throbber
bjh
`
`guideline
`
`Diagnosis and management of chronic graft-versus-host
`disease
`
`Fiona L. Dignan,1,2 Persis Amrolia,3 Andrew Clark,4 Jacqueline Cornish,5 Graham Jackson,6 Prem Mahendra,7
`Julia J. Scarisbrick,8 Peter C. Taylor,9 Bronwen E. Shaw1,10 and Michael N. Potter1 on behalf of the Haemato-oncology Task
`Force of the British Committee for Standards in Haematology and the British Society for Blood and Marrow
`Transplantation
`
`1Section of Haemato-oncology, The Royal Marsden NHS Foundation Trust, London, 2St John’s Institute of Dermatology, St Thomas’
`Hospital, London, 3Department of Bone Marrow Transplantation, Great Ormond Street Hospital, London, 4Bone Marrow Transplant
`Unit, Beatson Oncology Centre, Gartnavel Hospital, Glasgow, 5Department of Haematology, Bristol Royal Hospital for Children, Bris-
`tol, 6Department of Haematology, Freeman Road Hospital, Newcastle, 7Department of Haematology, University Hospital Birmingham,
`Birmingham, 8Department of Dermatology, University Hospital Birmingham, Birmingham, 9Department of Haematology, Rotherham
`General Hospital, Rotherham, and 10Anthony Nolan, London, UK
`
`Summary
`
`A joint working group established by the Haemato-oncology
`subgroup of the British Committee for Standards in Haema-
`tology (BCSH) and the British Society for Bone Marrow
`Transplantation (BSBMT) has reviewed the available litera-
`ture and made recommendations for the diagnosis and man-
`agement of chronic graft-versus-host disease (GvHD). This
`guideline includes recommendations for the diagnosis and
`staging of chronic GvHD as well as primary treatment and
`options for patients with steroid-refractory disease. The goal
`of treatment should be the effective control of GvHD while
`minimizing the risk of toxicity and relapse.
`
`graft-versus-host disease,
`Keywords: Chronic,
`management, diagnosis.
`
`transplant,
`
`Summary of recommendations
`
`1 Chronic graft-versus-host disease (GvHD) and overlap
`syndrome should be diagnosed primarily using clinical
`criteria, supported by biopsy when possible. (1B)
`2 Chronic GvHD should be graded as mild, moderate or
`severe according to National Institutes of Health (NIH)
`consensus criteria (Filipovich et al, 2005). (1A)
`All patients with signs or
`symptoms
`suggestive of
`chronic GvHD in one organ should be assessed for
`involvement of other organs. (1A)
`
`3
`
`Correspondence: BCSH Secretary, British Society for Haematology,
`
`100 White Lion Street, London, N1 9PF, UK.
`
`E-mail: bcsh@b-s-h.org.uk
`
`First published online 26 April 2012
`doi: 10.1111/j.1365-2141.2012.09128.x
`
`b'h
`~ British Journal of Haematology
`
`5
`
`7
`
`8
`
`10
`
`11
`
`12
`
`4 Corticosteroids are recommended in the first line treat-
`ment of chronic GvHD. (1A)
`An initial starting dose of 1 mg/kg prednisolone is rec-
`ommended. (1B)
`in the initial
`6 Calcineurin inhibitors may be helpful
`treatment of GvHD as a steroid-sparer. (2C)
`Extracorporeal photopheresis (ECP) may be considered
`as a second line treatment in skin, oral or liver chronic
`GvHD. (1B)
`ECP schedule should be fortnightly-paired treatments
`for a minimum assessment period of 3 months. (1C)
`9 Mammalian target of rapamycin (mTOR) inhibitors are
`suggested as a second line treatment option in refrac-
`tory chronic GvHD. (2C)
`Pentostatin is suggested as a second line treatment
`option in refractory chronic GvHD. (2B)
`Rituximab is
`suggested as a second line treatment
`option in refractory cutaneous or musculoskeletal
`chronic GvHD. (2B)
`Imatinib is suggested as a second line treatment option
`in refractory pulmonary or sclerodermatous chronic
`GvHD. (2C)
`ECP,
`imatinib and rituximab may be considered as
`third line treatment options in chronic GvHD involving
`other organs. (2C)
`The following agents are suggested as third line treat-
`ment options in refractory chronic GvHD: mycopheno-
`late mofetil, methotrexate, pulsed corticosteroids. (2C)
`There is insufficient evidence, at present, to support
`recommendations
`to use
`the
`following agents
`in
`the management of chronic GvHD: cyclophospha-
`mide, mesenchymal stem cells, thalidomide, retinoids,
`alemtuzumab, infliximab, etanercept, clofazimine, alefa-
`cept, daclizumab,
`basiliximab, hydroxychloroquine,
`thoraco-abdominal irradiation. (1C)
`
`13
`
`14
`
`15
`
`ª 2012 Blackwell Publishing Ltd
`British Journal of Haematology, 2012, 158, 46–61
`
`

`

`16
`
`Azathioprine is not recommended in the management of
`chronic GvHD due to the risk of oral malignancy. (1C)
`
`Introduction
`
`Chronic graft-versus-host disease (cGvHD) remains a major
`complication of allogeneic stem cell transplantation and is
`the leading cause of late non-relapse death (Lee et al, 2002).
`The prevalence varies from 25–80% in long-term survivors
`(Baird & Pavletic, 2006). A clear diagnostic and manage-
`ment strategy for cGvHD has been difficult to achieve due
`to the polymorphic nature of the disorder and the paucity
`of evidence for the majority of
`treatment options. The
`National Institutes of Health (NIH) consensus development
`project has tried to address this issue by developing criteria
`for clinical trials in cGvHD (Filipovich et al, 2005; Couriel
`et al, 2006a; Martin et al, 2006; Pavletic et al, 2006; Schultz
`et al, 2006; Shulman et al, 2006). Similarly, the German-
`Austrian-Swiss working party on bone marrow and blood
`stem cell
`transplantation held a consensus conference to
`define clinical management of cGvHD in 2009 and have
`recently published several papers,
`including a summary of
`first- and second-line management of cGvHD (Wolff et al,
`2010, 2011).
`At present there are no UK guidelines on the diagnosis
`and management of cGvHD. T-cell depletion is used widely
`in the UK and this practice may have an impact on the fre-
`quency and pattern of cGvHD and, therefore, management
`guidelines from other countries may be less applicable in this
`setting. This document attempts to provide a summary of an
`evidence-based approach to the diagnosis, staging and man-
`agement of cGvHD in clinical practice. The diagnosis and
`management of acute GvHD is discussed in a separate docu-
`ment (Dignan et al, 2012a) and the organ-specific manage-
`ment and supportive care of patients with GvHD is also
`discussed in a separate document (Dignan et al, 2012b).
`These guidelines are designed to be used together and to
`complement each other in order to provide an evidence-
`based approach to managing this complex disorder.
`
`Methodology
`
`The production of these guidelines involved the following steps:
`
`• Establishment of a working group comprising experts in
`the field of allogeneic transplantation followed by literature
`review to 17th June 2011 including Medline,
`internet
`searches and major conference reports.
`• Development of key recommendations based on random-
`ized, controlled trial evidence. Due to the paucity of
`randomized studies some recommendations are based on
`literature review and a consensus of expert opinion.
`• The GRADE nomenclature was used to evaluate levels of
`evidence and to assess the strength of recommendations.
`
`ª 2012 Blackwell Publishing Ltd
`British Journal of Haematology, 2012, 158, 46–61
`
`Guideline
`
`The GRADE criteria are specified in the British Committee
`for Standards in Haematology (BCSH) guideline pack and
`the GRADE working group website. See Appendix I. Fur-
`ther information is available from the following websites:
`
`o http://www.bcshguidelines.com/4_HAEMATOLOGY_
`GUIDELINES.html
`o http://www.gradeworkinggroup.org/index.htm
`
`• Review by the BCSH committees, British Society of Blood
`and Marrow Transplantation (BSBMT) executive commit-
`tee, the UK Photopheresis Society and the UK Paediatric
`Bone Marrow Transplant Group
`• Review by sounding board of
`the British Society for
`Haematology (BSH) and allogeneic transplant centres in
`the UK.
`
`Diagnosis
`
`Historically, cGvHD was defined as occurring more than
`100 d after transplant. The NIH consensus conference pro-
`posed two subcategories for cGvHD, classic and overlap syn-
`drome, based on clinical features rather than time of onset.
`This proposal recognized that classical features of cGvHD
`could occur within 100 d of transplant and that features of
`acute and cGvHD could occur together (Filipovich et al,
`2005). Futhermore, there is now evidence that this classifica-
`tion has clinical validity (Jagasia et al, 2009).
`The consensus conference also identified ‘diagnostic’ and
`‘distinctive’ features of cGvHD. Diagnostic signs are clinical
`features that establish the diagnosis of cGvHD without the
`need for further investigations. Diagnostic manifestations
`include poikiloderma and lichen planus-like features of the
`skin,
`lichen planus in the mouth or genitals, fasciitis and
`joint contractures. Distinctive signs are clinical features not
`associated with acute GvHD but which would be insufficient
`to make the diagnosis of cGvHD unless supported by posi-
`tive biopsy or
`laboratory findings. Distinctive findings
`include skin depigmentation, nail dystrophy, alopecia, xero-
`stomia, mucoceles, ulceration of the mouth, keratoconjuncti-
`vitis
`sicca and myositis. A full
`list of diagnostic and
`distinctive findings is detailed in the first report of the NIH
`consensus conference (Filipovich et al, 2005). Additional
`investigations are helpful
`in confirming the diagnosis of
`cGvHD in patients with distinctive features and excluding
`other conditions, e.g. infection or drug toxicities. The role of
`additional
`investigations is discussed in the organ-specific
`management document of
`these guidelines (Dignan et al,
`2012b).
`The new diagnostic definitions were designed for use in
`clinical trials and have yet to be fully validated in clinical
`practice. A recent report from a German, Austrian and Swiss
`consensus conference reported a high rate of acceptance of
`the new cGvHD subcategories and diagnostic classification
`(Greinix et al, 2011).
`
`47
`
`

`

`Guideline
`
`Recommendation
`
`• Chronic GvHD and overlap syndrome should be diag-
`nosed primarily using clinical criteria, supported by
`biopsy when possible (1B).
`
`Grading
`
`Chronic GvHD was originally staged as limited or extensive dis-
`ease based on the observations in 20 patients in a retrospective
`review (Shulman et al, 1980). The NIH consensus development
`project on criteria for clinical trials in cGvHD has reviewed
`staging of cGvHD (Filipovich et al, 2005). This document pro-
`posed a new clinical scoring system on a four point scale (0–3)
`with 0 representing no involvement, 1 mild involvement (no
`significant impairment of daily living), 2 moderate involvement
`(significant impairment of daily living) and 3 representing
`severe impairment (major disability). Chronic GvHD may then
`be classified as mild, moderate or severe. Patients with involve-
`ment of one or two organs with a score of 1 and no pulmonary
`GvHD are classified as having mild cGvHD. Moderate cGvHD
`is defined as involvement of three organs with a score of 1, at
`least one organ with a score of 2 or pulmonary GvHD with a
`score of 1. Patients who have major disability resulting in a
`score of 3 in any organ or site or patients who have pulmonary
`GvHD scoring 2 or 3 would be classified as having severe
`cGvHD. This classification is discussed in detail in Filipovich
`et al (2005) and has been reviewed by Devergie (2008). It is rec-
`ommended that all patients are scored using the NIH criteria
`(Filipovich et al, 2005) at 3 months following transplant. In
`patients diagnosed with GvHD, restaging using NIH criteria is
`recommended every 3 months.
`
`Prognostic factors
`
`The John Hopkins group showed in multivariate analysis
`that extensive (>50%) skin involvement, a platelet count of
`<100 9 109/l and progressive onset from acute GvHD were
`associated with poor prognosis (Akpek et al, 2001a). More
`recently, Arora et al (2011) reported a cGvHD risk score.
`Ten variables were identified as being significant in terms of
`overall survival and non-relapse mortality: age, prior acute
`GvHD, time from transplantation to cGvHD, donor type,
`disease status at transplantation, GvHD prophylaxis, gender
`mismatch,
`serum bilirubin, Karnofsky score and platelet
`count (Arora et al, 2011).
`
`Recommendation
`
`• Chronic GvHD should be graded as mild, moderate or
`severe according to NIH consensus criteria (Filipovich
`et al, 2005) (1A).
`• All patients with signs or
`suggestive of
`symptoms
`chronic GvHD in one organ should be assessed for
`involvement of other organs (1A).
`
`48
`
`Principles of cGvHD treatment
`
`A multi-disciplinary approach is mandatory. Patients may
`require joint care with specialist teams including the derma-
`tology, ophthalmology, gastroenterology, gynaecology and
`rheumatology teams as well as intensive input from physio-
`therapists and occupational
`therapists. Topical
`treatments
`and supportive agents also have an important role in effec-
`tive management of cGvHD and may be sufficient in those
`patients with mild disease. Detailed organ-specific manage-
`ment including diagnosis, topical treatment and supportive
`care are discussed in a separate document entitled ‘Organ
`specific management and supportive care in GvHD’ (Dignan
`et al, 2012b).
`
`First line systemic treatment for cGvHD
`
`Corticosteroids
`
`The NIH consensus conference recommended systemic treat-
`ment for moderate or severe GvHD (Filipovich et al, 2005).
`Corticosteroids have been used as first
`line treatment
`in
`cGvHD since the 1980s. Their effect is likely to be due to
`lympholytic effects and anti-inflammatory properties (Deeg,
`2007). The standard dose used has been 1 mg/kg in studies
`of steroids alone or in combination with other agents (Sulli-
`van et al, 1988a; Koc et al, 2002). There are no randomized
`studies comparing this dose to higher or lower steroid doses.
`Topical steroids may be used in conjunction with systemic
`steroids and may allow dose reduction in those patients with
`GvHD limited to the skin.
`At present, there is no consistent tapering protocol for ste-
`roid reduction in the UK. The Seattle group have reported
`on an alternate day dosing regimen for tapering steroids.
`This regimen involved using a daily dose of 1 mg/kg for two
`weeks and subsequently tapering to 1 mg/kg on alternate
`days over 4 weeks if cGvHD is stable or improving. The
`initial report (Sullivan et al, 1988a) used this schedule in com-
`bination with ciclosporin. In a recent review, Lee & Flowers
`suggested a similar initial schedule of 1 mg/kg for 2 weeks and
`then reducing the dose by 25% each week, aiming for a dose of
`1 mg/kg on alternate days after 6–8 weeks. In severe GvHD,
`this dose may be maintained for 2–3 months and then tapered
`by 10–20% per month for a total duration of 9 months. An
`alternative regimen is to miss out the period of stable dosing of
`2–3 months and to taper the dose by 10–20% per month until
`a dose of 0·5 mg/kg is reached. A slower steroid taper is
`advised thereafter depending on clinical response (reviewed by
`Lee & Flowers, 2008). Although there are no randomized
`studies comparing an alternate day approach to daily adminis-
`tration of corticosteroids in this setting, it is likely from studies
`undertaken in other patient groups that this approach may
`reduce side effects while maintaining efficacy (Dumler et al,
`1982; Jabs et al, 1996).
`
`ª 2012 Blackwell Publishing Ltd
`British Journal of Haematology, 2012, 158, 46–61
`
`

`

`In patients who are receiving other immunosuppressive
`agents it
`is recommended that steroids are tapered first.
`Other immunosuppressive agents can be tapered one at a
`time over a 3–9 month period with dose reductions every 2–
`4 weeks depending on clinical response (Lee & Flowers,
`2008). The median duration of immunosuppressive therapy
`is 2–3 years (Lee & Flowers, 2008).
`
`Calcineurin inhibitors
`
`Ciclosporin is commonly used in the prophylaxis of GvHD.
`Ciclosporin binds to cyclophilin and prevents generation of
`nuclear factor of activated T cells (NF-AT), which is a
`nuclear factor for initiating gene transcription for lympho-
`kines including interleukin 2 and interferon gamma. This
`action leads
`to suppression of cytokine production and
`subsequent inhibition of T-cell activation (reviewed in Grei-
`nix, 2008). Early reports suggested a possible benefit of ci-
`closporin in the primary treatment of cGvHD (Sullivan
`et al, 1988a). One randomized trial has been performed
`comparing the use of ciclosporin and daily 1 mg/kg pred-
`nisolone to prednisolone alone in the initial management
`of cGvHD. This study included 287 evaluable patients who
`
`Table I. Summary of the major toxicities of cGvHD treatments.
`
`Guideline
`
`had platelet counts >100 9 109/l at the start of treatment.
`The cumulative incidence of transplantation-related mortal-
`ity at 5 years was 17% in the combination arm compared
`to 13% in those patients who received prednisolone alone.
`There was no difference in efficacy as assessed by the need
`for secondary therapy at 5 years (11% vs. 17%) or the
`median interval to discontinuation of
`immunosuppression
`(1·6 vs. 2·2 years). A combination regimen of ciclosporin
`and prednisolone may have a steroid-sparing effect and
`reduce the incidence of
`steroid-associated complications:
`22% of patients in the prednisolone arm developed avascu-
`lar necrosis compared to 13% in the combination arm
`(Koc et al, 2002). These results may not be applicable to
`all types of transplant as this study group had received my-
`eloablative conditioning regimens and had all received bone
`marrow.
`There are limited data on the role of calcineurin inhibitors
`in the treatment of patients with refractory cGvHD. A pro-
`spective study of 17 patients with refractory disease reported
`a response to tacrolimus in six patients (Tzakis et al, 1991).
`In a larger Phase 2 study including 26 evaluable patients with
`cGvHD, a response to tacrolimus was observed in 12 patients
`(Kanamaru et al, 1995). In a single arm, open-label Phase 2
`
`Treatment
`
`Corticosteroids
`
`Calcineurin Inhibitors
`Mycophenolate mofetil
`
`Sirolimus
`
`Thalidomide
`Azathioprine
`Pentostatin
`Methotrexate
`
`Major toxicities
`
`Infection, hypertension, poor glycaemic control, mood swings,
`osteoporosis, weight gain, growth impairment
`Infection, renal impairment, thrombotic microangiopathy, hypertension
`Infection, deranged liver function tests, gastrointestinal disturbance,
`haematotoxicity, relapse
`Thrombotic microangiopathy, hyperlipidaemia, haematotoxicity
`
`Teratogenecity, peripheral neuropathy, constipation, thrombosis, fatigue
`Oral malignancies, pancytopenia
`Infection, pancytopenia
`Deranged liver function tests, cytopenias
`
`Hydroxychloroquine
`Clofazimine
`
`Gastrointestinal, ocular toxicity, rashes
`Skin pigmentation, gastrointestinal, methaemoglobinaemia
`
`Cyclophosphamide
`Extracorporeal photopheresis
`
`Alefacept
`Imatinib
`Rituximab
`Alemtuzumab
`
`Infliximab
`Etanercept
`
`Haematological, infection, urothelial toxicity
`Patients with poor vascular access require indwelling catheter,
`vaso-vagal episodes
`Infection
`Dyspnoea, fluid retention, pancytopenia, deranged liver function
`Infusional reactions/infection, progressive multifocal leucoencepthalopathy
`Infusional reactions/infections especially opportunistic e.g. cytomegalovirus
`
`Infusional reactions/ infection
`Infection
`
`Basiliximab
`Thoraco-abdominal irradiation
`Retinoids
`
`Infection/infusional reactions
`Haematotoxicty
`Teratogenicity, hyperlipidaemia, deranged liver function
`
`ª 2012 Blackwell Publishing Ltd
`British Journal of Haematology, 2012, 158, 46–61
`
`Reference
`
`Joint Formulary
`Committee (2011)
`Koc et al (2002)
`Martin et al (2009)
`Onishi et al (2010)
`Jurado et al (2007)
`Johnston et al (2005)
`Koc et al (2000)
`Curtis et al (2005)
`Pidala et al (2010)
`Inagaki et al (2008)
`Huang et al (2005)
`Gilman et al (2000)
`Lee et al (1997)
`Moreira et al (1998)
`Mayer et al (2005)
`Scarisbrick (2009)
`
`Shapira et al (2009)
`Stadler et al (2009)
`Kharfan-Dabaja et al (2009)
`Park et al (2009)
`Peleg et al (2007)
`Sleight et al (2007)
`Chiang et al (2002)
`Busca et al (2007)
`Willenbacher et al (2001)
`Robin et al (2005)
`Marcellus et al (1999)
`
`49
`
`

`

`Guideline
`
`study, 8/39 patients achieved a benefit of switching from
`ciclosporin to tacrolimus for refractory cGvHD (Carnevale-
`Schianca et al, 2000). Regular monitoring of
`levels
`is
`required when using calcineurin inhibitors to avoid toxicity.
`
`Recommendations
`
`• Corticosteroids are recommended in the first line treat-
`ment of chronic GvHD (1A).
`• An initial starting dose of 1 mg/kg prednisolone is rec-
`ommended (1B).
`• Calcineurin inhibitors may be helpful in the initial treat-
`ment of GvHD as a steroid sparer (2C).
`
`Second-line systemic treatment in cGvHD
`
`A number of agents have been used as second- and third-line
`therapy for cGvHD. The role of these therapies in the sys-
`temic management of cGvHD will be discussed in the follow-
`ing sections. The authors acknowledge that it is difficult to
`conduct randomized controlled trials in cGvHD and that the
`management suggestions made in this guideline are based on
`the interpretation of limited data available at time of review
`and widespread expert opinion. Many of these agents have
`significant toxicities, which are summarized in Table I.
`These agents may be helpful in the management of ste-
`roid-refractory disease or as steroid-sparing agents in patients
`who are steroid-dependent or intolerant to steroids. The def-
`inition of steroid-refractory disease varies between studies
`but may include progression on prednisolone at 1 mg/kg per
`day for two weeks, stable disease on  0·5 mg/kg per day of
`prednisolone for 4–8 weeks and inability to taper predniso-
`lone below 0·5 mg/kg per day (Martin et al, 2006; Wolff
`et al, 2011).
`Ideally, patients with steroid-refractory cGVHD should be
`entered in to clinical trials. Where this is not possible, the
`choice of agent is likely to depend on the toxicity profile,
`organ involvement, patient preference and availability. Some
`agents may be used in combination or sequentially depend-
`ing on clinical judgement. As there are no established predic-
`tors of response, second line therapy should, where possible
`avoid the changing of more than one agent at a time, with
`assessment at 8–12 weeks. Where there is progression within
`a 4-week period alternative therapies can be considered,
`although patients with sclerotic skin disease are likely to take
`longer to demonstrate response.
`
`Extracorporeal photopheresis
`
`Extracorporeal photopheresis (ECP) has been widely used as
`a second line therapy for the treatment of mucocutaneous
`cGvHD, with consistently high complete response rates of up
`to 80% with cutaneous manifestations, and significant
`improvement in sclerodermatous skin involvement (Couriel
`
`50
`
`et al, 2006b; Dignan et al, 2011). Flowers et al (2008) pub-
`lished the first multicentre, randomized controlled, prospec-
`tive Phase II trial of ECP in the treatment of patients with
`cGHVD. This study included patients who were steroid-
`dependent, steroid-refractory and those who were intolerant
`of steroids. Ninety-five patients were randomized to receive
`either ECP and standard therapy (corticosteroids plus other
`immunosuppressive agents including ciclosporin, tacrolimus
`or mycophenolate mofetil) or standard therapy alone. The
`study used percentage improvement in total skin scores after
`12 weeks of ECP treatment as the primary endpoint. The
`percentage reduction in total skin score from baseline was
`greater in the ECP arm compared to the non-ECP arm but
`this did not achieve statistical significance (P = 0·48). The
`proportion of patients who had at least a 50% reduction in
`steroid dose and at least a 25% decrease in total skin score
`was 8·3% in the ECP arm at week 12 and 0% in the control
`arm (P = 0·04) (Flowers et al, 2008). A major limitation of
`this study is that the study arm assignment was known to
`physicians who were controlling the prednisolone dose. This
`study has several other limitations due to the methodological
`challenges of conducting clinical
`trials
`in patients with
`cGvHD. These include the short duration of treatment, only
`using skin as the primary endpoint to assess response, the
`limited time allowed for reduction in steroids (6 weeks) and
`the large variation in immunosuppressive regimens used.
`The response reported in patients with visceral GvHD, e.g.
`liver, is more variable. Greinix et al (2006) reported a complete
`response rate of 68% for liver cGvHD (17/25 patients). Similarly,
`Couriel et al (2006b) reported a partial response rate of 15/21
`(71%) for liver cGvHD. These results have not been reflected
`in all studies (Seaton et al, 2003; Foss et al, 2005). Lung and
`gut involvement have demonstrated less consistent responses
`(Greinix et al, 1998; Child et al, 1999; Couriel et al, 2006b).
`There are mixed reports of the benefits of earlier (<12 months)
`versus delayed treatment with ECP (Child et al, 1999; Apisarn-
`thanarax et al, 2003; Messina et al, 2003; Foss et al, 2005).
`Response to ECP has been associated with increased survival and
`reduction in the use of corticosteroids (Foss et al, 2005).
`A UK consensus statement on the use of ECP in cGvHD
`suggested that patients with cutaneous, mucous membrane
`and hepatic manifestations of GvHD should be given priority
`for treatment as it is particularly efficacious in this setting
`(Scarisbrick et al, 2008). This consensus group recommended
`a treatment schedule of two ECP treatments on two consecu-
`tive days every 2 weeks with less frequent monthly treatment
`in those who respond (Scarisbrick et al, 2008). No benefit
`has
`been demonstrated for more
`regular
`treatments
`(reviewed in Scarisbrick et al, 2008; Foss et al, 2005). The
`median number of ECP cycles in a recent UK study was 15
`(30 treatments) and the median duration of treatment was
`330 d (Dignan et al, 2011).
`Although a number of biomarkers have been reported to
`predict response to ECP, none have been clinically validated.
`
`ª 2012 Blackwell Publishing Ltd
`British Journal of Haematology, 2012, 158, 46–61
`
`

`

`
`The proportions of immature CD19+ CD21
`B lymphocytes
`may predict the response to ECP in cGvHD (Kuzmina &
`Greinix, 2009). There is some evidence that the ECP-treated
`cell dose may correlate with ECP effect (Perseghin et al,
`2007; Whittle et al, 2011). Abnormalities of B-cell homeosta-
`sis with increased B-cell activating factor (BAFF) have been
`noted in cGvHD, with evidence of normalization in ECP
`responders (Whittle & Taylor, 2011). The use of ECP is asso-
`ciated with improved outcomes
`in classic and overlap
`cGvHD (Jagasia et al, 2009).
`ECP requires venous access, which is not always possible
`using peripheral veins. Central
`lines may be required with
`the attendant complications of infection and blockage (Scar-
`isbrick, 2009). Systemic infections requiring either oral or
`intravenous antibiotics are at least halved in patients receiv-
`ing ECP for 12 months (personal observations, PCT). Other
`side effects are minimal and include vasovagal episodes and
`fatigue.
`ECP facilities should be quality assured. ECP requires
`well-maintained and validated machines, specifically trained
`staff, as well as skilful overall management to ensure effective
`use of resources, a safe patient care pathway and achievement
`of desirable outcomes. Clinical standards for such a service
`should be identified and adhered to through a quality assur-
`ance programme.
`It is suggested that ECP may be helpful as a second line
`treatment in steroid-refractory cGvHD involving the skin,
`mouth or liver. There is less data supporting the use of ECP
`in cGvHD involving other organs but it may have a role as a
`third line option. The authors acknowledge that this special-
`ized form of therapy may not currently be uniformly avail-
`able in the UK.
`
`Mammalian target of rapamycin (mTOR) inhibitors
`
`Sirolimus is a macrolide antibiotic that exerts its immuno-
`suppressive effect by inhibiting cytokine-driven signalling
`pathways of the T cell via mTOR blockade and specifically
`inhibiting the progression of cells from the G1 phase to the
`S phase. Sirolimus has been used in combination with other
`immunosuppressive agents in the management of refractory
`cGvHD. A Phase 2 study reported an overall response rate of
`63% [6/35 complete response, 16/35 partial response (defined
`as > 50% improvement
`in clinical manifestations)]
`in
`patients with steroid-resistant cGvHD when sirolimus was
`used in combination with tacrolimus and corticosteroids
`(Couriel et al, 2005). Four patients developed thrombotic
`microangiopathy and 77% had infectious complications. The
`median survival was 15 months. A similar Phase 2 study
`enrolled 19 patients (Johnston et al, 2005). Only 16 were
`evaluable for response because three had discontinued the
`sirolimus after less than one month’s treatment due to toxic-
`ity. A partial response was defined as any improvement in
`symptoms. This study reported an initial clinical response in
`15/16 evaluable patients. Adverse events
`included renal
`
`ª 2012 Blackwell Publishing Ltd
`British Journal of Haematology, 2012, 158, 46–61
`
`Guideline
`
`impairment in four patients and haemolytic-uraemic syn-
`drome in two patients and relapse in one patient, leading to
`discontinuation of sirolimus in six patients (Johnston et al,
`2005). Sirolimus levels were not checked in all patients. Sirol-
`imus was used in a retrospective study in patients with
`refractory disease in combination with either calcineurin
`inhibitors, MMF or prednisolone in 47 patients (Jurado et al,
`2007). Clinical
`responses were seen in 81% (38/47) of
`patients. Eighteen had a complete response and 20 had a
`partial response (defined as improvement in 1 organ without
`evidence of progression in another). Survival was 54·7% at
`3 years. Four patients developed thrombotic microangiop-
`athy (Jurado et al, 2007).
`In an
`Everolimus
`is an alternative mTOR inhibitor.
`abstract of a single centre retrospective analysis including 29
`patients with steroid-resistant disease, responses were seen in
`69% of patients (two complete responses) treated with ever-
`olimus without an additional calcineurin inhibitor. No
`patients developed thrombotic microangiopathy (Klink et al,
`2008).
`mTOR inhibitors are suggested for use as a second line
`treatment option in refractory cGvHD. They should be used
`with caution in combination with calcineurin inhibitors in
`view of the risk of thrombotic microangiopathy and trough
`levels should be monitored. Patients should also be moni-
`tored for hyperlipidaemia. Care should be taken to avoid
`toxicity due to interactions with other medications, particu-
`larly azoles (Wolff et al, 2011).
`
`Pentostatin
`
`Pentostatin is a nucleoside analogue and is a potent inhibitor
`of adenosine deaminase. Cell death occurs as a result of
`accumulation of 2-deoxyadenosine 5-triphosphate particu-
`larly in T cells and Natural Killer cells. The drug also causes
`prolonged lymphopenia although it is not significantly myel-
`osuppressive (Margolis & Vogelsang, 2000). A Phase 2 study
`administered pentostatin fortnightly
`for 12 doses
`and
`reported a response rate of 55% in 58 heavily pre-treated
`patients with refractory cGvHD (Jacobsohn et al, 2007). A
`clearly defined scoring system was used to assess patients at
`3-monthly intervals and 31 patients were considered to have
`a major response according to the study criteria. Survival
`was 70% at 2 years and 11 infectious episodes were possibly
`related to pentostatin. The same investigators reported a 53%
`response rate in a Phase 2 study of 51 children with refrac-
`tory cGvHD (Jacobsohn et al, 2009). Similarly, a clearly
`defined scoring system was used to assess response at 3-
`monthly intervals and seven patients had a complete res-
`ponse and 20 had a partial response. Overall survival at one
`year was 84%. There were 27 episodes of infection occurring
`in 15 patients. A dose of 4 mg/m2 was administered intrave-
`nously every 2 weeks in these reports and the main toxicities
`were infection and haematotoxicity. In a retrospective series,
`10/18 patients with refractory cGvHD obtained a complete
`
`51
`
`

`

`Guideline
`
`or partial response to pentostatin treatment (Pidala et al,
`2010). As infections are frequent, it has been recommended
`that pentostatin is not used in the context of acute infection
`or in pulmonary cGvHD (Wolff et al, 2011).
`
`Rituximab
`
`Rituximab is an anti-CD20 monoclonal antibody used widely
`in the management of B-cell malignancies. Ratanatharathorn
`et al (2000) reported the first case of patient with cGvHD
`and immune thrombocytopenia having a complete response
`to four doses of 375 mg/m2 of rituximab. Cutler et al (2006)
`reported the results of a Phase 1/2 study that included 21
`patients with steroid-refractory cGvHD treated with 375 mg/
`m2 weekly of
`rituximab. A response rate of 70% was
`observed although many responses were partial and limited
`to cutaneous and musculoskeletal disease. In addition, many
`patients had relatively mild GvHD. Responses were durable
`for one year (Cutler et al, 2006). A further Phase 2 study of
`37 patients reported 8 complete and 24 partial responses
`with higher responses in skin, oral cavity and musculoskeletal
`systems (Kim et al, 2010). Similar results ha

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket