throbber

`
`For reprint orders, please contact reprints@expert-reviews.com
`
`|
`EXPERT
`| REVIEWS
`
`Current and future approaches
`for control of graft-versus-
`host disease
`
`Expert Rev, Hematol. 1(1), 111-128 (2008)
`
`John Koreth’ and
`Joseph H Antin
`‘Author for correspondence
`Division of Hematologic
`Malignancies, Dana Farber
`Cancer Institue, 44 Binney
`Street, Boston, MA 02115, USA
`Tel. +1 617 632 3470
`Fax: +7 617 632 5168
`john_koreth@dfci. harvard.edu
`
`Graft-versus-host disease (GVHD), both acute and chronic, remains one of the major barriers
`to improving outcomesafterallogeneic stem cell transplantation. The pathophysiology of GVHD
`is complex and incompletely understood. GVHD is believed to arise from the interaction of:
`tissue damage and proinflammatory cytokines causing activation of antigen-presenting cells
`(APCs, donor T-cell activation by APCs and cytokines and host tissue injury by effector
`T lymphocytes and proinflammatory cytokines. There is also a role for additional lymphocyte
`subtypes (naive and memoryT cells, regulatory T cells, naturalkiller T cells and B cells) in GVHD
`pathogenesis, Strategies to improve donor-recipient HLA match, and to minimize conditioning
`toxicity, cytokine release and APC and effector T-lymphocyte activation, will likely improve
`prophylaxis of acute (and possibly chronic) GVHD. Therapy of established acute and chronic
`GVHD isstill heavily dependent on corticosteroids, despite their limited efficacy and considerable
`toxicity. Novel agents (and/or combinations of agents) comprising pharmacologic, biologic and
`cellular therapies targeting specific steps or subsets involved in immune activation will
`likely
`comprise future advances in GVHD control. This article reviews the current state of knowledge
`regarding the prevention and treatment of acute and chronic GVHD. Novel approaches currently
`undergoing evaluation are also highlighted.
`
`Keyworops: allogeneic stem cell transplantation © graft-versus-host disease
`
`Allogeneic stem cell transplantation (alloSCT) is
`often the only curative option for patients with
`various hematologic and/or immune disorders,
`particularly those with aggressive or advanced
`hematologic malignancies. However, the toxic-
`ity of alloSCT remains a significant barrier to
`its wider utilization. Graft-versus-host disease
`(GVHD)remainsthe most frequent complication
`after alloSCT.
`Clinically, GVHD was categorized as ‘acute’
`and‘chronic’ based on time of presentation, Any
`GVHD before day 100 was knownas ‘acute’,
`and after day 100 it was known as ‘chronic’. The
`severity of GVHD was graded: acute GVHD
`was categorized as grade I-IV by modified
`Glucksberg criteria (A—D by the International
`Bone Marrow Transplant Registry index) (Tarte 1)
`1,2]; chronic GVHD was commonlycategorized
`as limited or extensive [3]. Based on these criteria,
`grade [I-IV acute GVHD is thought to occur
`in approximately 35%of recipients of matched,
`related donor transplants, and in up to 50%of
`unrelated or alternative donor transplantrecipi-
`ents. Chronic GVHD can affect up to 60%of
`
`recipients who survive beyond 100 days after
`matched donor alloSCT.
`While the simplicity of the day 100 definition
`is appealing, it is irrelevant biologically and clini-
`cally, For instance, in patients receiving reduced
`intensity conditioning (RIC) alloSCT,or after
`donor lymphocyte infusion (DLN),clinical acute
`GVHD may develop months after the proce-
`dure [4]. Hence, there is a current attempt by the
`National Institutes of Health chronic GVHD
`consensus project working group to reclassify
`acute GVHD into classic acute and late-onset
`acute; and chronic GHVDinto classic chronic
`and overlap syndrome[5], Classic acute GVHD
`is characterized by a maculopapular erythematous
`skin rash, gastrointestinal symptomsorcholestatic
`hepatic abnormalities occurring within 100 days
`ofalloSCT or DLI, while late acute GVHD pres-
`ents similarly beyond 100 days after alloSCT or
`DLI, Classic chronic GVHD consists solely of
`manifestations ascribable to chronic GVHD
`(without acute GVHD features) (Tanz 2), while
`overlap syndromehas clinical features of both
`acute and chronic GVHD occurring together.
`
`www.expert-reviews.com
`
`10.1586/17474086.1.1.111
`
`© 2008 Expert Reviews Lrd
`
`ISSN 1747-4086
`
`111
`
`SAN EX 1024, Page 01
`
`SAN EX 1024, Page 01
`
`

`

`Gut Organ stage Skin*
`
`oe
`
`Table 1. Modified Glucksberg criteria for acute graft-versus-host disease grading.
`aici
`
`1
`
`Rash < 25%
`
`Rash 25-50%
`
`Rash > 50%
`
`4
`
`Generalized erythroderma with
`bullae
`
`Overall grade
`I
`ll
`
`Stage 1 or 2
`Stage 3 or
`
`-
`I
`Stage 4 or
`Iv
`“Use ‘rule of nines’ to determine body surface area
`Data from [I].
`
`Risk factors for GVHD
`Therisk factors for GVHD include:
`
`* Donor-recipient match at the major histocompatibility com-
`plex (MHC) loci, for instance, HLA class I (HLA-A, -B and
`-C) and class Il (HLA-DR, -DP and -DQ). Mismatches at
`HLA-A,-B, -C or -DRB1 (and possibly also -DQ and -DP)
`increase the risk of GVHD (nonpermissive donor—recipient
`HLA mismatches may particularly influence GVHD severity)
`and negatively impact survival [6-10];
`Donor stem cell source: compared with bone marrow stem
`cells, peripheral blood stem cells (PBSCs) have a higher GVHD
`risk, while umbilical cord blood cells appear to have a lower
`risk [11-14];
`
`T-cell dose: compared with T-cell replete grafts, 2-3 log deple-
`tion of CD3* T lymphocytes ofthe graft can effectively reduce
`acute GVHD incidence (although the effect on chronic GVHD
`is less clear), while less-intensive log reductions of T cells have
`no significant impact [15.16]. However, the benefit of T-cell
`depletion is counteracted by increased risks of graft failure,
`opportunistic infection and disease relapse such that pan-T-cell
`depletion strategies are not currently favored [17];
`* Additional risk factors include donor and recipient age,
`donor-recipient sex mismatch (female donorto malerecipient),
`donorparity and allosensitization, disease stage and intensity
`ofconditioning (for acute GVHD). Acute GVHD isa powerful
`predictor of chronic GVHD risk [18].
`Measures to reduce GVHD risk would therefore include
`improvements in donorselection, improved HLA matching, as
`well as reduced intensity conditioning where possible. However,
`other trends, such as the increased use ofdonor PBSCsas a source
`of stem cells, extending alloSCT to older/sicker patients and the
`use of alternative donors (haploidentical and HLA-mismatched
`donors), suggest that GVHD control will remain a significant
`issue for the foreseeable future.
`
`112
`
`Bilirubin 2-2.9 mg/dl
`
`Diarrhea 500-1000cc/d or biopsy-proven upper GI
`involvement
`
`Bilirubin 3-6 mg/dl
`
`Bilirubin 6.1-15 mg/dl
`
`Bilirubin > 15 mg/dl
`
`None
`Stage 1 or
`
`Stage 2 or 3 or
`Stage 4
`
`Diarrhea 1000-1500cc/d
`
`Diarrhea 1500-2000cc/d
`
`Diarrhea > 2000 cc/d or severe abdominal pain with or
`without ileus
`
`None
`
`Stage 1
`
`Stage 2-4
`
`Etiopathogenesis of GVHD
`The etiology of GVHD is complex, but Billingham’s criteria
`still apply [19]. First, the graft must contain immunologically
`competent cells (T lymphocytes and possibly B lymphocytes).
`Second, the recipient must be incapable of rejecting the trans-
`plantedcells (achieved by conditioning chemotherapyorradia-
`tion). Third, the recipient must express tissue antigens that are
`not present in the donor (major or minor histocompatibility
`mismatch).
`Ourcurrent understanding ofacute GVHD,although incom-
`plete, is better than that of chronic GVHD.In part, this is due to
`the better availability of mouse models of acute GVHD.Broadly
`however, both forms of GVHD are believed to be caused by
`similar alloimmune responses that also underlie the beneficial
`graft-versus-leukemia (GVL) effect. Maintaining control of
`GVHD,while enabling the curative GVL response remains the
`holy grail of allorransplantation.
`The development of acute GVHD is frequently divided into
`three phases (Ficurr 1):
`
`* Tissue damage, owing to underlying disease, infections and
`conditioning regimentoxicity, resulting in leakage ofbacterial
`lipopolysaccharides across the damaged gut epithelium and a
`‘cytokine storm’ with the production of inflammatorycytok-
`ines, such as TNF-a, and IL-1 by injured cells, resulting in
`secondary changes in expression of adhesion molecules, MHC
`antigens and chemokines, which can act as danger signals
`and activate residual host and donor antigen-presenting cells
`{APCs) [20-24]. APC activation can occur via both Toll-like
`receptor (TLR) and non-TLR (e.g., nucleotide-binding
`oligomerization domain [NOD]) pathways(25.26);
`
`Donor T-cell activation, cytokine release, proliferation and
`tissue localization occurs in the context of the proinflamma-
`tory post-transplant milieu and after alloantigen presentation
`and costimulation by APCs (donor or host) [27-30];
`
`Expert Rew, Hematol. 1(1), (2008)
`
`SAN EX 1024, Page 02
`
`SAN EX 1024, Page 02
`
`

`

`|
`
`Current &future approachesforcontrolof graft-versus-hostdisease
`
` Table 2. Definite and probable manifestations of chronic graft-versus-host disease.
`
`Skin
`
`Mucous membranes
`
`GI tract
`
`Liver
`
`Scleroderma (superficial or fasciitis), lichen planus,
`vitiligo, scarring alopecia, hyperkeratosis pilaris,
`contractures from skin immobility, nail bed dysplasia
`
`Lichen planus, noninfectious ulcers, corneal erosions/
`noninfectious conjunctivitis
`
`Esophageal strictures, steatorrhea
`
`None
`
`GU tract
`Musculoskeletal/serosa
`
`Vaginal stricture,lichen planus
`
`Nonseptic arthritis, myositis, myasthenia, polyserositis,
`contractures from joint immobilization
`
`Hematologic
`
`None
`
`Lung
`
`Bronchiolitis obliterans
`
`GI: Gastrointestinal; GU: Genitourinary; GVHD: Graft-versus-host disease.
`
`* The effector phase of GVHD target organ damageinvolves a
`complex interaction of cytokine and cellular effectors, Cyto-
`toxic T lymphocytes (CTLs), both CD4" and CD8’, are the
`major cellular effectors of GVHD and cause cell death by a
`variety of pathways, such as Fas—Fas ligand (FasL), TNF recep-
`tor (TNFR)-like death receptors (e.g., TRAIL and TWEAK)
`and perforin—granzyme[31-36]. Inflammatory cytokines, such
`as TNF-a and IL-l, synergize with CTLs and can also act
`directly to promote tissue injury and inflammation in GVHD
`target organs [37-40].
`
`Eczematoid rash, dry skin, maculopapular rash, hair
`loss, hyperpigmentation
`
`Xerostomia, keratoconjunctivitis sicca
`
`Anorexia, malabsorption, weight loss, diarrhea,
`abdominal pain
`
`Elevation of alkaline phosphatase, transaminitis,
`cholangitis, hyperbilirubinemia
`
`Noninfectious vaginitis, vaginal atrophy
`Arthralgia
`
`Thrombocytopenia, eosinophilia, autoimmune
`cytopenias
`
`Bronchiolitis obliterans with organizing pneumonia,
`interstitial pneumonitis
`
`after alloSCT [59-1]. Similarly, polymorphisms in the non-TLR
`(NOD) pathway of adaptive immuneactivation can impact
`GVHD risk [62]. Genes inyolved in drug metabolism have also
`been linked to toxicity and GVHD after alloSCT [e3,64]. Finally,
`genes with onlyindirect associations with immuneactivity have
`also been linked to GVHD (és-67]. Both donor and recipient
`polymorphismsare often relevant with regards to GVHD risk,
`as in the case of IL-10 [eg].
`There is increasing awareness ofthe role of additional cellular
`subsets in GVHD:
`
`¢ Naive and memoryT cells: naive (CD62L") T cells, but not
`memory (CD62L))T cells, are often considered to have allore-
`active potential that can result in acute GVHD [69,70]. However,
`contrasting recentdataalso suggest a role for alloreactive mem-
`otyT cells and their precursor stem cells in the development of
`GVHD [71,72];
`
`Based on their cytokine expression pattern, there are at least
`two types of T helper (Th) effector cells involved in GVHD:
`Thl and Th2 cells. Th cells generate IL-2, TNF-a and IFN-¥,
`while Th2 cells produce IL-4 and IL-10. While the ‘cytokine
`storm’ phase of GVHD, which is amplified by Th1 cytokines,
`correlates with the development of acute GVHD, cytokines
`that polarize donor T cells to Th2 (e.g., granulocyte colony-
`stimulating factor [G-CSF], IL-4 and IL-18) can reduce acute
`GVHD [ai-44], However, this model may be an oversimplifica-
`tion, as Thl and Th2 subsets can each cause injury to distinct
`GVHD target organs in some mouse models of acute GVHD [as].
`Additional complexities involve possible roles for newly identi-
`fied Th17 cells in GVHD and the interaction between Th17
`effector cells and peripheral regulatory T cells (Tregs), given
`their alternate developmentalfates from commonnaive precursor
`T cells [46-48].
`Additionally, genetic polymorphismsthar lead to altered
`cytokine expressionlevels (e.g., IL-6, IL-10 and TNF-a) have
`also been linked to differences in acute and chronic GVHD
`incidence [49-58]. Furthermore, polymorphismsinvolving natural
`* NKTcells: host NKT cells also have immune suppressive
`killer (NK) cell receptor/ligand complex, collectively termed
`effects that can control GVHD in an IL-4-dependentfashion
`the killer immunoglobulin-like receptor family (KIR), have
`[30,81]. Human clinical data suggest that enhancing recipient
`been linked to differences in both GVHD and relapse rates
`
`* Tregs: CD4'CD25' FoxP3' Tregs from the donor have been
`shown to suppress the expansion of alloreactive donor T cells
`and the development of GVHD, without abrogating GVL in
`this MHC-mismatched murine model [73]. IL-2, initially iden-
`tified as a lymphocyte growth factor and thoughtprimarily to
`promote effector T-cell responses in vivo, is now identified as
`acytokinecritical for the development, expansion and activity
`of ‘regs [74,75]. In humans, FoxP3 mRNAlevels (considered a
`relatively specific marker for Tregs) wassignificantlydecreased
`in patients with GVHD [76,77]. The expression ofthe cell sur-
`face marker CD62L wasalso found to becritical for the ability
`of donor Tregs to control GVHD [73,79];
`
`WWW.expert-reviews.com
`
`113
`
`SAN EX 1024, Page 03
`
`SAN EX 1024, Page 03
`
`

`

`Koreth & Antin
`
`(I) Recipient conditioning
`tissue imaging
`
`|
`
`Small
`intestine
`
`aN ees
`IL-1
`
`Target cell
`apoptosis
`
`(II) Donor T-cell=\
`
`IL-2
`
`Prophylaxis of GVHD
`Pilot studies omitting GVHD prophylaxis
`indicated an acute GVHD incidence of
`nearly 100% [s9]. Studies using methotrex-
`ate as a single agent for GVHD prophylaxis
`via inhibition of rapidly dividing alloreac-
`tive T cells, indicated an acute grade II-IV
`GVHD rate of over 50%, even in the set-
`ting of HLA-matched sibling donors [90].
`The introduction of a calcineurin inhibi-
`tor, cyclosporine (and subsequently tac-
`rolimus), represented the next advance in
`the prevention of GVHD, with improved
`efficacy in GVHD control compared with
`methotrexate [91-93]. Cyclosporine and tac-
`rolimus bound to cyclophilin or FK-binding
`protein 12 (FKBP12), respectively, inhibit
`calcineurin (a protein phosphatase that is
`calcium- and calmodulin-dependent) and
`prevent the dephosphorylation and nuclear
`translocation of the transcription factor
`nuclear factor of activated T cells (NFAT).
`By blocking NFAT,one of the most impor-
`tant regulators of cytokine gene transcrip-
`tion following T-cell activation, calcineu-
`rin inhibitors block T-cell activation and
`proliferation (94,95). The combination of
`calcineurin inhibitor (cyclosporine) and
`methotrexate was moreeffective than either
`agentalone, with grade I-IV acure GVHD
`rates of 20-56% after HLA-matched sib-
`ling alloSCT (96,97), Compared with
`cyclosporine, tacrolimus has an improved
`toxicity profile and, more importantly,
`randomized data indicate improved acute
`GVHD prophylaxis in both HLA-matched siblings and unrelated
`donor allotransplants [93,99]. The length of immunosuppressive
`therapy appears to have no role in improving control of chronic
`GVHD. Patients with acute GV HD orbiopsy evidence of subclini-
`cal acute GVHD were randomly assignedto 6 versus 24 months of
`cyclosporine therapy. Therates ofclinical extensive chronic GVHD
`were 39 and 51%, respectively, a nonsignificant difference [100].
`Similarly, the presence or absence ofday 11 methotrexate does not
`likely impact chronic GVHD rates[101.102].
`Corticosteroids, the mainstay of therapy for established acute
`GVHD,do not have a significant role in GVHD prophylaxis.
`Various trials compared prednisone and cyclosporine to the three-
`drug combination ofmethotrexate, cyclosporine and prednisone.
`In one large trial, the acute GVHD rate in the cyclosporine
`and prednisone control arm was 23%, compared with only 9%
`in the three-drug arm of methotrexate, cyclosporine and pred-
`nisone [103]. However, subsequenttrials could not demonstrate
`similarly improved GVHD control, or improved long-term out-
`comes with the three-drug combination, and, currently, steroids
`are not routinely used in GVHD prophylaxis [104].
`
`activation
`
`~S IFN-y
`
`~ NK
`
`Tw sictetol
`
`elea uture Science GroupLtd (2008)
`
`Figure 1, Etiopathogenesis of acute graft-versus-host disease,
`Modified with permission from [245].
`
`NKTcells by total lymphoid irradiation (TLD in conjunction
`with anti-thymocyte globulin-based conditioning similarly
`promoted Th2 polarization and significantly reduced GVHD
`[82]. However, it is important to note that NKTcells are
`heterogeneous and their roles in GVHD are incompletely
`understood;
`
`B cells: traditionally, a major role for B cells and humoral
`immunityin the development of GVHD has not been con-
`sidered. However, recent work suggests that, in the context
`of matched sibling PBSC allotransplantation, the concentra-
`tion of CD20" B cells in the apheresis product may predict
`the development of acure GVHD [33]. Additionally, auto- and
`alloantibodies have been described in chronic GVHD, some
`of which may play a direct role in disease progression (e.g.,
`activating PDGF receptor antibodies) [84-37]. High circulat-
`ing levels of B-cell activation factor at 6-months post-trans-
`plant were a predictor of subsequent chronic GVHD, further
`supporting a role for B-cell dysfunction in chronic GVHD
`[38]. The role of humoral immunity in GVHD remains an
`area of controversy and further investigation.
`
`114
`
`Expert Rew, Hematol. 1(1), (2008)
`
`SAN EX 1024, Page 04
`
`SAN EX 1024, Page 04
`
`

`

` Current &future approachesforcontrol of graft-versus-hostdisease|§\=))=) |
`
`Cyclophosphamide has been used post-transplant since the
`1980s for GVHD prevention and act via inhibition of rapidly
`dividing T cells (in a manner similar to methotrexate) [195].
`Stem cells contain high levels of aldehyde dehydrogenase that
`converts the active metabolite 4-hydroxycyclophosphamide to
`an inactive nonalkylating metabolite, thus protecting the stem
`cell from the antiproliferative activity of the agent. Similarly, the
`gutepithelium has high levels of aldehyde dehydrogenasethatis
`protective against excess mucosal toxicity despite prior intensive
`conditioning, Used as a single agent after myeloablative condi-
`tioning in related and unrelated allotransplants, the grade [I-IV
`acute GVHD rate was 41%, with few late infections, attributed
`to the brief duration of immune suppressive therapy [100]. It is
`also currently being evaluated for alternative donor transplants
`(haploidentical donor) [107].
`Mycophenolate mofetil (MMF)isa potent, selective, noncom-
`petitive reversible inhibitor of inosine monophosphate dehydro-
`genase that inhibits the de move pathway ofguanosine nucleotide
`synthesis. It has potent cytostatic effects on lymphocytes (both
`T and B) whose proliferation is dependent on de novo purine
`synthesis. With good oral bioavailability, the optimal dosing
`interval remains uncertain, usually two- to three-times daily.
`It has been used for GVHD prophylaxis in various combina-
`tions (usually with a calcineurin inhibitor + methotrexate), The
`incidence of grade II-IV acute GVHD has ranged between 38
`and 62% [108,109]. In a single-center randomized study, the com-
`bination of cyclosporine plus MMF was associated with faster
`engraftment and reduced mucositis incidence, but with similar
`incidence of acute and chronic GVHD andsurvival comparable
`to cyclosporine plus methotrexate, possibly affected by limited
`sample size and follow-up duration for these secondary end
`points[110], Longer-term use of cyclosporine in combination with
`MMF after RIC alloSCT with matched related donors did not
`impact the rates of acute grade II-IV or chronic GVHD [u1).
`Sirolimus (also called rapamycin) binds uniquely to FKBP12
`and forms a complex with mammalian target of rapamycin
`(mTOR) that interacts with various upstream pathways includ-
`ing PTEN/PI3 kinase/Akt pathway and the Janus kinase path-
`way [112.113]. The sirolimus-mTOR complex inhibits several
`biochemical pathways, resulting in reduction of DNAtranscrip-
`tion/translation, protein synthesis and cell cycle progression,
`whichresults in T-cell immunosuppression [114.115]. Interestingly,
`there is apparentdifferential inhibition ofT-cell subsets, possibly
`involving selective inhibition of Thl cell responses, and sparing
`of Th2 and ‘Treg activity [116-120]. Despite theoretical concerns
`for competition for FKBP binding with calcineurin inhibitors,
`these agents appear to work synergistically, and sirolimus does
`not interact with calcineurin or its downstream effectors [112].
`In contrast to calcineurin inhibitors, sirolimus may also exert
`its immunosuppressive effects through suppression of APC
`activity via a reduction in antigen uptake, cellular processing,
`intracellular signaling and induction of apoptosis [121-123]. The
`combination ofsirolimus and tacrolimus appears more effective
`thansirolimus plus cyclosporine in reducingalloreactive memory
`T-cell production, abrogation of effector CTL induction and
`
`WWW.expert-reviews.com
`
`apoptosis induction [124]. Single-institution clinical studies of
`sirolimus and tacrolimus with and without low-dose methotrex-
`ate for GVHD prophylaxis after myeloablative conditioning with
`cyclophosphamide/total-body irradiation (TBI) indicate excel-
`lent efficacy and acceptable toxicity in the matched related and
`unrelated donor context, with grade [I-IV acute GVHD rates
`of 19 and 23%, respectively[125]. The rates of chronic GVHD,
`however, were not significantly impacted. Similar efficacy in
`acute GVHD control was noted despite omitting low-dose meth-
`otrexate, and toxicity was further reduced (126). Similar low-acute
`GVHD rates were also noted in the context of RIC. Other recent
`single-institution reports indicate concordant as well as variant
`estimates ofsirolimus efficacy for GVHD prophylaxis in the
`myeloablative alloSCT context [127.123]. Sirolimus plus tacroli-
`musis currently being evaluated in a Phase II multi-institution
`context in comparison to methotrexate plus tacrolimus.
`Biologic agents have also been evaluated for GVHD prophy-
`laxis. Jn vivo T-cell depletion with horse- or rabbit-derived poly-
`clonal antithymocyte globulin (ATG) has been evaluated for
`prevention of GVHD,as initially proposed by Ramseyet al. [129].
`Such agents administered pre- and peritransplant can simultane-
`ously target host and donor T cells to control both graft rejection
`and GVHD [130-132]. However, additional cellular components,
`such as B cells, NK cells and APCs, can also be affected by
`polyspecific antibodies. Their use does appear to reduce the inci-
`dence of chronic GVHD and chronic lung dysfunction, with
`improved late transplant-related mortality [133]. Whether the
`reduction in chronic GVHD is also associated with increased
`disease relapse remainsto be determined. Higher doses of rabbit
`ATG (thymoglobulin) are associated with increased infections
`that can abrogate its positive impact on GVHD [134]. TLI in
`conjunction with ATG-based conditioning also significantly
`reduced GVHD [2].
`Monoclonal antibodies, such as alemtuzumab (Campath-1H;
`anti-CD52), are widely used for i# vivo GVHD prophylaxis.
`It has been found to reduce GVHD and nonrelapse mortality
`after related and unrelated transplants, and can also facilitate
`engraftment [135]. Monoclonal antibodies targeting the IL-2
`receptor (CD25) may also show benefit [136]. However, IL-2
`is also critical for Treg development, expansion and activity,
`hence IL-2 targeting in GVHD may have the unintended
`consequence of impairing Tregs that are important to control
`GVHD [74,75]. Low-dose IL-2 is currently being evaluated for
`GVHD prophylaxis. Some biologic agents that may have activ-
`ity in established active GVHD,such as IL-1 antagonists and
`ricin-conjugated CD5 antibody, do not show benefit in the
`prophylactic setting (137-141). Interestingly, rituximab, a mono-
`clonal CD20 antibodythat depletes B cells, may independently
`decrease acute GVHD risk [142]. It is also being evaluated for
`the prophylaxis of chronic GVHD.
`in vitro T-cell depletion (TCD) has also been attempted to
`control GVHD,with some success in controlling acute (and pos-
`sibly chronic) GVHD. However, in a randomized study compar-
`ing GVHD prophylaxis with approximately 1-log TCD (with
`monoclonal antibody T10B9 targeting the T-cell receptor) plus
`
`115
`
`SAN EX 1024, Page 05
`
`SAN EX 1024, Page 05
`
`

`

` oe
`
`cyclosporine versus methotrexate and cyclosporine, improved
`acute GVHD control did not lead to improvement in long-term
`survival, and disease relapse and infection risk was significantly
`increased after T-cell depletion [16,143]. In an attempt to circum-
`vent problems associated with global TCD,selective depletion
`strategies focused on [T-cell subsets (e.g., CD4*, CD6* and CD8*
`T cells) have been utilized, with limited success [144-146]. Orher
`studies have combined TCD and scheduled DLI post-alloSCT,
`to improve relapse rates and outcomes after TCD, with mixed
`results (147,148). In an alternative strategy to induce anergy to donor
`alloantigens, a smalltrial utilized costimulation blockade of HLA
`haploidentical donorT cells by ex vive incubation with CTLA4
`antibody and donor APCs with some reported success [149].
`Proteasomeinhibition may have a role in GVHD control. The
`transcription factor NF-«B plays an importantrole in cytokine
`signaling and the generation of cell-mediated immuneresponses.
`In addition, the proteasome has been shown to play a critical role
`in T-cell activation, proliferation and apoptosis, largely through
`NF-«B activation [150-152]. In addition to direct cytotoxic effects,
`the proteasome inhibitor bortezomib demonstrates immuno-
`modulatory effects through NF-«B [153]. It can attenuate TLR4-
`mediated APC activation, with reduced cytokine production and
`immunostimulatoryactivity [154]. Additionally, in the allogeneic
`setting, bortezomib preferentially and specifically depletes allore-
`active T lymphocytes [155]. In murine models of GVHD,bort-
`ezomibearly after stem cell infusion protected against GVHD
`without impairing engraftment[156,157]. Phase I and II trials for
`prevention and treatment of acute GVHD are ongoing, with
`interesting preliminary results [158].
`Additional agents that haye efficacy in the treatment of estab-
`lished acute GVHD are also being evaluated for primary GVHD
`prophylaxis. Examples include pentostatin and etanercept (dis-
`cussed later). Novel approaches include blocking lymphocyte
`migration to GVHD target organs using chemokine blockade
`(although there is significant redundancy in this system, com-
`plicating targeting efforts) and the use of extracorporeal photo-
`pheresis, which mayalter host antigen presentation and enhance
`Tregs for GVHD control[159-162], Ursediol, utilized for control
`of hepatotoxicity and treatment-related mortality (TRM) peri-
`transplant, was reported to also control severe acute GVHD [163].
`However, a meta-analysis confirmed the hepatotoxic and TRM
`benefit of ursodiol, but did not note improved GVHD control
`[164], Thalidomide was evaluated for chronic GVHD prophylaxis
`ina Phase III trial, with negative impact on mortality and chronic
`GVHD incidence [165]. Revlimid® or newer congeners may be
`more useful. Attempts to prevent thymic atrophy and associated
`chronic GVHD with thymic tissue implants, thymic epithelial
`cells or thymic hormones have not hadpositive results [166,167].
`
`Treatment of established GVHD
`In patients with established acute GVHD,the goal of therapy is
`to achieve rapid control, since the probability of survival depends
`upon theinitial stage of GVHD atpresentation and response to
`therapy [168-170], Long-term survival of patients with grade 0—-I
`acute GVHD is 50%, while long-term survival of those with
`
`116
`
`grade IV acute GVHD is as low as 11%[168]. Response to therapy
`is a key predictor of outcome, as mortality in acute grade J-IV
`GVHD is lowest in those with a complete response to initial
`therapy [169,171,172]. Corticosteroids are lympholytic and inhibit
`inflammatory cytokine cascades. Other agents have been uti-
`lized as first-line therapy, however none have proven superior to
`corticosteroids [169,173]. Corticosteroids also remain the primary
`front-line therapy for chronic GVHD,often in combination with
`a calcineurin inhibitor, as discussed later.
`
`First-line therapy of GVHD
`Corticosteroids (typically prednisone or methylprednisolone)
`dosed at approximately 2 mg/kg/day are the standard therapy
`for acute grade I-IV GVHD [174.175]. After single-agent steroid
`therapy, response rates were approximately 50% [169,171]. Higher
`doses have not been associated with improvedresponserates. Ina
`prospective randomized study comparing methylprednisolone at 2
`versus 10 mg/kg/day in patients with acute grade I-IV GVHD,
`response rates, TRM, GVHD progression and overall survival
`were similar [176]. TRM andlong-term survival were significantly
`improved in patients with early acute GVHD response that per-
`mitted steroid taper by day 5 of therapy [172]. In order to reduce
`the toxicities of prolonged systemic steroids, adjuvant topical ster-
`oids have been evaluated. In one randomized study, prednisone
`with and without oral nonabsorbable steroids (enteric-coated
`beclomethasone) were evaluated for therapy of gastrointestinal
`acute GVHD.Prednisone taper wasinitiated on day 10 if clinical
`response occurred. Durable responses and day 200 mortality were
`improved in the beclomethasone plus prednisone arm [177].
`The limited response to systemic steroids alone has prompted
`evaluation ofadditional immunosuppressive agents in the initial
`therapy of GVHD, This strategy has had only limited success,
`given the increased risks of infection and TRM. ATGis the most
`widely studied in this setting. Initial studies of upfront therapy
`with ATG plus steroids reported impressive response rates of
`67-80% [178.179], However, in a randomized study,initial therapy
`of acute grade I-IV GVHD with prednisone with and without
`ATG failed to demonstrate an improvementin response rates or
`survival in the ATG arm.Infectious complications were more
`commonin the combination ATG arm [iso].
`Otherbiologic agents have been evaluated in combination with
`steroids for initial therapy of acute GVHD.One randomized study
`evaluated systemic steroids with and without the monoclonal anti-
`body daclizumab that targets CD25 (the IL-2 receptor a-chain)
`present on activated T cells, dosed at 1 mg/kg on days | and 4 and
`weekly thereafter. Overall response rates were similar in the two
`groups, but survival at 100 days and 1 year wasinferiorin the dacli-
`zumab plus steroid group [11]. Similar lack of benefit was noted
`in a randomized study evaluating prednisone and cyclosporine
`with and without another monoclonalantibodytargeting the IL-2
`receptor (BT'563) [182]. CD5, found on the majority ofT cells, acts
`as a costimulation molecule to regulate signaling via the T-cell
`receptor. A randomized trial utilizing a CD5-specific immuno-
`toxin or placebo in combination with methylprednisolone found
`improved early response of acute GV HD in the immunoconjugate
`
`Expert Rew, Hematol. 1(1), (2008)
`
`SAN EX 1024, Page 06
`
`SAN EX 1024, Page 06
`
`

`

`Current &future approachesforcontrol of graft-versus-hostdisease|§\=))=) |
`
`arm, but comparable long-term outcomes [140]. The TNF-a. inhibi-
`tor etanercept in combination with corticosteroids plus tacrolimus
`demonstrated superior acute GVHD control in comparison to a
`historical cohort treated with steroids alone, in a small study that
`was recently updated (133,134).
`Currently, none of these agents have displaced corticosteroids
`as upfront therapy for acute GVHD,butthere is significant
`interest in finding better therapies for initial treatment of acute
`GVHD.Adjunctive agents that are currently being evaluated in
`an ongoing randomized trial by the Bone Marrow Transplant
`Clinical Trials Network include etanercept, denileukin diftitox,
`mycophenolate mofetil and pentostatin, each in combination
`with corticosteroids.
`Corticosteroids are also the mainstay of therapy for chronic
`GVHD. Other single agents are associated with a low response
`rate, Currently, there is no standard second-line therapy for
`chronic GVHD and therapy typically consists of prolonged
`administration of a corticosteroid combined with other immu-
`
`nosuppressive medications, such as calcineurin inhibitors
`(cyclosporine or tacrolimus). In a randomized trial compari

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