throbber
C\ Taylor & Francis
`
`~
`
`Taylor&Fr•ncisGroup
`
`rom
`OPINION
`
`Expert Opinion on Orphan Drugs
`
`ISSN: (Print) 2167-8707 (Online) Journal homepage: https://www.tandfonline.com/loi/ieod20
`
`Therapeutic targets and emerging treatment
`options in gastrointestinal acute graft-versus-host
`disease
`
`Anne S. Renteria, John E. Levine &James L. M. Ferrara
`
`To cite this article: Anne S. Renteria, John E. Levine & James L. M. Ferrara (2016) Therapeutic
`targets and emerging treatment options in gastrointestinal acute graft-versus-host disease , Expert
`Opinion on Orphan Drugs, 4:5, 469-484, DOI: 10.1517/21678707.2016.1166949
`
`To link to this article: https://doi.org/10.1517 /21678707.2016.1166949
`
`Accepted author version posted on line: 29
`Mar 2016.
`Published online: 06 Apr 2016.
`
`Submit your article to this journal 8"
`
`Wtl. Article views: 196
`~ ... ~ View related articles 8"
`
`(I} View Crossmark data 8"
`
`CronM.irk
`
`~ Citing articles: 1 View citing articles 8"
`
`Full Terms & Conditions of access and use can be found at
`https://www.tandfonline.com/action/journallnformation?journa1Code=ieod20
`
`Pharmacvclics Exhibit 2049
`Sandoz v. Pharmacyclics
`IPR20 19-00865
`
`

`

`EXPERT OPINION ON ORPHAN DRUGS, 2016
`VOL. 4, NO. 5, 469- 484
`http://dx.doi.org/1 O. l 517/21678707.2016. l 166949
`
`REVIEW
`
`W Taylor & Francis
`~ Taytor &.Francis Group
`
`Therapeutic targets and emerging treatment options in gastrointestinal acute
`graft-versus-host disease
`Anne S. Renteria•, John E. Levine" and James L. M. Ferrarab
`
`•Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; bHematologic
`Malignancies Translational Research Center, Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount
`Sinai, New York, NY, USA
`
`ABSTRACT
`Introduction: Graft-versus-host disease (GVHD) continues to be the major lethal complication of
`allogeneic hematopoietic stem cell transplantation (HCT) but the standard of care, high dose steroids,
`has not changed in 40 years. Approximately 50% of GVHD patients will develop steroid refractory
`disease, typically involving the gastrointestinal (GI) tract, which has a very poor prognosis. Newly
`developed GVHD biomarker-based risk scores provide the first opportunity to treat patients at the
`onset of symptoms according to risk of steroid failure. Furthermore, improvements in our understand(cid:173)
`ing of the pathobiology of GVHD, its different signaling pathways, involved cytokines, and the role of
`post-translational and epigenetic modifications, has identified new therapeutic targets for clinical trials.
`Areas covered: This manuscript summarizes the pathophysiology, diagnosis, staging, current and new
`targeted therapies for GVHD, with an emphasis on GI GVHD. A literature search on PubMed was
`undertaken and the most relevant references included.
`Expert Opinion: The standard treatment for GVHD, high dose steroids, offers less than optimal out(cid:173)
`comes as well as significant toxicities. Better treatments, especially for GI GVHD, are needed to reduce
`non-relapse mortality after allogeneic HCT. The identification of high risk patients through a biomarker(cid:173)
`defined scoring system offers a personalized approach to a disease that still requires significant research
`attention.
`
`ARTICLE HISTORY
`Received 13 January 2016
`Accepted 14 March 2016
`Published online
`5 April 2016
`
`KEYWORDS
`Biomarkers; GVHD;
`graft-versus-host disease;
`microbiome; Paneth cells;
`steroid-refractory GVHD;
`treatment
`
`1. Introduction
`
`Allogeneic hematopoietic stem cell transplantation (HCTI is
`increasingly used to cure malignant and benign hematologic
`diseases, with over 8000 transplants performed in the year
`2013.[1] Transplanted T cells from the donor can recognize
`and eradicate hematologic malignancies through the immu(cid:173)
`nologic graft-versus-leukemia
`(GVL) effect. Unfortunately,
`donor conventional T cells (Tcons) recognize normal recipient
`tissues and attack them, causing graft-versus-host disease
`(GVHD). The skin, liver, and gastrointestinal (GI) tracts are the
`primary targets of acute GVHD, which is the major cause of
`nonrelapse mortality (NRM) after HCT, [2] and develops in 40-
`60% of patients.[3]
`GVHD in the skin is the most frequently involved target
`organ and presents as an erythematous maculopapular rash;
`liver GVHD, the least common, causes hyperbilirubinemia; GI
`GVHD involves the upper GI tract, causing nausea, vomiting,
`and anorexia and, more often, the lower GI tract, cau sing
`diarrhea and abdominal pain. Each GVHD target organ is
`staged on a 0-4 severity scale, and the individual stages are
`used to create a composite clinical severity grade (Table 1).
`[4-6] Onset clinical severity does not correlate as well with
`survival as does maximal clinical severity, which also reflects
`treatment response. Patients with significant (;:,:Grade 2)
`
`GVHD are all treated similarly with high-dose steroid s, with
`intensification reserved for primary treatment failure. The
`high rate of treatment failure for lower GI GVHD accounts
`for the majority of NRM in the first 6 months after HCT.[7,8]
`Therefore, this manuscript will emphasize GI GVHD and the
`available and emerging therapies for its treatment.
`
`2. Pathophysiology
`
`The graft-versus-host (GVH) reaction is initiated when donor
`Tcons
`respond
`to genetically defined protein antigens
`expressed on host antigen-presenting cells (APCs).[2] Donor
`Tcons proliferate and differentiate during GVH, and the balance
`between effector and regulatory T cells (Tregs) plays an impor(cid:173)
`tant role in its progression and resolution. This paradigm for
`GVHD pathophysiology involves three distinct phases. In the
`first phase, which commences weeks before the onset of symp(cid:173)
`toms, tissue damage from the radiation and/ or chemotherapy
`given in the conditioning regimen initiates an inflammatory
`immunologic cascade involving both the innate and adaptive
`immune systems. The release of proinflammatory cytokines (e.g.
`TNFa, IL-1, and IL-6) promotes the activation of host APCs,
`which in turn drive donor Tcon proliferation, differentiation,
`and migration to target tissues.[1 OJ Damage to the GI epithe(cid:173)
`lium allows the translocation from the gut lumen of danger
`
`CONTACT Anne S. Renteria Q Anne.Renteria@mountsinai.org G) Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at
`Mount Sinai, New York, NY 10029, USA
`© 2016 lnforma UK Limited, trading as Taylor & Francis Group
`
`

`

`470 @ A. S. RENTERIA ET AL.
`
`Article highlights
`
`• Acute GVHD develops in 40-60% of allogeneic HCT recipients and is
`the major cause of NRM. The standard first-line therapy for acute
`GVHD is high-dose steroids, but SO% of cases are steroid refractory.
`GI GVHD accounts for the majority of NRM because of its high rate of
`treatment failure.
`• New treatments for GVHD under study that exploit new t argets
`involved in GVHD pathophysiology include blockade of leukocyte
`trafficking, JAK inhibition, hi stone deacetylase inhibitors, alpha-1
`antitrypsin, induction of regulatory T cells, and restoration of GI
`barrier function through cytokines.
`• A va lidated scoring system based on GVHD biomarkers (Ann Arbor
`risk scores) objectively stratifies patients according to risk of primary
`treatment failure and can identify patients for clinical trials before
`steroid refractory GVHD ha s developed.
`• The loss of diversity in the GI microbiota is associated with increased
`GVHD mortality and relates to different factors, including exposure t o
`antibiotics. Preservation or restoration of a healthy GI microbiome is
`an alternative strategy to treat GI GVHD.
`
`This box summarizes key points contained in the article.
`
`signals and pathogen-associated molecular patterns (PAMPs)
`such as bacterial cell wall components (e.g. lipopolysaccharide
`- LPS) and damage-associated molecular patterns (DAMPs) (e.g.
`ATP and extracellular matrix proteins) to amplify the cytokine
`cascade and trigger the development of chemokine gradients
`that attract donor Tcons.[11 ] Tregs are transcription factor fork(cid:173)
`head box P3 (Foxp3+) CD4 + T cells that by suppressing allor(cid:173)
`eactive lymphocytes can dampen the effect of GVHD caused by
`Tcons [12] (Figure 1 ). In the second phase, also days to weeks
`before the onset of symptoms, T-cell traffic to target organs in a
`highly regulated process, during which interactions between
`adhesion molecules (e.g. MAdCAM-1) and integrins (e.g. a4~7)
`result in leukocyte adherence to the capillary endothelium and
`migration into the subendothelium [13] as shown in Figure 2.
`Chemokines regulate not only the trafficking of leukocytes, but
`also their activation and differentiation by binding to specific
`receptors, such as chemokine (C-C motif) receptor 5 (CCRS),
`CCR6, and CCR7.[l 4, 15] In Phase 3, clinical symptoms com(cid:173)
`mence when Tcons cause tissue destruction through direct
`cytotoxic activity, mostly through Fas ligand: Fas and perforin(cid:173)
`granzyme pathways [16], as well as through cytokines such as
`TNFa (Figure 3).
`
`3. Diagnosis
`
`The diagnosis of acute GI GVHD is based on clinical signs and
`symptoms
`(which do not occur until Phase 3 of its
`pathophysiology).
`It is common practice to rule out infectious enteritis by
`testing the stool for adenovirus, rotavirus, or Clostridium diffi(cid:173)
`cile.[17] Diarrhea
`from pretransplant chemoradiotherapy(cid:173)
`induced GI damage is common prior to engraftment and
`when GVHD symptoms occur; most centers perform diagnos(cid:173)
`t ic endoscopy to establish the diagnosis. However, endoscopic
`abnormalities are seen in less than one-third of GI GVHD cases
`and are nonspecific [18]; biopsies are usually performed to
`obtain histologic confirmation of the diagnosis. Flexi ble sig(cid:173)
`moidoscopy is as sensitive as full colonoscopy.[19] Upper GI
`endoscopy is often performed, as the small bowel may be the
`major source of diarrhea, although this approach is not uni(cid:173)
`versally accepted.[20] Although GI GVHD symptoms may start
`as early as 9 days after HCT, they often do not start until
`patients have been discharged from the hospital, and accurate
`measurements of outpatient diarrheal output are not routinely
`available. The average volume per episode of diarrhea in an
`adult has been estimated to be - 200 ml, allowing for staging
`and grading of GI GVHD when only the number of diarrhea
`episodes is known.[9]
`
`4. Histology
`
`GI crypt dropout is characteristic of histologic GVHD, and
`intestinal stem cells (ISCs) are considered targets of the dis(cid:173)
`ease,[2 1,22] with crypt cell apoptosis a histologic hallmark.
`Nonetheless, GI GVHD histology is often not straightforward.
`Crypt damage from conditioning chemotherapy ± radiation
`can take weeks to heal, and persistent changes can overlap
`with the onset of GI GVHD symptoms. Other causes of crypt
`damage include infections and mycophenolate mofetil,[23] a
`commonly used immunosuppressant. The patchy nature of GI
`GVHD histology sometimes leads to false-negative biopsies
`through sampling error. Finally, the histologic grading system
`(Table 2) [24] has never been standardized for a number of key
`parameters, including the number of tissue sections or high(cid:173)
`powered fields to be analyzed, or the number of apoptotic
`
`Table 1. GVHD target organ staging.
`
`Stage
`0
`
`Skin (active erythema only)
`No active (erythematous) GVHD rash
`
`Liver (bilirubin)
`
`Upper GI
`
`<2 mg/ di
`
`2-3 mg/d i
`
`No or intermittent nausea,
`vomiting, or anorexia
`Persistent nausea,
`vom iting, or anorexia
`
`2
`
`4
`
`3.1 - 6 mg/ di
`
`6.1-15 mg/ di
`
`>15 mg/di
`
`Maculopapular ra sh
`< 25% BSA
`Maculopapular ra sh
`25-50% BSA
`Maculopapular rash
`>50% BSA
`Generalized erythroderma (>50% BSA) plus
`bullous formation and desquamation >5% BSA
`*When stool volume is not quantified, a 200-ml/episode can be used as an estimate for adults [9].
`Overall cli nical grade (based on most severe target organ involvement):
`Grade 0: No stage 1-4 of any organ.
`Grade I: Stage 1-2 skin without liver, upper GI, or lower GI involvement.
`Grade II: Stage 3 rash and/o r Stage 1 liver and/or Stage 1 upper GI and/or Stage 1 lower GI.
`Grade Ill: Stage 2-3 liver and/or Stage 2- 3 lower GI, with Stage 0-3 skin and/or Stage 0-1 upper GI.
`Grade IV: Stage 4 skin, liver, or lower GI involvement, with Stage 0-1 upper GI.
`
`Lower GI (stool output/day)*
`Adult: <500 ml/ day or <3 episodes/ day
`Child: < 10 ml/ kg/ day or <4 episodes/ day
`Adult: 500- 999 ml/ day or 3-4 episodes/day
`Child: 10- 19.9 ml/ kg/ day or 4-6 episodes/ day
`Adult: 1000- 1500 ml/ day or 5-7 episodes/day
`Child: 20- 30 ml/ kg/day or 7-1 0 episodes/ day
`Adult: > 1500 ml/ day or > 7 episodes/ day
`Child: > 30 ml/kg/ day or > 10 episodes/day
`Severe abdominal pain with or without ileus or
`grossly bloody stool (regardless of stool vo lume)
`
`

`

`EXPERT OPINION ON ORPHAN DRUGS @ 471
`
`Donor Tcon Activation
`
`Figure 1. Blockade of donor Tcon activation (first phase of acute GVHD).
`This schematic figure depicts key events in early acute GVHD pathophysio logy in the GI tract, the most important GVHD target organ. A villus of the small intestine
`(left) is highly magnified (right).
`The tran splant conditioning regimen leads to generation of danger signals and pro-inflammatory cytokines (e.g. TNFa, IL-6) that activate APCs. For illustration
`purposes, one APC is shown interacting with one conventional T cell (Tcon) and one regulatory regulatory T cell (Treg). Physiologically, these cells are clustered
`beneath crypts, in the lamina propria. Etanercept and infliximab, two anti-TNFa antibodies, neutralize TNFa directly. Tocilizumab, prevents APC activation by
`inhibiting binding of IL-6 to the IL-6 receptor.
`Steroids, ATG, a/emtuzumob, and MSCs are broad immunosuppressors that act on multiple immunologic processes, including the activation and differentiation of
`Tcons through different mechanisms (see text). ECP and vorinostat suppress host APC and promote Treg expansion.
`Abbreviations: APC, antigen-presenting cell; ATG, antithymocyte globulin; ECP, extracorporeal photopheresis; MSC, mesenchymal stromal cell.
`
`cells required to diagnose Grade 1 GVHD. As a consequence,
`interobserver variability is high, histologic severity does not
`correlate well with clinical symptoms, and a negative biopsy
`does not necessarily rule out GVHD.[25] Paneth cells are pri(cid:173)
`marily located in the crypts of the small intestine, and their
`number inversely correlates with high-risk disease.[26] Paneth
`cells are readily identified by their location in the GI tract and
`their histochemical staining with lysozyme. Their straightfor(cid:173)
`ward quantification can aid in establishing the diagnosis and
`in prognosticating its severity.
`
`antimicrobial peptide and regulator of intestinal gram-positive
`bacteria. As already mentioned, Paneth cell loss correlates with
`GI GVHD severity and long-term outcomes.[26] As a biomarker
`specific for GI GVHD, REG3a discriminates between GVHD and
`non-GVHD causes of diarrhea.[32] ST2 is secreted in response
`to inflammatory stimuli, and it functions as a decoy receptor
`for interleukin-33 (IL-33), which drives Tcons toward a proin(cid:173)
`flammatory phenotype.[33] ST2 plasma concentrations at the
`initiation of GVHD treatment strongly correlates with eventual
`resistance to treatment and 6-month NRM, which is primarily
`driven by steroid-refractory (SR) GI GVHD.[34]
`
`5. GVHD biomarkers
`
`Despite a large number of plasma proteins, DNA single
`nucleotide polymorphisms, microRNA molecules, and periph(cid:173)
`eral blood cellular subsets with associations to GVHD,[27-29]
`only a small number of candidate biomarkers have been
`validated in multicenter patient cohorts. Of these validated
`biomarkers, those with greatest relevance to GI GVHD are
`tumor necrosis factor receptor-1 (TNFRl ), regenerating islet(cid:173)
`derived protein-3-a (REG3a}, and suppression of tumorigeni(cid:173)
`city 2 (ST2). TNFRl, a membrane receptor for TNF-a that
`becomes soluble after binding its ligand, is not specific for GI
`GVHD but has been shown to strongly correlate with overall
`GVHD severity, response to treatment, NRM, and survival.
`[30,31] REG3a, which
`is secreted by Paneth cells,
`is an
`
`5. 1. Biomarker-defined risk stratification
`
`Individual GVHD biomarker concentrations vary widely among
`centers. To overcome this limitation, the Ann Arbor (AA) scor(cid:173)
`ing system used several biomarkers from a multicenter cohort.
`An algorithm combining the plasma concentrations of TNFRl,
`REG3a, and ST2 at GVHD onset was developed to categorize
`patients according to risk of primary treatment failure and
`NRM.[35] Thresholds define three distinct scores: AA 1, NRM
`-1 0%; AA2, NRM -25%; and AA3, NRM -40%. Because relapse
`rates do not differ among the AA scores, these differences in
`NRM translate into significant differences in overall survival. It
`is important to note that AA scores identify patients who will
`later develop lower GI GVHD but who present with only a rash.
`
`

`

`472 @ A. S. RENTERIA ET AL.
`
`lnolimomab
`Denlleukin diftitox
`
`Nucleus:
`HDACi
`
`Cytoplasm:
`SYKi
`JAKi
`AAT
`
`Tcon Proliferation & Migration
`
`Figure 2. Blockade of Tcon proliferation and migration (second phase of acute GVHD).
`Following activation by host APCs, donor Tcons proliferate and migrate from secondary lymphoid organs and tissues associated with the mucosa (e.g. Peyer's
`patches). Activated Tcons release cytokines (e.g. IL-2) that promote further proliferation and differentiation. Basiliximab, daclizumab, inolimomab, and denileukin
`diftitox all bind to IL-2 receptor (IL-2R). HiDAC inhibitors suppress APCs activity, enhance Treg activity, and reduce pro-inflammatory cytokines through DNA
`methylation (azacitidine) and histone acetylation (vorinostat), whi le inhibitors of JAK1/2 (ruxolitinib), SYK (fostamatinib, and a-1-antitrypsin (AAD suppress cytokine
`production through cytoplasmic receptors. The migration of activated Tcon s (T cel l trafficking) into the GI subendothelium requires interaction between integrins
`(e.g. a4P7-integrins) expressed on Tcons and their receptors. Natalizumab and vedolizumab are antibodies that block a4 or a4P7, respectively. Intestinal stem cells
`(ISCs) are located at the base of th e crypts, interspersed with Paneth cells, and are responsible for crypt regeneration. Type 3 innate lym phoid cells (I LC3 s) produce
`IL-22 which is trophic for ISCs and which induces regenerating islet derived protein-3-a (REG3a), an antimicrobial peptide which destroys gram-positive bacteria.
`Abbreviations: APC, antigen-presenting cell; GI, gastrointestinal; GVHD, graft-versus-host disease.
`
`Patients whose skin was the only target organ affected at the
`time of diagnosis and who were classified as AA3 by biomar(cid:173)
`kers were twice as likely to develop lower GI GVHD later as
`patients classified as AA 1.[35] Biomarker-defined GVHD sever(cid:173)
`ity thus appears promising as an opportunity for early inten(cid:173)
`sive intervention in patients diagnosed with high-risk GVHD.
`
`6. Therapeutic options
`
`6. 1. Broad immunosuppression
`
`Most therapeutic options for GVHD have been evaluated in
`the context of systemic disease where sometimes, only overall
`grades are reported. In this review, we highlight outcomes for
`GI GVHD wherever possible; otherwise, the results described
`apply to GVHD in general.
`Intensified immunosuppression with systemic steroids is
`the only proven treatment for GVHD. Unfortunately, SR
`GVHD develops in - 50% of patients,[7,36,37] more often in
`patients with lower GI involvement,[?] and there is no estab(cid:173)
`lished second-line therapy. The response rates to second-line
`treatment are typically low (20-40%), and survival is poor,
`highlighting the urgent need
`for better therapies.[36,38]
`Thus, SR GVHD remains a significant contributor to treat(cid:173)
`ment-related mortality (TRM) and morbidity. As reviewed in
`the following section and summarized in Tables 3 and 4,
`
`several treatments have shown benefit in single-arm studies;
`however, that benefit has not been confirmed in the few that
`have been evaluated by more rigorous randomized trials.
`
`6.2. Steroids
`
`GVHD treatment usually begins with high doses of prednisone
`or, for patients unable to take oral steroids, methylpredniso(cid:173)
`lone (MP) (Figure 1 ). A randomized, prospective clinical trial
`showed that a 50% reduction in the starting dose of steroids
`was effective, although patients with more severe GVHD were
`more likely to require addition of secondary immunosuppres(cid:173)
`sion.[39] Upper GI GVHD may also respond well to the combi(cid:173)
`nation of 1 mg/ kg/d of prednisone and nonabsorbable steroid
`therapy.[40] Conversely, doses higher than 2 mg/ kg/day do
`not improve outcomes.[41]
`
`6.2. 1. Nonabsorbable steroids
`Oral nonabsorbable steroids, such as budesonide and beclo(cid:173)
`methasone, theoretically deliver high steroid doses to the GI
`tract without incurring the side effects of systemic steroid
`therapy. These agents have primarily been tested in combina(cid:173)
`tion with systemic steroids as part of first-line therapy for GI
`GVHD. It should be noted, however, that the term 'nonabsorb(cid:173)
`able' is a misnomer, because synthetic steroid screens often
`demonstrate significant absorption.[42] Results of
`this
`
`

`

`EXPERT OPINION ON ORPHAN DRUGS @ 473
`
`I
`"
`
`\
`
`' /
`Enter~ occus -' aJ 11u,
`
`I
`
`;
`
`'
`
`....
`Target Cell
`Destruction
`
`• Reg3a•enters
`bloodstream
`
`!£/--JU/
`
`o:01sMountSmaf
`IJN1th.5yllm
`
`Figure 3. Prevention of target cell destruction (third phase of acute GVHD).
`Activated Tcons that migrated to the GI epithelium secrete inflammatory cytokines and lyse ISCs and Paneth cells, disrupting the GI mucosal barrier. REG3a stored
`intracellularly and in the mucus leaks into the systemic circulation along with luminal contents, such as bacteria. Strategies to restore the integrity of the mucosal
`barrier are currently explored via clinical trial of recombinant IL-22 (r/L-22) administration.
`Abbreviations: GI, gastrointestinal; GVHD, graft-versus-host disease.
`
`Table 2. Histologic grading for acute gastrointestinal graft-versus-host disease
`(24].
`
`Grade
`0
`1
`2
`3
`4
`
`Histology
`
`Normal mucosa
`Isolated apoptotic epithelial cells without crypt loss
`Loss of isolated crypts without loss of contiguous crypts
`Loss of two or more contiguous crypts
`Extensive crypt loss with mucosal denudation
`
`Marrow Transplant Clinical Trials Network. As a salvage ther(cid:173)
`apy for SR GVHD, response rates of 57-76% have been
`reported for sirolimus.[48,53] Its side effect profile differs
`from other immunosuppressants and
`includes transplant(cid:173)
`related microangiopathy (TMA), particularly when combined
`with calcineurin inhibitors, as well as hyperlipidemia.[53]
`
`approach have been mixed, [43,44] and the only Phase Ill trial
`involving budesonide did not meet its primary end point of
`time to treatment failure.[40] Steroid delivery through arterio(cid:173)
`graphy to the arterial blood supply to the gut has been
`explored in several small studies. In two prospective series,
`GI GVHD response rates of 70-85% were observed and
`appeared better than historical controls.[45-47] The need for
`trained and available interventional radiology specialists may
`explain the lack of larger and adequately powered rando(cid:173)
`mized studies.
`
`6.3. Sirolimus
`
`Sirolimus, an mTOR inhibitor, blocks late Gl cell cycle progres(cid:173)
`sion, most prominently in T lymphocytes,[48] and also main(cid:173)
`tains Treg populations.[49] Sirolimus has proven efficacy in
`GVHD prophylaxis [50,51 ] and showed comparable activity to
`that of high-dose steroids as primary therapy for acute GVHD
`in one study.[52] Sirolimus is currently being compared to
`prednisone as a single agent for primary therapy of low-risk
`GVHD in a randomized trial conducted by the Blood and
`
`6.4. Purine synthesis inhibitors
`
`Mycophenolate mofetil (MMF) blocks de nova purine synthesis
`through inhibition of inosine monophosphate dehydrogenase
`(IMPDH), which arrests
`lymphocytes,
`in S-phase.[33]
`Encouraging results of MMF as primary treatment for GVHD
`were observed in several small series, [54,55] including multi(cid:173)
`center, randomized, Phase II trial.[56] However, a randomized,
`placebo-controlled trial was closed to accrual early for futility.
`[57] A significant drawback to MMF is its GI toxicity profile,
`which includes ulcerative esophagitis, reactive gastropathy,
`and pathologic changes very similar to GVHD that can cloud
`diagnostic biopsy interpretation.[58]
`Pentostatin, a nucleoside analog, inhibits adenosine deami(cid:173)
`nase and induces cell death particularly in T and NK cells.[59]
`Pentostatin has shown mixed results for SR GVHD in small
`series, with survival ranging from 7 to 43%.[60- 62]
`
`6.5. Anti-T-ce/1 serotherapy
`
`Antithymocyte globulin (ATG) consists of polyclonal lgG anti(cid:173)
`bodies against human T cells (Figure 1 ). The role of both
`
`

`

`)>
`----l
`m
`►
`::0
`m
`----l
`m z
`::0
`~
`?>

`... .._, ...
`
`'
`
`(Continued)
`
`were used. High rate of infections
`
`Limited activity. Variety of doses and schedules
`
`ORR 38% (CR 21%); 2 year Schmitt (2011) [62]
`ORR 50% (CR 33%)
`Pida/a (2010) [61]
`
`Alam (2013) [60]
`
`additional therapies
`
`ORR 33%; 53% received
`
`05 17%
`
`(2015) [55]
`
`Inagaki
`
`[57]
`
`Bolanos-Meade (2014)
`
`3 year OS 86%
`day 56 CR 100% (GI only);
`Day 28 ORR 83% (GI only);
`
`from MMF
`(p = 0.08); no benefit
`1 year relapse 24 vs. 16%
`(p = 0.34);
`1 year OS 58% vs. 65%
`(p = 0.34);
`
`Day 56 CR 60 vs. 54%
`
`16 vs. 52%
`pts for toxicity; 1 year OS
`MMF discontinued in 21%
`31%);
`
`responses
`
`survival
`
`Furlong (2009) [54]
`
`ORR 47 vs. 48% (CR 31 vs.
`
`Day 35 response
`
`ORR 62% (CR 46%); 1 year Meunier (2014) [72]
`
`05 33%; one case of PTLD
`
`myelosuppressive
`mimic GVHD. Increased infections,
`diarrhea; can induce GI pathology changes that
`
`Usually well tolerated but can cause nausea and
`
`EBV reactivation/PTLD due to B cell depletion
`treatment. Increased risk for CMV; less risk for
`
`Effective GVHD prophylaxis; less studied for GVHD
`
`(2008) [71]
`
`Gomez-Almaguer
`
`ORR 83% (CR 33%)
`one case of PTLD
`higher TRM with ATG;
`33 vs. 24%, p = NS);
`
`one case of PTLD
`survival. 1 year OS 32%;
`and earlier. No impact on
`response for skin GVHD
`
`p = 0.11
`6 month OS 79 vs. 50%
`(CR 63 vs. 21%, p = 0.03);
`
`ORR 79 VS. 42%, p = 0.07
`
`ORR 54% (CR 20%). Best
`
`Day 28 response
`
`Day 30 response
`
`Day 28 response
`
`ORR 55 vs. 48%, p = NS (CR Van Lint (2006) [67]
`
`increased risk for PTLD
`to use; significant infectious complications,
`proof of efficacy; no consensus on which brand
`
`MacMillan (2002) [65] Commonly used agent for SR GVHD despite lack of
`
`Nishimoto (2015) [47]
`
`Safe procedure and no increase in infection rates.
`
`Burg/er (2014) [46]
`
`[40]
`
`Hackenbery (2007)
`
`radiology service is needed
`Pitfalls: trained and available interventional
`
`Response at discharge ORR 72%; 1.5 year OS 27% Weintraub (2010) [45]
`
`setting of GVHD and antacid therapy
`action of the drug, and not always optimal in
`suppression; pH milieu is important for effective
`absorption does occur with reports of adrenal
`
`Nonabsorbed is a misnomer, variable systemic
`
`Comments
`
`ORR 77% (CR 65.5%), 5 pts Castilla (2006) [44]
`
`Day 28 response
`
`Bertz (1999) [43]
`Author
`
`ORR 77 vs. 32%, p < 0.0,
`
`Results
`
`movement
`frequency of bowel
`
`p = 0.04
`1 year OS 71 vs. 58%,
`50 (31 vs. 48%, p = 0.12);
`BDP group through day
`Fewer treatment failure in
`
`ORR 83%; 1 year OS 50%
`
`systemic steroids
`50% pts required
`in CR relapsed; overall
`
`survival
`
`Day 28 response
`
`Retrospective
`
`GI)
`
`15 SR GVHD (12
`
`Day 28 response
`Day 28 response
`
`Retrospective
`Retrospective
`
`24 SR GI GVHD
`12 SR GI GVHD
`
`(purine analog)
`
`Pentostatin
`
`only, high dose MMF
`
`Retrospective, children Day 28 and 56
`
`with GI GVHD)
`14 SR GVHD (12
`
`Day 56 GVHD-free
`
`placebo
`MMF 1 g Q8 h VS.
`multicenter; PRD +
`placebo controlled,
`Phase Ill prospective,
`
`onset GVHD
`
`116 vs. 119 new
`
`ANC < 1500 excluded
`controls; pts NPO or
`with historical
`
`with GI)
`controls (11
`29 historical
`
`synthesis)
`(inhibitor of purine
`(MMF)
`
`19 (10 with GI) vs. Phase II prospective
`
`ANTI METABOLITES Mycophenolate mofetil
`
`Survival
`
`Retrospective
`
`24 SR GI GVHD
`
`Prospective, 2nd line
`
`with GI GVHD)
`
`antibody)
`emtuzumab (anti-CD52 18 SR GVHD, (8
`
`Al
`
`kg x 5 doses)
`days± ATG (1.25 mg/
`MP 5 mg/kg/d x 10
`211 pts; 61 with SR Phase 111, multicenter;
`
`(rabbit ATG)
`acute GVHD
`
`days+ GC
`15 mg/kg bid x 5
`1-5 courses ATG
`
`(horse ATG)
`lower GI.
`
`79 SR GVHD; 52% Retrospective; 2nd line; Day 28 response
`
`dose MP 180 mg
`19 SR GI GVHD vs. Phase 11, mean total
`
`controls
`14 historical
`
`Pilot study, ;,, 2nd line,
`
`)
`
`(MP 75 mg/m2
`
`11 SR GI GVHD
`
`Prospective, mean total Day 28 response
`
`12 SR GI GVHD
`
`dose MP 180 mg
`
`1 I diarrhea excluded
`placebo, pts with >
`1 mg/kg/d +
`PRD + BDP vs. PRD
`
`Phase 111, biopsy proven, Day 50 response;
`
`only
`with GI GVHD
`62 vs. 67 pts, all
`
`(ATG)
`
`Antithymocyte globulin
`
`(IAS)
`
`Intra-arterial steroids
`
`monotherapy BDP
`
`GI GVHD
`
`26 pts ,;; Grade 2 Prospective,
`
`proprionate (BOP)
`Oral beclomethasone
`
`di
`
`GC + BUD vs. GC only
`
`historical control
`GI GVHD vs. 19
`
`22 pts ;,, Grade 2 Retrospective, systemic Stool volume and
`End points
`Patient population
`
`Study design
`
`Budesonide (BUD)
`Agent
`
`AGENTS
`DEPLETING
`LYMPHOmE-
`
`Table 3. Nonspecific T-cell targeting.
`
`

`

`C:
`
`QJ .s
`"' %
`
`>(cid:173)
`"' Cl
`
`"' QJ
`cc
`
`QJ
`
`QJ
`
`> ·e
`5;(cid:173)
`g
`
`QJ
`Ct::
`
`C:
`C,
`·.;;
`QJ
`"O
`
`-C:
`
`QJ
`
`C, <
`
`EXPERT OPINION ON ORPHAN DRUGS 9 475
`
`horse- and rabbit-derived formulations of ATG in GVHD pro(cid:173)
`phylaxis is well established.[63,64] Rabbit-derived ATG induces
`more profound lymphopenia, but titers in the two brands vary
`widely and with no consensus as to which brand should be
`preferred. A retrospective series of 79 patients treated with
`horse ATG for SR GVHD showed a 54% CR/PR rate [65];
`patients in that study were more likely to have skin involve(cid:173)
`ment and less likely to have GI
`involvement compared to
`studies with less favorable results.[66] A large randomized
`study of rabbit ATG for SR GVHD did not improve outcomes,
`and its use was associated with a significantly increased risk of
`TRM.(67] Thus, improvements in response rates with ATG may
`not translate to survival benefit because of increased infec(cid:173)
`tious complications and Epstein-Barr virus (EBV)-associated
`lymphoproliferative disease.(68]
`Alemtuzumab, a humanized lgG monoclonal antibody to
`CD52 that is expressed on lymphocytes, monocytes, and APCs
`(Figure 1), is a more potent alternative to ATG,(69] and is
`effective as GVHD prophylaxis.(70] In small studies of SR
`GVHD patients, alemtuzumab provides overall response rates
`from 62 to 83%, with overall survival of 33-70%.(71,72] As
`expected, infectious complications frequently develop.
`
`6.6. Targeted therapies
`
`6.6.7. TNFa
`lnfliximab. lnfliximab (Figure 1) is a chimeric anti(cid:173)
`6.6. 7. 7.
`body that neutralizes TNFa and lyses the cells that produce it
`by binding to its membrane-bound form.(73] In retrospective
`series of patients with SR GVHD, infliximab produced overall
`response rates ranging from 15 to 60%, but these were asso(cid:173)
`ciated with high rates of fungal infections.(73-75] A rando(cid:173)
`mized, Phase Ill trial showed no benefit from the addition of
`infliximab to steroids as primary GVHD therapy.(76]
`
`6.6. 7 .2. Etanercept. Etanercept, which consists of
`two
`recombinant human TNFR (p75) monomers fused to the Fe
`portion of human lgG, neutralizes soluble TNFa (Figure 1), but
`does not lyse the cells producing it, and has a good safety
`profile.[77] In a prospective single-center study as primary
`GVHD therapy, the combination of etanercept and steroids
`was significantly superior to steroids alone (CR 69 vs. 33%);
`(30] however, these results were not reproduced in a rando(cid:173)
`mized, Phase II study.(56] The drug is not as effective in the SR
`GVHD setting, with response rates of 40-50%, including for
`patients with severe GI GVHD.[78,79]
`
`6.6.2. /L-6 receptor
`Tocilizumab is a humanized anti-lL6 receptor antibody that
`blocks IL-6 signaling, which is pivotal in the differentiation of
`CD4+/IL-17-secreting T (Th17) cells
`from na"i"ve T cells
`(Figure 1 ). In murine models, loss

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