throbber
Bone Marrow Transplantation (2013) 48, 203–209
`& 2013 Macmillan Publishers Limited All rights reserved 0268-3369/13
`
`www.nature.com/bmt
`
`•
`
`REVIEW
`
`Chronic GVHD: Where are we? Where do we want to be?
`Will immunomodulatory drugs help?
`YPL Linhares1, S Pavletic2 and RP Gale3
`
`Chronic GVHD (cGVHD) is an important problem after allotransplants. Some risk factors for cGVHD are similar to those of acute
`GVHD (aGVHD) but others are distinct indicating sometimes overlapping but unique pathogeneses. Precise incidence and
`prevalence data of cGVHD are lacking because of diverse diagnostic criteria but a 50% risk is a reasonable estimate. Incidence and
`prevalence of cGVHD are probably growing because of increased use of unrelated donors, blood rather than bone marrow (BM)
`grafts, decreased early transplant-related mortality (TRM) and increasing frequency of allotransplants. Pathophysiology of cGVHD is
`complex and poorly understood. Notably, no reliable surrogate end point to predict mechanism(s) of cGVHD has been identified.
`Therapy of cGVHD is unsatisfactory. Corticosteroids are effective but other drugs are controversial and few are rigorously tested in
`randomized trials. Highly variable response rates are reported because of small sample sizes and inconsistencies in eligibility,
`diagnostic and response criteria. We focus on the possible role of immunomodulatory drugs (IMiDs), thalidomide lenalidomide and
`pomalidomide, in preventing and treating cGVHD. The data suggest activity of thalidomide but at doses not clinically practical in
`many instances. There are few data with lenalidomide. Trials of pomalidomide, which has immune activities like thalidomide but
`with fewer adverse effects, are beginning. Because cGVHD is not recently reviewed in Bone Marrow Transplantation, we give a brief
`background and discuss challenges in diagnosing, understanding and treating cGVHD including the recently proposed National
`Institutes of Health consensus criteria for cGVHD.
`
`Bone Marrow Transplantation (2013) 48, 203–209; doi:10.1038/bmt.2012.76; published online 14 May 2012
`
`Keywords: Chronic GVHD; IMiDs; thalidomide; lenalidomide; pomalidomide
`
`INTRODUCTION
`Chronic GVHD (cGVHD) is an important complication of allogeneic
`hematopoietic cell transplantation.1–3 It occurs in about 50% of
`persons surviving 41 year post transplant and causes substantial
`morbidity and mortality. There has been little progress over the
`past 30 years in preventing and/or treating cGVHD. Moreover,
`incidence and prevalence are increasing because of several factors
`including: (1) increased use of blood cell rather than BM grafts; (2)
`increasing use of
`incompletely HLA-matched related and
`unrelated donors;
`(3)
`increased use of donor-lymphocyte
`infusions,
`especially
`in the
`context of
`reduced-intensity
`allotransplants; (4) increased number of transplants done each
`year; and (5)
`increased proportion of
`transplants in older
`persons.4-6 The focus of our review is on the use and potential
`of IMiD-class drugs to prevent and/or treat cGVHD. These drugs
`have unique, complex immune regulatory activities. As a prelude,
`we review some relevant definitional,
`laboratory and clinical
`features of cGVHD.
`
`CHRONIC GVHD
`Reported incidences of cGVHD vary dramatically: 6–80% but 50%
`is a reasonable estimate.7–9 Several important subject-, disease-
`and transplant-related variables correlate with the risk of cGVHD
`including: (1) recipient and/or donor genetic disparity (like related
`or unrelated, degree of HLA matching); (2) graft type (blood, BM or
`umbilical cord blood): (3) graft manipulation (like T-cell depletion);
`
`and (4) whether donor-lymphocyte infusions are given post
`transplant, which are unadjusted for in these reports with diverse
`incidences.10 Some data indicate that male recipients of grafts
`from female donors, especially those who are multiparous, have
`an increased risk of cGVHD. This increased risk from multiparity
`may also apply to female recipients.
`It is unknown whether
`progeny gender of multiparous donors correlates with risk of
`cGVHD.11 Detection of antibodies to Y-chromosome encoded
`antigens in male recipients of grafts from female donors correlates
`with an increased risk of cGVHD.12 Whether this is a cause: effect
`relationship is unknown.
`Some studies report prior acute GVHD (aGVHD) strongly
`correlated with cGVHD.13 A recent multivariate analysis by
`Flowers et al.14 in which National
`Institutes of Health (NIH)
`definitions for aGVHD and cGVHD were used showed grade-3/-4
`aGVHD was a risk factor for cGVHD. However, there were also
`some unique risk factors correlated solely with cGVHD including:
`(1) blood cell grafts and (2) older donor age. Also, use of female
`donors for male recipients had a greater effect on risk of cGVHD
`than on aGVHD. There was little change in hazard ratios with other
`variables after adjusting the hazard ratio of cGVHD for aGVHD.
`This suggests that the association of certain risk factors with
`cGVHD is independent of aGVHD. These data support the notion
`cGVHD is not merely a time-dependent expression of aGVHD.
`aGVHD and cGVHD were previously considered to have distinct
`clinical and temporal features. By convention, the temporal cutoff
`for classifying a clinical syndrome as aGVHD or cGVHD was 100
`
`1Department of Medicine, Division of Hematology/Medical Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; 2GVHD and Autoimmunity Unit,
`Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD, USA and 3Department of Medicine, Section of Haematology, Division of
`Experimental Medicine, Imperial College, London, UK. Correspondence: Dr RP Gale, Department of Medicine, Section of Haematology, Division of Experimental Medicine, Imperial
`College, 11693 San Vicente Boulevard, Suite 335, Los Angeles, CA 90049-5105, USA.
`E-mail: robertpetergale@aol.com
`Received 29 March 2012; accepted 29 March 2012; published online 14 May 2012
`
`

`

`•
`
`204
`
`Immune modulators in chronic GVHD
`YPL Linhares et al
`
`this binary classification is
`transplant. However,
`days post
`obscured when aGVHD occurs after day 100, when there
`is delayed development of hematopoietic chimerism, when
`there are flares after tapering post transplant immune suppression
`or following post transplant infusions of donor lymphocytes. In
`2005, the NIH consensus project proposed two main GVHD
`categories each with two subcategories.10,15 For aGVHD there is
`(1) classic aGVHD occurring within 100 days post transplant and
`(2) persistent, recurrent or late onset aGVHD, a clinical syndrome
`resembling aGVHD but persisting beyond or developing after 100
`days post transplant commonly referred to just as ‘late’ aGVHD.
`For cGVHD there is (1) classic cGVHD subtype (without features
`characteristic of aGVHD) and (2) overlap syndrome with
`synchronous clinical
`features of cGVHD and aGVHD. The old
`‘limited’ and ‘extensive’ cGVHD staging nomenclature is replaced
`with the more informative individual organ severity scoring
`(grades 0–3) and global cGVHD (mild–moderate–severe) stage
`which in addition to cGVHD manifestations also account for
`subjects symptoms and quality of life.10,16 Feasibility of use and
`clinical utility of the NIH consensus cGVHD classification and
`severity scoring is validated in several prospective studies.8,16,17
`Several
`retrospective
`and prospective
`analyses
`identified
`additional prognostic variables which complement
`the NIH
`consensus staging system including low platelet levels, poor per-
`formance score when cGVHD is diagnosed, and gastrointestinal
`tract
`involvement which significantly influence outcome and
`progressive onset
`type of
`cGVHD as adversely affecting
`survival.18,19 These correlations need confirmation.
`cGVHD is a multiorgan alloimmune and autoimmune disorder
`characterized by immune dysregulation,
`immune deficiency,
`impaired end-organ function and decreased survival.20 Although
`these features of cGVHD were noted 430 years ago there has
`been little progress sorting out the precise mechanism(s). Therapy
`of cGVHD, whatever the cause(s), is typically prolonged immune
`suppression which may
`further
`aggravate
`the
`immune
`suppression intrinsic to cGVHD.21
`
`PATHOPHYSIOLOGY
`The pathophysiology of cGVHD is poorly understood (to say the
`least) despite several decades of research. T cells (Th1 and Th2) and
`B cells are important suggesting a global loss of immune tolerance
`including abnormalities in the function of regulatory T cells (Tregs;
`CD4 þ , CD25 þ , FoxP3 þ ) which maintain self-tolerance.22
`Studies in mice indicate that normal Tregs can suppress aGVHD
`and cGVHD and that deficient and/or defective Tregs worsens
`aGVHD and cGVHD.23 There are conflicting data in humans
`concerning the role of Tregs in the development of cGVHD.24 A
`recent study suggests that therapy with low-dose IL-2 can increase
`Tregs and improve severe cGVHD.25
`Several
`studies
`suggest
`that aGVHD is associated with
`predominant Th1-type immune response and cGVHD with a
`predominant Th2-type immune response.26 Th1 cells produce
`IFN-g that mediates cell-mediated immunity whereas Th2 cells
`produce IL-4, IL-5 and IL-13 that mediate humoral immunity. T-cell
`dysregulation results in cytokine abnormalities which may be
`important in cGVHD. High levels of TNF-a, IL-6, TGF-b and IL-1b
`and low levels of INF-g and IL-10 are reported in persons with
`cGVHD.27–31 A recent study by Imanguli et al.31 challenges this
`paradigm of cGVHD as type-II cytokine-mediated disorder. Data
`from this study suggest that activation of the type-I IFN axis is
`important in oral cGVHD.
`Autoimmunity and autoreactive T cells are important in cGVHD.
`Some studies report a functional host thymus is not required to
`induce cGVHD and that quiescent autoreactive T and B cells in
`transplants from non-autoimmune donors can be activated and
`expanded to induce cGVHD.32 In contrast, involvement of thymus-
`dependent T-cell pathways in cGVHD development begins with
`
`injury to the thymus from chemotherapy and/or radiation and/or
`from aGVHD leading to loss of self-tolerance.26,33,34 Some data in
`mice suggest that during aGVHD graft-donor CD4 þ T cells that
`can recognize and react against host tissues develop in the
`thymus in and mediate cGVHD.34
`Some data suggest an important role for B cells in cGVHD.
`Because B cells produce antibodies and can, in some instances,
`expose or present antigen to T cells, it may enhance development
`of cGVHD. Anti-nuclear, -mitochondrial, -parietal, -smooth muscle
`and -parotid autoantibodies are present in some persons with
`cGVHD.35,36 Also, persons with autoantibodies have more cGVHD-
`associated symptoms than persons without autoantibodies.35
`Autoantibodies against platelet-derived growth factor receptor
`may have a role in cGVHD.37 These platelet-derived growth factor
`receptor-a autoantibodies stimulate thyrosine phosphorylation,
`resulting in a cascade of events that may contribute to inflamma-
`tion and fibrosis.
`Some studies report elevated levels of BAFF (B-cell activating
`factor of the TNF family) in persons with cGVHD. BAFF is produced
`by T cells and granulocytes and supports differentiation and
`survival of normal B cells in persons with cGVHD and autoimmune
`diseases.23,38 Fujii and coworkers reported persons with early
`onset cGVHD have elevated levels of sBAFF, sIL-2Ra, sCD13 and
`anti-dsDNA autoantibodies. sBAFF, anti-dsDNA and antinuclear
`antibody are elevated in late onset cGVHD, suggesting B-cell
`activation.39–41 These observations may explain occasional reports
`of benefit of therapy of cGVHD with anti-B-cell antibodies like anti-
`CD20 (rituximab for instance).40
`These T- and B-cell pathways and others are potential targets
`for treating cGVHD. However, it is important to note no immune
`parameter(s)
`is a reliable biomarker of diagnosis,
`severity,
`therapy outcome of cGVHD.42 Consequently,
`prognosis or
`clinical
`trials,
`ideally
`randomized, blinded and placebo
`controlled, are the sole way to know whether a therapy
`intervention in cGVHD is safe and effective.
`
`CLINICAL FEATURES
`lungs,
`liver,
`cGVHD usually targets the skin, eyes, mouth, gut,
`joints and genitourinary system. Typical skin manifestations are
`sclerosis and poikiloderma and lichen-type lesions. There are
`often hyperkeratotic oral plaques. A lung biopsy may show
`bronchiolitis obliterans. These clinical
`features resemble auto-
`immune diseases like progressive systemic sclerosis, systemic
`lupus erythematosis and Sjo¨ gren’s syndrome.10
`cGVHD is categorized in the NIH global scoring system as mild,
`moderate or severe depending on the number of organs involved
`and the severity of the abnormality(ies).10,15 Systemic immune
`suppression is usually advised for persons with moderate or
`severe cGVHD. Systemic therapy is also considered in persons with
`thrombocytopenia (platelets o100  10e9/L) or progression while
`prednisone.43
`receiving
`cGVHD eventuates
`in
`impaired
`performance score, poor quality-of-life and death. 8-10,21,44-46
`
`TREATMENT
`interventions that prevent aGVHD are not
`Pharmacological
`effective in preventing cGVHD. Strategies using anti-thymocyte
`globulins for in-vivo T-cell depletion show promise but no benefit
`on survival.47 The standard initial therapy of cGVHD is prednisone
`with or without a calcineurin inhibitor. However, only about 50%
`of persons have a durable response.22,43 There is no standard next
`therapy. Recommended interventions include about 40 drugs, all
`studied in poorly standardized, phase-2 trials or reported in
`retrospective case analyses, including sirolimus, tacrolimus, myco-
`phenolate, MTX, MoAbs, pentostatin,
`imatinib, extracorporeal
`low-dose IL-2 and many others.25,48-49 Choice
`photopheresis,
`
`Bone Marrow Transplantation (2013) 203 – 209
`
`& 2013 Macmillan Publishers Limited
`
`

`

`Immune modulators in chronic GVHD
`YPL Linhares et al
`
`Thalidomide
`
`Lenalidomide
`
`Pomalidomide
`
`•
`
`205
`
`~
`
`0
`
`NH2
`
`0
`
`o QOYso
`8:$
`-µ=o
`
`NH
`
`O 0
`
`++++
`
`++++
`
`+++
`
`+++
`
`+++
`
`+++++
`
`+++++
`
`++++
`
`++++
`
`++++
`
`+ +
`
`Anti-inflammation
`
`T-cell stimulation
`
`Anti-angiogenesis
`
`++++
`
`+ +
`
`Anti-proliferation
`
`Pro-erythroid
`
`Figure 1. Biological activities of IMiDs.
`
`Survival was 64% with 55 months follow-up and 76% in persons
`failing prior therapy and 48% in those with previously untreated
`high-risk cGVHD. Confidence intervals were not reported and
`there was no comparator cohort. Main adverse effects were
`sedation, neuropathy and constipation.58 Heney et al.61 reported
`responses in five of six persons receiving thalidomide, 100–200 mg/day.
`Response was best in skin cGVHD. Two subjects developed
`neuropathy. Cole et al.62 reported five children with advanced
`cGVHD treated with thalidomide, 12–25 mg/kg/day. There was
`one complete response and four partial responses. Adverse effects
`were minimal and there was no neuropathy. Parker et al.63 treated
`80
`subjects with
`advanced
`cGVHD with
`thalidomide,
`400–1200 mg/day. There were nine complete and seven partial
`responses. Twenty-nine subjects discontinued because of adverse
`effects, including sedation, constipation, neuritis, neutropenia and
`rash. Rovelli et al.64 used thalidomide, 3–12 mg/kg/day in 14
`children with cGVHD. Six complete responses and four partial
`responses were reported. Browne et al.65 treated 37 subjects with
`advanced cGVHD with thalidomide, 200–800 mg/day.
`In all, 1
`subject had a complete and 13, partial responses. Responses were
`more common in children than in adults. Kulkarni et al.66 reported
`data on 59 subjects with advanced cGVHD using thalidomide,
`600–1200 mg/day. Thirteen subjects had a complete and eight, a
`partial response. Two subjects developed poly-neuropathy, two,
`deep vein thromboses and one, thrombocytopenia. There are a
`case series and two case reports not included in Table 1. Mehta
`treated two children with cGVHD. One had a complete and the
`other a PR. Adverse effects were sedation and constipation.67
`Forsyth reported a response to thalidomide, 400 mg/day, in a case
`of bronchiolitis obliterans from cGVHD.68 Staumont-Salle reported
`a response to thalidomide, 100 mg/day, in a subject with lichenoid
`vulvar lesions from cGVHD.69
`Table 2 summarizes data from three randomized trials. Two
`it.70–72
`were attempts to treat cGVHD and one,
`to prevent
`Koc et al.70 reported a placebo-controlled trial of thalidomide,
`200–800 mg/day,
`in adults and 3–12 mg/kg/day in children.
`Thalidomide was discontinued in 23 of 25 subjects in the
`thalidomide cohort and 17 of 26 subjects in the placebo cohort
`because of
`intolerance, mostly neutropenia,
`sedation and
`neuropathy. This high discontinuation rate in the placebo cohort
`underscores the variable natural course of cGVHD and subsequent
`difficulty of performing clinical trials in persons with advanced
`cGVHD.
`It also underscores the need for placebo-controlled
`double-bind studies.
`It is also possible that thalidomide was
`ineffective in this population. Arora et al.71 reported a randomized,
`placebo-controlled trial of
`initial
`treatment of cGVHD with
`thalidomide. There was no benefit of adding thalidomide,
`200–800 mg/day, vs placebo to cyclosporine and prednisone.
`Chao et al.72 reported thalidomide increased the incidence and
`severity of cGVHD when given in a prevention study.
`
`between drugs is based on logistics; cost, failed prior treatments,
`toxicity profile and subject or clinician preference.
`Therapy of cGVHD is a difficult problem to address because of
`the logistical challenges in conducting such trials and the lack of
`standardized criteria for study design. To improve conduct and
`interpretation of clinical
`trials,
`the NIH-Sponsored Consensus
`Development Project published guidelines addressing diagnosis
`and staging, histopathology, biomarkers, assessment of therapy
`response, ancillary therapy and supportive care and the design of
`cGVHD clinical trials.10,50–54
`cGVHD typically is diagnosed within 6 months post transplant
`and lasts 2–5 years. About 85% of survivors can discontinue
`systemic immune suppression by that time. Five-year survival of
`persons with cGVHD is 30–40% for those with high-risk disease
`and for persons failing corticosteroids. There is better 5-year
`survival, about 70–80%,
`in persons with lower risk cGVHD and
`those responding to corticosteroids.9,55,56
`Treatment goals for cGVHD include reversing symptoms and
`signs, preventing recurrence, disability or death. A goal could be
`also correcting associated immune abnormalities. This is, of
`course, challenging, as therapy of cGVHD typically involves
`immune-suppressive drugs that have multiple acute and cumu-
`lative toxicities. The therapy of cGVHD is largely unsatisfactory and
`most persons, especially those who fail corticosteroids, should be
`treated on investigational protocols whenever possible.45
`
`IMID-CLASS DRUGS
`Thalidomide
`Thalidomide is effective in modulating cGVHD in rodents and is
`studied as therapy and prevention of cGVHD in humans.57
`Vogelsang et al.58 reported that
`thalidomide is a safe and
`effective treatment
`for severe cGVHD. Several other studies
`reproduced these findings. Thalidomide is
`the third most
`commonly used drug in phase-2 trials of therapy of cGVHD in
`persons failing corticosteroids.59
`Thalidomide has diverse immune-modulating effects including:
`(1) reduced levels of TNF-a; (2) co-stimulation of T cells to produce
`IL-2 and IFN-g; (3) inhibition of other cytokines like IL-1b,
`IL-6,
`IL-12;
`(4) downregulation of cell surface adhesion molecules
`involved in leukocyte migration; and (5) anti-angiogenesis.48,57,60
`Its biological activities are contrasted with other IMiDs in Figure 1.
`There are seven phase-2 and three phase-3 trials of thalidomide in
`cGVHD. Analysis of the trials is complex for several reasons:
`(1) different definitions of cGVHD; (2) different response criteria;
`(3) inclusion of children in some studies; (4) different doses of
`thalidomide;
`(5) different prior
`therapy (ies);
`(6) different
`objectives
`(for example,
`therapy vs prevention and initial
`therapy vs second-line therapy after corticosteroid failure); and
`(7) thalidomide alone vs combinations and other variables. This
`heterogeneity, not uncommon in cGVHD trials, makes it difficult/
`impossible to draw precise conclusions regarding safety and
`efficacy of thalidomide in cGVHD.
`Table 1 summarizes data from 7 trials in 245 subjects with
`cGVHD receiving thalidomide after failing initial therapy at dosesof
`100–1600 mg/day. These data include children and adults in
`diverse therapy settings and using diverse response criteria. In all,
`46 subjects are reported to have had a complete (19%) and 51 a
`partial response (21%) for an overall response in 97 subjects (40%).
`This result is encouraging but there is the undoubtedly important
`issue like possible of selective reporting of favorable outcomes.
`These trials are reviewed in below.
`Vogelsang et al.58 used thalidomide, 800–1600 mg/day, as initial
`therapy of 21 subjects with high-risk cGVHD and as salvage
`therapy for 23 subjects with cGVHD failing initial therapy. Initial
`dose in children was 3 mg/kg given four times daily. Complete
`response was reported in 14 subjects and partial response in 12.
`
`& 2013 Macmillan Publishers Limited
`
`Bone Marrow Transplantation (2013) 203 – 209
`
`

`

`•
`
`206
`
`Immune modulators in chronic GVHD
`YPL Linhares et al
`
`Table 1. Phase-2 trials of thalidomide in advanced cGVHD
`
`Vogelsang et al.59
`Heney et al.62
`Cole et al.63
`Parker et al.64
`Rovelli et al.65
`Browne et al.66
`Kulkarni et al.67
`
`Abbreviation: cGVHD ¼ chronic GVHD.
`
`Dose
`
`800–1600 mg/day (X3 mg/kg/day children)
`100–200 mg/day
`12–25 mg/kg/day
`400–1200 mg/day
`3–12 mg/kg/day
`200–800 mg/day
`600–1200 mg/day
`
`Table 2. Phase-3 trials of thalidomide in initial therapy or prevention of cGVHD
`
`N
`
`44
`6
`5
`80
`14
`37
`59
`
`Response
`
`CR-14; PR-12
`CR-2; PR-3
`1 CR; PR-4
`CR-9; PR-7
`CR-6; PR-4
`CR-1; PR-13
`CR-13; PR-8
`
`First therapy
`Koc et al.71
`Arora et al.72
`
`Prevention
`Chao et al.73
`
`Abbreviation: cGVHD ¼ chronic GVHD.
`
`Dose
`
`200–800 mg/day
`200–800 mg/day
`
`400 mg/day
`
`N
`
`51
`54
`
`59
`
`Response
`
`Early discontinuation due to toxicity
`No difference
`
`Increased cGVHD
`
`In summary, several phase-2 trials report about 50% responses
`to thalidomide doses of 100–1600 mg/day. Much of the variability
`between trials reflects small sample sizes, heterogeneous subjects
`and diverse, poorly defined response criteria among other
`complexities discussed above. Thalidomide doses of 4200 mg/
`day were poorly tolerated. Phase-3 trials show no convincing
`benefit of thalidomide for prevention or initial therapy of cGVHD.
`Although thalidomide may be active against cGVHD at high doses
`in rodents,
`these doses cannot be reproducibly and safely
`achieved in humans in most
`instances. An alternative, not
`mutually exclusive problem is the variable course of cGVHD with
`exacerbations and improvements unrelated to therapy interven-
`tions, which may mimic drug adverse effects and/or therapy
`response.
`
`Lenalidomide
`Given the many reports and widespread use of thalidomide in
`corticosteroid-resistant cGVHD, there are remarkably few data on
`use of lenalidomide in this setting. A recent Boolean PUBMED
`search of the English-language literature 1966-present using the
`search terms lenalidomide AND chronic graft-versus-host disease
`identified fewer than 10 reports most of which were anecdotes.
`One phase-2 study of lenalidomide maintenance for myeloma
`after allogeneic transplantation was discontinued because of a
`claimed increased risk of aGVHD.73 However,
`there was no
`concurrent
`control
`arm. Another
`report
`suggested that
`lenalidomide induced a syndrome resembling aGVHD in
`autotransplant recipients.74 It is difficult to interpret these few
`data. One possibility is that concerns about BM toxicity of
`lenalidomide preclude widespread use. Another is publication
`bias: trials or treatments may have been done but were not
`reported because of unfavorable results. The bottom line is
`efficacy of lenalidomide in corticosteroid-resistant cGVHD is not
`known because it appears not extensively studied.
`
`Pomalidomide
`Pomalidomide is a novel immune-modulating drug with 4000-fold
`greater inhibition of TNF-a production compared with thalido-
`mide.75 A comparison of biological activities of pomalidomide
`with thalidomide and lenalidomide is included in Figure 1.
`
`Pomalidomide is extensively used in humans in the setting of
`clinical trials primarily for the treatment of multiple myeloma and
`myeloproliferative neoplasm-associated myelofibrosis.76,77 It offers
`high potency without the dose-limiting toxicities of neuropathy
`and sedation. In persons with multiple myeloma, the dose-limiting
`toxicity is BM suppression with a maximum-tolerated dose of
`2 mg/day.78 Several features of pomalidomide suggest it may be
`useful in treating cGVHD including: (1) in-vitro suppression of TNF-
`a (human monocytes);79 (2) increasing Th1 (mouse cancer vaccine,
`human CD4 þ T cells in vitro);80,81 (3) suppression of Th2 (mouse
`cancer vaccine);80 and (4) stimulation of
`IL-12 and sIL-2Ra
`(humans).78,80 However, other effects of pomalidomide have
`potential
`adverse
`effects
`in
`treating
`cGVHD including:
`(1)
`increased CD45RO þ (memory) CD4 and CD8 T cells
`(humans);78 (2) decreased Tregs;82 (3)
`increased Th2 (polarized
`human CD4 þ T cells in vitro);81 and (4) increased B cells (in-vitro
`human CD19 þ cells).83 Recently, cereblon was identified as an
`essential mediator of Ienalidomide and pomalidomide anticancer
`activity in multiple myeloma. These drugs react with cereblon to
`mediate IFN-regulator factor downregulation. This may affect
`development of Th-17 cells
`sometimes
`implicated in the
`development of cGVHD.84,85 Whether this is a possible target of
`activity of pomalidomide in cGVHD is unknown. Pomalidomide is
`effective in treating experimental scleroderma in mice and
`in a model of bleomycin-induced skin fibrosis (Celgene Corp;
`unpublished data). Because, as discussed, the precise patho-
`genesis of cGVHD is unknown (and may differ
`in different
`persons), it is impossible to predict the impact of therapy with
`pomalidomide outside the context of a controlled clinical trial.
`There is one report of a small phase-2 study of pomalidomide in
`cGVHD. Pusic et al.86 treated eight subjects failing corticosteroids.
`Subjects received 3 mg/day with dose reductions to 2 mg, 1 mg
`and 0.5 mg/day. Seven subjects had dose reductions because of
`muscle cramps, tremor and fatigue. Five subjects discontinued
`therapy for worsening cGVHD of the skin and mouth (N ¼ 1), pain
`(N ¼ 1) and no response (N ¼ 3). There was no BM suppression,
`somnolence, constipation or
`thromboembolic events. Three
`persons reached the primary evaluation end point at 6 months
`at the 2 mg (N ¼ 2) or 1 mg dose (N ¼ 1). These three had global
`PRs per NIH criteria (erythema and gastrointestinal) and oPR
`ongoing improvements (skin, mouth and eyes). This study shows
`
`Bone Marrow Transplantation (2013) 203 – 209
`
`& 2013 Macmillan Publishers Limited
`
`

`

`•
`
`207
`
`feasibility of giving pomalidomide to humans with cGVHD and
`absence of serious side effects at doses of p2 mg/day. A
`randomized phase-2 trial of pomalidomide in persons with
`corticosteroid-resistant cGVHD is planned. There are also large,
`ongoing studies of pomalidomide in multiple myeloma and
`myeloproliferative neoplasm-associated myelofibrosis.
`
`CONCLUSIONS
`thalidomide
`Considerable preclinical data support efficacy of
`therapy for cGVHD. Data from uncontrolled clinical
`trials of
`therapy after failure of other drugs, mostly corticosteroids, support
`this notion. Randomized trials have not been performed
`for the second-line cGVHD treatment. Data from randomized trials
`of cGVHD prevention or
`initial
`therapy are less convincing.
`One randomized therapy trial could not be completed because
`few subjects could tolerate the prescribed dose (nor could
`subjects receiving placebo) whereas another study showed
`no benefit when thalidomide was added to standard post
`transplant immune suppression. The one prevention trial showed
`no benefit.
`Disparate results of intervention to prevent and/or treat aGVHD
`or cGVHD are common. Examples include daclizumab that seems
`effective in corticosteroid-resistant aGVHD but detrimental when
`added to corticosteroids as initial therapy or mycophenolate that
`is ineffective when added to steroids for the initial treatment of
`cGVHD.87,88 Reasons for
`this are complex and incompletely
`understood.
`Its possible corticoisteroid-resistant cases of GVHD
`are biologically different than untreated cases. We conclude
`thalidomide is likely to be effective as second-line therapy of
`cGVHD therapy but that it is difficult to give doses compatible
`with those effective in preclinical models.
`There are few data regarding lenalidomide therapy or preven-
`tion of cGVHD.
`It is unclear whether this represent publication
`bias, limited use or other factors. Persons with cGVHD typically
`have various degrees of BM dysfunction and the BM toxicity of
`lenalidomide poses a substantial challenge. Although low-dose
`lenalidomide has not been extensively evaluated, it seems unlikely
`this will be a useful approach to therapy or prevention of cGVHD.
`There are few data of pomalidomide in cGVHD. The single
`phase-1/-2 trial
`is difficult to interpret. A randomized phase-2
`study will start soon. Lack of neurotoxicity and BM toxicity is
`attractive but additional clinical data are needed.
`There are major challenges to developing therapy for cortico-
`steroid-resistant cGVHD. Prominent among these are (1) cGVHD is
`complex and there are no convincing surrogate in-vitro or in-vivo
`parameters to predict benefit. We are left with clinical trials and
`ultimately, placebo-controlled randomized trials, which are
`difficult and costly to perform; (2) although there are considerable
`efforts to define cGVHD and therapy response, none is prospec-
`tively validated. This confounds design and execution of clinical
`trials using end points other than survival; (3) one of the major
`consequences of cGVHD is immune suppression. However,
`immune-suppressive drugs are our dominant therapy interven-
`tion. This may aggravate rather than help cGVHD outcomes;
`(4) cGVHD has multiple impacts confounding outcomes analyses.
`cGVHD is correlated with decreased survival and with disability,
`but preventing or decreasing cGVHD is correlated with increased
`graft failure, infections and cancer recurrence. Consequently, the
`most convincing outcome of a trial of cGVHD intervention is a
`survival benefit. This is difficult to show in a chronic disease and is
`confounded by competing, unrelated causes of therapy failure.
`Progress in treating and/or preventing cGVHD is a substantial
`challenge in improving survival of recipients of blood cell or BM
`allotransplants. Whether IMiD-class drugs will be useful
`in this
`setting remains to be determined. Current focus is on pomalido-
`mide; trials are progressing.
`
`Immune modulators in chronic GVHD
`YPL Linhares et al
`
`FUTURE CONSIDERATIONS
`Progress in diagnosing, staging and treating cGVHD is ideally
`based on an accurate and reliable understanding of pathogenesis.
`Unfortunately, this is unlikely and we remain with empirical clinical
`trials of drugs that seem promising. We can make progress in
`cGVHD by standardizing diagnosis, staging and evaluation of
`response using the proposed NIH consensus criteria, perhaps with
`some added variables. Use of validated biomarkers may also help
`but none are currently available. There is progress in developing
`collaborations and further
`testing the NIH criteria in large
`prospective observational and interventional trials.89–91 The end
`point of any therapy intervention in cGVHD must be a clinically
`important
`improvement
`in transplant outcomes, especially
`survival and quality of life needs to be confirmed in a double-
`blind randomized clinical trial. Substantial progress in preventing
`and treating aGVHD was made 30 years ago without a precise
`understanding of etiology or pathogenesis. We hope similar
`progress may be made in preventing and treating cGVHD which is
`an even more complex and challenging problem because of
`multiple confounded outcomes of therapy interventions. The goal
`is to prevent cGVHD-associated morbidity and mortality without
`losing graft-vs-cancer effects in diseases where it exists. Lack of
`specificity of current cGVHD therapies makes achieving this goal
`challenging and difficult. Currently, we need to focus on
`developing better treatment strategies.
`
`CONFLICT OF INTEREST
`SZP is an employee of the Center for Cancer Research, National Cancer Institute and
`National Institutes of Health. RPG is a parttime employee of Celgene Corp.
`
`ACKNOWLEDGEMENTS
`RPG acknowledges support from the NIHR Biomedical Research Centre funding
`scheme. Statements included in this article do not represent the official position of
`the NCI, NIH or the US government. SZP receives partial research funding support
`through the Cooperative Research and Development Agreement for intramural-PHS
`clinical re

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