throbber
Review in translational hematology
`
`New approaches for preventing and treating chronic graft-versus-host disease
`Stephanie J. Lee
`
`Despite improvements in the practice of
`allogeneic hematopoietic stem cell trans-
`plantation (HCT) over the last 25 years,
`chronic graft-versus-host disease (GVHD)
`remains a substantial problem with little
`change in the incidence, morbidity, and
`mortality of this complication.
`In fact,
`with increased use of peripheral blood,
`Introduction
`
`transplantation of older patients, and less
`immediate transplantation-related mortal-
`ity, the prevalence of chronic GVHD may
`increase. One of the difficulties in combat-
`ing chronic GVHD is a lack of understand-
`ing about the pathophysiology of the syn-
`drome. Inherent difficulties in conducting
`human clinical trials also contribute to
`
`the lack of meaningful progress. This
`review covers potential new approaches
`to the prevention and treatment of chronic
`GVHD. (Blood. 2005;105:4200-4206)
`
`© 2005 by The American Society of Hematology
`
`Chronic graft-versus-host disease (GVHD) is the most serious and
`common long-term complication of allogeneic hematopoietic stem
`cell transplantation (HCT), occurring in 20% to 70% of people
`surviving more than 100 days.1,2 Approximately half of affected
`people have 3 or more involved organs, and treatment typically
`requires immunosuppressive medications for a median of 1 to 3
`years. Because of higher treatment-related (nonrelapse) mortality,
`chronic GVHD remains the major cause of late death despite its
`association with a lower relapse rate.3,4 In addition, secondary
`malignancies are more common in people with chronic GVHD,
`particularly of commonly involved tissues such as mouth and skin,
`suggesting that chronic inflammation, prolonged exposure to
`immunosuppressive medications, or immune dysregulation facili-
`tates the development of new cancers.5 Finally, the functional
`consequences of chronic GVHD organ involvement are major
`determinants of the health and quality of life of survivors.6,7
`Despite the well-recognized adverse effects of chronic GVHD on
`the long-term success of allogeneic transplantation, its pathophysi-
`ology is poorly understood, and management strategies beyond
`systemic corticosteroids have not been established.
`While there are some lessons that can be translated from basic
`and clinical studies of acute GVHD, several lines of evidence
`suggest that chronic GVHD is not simply a continuation of acute
`GVHD, and that separate approaches will be required for its
`prevention and management. First, except for T-cell depletion and
`use of umbilical cord blood, the major innovations that have
`improved acute GVHD rates do not seem to have affected chronic
`GVHD incidence. Second, while there is significant overlap
`between the organs involved in acute and chronic GVHD, the
`distribution of affected organs in chronic GVHD is much broader.
`Fully evolved chronic GVHD is largely an inflammatory and
`fibrotic process, while acute GVHD is more likely to reflect
`apoptosis and necrosis. Although traditionally the boundary be-
`tween acute and chronic GVHD has been set at 100 days after
`transplantation, more recent definitions hinge on different clinical
`manifestations rather than time of onset. Third, while acute GVHD
`
`is highly associated with subsequent chronic GVHD, approxi-
`mately 25% to 35% of chronic GVHD is de novo without any
`preceding acute manifestations, while 20% to 30% of people who
`had acute GVHD do not go on to develop chronic GVHD later.
`This paper will review current beliefs about the pathophysiol-
`ogy of chronic GVHD and discuss the evidence for emerging
`approaches to prevent or
`treat
`this complication. Readers
`interested in reviews focusing on clinical management are referred
`to other sources.8-10
`
`Rodent models
`
`Both human and murine studies will be reviewed throughout this
`article, although it is important to recognize the similarities and
`differences between the 2 species. In mice, chronic GVHD
`manifestations are highly dependent on the age of the mice, the
`strain combinations selected, the number and type of donor cells
`injected, and the preparative regimen. In contrast to humans, mice
`are not given pharmacologic prophylaxis against GVHD or treat-
`ment for GVHD.
`Murine chronic GVHD can be induced by transplantation
`across class I, class II, or minor histocompatibility antigen barriers
`using irradiation-based regimens. Clinically, these models produce
`late weight loss, lymphoid atrophy, and lymphocyte infiltration of
`affected organs. Fibrosis of the skin, liver, lung, and exocrine
`glands is seen.11 Autoreactive T helper (Th) clones can be
`isolated.12 One minor mismatch model mimics human scleroderma
`with skin and lung fibrosis, a process that can be blocked by
`anti–transforming growth factor ␤ (TGF-␤) antibody treatment.13
`Another murine model mimics systemic lupus erythematosis
`with splenomegaly, B-cell expansion, autoantibodies, and glomeru-
`lonephritis. This syndrome is induced by transplantation of parental
`(P1) cells into F1 (P1 crossed with P2) nonirradiated recipients in
`strain combinations with inherent or induced deficiencies in donor
`CD8⫹ T cells. Th2 cells secreting interleukin-4 (IL-4), IL-6, and
`
`From the Department of Medical Oncology, Dana-Farber Cancer Institute,
`Boston, MA.
`
`Supported in part by P01 HL070149 from the National Heart, Lung, and Blood
`Institute.
`
`Submitted October 22, 2004; accepted February 2, 2005. Prepublished online as
`Blood First Edition Paper, February 8, 2005; DOI 10.1182/blood-2004-10-4023.
`
`Reprints: Stephanie Lee, Dana-Farber Cancer Institute, Boston, MA 02115; e-
`mail: stephanie_lee@dfci.harvard.edu.
`
`© 2005 by The American Society of Hematology
`
`4200
`
`BLOOD, 1 JUNE 2005 䡠 VOLUME 105, NUMBER 11
`
`This material may be protected by Copyright law (Title 17 U.S. Code)
`
`

`

`BLOOD, 1 JUNE 2005 䡠 VOLUME 105, NUMBER 11
`
`NEW APPROACHES TO CHRONIC GVHD
`
`4201
`
`IL-10 appear responsible for clinical manifestations. Notably,
`transplantation from the other parental (P2) strain often results in
`an acute GVHD syndrome unless CD8⫹ T cells are depleted.14
`Administration of cytokines (IL-12, IL-18), costimulatory block-
`ade (4-1BB, cytotoxic T-lymphocyte antigen 4 [CTLA-4], induc-
`ible costimulator [ICOS], CD2815,16), and chemokine antagonists
`(CC chemokine receptor 7 [CCR7])17 can interfere with develop-
`ment of the chronic syndrome. However, the relevance of this
`model has been questioned, as splenomegaly and glomerulonephri-
`tis are not components of human chronic GVHD. Also, most
`human transplantation occurs between major histocompatibility
`complex (MHC)–matched individuals using some form of recipient
`conditioning.
`As with humans, some chronic GVHD manifestations are seen
`in nontransplant situations. The tight skin (TSK) mouse contains a
`partially duplicated fibrillin-1 gene that results in lung, heart, and
`skin lesions and an antibody profile akin to human scleroderma.18
`Transgenic mice with tumor necrosis factor ␣ (TNF-␣) expressed
`in keratinocytes under the keratin-14 promoter have skin fibrosis
`and evidence of cachexia.19 While these models show some
`resemblance to chronic GVHD, their relevance to the human
`syndrome following HCT is questionable.
`
`Human pathophysiology
`
`In humans, chronic GVHD is exceedingly rare after autologous or
`syngeneic transplantation despite similar preparatory regimens. In
`the allogeneic setting, the onset of chronic GVHD is usually
`delayed until 4 to 6 months after transplantation, rarely appearing
`before day ⫹80 and with less than 5% of cases developing after 1
`year. These observations suggest that alloreactivity is a key
`requirement, and that the processes leading to chronic GVHD
`either have a long latency or they exert their effects slowly on
`target tissues.
`The current understanding of human chronic GVHD etiology
`starts with pathogenic donor T cells that expand in response to
`alloantigens or autoantigens unchecked by normal
`thymic or
`peripheral mechanisms of deletion. Critical donor or recipient
`tolerance-promoting cells may be absent. These pathologic T cells
`then attack target tissue directly through cytolytic attack, secretion
`of inflammatory and fibrosing cytokines, or promotion of B-cell
`activation and autoantibody production. Tissue damage leads to
`fibrosis and dysfunction. Chronic GVHD or its treatment leads to
`death from organ failure or infection. Thus, prevention and
`treatment of chronic GVHD has focused on interrupting this
`process through elimination or inhibition of pathogenic T cells,
`induction of tolerance, cytokine therapy, elimination of B cells, or
`modulating effects on local tissues.
`
`Approaches to chronic GVHD prevention
`
`Choice of donor, graft source, and GVHD prophylaxis
`
`Clinical studies have identified many recipient, donor, and trans-
`plant factors associated with higher rates of chronic GVHD.
`Children experience lower rates of chronic GVHD, but the major
`risk factors for, organ manifestations in, and clinical impact on
`affected children appear similar.2 Many recipient risk factors
`associated with increased chronic GVHD are not modifiable, and
`include older age, certain diagnoses (eg, chronic myeloid leukemia,
`
`aplastic anemia), and lack of an HLA-matched donor. Other
`modifiable factors are associated with lower rates of chronic
`GVHD, and although causality is not proved, avoidance of
`high-risk factors may decrease the risk of clinically significant
`chronic GVHD. Assuming multiple HLA-matched donors are
`available, then selection of a younger related donor, use of bone
`marrow rather than peripheral blood,20 and limitation of CD34⫹21
`and T-cell dose infused may minimize the risk of chronic GVHD.
`Two reports suggest that chronic GVHD is also more likely to be
`extensive and difficult to treat in recipients of related and unrelated
`peripheral blood compared with bone marrow.1,22 Comment on the
`incidence and clinical manifestations of chronic GVHD after
`nonmyeloablative or reduced-intensity conditioning regimens awaits
`more definitive reports. If the recipient is male, then avoidance of a
`female donor, especially someone multiparous, may decrease the
`risk of chronic GVHD.23 Donor ABO compatibility and cytomega-
`lovirus (CMV) seronegativity have also been associated with lower
`risks of chronic GVHD. While umbilical cord blood is currently a
`graft source of last resort in adults, it appears to be associated with
`lower rates of chronic GVHD.24
`As most HCT procedures use HLA-matched donors, so-called
`“minor” histocompatibility antigens (mHAs) must contribute to the
`pathophysiology of acute and chronic GVHD. Minor antigens are
`polymorphic proteins encoded in the genome that are degraded and
`presented to T cells in the context of HLA,
`thus inducing
`MHC-restricted immune responses. Conceivably,
`identification
`and avoidance of important minor mismatches could prevent both
`acute and chronic GVHD. In human transplantation, studies of
`predictors of acute GVHD have primarily focused on mHA-1 and
`mHA-2, expressed solely on hematopoietically derived cells (includ-
`ing dendritic and Langerhans cells) and presented in the context of
`HLA-A*0201. Other antigens are broadly expressed on tissues:
`mHA-3 is presented by HLA-A*0101, while mHA-8 is presented
`by HLA-A*0201 and HLA-A*0202. However, none of these
`mHAs has been associated with chronic GVHD.25,26
`An increased risk of chronic GVHD has long been recognized
`when a male recipient receives a graft from a female donor,
`particularly one who may have been alloimmunized by pregnancy
`or transfusion.27 The best explanation for this clinical observation
`is that mHAs encoded on the Y chromosome can elicit responses
`from female donors in male recipients.28 In a murine skin explant
`model, female cytotoxic T lymphocytes (CTLs) specific for the
`H-Y antigens found in males caused severe changes consistent with
`acute GVHD when exposed to male but not female skin.29
`
`Prevention of acute GVHD
`
`Acute GVHD is a major predictor of chronic GVHD, and 70% to
`80% of people with grades II to IV acute GVHD develop chronic
`GVHD.30 The nature of the observed association is highly contro-
`versial, and at least 4 explanations have been offered. Chronic
`GVHD has been suggested to be a later manifestation of alloreac-
`tive acute GVHD, a result of tissue damage (particularly thymus)
`caused by acute GVHD,31 a result of treatment (particularly
`steroids) for acute GVHD,32 or an epiphenomenon that is associ-
`ated with but not etiologically linked to acute GVHD. These
`distinctions are important because interfering with the development
`of acute GVHD may prevent chronic GVHD if any of the first 3
`mechanisms is operative, but would not affect chronic GVHD
`incidence if the last were true. In fact, some successful attempts to
`decrease acute GVHD may have actually increased chronic GVHD
`rates. Two reports have suggested that exposure to steroids as
`
`

`

`4202
`
`LEE
`
`BLOOD, 1 JUNE 2005 䡠 VOLUME 105, NUMBER 11
`
`prophylaxis for acute GVHD tends to increase the rate of subse-
`quent chronic GVHD.32,33
`Some attention has recently focused on the role of recipient
`and donor APCs in prevention of acute GVHD while preserving
`graft-versus-tumor effects. Shlomchik et al34 reported murine
`studies in which recipient antigen-presenting cells are critical
`for initiation of acute GVHD by donor CD8 cells. Human
`reports also suggest that APCs may be important. In patients
`with myeloid malignancies undergoing haplotype-mismatched
`and killer immunoglobulin-like receptor (KIR) mismatched
`transplantations, decreased rates of acute GVHD, decreased
`relapse rates, and higher overall survival in patients were reported.
`Chronic GVHD rates were not reported. The proposed mechanism
`of action is elimination of host APCs (decreasing acute GVHD) and
`host tumor cells (decreasing relapse rate) by uninhibited donor
`natural killer (NK) cells.35 This interpretation was supported by a
`similar study in unrelated donor transplantation that
`included
`anti–thymocyte globulin (ATG) for GVHD prophylaxis. KIR
`mismatching was associated with better survival and marginally
`lower rates of acute GVHD, although rates of chronic GVHD were
`comparable.36 Studies in other populations have disputed these
`findings or identified other important NK factors; however, chronic
`GVHD rates are not mentioned.37,38 A second approach to depletion
`of host APCs uses extracorporeal photopheresis (ECP) prior to
`infusion of donor stem cells. In a small series, Chan et al39 reported
`lower rates of extensive chronic GVHD, especially when donor
`dendritic cell (DC) chimerism is achieved by day 100.
`Two other studies looked at donor APCs, specifically plasmacy-
`toid DC2 cells. Clark et al40 reported that people with chronic
`GVHD had normal numbers of donor-derived plasmacytoid DC2s
`in their blood compared with reduced numbers in posttransplanta-
`tion controls without chronic GVHD. In contrast, Waller et al41
`reported that patients receiving bone marrow grafts with higher
`numbers of CD3⫺CD4bright, presumably DC2 cells, had a lower
`incidence of chronic GVHD. A better understanding of the role of
`APCs in chronic GVHD awaits additional studies.
`
`Chronic GVHD prophylaxis
`
`Attempts to prevent chronic GVHD through prolonged use of
`immunosuppressive medications or addition of other agents have
`been unsuccessful. Based on observational reports that extended
`calcineurin inhibitor treatment may have decreased the incidence
`of chronic GVHD,42 a randomized trial by the Seattle group
`compared 6 months versus 24 months of cyclosporine in patients
`with prior acute GVHD or evidence of subclinical chronic GVHD
`on skin biopsy. No statistically significant difference was seen in
`rates of clinical extensive chronic GVHD.43 Chao et al44 studied
`thalidomide beginning 80 days after transplantation in a random-
`ized, double-blinded, placebo-controlled study and found a higher
`rate of chronic GVHD and mortality in patients receiving active
`drug. Ringden et al45 treated a small number of patients with grade I
`or higher acute GVHD with steroids until 6 months after transplan-
`tation in an attempt to prevent chronic GVHD. They abandoned
`this approach when a higher than expected incidence of severe
`chronic GVHD was noted.
`
`Preemptive treatment of minimal chronic GVHD
`
`A provocative study that attempted to avert clinical chronic GVHD
`by preemptive treatment of subclinical chronic GVHD noted that
`
`most (70%) went on to develop chronic GVHD, and that the relapse
`rate was higher in patients who underwent
`transplantation in
`relapse who did not develop chronic GVHD.46
`Eosinophilia is associated with Th2 allergic disorders and may
`precede the diagnosis of clinical chronic GVHD, leading at least
`one pediatric center to start treatment when eosinophilia alone is
`noted after transplantation. No long-term results of this strategy
`were reported.47
`
`New approaches to treatment
`
`More than 20 years ago, corticosteroid therapy was shown to
`improve survival in patients with chronic GVHD compared with no
`therapy.48 However, extended corticosteroid therapy has well-
`known, long-term adverse effects, and alternative treatments are
`generally unsatisfactory.49 Other reviews concisely summarize data
`on primary and salvage therapies currently available to treat
`chronic GVHD, so this information will not be reviewed here.
`Instead, potential new approaches to control of chronic GVHD are
`emphasized. Two ongoing randomized, double-blinded multicenter
`studies of hydroxychloroquine or mycophenolate mofetil added to
`standard corticosteroid initial treatment seek to improve initial
`therapy (A. L. Gilman, University of North Carolina at Chapel Hill,
`and P. J. Martin, Fred Hutchinson Cancer Research Center, oral
`communication, July 2004).
`
`Elimination or inhibition of pathogenic T cells
`through pharmacologic therapy
`
`Pharmacologic inhibition of T cells forms the backbone of modern
`chronic GVHD therapy. In human cutaneous chronic GVHD, most
`infiltrating lymphocytes are CD8⫹,50,51 although one recent report
`suggests that alloreactive CD4⫹ infiltrating cells are also important
`and pre-exist in the donor.52 Newer agents under study include
`mycophenolate mofetil,53 sirolimus,54 daclizumab,55 pentostatin,56
`and alemtuzumab. When pharmacologic therapy is stopped, be-
`tween 10% to 25% of patients flare and require reinstitution of
`systemic treatment.57 Optimally, novel methods of T-cell inhibition
`would target specifically the T cells responsible for chronic GVHD
`while sparing other cells that could provide protective immunity.
`For example, given the relative lymphopenia with fewer recent
`thymic emigrants (as measured by T-cell receptor excision circle
`levels) and a relative deficit of central memory populations
`(CD45⫺CCR7⫹, precursor effector cells) noted in chronic GVHD,
`preservation of any nonpathologic T cells might help decrease
`serious infections.31,58,59
`In humans, OX-40 (CD134, a member of TNF receptor
`superfamily responsible for costimulation) expressing CD4⫹ cells
`are reportedly associated with onset of chronic GVHD and
`decreased response to initial therapy.60 If OX-40 is a marker of cells
`involved in chronic GVHD, then targeting these cells or their
`interactions at the onset of chronic GVHD may be therapeutic. In
`mice, antibody to the OX-40 ligand (OX-40L) decreases acute
`GVHD, but to this point, human trials have not been conducted.
`
`Inhibition of pathogenic T cells through cellular therapy
`
`Cellular approaches to inhibiting T cells have recently focused on
`so called T regulatory cells (Tregs), a small subset of T cells that can
`suppress proliferation and function of T effector cells, particularly
`
`

`

`BLOOD, 1 JUNE 2005 䡠 VOLUME 105, NUMBER 11
`
`NEW APPROACHES TO CHRONIC GVHD
`
`4203
`
`of the Th1 class.61 Tregs are activated in an antigen-specific
`manner, perhaps with involvement of IL-10, TGF-␤, and immature
`dendritic cells, causing them to express high levels of CD25⫹ (IL-2
`receptor alpha chain) constitutively.53 They are unresponsive to
`mitogens, express CTLA-4, and are able to block production of
`IL-2 and interferon ␥ (IFN-␥) by effector T cells in a manner that
`relies on cell-to-cell contact. Most reports suggest
`that Tregs
`function in an antigen-nonspecific manner as tested in vitro,
`although at least one report suggested that they may be antigen
`specific in vivo.62 Tregs may also have inhibitory effects on APCs,
`and their high expression of CCR4 and CCR8 suggests aberrant
`trafficking.53 Recently, a distinction has been made between
`“natural” Tregs, important in preventing autoimmune responses to
`continually expressed antigens in the noninflammatory setting, and
`“adaptive” Tregs, which are important in developing tolerance to
`foreign antigens and quelling inflammatory processes.63
`In murine models, infusion of CD4⫹CD25⫹ T cells is able to
`prevent acute GVHD while graft-versus-tumor is maintained.64,65
`Removal of CD4⫹CD25⫹ T cells from the graft or blockade by
`CD25⫹ antibodies worsened acute GVHD.66 Another model sug-
`gested that chronic GVHD incidence and severity is higher in the
`absence of recipient CD4⫹CD25⫹ cells, and that repletion with
`recipient or host Tregs is protective.11 These murine observations
`led to the hypothesis that human chronic GVHD results from a low
`Treg population, and that expansion ex vivo and replacement could
`help control chronic GVHD. However, a small study of 17 patients
`with chronic GVHD showed higher numbers of CD4⫹CD25⫹ cells
`in people with chronic GVHD but lower CD62 ligand (CD62L)
`expression compared with people without chronic GVHD, al-
`though absolute numbers and functionality were similar to con-
`trols.67 A more recent study of people considered to have allogeneic
`or autologous chronic GVHD quantified Foxp3 expression as a
`marker of Tregs and suggested that they may indeed be deficient.68
`Thus,
`it remains controversial whether Tregs are involved in
`chronic GVHD, and further studies are warranted.
`
`Interventions to induce tolerance
`
`It is not clear when donor tolerance to the recipient is established.
`Possibilities include before engraftment due to lack of critical
`donor-recipient differences, early after transplantation due to
`deletion or tolerization of donor T cells, or in an ongoing process
`later after transplantation. The role of immunosuppressive medica-
`tions in promoting or interfering with tolerance development is
`unknown. Anecdotal reports of exacerbation or induction of
`chronic GVHD with sun exposure, sunburns, and infections
`suggest that a state of apparent tolerance can be broken by aberrant
`antigen presentation or inflammatory states.
`Improvement of thymic function has the potential both to
`improve protection against pathogens and decrease autoimmunity,
`either by increasing the thymus’ ability to delete autoreactive cells
`or by fostering development of natural Tregs. Loss of thymic
`function in older patients may explain the higher observed rates of
`chronic GVHD compared with children. IL-7 is produced by
`stromal cells in the thymus and bone marrow and plays a critical
`role in T- and B-cell development. In children, posttransplantation
`IL-7 levels inversely correlate with the absolute lymphocyte
`number.69 The effects of exogenous IL-7 have been tested on
`human thymic cultures or human stem cells injected into immunoin-
`competent mice, showing that T-cell production can be increased.70,71
`
`Other approaches to T-cell tolerization rely on immunomodu-
`lation, such as might be provided by extracorporeal photophere-
`sis (ECP). Subjects who responded to ECP were more likely to
`have a clonal T-cell population, although these populations were
`found equally in people with and without chronic GVHD.72
`Approximately 5% of mononuclear cells undergo apoptosis
`after ECP, and these degrading cells may cause autoimmuniza-
`tion and an increased production of IL-10 and IL-1 receptor
`antagonist (IL-1Ra).73 Clinical responses are typically delayed
`until 2 to 3 months of therapy.
`tolerance
`There are several reports of oral and intranasal
`induced in murine models. In a minor mismatch chronic GVHD
`model, recipients fed recipient splenocyte proteins for 11 days after
`transplantation developed less fibrosis, less organ inflammation,
`and higher levels of IL-10.74 In a parent into F1 acute GVHD
`model, tolerance was induced by posttransplantation oral adminis-
`tration of recipient splenocytes.75 The intranasal route has been
`used to tolerize female mice to male HY peptides, resulting in
`acceptance of male skin grafts and bone marrow cells that are
`rejected in controls.76 The mechanism of oral tolerance is hypoth-
`esized to be induction of a regulatory T cell of Th3 phenotype that
`secretes high amounts of TGF-␤.77
`
`Cytokine therapy
`
`Cytokines secreted by T cells, APCs, and damaged target tissues
`may contribute to chronic GVHD. T-cell cytokines are generally
`classified as Th1-type (IL-2, IFN-␥) and Th2-type (IL-4, IL-5,
`IL-10, IL-13).78 APCs and damaged target
`tissue can secrete
`TNF-␣ and IL-1. While animal studies have demonstrated the
`importance of cytokine availability to chronic GVHD develop-
`ment, documentation in human systems has been less convincing.
`Human studies in chronic GVHD have focused on circulating
`cytokine levels (IL-10, TGF-␤1), tissue cytokine expression (IL-2,
`IFN-␥, IL-4, IL-5, IL-10, IL-1␣, TNF-␣, platelet-derived growth
`factor [PDGF], TGF-␤), and donor or recipient cytokine polymor-
`phisms (IFN-␥, IL-6, IL-10, IL-1␣, IL-1␤, IL-1Ra, TNF-␣, TNF-
`␤). To date, only cytokine blockade (TNF-␣) has advanced to
`human trials for treatment of chronic GVHD.
`Studies with limited numbers of patients suggest that circulating
`IL-10 levels are lower in people with chronic GVHD, while IL-1␤,
`IL-6, TNF-␣, and TGF-␤1 levels are higher.79,80 A small study
`suggested that in vitro mononuclear cell IL-10 secretion after
`stimulation was lower in patients with chronic GVHD, while use of
`IL-10 blocking antibodies increased IFN-␥ secretion.81 This con-
`trasts with the murine parent into F1 studies in which high IL-10 is
`associated with chronic GVHD and IL-10 antibodies can block
`chronic GVHD manifestations. Of note, any human trials of
`exogenous IL-10 supplementation will have to be undertaken
`carefully as observational studies in human transplant and nontrans-
`plant settings document an association between higher serum IL-10
`levels, greater severity of septic shock, and fatal outcome.82-84
`Ochs et al30 used reverse-transcription–polymerase chain reac-
`tion (RT-PCR) to study transcription of cytokines in a limited
`number of skin biopsies from people with active chronic GVHD,
`people without active chronic GVHD, and healthy controls.
`Although IFN-␥ transcript levels were elevated in people with
`active chronic GVHD, the other cytokine levels studied were not
`different (IL-1␣, TNF-␣, IL-2, IL-4, IL-5, IL-10, PDGF, TGF-␤).
`Certain donor and recipient cytokine polymorphisms have been
`associated with chronic GVHD, but results are conflicting. In
`contrast with the findings of studies measuring circulating IL-10
`
`

`

`4204
`
`LEE
`
`BLOOD, 1 JUNE 2005 䡠 VOLUME 105, NUMBER 11
`
`levels, polymorphisms of IL-10 associated with higher donor85 or
`recipient86 IL-10 production were associated with chronic GVHD.
`Two recipient IL-1␣ polymorphisms were associated with chronic
`GVHD, although one polymorphism is associated with increased
`IL-1␣ while the other, with decreased IL-1␣ production.87 In
`another study, neither recipient IL-10 nor IL-1 polymorphisms
`were associated with chronic GVHD.88 Recipient IL-1Ra polymor-
`phisms associated with lower production of IL-1Ra, and thus
`perhaps more biologic activity of IL-1 were also associated with
`chronic GVHD.86
`Recipient IL-6 polymorphisms have been associated with
`chronic GVHD in 2 reports,89,90 though these studies failed to find
`an association with other donor or recipient polymorphisms
`(TNF-␣, TNF-␤, IL-1, IL-10, IFN-␥). A study by Stark et al90
`identified an association between chronic GVHD and donor
`homozygosity for the 196R allele of the TNF type II receptor
`(TNFRII), associated with decreased TNFRII levels and thus
`increased functional levels of TNF. Eternacept, a recombinant
`soluble TNF inhibitor, has reported activity in 10 steroid-refractory
`patients with chronic GVHD.91 Another TNF inhibitor, infliximab,
`has been associated with invasive fungal infections when used to
`treat steroid-refractory acute GVHD, and there are no reports of its
`use in chronic GVHD.
`
`Eliminate B cells
`
`Circulating autoantibodies and polyclonal hypergammaglobuline-
`mia were noted in the earliest reports of human chronic GVHD,92
`but more recent studies show hypogammaglobulinemia and fewer
`precursor and mature B cells.58,93 Nevertheless,
`the similarity
`between many autoimmune diseases associated with autoantibod-
`ies and the clinical manifestations of chronic GVHD has always
`been intriguing. Patients with CMV infection may have anti-CD13
`antibodies, which recognize normal structures in skin.94 Patients
`with sclerodermatous chronic GVHD are more likely to have
`immunoglobulin G (IgG) antibodies to nuclear proteins (95%), but
`these antibodies are also common in patients without chronic
`GVHD.95 Miklos et al96 have found antibody responses to H-Y
`antigens in male recipients with female donors, and these antibod-
`ies were associated with development of chronic GVHD.
`Rituximab is a chimeric murine-human CD20⫹ antibody that
`has reported activity in treating chronic GVHD and other autoim-
`mune diseases. Two case series suggest
`that
`transient B-cell
`depletion can improve clinical manifestations of chronic GVHD
`with durable responses.97,98
`
`Minimize effects on target tissues
`
`Even if the systemic causes of chronic GVHD cannot be controlled,
`treatments aimed at target tissues may still minimize morbidity and
`improve functionality. One of the major debilitating tissue re-
`sponses is fibrosis. Halofuginone has been given topically or
`systemically to inhibit TGF-␤–induced collagen ␣1 gene overex-
`pression. Halofuginone inhibits smad3 phosphorylation, particu-
`larly in fibroblasts induced to oversecrete collagen by activation
`with TGF-␤ or activating mutations, via a mechanism that relies on
`protein synthesis.99 If applied topically, halofuginone is not ab-
`sorbed and effects are reversible after 3 months. In a rat model of
`liver fibrosis, halofuginone was able to reverse fibrosis.100 In
`human and murine studies, normal collagen production was
`unaffected. Despite great interest in studying halofuginone for
`
`sclerodermatous chronic GVHD, this drug is not yet available for
`studies of chronic GVHD in the United States.
`Excess collagen deposition may also be combated through
`physical rehabilitation, similar to the treatment of burn victims and
`people with scleroderma, many of whom also suffer from excess
`collagen deposition. Aggressive heat therapy, massage, and passive
`range-of-motion exercises can help maintain function until the
`sclerotic process can be controlled.101 To date, there are no studies
`specifically evaluating rehabilitative methods in people with
`chronic GVHD.
`Ursodeoxycholic acid has reportedly improved the biochemical
`profile of patients with hepatic chronic GVHD. As a less hydropho-
`bic acid, ursodeoxycholic acid replaces the human bile acids and
`may result in less cholestatic damage. It may also cause decreased
`expression of HLA class I molecules on hepatocytes. However, this
`medication is very expensive and in one study of patients with
`chronic GVHD, biochemical improvements were lost upon discon-
`tinuation of the drug. In primary biliary sclerosis, ursodeoxycholic
`acid has improved liver function tests but not changed the natural
`history of the disease.102 No long-term studies of this drug in
`chronic GVHD have been reported.
`Biedermann et al103 studied skin biopsies in a small number of
`people with acute, chronic, or no GVHD after allogeneic transplan-
`tation, and found fewer microvessels and higher levels of von
`Willebrand factor in people with chronic GVHD. They hypothesize
`that endothelial damage happens first, followed by fibrosis akin to
`what happens in solid organ rejection. Thus, methods to prevent
`endothelial damage may have efficacy in chronic GVHD preven-
`tion or treatment.
`Topical immunosuppressives such as corticosteroids and cal-
`cineurin inhibitors can improve local symptoms in the eyes, mouth,
`skin, and vaginal area. Small series report efficacy for autologous
`serum eyedrops in Sjo¨gren syndrome and UVB laser for lichen
`planus. As new approaches to local therapy are developed for
`autoimmune diseases, rapid translation and testing in the chronic
`GVHD population seems warranted.
`
`Summary
`
`A better understanding of chronic GVHD and discovery of ways to
`prevent or control this complication without compromising disease-
`free survival stand as major barriers to the success of allogeneic
`transplantation. Far too many people

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