samarasinghe et al-2012-british journal of haematology

15
 How I manage aplastic anaemia in children Sujith Samarasinghe 1 and David K. H. Webb 2 1 Paediatric Haematopoietic Stem Cell Transplant Unit, Department of Adolescent and Paediatric Haematology and Oncology, Great  North Children’s Hospital, Royal Victoria Inrmary, Newcastle Upon Tyne, UK and  2 Department of Haematology, Great Ormond Street Hospital, London, UK Summary Aplast ic anaemia (AA) is a rare heterog eneous condition in chil dre n. 15   20% of cases ar e const itu tio nal and cor rect diagnosis of thes e inherited causes of AA is impo rta nt for appropriate management. For idiopathic severe aplastic anae- mia, a mat ched sibling donor (MS D) hae mat opoietic ste m cell transplant (HSCT) is the treatment of choice. If a MSD is not available, the options include immunosuppressive ther- apy (IST) or unr ela ted donor HSC T. IST with hor se ant i- thymocyte globulin (ATG) is superior to rabbit ATG and has good long-term results. In contrast, IST with rabbit ATG has an overall response of only 30   40%. Due to improvements in outcome over the last two decades in matched unrelated donor (MUD) HSCT, results are now similar to that of MSD HSCT. The decision to proceed with IST with ATG or MUD HSCT will depend on the likelihood of nding a MUD and the differing risks and benets that each therapy provides. Keywords:  pae diatric aplastic anaemia, inherited bone mar- row failure syndrome, trans planta tion in aplas tic anaemia, anti-thymocyte globulin. Denition Aplastic anaemia (AA) is dened as pancytopenia with a hyp- ocellular bone marrow in the absence of an abnormal inl- trate or marrow brosis. To diagnose AA, there must be at least two of the following: (i) haemoglobin  <100 g/l (ii) plate- let count  <50  9 10 9 /l (iii) neutroph il count <15  9 10 9 /l. The modied Camitta criteria are used to assess severity (Camitta  et al  , 1975). The severity of AA is determined on the full bl ood cou nt (FBC) and bone marr ow ndi ngs (Marsh et al  , 2009; Table I). Incidence The incidence is 2   3 million per year (all age groups) in Eur- ope, but higher in East Asia (Montane  et al  , 2008). There is a biphasic distribution, with peaks at 10   25 years and over 60 years. Aetiology This review wil l focus on constit uti onal and acquired idio- pathic AA. The majority (70   80%) of cases are classied as idiopat hic because their aetiolo gy is unknown (Mars h et al  , 2009). The remainder mainly consist of inherited bone mar- row fai lur e syn dromes (IB MFS ; 15   20%), the commones t bei ng Fanconi ana emi a (FA; see Table II ). Fiv e per cent of idi opat hic AA ha ve undi agnosed IBMFS where the full dise ase phe noty pe has not beco me apparent (Fogarty  et al  , 2003; Calado & You ng, 20 08). The cor rect diagnosis of  IBMFS is vital for approp riate managemen t, educat ion and genetic counselling. A classication of AA based on aetiology can be seen in Table II. For further information on patho- physiology please refer to a recent review (Young et al , 2010). Clinical evaluation The dist inc tion between inherited and acquir ed AA can be difcult because of the clinical and genetic heterogeneity of IBMFS (Table I II ). A fami ly hi st ory should be ta ken for blood diso rde rs, consangui nit y, mal ignanc y or cong eni tal anomalies as their presence may suggest an IBMFS. Examination The physical examination in children with AA may reveal fea- tures of an IBMFS, with features unique to each of the syn- dromes. The commonest phy sical nd ings in FA are short stature, skin hyper/hypo pigmented areas and skeletal abnor- malities, par ticu lar ly affecti ng the thumb (Sh ima mur a & Alter, 2010). A proportion of FA patients may be of normal stature with no appar ent anoma lies. Dyskeratos is congeni ta (DKC) has the classic diagnostic triad of nail dystrophy, retic- ular skin pigmentation and oral leucoleucoplakia (Shimamura Correspondence: Dr Sujith Samarasinghe, Department of Adolescent and Paediatric Haematology and Oncology and Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children’s Hospital, Royal Victoria Inrmary, Newcastle Upon Tyne NE1 4LP, UK. E-mail: [email protected] First published online 20 February 2012 doi: 10.1111/j.1 365-2141.2 012.09058.x ª 2012 Blackwell Publishing Ltd British Journal of Haematology , 2012,  157, 26–40 state of the art review

Upload: fadmawati-andri

Post on 01-Nov-2015

225 views

Category:

Documents


0 download

DESCRIPTION

n

TRANSCRIPT

  • How I manage aplastic anaemia in children

    Sujith Samarasinghe1 and David K. H. Webb2

    1Paediatric Haematopoietic Stem Cell Transplant Unit, Department of Adolescent and Paediatric Haematology and Oncology, Great

    North Childrens Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, UK and 2Department of Haematology, Great Ormond

    Street Hospital, London, UK

    Summary

    Aplastic anaemia (AA) is a rare heterogeneous condition in

    children. 1520% of cases are constitutional and correct

    diagnosis of these inherited causes of AA is important for

    appropriate management. For idiopathic severe aplastic anae-

    mia, a matched sibling donor (MSD) haematopoietic stem

    cell transplant (HSCT) is the treatment of choice. If a MSD

    is not available, the options include immunosuppressive ther-

    apy (IST) or unrelated donor HSCT. IST with horse anti-

    thymocyte globulin (ATG) is superior to rabbit ATG and has

    good long-term results. In contrast, IST with rabbit ATG has

    an overall response of only 3040%. Due to improvements

    in outcome over the last two decades in matched unrelated

    donor (MUD) HSCT, results are now similar to that of MSD

    HSCT. The decision to proceed with IST with ATG or MUD

    HSCT will depend on the likelihood of finding a MUD and

    the differing risks and benefits that each therapy provides.

    Keywords: paediatric aplastic anaemia, inherited bone mar-

    row failure syndrome, transplantation in aplastic anaemia,

    anti-thymocyte globulin.

    Definition

    Aplastic anaemia (AA) is defined as pancytopenia with a hyp-

    ocellular bone marrow in the absence of an abnormal infil-

    trate or marrow fibrosis. To diagnose AA, there must be at

    least two of the following: (i) haemoglobin

  • & Alter, 2010). This triad however is typically absent in early

    life and may remain absent even in a subset of adults.

    Investigations

    To confirm the diagnosis, assess severity and confirm/exclude

    constitutional AA, the following investigations are recom-

    mended (see Table III). The FBC typically shows pancytope-

    nia, although isolated thrombocytopenia may occur early on.

    A macrocytic anaemia with reticulocytopenia is normally

    present. A bone marrow aspirate shows hypocellular particles,

    with increased fat cells, macrophages, mast cells, and plasma

    cells. Erythrocytes, megakaryocytes and granulocytes are

    reduced or absent. Dyserythopoeisis is very common (Marsh

    et al, 2009) but dysplastic megakaryocytes and granulocytes

    are not seen in AA. A bone marrow trephine is required to

    exclude abnormal fibrosis or an abnormal infiltrate. Baseline

    immunological investigations are required as some IBMFS are

    associated with immunodeficiency (Dokal, 2000; Dror et al,

    2001; Knudson et al, 2005; Filipovich et al, 2010).

    Screening for IBMFS

    A diagnosis of FA can be confirmed by demonstration of

    increased chromosomal breakage following exposure of periph-

    eral blood lymphocytes to clastogens, such as mitomycin C or

    diepoxybutane (Table IV). All children with AA should have

    a clastogen stress test. Somatic reversion of the FA gene

    mutation can result in a false negative test (found in at least

    10%; Lo Ten Foe et al, 1997). Where there is a high clinical

    suspicion, but a negative stress test, the diagnosis of FA can

    be confirmed by testing skin fibroblasts for increased chro-

    mosomal breakage. Complementation group and mutation

    analysis facilitates genetic counselling.

    Table II. Classification of aplastic anaemia based on aetiology.

    Constitutional

    Fanconi anaemia (FA)

    Dyskeratosis congenita (DKC)

    ShwachmanDiamond syndrome (SDS)

    Congenital amegakaryocytic thrombocytopenia (CAMT)

    Acquired

    Radiation

    Drugs and chemicals e.g. chloramphenicol, benzene,

    anti-epileptics, chemotherapy

    Viruses e.g. Hepatitis (non-A,-B,-C,-E or -G), EpsteinBarr virus

    Graft-versus-host disease (GVHD)

    Paroxysmal nocturnal haemoglobinuria (PNH)

    Immune-systemic lupus erythematous (rare)

    Idiopathic

    Table I. Classification of aplastic anaemia based on severity.

    Severe aplastic anaemia (SAA)

    Marrow cellularity

  • Tab

    leIV

    .Characteristicsofinherited

    bonemarrowfailure

    syndromes

    that

    predispose

    toaplasticanaemia.

    Syndrome

    Age

    atpresentation

    (years)

    Haematologicalfeatures

    Nonhaematologicalfeatures

    Genemutation/Inheritance

    Screening/Diagnostictests

    Fanconianaemia

    (FA)

    Medianage65

    (range

    049)1

    M=F

    Progressive

    thrombocytopenia

    followed

    byAA.Macrocytosis

    Increasedrisk

    ofMDS/AML

    Lim

    b/thumbabnorm

    alities,

    cafe-au-laitspots,shortstature,

    microcephaly,urogenital

    anomalies

    Solidtumours

    (cumulative

    probabilityofmalignancy

    by

    50years85%)

    15genes

    (AR/X-linked)

    (FANCA,FANCB,FANCC,

    BCRA2(FANCD1),FANCD2,

    FANCE,FANCF,FANCG,

    FANCI,BRIP1(FANCJ),

    FANCL,FANCM,PALB2

    (FANCN),RAD15C(FANCO)

    andSL

    X4(FANCP)2

    Increasedchromosomal

    breakagebyDNAcross

    linkers

    inhaematopoieticcells(90%

    )

    orfibroblasts(100%)

    Dyskeratosis

    Congenita(D

    KC)

    Medianage14

    (range

    075)

    M>F

    AA,macrocytosis,

    MDS/AML

    90%

    developAAbythird

    decade

    Dystrophicnails,lacy

    reticular

    pigmentation,oralleucoplakia,

    solidtumours,pulm

    onary

    fibrosis,osteoporosis,cirrhosis

    8genes

    (AD/AR/X-linked)

    Flow-FISH

    fortelomerelength

    MolecularanalysisofDKC1,

    TERC,TERT,NOP10,NHP2,

    TIN

    F2,C16orf57andWRAP53

    (TCAB1)

    3

    Shwachman

    DiamondSyndrome

    (SDS)

    Range

    011

    M:F

    Neutropenia

    (77100%

    ),

    pancytopenia

    (1044%),

    MDS/AML(1333%)4

    Excocrinepancreaticfailure,

    metaphysealdysostosis,short

    stature

    SBDS(90%

    )

    AR

    Decreased

    serum

    trypsinogen/

    isoam

    ylase

    Reducedstoolelastase

    Imagingforfattypancreas

    MolecularanalysisofSB

    DSgene

    Congenital

    Amegakaryocytic

    Thrombocytopenia

    (CAMT)

    Range

    05

    M:F

    Thrombocytopenia

    with

    absentmegakaryocytesin

    bonemarrow

    Followed

    byAAin

    majority

    5

    Usually

    nosomatic

    abnorm

    alities

    MPL(thrombopoeitinreceptor

    gene)

    AR

    MolecularanalysisofMPLgene

    (However

    notallpatientshave

    MPLmutations)

    M,male;F,female;AA,aplasticanaemia;MDS,

    myelodysplasticsyndrome;AML,acute

    myeloid

    leukaem

    ia;AD,autosomaldominant;AR,autosomalrecessive.

    1Shim

    amura

    andAlter

    (2010),

    2Crossan

    andPatel(2012),

    3DokalandVulliamy(2010),Walneet

    al(2010),Zhonget

    al(2011),

    4Sm

    ithet

    al(1996),

    5Ballm

    aier

    andGermeshausen(2009).

    28 2012 Blackwell Publishing LtdBritish Journal of Haematology, 2012, 157, 2640

    Review

  • Testing for other IBMFS depends on clinical suspicion.

    DKC is typically characterized by the detection of very

    short telomeres in blood leucocytes (typically less than the

    1st centile for age; Alter et al, 2007). Flow cytometry with

    fluorescent in situ hybridization (Flow-FISH) can be used

    to screen for abnormal telomere length. However this test

    is not currently available as a routine clinical service. Fur-

    thermore, not all patients with DKC have short telomeres

    (Walne et al, 2010). Alternatively, blood can be sent for

    mutation screening but there are probably many unidenti-

    fied mutations (Dokal & Vulliamy, 2010). Thus a negative

    genetic screen is insufficient to exclude DKC. Children with

    mutations in TERC, TINF2 and TERT can present with just

    AA, without the other manifestations of DKC; thus it is

    reasonable to screen all children with idiopathic AA for

    these mutations if the FA screen is negative. The other

    genes can be screened for if there are additional features of

    DKC (personal communication Inderjeet Dokal, Centre for Pae-

    diatrics, Blizard Institute of Cell and Molecular Science, Barts

    and The London School of Medicine and Dentistry, London).

    If there is a clinical suspicion of ShwachmannDiamond

    Syndrome (SDS), exocrine pancreatic insufficiency can be

    screened for, though pancreatic function tends to improve

    with age. Molecular analysis of the SBDS gene can help con-

    firm the diagnosis, though 10% of children with SDS do not

    have a mutation in the SBDS gene (Boocock et al, 2003).

    Congenital amegakaryocytic thrombocytopenia (CAMT) is

    usually diagnosed on clinical features and confirmed by

    molecular analysis of the MPL gene (Ihara et al, 1999; Ball-

    maier & Germeshausen, 2009).

    Differential diagnosis

    Around 12% of childhood lymphoblastic leukaemia (ALL)

    cases are preceded by a period of pancytopenia, often with a

    hypocellular marrow, which subsequently develops into overt

    ALL around 19 months later (Breatnach et al, 1981). Dys-

    plastic granulopoeitic or megakaryocytes, increased reticulin,

    abnormal localization of immature precursors and increased

    blasts are seen in hypoplastic myelodysplastic syndrome

    (MDS) and not in AA (Bennett & Orazi, 2009). The detec-

    tion of monosomy 7 or 5q- should be considered as MDS

    (Marsh et al, 2009). The presence of isolated thrombocytope-

    nia can make distinguishing between ITP and AA (especially

    CAMT) difficult. ITP rather than AA is suggested by the

    presence of normal or increased numbers of megakaryocytes

    and increased reticulated platelet count.

    Key points

    AA is a rare disorder. About 7080% of cases are idio-

    pathic.

    Other potential causes of pancytopenia should be

    excluded.

    It is important to exclude IBMFS. Their presentation is

    heterogeneous and management different to idiopathic

    severe aplastic anaemia (SAA).

    All children should be screened for FA.

    Screening for other IBMFS will depend on clinical suspi-

    cion.

    Children with SAA and their families should be tissue

    typed at diagnosis. If there is no matched family member,

    an unrelated donor search should be undertaken.

    Management

    Transfusional support

    Red cell transfusions should be used only to treat definite

    symptoms/signs rather than maintain an arbitrary level. Leu-

    codepleted blood products (routine in the UK) should be

    given to reduce the risk of human leucocyte antigen (HLA)

    sensitization. Cytomegalovirus (CMV)-negative blood prod-

    ucts should be given until the patients CMV status is

    known. The European Bone Marrow Transplant Severe

    Aplastic Anaemia Working Party (EBMT SAAWP) currently

    recommends that children should receive irradiated blood

    products following immunosuppressive therapy (IST) and

    that this should continue for as long as they receive ciclospo-

    rin (Marsh et al, 2010). Repeated transfusions will lead to

    secondary iron overload. Iron chelation should be considered

    when the liver iron is >7 mg/g dry weight or when the totalred cell transfusion volume is >200 ml/kg. If liver iron mea-surement is not available, a persistently elevated ferritin level

    >1000 lg/l may be used as a surrogate marker of iron over-load, though this is a non-specific marker (Marsh et al,

    2009). Deferiprone should be avoided because of the risk of

    agranulocytosis. Desferrioxamine or deferasirox are however

    suitable iron chelators, the latter having the advantage of oral

    administration (Lee et al, 2010). Following successful treat-

    ment with IST or HSCT, venesection should be used to

    remove excess iron.

    Platelet transfusions can be given when the platelet count

    is

  • Infection prevention and treatment

    Anti-fungal prophylaxis should be given when the neutrophil

    count is

  • Predictors of response to IST

    Good prognostic factors that increase the likelihood of

    response to IST include severity [very severe aplastic anaemia

    (vSAA) better than SAA; Fuhrer et al, 2005], younger age,

    higher pre-treatment reticulocyte count and lymphocyte count

    (Scheinberg et al, 2008), male gender, and a leucocyte count

  • suppression. As late graft rejection is a characteristic com-

    plication of HSCT for SAA, ciclosporin should be contin-

    ued for at least 9 months, even in the absence of GVHD

    and tailed over the following 3 months. Chimerism should

    be monitored at 1, 3, 6 and 12 months post-HSCT and

    during tapering of immunosuppression. If there is an

    increase in recipient cells during this time period, ciclospo-

    rin should be increased to maintenance levels and a further

    attempt at withdrawal 3 months later (Lawler et al, 2009).

    Stable mixed chimerism is associated with very low rates of

    chronic GVHD and excellent outcome (McCann et al,

    2007). Late effects remain a potential concern. In a single

    centre analysis, fertility was preserved in 8090% of females

    and c. 60% of males with normal growth (Sanders et al,

    2011). Malignancy was reported in 713% on long-term

    follow up (Kahl et al, 2005; Sanders et al, 2011). Chronic

    GVHD and use of total body irradiation (TBI) regimens

    remain the major risk factors for the development of malig-

    nancy post-MSD HSCT.

    The conditioning regimen for MSD HSCT favoured to

    date in the UK is:

    1 Cyclophosphamide 50 mg/kg per d 5 to 2 (total dose200 mg/kg).

    2 Serotherapy is optional. If serotherapy is used, ale-

    mtuzumab is the preferred option, 03 mg/kg per d 6 to4 (total dose 09 mg/kg).

    3 Ciclosporin and methotrexate for GVHD prophylaxis.

    Methotrexate may be omitted if alemtuzumab is used.

    In view of the potential for impaired fertility seen with

    high dose cyclophosphamide, an alternative approach is a

    fludarabine-based regimen but using a lower dose of cyclo-

    phosphamide (fludarabine 150 mg/m2, cyclophosphamide

    120 mg/kg and alemtuzumab 09 mg/kg). A small seriesusing a similar approach showed promising results (Resnick

    et al, 2006).

    Key points

    MSD HSCT is first-line therapy for idiopathic SAA.

    Serotherapy is optional.

    GVHD prophylaxis consists of ciclosporin and methotrex-

    ate. Methotrexate may be omitted if alemtuzumab is used.

    Serial chimerism should be monitored post-HSCT.

    Unrelated donor HSCT

    Children who fail IST are eligible for a MUD HSCT. Out-

    comes over the last two decades have improved dramatically

    for MUD HSCT in SAA (Perez-Albuerne et al, 2008; Viollier

    et al, 2008), mainly due to the use of leucodepleted blood

    products, improvements in tissue typing donor/recipient and

    development of improved conditioning regimens (Maury

    et al, 2007). There are currently two approaches to condition-

    ing in MUD HSCT: a conditioning regimen incorporating

    low dose TBI (2 Gy; Deeg et al, 2006) and a radiation-free

    regimen using fludarabine (Bacigalupo et al, 2010). Deeg et al

    (2006) demonstrated improved outcomes using a regimen of

    2 Gy TBI, cyclophosphamide and ATG compared to higher

    doses of TBI. With this regimen, children who were trans-

    planted within a year of diagnosis had an 85% survival. The

    aim of the low dose TBI regimen was to optimize engraftment

    but minimize the long-term side effects associated with irradi-

    ation. There was however a relatively high incidence of acute

    GVHD (Grade IIIV 70%) and chronic GVHD (62%). The

    high levels of GVHD may be partly due to the pro-inflamma-

    tory effects of TBI. Administration of TBI also remains a

    long-term concern in children because of the effects on fertil-

    ity, growth, endocrine problems and potential for malignancy.

    This is all the more worrying because of the inherent risk in

    SAA to develop malignancy. In order to minimize these long-

    term side effects, a radiation-free regimen was developed by

    the EBMT (fludarabine 120 mg/kg, low dose cyclophospha-

    mide 1200 mg/m2 and rATG 15 mg/kg; Bacigalupo et al,

    2005). However this regimen has been complicated by rela-

    tively high rates of graft failure in older children (5%

    14 years, 32% if 15 years), post-transplant lymphopro-liferative disease (PTLD) and GVHD (Bacigalupo et al, 2005,

    2010). In view of these complications, a modified EBMT pro-

    tocol is now proposed (aged 14 years and not sensitized,fludarabine 120 mg/kg, cyclophosphamide 120 mg/kg, rATG

    75 mg/kg and prophylactic rituximab; 2 Gy TBI is added tothe aforementioned regimen for those patients aged

    15 years or sensitized; Kojima et al, 2011).In the UK, there has been considerable enthusiasm for

    using alemtuzumab (campath-1H), a monoclonal anti-CD52

    antibody. A retrospective analysis of 44 consecutive children

    who received HLA-A, -B, -C, -DRB1, -DQ matched unrelated

    donor HSCTs using a fludarabine (150 mg/kg), cyclophos-

    phamide (120 or 200 mg/kg) and alemtuzumab regimen (091 mg/kg; FCC regimen) demonstrated excellent outcome.

    There were no cases of graft failure, with an estimated 5 year

    OS/FFS of 95% (Samarasinghe et al, 2012). At a median of

    29 years follow up, there was a low rate of severe acuteGVHD (grades IIIIV 23%) and chronic GVHD (68%). Thelow rates of chronic GVHD are similar to other groups who

    have also used alemtuzumab-based conditioning regimens

    (Marsh et al, 2011). There were no cases of PTLD although

    two children did require rituximab because of EpsteinBarr

    Virus reactivation. This data, along with that reported by

    others (Kennedy-Nasser et al, 2006), suggests that outcomes

    following MUD HSCT (10/10 by high resolution typing) for

    paediatric SAA are similar to that of MSD. As excellent

    engraftment can be achieved in the absence of TBI, the best

    approach would involve a radiation-free regimen (such as the

    FCC regimen) in an attempt to minimize long-term side

    effects. However until long-term data is available, it is difficult

    to be certain whether radiation-free regimens will prevent

    32 2012 Blackwell Publishing LtdBritish Journal of Haematology, 2012, 157, 2640

    Review

  • long-term side effects. Therefore, post-pubertal males receiv-

    ing a HSCT should have sperm cryopreserved.

    The conditioning regimen recommended by the UK Pae-

    diatric BMT group for 10/10 (HLA-A, -B, -C, -DRB1, -DQ

    matched by high resolution) MUD HSCT is (FCC regimen):

    1 Fludarabine 30 mg/m2 per d for 5 d: days 6 to 2(Total dose 150 mg/m2).

    2 Cyclophosphamide 60 mg/kg per d for 2 d: days 3 to2 (Total dose 120 mg/kg).

    3 Alemtuzumab 03 mg/kg per d for 3 d: days 6 to 4(Total dose 09 mg/kg: cap dose at 50 mg).With ciclosporin prophylaxis.

    Data on mismatched unrelated donor HSCT and unre-

    lated donor umbilical cord HSCT in idiopathic SAA is lim-

    ited. Retrospective data suggest that single allele mismatched

    unrelated donor (MMUD) HSCT have a reasonably good

    outcome (78% 2-year OS for 8/8 vs. 60% for 7/8; Eapen &

    Horowitz, 2010). Furthermore, with the advent of high reso-

    lution typing, single antigen/allele MMUD may have a simi-

    lar outcome to MUD HSCT (Yagasaki et al, 2011). Due to

    the low cell dose in umbilical cord donations, initial reports

    of using unrelated donor umbilical cord HSCT for idiopathic

    SAA have been discouraging because of the high graft failure

    rate and treatment-related mortality (TRM). OS in the two

    largest retrospective analyses to date have ranged from 30%

    to 40% (Yoshimi et al, 2008; Peffault de Latour et al, 2011).

    Improved results were seen with higher total nucleated cell

    (TNC) doses (OS was 45% for TNC > 39 9 107/kg vs. 18%for TNC 39 9 107/kg; Peffault de Latour et al, 2011).An impressive OS was seen in a small series of adults who

    received unrelated umbilical cord HSCT using a fludarabine,

    melphalan and 4 Gy TBI conditioning regimen (3-year OS of

    83%), but these results will need to be confirmed in further

    studies (Yamamoto et al, 2011). Selecting units to which the

    recipient does not have anti-HLA antibodies may also

    improve outcomes (Takanashi et al, 2010).

    Key points

    MUD HSCT (a HLA-A, -B, -C, -DRB1, -DQ matched

    donor on high resolution typing) has an excellent outcome

    in idiopathic SAA.

    Radiation-free conditioning regimens are favoured in Eur-

    ope.

    Alemtuzumab-based conditioning regimens have low rates

    of chronic GVHD.

    Stem cell choice

    The recommended stem source is bone marrow. PBSCs lead

    to an increased risk of chronic GVHD and inferior outcome

    (Schrezenmeier et al, 2007; Eapen et al, 2011). However, use

    of alemtuzumab-based conditioning regimens may minimize

    the effect of PBSCs on chronic GVHD (Marsh et al, 2011;

    Shaw et al, 2011). Umbilical cord stem cells from a MSD are

    also acceptable though rarely available.

    Key points

    Bone marrow is stem cell of choice.

    Algorithm for idiopathic paediatric SAA

    A recent algorithm for childhood SAA (Marsh et al, 2009)

    states that a MSD HSCT is the treatment of choice. Those

    children lacking a MSD should receive IST with ATG/ciclo-

    sporin as second choice. Should they fail IST (response

    assessment at 34 months), then they should proceed with

    MUD HSCT as third choice. However, with recent data

    demonstrating the low efficacy of rATG and the steady

    improvement in MUD HSCT, a new algorithm is proposed

    (see Fig 1). In the proposed algorithm, MSD HSCT remains

    the first choice. Whether IST or MUD HSCT should how-

    ever be second choice is contentious. Advantages of IST

    with hATG include excellent long-term survival and low

    TRM. However, IST has a significant RR (at least 10%; Sar-

    acco et al, 2008), and a 10-year risk of developing a clonal

    disorder of between 10% and 15%. Furthermore, IST takes

    at least 34 months for cellular recovery, which is particu-

    larly relevant if a child develops infectious complications.

    MUD HSCT now has an excellent outcome with a much

    quicker neutrophil recovery and a marked reduction in

    development of secondary clonal disorders and relapse com-

    pared to IST (Socie et al, 1993). Although MUD HSCT car-

    ries a higher risk of early mortality, the subsequent survival

    curve is stable. In contrast, due to the increased risk of sec-

    ondary clonal disorders, no such plateau in survival is seen

    following IST. Historically, HSCT was associated with the

    upfront toxicities of GVHD, graft failure and infectious

    complications. With improvements in supportive care, tis-

    sue typing and conditioning regimens, the TRM for a

    MUD HSCT in children is similar to that of MSD. The dis-

    advantage with MUD HSCT includes the time taken to find

    a suitable donor (typically 3 months before HSCT can pro-

    ceed) and the difficulty finding a MUD in children with

    more rare HLA haplotypes.

    In those countries where hATG is available, the choice

    between IST and MUD HSCT (when a 10/10 MUD donor

    exists) will require a careful discussion between the physician

    and family regarding the different risks. To help determine

    whether one should proceed with hATG or MUD HSCT,

    urgent tissue typing should be done and a judgement on the

    likelihood of finding a MUD can then be made. Should IST

    fail with hATG, the child should proceed directly to MUD

    HSCT (see Fig 1). Horse ATG (ATGAM) is not currently

    available in Europe, though the EBMT SAAWP is urgently

    trying to change this. Until then, in Europe due to the disap-

    2012 Blackwell Publishing Ltd 33British Journal of Haematology, 2012, 157, 2640

    Review

  • pointing results with rATG, a MUD HSCT should be consid-

    ered as the second choice (where a suitable donor exists).

    The EBMT SAAWP have issued similar guidance (EBMTG

    SAAWP, 2011).

    In the proposed algorithm, IST with rATG/ciclosporin

    would be the third choice. The EBMT SAAWP have sug-

    gested that rATG be used at 25 mg/kg per d for 5 d ratherthan 375 mg/kg per d for 5 d (http://www.bcshguidelines.com/documents/Use_of_rabbit_ATG_July_2011.pdf) as rATG

    is more immunosuppressive than hATG. Single allele/antigen

    MMUD may be considered a suitable alternative to IST with

    rATG. For those children lacking a suitable unrelated donor

    (10/10 or 9/10) and failing IST, possible options include a

    second course of ATG, an alternative IST or umbilical cord/

    haploidentical HSCT. Alternative IST options include high

    dose cyclophosphamide (Brodsky et al, 2010) or ale-

    mtuzumab (Risitano et al, 2010). However further studies

    are required in children to determine their long-term efficacy

    and optimal dosing.

    Non-severe aplastic anaemia (NSAA)

    Transfusion-independent children with a neutrophil count

    above 05 9 109/l should be observed. If they are transfu-sion-dependent, or have a neutrophil count

  • conditioning regimens were associated with high TRM. As

    such, Gluckman et al (1984) proposed the use of a condi-

    tioning regimen of low dose cyclophosphamide (2040 mg/

    kg) combined with 400600 cGy of thoraco-abdominal irra-

    diation or TBI with ciclosporin for GVHD prophylaxis. OS

    rates of between 80% and 90% have been reported using this

    approach (Dufour et al, 2001; Farzin et al, 2007). FA patients

    demonstrate increased propensity to severe GVHD (Guardi-

    ola et al, 2004). The occurrence of severe acute GVHD

    (grades IIIV) and chronic GVHD strongly increased the risk

    of oropharngeal/anogenital squamous cell carcinomas in

    long-term FA survivors, with a 15-year incidence of head

    and neck cancers of 53% in one series (Guardiola et al,

    2004). There is no evidence currently that low dose chemora-

    diotherapy conditioning regimens used in FA HSCTs increase

    the risk of long-term malignancy. However there has been a

    move to avoid irradiation-based conditioning regimens and

    incorporate T-cell depletion to minimize the risk of GVHD,

    with the aim of reducing the risk of malignancy. A retrospec-

    tive comparison of irradiation-based regimens with radia-

    tion-free regimens showed no difference in OS (Pasquini

    et al, 2008). Prior use of androgens, age >10 years and CMVpositivity in either donor/recipient were adverse factors.

    The optimal radiation-free conditioning regimen is

    unknown. The favoured conditioning regimen for FA within

    the UK incorporates fludarabine and low dose cyclophospha-

    mide with serotherapy (de la Fuente et al, 2003). Whether

    such regimens can reduce late effects will require long-term

    follow up. All matched family donors should be screened

    very carefully for FA even if they do not show features of the

    disorder. Bone marrow rather than PBSCs is the stem cell of

    choice.

    Current indications for a MSD HSCT in FA are (please

    see www.fanconi.org.uk/clinical-network/standards-of-care/):

    1 Significant cytopenia/moderate BMF (haemoglobin

  • Dyskeratosis congenita (DKC)

    Dyskeratosis congenita is an inherited disorder of telomere

    maintenance characterized by mucocutaneous abnormalities,

    BMF and increased predisposition to malignancy. There is

    considerable genetic and phenotypic heterogeneity, which

    can make diagnosis difficult (Vulliamy et al, 2006). For a

    review of the genetics please see recent article (Dokal & Vul-

    liamy, 2010). BMF/immunodeficiency is the major cause of

    death, followed by pulmonary complications and malignancy

    (Walne & Dokal, 2009). Allogeneic HSCT is the only curative

    option for patients with BMF. However, high TRMs were

    encountered using myeloablative conditioning regimens. To

    counter this, reduced-intensity conditioning (RIC) regimens

    have successfully been used (Dietz et al, 2011). However, the

    numbers are too small to make any recommendations

    regarding the optimal RIC regimen.

    For those who lack a MSD or MUD, oxymetholone or

    growth factors can be used. The same caveats that guide use

    of androgens in FA apply in DKC. GCSF should not be used

    with oxymethalone because of the risk of splenic rupture

    (Shimamura & Alter, 2010).

    ShwachmannDiamond syndrome (SDS)

    ShwachmannDiamond syndrome is an autosomal recessive

    disorder characterized by exocrine pancreatic insufficiency,

    BMF and other somatic abnormalities (especially metaphyseal

    dysostosis). A trial of GCSF may be considered to ameliorate

    infection or prevent recurrent sepsis. The lowest possible

    dose that maintains adequate levels of neutrophils should be

    used. No association between GCSF administration and

    malignancy has been demonstrated (Rosenberg et al, 2006).

    Indications for HSCT include significant cytopenias, transfu-

    sion dependence and severe recurrent sepsis secondary to

    persistent neutropenia (Burroughs et al, 2009). However,

    HSCT in SDS using myeloablative conditioning is associated

    with increased TRM. The optimal conditioning regimen is

    unknown but small case series have demonstrated the efficacy

    of a RIC approach (Bhatla et al, 2008).

    Congenital amegakaryocytic thrombocytopenia(CAMT)

    This is an autosomal recessive IBMFS characterized by severe

    thrombocytopenia at birth with a lack of or absence of

    megakaryocytes in the bone marrow. The disorder is charac-

    terized by a mutation in the thrombopoeitin receptor (MPL

    gene). The diagnosis is one of exclusion; please see the recent

    review by Ballmaier and Germeshausen (2009). HSCT from a

    MSD after development of SAA has a good outcome (Ball-

    maier & Germeshausen, 2009). CAMT patients do not show

    increased TRM like FA or DKC patients following myeloab-

    lative regimens. Increasing success has also been seen using

    unrelated donor HSCTs with RIC regimens, although num-

    bers remain small (Tarek et al, 2011).

    Acknowledgements

    The authors are grateful for Professor Irene Roberts and Dr.

    Rod Skinner for reviewing the script.

    Author contributions

    SS and DW wrote and edited the review.

    Financial disclosures

    There are no financial disclosures.

    References

    Alter, B.P., Baerlocher, G.M., Savage, S.A.,

    Chanock, S.J., Weksler, B.B., Willner, J.P.,

    Peters, J.A., Giri, N. & Lansdorp, P.M.

    (2007) Very short telomere length by flow

    fluorescence in situ hybridization identifies

    patients with dyskeratosis congenita. Blood,

    110, 14391447.

    Bacigalupo, A., Locatelli, F., Lanino, E., Marsh, J.,

    Socie, G., Maury, S., Prete, A., Locasciulli, A.,

    Cesaro, S. & Passweg, J. (2005) Fludarabine,

    cyclophosphamide and anti-thymocyte globulin

    for alternative donor transplants in acquired

    severe aplastic anemia: a report from the

    EBMT-SAA Working Party. Bone Marrow Trans-

    plantation, 36, 947950.

    Bacigalupo, A., Socie, G., Lanino, E., Prete, A., Lo-

    catelli, F., Locasciulli, A., Cesaro, S., Shimoni,

    A., Marsh, J., Brune, M., Van Lint, M.T., Oneto,

    R. & Passweg, J. (2010) Fludarabine, cyclophos-

    phamide, antithymocyte globulin, with or with-

    out low dose total body irradiation, for

    alternative donor transplants, in acquired severe

    aplastic anemia: a retrospective study from the

    EBMT-SAA Working Party. Haematologica, 95,

    976982.

    Ballmaier, M. & Germeshausen, M. (2009)

    Advances in the understanding of congenital

    amegakaryocytic thrombocytopenia. British Jour-

    nal of Haematology, 146, 316.

    Bennett, J.M. & Orazi, A. (2009) Diagnostic crite-

    ria to distinguish hypocellular acute myeloid

    leukemia from hypocellular myelodysplastic syn-

    dromes and aplastic anemia: recommendations

    for a standardized approach. Haematologica, 94,

    264268.

    Bhatla, D., Davies, S.M., Shenoy, S., Harris, R.E.,

    Crockett, M., Shoultz, L., Smolarek, T., Bleesing,

    J., Hansen, M., Jodele, S., Jordan, M., Filipovich,

    A.H. & Mehta, P.A. (2008) Reduced-intensity

    conditioning is effective and safe for transplan-

    tation of patients with ShwachmanDiamond

    syndrome. Bone Marrow Transplantation, 42,

    159165.

    Boocock, G.R., Morrison, J.A., Popovic, M., Rich-

    ards, N., Ellis, L., Durie, P.R. & Rommens, J.M.

    (2003) Mutations in SBDS are associated with

    ShwachmanDiamond syndrome. Nature Genet-

    ics, 33, 97101.

    Breatnach, F., Chessells, J.M. & Greaves, M.F.

    (1981) The aplastic presentation of childhood

    leukaemia: a feature of common-ALL. British

    Journal of Haematology, 49, 387393.

    Brodsky, R.A., Chen, A.R., Dorr, D., Fuchs, E.J.,

    Huff, C.A., Luznik, L., Smith, B.D., Matsui, W.

    H., Goodman, S.N., Ambinder, R.F. & Jones, R.

    J. (2010) High-dose cyclophosphamide for

    severe aplastic anemia: long-term follow-up.

    Blood, 115, 21362141.

    Burroughs, L., Woolfrey, A. & Shimamura, A.

    (2009) ShwachmanDiamond syndrome: a

    review of the clinical presentation, molecular

    36 2012 Blackwell Publishing LtdBritish Journal of Haematology, 2012, 157, 2640

    Review

  • pathogenesis, diagnosis, and treatment. Hematol-

    ogy/Oncology Clinics of North America, 23, 233

    248.

    Calado, R.T. & Young, N.S. (2008) Telomere

    maintenance and human bone marrow failure.

    Blood, 111, 44464455.

    Camitta, B.M., Rappeport, J.M., Parkman, R. &

    Nathan, D.G. (1975) Selection of patients for

    bone marrow transplantation in severe aplastic

    anemia. Blood, 45, 355363.

    Champlin, R., Perez, W., Passweg, J., Klein, J., Ca-

    mitta, B., Gluckman, E., Bredeson, C., Eapen,

    M. & Horowitz, M. (2007) Bone marrow trans-

    plantation for severe apalstic anaemia: a ran-

    domized controlled study of conditioning

    regimens. Blood, 109, 45824585.

    Crossan, G. & Patel, K. (2012) The Fanconi Anae-

    mia Pathway Orchestrates incisions at sites of

    crosslinked DNA. Journal of Pathology, 226, 326

    337.

    Deeg, H.J., ODonnell, M., Tolar, J., Agarwal, R.,

    Harris, R.E., Feig, S.A., Territo, M.C., Collins, R.

    H., McSweeney, P.A., Copelan, E.A., Khan, S.P.,

    Woolfrey, A. & Storer, B. (2006) Optimization

    of conditioning for marrow transplantation

    from unrelated donors for patients with aplastic

    anemia after failure of immunosuppressive ther-

    apy. Blood, 108, 14851491.

    Dietz, A.C., Orchard, P.J., Baker, K.S., Giller, R.H.,

    Savage, S.A., Alter, B.P. & Tolar, J. (2011) Dis-

    ease-specific hematopoietic cell transplantation:

    nonmyeloablative conditioning regimen for dys-

    keratosis congenita. Bone Marrow Transplanta-

    tion, 46, 98104.

    Dokal, I. (2000) Dyskeratosis congenita in all its

    forms. British Journal Haematology, 110, 768

    779.

    Dokal, I. & Vulliamy, T. (2010) Inherited bone

    marrow failure syndromes. Haematologica, 95,

    12361240.

    Dror, Y., Ginzberg, H., Dalal, I., Cherepanov, V.,

    Downey, G., Durie, P., Roifman, C.M. & Freed-

    man, M.H. (2001) Immune function in patients

    with ShwachmanDiamond syndrome. British

    Journal Haematology, 114, 712717.

    Dufour, C., Rondelli, R., Locatelli, F., Miano, M.,

    Di Girolamo, G., Bacigalupo, A., Messina, C.,

    Porta, F., Balduzzi, A., Iorio, A.P., Buket, E.,

    Madon, E., Pession, A., Dini, G. & Di Bartolo-

    meo, P. (2001) Stem cell transplantation from

    HLA-matched related donor for Fanconis anae-

    mia: a retrospective review of the multicentric

    Italian experience on behalf of AIEOP-GITMO.

    British Journal Haematology, 112, 796805.

    Eapen, M. & Horowitz, M.M. (2010) Alternative

    donor transplantation for aplastic anemia.

    Hematology American Society of Hematology Edu-

    cation Program, 2010, 4346.

    Eapen, M., Le Rademacher, J., Antin, J.H., Cham-

    plin, R.E., Carreras, J., Fay, J., Passweg, J.R.,

    Tolar, J., Horowitz, M.M., Marsh, J.C. & Deeg,

    H.J. (2011) Effect of stem cell source on out-

    comes after adult unrelated donor transplanta-

    tion in severe aplastic anemia. Blood, 118, 2618

    2621.

    European Blood and Marrow Transplant Group,

    Severe Aplastic Anaemia Working Party (2011)

    Rabbit ATG for aplastic anaemia treatment: a

    backward step? Lancet, 378, 18311833.

    Farzin, A., Davies, S.M., Smith, F.O., Filipovich,

    A., Hansen, M., Auerbach, A.D. & Harris, R.E.

    (2007) Matched sibling donor haematopoietic

    stem cell transplantation in Fanconi anaemia: an

    update of the Cincinnati Childrens experience.

    British Journal Haematology, 136, 633640.

    Filipovich, A.H., Zhang, K., Snow, A.L. & Marsh,

    R.A. (2010) X-linked lymphoproliferative syn-

    dromes: brothers or distant cousins? Blood, 116,

    33983408.

    Fogarty, P.F., Yamaguchi, H., Wiestner, A., Baerl-

    ocher, G.M., Sloand, E., Zeng, W.S., Read, E.J.,

    Lansdorp, P.M. & Young, N.S. (2003) Late pre-

    sentation of dyskeratosis congenita as apparently

    acquired aplastic anaemia due to mutations in

    telomerase RNA. Lancet, 362, 16281630.

    Frickhofen, N., Heimpel, H., Kaltwasser, J.P. &

    Schrezenmeier, H. (2003) Antithymocyte globu-

    lin with or without cyclosporin A: 11-year fol-

    low-up of a randomized trial comparing

    treatments of aplastic anemia. Blood, 101, 1236

    1242.

    de la Fuente, J., Reiss, S., McCloy, M., Vulliamy,

    T., Roberts, I., Rahemtulla, A. & Dokal, I.

    (2003) Non-TBI stem cell transplantation proto-

    col for Fanconi anamia using HLA-compatible

    sibling and unrelated donors. Bone Marrow

    Transplantation, 32, 653656.

    Fuhrer, M., Rampf, U., Baumann, I., Faldum, A.,

    Niemeyer, C., Janka-Schaub, G., Friedrich, W.,

    Ebell, W., Borkhardt, A. & Bender-Goetze, C.

    (2005) Immunosuppressive therapy for aplastic

    anemia in children: a more severe disease pre-

    dicts better survival. Blood, 106, 21022104.

    Gluckman, E., Berger, R. & Dutreix, J. (1984) Bone

    marrow transplantation for Fanconi anemia.

    Seminars in Hematology, 21, 2026.

    Gluckman, E., Rocha, V., Ionescu, I., Bierings, M.,

    Harris, R.E., Wagner, J., Kurtzberg, J., Cham-

    pagne, M.A., Bonfim, C., Bittencourt, M., Dar-

    byshire, P., Fernandez, M.N., Locatelli, F. &

    Pasquini, R. (2007) Results of unrelated cord

    blood transplant in fanconi anemia patients: risk

    factor analysis for engraftment and survival.

    Biology of Blood and Marrow Transplantation,

    13, 10731082.

    Guardiola, P., Socie, G., Li, X., Ribaud, P., Dever-

    gie, A., Esperou, H., Richard, P., Traineau, R.,

    Janin, A. & Gluckman, E. (2004) Acute graft-

    versus-host disease in patients with Fanconi ane-

    mia or acquired aplastic anemia undergoing

    bone marrow transplantation from HLA-identi-

    cal sibling donors: risk factors and influence on

    outcome. Blood, 103, 7377.

    Gupta, V., Ball, S.E., Yi, Q.L., Sage, D., McCann,

    S.R., Lawler, M., Ortin, M., Freires, M., Hale,

    G., Waldmann, H., Gordon-Smith, E.C. &

    Marsh, J.C. (2004) Favorable effect on acute and

    chronic graft-versus-host disease with cyclophos-

    phamide and in vivo anti-CD52 monoclonal

    antibodies for marrow transplantation from

    HLA-identical sibling donors for acquired aplas-

    tic anemia. Biology of Blood and Marrow Trans-

    plantation, 10, 867876.

    Gupta, V., Gordon-Smith, E.C., Cook, G., Parker,

    A., Duguid, J.K., Wilson, K.M., Yi, Q.L. &

    Marsh, J.C. (2005) A third course of anti-thy-

    mocyte globulin in aplastic anaemia is only ben-

    eficial in previous responders. British Journal

    Haematology, 129, 110117.

    Hendry, C.L., Sivakumaran, M., Marsh, J.C. &

    Gordon-Smith, E.C. (2002) Relapse of severe

    aplastic anaemia after influenza immunization.

    British Journal Haematology, 119, 283284.

    Ihara, K., Ishii, E., Eguchi, M., Takada, H., Sumi-

    noe, A., Good, R.A. & Hara, T. (1999) Identifi-

    cation of mutations in the c-mpl gene in

    congenital amegakaryocytic thrombocytopenia.

    Proceedings of the National Academy of Sciences

    of the United States of America, 96, 31323136.

    Kahl, C., Leisenring, W., Deeg, H.J., Chauncey, T.

    R., Flowers, M.E., Martin, P.J., Sanders, J.E. &

    Storb, R. (2005) Cyclophosphamide and antithy-

    mocyte globulin as a conditioning regimen for

    allogeneic marrow transplantation in patients

    with aplastic anaemia: a long-term follow-up.

    British Journal Haematology, 130, 747751.

    Kamio, T., Ito, E., Ohara, A., Kosaka, Y., Tsuchida,

    M., Yagasaki, H., Mugishima, H., Yabe, H.,

    Morimoto, A., Ohga, S., Muramatsu, H., Hama,

    A., Kaneko, T., Nagasawa, M., Kikuta, A., Osugi,

    Y., Bessho, F., Nakahata, T., Tsukimoto, I. &

    Kojima, S. (2011) Relapse of aplastic anemia in

    children after immunosuppressive therapy: a

    report from the Japan Childhood Aplastic Ane-

    mia Study Group. Haematologica, 96, 814819.

    Kennedy-Nasser, A.A., Leung, K.S., Mahajan, A.,

    Weiss, H.L., Arce, J.A., Gottschalk, S., Carrum,

    G., Khan, S.P., Heslop, H.E., Brenner, M.K.,

    Bollard, C.M. & Krance, R.A. (2006) Compara-

    ble outcomes of matched-related and alternative

    donor stem cell transplantation for pediatric

    severe aplastic anemia. Biology of Blood and

    Marrow Transplantation, 12, 12771284.

    Knudson, M., Kulkarni, S., Ballas, Z.K., Bessler, M.

    & Goldman, F. (2005) Association of immune

    abnormalities with telomere shortening in auto-

    somal-dominant dyskeratosis congenita. Blood,

    105, 682688.

    Kobayashi, R., Yabe, H., Hara, J., Morimoto, A.,

    Tsuchida, M., Mugishima, H., Ohara, A., Tsu-

    kimoto, I., Kato, K., Kigasawa, H., Tabuchi, K.,

    Nakahata, T., Ohga, S. & Kojima, S. (2006) Pre-

    ceding immunosuppressive therapy with antithy-

    mocyte globulin and ciclosporin increases the

    incidence of graft rejection in children with

    aplastic anaemia who underwent allogeneic bone

    marrow transplantation from HLA-identical sib-

    lings. British Journal Haematology, 135, 693696.

    Kojima, S., Horibe, K., Inaba, J., Yoshimi, A., Ta-

    kahashi, Y., Kudo, K., Kato, K. & Matsuyama,

    T. (2000) Long-term outcome of acquired aplas-

    tic anaemia in children: comparison between

    immunosuppressive therapy and bone marrow

    transplantation. British Journal Haematology,

    111, 321328.

    2012 Blackwell Publishing Ltd 37British Journal of Haematology, 2012, 157, 2640

    Review

  • Kojima, S., Nakao, S., Young, N., Bacigalupo, A.,

    Gerard, G., Hirano, N., Maciejewski, J., Deeg, J.,

    Marsh, J., Zhang, F.K., Lee, J.W. & Ozawa, K.

    (2011) The Third Consensus Conference on the

    treatment of aplastic anemia. International Jour-

    nal of Hematology, 93, 832837.

    Kosaka, Y., Yagasaki, H., Sano, K., Kobayashi,

    R., Ayukawa, H., Kaneko, T., Yabe, H.,

    Tsuchida, M., Mugishima, H., Ohara, A.,

    Morimoto, A., Otsuka, Y., Ohga, S., Bessho,

    F., Nakahata, T., Tsukimoto, I. & Kojima, S.

    (2008) Prospective multicenter trial compar-

    ing repeated immunosuppressive therapy with

    stem-cell transplantation from an alternative

    donor as second-line treatment for children

    with severe and very severe aplastic anemia.

    Blood, 111, 10541059.

    Kutler, D.I., Singh, B., Satagopan, J., Batish, S.D.,

    Berwick, M., Giampietro, P.F., Hanenberg, H. &

    Auerbach, A.D. (2003) A 20-year perspective on

    the International Fanconi Anemia Registry

    (IFAR). Blood, 101, 12491256.

    Lawler, M., McCann, S.R., Marsh, J.C., Ljungman,

    P., Hows, J., Vandenberghe, E., ORiordan, J.,

    Locasciulli, A., Socie, G., Kelly, A., Schrezenme-

    ier, H., Marin, P., Tichelli, A., Passweg, J.R.,

    Dickenson, A., Ryan, J. & Bacigalupo, A. (2009)

    Serial chimerism analyses indicate that mixed

    haemopoietic chimerism influences the probabil-

    ity of graft rejection and disease recurrence fol-

    lowing allogeneic stem cell transplantation

    (SCT) for severe aplastic anaemia (SAA): indica-

    tion for routine assessment of chimerism post

    SCT for SAA. British Journal of Haematology,

    144, 933945.

    Lee, J.W., Yoon, S.S., Shen, Z.X., Ganser, A., Hsu,

    H.C., Habr, D., Domokos, G., Roubert, B. &

    Porter, J.B. (2010) Iron chelation therapy with

    deferasirox in patients with aplastic anemia: a

    subgroup analysis of 116 patients from the EPIC

    trial. Blood, 116, 24482454.

    Lo Ten Foe, J.R., Kwee, M.L., Rooimans, M.A.,

    Oostra, A.B., Veerman, A.J., van Weel, M., Pau-

    li, R.M., Shahidi, N.T., Dokal, I., Roberts, I., Al-

    tay, C., Gluckman, E., Gibson, R.A., Mathew, C.

    G., Arwert, F. & Joenje, H. (1997) Somatic

    mosaicism in Fanconi anemia: molecular basis

    and clinical significance. European Journal of

    Human Genetics, 5, 137148.

    Locatelli, F., Bruno, B., Zecca, M., Van-Lint, M.T.,

    McCann, S., Arcese, W., Dallorso, S., Di Bartol-

    omeo, P., Fagioli, F., Locasciulli, A., Lawler, M.

    & Bacigalupo, A. (2000) Cyclosporin A and

    short-term methotrexate versus cyclosporin A as

    graft versus host disease prophylaxis in patients

    with severe aplastic anemia given allogeneic

    bone marrow transplantation from an HLA-

    identical sibling: results of a GITMO/EBMT ran-

    domized trial. Blood, 96, 16901697.

    MacMillan, M.L. & Wagner, J.E. (2010) Haemato-

    poeitic cell transplantation for Fanconi anaemia

    when and how? British Journal of Haematology,

    149, 1421.

    Marsh, J.C., Ball, S.E., Cavenagh, J., Darbyshire, P.,

    Dokal, I., Gordon-Smith, E.C., Keidan, J., Lau-

    rie, A., Martin, A., Mercieca, J., Killick, S.B.,

    Stewart, R. & Yin, J.A. (2009) Guidelines for the

    diagnosis and management of aplastic anaemia.

    British Journal Haematology, 147, 4370.

    Marsh, J., Socie, G., Tichelli, A., Schrezenmeier,

    H., Hochsmann, B., Risitano, A.M., Fuehrer, M.,

    Bekassy, A.N., Korthof, E.T., Locasciulli, A.,

    Ljungman, P., Bacigalupo, A., Camitta, B.,

    Young, N.S. & Passweg, J. (2010) Should irradi-

    ated blood products be given routinely to all

    patients with aplastic anaemia undergoing

    immunosuppressive therapy with antithymocyte

    globulin (ATG)? A survey from the European

    Group for Blood and Marrow Transplantation

    Severe Aplastic Anaemia Working Party. British

    Journal Haematology, 150, 377379.

    Marsh, J.C., Gupta, V., Lim, Z., Ho, A.Y., Ireland,

    R.M., Hayden, J., Potter, V., Koh, M.B., Islam,

    M.S., Russell, N., Marks, D.I., Mufti, G.J. & Pa-

    gliuca, A. (2011) Alemtuzumab with fludarabine

    and cyclophosphamide reduces chronic graft-

    versus-host disease after allogeneic stem cell

    transplantation for acquired aplastic anemia.

    Blood, 118, 23512357.

    Maury, S., Balere-Appert, M.L., Chir, Z., Boiron, J.

    M., Galambrun, C., Yakouben, K., Bordigoni,

    P., Marie-Cardine, A., Milpied, N., Kanold, J.,

    Maillard, N. & Socie, G. (2007) Unrelated stem

    cell transplantation for severe acquired aplastic

    anemia: improved outcome in the era of high-

    resolution HLA matching between donor and

    recipient. Haematologica, 92, 589596.

    McCann, S., Passweg, J., Bacigalupo, A., Locasciul-

    li, A., Locatelli, F., Ryan, J., Schrezenmeier, H. &

    Lawler, M. (2007) The influence of cyclosporin

    alone, or cyclosporin and methotrexate, on the

    incidence of mixed haematopoietic chimaerism

    following allogeneic sibling bone marrow trans-

    plantation for severe aplastic anaemia. Bone

    Marrow Transplantation, 39, 109114.

    Montane, E., Ibanez, L., Vidal, X., Ballarin, E.,

    Puig, R., Garcia, N. & Laporte, J.R. (2008) Epi-

    demiology of aplastic anemia: a prospective

    multicenter study. Haematologica, 93, 518523.

    Niederwieser, D., Pepe, M., Storb, R., Loughran,

    Jr, T.P. & Longton, G. (1988) Improvement in

    rejection, engraftment rate and survival without

    increase in graft-versus-host disease by high

    marrow cell dose in patients transplanted for

    aplastic anaemia. British Journal of Haematology,

    69, 2328.

    Pasquini, R., Carreras, J., Pasquini, M.C., Camitta,

    B.M., Fasth, A.L., Hale, G.A., Harris, R.E.,

    Marsh, J.C., Robinson, A.J., Zhang, M.J., Eapen,

    M. & Wagner, J.E. (2008) HLA-matched sibling

    hematopoietic stem cell transplantation for fan-

    coni anemia: comparison of irradiation and

    nonirradiation containing conditioning regi-

    mens. Biology of Blood and Marrow Transplanta-

    tion, 14, 11411147.

    Passweg, J.R. & Marsh, J.C. (2010) Aplastic ane-

    mia: first-line treatment by immunosuppression

    and sibling marrow transplantation. Hematology

    American Society of Hematology Education Pro-

    gram, 2010, 3642.

    Passweg, J.R. & Tichelli, A. (2009) Immunosup-

    pressive treatment for aplastic anemia: are we

    hitting the ceiling? Haematologica, 94, 310312.

    Peffault de Latour, R., Purtill, D., Ruggeri, A.,

    Sanz, G., Michel, G., Gandemer, V., Maury, S.,

    Kurtzberg, J., Bonfim, C., Aljurf, M., Gluckman,

    E., Socie, G., Passweg, J. & Rocha, V. (2011)

    Influence of nucleated cell dose on overall sur-

    vival of unrelated cord blood transplantation for

    patients with severe acquired aplastic anemia: a

    study by eurocord and the aplastic anemia

    working party of the European group for blood

    and marrow transplantation. Biology of Blood

    and Marrow Transplantation, 17, 7885.

    Perez-Albuerne, E.D., Eapen, M., Klein, J., Gross,

    T.J., Lipton, J.M., Baker, K.S., Woolfrey, A. &

    Kamani, N. (2008) Outcome of unrelated donor

    stem cell transplantation for children with severe

    aplastic anemia. British Journal of Haematology,

    141, 216223.

    Resnick, I., Aker, M., Shapira, M., Tsirigotis, P.,

    Bitan, M., Abdul-Hai, A., Samuel, S., Ackerstein,

    A., Gesundheit, B., Zilberman, I., Miron, S.,

    Yoffe, L., Lvovich, A., Salvin, S. & Or, R. (2006)

    Allogeneic stem cell transplantation for severe

    acquired aplastic anaemia using a fludarabine-

    based preparative regimen. British Journal of

    Haematology, 133, 649654.

    Risitano, A.M., Selleri, C., Serio, B., Torelli, G.F.,

    Kulagin, A., Maury, S., Halter, J., Gupta, V.,

    Bacigalupo, A., Socie, G., Tichelli, A.,

    Schrezenmeier, H., Marsh, J., Passweg, J. &

    Rotoli, B. (2010) Alemtuzumab is safe and

    effective as immunosuppressive treatment for

    aplastic anaemia and single-lineage marrow

    failure: a pilot study and a survey from the

    EBMT WPSAA. British Journal of Haematology,

    148, 791796.

    Rosenberg, P.S., Alter, B.P., Bolyard, A.A., Bonilla,

    M.A., Boxer, L.A., Cham, B., Fier, C., Freedman,

    M., Kannourakis, G., Kinsey, S., Schwinzer, B.,

    Zeidler, C., Welte, K. & Dale, D.C. (2006) The

    incidence of leukemia and mortality from sepsis

    in patients with severe congenital neutropenia

    receiving long-term G-CSF therapy. Blood, 107,

    46284635.

    Roth, A. & Duhrsen, U. (2011) Treatment of Par-

    oxysmal Nocturnal Hemoglobinuria in the Era

    of Eculizumab. European Journal of Haematolo-

    gy, 87, 473579.

    Samarasinghe, S., Steward, C., Hiwarkar, P., Saif,

    M.A., Hough, R., Webb, D., Norton, A., Law-

    son, S., Qureshi, A., Connor, P., Carey, P., Skin-

    ner, R., Vora, A., Pelidis, M., Gibson, B.,

    Stewart, G., Keogh, S., Goulden, N., Bonney, D.,

    Stubbs, M., Amrolia, P., Rao, K., Meyer, S.,

    Wynn, R. & Veys, P. (2012) Excellent Outcome

    of Matched Unrelated Donor Transplantation in

    Paediatric Aplastic Anaemia following Failure

    with Immunosuppressive Therapy: a United

    Kingdom Retrospective Multicentre Experience.

    British Journal of Haematology, (in press).

    Sanders, J.E., Woolfrey, A.E., Carpenter, P.A.,

    Storer, B.E., Hoffmeister, P.A., Deeg, H.J., Flow-

    ers, M.E. & Storb, R.F. (2011) Late effects

    38 2012 Blackwell Publishing LtdBritish Journal of Haematology, 2012, 157, 2640

    Review

  • among pediatric patients followed for nearly 4

    decades after transplantation for severe aplastic

    anemia. Blood, 118, 14211428.

    Saracco, P., Quarello, P., Iori, A.P., Zecca, M.,

    Longoni, D., Svahn, J., Varotto, S., Del Vecchio,

    G.C., Dufour, C., Ramenghi, U., Bacigalupo, A.

    & Locasciulli, A. (2008) Cyclosporin A response

    and dependence in children with acquired aplas-

    tic anaemia: a multicentre retrospective study

    with long-term observation follow-up. British

    Journal Haematology, 140, 197205.

    Scheinberg, P., Nunez, O., Wu, C. & Young, N.S.

    (2006a) Treatment of severe aplastic anaemia

    with combined immunosuppression: anti-thy-

    mocyte globulin, ciclosporin and mycophenolate

    mofetil. British Journal Haematology, 133, 606

    611.

    Scheinberg, P., Nunez, O. & Young, N.S. (2006b)

    Retreatment with rabbit anti-thymocyte globulin

    and ciclosporin for patients with relapsed or

    refractory severe aplastic anaemia. British Journal

    Haematology, 133, 622627.

    Scheinberg, P., Wu, C.O., Nunez, O. & Young, N.

    S. (2008) Long-term outcome of pediatric

    patients with severe aplastic anemia treated with

    antithymocyte globulin and cyclosporine. Jour-

    nal of Pediatrics, 153, 814819.

    Scheinberg, P., Wu, C.O., Nunez, O., Boss, C.,

    Sloand, E.M. & Young, N.S. (2009) Treat-

    ment of severe aplastic anemia with a combi-

    nation of horse antithymocyte globulin and

    cyclosporine, with or without sirolimus: a

    prospective randomized study. Haematologica,

    94, 348354.

    Scheinberg, P., Nunez, O., Scheinberg, P. & Young,

    N. (2011a) Cyclosporin Taper does not Prevent

    Relapse in Severe Aplastic Anaemia. Blood

    (Annual ASH Meeting Abstracts), 118, 2406.

    Scheinberg, P., Nunez, O., Weinstein, B., Biancot-

    to, A., Wu, C.O. & Young, N.S. (2011b) Horse

    versus rabbit antithymocyte globulin in acquired

    aplastic anemia. New England Journal of Medi-

    cine, 365, 430438.

    Schrezenmeier, H., Passweg, J.R., Marsh, J.C., Ba-

    cigalupo, A., Bredeson, C.N., Bullorsky, E., Ca-

    mitta, B.M., Champlin, R.E., Gale, R.P.,

    Fuhrer, M., Klein, J.P., Locasciulli, A., Oneto,

    R., Schattenberg, A.V., Socie, G. & Eapen, M.

    (2007) Worse outcome and more chronic

    GVHD with peripheral blood progenitor cells

    than bone marrow in HLA-matched sibling

    donor transplants for young patients with

    severe acquired aplastic anemia. Blood, 110,

    13971400.

    Shaw, B.E., Apperley, J.F., Russell, N.H., Craddock,

    C., Liakopoulou, E., Potter, M.N., Wynn, R.,

    Gibson, B., Pearce, R.M., Kirkland, K., Lee, J.,

    Madrigal, J.A., Cook, G. & Byrne, J.L. (2011)

    Unrelated donor peripheral blood stem cell

    transplants incorporating pre-transplant in-vivo

    Alemtuzumab are not associated with any

    increased risk of significant acute or chronic

    graft-versus-host disease. British Journal Haema-

    tology, 153, 244252.

    Shimamura, A. & Alter, B.P. (2010) Pathophysi-

    ology and management of inherited bone mar-

    row failure syndromes. Blood Reviews, 24, 101

    122.

    Skinner, R., Cant, A., Davies, G., Finn, A. & Foot,

    A. (2002)Immunisation of the Immunocompro-

    mised child. Best Practice Statement. Royal Col-

    lege of Paediatrics and Child Health, London.

    Smith, O.P., Hann, I.M., Chessells, J.M., Reeves, B.

    R. & Milla, P. (1996) Haematological abnormal-

    ities in ShwachmanDiamond syndrome. British

    Journal Haematology, 94, 279284.

    Socie, G., Henry-Amar, M., Bacigalupo, A., Hows,

    J., Tichelli, A., Ljungman, P., McCann, S.R.,

    Frickhofen, N., Vant Veer-Korthof, E. & Gluck-

    man, E. (1993) Malignant tumors occurring

    after treatment of aplastic anemia. European

    Bone Marrow Transplantation-Severe Aplastic

    Anaemia Working Party. New England Journal

    of Medicine, 329, 11521157.

    Socie, G., Mary, J.Y., Schrezenmeier, H., Marsh, J.,

    Bacigalupo, A., Locasciulli, A., Fuehrer, M., Bek-

    assy, A., Tichelli, A. & Passweg, J. (2007) Granu-

    locyte-stimulating factor and severe aplastic

    anemia: a survey by the European Group for

    Blood and Marrow Transplantation (EBMT).

    Blood, 109, 27942796.

    Stern, M., Passweg, J.R., Locasciulli, A., Socie,

    G., Schrezenmeier, H., Bekassy, A.N., Fuehrer,

    M., Hows, J., Korthof, E.T., McCann, S., Tich-

    elli, A., Zoumbos, N.C., Marsh, J.C., Baciga-

    lupo, A. & Gratwohl, A. (2006) Influence of

    donor/recipient sex matching on outcome of

    allogeneic hematopoietic stem cell transplanta-

    tion for aplastic anemia. Transplantation, 82,

    218226.

    Storb, R., Prentice, R.L., Sullivan, K.M., Shulman,

    H.M., Deeg, H.J., Doney, K.C., Buckner, C.D.,

    Clift, R.A., Witherspoon, R.P., Appelbaum, F.A.,

    Sanders, J.E., Stewart, P.S. & Thomas, E.D.

    (1983) Predictive factors in chronic graft-versus-

    host disease in patients with aplastic anemia

    treated by marrow transplantation from HLA-

    identical siblings. Annals of Internal Medicine,

    98, 461466.

    Storb, R., Deeg, V., Farewell, K., Doney, F., Appel-

    baum, F., Beatty, P., Bensinger, W., Buckner, C.

    & Clift, R. (1986) Marrow transplantation for

    severe aplastic anaemia: methotrexate alone

    compared with a combination of methotrexate

    and cyclosporine for prevention of graft-versus-

    host disease. Blood, 68, 119125.

    Takanashi, M., Atsuta, Y., Fujiwara, K., Kodo, H.,

    Kai, S., Sato, H., Kohsaki, M., Azuma, H., Ta-

    naka, H., Ogawa, A., Nakajima, K. & Kato, S.

    (2010) The impact of anti-HLA antibodies on

    unrelated cord blood transplantations. Blood,

    116, 28392846.

    Tarek, N., Kernan, N.A., Prockop, S.E., Scarad-

    avou, A., Small, T.N., OReilly, R.J. & Boulad, F.

    (2011) T-cell-depleted hematopoietic SCT from

    unrelated donors for the treatment of congenital

    amegakaryocytic thrombocytopenia. Bone Mar-

    row Transplantation, 13. doi:10.1038/bmt.2011.

    142

    Tichelli, A., Passweg, J., Nissen, C., Bargetzi, M.,

    Hoffmann, T., Wodnar-Filipowicz, A., Signer,

    E., Speck, B. & Gratwohl, A. (1998) Repeated

    treatment with horse antilymphocyte globulin

    for severe aplastic anaemia. British Journal Hae-

    matology, 100, 393400.

    Tichelli, A., Schrezenmeier, H., Socie, G., Marsh,

    J., Bacigalupo, A., Duhrsen, U., Franzke, A.,

    Hallek, M., Thiel, E., Wilhelm, M., Hochsmann,

    B., Barrois, A., Champion, K. & Passweg, J.R.

    (2011) A randomized controlled study in

    patients with newly diagnosed severe aplastic

    anemia receiving antithymocyte globulin (ATG),

    cyclosporine, with or without G-CSF: a study of

    the SAA Working Party of the European Group

    for Blood and Marrow Transplantation. Blood,

    117, 44344441.

    Viollier, R., Socie, G., Tichelli, A., Bacigalupo, A.,

    Korthof, E.T., Marsh, J., Cornish, J., Ljungman,

    P., Oneto, R., Bekassy, A.N., Fuehrer, M., Mau-

    ry, S., Schrezenmeier, H., van Lint, M.T., Wo-

    jcik, D., Locasciulli, A. & Passweg, J.R. (2008)

    Recent improvement in outcome of unrelated

    donor transplantation for aplastic anemia. Bone

    Marrow Transplantation, 41, 4550.

    Vulliamy, T.J., Marrone, A., Knight, S.W., Walne,

    A., Mason, P.J. & Dokal, I. (2006) Mutations in

    dyskeratosis congenita: their impact on telomere

    length and the diversity of clinical presentation.

    Blood, 107, 26802685.

    Wagner, J.E., Eapen, M., MacMillan, M.L., Harris,

    R.E., Pasquini, R., Boulad, F., Zhang, M.J. &

    Auerbach, A.D. (2007) Unrelated donor bone

    marrow transplantation for the treatment of

    Fanconi anemia. Blood, 109, 22562262.

    Walne, A.J. & Dokal, I. (2009) Advances in the

    understanding of dyskeratosis congenita. British

    Journal Haematology, 145, 164172.

    Walne, A.J., Vulliamy, T., Beswick, R., Kirwan, M. &

    Dokal, I. (2010) Mutations in C16orf57 and nor-

    mal-length telomeres unify a subset of patients

    with dyskeratosis congenita, pokiloderma with

    neutropenia and Rothmund-Thomson syndrome.

    Human Molecular Genetics, 19, 44534461.

    Yagasaki, H., Kojima, S., Yabe, H., Kato, K., Kigas-

    awa, H., Sakamaki, H., Tsuchida, M., Kato, S.,

    Kawase, T., Morishima, Y. & Kodera, Y. (2011)

    Acceptable HLA-mismatching in unrelated

    donor bone marrow transplantation for patients

    with acquired severe aplastic anemia. Blood, 118,

    31863190.

    Yamamoto, H., Kato, D., Uchida, N., Ishiwata, K.,

    Araoka, H., Takagi, S., Nakano, N., Tsuji, M.,

    Asano-Mori, Y., Matsuno, N., Masuoka, K., Izu-

    tsu, K., Wake, A., Yoneyama, A., Makino, S. &

    Taniguchi, S. (2011) Successful sustained

    engraftment after reduced-intensity umbilical

    cord blood transplantation for adult patients

    with severe aplastic anemia. Blood, 117, 3240

    3242.

    Yoshida, N., Yagasaki, H., Hama, A., Takahashi,

    Y., Kosaka, Y., Kobayashi, R., Yabe, H., Kaneko,

    T., Tsuchida, M., Ohara, A., Nakahata, T. &

    Kojima, S. (2011) Predicting response to immu-

    2012 Blackwell Publishing Ltd 39British Journal of Haematology, 2012, 157, 2640

    Review

  • nosuppressive therapy in childhood aplastic ane-

    mia. Haematologica, 96, 771774.

    Yoshimi, A., Kojima, S., Taniguchi, S., Hara,

    J., Matsui, T., Takahashi, Y., Azuma, H.,

    Kato, K., Nagamura-Inoue, T., Kai, S. &

    Kato, S. (2008) Unrelated cord blood trans-

    plantation for severe aplastic anemia. Biology

    of Blood and Marrow Transplantation, 14,

    10571063.

    Young, N.S., Bacigalupo, A. & Marsh, J.C. (2010)

    Aplastic anemia: pathophysiology and treatment.

    Biology of Blood and Marrow Transplantation,

    16, S119S125.

    Zhong, F., Savage, S., Shrekli, M., Giri, N., Jes-

    sop, L., Myers, T., Chen, R., Alter, B. & Art-

    andi, S. (2011) Disruption of telomerase

    trafficking by TCAB1 mutation causes dysker-

    atosis congenita. Genes and Development, 25,

    1116.

    40 2012 Blackwell Publishing LtdBritish Journal of Haematology, 2012, 157, 2640

    Review