throbber
Clinical Immunology (2013) 147, 105419
`
` www.elseviercom/localelyolim
`
`available at www.3clencedireclcom
`
`Clinical Immunology
`
`
`
`Modulation of immune function occurs within hours of
`
`therapy initiation for multiple sclerosis
`
`Chris L. Ayersa’1’2, Jason P. Mendozaa’1’3, Sushmita Sinhaa’ 1,
`Khrishen Cunnusamy 3, Benjamin M. Greenbergb,
`Elliot M. Frohmanb, Nitin J. Karandikar“
`
`a Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-9072, USA
`b Department of Neurology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
`
`Received 15 January 2013; accepted with rew‘sion 15 February 2013
`Available online 6 March 2013
`
`a
`
`KEYWORDS
`Glatiramer acetate;
`Multiple 55130515;
`Immunomodulation
`
`
`
`Daily administration of FDA-approved glatiramer acetate (GA) has beneficial effects
`Abstract
`on clinical course of relapsing remitting multiple sclerosis (RRMS). Although mechanisms of
`GA-action have been widely investigated and partially understood, immediate immune dynamics
`following (SA-therapy are unknown.
`In the present study, we characterized the immediate
`effects of GA on phenotype, quantity and function of immune cells in MS patients. Prominent
`changes in immune cells were detected within 4712 h post-first (SA-injection. T-cell modulation
`included significantly decreased CD4ICD8 ratio, perturbed homeostasis of predominantly CD8+
`T~cells, significant enhancement in CD8+ T-celt mediated suppression and inhibitory potential of
`induced CD4-suppressors. Changes in APC were restricted to monocytes and included reduced
`stimulatory capacity in MLR and significantly increased iLafO and TNF-u production. Our study
`provides the first evidence that GA treatment induces rapid immunologic changes within hours
`of first dose. Interestingly, these responses are not only restricted to innate immune cells but
`also include complex modulation of T-ceil functionality.
`© 2013 Elsevier Inc. All rights reserved.
`
`
`" Corresponding author at: University of Iowa Healthcare, Carver
`College of Medicine, Iowa City, IA 52240, USA. Fax: +1 319 384 9613.
`E-mail address: nitin-karandikar@uiowa.edu (NJ. Karandikar).
`I Equal comm-bum“ (wfirst authors).
`2 Currently at: Opexa Therapeutics, The Woodlands, TX 77381,
`USA.
`3 Currently at: Teva Pharmaceuticals, Kansas City, MO 64131, USA.
`
`1 . Introduction
`
`Underlying immune—pathogenesis of multiple sclerosis (MS)
`involves multi-focal demyelination and axonal loss caused by
`infiltration of myelinereactive T cells into the CNS. However,
`the disease is much more complex and tissue injury in active
`CNS lesions is also associated with activated microglia and
`macrophages. Glatiramer acetate (GA) is an FDA-approved
`immunomodulatory agent for the treatment 0f relapsing
`remitting-MS (RRMS). GA is a synthetic polypeptide composed
`of glutamic acid,
`lysine, alanine and tyrosine in a defined
`
`15217661615 - see front matter © 2013 Elsevier Inc. All rights reserved.
`
`http:/ldx.doi.org/10.1016/j.clim.2013.02.015
`
`Page 1 0f 15
`
`Biogen Exhibit 2227
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 1 of 15
`
`Biogen Exhibit 2227
`Mylan v. Biogen
`IPR 2018-01403
`
`

`

`106
`C.L. Ayers et al.
`
`molar ratio. Although the precise mechanism of GA action is
`still unclear, several immunologic effects of GA therapy
`have been described,
`including 1)
`immune deviation of
`CD4+ T cell responses from Th1 to Th2 [1—5], 2) induction
`of CD4+CD25+FoxP3+ regulatory T cells [6], 3) induction
`of Oligoclonal, cytotoxic/suppressor CD8+ T cells [7—10],
`4) induction of immune modulatory Type II monocytes [11], and
`5) induction of IL-10 producing B cells [12,13]. However, the
`sequence of these interactions and the immediate immunologic
`effects of GA that may determine downstream effects on
`disease remain poorly understood.
`In fact, there has been no
`systemic dissection of the effects of early GA administration on
`immune cell phenotype and function. Several studies have
`documented altered immune cell effector functions upon short
`term exposure to GA. In accordance with this, GA is capable of
`inducing changes in antigen presenting cell
`(APC) function
`within hours of overnight in vi tro cultures. Similarly, even in the
`absence of prior GA therapy, GA is able to induce in vitro CD4+
`and CDS+ T cell responses from PBMC derived from healthy
`subjects and MS patients within a few days of culture [7,9].
`Therefore it
`is conceivable that
`following the first
`few
`injections, GA would show immediate in viva immune effects
`that might dictate the eventual ability to develop a sustained
`immune regulatory response.
`The present study is a novel comprehensive evaluation of
`immune alterations induced in T cell and APC populations
`during the first 72 h of GA therapy. Treatment naive RRMS
`patients initiating GA therapy were recruited for the study.
`Phenotypic and functional assays were performed on CD4+ T
`cell, C08+ T cell, CD14+ monocyte, CD19+ B cell, BDCA1+
`myeloid dendritic cell
`(MDC) and BDCA4+ plasmacytoid
`dendritic cell (PDC) populations. The results were compared
`to the control subjects comprising of healthy donors (HD) as
`well as untreated-treatment na'ive RRMS patients, all of
`whom underwent a mock admission for specimen collection.
`We found that GA induces prominent phenotypic and func-
`tional changes in not only innate APC populations but also
`complex changes in T cells, particularly in the functional
`status of C08+ T cells as early as 12 h after the first injection.
`These studies provide important insights into the timeline of
`immune alterations and emphasize the need for longitudinal
`studies to assess their significance in determining long-term
`immune and clinical consequences.
`
`2. Materials and methods
`
`2.1 . Patients and control subjects
`
`After obtaining informed consent, 7 healthy donors, 8
`treatment-naive RRMS patients initiating glatiramer ace-
`tate (GA) therapy, and 4 "untreated" treatment na'l've RRMS
`patients were recruited for the study. At the time of
`monitoring, MS patients were free of steroid therapy for at
`least 3 months, and had no record of acute relapse within
`3 months. None had a history of disease modifying therapy.
`All participants were admitted to the Clinical and Transla-
`tional Research Center (CTRC) for overnight blood draws
`(0 h baseline usually between 6 and 8 PM, followed by 4, 12
`and 24 h post-first
`injection). The 24 h collection was
`performed prior to the second daily GA injection. Partici~
`pants were then released and asked to return for a 72 h post-
`
`Page 2 of 15
`
`baseline blood draw (before their fourth daily 5th of GA).
`Treatment decisions were determined by routine standard of
`care and patients were provided injection training during their
`first two GA injections. The healthy subjects and the untreated
`subjects served as important cohorts to control for potential
`diurnal variation of measured parameters. Thus, only the
`parameters that changed in the GA-treated cohort but not in
`the other two cohorts were considered an effect of GA therapy.
`All studies were approved by the UT Southwestern IRB according
`to Declaration of Helsinki principles.
`
`2.2. Cell preparation and bead sorting
`
`PBMC were isolated from whole blood using Ficoll Hypaque
`(GE Healthcare Biosciences, Pittsburg, PA) density gradient.
`In all cases, the 0 h, 4 h and 12 h specimens were processed
`simultaneously and the 24 h and 72 h specimens were
`processed independently. This design was based on initial
`stability studies for ex vivo subset quantification (not shown).
`From PBMC preparations, purified C08+, C014+ and CD19+
`cells were isolated using respective Miltenyi microbead positive
`selection kits. The C019 depleted fraction was used for positive
`selection of BDCA1+ (MDC), and BDCA4+ (PDC) populations
`using respective microbeads. "Untouched" CD4+ T cells were
`then isolated using negative selection kits. C025+ T-cells were
`positively sorted from the purified CD4+ fraction using C025
`microbeads. To prepare third party Teff (CD4+CD25-) cells
`and APC, PBMC were isolated from buffy coats of healthy
`donors using Ficoll Hypaque. APC fraction was prepared by
`depleting CD3 + T cells from PBMC using CD3+ microbeads.
`CD4+CD25— (responder) cells were obtained by negative
`sorting for CD4+ T cells followed by depletion of C025+
`cells. Both responder cells and APC were stored in freezing
`media in liquid nitrogen until further use in multiple assays.
`All magnetic microbeads were purchased from Miltenyi
`Biotech
`(Auburn,
`CA)
`and
`used
`according
`to
`the
`manufacturer‘s instructions, resulting in population purities
`>90—9596.
`
`2.3. CFSE staining
`
`Third party CD4+CD25+ responder cells used in suppression
`assays ware labeled with carboxyfluorescein diacetate suc-
`cinimidyl ester (CFSE) (lnvitrogen Molecular Probes, Eugene,
`OR), as described previously [14]. Briefly cells were suspended
`at 1 x 10" cells/mL and incubated for 7 min at 37 ”C with
`0.25 (1M CFSE, then washed twice with media containing 5%
`heat inactivated (HI) human serum.
`
`2.4. Flow cytometry based suppression assays
`
`Allogeneic 0.1 x 10" CFSE-stained CD4+C025 — (Terr) cells
`and APC were co-cultured with varying ratios of CD4+C025+
`or CD8+ cells isolated from study participants in an anti-CD3
`coated 96 Well round bottom plates (Costar, Corning, NY) for
`6 days. Cells were subsequently washed and stained for flow
`cytometry, as described later. Suppressive potential of
`regulatory T cells was assessed by normalizing the percent
`proliferative responses (%CFSE low CD4+) to the response
`without suppressors (defined as 100%), as described before
`[15].
`
`Page 2 of 15
`
`

`

`107
`GA-induced early immune changes
`
`2.5. Induced suppression assay
`
`2.9. Statistical analysis
`
`Induced suppressors from project participants were gener-
`ated by culturing 2 x 10" CD4+CDZS— T cells with plate
`bound anti-CD3 for 6 days, as described previously [16]. At
`the end of
`the culture period,
`the induced cells were
`harvested and washed twice and then co-culturecl in varying
`ratios with CFSE-labeled allogeneic third party CD4+C025—
`responder cells and APC in anti-CD3 coated round bottom
`plates for 6days. Cells were subsequently washed and
`stained for flow cytometry, as described below. Inhibitory
`potential of induced suppressors was evaluated by normal-
`izing the percent proliferative responses (%CFSE low CD4+)
`to the response without suppressors (defined as 100%).
`
`2.6. Mixed lymphocyte reaction
`
`Antigen presenting cells (CD14+ Et CD19+) and responder
`cells were isolated from PBMC by using microbeads as described
`earlier. APC were collected from project participants, while
`the responder cells were obtained from third party donors. For
`the allogeneic response assay, APC and responder cells were
`incubated together at a 1:10 (APC:T9«) ratio in a 96-well plate
`(Costar, Corning, NY), as described previously [17]. The MLR
`was harvested after 6 days in culture and stained for flow
`cytometry. APC stimulatory capacity was evaluated by enu-
`merating C025 expression and proliferation on responding T
`cells [17]. Additionally, responder cells isolated from project
`participants were co-cultured with APC from third party donors
`to assess the T cell activation potential of MS patients on GA
`therapy as compared to controls.
`
`2.7. Flow cytometric antibody staining
`
`For all the subjects, PBMC subset expreSsion markers were
`evaluated on freshly isolated samples (1 x 10" cells/tube).
`Anti-human antibodies used for multi-color flow cytometric
`analysis included: CD4-PeGC-Cy5.5, CDB-AmCyan, CD45RO-
`Pacific Blue, CDZ7-FITC, CD19-PE-Cy7, CDi4-PerCP, BDCAi-
`APC, and BDCA4-PE. All antibodies Were obtained from BD
`Biosciences (San Jose, CA). Cells from suppression assays
`and MLR were washed with 0.1% (w/v) sodium azide/
`phosphate-buffered saline (Mediatech Cellgro) and stained
`with anti-human CD4-PE & CD25-APC, then resuspended in
`1% paraformaldehyde
`(Electron Microscopy Sciences,
`Hatfield, PA). Flow cytometric data were acquired on a
`4-Laser LSRII using FACSDiva software (Becton Dickinson).
`Data were analyzed using FlowJo (Tree Star, Ashland, OR).
`
`2.8. Antigenic stimulation
`
`2 x 105 B cells, monocytes and MDC were separately
`placed in 96-Well
`round bottom plates and stimulated
`with Staphylococcus aureus (pansorbin) (1:1000 dilution),
`LPS (5 pg/mL) and Poly I:C (1 pg/mL) respectively, for 48 h
`in activation media (RPMI + 5% HI human serum and 5 mM
`Pen/ Strep). All culture supernatants were harvested and were
`stored at —80 ”C for cytokine determination using multiplex
`MSD cytokine kits (Meso Scale Discovery, Gaithersburg, MD).
`Cytokines evaluated included TNF-u, |L-10, GMCSF, |L-8, |L-1is,
`iL-6, lL-12, and |L-2.
`
`Page 3 0f 15
`
`Data between the groups was analyzed with unpaired two-
`tailed Student‘s t-test and p < 0.05 was considered significant.
`Paired two-tailed Student's t-test was used to compare data
`within a group and p < 0.05 was considered significant.
`
`3. Results
`
`3.1. Subject characteristics and study design
`
`This study was designed as the first comprehensive evaluation
`of longitudinal changes in phenotype, quantity, and effector
`functions of immune cells in peripheral blood of MS patients
`within the first 72 h of GA therapy. The subjects comprised 12
`RRMS patients and 7 healthy donors (HD). Both the GA treated
`(n = 8) and untreated (n = 4) MS patients were in the quiescent
`phase of relapsing remitting disease {not in active relapse)
`and were treatment naive (no history of disease modifying
`therapy). The healthy subjects and the untreated subjects
`served as important cohorts to control for potential diurnal
`variation of measured parameters. Thus, only the parameters
`that changed in the GA-treated cohort but not in the other two
`cohorts were considered an effect of GA therapy.
`The MS groups consisted of 2 males/8 females and 4 females
`respectively, with an age range and mean age of 22759
`(42 years) and 38—56 (46.3 years). The HD cohort included 3
`males/4 females, 27—54 years (mean: 37.3 years). Peripheral
`blood was collected just before treatment initiation and this
`was considered time point 0(h). Patients in the GA cohort
`initiated their GA therapy as part of an overnight admission in
`the UT Southwestern CTRC. Clinical oversight with injection
`training was provided as part of routine care procedures. Blood
`samples were then collected at 4 h, 12 h and 24 h from
`the time of GA injection, as part of the admission. The
`24 h time point was collected before the administration of
`the second injection. Subjects then returned for the 72 h
`time point before their fourth dose of GA. Throughout the
`study, untreated and GA-treated MS patients were pooled
`together for baseline (0 h) comparisons with HD. At baseline,
`percentages and absolute numbers of CD4+, CD8+, CDi4+,
`CD19+, myeloid dendritic cells (MDC) and plasmacytoid
`dendritic cells (PDC) were not different betWeen HD and MS
`patients (Figs. 1A, B and C). We then sought to evaluate
`whether GA induced any phenotypic, quantitative, and/or
`functional changes in these cell populations within 72 h of
`therapy initiation.
`
`3.2. GA induces a rapid increase in CD8 + T cell
`percentages in the blood of MS patients
`
`The percentage of CD4+ T lymphocytes in peripheral blood
`of GA-treated MS patients remained stable during the first
`72 h of therapy, except for a slight but significant increase
`at 4 h (GA-CD4+ T cells, 0 h vs. 4 h, p < 0.01, Fig. 2A). Of
`note, a similar increase in the percentage of CD4+ T cells
`was also seen in the peripherai blood of healthy donors
`(HD-CD4+ T cells, 0 h vs. 4 h, p < 0.01, Fig. 2A), and
`therefore did not represent a GA-selective effect. interest-
`ingly, GA-treated MS patients showed a significant increase
`
`Page 3 of 15
`
`

`

`
`108
`C.L. Ayers et al.
`
`0
`-=
`E60
`to
`0
`5"”
`1:
`0
`“20
`E,
`'8 o
`32
`
`+
`E
`O
`
`+
`B
`O
`
`+
`93
`D
`U
`
`:20
`O
`1515
`0
`III
`E13
`fl-
`"5
`3:
`
`1
`o
`
`+
`.‘I
`D
`U
`

`

`
`1.
`
`5
`g
`u 1.0
`m 05
`g
`’
`3
`.3.
`Eonzs
`<
`
`IHD
`D MS
`
`I... an __ _
`
`+
`3
`U
`
`+
`3
`U
`
`+
`92
`D
`U
`
`+
`3
`D
`0
`
`E
`
`g
`
`Similar percentages and absolute numbers of T, B and antigen presenting cells at baseline. PBMC from healthy donors and
`Figure 1
`treatment-naive MS patients were analyzed by flow cytometry for determining the frequency (A Et B) and absolute numbers (C) of
`CD4+, CD8+, CD14 +, CDi9+, BDCA1 + (myeloid dendritic cells) and BDCA4+ (plasmacytoid dendritic cells) cells at the O h time point.
`
`HD
`
`MS (Untreated)
`
`p<0.0‘|
`
`MS (GA)
`
`A
`p<o.o1
`no
`5
`In
`$060 I
`3% a; l'
`'
`#240
`3%
`0220
`;E
`
`‘
`"33 an: act
`‘
`v
`o
`
`a?
`'
`O * ‘
`e -
`
`’
`‘5‘
`t .
`
`so
`60
`
`40
`
`20
`
`c
`
`
`
`
`on
`
`4n
`
`12h
`
`24:1
`
`72h
`
`on
`
`4n
`
`12h
`
`24h
`
`72h
`
`on
`
`4n
`
`12h
`
`24h
`
`72h
`
`on
`
`an
`
`12h
`
`24h
`
`72h
`
`on
`
`4h
`
`12?]
`
`24h
`
`72h
`
`on
`
`4h
`
`12h
`
`24h
`
`72h
`
`52aDp
`
`—+ODo3
`
`3
`
`C
`.020
`1'6
`'='15
`$
`i—1o
`
`g 5
`"g
`o
`
`20
`
`15
`
`10
`
`5
`
`u
`
`O
`.
`I
`file-rater?
`on
`431
`12h
`24h
`72h
`
`
`
`'
`
`'
`
`.
`
`% V %‘
`"-u‘fi.
`on
`4n
`12h
`24h
`72h
`
`
`
`on
`
`an
`
`12h
`
`24h
`
`72h
`
`E) MS (GA)
`D) MS (GA)
`3‘”
`[343.05
`25
`i—P—lm'us
`'
`|
`Ea “é
`gaze
`a, W =
`£815
`85
`.|: WA )— R
`$110
`004
`DE
`D
`02‘ 5 5 :
`o 2 §
`g D
`325
`U
`
`0
`
`0h
`
`12"!
`
`CH1
`
`12h
`
`GA treatment reduces CD4/CD8 T cell ratio in the peripheral blood 12 h post injection. CDé+ (panel A), CD8+ (panel B)
`Figure 2
`percentages and CD4/C08 T cell ratio (panel C) were determined in the peripheral blood of HD, untreated and (SA-treated MS patients
`at indicated time points. Each point represents a single subject. Panels D and E show trends for CD8+ T cell percentages and CD4! CD8
`ratio. respectively, at 0 vs. 12 h in the (SA-treated group. Significant p values are indicated.
`
`Page 4 of 15
`
`Page 4 of 15
`
`

`

`109
`GA—induced early immune changes
`
`in C08 + T cell percentages at 12 h post‘first injection when
`compared to 0 h time point (p< 0.01, Figs. 28 and D).
`Concomitantly, the CD4/ CD8 T cell ratio was significantly
`reduced at 12 h in the peripheral blood of GA-treated
`patients (p < 0.05, Figs. 2C and E). No changes in C08+ T cell
`percentages were seen in the peripheral blood of HD or
`untreated MS patients. Within the patient cohort, increased
`CD8+ T cell percentages and reduced CD4/ C D8 T cell ratio at
`12 h were seen in the blood of 6/7 GA-treated MS patients
`(Figs. 2D and E, respectively). At the 24 h and 72 h time
`points, both representing "trough” points for drug (right
`before daily shots), the cell percentages were somewhat
`elevated but not significantly different from 0 h.
`
`3.3. GA predominantly alters CD8 + T cell homeostasis
`in the peripheral blood 12 h post injection
`
`To further characterize the early effects of GA on peripheral T
`cells, percentages of naive (CDZ7+CD45R0—), central memory
`(CM; CDZ7+CD45RO+), effector memory (EM; CDZYFCD45RD+)
`and terminal (Ter; CD27—CD45RO—) CD4+ and CD8+ T-cells
`were examined in the peripheral blood of MS patients treated
`with GA. Gating strategy for these populations is shown in
`
`Suppl. Fig. 1. In CD4+ T cells of MS patients treated with GA, the
`percentages of EM and Ter CD4+ T cells were significantly
`increased at 12 h (after first GA injection) when compared to
`the 0 h time point (GA-EM CD4+ Tcells, p < 0.05; GA-Ter C04+
`T cells, p < 0.05, Fig. 3C). While HD also showed a significant
`increase in EM CD4+ T cells at 12 h as compared to 0 h (HD-EM
`CD4+ Tcells, p < 0.05, Fig. 3A), the increase in Ter CD4+ T cells
`at 12 h was selective to GA—treated MS patients. The only
`diumal fluctuation in CD4+ T cell compartment of untreated MS
`patient was a significant decrease in EM population at 24 h
`when compared with O h time point (MS—untreated EM CD4+ T
`cells, 0 h vs. 12 h, p < 0.05, Fig. 3B).
`GA-treated MS patients, selectively, showed interesting
`changes in the peripheral blood CD8+ T cell homeostasis
`within 12 h of therapy initiation. Naive CD8+ T cells Were
`significantly decreased at 12 h when compared to 0 h in the
`blood of GA-treated MS patients (GA~Nai've CD8+ T cells, 0 h
`vs. 12 h, p < 0.05, Fig. 4C). Further, at4 h, EM CD8+ Tcells
`showed a decreasing trend (as compared to 0 h)
`in GA
`treated and untreated MS patients and this was in contrast to
`increasing trend of EM CD8+ T cells from HD (Figs. 4A vs. B E C).
`While the decreasing trend in the EM CDB + T cell population in
`untreated MS patients became significant at 12 h (MS-untreated
`EM C084» T cells, 0 h vs. 12 h, p < 0.05, Fig. 4B), notably, the
`
`
`
`["1
`
`4!!
`
`12h
`
`24h
`
`72h
`
`0h
`
`4h
`
`12h
`
`24h
`
`72h
`
`B) MS (Untreated)
`5
`
`an;ace%InCD4+Tcell 33 6
`
`
`Of!
`
`4n
`
`12h
`
`24h
`
`72h
`
`0h
`
`4h
`
`12h
`
`24h
`
`72h
`
`0) Ms (GA)
`5
`«3
`acoo
`
`“AInCD4+Tcell 38 D
`
`on
`
`4h
`
`12h
`
`24h
`
`72h
`
`0
`
` 12h
`
`an
`
`All
`
`24h
`
`72h
`
`
`
`Terminal
`
`12h
`
`24h
`
`0h
`
`4h
`
`?2h
`
`0h
`
`4h
`
`12h
`
`24h
`
`72h
`
`0h
`
`4h
`
`12!!
`
`24h
`
`72h
`
`p<0.05
`
`p<0.05
`
` 0h
`
`4!!
`
`12h
`
`24h
`
`72h
`
`Uh
`
`4h
`
`12h
`
`24h
`
`72h
`
`Figure 3
`
`No GA-induced changes in CD4+ Tcell compartment during the first 72 h of therapy. Fluorescent conjugated anti-human C027
`and CD45R0 were used for flow cytometry analysis to further differentiate CD4+ T cells into naive (CD27+CD45RO+), central memory
`(CM, CD27+CD45RO+), effector memory (EM, CD27—CD45RO+) and terminal (Ter, CD27—CD45RO +) populations (Suppl. Fig. 1). Panels A, B
`and C represent percentages of naive, CM, EM and terminal CD4+ Tcells in HD, untreated, and (SA-treated MS patients, respectively. Except
`for the terminal CD4+ T cells, similar diurnal fluctuations in CD4+ T cell subpopulations were present in all the study groups.
`
`Page 5 0f 15
`
`Page 5 of 15
`
`

`

`
`
`C.L. Ayers et al.
`
`
`
`0h
`
`4“
`
`12h
`
`24h
`
`72"
`
`0h
`
`4h
`
`12h
`
`24h
`
`72“
`
`“loInCDB+Tcell
`
`30
`
`20
`
`1|]
`
`DI'I
`
`4h
`
`12h
`
`24h
`
`72h
`
`Terminal
`
`“0.05
`l L..
`a¥§¥é
`0h
`4h
`12h
`24h
`72h
`
`
`
`30
`
`oo
`40
`
`
`
`
`B) MS (Untreated)
`5 .s
`canon
`
`I
`
`4 4
`
`"loInCDB+Tcell 33 m +1 H—i
`
`
`on
`411
`12h
`24h
`72h
`on
`411
`12h
`24h
`7211
`0h
`4h
`12h
`24h
`72h
`01]
`4h
`12!]
`24h
`72h
`
` Oh
`
`0h
`
`4h
`
`12h
`
`24h
`
`72!!
`
`4h
`
`12h
`
`24h
`
`72h
`
`C) MS (GA)
`2100
`'6o
`s...
`
`
`
`on
`
`an
`
`12h
`
`24h
`
`7211
`
`on
`
`41-:
`
`12h
`
`24h
`
`7211
`
`+o
`
`nOO Ea
`
`?
`
`Figure 4
`Frequency of 038+ T cell subpopulations is rapidiy altered in the blood post GA therapy. Fluorescent conjugated
`anti-human C027 and CD45RO were used for flow cytometry analysis to further differentiate C08+ T cells into naive (CDZ7+CD45RO—),
`central memory (CM, CDZ7+CD45RO+), effector memory (EM, CD27—CD45RO+) and terminal (Ter, CD27—CD45R0—) populations
`(Suppl. Fig. 1). Panels A, B and C represent percentages of naive, CM, EM and terminal CD4 + T cells in HD, untreated, and GA-treated
`MS patients, respectively. GA induced significant decrease in naive (panel C) and increase in EM (panel C) CD8 + T cells in the blood as
`early as 12 h after therapy initiation. No such changes in (208+ T cell subpopulations were seen in HD (panel A) or untreated MS
`patients (panel B). Significant p values are indicated.
`
`initial decrease in EM CDB+ T cells in GA-treated MS patients
`was followed by a significant rebound at 12 h (GA-EM CDS+ T
`cells, 4 h vs. 12 h, p < 0.01) post first GA injection (Fig. 4C).
`Fluctuations in the terminal CDB+ T cell compartment were
`seen in all three groups (Figs. 4A, B and C).
`
`3.4. Ex vivo suppression of allogeneic responder]
`effector T cells (Teff) by CD4+CD25 + T,egs obtained
`from GA-treated MS patients remains unaltered
`during the first 72 h of therapy
`
`To analyze the effects of GA on suppressive function of
`CD4+CD25 + regulatory T cells (ngs) during the first 72 h of
`therapy, Te" (CD4+CDZS—) and irradiated APC, obtained
`from third-party healthy volunteers, were co-cultured with
`Tress isolated from peripheral blood of either MS patients
`treated with GA or control cohorts. Representative dot plots
`are shown in Fig. 5A. Surprisingly, at 0 h the mean sup-
`pression index of CD4+CDZS+ ng cells in MS patients and HD
`was not significantly different
`(Suppl. Fig. 2). Moreover,
`there were no changes in percent suppression of anti-CD3
`stimulated ailogeneic Tefg cells by CD4+C025 + Tregs obtained
`
`either from (SA-treated MS patients or controls during the
`first 72 h of therapy (Fig. 53).
`
`3.5. CD8 suppressor ability is significantly improved
`as early as 12 h after the first GA injection
`
`Expansion of GA-specific suppressor 038+ T cell responses
`has been shown in MS patients on GA therapy [7,9],
`implicating a role of CDB+ T cells in GA-mediated immune
`modulation of MS. To test whether improvement in giobal
`suppressive function of CD8+ T cells is initiated as early
`immune change during GA therapy, suppression of anti-CD3-
`stimulated allogeneic Te” cells was carried out with CD8+ T
`cells sorted from GA-treated MS patients or control subjects.
`Representative dot plots of suppression of allogeneic Teff
`cells by CD8 T cells obtained from a GA-treated patient are
`shown in Fig. 6A. Similar to CD4+CDZS+ Tregs, at 0 h, CDS+
`suppressors from MS patients and HD displayed no significant
`difference in the mean suppression indices (Suppl. Fig. 2).
`lmportantty, there was a significant enhancement of (208+ T
`cell-mediated suppression at 12 h after first GA injection
`(%suppression GA, 0 h vs. 12 h, p < 0.05, Figs. 63, C and D).
`
`Page 6 0f 15
`
`Page 6 of 15
`
`

`

`GA-induced early immune changes
`
`111
`
`A
`
`1 :0
`
`1 :0.125
`
`
`
`aQ
`
`Proliferation: 38%
`
`Proliferation: 36%
`Proliferation: 32%
`
`
`
`Suppression: 17%
`Suppression: 31%
`Suppression: 22%
`
`
`WW
`wool
`
`
`
`
`’3
`
`CH
`
`Proliferation: 31% Proliferation: 28%
`
`Suppression: 25% Suppression: 33%
`
`
`
`
`Proliferation: 46%
`
`Proliferation: 42%
`
`GA 0h
`
`GA 12h
`
`CD4 {
`
`n
`
`
`CI l
`“InSuppressio
`
`1 _-.,
`
`
`Proliferation: 36%
`
`
`Suppression: 15%
`
`CFSE
`
`
`
`
`
`MS(GA)
`
`
`
`A a o
`.-
`o.
`a it it .. e-
`
`
`
`
`12h
`24h
`72h
`Uh
`4h
`12h
`24h
`72h
`0h
`4h
`
`a OD
`
`on
`
`4h
`
`12h
`
`24h
`
`72h
`
`Figure 5
`Ex vivo suppression by CD4+CDZS+ Tregs remains unaffected during the first 72 h of GA therapy. Increasing numbers of
`CD4+CD25+ ngs, isolated from the indicated cohorts were used in anti-CD3 stimulated CFSE-based suppression assays using
`third-party Teff cells and irradiated APC. Representative clot plots from a GA-treated patient are shown in panel A. Proliferation
`percentages in the dot plots indicate gated percentages of CD4 +/ CFSE low (proliferating) cells. Percent proliferative responses were
`normalized to the response without suppressors (defined as 100%), indicated as 1:0 in the dot plots and %suppression was calculated
`(also indicated). Panel B represents %suppression by CD4+CD25+ ng cells isolated from HD, untreated and GA-treated MS patients.
`
`Additionally, suppression remained elevated in the GA group
`at 24 and 72 h (as compared to 0 h), although the difference
`did not reach statistical significance at these "trough" time
`points early in therapy (mean %suppression GA, 0 h vs. 24 h &.
`72 h, 21.5 1 45.9 vs. 34.9 :19.0 &. 33.4 s 29.8). At 12 h, 6/7
`GA treated patients had improvement in CD8+ suppressive
`potential as shown in Fig. 6D. The control cohorts showed no
`differences in CD8 suppressive ability over time (Fig. 6C).
`
`3.6. Significant improvement in the inhibitory
`potential of induced suppressors at 12 h post
`GA therapy
`
`Several studies from us and others have shown that human
`CD4+CDZ5— T cells, upon activation, can be converted into
`CD4+CDZS+FOXP3+ cells and acquire suppressive potential
`[16,18,19]. To test whether GA therapy altered the ability
`of induced Treg generation, we assessed the ability of anti-CD3-
`stimulated CD4+CD25 * T cells from GA- treated MS patients and
`controls to suppress allogeneic Teff cells. CD4+CD257 T cells
`sorted from the peripheral blood were cultured with irradiated
`(third party) APC in the presence of plate bound anti~CD3.
`Cells were subsequently washed and used as suppressors in a
`
`secondary in vitro suppression assay. Representative dot
`plots of induced suppressors from a GA-treated patient
`co—cultured with allogeneic Te” cells are shown in Figs. 7A
`and B. Interestingly, at the untreated state (0 h], induced
`suppressors generated from MS patient's CD4+CD257 cells
`had significantly reduced Suppressive potential towards
`allogeneic Te” cells, compared to healthy donors (Fig. 7C).
`Importantly, the suppressive ability of CD4+CD25+ Tregs
`generated from CD4+CD25— T cells from (SA-treated MS
`patients was significantly improved at 12 h (%suppression
`GA, 0 h vs. 12 h, p < 0.05, Fig. 70). Within the cohort, 5/6
`patients showed improvement in induce Treg suppression
`ability (Fig. 7E). induced suppressors generated from the
`control cohort CD4+CDZS — Ten cells displayed no significant
`differences over time in suppressing third party Te" cells
`(Fig. 7D).
`
`3.7. Similar activation profiles of CD4+CDZS— T cells
`from GA-treated MS patients and HD when cultured
`with third party allogeneic APC
`
`The effect of GA on the activation potential of CD4+CDZS— Te”
`cells in response to allogeneic APC was tested in an MLR. C025
`
`Page 7 0f 15
`
`Page 7 of 15
`
`

`

`
`
`C.L. Ayers et al.
`
`B) GA 12h
`
` 0
`
`MS (GA)
`
`ms (GA)
`
`
`
`
`
`_%Suppression Sa8
`
`.n OO
`0h
`4!!
`12h
`24h
`72h
`0h
`4h
`
`Suppression2‘iiaaa
`"A.
`
`€41.05
`
`
`r-—'—"‘_—.
`/'
`
`uh
`
`uh
`
`12h
`
`2411 no
`
`12!!
`
`24h
`
`72h
`
`031
`
`4“
`
`Figure 6
`Significant improvement in CDS+ T cell suppressive potential within 12 h of GA therapy initiation. Ex vivo purified CDB+
`cells were used in anti-CD3-stimulated CFSE-based suppression assays using third-party Te" cells and irradiated APC. Panels A and B
`show representative dot plots from a GA-treated patient at 0 h and 12 h, respectively. Percentages in dot plot indicate gated
`percentages of CD4 +/CFSE low (proliferating) cells. Percent proliferative responses were normalized to the response without
`suppressors (defined as 100%), indicated as 1:0 in the dot plots. Panel C represents %suppression by C08+ T cells isolated from HD,
`untreated, and GA-treated MS patients. GA-treateci patients showed significant improvement in CD8+ T cell mediated suppression at
`12 h (D). No such change was seen in HD and untreated MS patients. Per patient trends in GA treated group at 12 h are shown in D.
`
`upregulation response of CD4+ T cells, after co-culture with
`allogeneic APC, from a representative (SA-treated MS patient
`is shown as a dot plot in Suppl. Fig. 3A. At baseline, CD4+ T
`cells from MS patients and HD showed similar activation by
`allogeneic APC (Suppl. Fig. BB). Further, following GA therapy,
`there were no significant changes in the MLR responses of
`CD4+ T cells from any of the cohorts (Fig. 8).
`
`3.8. Reduced stimulatory potential of MS patient
`monocytes within 12 h of GA treatment
`
`Several reports have implicated immune modulatory effects
`of GA on APC populations such as reduced production of
`proinflammatory cytokines [L-12, TNF-a and elevated anti-
`inflammatory IL— 10 from monocytes isolated from GA-treated
`MS patients {20,21}. GA therapy-induced increase in lL-10
`from B cells has also been reported. We sought to evaluate
`GA-induced phenotypic and functional changes in APC during
`the first 72 h of therapy.
`lmmunophenotyping revealed no
`significant differences in total monocytes, B cell, MDC, and PDC
`percentages or absolute numbers in MS patients at baseline,
`when compared to HD (Figs. 13 & C). Moreover, unlike the
`dynamic changes in certain T cell subsets, the percentages of
`APC subsets in the peripheral blood remained relatively stable
`in all three groups when compared to baseline levels (Fig. 9),
`with no statistically significant increase or decrease at any time
`point.
`
`In addition to numbers, we also tested the capacity of
`monocyte and B cells to stimulate MLR responses to evaluate
`whether GA induces functional changes in the APC activating
`potential within 72 h of treatment. Representative dot plots
`of C025 upregulation by third party CD4+CD25- Te” cells
`when cultured with GA patient's monocytes and B cells are
`shown in Figs. 10A and B, respectively. Monocytes from HD
`vs. untreated MS patients at baseline (0 h) displayed no
`significant differences in their stimulatory capacity (Suppl.
`Fig. 3C). Importantiy, while control groups did not show any
`variation in the monocytes stimulatory capacity over time,
`there was an appreciable trend of reduced activation potential
`of monocytes in GA-treated MS patients at 12 h (GA monocyte
`MLR, 0 h vs. 12 h, p = 0.06, Fig. 10C). Within the cohort, the
`activation potential of monocytes showed a downward trend in
`7/ 8 patients at 12 h post-first GA injection (Fig. 10E). Similar to
`monocytes, B cells from MS patients and HD at 0 h showed no
`significant difference in overall stimulatory capacity (Suppl.
`Fig. 3C). Furthermore, there were no significant changes in B
`cell stimulatory potential over time when compared to baseline
`levels after GA therapy (Fig. 10D).
`
`3.9. Significant increase in lL-10 and TNF-u from
`monocytes early during GA therapy
`
`Cytokine levels were determined in culture supernatants of
`monocytes, B cells and MDC obtained from GA-treated MS
`
`Page 8 0f 15
`
`Page 8 of 15
`
`

`

`GA-induced early immune changes
`
`113
`
`C
`
`g so
`E 40
`_§In
`
`O
`
`g3
`
`A) GA Oh
`B) GA 12h
`
`1:0
`1:0.125
`1:0
`I
`i
`2“
`1
`./
`I a-~
`
`_4
`
`w
`1
`W $20
`
`
`
`
`
`
`
`D
`100
`5
`a so
`t
`‘1 0
`g
`a; -50
`32
`.190
`
`100
`
`D
`
`5
`a so
`s
`'6.
`n.
`3 .50
`3-3
`-100
`
`on
`
`4h
`
`12h
`
`2

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