throbber
I
`
`Seminar
`
`Lancet 2009; 373: 1550–61
`Published Online
`March 12, 2009
`DOI:10.1016/S0140-
`6736(09)60237-3
`University of Michigan,
`Pediatrics and Internal
`Medicine, Blood and Marrow
`Transplantation Program,
`Ann Arbor, MI, USA
`(Prof J L M Ferrara MD,
`Prof J E Levine MD, P Reddy MD);
`and Internal Medicine and
`Department of Haematology/
`Oncology, University Hospital,
`Regensburg, Germany
`(Prof E Holler MD)
`Correspondence to:
`Prof James L M Ferrara,
`University of Michigan,
`1500 East Medical Center Drive,
`6303 CCC, Ann Arbor,
`MI 48109-5942, USA
`ferrara@umich.edu
`
`Graft-versus-host disease
`
`James L M Ferrara, John E Levine, Pavan Reddy, Ernst Holler
`
`Haemopoietic-cell transplantation (HCT) is an intensive therapy used to treat high-risk haematological malignant
`disorders and other life-threatening haematological and genetic diseases. The main complication of HCT is graft-
`versus-host disease (GVHD), an immunological disorder that aff ects many organ systems, including the
`gastrointestinal tract, liver, skin, and lungs. The number of patients with this complication continues to grow, and
`many return home from transplant centres after HCT requiring continued treatment with immunosuppressive drugs
`that increases their risks for serious infections and other complications. In this Seminar, we review our understanding
`of the risk factors and causes of GHVD, the cellular and cytokine networks implicated in its pathophysiology, and
`current strategies to prevent and treat the disease. We also summarise supportive-care measures that are essential for
`management of this medically fragile population.
`
`Introduction
`The number of allogeneic haemopoietic-cell trans-
`plantations (HCTs) continues to rise, with more than
`25 000 procedures undertaken annually. The graft-versus-
`leukaemia or graft-versus-tumour eff ect during this
`procedure eff ectively eradicates many haema tological
`malignant diseases.1 Development of novel strategies
`that use donor leucocyte infusions, non-myeloablative
`conditioning, and umbilical-cord blood transplantation
`has helped expand the indications for allogeneic HCT
`over the past few years, especially for older patients.2
`Improvements
`in
`infectious prophylaxis,
`immuno-
`suppressive treatments, suppor tive care, and DNA-based
`tissue typing have also contributed to enhanced outcomes
`after the technique.1 Yet, the major complication of
`allogeneic HCT—graft-versus-host disease (GVHD)—
`remains lethal and limits use of this important procedure.2
`In view of current trends, the number of transplants
`from unrelated donors is expected to double within the
`next 5 years, substantially increasing the population of
`patients with GVHD. In this Seminar, we review advances
`made in identifi cation of genetic risk factors and
`pathophysiology of this major HCT complication and its
`prevention, diagnosis, and treatment.
`
`Cause and clinical features
`50 years ago, Billingham formulated three requirements
`for development of GVHD: (1) the graft must contain
`immunologically competent cells; (2) the recipient must
`express tissue antigens that are not present in the
`transplant donor; and (3) the patient must be incapable
`
`Search strategy and selection criteria
`
`We searched PubMed and Medline with the term ‘‘GVHD’’
`and cross-referenced it with the following words: ‘‘clinical’’,
`‘‘cytokines’’, ‘‘MHC’’, ‘‘HLA antigens’’, ‘‘biology’’, and
`‘‘immunology’’. We included mostly peer-reviewed original
`and review journal articles published within the past
`decade, except for seminal articles that initially described
`the clinical features. All non-peer-reviewed manuscripts,
`supplements, and textbooks were excluded.
`
`of mounting an eff ective response to eliminate the
`transplanted cells.3 We know now that the immuno-
`logically competent cells are T cells and that GVHD can
`develop
`in various clinical settings when tissues
`containing T cells (blood products, bone marrow, and
`solid organs) are transferred from one person to another
`who is not able to eliminate those cells.4,5 Patients whose
`immune systems are suppressed and who receive white
`blood cells from another individual are at especially high
`risk for the disease.
`GVHD arises when donor T cells respond to
`genetically defi ned proteins on host cells. The most
`important proteins are human leucocyte antigens
`(HLAs),2,6,7 which are highly polymorphic and are
`encoded by the major histocompatibility complex
`(MHC). Class I HLA (A, B, and C) proteins are expressed
`on almost all nucleated cells of the body at various
`densities. Class II proteins (DR, DQ, and DP) are mainly
`expressed on haemopoietic cells (B cells, dendritic cells,
`and monocytes), but their expression can be induced on
`many other cell types after infl ammation or injury.
`High-resolution DNA typing of HLA genes with
`PCR-based techniques has now largely replaced earlier
`methods. The frequency of acute GVHD is directly
`related to the degree of mismatch between HLA
`proteins,8,9 and thus ideally, donors and recipients are
`matched at HLA A, B, C, and DRB1 (referred to as
`8/8 matches), but mismatches can be tolerated for
`umbilical-cord blood grafts (see Clinical features of
`acute GVHD).10–12
`Despite HLA identity between a patient and donor,
`about 40% of recipients of HLA-identical grafts develop
`systemic acute GVHD that needs treatment with
`high-dose steroids. This disorder is due to genetic
`diff erences that lie outside the HLA loci and that encode
`proteins referred to as minor histocompatibility antigens.
`Some minor histocompatibility antigens, such as HY
`and HA-3, are expressed on all tissues and are targets for
`both GVHD and graft-versus-leukaemia.13 Others, such
`as HA-1 and HA-2, are expressed most abundantly on
`haemopoietic cells (including leukaemic cells) and could,
`therefore, induce an enhanced graft-versus-leukaemia
`eff ect with diminished GVHD.13,14
`
`1550
`
`www.thelancet.com Vol 373 May 2, 2009
`
`

`

`I
`
`Seminar
`
`Panel 1: Acute GVHD symptoms
`
`Skin
`• Maculopapular skin rash
`
`Upper gastrointestinal tract
`• Nausea, anorexia, or both, and positive histological fi ndings
`
`Lower gastrointestinal tract
`• Watery diarrhoea (≥500 mL)
`• Severe abdominal pain
`• Bloody diarrhoea or ileus (after exclusion of infectious
`causes)
`
`Liver
`• Cholestatic hyperbilirubinaemia
`
`Polymorphisms in both donors and recipients of
`cytokines that have a role in the classic cytokine storm of
`GVHD (see Pathophysiology of acute GVHD) have been
`implicated as risk factors for the disorder.15 Tumour
`necrosis factor (TNF) α, interleukin 10, and interferon γ
`variants have correlated with GVHD in some, but not all,
`studies.16–18 Genetic polymorphisms of proteins connected
`with innate immunity, such as nucleotide oligomerisation
`domain 2 and keratin 18 receptors, have also been
`associated with the disorder.19–22 Future strategies to
`identify the best possible transplant donor will probably
`incorporate both HLA and non-HLA genetic factors.
`
`Clinical features of acute GVHD
`On the basis of early work, acute GVHD was defi ned as
`arising before day 100 post-transplant, whereas chronic
`disease happened after that time.23–25 This defi nition is
`far from satisfactory, and a National Institutes of Health
`classifi cation includes late-onset acute GVHD (after
`day 100) and an overlap syndrome with features of both
`acute and chronic disorder.26,27 Late-onset acute GVHD
`and the overlap syndrome arise with greater frequency
`after reduced-intensity conditioning, an increasingly
`widespread technique (see Prevention of GVHD). Panel 1
`shows the clinical manifestations of acute GVHD. In a
`comprehensive review, Martin and colleagues noted that
`at onset of acute GVHD, aff ected regions included skin
`(81% of patients), gastrointestinal tract (54%), and
`liver (50%).23
`Skin is most frequently aff ected and is usually the fi rst
`organ involved, generally coinciding with engraftment of
`donor cells. The characteristic maculopapular rash is
`pruritic and can spread throughout the body, sparing the
`scalp (fi gure 1). In severe cases the skin can blister and
`ulcerate.28 Apoptosis at the base of epidermal rete pegs is
`a characteristic pathological fi nding. Other features
`include dyskeratosis, exocytosis of lymphocytes, satellite
`lymphocytes
`adjacent
`to dyskeratotic
`epidermal
`keratinocytes, and perivascular lymphocytic infi ltration
`in the dermis.29,30
`
`Figure 1: Acute GVHD of the skin (grade I)
`Photograph courtesy of J Levine.
`Gastrointestinal-tract involvement of acute GVHD
`usually presents as diarrhoea but can also include
`vomiting, anorexia, abdominal pain, or a combination
`when severe.29 Diarrhoea in GVHD is secretory and
`usually voluminous (>2 L per day). Bleeding, which has
`poor prognosis, happens as a result of mucosal
`ulceration,31 but patchy involvement of mucosa generally
`leads to a normal appearance on endoscopy.32 Radiological
`fi ndings of the gastrointestinal tract include luminal
`dilatation with thickening of the wall of the small bowel
`(ribbon sign on CT) and air or fl uid levels suggestive of
`an ileus.28 Histological features include patchy ulcerations,
`apoptotic bodies in the base of crypts, crypt abscesses,
`and loss and fl attening of surface epithelium.33
`Liver disease caused by GVHD can be diffi cult to
`distinguish from other causes of liver dysfunction after
`bone-marrow transplantation, such as veno-occlusive
`disease, toxic drug eff ects, viral infection, sepsis, or iron
`overload. The histological features of hepatic GVHD are
`endothelialitis, lymphocytic infi ltration of the portal
`areas, pericholangitis, and bile-duct destruction.34,35
`However, biopsy specimens of liver are taken rarely
`because thrombocytopenia early after transplantation
`greatly increases the risks of the biopsy procedure,
`making the diagnosis of GVHD one of exclusion.
`Severity of acute GVHD is ascertained by the extent of
`involvement of the three main target organs. Overall
`grades are I (mild), II (moderate), III (severe), and
`IV (very severe). Severe GVHD has poor prognosis, with
`25% long-term survival (5 years) for grade III disease and
`5% for grade IV.36
`
`www.thelancet.com Vol 373 May 2, 2009
`
`1551
`
`

`

`I
`
`Seminar
`
`Panel 2: Chronic GVHD symptoms
`
`Skin
`Dyspigmentation, new-onset alopecia, poikiloderma, lichen
`planus-like eruptions, or sclerotic features
`
`Nails
`Nail dystrophy or loss
`
`Mouth
`Xerostomia, ulcers, lichen-type features, restrictions of
`mouth opening from sclerosis
`
`Eyes
`Dry eyes, sicca syndrome, cicatricial conjunctivitis
`
`Muscles, fascia, joints
`Fasciitis, myositis, or joint stiff ness from contractures
`
`Female genitalia
`Vaginal sclerosis, ulcerations
`
`Gastrointestinal tract
`Anorexia, weight loss, oesophageal web or strictures
`
`Liver
`Jaundice, transaminitis
`
`Lungs
`Restrictive or obstructive defects on pulmonary function
`tests, bronchiolitis obliterans, pleural eff usions
`
`Kidneys
`Nephrotic syndrome (rare)
`
`Heart
`Pericarditis
`
`Marrow
`Thrombocytopenia, anaemia, neutropenia
`
`Prevalence of acute GVHD is directly related to the
`degree of mismatch between HLA proteins. It ranges
`from 35–45% in recipients of full-matched sibling donor
`grafts8,9 to 60–80% in people receiving one-antigen
`HLA-mismatched unrelated-donor grafts.6,37–39 The same
`amount of mismatch causes diminished GVHD with
`umbilical-cord blood grafts, and frequency of acute
`GVHD is low after transplantation of partly matched
`umbilical-cord blood units (35–65%).12
`
`Clinical features of chronic GVHD
`Chronic GVHD is the major cause of late non-relapse
`death after HCT.40 Its presentation can be progressive
`(active or acute GVHD merging into chronic), quiescent
`(acute disease that resolves completely but is followed
`later by chronic), or de novo. Older recipient age and a
`history of acute GVHD are the greatest risk factors for
`chronic disease.41 Therefore, strategies for acute GVHD
`prevention could help to prevent chronic disease. Panel 2
`shows that manifestations of chronic GVHD are
`
`somewhat protean and typically of an autoimmune
`nature. Clinical signs are generally seen fi rst in the buccal
`mucosa (fi gure 2). New consensus criteria for diagnosis
`and staging of chronic GVHD have been developed.26
`
`Pathophysiology of acute GVHD
`Two important principles should be considered with
`respect to the pathophysiology of acute GVHD. First,
`the disease is indicative of exaggerated but typical
`infl ammatory mechanisms mediated by donor
`lymphocytes infused into the recipient, in whom they
`function appropriately
`in view of
`the
`foreign
`environment they encounter. Second, the recipient’s
`tissues that stimulate donor lymphocytes have usually
`been damaged by underlying disease, previous
`infections, and the transplant conditioning regimen.29
`As a result, these tissues produce molecules such as
`proinfl ammatory cytokines and chemokines, which
`increase
`expression
`of
`key
`receptors
`on
`antigen-presenting cells (APCs), thereby enhancing
`cross-presentation of polypeptide proteins (eg, minor
`histocompatibility antigens) to the donor immune cells
`that mediate GVHD.42–45
`Mouse models have been central to identifi cation and
`understanding of pathophysiological mechanisms of
`GVHD, and work undertaken in dogs has been vital for
`development of clinically useful strategies for GVHD
`prophylaxis and treatment advances in donor leucocyte
`infusions.36,46,47 Largely on the basis of these experimental
`data, progression of acute GVHD can be summarised in
`three sequential steps or phases: (1) activation of APCs;
`(2) donor T-cell activation, proliferation, diff erentiation,
`and migration; and
`(3)
`target
`tissue destruction
`(fi gure 3).
`The fi rst step entails activation of APCs by the
`underlying disease and the HCT conditioning regimen.
`Damaged host tissues respond by producing so-called
`danger signals, including proinfl ammatory cytokines (eg,
`TNFα and interleukins 1 and 6), chemokines, and
`amplifi ed expression of adhesion molecules, MHC
`antigens, and costimulatory molecules on host APCs.42,48–50
`Findings of a report showed that 1 week after HCT,
`increased amounts of TNFα receptor 1—a surrogate
`marker
`for TNFα—correlated strongly with
`later
`development of GVHD.51 Injury to the gastrointestinal
`tract from conditioning is especially important because it
`allows
`for
`systemic
`translocation of
`additional
`infl ammatory stimuli, such as microbial products
`including lipopolysaccharide or other pathogen-associated
`molecular patterns, that further enhance activation of
`host APCs.49
`The secondary lymphoid tissue in the gastrointestinal
`tract is probably the initial site of interaction between
`activated APCs and donor T cells.52 These observations
`have led to an important clinical strategy to diminish
`acute GVHD by reducing the intensity of the conditioning
`regimen.53,54 Experimental GVHD can also be decreased
`
`1552
`
`www.thelancet.com Vol 373 May 2, 2009
`
`

`

`I
`
`Seminar
`
`by manipulation of distinct subsets of APCs.55,56
`Furthermore, non-haemopoietic stem cells, such as
`mesenchymal stromal cells, can reduce allogeneic T-cell
`responses and ameliorate GVHD, although
`the
`mechanism for such inhibition remains unclear.57
`The idea that amplifi ed activation of host APCs
`increases the risk for acute GVHD unifi es several
`seemingly disparate clinical associations with that risk,
`such as advanced stages of malignant disease, more
`intense transplant conditioning regimens, and history of
`viral infection. APCs detect infections with receptors on
`their cell surfaces, such as Toll-like receptors, which
`recognise conserved molecular patterns of microbes.27,58
`Toll-like receptors specifi c for viral DNA or RNA activate
`APCs and could boost GVHD, providing a potential
`mechanistic basis for enhanced disease associated with
`viral infections such as cytomegalovirus.59
`The core of the graft-versus-host reaction is the second
`step, in which donor T cells proliferate and diff erentiate
`in response to host APCs (fi gure 3). The danger signals
`generated in the fi rst phase augment this activation, at
`least in part, by increasing expression of costimulatory
`molecules.60 Blockade of costimulatory pathways to
`prevent GVHD is successful in animal models, but this
`approach has not yet been tested in large clinical trials.2
`In mouse models, in which genetic diff erences between
`donor and recipient strains can be tightly controlled,
`CD4+ cells induce acute GVHD to MHC class II
`diff erences and CD8+ cells induce acute disease to class I
`diff erences.61,62 In most HLA-identical HCTs, both CD4+
`and CD8+ subsets respond to minor histocompatibility
`antigens and can cause GVHD
`in HLA-identical
`procedures.
`Regulatory T cells can suppress proliferation of
`conventional T cells and prevent GVHD in animal
`models when added to donor grafts containing con-
`ventional T cells,63 but use of regulatory T cells in clinical
`acute GVHD will need enhanced techniques to identify
`and expand them. Natural killer T-cell 1.1+ subsets from
`the host and donors have also been shown to modulate
`acute GVHD.64 In a clinical trial of total lymphoid
`irradiation (as conditioning), GVHD was reduced
`signifi cantly and host natural killer T-cell function was
`amplifi ed.65
`Activation of immune cells results in rapid intracellular
`biochemical cascades that induce transcription of genes
`for many proteins,
`including cytokines and their
`receptors. T-helper 1 cytokines (interferon γ, interleukin 2,
`and TNFα) are released in large amounts during acute
`GVHD. Production of interleukin 2 by donor T cells
`remains the main target of many current clinical
`therapeutic and prophylactic approaches to GVHD, such
`as cyclosporine, tacrolimus, and monoclonal antibodies
`directed against this cytokine and its receptor.9 However,
`emerging data indicate an important role for interleukin 2
`in the generation and maintenance of CD4+CD25+
`regulatory T cells, suggesting that prolonged interference
`
`Figure 2: Lichenoid changes of buccal mucosa in chronic GVHD
`Photograph courtesy of J Ferrara and J Levine.
`
`with this cytokine could unintentionally stop development
`of long-term tolerance after allogeneic HCT.66
`Interferon γ has many functions and can either amplify
`or reduce GVHD.67,68 It could boost disease by increasing
`expression of molecules such as chemokine receptors,
`MHC proteins, and adhesion molecules; it also raises
`the sensitivity of monocytes and macrophages to stimuli
`such as lipopolysaccharide and accelerates intracellular
`cascades in response to these stimuli.69 Early polarisation
`of donor T cells so that they secrete less interferon γ and
`more interleukin 4 can also attenuate experimental acute
`GVHD.70 Interferon γ might amplify GVHD by direct
`damage to epithelium in the gastrointestinal tract and
`skin and by induction of immunosuppression by
`generation of nitric oxide.71 By contrast, this cytokine
`could suppress GVHD by hastening apoptosis of
`activated donor T cells.68,72 This complexity means
`manipulation of interferon γ could have diverse eff ects
`in vivo, making the cytokine a challenging target with
`respect to therapeutic intervention.
`Interleukin 10 has a key role in suppression of immune
`responses, and clinical data suggest it might regulate
`acute GVHD.17 Transforming growth factor β, another
`suppressive cytokine, can subdue acute GVHD but
`exacerbate chronic disease.73 Thus, timing and duration
`of secretion of any given cytokine could establish the
`specifi c eff ects of that molecule on GVHD severity.
`
`www.thelancet.com Vol 373 May 2, 2009
`
`1553
`
`

`

`I
`
`Seminar
`
`Conditioning: tissue damage
`
`(1) Host APC
`(1) HostAPC
`activation
`activation
`
`Host
`tissues
`
`TNFα
`IL1
`LPS
`
`Small
`intestine
`
`LPS
`
`--- --►
`.. .... -..
`..
`..
`..
`..
`
`IFNγ
`
`;
`
`;
`
`;
`
`;
`
`;
`
`;
`I
`
`Mφ
`
`TNFα
`IL1
`
`CD4
`CTL
`
`TNFα
`IL1
`
`CD8
`CTL
`
`Target cell
`apoptosis
`
`CD8
`CTL
`
`(3) Cellular and
`inflammatory
`effectors
`
`Treg
`
`T cell
`
`Treg
`
`(2) Donor T-cell
`activation
`
`Th1
`
`Figure 3: Pathophysiology of acute GVHD
`IL 1=interleukin 1. IFN γ=interferon γ. LPS=lipopolysaccharide. Treg=regulatory T cell. Th1=T-helper 1 cell. CTL=cytotoxic T lymphocyte.
`
`The third eff ector phase of the graft-versus-host process
`(fi gure 3) is a complex cascade of cellular mediators (such
`as cytotoxic T lymphocytes and natural killer cells) and
`soluble infl ammatory agents (eg, TNFα, interferon γ,
`interleukin 1, and nitric oxide).2,29 These molecules work
`synergetically to amplify local tissue injury and further
`promote infl ammation and target tissue destruction.
`The cellular eff ectors of acute GVHD are mainly
`cytotoxic T lymphocytes and natural killer cells.49 Cytotoxic
`T lymphocytes that prefer to use the Fas and FasL pathway
`of target lysis seem to predominate in GVHD liver
`damage (hepatocytes express large amounts of Fas)
`whereas cells that use the perforin and granzyme
`pathways are more important in the gastrointestinal tract
`and skin.2,74 Chemokines direct migration of donor T cells
`from lymphoid tissues to the target organs in which they
`cause damage. Macrophage infl ammatory protein 1α and
`other chemokines (such as CCL2–CCL5, CXCL2, CXCL9,
`CXCL10, CXCL11, CCL17, and CCL27) are overexpressed
`and enhance homing of cellular eff ectors to target organs
`during experimental GVHD.75 Expression of integrins,
`such as α4β7 and its ligand MADCAM1, is also important
`
`for homing of donor T cells to Peyer’s patches during
`intestinal GVHD.52,76,77
`Microbial products such as lipopolysaccharide, which
`leak through damaged intestinal mucosa or skin, can
`stimulate secretion of infl ammatory cytokines through
`Toll-like receptors.49,78 The gastrointestinal
`tract
`is
`especially susceptible to damage from TNFα, and the
`gastrointestinal tract has a major role in amplifi cation
`and propagation of the cytokine storm characteristic of
`acute GVHD.49 TNFα can be produced by both donor and
`host cells and it acts in three diff erent ways: (1) it activates
`APCs and enhances alloantigen presentation; (2) it
`recruits eff ector cells to target organs via induction of
`infl ammatory chemokines; and (3) it directly causes
`tissue necrosis (as its name suggests).79–81
`
`Prevention of GVHD
`On the basis of evidence from animal models for the
`central role of T cells in initiation of GVHD, many clinical
`studies of T-cell depletion as prophylaxis for the disease
`were undertaken in the 1980s and 1990s. Three main
`depletion strategies were studied: (1) negative selection
`
`1554
`
`www.thelancet.com Vol 373 May 2, 2009
`
`

`

`I
`
`Seminar
`
`of T cells ex vivo; (2) positive selection of CD34+ stem
`cells ex vivo; and (3) antibodies against T cells in vivo.82
`Most approaches showed substantial limitation of both
`acute and chronic GVHD.83–85 Unfortunately, the lowest
`frequency of severe GVHD was off set by high rates of
`graft failure, relapse of malignant disease, infections,
`and Epstein-Barr virus-associated lymphoproliferative
`disorders. Negative-selection purging strategies with
`various antibodies against T cells achieved similar
`long-term results irrespective of the breadth of antibody
`specifi city.86–88 Findings of one large registry study showed
`that purging techniques that used antibodies with broad
`specifi cities produced inferior leukaemia-free survival
`than standard immunosuppression in patients receiving
`unrelated donor transplants.89
`Several research groups have investigated partial T-cell
`depletion, either by elimination of specifi c T-cell subsets
`(eg, CD8+) or by titration of the dose of T cells present in
`the inoculum.90–92 None of these approaches, however,
`has been shown convincingly to be the best strategy that
`enhances long-term survival.
`Alemtuzumab is a monoclonal antibody that binds
`CD52, a protein expressed on a broad range of leukocytes
`including lymphocytes, monocytes, and dendritic cells.
`Its use in a phase II trial of GVHD prophylaxis lowered
`incidence of
`acute
`and
`chronic GVHD
`after
`reduced-intensity transplant.93 In two prospective studies,
`patients who received alemtuzumab rather
`than
`methotrexate showed signifi cantly lower rates of acute
`and chronic GVHD,94 but they had more infectious
`complications and higher rates of relapse, so no overall
`survival benefi t was recorded. Alemtuzumab might also
`contribute to graft failure when used with minimum-
`intensity conditioning regimens.95
`An alternative strategy to T-cell depletion attempted to
`induce anergy in donor T cells by ex-vivo antibody
`blockade of costimulatory pathways before transplantation.
`Findings of a small study of this approach in patients
`undergoing haploidentical HCT was quite encouraging,
`but they have not yet been replicated.96 Thus, the focus of
`most preventive strategies remains pharmacological
`manipulation of T cells after transplant.
`Administration of antibodies against T cells in vivo as
`GVHD prophylaxis has also been tested extensively. The
`best studied drugs are anti-thymocyte globulin or
`anti-lymphocyte globulin preparations. These serum
`samples, which have high titres of polyclonal antibodies,
`are made by immunisation of horses or rabbits to
`thymocytes or lymphocytes, respectively. A complicating
`factor in establishing the role of these polyclonal serum
`samples in transplantation is the observation that even
`diff erent brands of the same class exert diverse biological
`eff ects.97 However, the side-eff ects of anti-thymocyte
`globulin and anti-lymphocyte globulin infusions are
`similar across diff erent preparations and include fever,
`chills, headache, thrombocytopenia (from cross-reactivity
`to platelets), and, infrequently, anaphylaxis.
`
`In retrospective studies, rabbit anti-thymocyte globulin
`reduced the frequency of GVHD in related-donor
`haemopoietic stem-cell transplant recipients without
`seeming to enhance survival.98,99 In patients receiving
`unrelated-donor haemopoietic stem cells, addition of
`anti-lymphocyte globulin to standard GVHD prophylaxis
`prevented severe GVHD eff ectively but did not result in
`better survival because of increased infections.86 In a
`long-term
`follow-up study, however, pretransplant
`anti-thymocyte globulin provided signifi cant protection
`against extensive chronic GVHD and chronic lung
`dysfunction.100
`The primary pharmacological strategy to prevent GVHD
`is inhibition of the cytoplasmic enzyme calcineurin,
`which is important for activation of T cells. The calcineurin
`inhibitors cyclosporine and tacrolimus have similar
`mechanisms of action, clinical eff ectiveness, and toxic
`eff ects, including hypomagnesaemia, hyperkalaemia,
`hypertension, and nephrotoxicity.9,101 Serious side-eff ects
`include transplant-associated thrombotic microangio-
`pathy and neurotoxic eff ects that can lead to premature
`discontinuation. Although clinically similar to thrombotic
`thrombocytopenic
`purpura,
`transplant-associated
`thrombotic micro angio pathy does not respond reliably to
`therapeutic plasmapheresis, carries a high mortality rate,
`and removal of the off ending agent does not always result
`in improvement.102 Posterior reversible encephalopathy
`syndrome includes mental status changes, seizures,
`neurological defi cits, and characteristic fi ndings on MRI;
`this syndrome has been seen in 1–2% of patients
`undergoing HCT and taking calcineurin inhibitors.103
`Side-eff ects of these drugs fall as the dose is tapered,
`usually 2–4 months after transplantation.
`Calcineurin inhibitors are usually administered in
`combination with other immunosuppressants, such as
`methotrexate, which is given at low doses in the early
`post-transplant period.9,101 The toxic eff ects of methotrexate
`(neutropenia and mucositis) have led some investigators
`to replace it with mycophenolate mofetil. In a prospective
`randomised trial, patients who received mycophenolate
`mofetil as part of GVHD prophylaxis had signifi cantly less
`severe mucositis and more rapid neutrophil engraftment
`than did those who received methotrexate.104 Frequency
`and severity of acute GVHD was similar between the two
`groups, but the study closed early because of superiority of
`the mycophenolate mofetil arm with respect to reduced
`mucositis and speed of haemopoietic engraftment. A
`desire for faster neutrophil engraftment has led to use of
`mycophenolate mofetil in umbilical-cord blood transplants
`for which graft failure is a major concern.105 This drug is
`also sometimes used after reduced-intensity conditioning
`regimens for similar reasons.106,107
`Sirolimus is an immunosuppressant that is structurally
`similar to tacrolimus but does not inhibit calcineurin. In
`phase II trials, sirolimus was very eff ective in combination
`with tacrolimus;108,109 the drug damages endothelial cells,
`however, and it might enhance transplant-associated
`
`www.thelancet.com Vol 373 May 2, 2009
`
`1555
`
`

`

`I
`
`Seminar
`
`thrombotic microangiopathy, which is associated with
`calcineurin inhibitors.110 The combination of tacrolimus
`and sirolimus is currently being compared in a large
`randomised multicentre trial.
`Reduced-intensity conditioning regimens attempt to
`suppress the host immune system suffi ciently so
`that donor T cells can engraft and then ablate the
`lymphohaemopoietic compartment of the recipient.
`The term non-myeloablative is therefore somewhat
`misleading. Reduced-intensity conditioning regimens
`diminish tissue damage and lead to decreased amounts
`of infl ammatory cytokines, which are important in the
`initiation of GVHD pathophysiology; this eff ect could
`account for the reduced frequency of severe GVHD after
`reduced-intensity conditioning versus
`full-intensity
`conditioning used in historical controls.53,54,93,111 Onset of
`acute GVHD can be delayed after reduced-intensity
`conditioning until after day 100, however, and acute
`disease could present simultaneously with elements of
`chronic GVHD (known as overlap syndrome).111–113
`
`Treatment of acute GVHD
`GVHD fi rst develops, generally, in the second month
`after HCT during calcineurin-based prophylaxis.114
`Steroids, with
`their potent anti-lymphocyte and
`anti-infl ammatory activity, are the gold standard for
`treatment of GVHD. Many centres treat mild GVHD of
`the skin (grade I) with topical steroids alone, but for more
`severe disease and any degree of visceral GVHD
`involvement high-dose systemic steroids are usually
`initiated. Administration of steroids results in complete
`remission in less than half of patients,115 and more severe
`GVHD is less likely to respond to treatment.116 In a
`prospective randomised study, addition of anti-thymocyte
`globulin to steroids as primary treatment did not increase
`the response rate.116 In a retrospective study, use of
`anti-thymocyte globulin in patients who showed early
`signs of steroid resistance was benefi cial,115 but not all
`study fi ndings show such benefi t, and this antibody
`preparation is not used as standard because of raised
`infection risks.100,117 Infusion of mesenchymal stromal
`cells—expanded in culture either from the original HCT
`donor or from a third party—is a promising approach,
`which produced 55% complete responses in a phase II
`study of patients with steroid-resistant GVHD.57
`An increasingly frequent treatment for GVHD is
`extracorporeal photopheresis. During this procedure, the
`patient’s white blood cells are gathered by apheresis,
`incubated
`with
`the DNA-intercalating
`agent
`8-methoxypsoralen, exposed to ultraviolet light, and
`returned to the patient. Extracorporeal photopheresis is
`known to induce cellular apoptosis, which has strong
`anti-infl ammatory eff ects in several systems, including
`prevention of rejection of solid organ grafts.118 Work done
`in animals shows that extracorporeal photopheresis
`reverses acute GVHD by increasing the number of
`regulatory T cells.119 Data from a phase II clinical study of
`
`steroid-dependent or steroid-refractory GVHD showed
`resolution of disease in most patients, with 50% long-term
`survival in this very-high-risk group.120 Randomised
`multicentre studies of this approach are needed to
`establish its place in management of acute GVHD.
`Another strategy to treat GVHD is blockade of the
`infl ammatory cytokine TNFα. TNFα can activate APCs,
`recruit eff ector cells, and cause direct tissue damage (see
`Pathophysiology of acute GVHD).121 Data from a phase II
`trial of etanercept (solubilised TNFα receptor 2) showed
`signifi cant eff ectiveness of the drug when added to
`systemic steroids as primary treatment for acute GVHD.
`70% of patients had complete resolution of all GVHD
`symptoms within 1 month, with 80% complete responses
`in the gastrointestinal tract and skin. The researchers also
`reported that concent

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