survival probability

Patients With

Beta

-Thalassemia Major (N=103)

Survival by Birth Cohort at

University College London Hospitals

Analysis September 2000Slide71

Falling Cardiac and other complications with Deferoxamine

Borgna-Pignatti, 2004

Thal Major born after 1960, n=977

Birth 1970-74* Birth 1980-84^

Death at 20 5% 1%

Hypogonadism 64.5% 14.3%

Diabetes 15.5% 0.8%

Hypothyroidism 17.7% 0. 0%

* IM DFO 1975 ^ sc DFO 1980

1995 121 switched to L1 (censored at this time)Slide72

Rescue With Continuous IV DFO

for Cardiac DiseaseRationaleContinuous chelation because labile chelatable iron pools (NTBI) re-emerge rapidlyHigh doses may not be necessary

Outcome at 16y follow-upLVEF improvement within 6-8monthsReversal of arrhythmias in 6/6 (24h-6 mo)Good long term survival if chelation subsequently maintained

20

30

40

50

60

70

LVEF (%)

Pre–IV DFO

Post–IV DFO

6-8 mo

Mean 49%

Mean 36%

P

=0.002

LVEF = left ventricular ejection fraction; DFO = deferoxamine.

Davis and Porter.

Blood.

2000;95:1229.Slide73

New developments in chelation

Deferasirox monotherapyNew combinationsCombination studies with desferrioxamine (3)Tolerability at low ferritinFerrokinDesferrithiocin derivative

Phase 2 study results submitted for publicationSlide74

Deferasirox:recent publicationsLong-term cardiac effects

Liver effectsLong-term efficacy and tolerabilitySafety and efficacy of dose escalationTolerability at low levels of iron loadEffect of administration regime on efficacy and tolerabilityUse in combination with other chelators

Use in conditions other than transfusion-dependent thalassaemiaSlide75

5-year follow-up in patients with β-thalassaemia major: changes in serum ferritin

(n)

(422)

(433)

(375)

(343)

(286)

(253)

(244)

(220)

(154)

Cappellini MD, et al. Blood. 2008;112:[abstract 5411].

Studies 105–108: 4.5-year data

3,000

2,500

2,000

1,500

1,000

500

0

Time (months)

Median serum ferritin (

μ

g/L)

Mean deferasirox dose (mg/kg/day)

30

25

20

15

10

5

0

Deferasirox dose

Serum ferritin

BL 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 54

Mean actual daily dose of DFX:

22.1

± 6.4 mg/kg/day (range (6–37)

N = 472 at

baseline (BL)

Core

(DFO)

Extension (deferasirox)Slide76

Porter JB, et al. Blood. 2008;112:[abstract 5423].

Experience

with serum ferritin

< 1,000

μg/L

174 adult and paediatric patients (out of 474) were chelated to serum ferritin levels < 1,000

μ

g/L

% of patients achieving serum ferritin < 1,000 µg/L

Year 1

Year 2

Year 3

Year 4

Year 5

The incidence of drug-related AEs did not appear to increase during the periods after serum ferritin levels first decreased < 1,000 μg/L

YearsSlide77

Safety profile over time in patients with β-thalassaemia major

Cappellini MD, et al. Blood. 2011;118:884-93.

Patients (%)

Adverse event

10

8

6

4

2

0

9

7

5

3

1

Increased blood

creatinine

Abdominal

pain*

Nausea

Vomiting

Rash

Diarrhoea

Year 1 (n = 296)

Year 2 (n = 282)

Year 3 (n = 234)

Year 4 (n = 213)

Year 5 (n = 196)

* Reports of abdominal pain and abdominal pain are combined

and presented as abdominal pain.Slide78

Stable creatinine clearance in children and adults with β-thalassaemia major over 5 years

Adapted from: Cappellini MD, et al. Blood. 2011;118:884-93.

37 (6.7%) patients with a normal serum creatinine at baseline

had 2 consecutive values > 33% and > ULN

Creatinine clearance (mL/min)

400

200

0

300

100

BL

3

6

9

12

15

18

21

24

27

30

33

36

39

42

45

48

51

54

57

60

Deferasirox

Crossover

Time (months)

Core

Extension

ULN = upper limit of normal.Slide79

Patients, n

< 10 ms

24

24

24

24

10–< 20 ms

47

47

47

44

All patients

71

71

71

68

Cardiac iron reduction with deferasirox: continued improvement in cardiac T2*

Pennell D, et al.

Haematologica. 2012 Jan 22. [Epub ahead of print].

CI = confidence interval; LOCF = last observation carried forward.

p = 0.0012 versus baseline;

p < 0.001 versus baseline

Dashed line indicates normal cardiac T2* of ≥

20 ms

10.5

7.7

8.6

9.4

15.0

17.7

20.3

22.3

Baseline

12

24

36

Time (months)

Geometric mean T2*

± 95% CI (ms)

> 5

–< 10 ms

10

–< 20 ms

All patients

0

5

10

20

30

15

25

17.1

15.6

13.9

12.0Slide80

CORDELIA: RCT deferasirox vs DFOObjective: to prospectively compare the efficacy of deferasirox to DFO in patients with a MRI-measured LVEF of

 56% but with evidence of cardiac iron deposition depicted by a myocardial T2* of  20 ms

Screening

23 days

1-year study Rx in core study

96 patients* deferasirox

96 patients* DFO

1-year study Rx in extension study

96 patients deferasirox

96 patients DFO

Randomize eligible patients

(1:1 ratio)

Followed by 5-day washout

Screening

23 days

End core / start extension

End

extension

* = Patients with β-thalassaemia major or Diamond-Black anaemia or sideroblastic anaemia on chronic transfusion therapy.Slide81

82.6% of patients experienced either stabilization or improvement in fibrosis stagingImprovements in fibrosis staging were observed in patients who met the LIC response criteria and in those who did not

Figure 1

Deugnier Y, et al. Gastroenterology. 2011;141:1102-11.

Improvement in liver pathology with at least 3 years of deferasirox treatmentSlide82

Combination Tharapies

Desferrioxamine + Deferiprone

Desferrioxamine + Deferasirox

Deferiprone + DeferasiroxSlide83

Deferasirox + DFO metabolic iron balance studies

Grady et al, 2010 116: Abstract 5163

Introduction: Deferasirox (Exjade, ICL670) is an orally effective iron chelating agent approved for use in patients 2 years of age and older with transfusional iron overload. While it is effective as a single agent, there are patients who do not attain net negative

iron balance despite being at the upper limit of approved dosing (40 mg/kg/day). Deferiprone (Ferriprox, L1) is another orally effective iron chelator. Through a series of metabolic iron balance studies we were able to demonstrate that various regimens

involving the combined use of deferiprone and deferoxamine (Desferal, DFO) were capable of placing all patients into net negative iron balance with the potential to adjust dosing schedules and the ratio of drugs to maximize effectiveness while minimizing toxicity. A variety of such regimens are now in widespread clinical use. We have taken the same approach in order to optimize the use of deferasirox.Methods: Six patients with thalassemia major were enrolled in a 34-day

metabolic iron balance study wherein

deferasirox

and deferoxamine

were evaluated alone and in combination, each patient serving

as his/her own control. Deferoxamine (40 mg/kg) was administered

on days 5 – 10 as an 8-hour subcutaneous infusion during

the night. On days 15 – 20, deferasirox (30 mg/kg) was given orally, 30 minutes prior to breakfast. Both drugs were given on days 25 – 30, the same doses and dosing schedules being employed. Non drug days allowed for washout of stool iron induced by the previous treatment. The patients consumed a fixed low-iron diet consisting of four individualized meal plans. Daily collections of urine and stool were made and their iron content determined by atomic absorption, a correction being made for all uneaten food.Results: As in previous studies, there was significant patient to patient variability in terms of the amount of drug-induced iron excretion. Combination therapy placed all patients into iron balance exceeding

200% (206% - 270%, mean 251%). The combination was synergistic

in two patients (35% and 57%), additive in three patients, and

less than additive in one patient. Where iron excretion was

more than additive, all of the excess appeared in the urine

(a 2.1- and 3.4-fold increase). Deferasirox proved to be less effective than deferoxamine in all six patients (relative iron

excretion: 23% - 59%, mean 42%), stool iron excretion in response

to the respective drugs being 94% -100% (mean 98%) and 49% - 74% (mean 59%). Net negative iron balance was achieved in 2/6 patients when on deferasirox (iron balance 28% - 129%, mean

72%) and 6/6 patients when infusing deferoxamine (iron balance 125% - 219%, mean 167%).Discussion: These results suggest that chelation therapy can be tailored

to the individual needs of each patient, selectively removing iron from different pools while minimizing any side effects. Deferasirox appears to shuttle iron to deferoxamine for excretion

in the urine, not unlike the situation when deferiprone and deferoxamine are combined, although the source of the iron may be somewhat different. It is hoped that these results will provide patients with further options to optimize their chelation regimens

.

Patients: - 6 with TM

- 34-day metabolic iron balance study - each patient serving as his/her own control

- fixed low-iron diet consisting of four individualized meal plansDosing: - Deferoxamine

(40 mg/kg) days 5 – 10 as an 8-hour sc nocte - Deferasirox (30 mg/kg) days 15 – 20, 30 minutes prior to breakfast. - Washout - then both drugs

were given on days 25 – 30Results: - Combination – Mean -

ve iron balance -251%,( range 206% - 270%) - Combination > additive 2 patients (35% and 57%) - additive in three patients

< additive in one patient Slide84

Duration of therapy: 52 weeks

Deferasirox 20-30 mg/kg/dayDFO 35-50 mg/kg/infusion infused 3-7 days/week

Combined Chelation Therapy with Deferasirox

and Deferoxamine in Transfusion-Dependent Thalassemia

Lal A, et al.

Blood.

2010;116: Abstract 4269.

Aim: To explore safety and efficacy of combined

deferasirox

and DFO in patients with transfusion-dependent thalassemia who had failed standard chelation therapy with single drug (US24T)

15 patients enrolled and randomized into 3 equally sized groups

Group A

Adults

LIC <15 mg/g dw

Group B

Adults

LIC >15 mg/g dw

Group C

8–18 years

LIC >5 mg/g dwSlide85

Improvements in Iron Overload

Median LIC (mg/g)

Cardiac improvements (in three patients who had T2* <20

ms

at baseline)

T2* <20

ms

at baseline (6.5 to 19.5

ms

): improved +2.43

ms

(8.8 to 21.3

ms

) [

p

=.027]

LVEF <60% at baseline (47.4 to 58.1%): improved to 60.6 to 64.4%Median labile plasma iron (LPI) decreased: 0.87 µM to 0.05 µM (P = .004)

Median plasma NTBI (µM)

DFO

DFO + deferasirox

P

<.001

Median SF (ng/mL)

BL 1 year

48%

(

P

= .003)

43%

(

P

= .008)

BL 1 year

LIC

Serum ferritin

Non-transferrin-

bound iron (NTBI)

0

500

1000

1500

2000

0

5

10

15

0

1

2

3

Lal A, et al.

Blood.

2010;116: Abstract 4269.Slide86

Deferiprone + Deferasirox ?34yo female TM, 2 units of packed red blood cell, every 20 dDeferoxamine - failed to comply

- T2* liver 1.1ms, cardiac T2* 9.4ms - serum ferritin > 2800 lg/lDeferasirox, 20 mg/kg for 12 mo liver T2* 3.33 ms,

Cardic T2* 10.6 ms Deferasirox 30 mg/kg, 24mo Liver 7.81ms, cardiac T2* 13.

8 ms, SF 2080 lg/l Deferasirox 30 mg/kg/d

+ deferiprone 75 mg/kg/d for 12 mo - Serum ferritin (397 lg/l), - Liver T2* 15.3, Cardiac 21.1 ms

Voskaridou,, et al. (2011).

Brit. J Haematol

154

(5): 654-656.

combination

Cardiac

LiverSlide87

Berdoukas et al , Blood. Blood 2010 116

Abstract 2064

Deferiprone + Deferasirox

Patient selection

4 case reports - adult patients with TM Reduced LVEF + either severe allergy or intolerance to DFOHigh liver iron concentrations (LIC)

Previous treatments DFX or DFO

Treatment

DFX (15–40 mg/kg/day ) + DFP (75–100 mg/kg/day), 6 - 60

mo

(mean 18 )

Outcome

Cardiac T2* improved

from 5.8 ±1.5 to 7.0 ± 1.5

ms

(mean) (p=0.15).

LVEF 52.8% to 58.9% (p=0.02). Ferritin fell from a mean of 5826 to 5544 ng/L (p=0.86).LIC increased from 20.7 to 28.1 mg/g dry weight (p=0.36)Tolerability No drug-related neutropenia, agranulocytosis or arthralgia No significant proteinuria and mean

creatinine

levels were unchanged.

ALT's showed fluctuations

Compliance

Highly variable

Conclusions

Well tolerated, prospective studies neededSlide88

Deferiprone + Deferasirox

Patient selection

16 TM >20yo either intolerance to DFO or ‘Inconvenience to DFO’

Ferritin >500µg/L >1 IOL complication (clinical or laboratory)Treatment; up to 2years of DFX (20–25 mg/kg/day ) + DFP (75–100 mg/kg/day )OutcomeReversal of cardiac dysfunction in 2/4

Mean LVEF increased significantly.

GTT improved in 2/8 with impaired GTT

Improvement in gonadal function (1)

Tolerability

No serum

creatinine

>ULN

No

agranulocytosis,neutropenia

thrombocytopenia

3/15(20%) minor GI disturbance______________________________ Baseline After______________________________Ferritin(µg/L)

581

±346

103

± 60

LIC

(mg/g

dw

)

1.6

±1.1 1.0 ± 0.2cT2* ms 34.1 ± 5.8

36.9 ±5.6 *LVEF (%) 61

± 6.0 65 ± 7.6 *2h GTTmg/dl

150 ± 87 111 ± 24Creatinine(mg/dl) 0.9 1.0

_______________________________*p<0.001

Farmaki et al, Blood Cells, Mol, and Dis 47 (2011) 33–40

cT2* = cardiac T2, GTT glucose tolerance test,

ULN upper limit normal, IO: iron overlloadSlide89

Desferrithiocin and derivatives

Desferrithiocin

Bergeron RJ, et al. Biometals. 2011;24:239-58.

Tridentate chelator

Renal toxicity is a class

effect but minimized by

derivatization in animal

studies

Numerous analogues

synthesized

Def

erritrin, nephrotoxic in clinical studies

Deferitrin (1)

CH

3

CO

2

H

N

S

OH

HO

(

S

)-3′-(HO)-DADFT-PE (9)

OH

CH

3

CO

2

H

OCH

3

N

S

O

O

O

By replacing the 4

-(HO) of 1 with a 3,6,9-trioxadecyloxy group nephrotoxicity could be controlledSlide90

Phase 1b dose-escalation study: safety, tolerability, and pharmacokinetics16 adult patients with transfusional overloaded Once daily for 7 days at doses up to 32 mg/kg

Well tolerated at all dose levelsPharmacokinetics showed dose-proportionalityCmax at 60–90 min

Rapidly distributed at the predicted therapeutic dosesPlasma t1/2 – approximately 19 hours

Clinical studies witha desferrithiocin

derivative FBS0701Rienhoff HY Jr, et al. Haematologica. 2011;96:521-5.Slide91

51 patients, stratified by transfusional iron intake FBS0701 at 14.5 or 29 mg/kg/day p.o. once daily49 patients (96%) completed the studyNo AEs showed dose-dependency Commonest AE was increased transaminases (16%, n = 8)

Mean serum creatinine did not change significantlyΔLIC mean at 14.5 mg/kg/day was +3.1 mg/g (dry wt) 29% achieved a decrease in LICΔLIC mean at 29 mg/kg/day was −0.3 mg/g (dry wt) 44% achieved a decrease in LIC

24-week multicentre phase 2 studywith FBS0701

Neufeld EJ, et al. Blood. 2012 Jan 17;[Epub ahead of print].Slide92

Conclusions

Cardiac iron overload is no longer the leading cause of mortality in Thal major patients if treated with full range of chelator options and monitored (including MRI) and supported

appropraitelyAll chelator regimes remove cardiac iron; choice of regime depends on severity of loading and heart functionLiver disease is becoming a serious issue in undertreated patients especially SCD

New combinations of chelators

are at early stage of assessment but may provide useful treatment options in future for difficult patientsFBS entering Phase III , when efficacy/toxicty will be scrutinisedSlide93

Treatment of Iron overloadWhich conditions ?Transfusion dependent Thalassaemia MajorMulti transfused SCDDBASideroblastic

Aplastic AnaemiaIron loaded NTDTMDSSlide94

Overview of Iron Chelators

Property

Deferoxamine (DFO)

Deferiprone (DFP)

Deferasirox

Usual dose

25-60 mg/kg/day

75 mg/kg/day

20-40 mg/kg/day

Route

s.c., i.v.

8-12 h, 5 days/week

p.o.

3 times daily

p.o.

once daily

Half-life

20-30 min

3-4 h

8-16 h

Excretion

Urinary, fecal

Urinary

Fecal

Adverse effects

Local reactions, ophthalmological, auditory, growth retardation, allergic

GI disturbances,

agranulocytosis

/ neutropenia,

arthralgia, elevated liver enzymes

GI disturbances, rash,

mild non-progressive creatinine increase, ophthalmological, auditory, elevated liver enzymes

Approved indications

Treatment of chronic iron overload due to transfusion-dependent anaemias

Thalassemia major

Treatment of chronic iron overload due to frequent blood transfusions

Deferoxamine Prescribing Information.

Deferasirox Summary of Product Characteristics.

Deferiprone Summary of Product Characteristics.

GI = gastrointestinal; i.v. = intravenous; p.o. = per orum; s.c. = subcutaneous.Slide95

Univariate analysis of biochemical, virological, and histological features associated with severe fibrosis

Di Marco V, et al. Haematologica. 2008;93:1243-6.

The majority of HCV-RNA negative patients with low iron load did not develop liver fibrosis, while hepatitis virus C-RNA positive patients infected with genotype 1 or 4 and iron overload more frequently developed advanced fibrosis

Fibrosis stage

0–1–2

(104 patients)

Fibrosis stage

3–4

(22 patients)

p value

Age, mean ± SD

16.8±8.7

19.7±9.2

0.2

Gender, M/F

50/54

17/5

0.01

ALT, mean ± range

69.1±80.1

112.5±61.2

< 0.001

Serum ferritin, median (range)

1,583 (141–5,952)

2,115 (188–5,503)

0.3

HCV-RNA positive

39 (37.5%)

19 (86.4%)

< 0.001

LIC, median (range)

2.3 (0.3–22)

2.9 (0.4–11.8)

0.3

Histological inflammation (grading)

Grade 0

23 (22%)

0

Grade 1/Grade 2

79 (76%)

22 (100%)

Grade 3

2 (2%)

0

0.2Slide96

Rate of fibrosis progression in transfusion-dependent β-thalassaemia patients

Patients (n)

Rate of fibrosis progression

(per year)

Expected duration for progression to cirrhosis (years)

All patients

117

0.087

(0.077–0.107)

57

(47–65)

HCV RNA+

80

0.101

(0.083–0.120)

49

(42–60)

HCV RNA−

37

0.075

(0.058–0.111)

67

(45–85)

Prati D, et al. Haematologica.

2004;89:1179-86

.Slide97

LIC Increases With Time in the Absence of Effective Chelation

Adapted from Olivieri NF, et al. N Engl J Med. 1999;341:99-109.

Homozygoushemochromatosis

Estimate for

NTDT

Age (years)

Non-chelated thalassemia major

0

0

10

20

30

40

50

Hepatic Iron (mg/g of liver, dry weight

)

Increased risk of iron-related morbidity

Normal hepatic iron concentration

Homozygous hemochromatosis

10

20

30

40

50Slide98

labile

iron

Cell death

Fibrosis

Organelle

damage

TGF-

β

1

ROS

Lipid peroxidation

Lysosomal

fragility

Enzyme leakage

Collagen synthesis

NF-

к

B

activation

DNA damage

Genomic Instability

Caspase

activation

Anti-

-apoptotic

Blood

Transfusion

+

Iron

Chelation

-

Neoplasia

High Iron

absorption

Infection

storage

iron

Pathophysiology of Iron OverloadSlide99

Organs susceptible to iron overload

Diabetes

Pancreas

Hypogonadotrophic Hypogonadism

Gonads

Cirrhosis, carcinoma

Liver

Cardiomyopathy

Heart

Hypoparathyoidism

Parathyroid

Hypothyroidism

Thyroid

Hypogonadotrophic Hypogonadism

Pituitary

Consequences

Organ