throbber
Downloaded from https://ashpublications.org/blood/article-pdf/113/16/3865/1307974/zh801609003865.pdf by NEW YORK UNIVERSITY user on 20 January 2020
`
`TRANSPLANTATION
`
`Altered B-cell homeostasis and excess BAFF in human chronic graft-versus-host
`disease
`Stefanie Sarantopoulos,1 Kristen E. Stevenson,2 Haesook T. Kim,2 Corey S. Cutler,1 Nazmim S. Bhuiya,1
`Michael Schowalter,1 Vincent T. Ho,1 Edwin P. Alyea,1 John Koreth,1 Bruce R. Blazar,3 Robert J. Soiffer,1 Joseph H. Antin,1
`and Jerome Ritz1,4,5
`
`1Division of Hematologic Malignancies and Department of Medical Oncology and 2Department of Biostatistics and Computational Biology, Dana-Farber Cancer
`Institute, Boston, MA; 3Cancer Center and Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota, Minneapolis; 4Cancer
`Vaccine Center, Dana-Farber Cancer Institute, Boston, MA; and 5Harvard Stem Cell Institute, Harvard Medical School, Boston, MA
`
`Chronic graft-versus-host disease (cGVHD)
`causes significant morbidity and mortal-
`ity in patients otherwise cured of malig-
`nancy after hematopoietic stem cell
`transplantation (HSCT). The presence of
`alloantibodies and high plasma B cell–
`activating factor (BAFF) levels in patients
`with cGVHD suggest that B cells play a
`role in disease pathogenesis. We per-
`formed detailed phenotypic and func-
`tional analyses of peripheral B cells in 82
`patients after HSCT. Patients with cGVHD
`Introduction
`
`had significantly higher BAFF/B-cell ra-
`tios compared with patients without
`cGVHD or healthy donors.
`In cGVHD,
`increasing BAFF concentrations corre-
`lated with increased numbers of circulat-
`ing pre–germinal center (GC) B cells and
`post-GC “plasmablast-like” cells, sug-
`gesting in vivo BAFF dependence of these
`2 CD27ⴙ B-cell subsets. Circulating CD27ⴙ
`B cells in cGVHD comprised in vivo acti-
`vated B cells capable of IgG production
`without requiring additional antigen stim-
`
`ulation. Serial studies revealed that pa-
`tients who subsequently developed
`cGVHD had delayed reconstitution of
`naive B cells despite persistent BAFF
`elevation as well as proportional increase
`in CD27ⴙ B cells in the first year after
`HSCT. These studies delineate specific
`abnormalities of B-cell homeostasis in
`patients with cGVHD and suggest that
`BAFF targeting agents may be useful in
`this disease. (Blood. 2009;113:3865-3874)
`
`The limited understanding of the immune mechanisms that result in
`chronic graft-versus-host disease (GVHD) hinders our ability to
`develop effective targeted therapies that would improve the sur-
`vival of patients undergoing allogeneic hematopoietic stem cell
`transplantation (HSCT).1-3 In acute GVHD, tissue injury is medi-
`ated primarily by donor T cells that specifically target minor
`histocompatibility antigens (mHAs) in affected organs.4,5 However,
`despite effective prevention of acute GVHD with agents that
`primarily inhibit T cells (calcineurin inhibitors, mTOR inhibitors,
`and purine analogues),2,6 the incidence and severity of chronic
`GVHD (cGVHD) remain high.1,2 This suggests that
`immune
`mechanisms of cGVHD are distinct from acute GVHD. Studies in
`mHA-mismatched murine transplantation models demonstrate in-
`volvement of B cells in the development of cGVHD.7,8 After
`allogeneic HSCT in humans, alloantibodies to Y chromosome–
`encoded proteins are detectable in 50% of male recipients if they
`received hematopoietic stem cells from a female donor.9 Such
`alloantibodies develop 4 to 9 months after HSCT, and the presence
`of alloantibodies correlated significantly with clinical cGVHD
`development.10 HY antibodies were not associated with acute
`GVHD and did not develop when recipient and donor were sex
`matched. When studied in greater detail, patients with HY antibod-
`ies were also found to have coordinated T-cell responses to distinct
`epitopes in the same HY mHA (DBY).11 These studies have
`suggested that donor B cells play a role in the development of
`cGVHD in humans, and several phase 1 or 2 trials of B cell–
`
`directed therapy with rituximab in steroid-refractory cGVHD have
`revealed clinical responses.12-14 Although these observations pro-
`vide compelling evidence that B cells play an important role in the
`immune pathology of human cGVHD,
`the mechanisms that
`promote and sustain B-cell involvement have not been elucidated.
`HSCT conditioning depletes normal recipient B cells and the
`expansion of normal donor B cells is altered as reconstitution
`occurs in the setting of constant exposure to “foreign” minor
`histocompatibility antigens. Although genetic disparity between
`donor and recipient must exist for cGVHD to develop, sustainable
`autoreactivity occurs after alloreactivity in murine models. Along
`with a more prolonged humoral immune deficiency, a complex
`autoimmune disease–like phenotype is found in cGVHD.15,16 The
`frequent production of autoantibodies in patients with cGVHD17-19
`suggests that disease pathogenesis reflects a critical breakdown in
`peripheral B-cell tolerance after allogeneic HSCT. Furthermore, a
`relative decrease in immature B-cell number in cGVHD compared
`with other post-HSCT patients20 suggests that altered B-cell
`homeostasis is a component of this disease.
`B cell–activating factor (BAFF) plays a critical role in B-cell
`reconstitution and homeostasis after myeloablation.21 Studies of
`B-cell development in murine models have shown that survival of
`normal mature B cells depends on the relative balance of both
`B-cell receptor (BCR) and BAFF signaling.22,23 In addition,
`maintenance of B-cell homeostasis in mice depends on in vivo
`soluble BAFF concentration.24,25 Murine models demonstrate that a
`
`Submitted September 30, 2008; accepted January 11, 2009. Prepublished
`online as Blood First Edition paper, January 23, 2009; DOI 10.1182/blood-
`2008-09-177840.
`
`payment. Therefore, and solely to indicate this fact, this article is hereby
`marked ‘‘advertisement’’ in accordance with 18 USC section 1734.
`
`The publication costs of this article were defrayed in part by page charge
`
`© 2009 by The American Society of Hematology
`
`BLOOD, 16 APRIL 2009 䡠 VOLUME 113, NUMBER 16
`
`3865
`
`SAN EX 1025, Page 01
`
`

`

`Downloaded from https://ashpublications.org/blood/article-pdf/113/16/3865/1307974/zh801609003865.pdf by NEW YORK UNIVERSITY user on 20 January 2020
`
`3866
`
`SARANTOPOULOS et al
`
`BLOOD, 16 APRIL 2009 䡠 VOLUME 113, NUMBER 16
`
`diverse B-cell pool is required for antigen-induced anergy and
`exclusion of autoreactive B cells from follicular niches.26,27 A set of
`elegant studies subsequently identified a BAFF-mediated B-cell
`tolerance checkpoint in which limiting amounts of BAFF are
`required for ongoing B-cell turnover and avoidance of B-cell
`autoreactivity.28,29 When the pool of normal B cells is reduced,
`excess BAFF promotes survival of autoreactive B cells.29 How-
`ever, even in the presence of high BAFF, high numbers of
`nonautoreactive B cells outcompete autoreactive B cells for avail-
`able soluble BAFF, causing autoreactive B cells to undergo apopto-
`sis.28 Thus, B-cell autoimmunity in transgenic mouse models is
`determined by both the level of soluble BAFF and the numbers of
`competing naive B cells.
`Defective censoring of autoreactive B cells in patients with
`systemic lupus erythematosus (SLE) and rheumatoid arthritis has
`been observed30-32 and high BAFF has been found in patients with a
`variety of autoimmune diseases,33-36 suggesting that excessive
`BAFF stimulation in humans contributes to development of
`autoimmunity. Since high BAFF levels are significantly associated
`with presence of active cGVHD more than 1.5 years after
`HSCT,37,38 we hypothesized that relative B lymphopenia and high
`BAFF after HSCT could support potentially pathologic activated
`alloreactive and autoreactive B-cell populations in cGVHD pa-
`tients. Examining peripheral blood after allogeneic HSCT, we
`found that patients with cGVHD had significantly higher “BAFF/B-
`cell ratio” compared with patients without cGVHD. Serial analysis
`of patients for 1 year after HSCT showed that, in patients without
`cGVHD, BAFF levels gradually normalized as B-cell numbers
`recovered, indicating a normal homeostatic response to B lymphope-
`nia. In contrast, patients who subsequently developed cGVHD
`exhibited delayed reconstitution of naive B cells despite persistent
`BAFF elevation. Using CD27⫹ as a marker of antigen experience
`and ex vivo IgG production as a measure of B-cell activation, we
`found that circulating B cells, including pre-GC B cells, were
`activated in cGVHD. These results suggest that altered B-cell
`homeostasis and excess BAFF contribute to promotion of activated
`B cells in patients with cGVHD.
`
`Methods
`
`Patient characteristics
`
`All patient samples were collected after written informed consent was
`obtained in accordance with the Declaration of Helsinki and approved by
`the Human Subjects Protection Committee of the Dana-Farber/Harvard
`Cancer Center.
`
`Patient group 1
`
`Blood samples for analysis of B-cell reconstitution were obtained from
`57 patients who had undergone allogeneic HSCT between 1994 and 2005
`and were more than 12 months after HSCT. Within this group, BAFF levels
`from 28 samples have been reported previously.37 Clinical characteristics of
`the 57 patients are summarized in Table 1. The study included patients who
`received nonmyeloablative or myeloablative conditioning regimens and
`bone marrow or mobilized peripheral blood stem cell grafts. Patients whose
`primary disease relapsed within 1 year of transplantation and patients
`receiving high-dose steroids (⬎ 0.5 mg/kg prednisone) at
`the time of
`sample collection were excluded. Chronic GVHD status at the time of
`sample collection was categorized according to documented clinical
`examination and laboratory studies using both Seattle criteria and National
`Institutes of Health (NIH) cGVHD consensus criteria. Twenty-two patients
`had “active cGVHD” at the time of primary analysis. This included patients
`with either limited or extensive cGVHD based on clinical severity and
`
`target organs affected per modified Seattle criteria.39 Twenty-three patients
`had “inactive cGVHD” at the time of sample collection. Inactive cGVHD
`was used to describe patients with cGVHD who had achieved a complete
`response to immune-suppressive therapy at the time of analysis. Patients
`who never developed cGVHD after HSCT were designated as having “no
`cGVHD.” Unlike patients with inactive or active cGVHD, patients with no
`cGVHD were receiving no immune-suppressive agents, except one patient
`who received low-dose steroids for a non-GVHD indication (Table 1).
`Median time of analysis was 20.9 months after transplantation for patients
`with active cGVHD compared with 26.6 months after transplantation for
`patients with no cGVHD (P ⫽ .10). Patients with inactive cGVHD were
`significantly later in their post-HSCT course (30.9 months) compared with
`patients with active cGVHD (20.9 months, P ⫽ .007). All other clinical
`characteristics including age, sex,
`type of transplant, and underlying
`hematologic malignancy were similar in patients with active and inactive
`cGVHD. We also studied 33 healthy controls.
`
`Patient group 2
`
`We prospectively followed an additional group of 25 patients from day of
`stem cell transplantation (day 0), with samples obtained every 3 months
`over a 19-month period. These patients received either myeloablative or
`nonmyeloablative conditioning regimens and tacrolimus and/or sirolimus
`as GVHD prophylaxis.6,40 Within this cohort, 8 patients did not develop
`cGVHD by 12 months after HSCT and 17 patients developed cGVHD
`between 3 and 12 months after HSCT. All patients in group 2 were
`analyzed, regardless of prednisone dose or treatment with other immunosup-
`pressive agents. Six of the 17 patients who developed cGVHD had prior
`grades 2 to 4 acute GVHD. None of the patients without cGVHD were
`taking immunosuppressive agents after the 6-month time point. Control
`blood samples were obtained from 8 patients with multiple myeloma after
`the second of a planned tandem autologous transplantation.
`
`Processing of patient plasma and peripheral blood cells
`
`Whole blood was drawn into standard EDTA-containing collection tubes.
`Plasma was separated from whole blood cells by centrifugation at 600g.
`Plasma was stored in aliquots at ⫺80°C and used after first thaw for BAFF
`measurements. Whole blood for flow cytometry studies was also collected
`on day of use in EDTA-containing tubes. Peripheral blood leukapheresis
`products were obtained from 2 cGVHD patients. Discarded leukocyte
`filters from anonymous healthy platelet donors were obtained from the
`Kraft Blood Donor Center at Dana-Farber Cancer Institute (DFCI).
`
`BAFF enzyme-linked immunosorbent assay
`
`Soluble BAFF in patient plasma samples was measured using a
`commercially available enzyme-linked immunosorbent assay (ELISA)
`and the manufacturer’s recommended procedures (R&D Systems,
`Minneapolis, MN).
`
`Flow cytometric analysis of peripheral B cells
`
`Antibodies used for flow cytometry were as follows: CD19 ECD or CD19
`PC7 (both clone J4.119), CD20 ECD (clone B9E9), CD38 PE (clone
`LS198), CD27 PCD5 (clone 1A4), and IgD FITC (clone IA6-2) from BD
`Biosciences Pharmingen (San Diego, CA); BR3-FITC (eBioscience, San
`Diego, CA); and TACI-PE (R&D Systems). Whole blood was processed for
`flow cytometry using the Prep Plus 2 system (Beckman Coulter, Fullerton,
`CA). The lymphocyte gate was established using forward and side scatter.
`A minimum of 50 000 lymphocytes were collected for all samples to ensure
`adequate subset analysis. Gates positive for CD19, IgD, CD38, and CD27
`were first set according to isotype controls. Only CD19⫹ cells were
`analyzed to ensure that CD3⫹ cells were not included in the analysis of
`CD38 or CD27 expression. Anti-CD19 PC7 (Beckman Coulter) was used
`per the manufacturer’s instructions. Red blood cells (RBCs) were lysed and
`leukocytes were fixed using the Beckman Coulter TQPrep system before
`analysis. Cells were analyzed using a Cytomics FC 500 instrument and
`CXP Analysis 2.0 software (Beckman Coulter). To determine BAFF
`receptor profiles, CD19⫹ cells positively selected from large volume
`
`SAN EX 1025, Page 02
`
`

`

`Downloaded from https://ashpublications.org/blood/article-pdf/113/16/3865/1307974/zh801609003865.pdf by NEW YORK UNIVERSITY user on 20 January 2020
`
`BLOOD, 16 APRIL 2009 䡠 VOLUME 113, NUMBER 16
`
`ALTERED B-CELL HOMEOSTASIS IN CHRONIC GVHD
`
`3867
`
`Table 1. Clinical characteristics of 57 patients who underwent allogeneic HSCT
`
`Chronic GVHD
`
`Characteristic
`
`No. (%)
`Median age, y (range)
`Female sex (%)
`Conditioning regimen (%)
`Myeloablative
`Nonmyeloablative
`Cell source (%)
`Peripheral blood stem cells
`Bone marrow
`HLA matching (%)
`Matched, unrelated
`Matched, related
`Mismatched
`Prednisone dose (%)
`0 mg/day
`0-30 mg/day
`Cellcept (%)
`Tacrolimus (%)
`Rapamycin (%)
`Months after transplantation, median (range)
`BAFF level, ng/mL, median (range)
`Grades II-IV aGVHD (%)
`Disease (%)
`AML/AML from MDS
`ALL
`CML
`CLL
`MDS
`NHL
`HL
`MM
`Other
`
`Active
`
`22 (45)
`46 (24-64)
`10 (45)
`
`14 (64)
`8 (36)
`
`19 (86)
`3 (14)
`
`10 (45)
`11 (50)
`1 (5)
`
`9 (30)
`13 (59)
`7 (32)
`8 (36)
`8 (36)
`20.9 (13.6-79.2)
`7.0 (1.6-24.5)
`2 (9)
`
`8 (36)
`2 (9)
`0 (0)
`3 (14)
`4 (18)
`2 (9)
`1 (5)
`0 (0)
`2 (9)
`
`Inactive
`
`23 (36)
`47 (19-66)
`11 (48)
`
`17 (74)
`6 (26)
`
`15 (65)
`8 (35)
`
`13 (57)
`9 (39)
`1 (4)
`
`10 (42)
`13 (57)
`5 (21)
`3 (13)
`3 (13)
`30.9 (16.7-134.6)
`5.5 (1.5-24.3)
`5 (22)
`
`5 (22)
`1 (4)
`9 (39)
`0 (0)
`4 (17)
`1 (4)
`0 (0)
`1 (4)
`2 (9)
`
`No
`
`12 (18)
`44 (25-58)
`4 (33)
`
`10 (83)
`2 (17)
`
`9 (75)
`3 (25)
`
`2 (17)
`10 (83)
`0 (0)
`
`11 (92)
`1 (8)
`0 (0)
`0 (0)
`0 (0)
`26.6 (18.7-93.2)
`3.0 (1.4-4.9)
`2 (17)
`
`2 (17)
`3 (25)
`3 (25)
`0 (0)
`0 (0)
`4 (33)
`0 (0)
`0 (0)
`0 (0)
`
`P
`
`.65
`.73
`
`.52
`
`.26
`
`.11
`
`.01*
`.001
`.52
`
`AML indicates acute myeloid leukemia; MDS, myelodysplastic syndrome; ALL, acute lymphoblastic leukemia; CML, chronic myeloid leukemia; CLL, chronic lymphoblastic
`leukemia; NHL, non-Hodgkin leukemia; HL, Hodgkin lymphoma; and MM, multiple myeloma.
`*P ⫽ .101 for active group versus none; P ⫽ .007 for active versus inactive group.
`
`leukapheresis samples (Miltenyi Biotec, Auburn, CA) were stained with
`anti–BAFF-R, BR3 PE (clone 8A7; eBioscience), or biotinylated BMCA or
`TACI (R&D Systems) followed by streptavidin-APC antibody (Invitrogen,
`Carlsbad, CA).
`
`Ex vivo purification of B-cell subsets
`
`For purification of CD19⫹CD27⫹ and CD19⫹CD27⫺ B cells, whole EDTA
`anticoagulated blood (10-12 mL) was obtained from post-HSCT patients or
`healthy individuals. For purification of IgD versus CD38 B-cell subsets (Table 2),
`large volume leukapheresis products from 2 cGVHD patients or leukocyte filters
`from 3 healthy donors were obtained. Lymphocytes were isolated in a sterile
`fashion using Ficoll-Paque PLUS (GE Healthcare, Uppsala, Sweden). CD27⫹ or
`CD27⫺ B cells were sorted by flow cytometry to more than 98% purity (BD
`FACSAria Special Order Cell-Sorting System; Becton Dickinson, San Jose, CA).
`Purified CD27⫹ or CD27⫺ B cells (⬎ 95%) were also obtained using the indirect
`
`magnetic labeling system B Cell Isolation Kit II, human (Miltenyi Biotec). In
`brief, non-B cells were retained on magnetic-activated cell sorting (MACS)
`columns after staining with a cocktail of biotin-conjugated antibodies against
`CD2, CD14, CD16, CD36, and CD43 and binding to streptavidin magnetic
`microbeads. B cells passed through the column and were positively selected with
`CD27 magnetic beads and enriched to more than 95% CD20⫹CD27⫹. IgD
`versus CD38 B-cell subsets were isolated using a BD FACSAria cell sorter (BD
`Biosciences, San Jose, CA).
`
`IgG measurement
`
`B cells purified to more than 98% purity as assessed by CD20 staining were
`incubated in vitro in complete RPMI medium with 100 ng/mL IL-10 (R&D
`Systems); 250 U IL-2 (BD Biosciences); and 0, 25, 250, or 2500 ng/mL
`BAFF (Axxora LLC, San Diego, CA). IgG was measured using the Human
`IgG Elisa Quantitation Kit (Bethyl Laboratories, Montgomery, TX).
`
`Table 2. “Antigen-naive” (naive and transitional) and CD27ⴙ “antigen-experienced” B cells in patients after HSCT
`Active, n ⴝ 22
`Inactive, n ⴝ 23
`No, n ⴝ 12
`B-cell subset
`
`Healthy, n ⴝ 33
`
`Total naive B cells ⴛ1000/L
`P vs none
`Total transitional B cells ⴛ1000/L
`P vs none
`Total CD27ⴙCD19ⴙ B cells ⴛ1000/L
`P vs none
`CD27ⴙ B cells, %
`P vs none
`
`79.8
`.001
`8.1
`.04
`17.5
`.14
`19.0
`.06
`
`99.1
`.001
`8.5
`.04
`25.3
`.37
`8.4
`.21
`
`260.5
`
`28.7
`
`24.9
`
`6.05
`
`89.5
`.001
`14.4
`.02
`68.1
`.01
`27.3
`⬍ .001
`
`SAN EX 1025, Page 03
`
`

`

`3868
`
`SARANTOPOULOS et al
`
`BLOOD, 16 APRIL 2009 䡠 VOLUME 113, NUMBER 16
`
`Downloaded from https://ashpublications.org/blood/article-pdf/113/16/3865/1307974/zh801609003865.pdf by NEW YORK UNIVERSITY user on 20 January 2020
`
`Figure 1. High BAFF levels and low B-cell numbers result in high BAFF/B-cell ratios in patients with cGVHD. Plasma BAFF concentrations and numbers of CD19⫹
`B cells were measured in 3 groups after allogeneic HSCT: 22 patients with active cGVHD, 23 with inactive cGVHD, and 12 who did not develop cGVHD. Results were
`compared with 33 healthy donors. (A) Plasma BAFF concentrations in each patient group after HSCT and healthy donors. (B) Total number of CD19⫹ B cells in each patient
`group and healthy donors. (C) Median BAFF/B-cell ratio for each patient group and healthy donors. The BAFF/B-cell ratio was defined as nanograms of BAFF per 103 CD19⫹
`B cells. Box plots in each figure depict 75th percentile; median and 25th percentile values and whiskers represent maximum and minimum values.
`
`Statistical analyses
`
`For 2-sample comparison of continuous variables, a Wilcoxon rank sum test
`was performed. The Fisher exact test was used to compare categoric
`variables and the Spearman rank test was used for correlation analysis. All
`tests performed were 2-sided and considered significant at the .05 level.
`
`Results
`
`Relative B lymphopenia and high BAFF/B-cell ratio are
`associated with cGVHD
`
`A detailed analysis of plasma BAFF levels and B-cell phenotype
`was carried out in 57 patients who underwent allogeneic HSCT
`more than 12 months ago (Table 1). As shown in Figure 1A, all
`patients after HSCT had significantly higher BAFF levels com-
`pared with healthy individuals. Patients with active cGVHD had
`BAFF levels that were significantly higher than patients without
`cGVHD (no cGVHD; P ⬍ .001) or healthy donors (P ⬍ .001).
`Patients without cGVHD also had higher than normal BAFF levels
`(3.0 ng/mL vs 1.9 ng/mL, P ⫽ .003). Consistent with a previously
`described “surge” in B-cell number after HSCT,20 B-cell numbers
`in patients without cGVHD were significantly higher than normal
`(Figure 1B). In contrast, patients with active or inactive cGVHD
`had significantly lower numbers of total CD19⫹ B cells compared
`with patients without cGVHD (P ⫽ .001 and P ⫽ .01, respec-
`tively). Since BAFF level per B cell has been determined in murine
`models to be a critical determinant of autoreactive B-cell sur-
`vival,28,29 we calculated BAFF/B-cell ratios (level of BAFF per 103
`B cells) for each patient in each post-HSCT group. Figure 1C
`shows that despite higher than normal BAFF levels, supranormal
`B-cell numbers in patients without cGVHD resulted in BAFF/B-
`cell ratios that were similar to those found in healthy individuals. In
`contrast, patients with cGVHD had significantly higher BAFF/B-
`cell ratios compared with other post-HSCT patients and healthy
`individuals (P ⬍ .001 each). As shown in Figure 1A and B, high
`BAFF/B-cell ratios in patients with cGVHD were due to both high
`BAFF levels and B lymphopenia.
`
`Chronic GVHD is associated with decreased numbers of naive
`B cells
`
`In murine models, high BAFF/B-cell ratios promote survival of
`autoreactive B-cell clones in the absence of competition for available
`soluble BAFF by sufficient numbers of antigen-naive B cells.28 We
`
`therefore examined the relative numbers of antigen-naive B cells and
`antigen-experienced B cells in post-HSCT patients (Table 1) and in
`healthy individuals. In this analysis, antigen-naive B cells were de-
`fined as naive (CD19⫹IgDCD38LoCD27⫺) B cells and transitional
`(CD19⫹IgD⫹CD38HiCD27⫺) B cells; antigen-experienced B cells were
`defined as CD19⫹CD27⫹ B cells. As shown in Table 2, patients with
`active cGVHD had low numbers of naive B cells compared with
`patients with no cGVHD (median time after HSCT, 21 months vs
`27 months, respectively, P ⫽ .01). Patients without cGVHD had su-
`pranormal numbers of both naive and transitional B cells (260.5 ⫻ 106/L
`vs 89.5 ⫻ 106/L [P ⫽ .001] and 28.7 ⫻ 106/L in cGVHD vs
`14.4 ⫻ 106/L [P ⫽ .02], respectively). High total numbers of
`circulating recent bone marrow emigrants or transitional cells and
`naive B cells in patients greater than 1 year after HSCT without
`cGVHD suggests increased bone marrow output and prolonged
`survival of naive B cells compared with healthy individuals and
`with patients with cGVHD. In contrast to naive B cells, numbers
`and frequencies of CD27⫹ antigen-experienced B cells were signifi-
`cantly decreased compared with healthy donors but these cells
`were not different among post-HSCT groups (Table 2). In the
`setting of relative naive B lymphopenia, active cGVHD patients
`tended to have higher frequencies of CD27⫹CD19⫹ B cells (19.0%
`in active cGVHD vs 6.1% in patients without cGVHD), but this
`difference did not reach significance (P ⫽ .06).
`
`Identification of circulating pre-GC (IgDⴙCD38HiCD27ⴙ) B cells
`in cGVHD
`
`In humans, CD27 expression identifies antigen-experienced B cells
`committed to plasma cell differentiation.41 CD27⫹ B-cell subsets
`include IgDLo post–germinal center (GC) memory and plasmablast-
`like (PB)/plasma cell–like (PC) populations. CD27⫹ IgD⫹ B-cell
`populations include “IgD⫹ memory” V region–mutated memory
`B cells.42 In addition, the presence of CD27 on IgD⫹ CD38Hi–
`expressing B cells allows distinction of human peripheral transi-
`tional B cells from pre-GC B cells.43 Pre–germinal center cells
`(GCs), also called BM2⬘, GC founder, or preplasmablast cells, are
`IgD⫹CD38HiCD27⫹. These cells are typically found in human
`tonsils and have also been described in peripheral blood in SLE
`patients.44,45 Table 3 summarizes peripheral CD27⫹ B-cell subset
`phenotypes and their corresponding functional characteristics. We
`used multiparameter flow cytometry to determine whether any of
`these distinct circulating CD27⫹ B-cell subsets were more preva-
`lent in cGVHD. As described in “Flow cytometric analysis of
`peripheral B cells” in “Methods,” we first gated on CD19⫹ B cells
`
`SAN EX 1025, Page 04
`
`

`

`Downloaded from https://ashpublications.org/blood/article-pdf/113/16/3865/1307974/zh801609003865.pdf by NEW YORK UNIVERSITY user on 20 January 2020
`
`BLOOD, 16 APRIL 2009 䡠 VOLUME 113, NUMBER 16
`
`ALTERED B-CELL HOMEOSTASIS IN CHRONIC GVHD
`
`3869
`
`Table 3. Peripheral blood B-cell subsets analyzed in this study
`Cell-surface
`phenotype
`
`Peripheral blood B-cell subset
`
`Table 4. Proportions of CD27ⴙ B-cell subsets in patients after HSCT
`Active,
`Inactive,
`No,
`Healthy,
`n ⴝ 22
`n ⴝ 23
`n ⴝ 12
`n ⴝ 33
`
`CD19ⴙ subset
`
`CD27ⴚ subsets
`Naive B cells
`Transitional B cells42,43
`CD27ⴙ subsets
`IgD⫹ memory B42,45
`Pre-GC (or “GC founder”)42
`Post-GC memory B
`Plasmablast/plasma cell (PB/PC)
`
`IgD⫹CD38LoCD27⫺
`IgD⫹CD38HiCD27⫺
`
`IgD⫹CD38LoCD27⫹
`IgD⫹CD38HiCD27⫹
`IgDLo/⫺CD38LoCD27⫹
`IgDLo/⫺CD38HiCD27⫹
`
`IgDⴙ memory
`P vs none
`Post-GC memory, %
`P vs none
`Pre-GC, %
`P vs none
`PB/PC, %
`P vs none
`
`8.4
`.04
`20.4
`.06
`41.3
`.06
`86.9
`.08
`
`2.8
`.45
`18.4
`.06
`9.7
`.42
`66.4
`.23
`
`2.3
`
`10.8
`
`2.6
`
`36.8
`
`4.1
`.11
`32.8
`⬍ .001
`4.7
`.65
`40.7
`.80
`
`and examined relative levels of surface IgD, CD38, and CD27⫹ on
`these cells. Figure 2 shows representative examples of flow
`cytometric profiles of CD27⫹ B-cell subgroups identified in
`patients after HSCT and in healthy individuals. Table 4 compares
`the median frequency of each CD27⫹ B-cell subset in patients with
`and without cGVHD and in healthy individuals. Consistent with
`previous reports, all post-HSCT patients had fewer post-GC (IgDLo).46
`We found that the post-GC memory (CD27⫹IgDLoCD38Lo) B-cell
`subgroup was less frequent and lower in number in each post-
`HSCT group compared with healthy individuals, whereas all other
`CD27⫹ subsets were not proportionally or numerically different
`
`from normal (Table 4 and data not shown). As shown in Table 4,
`median IgD⫹ naive (CD38Lo) or pre-GC (CD38Hi) percentages in
`all post-HSCT groups were the same, but these populations tended
`to have higher CD27⫹ expression in cGVHD patients compared
`with patients without cGVHD (although differences did not reach
`significance).
`Overall increased CD27⫹ expression on B-cell subsets in the
`active cGVHD group could not be attributed to time after HSCT,
`since the active cGVHD group was closest in time to HSCT
`(median, 20.9 months) compared with the patients with inactive or
`without cGVHD, and therefore would have been expected to have
`
`Figure 2. Flow cytometric gating algorithm demonstrating circulating CD27ⴙ B-cell subsets in patients after HSCT. Gated CD19⫹ B cells were first examined for IgD
`and CD38 expression (far left column). Subsequently, each subset (a-d) was analyzed for CD27⫹ expression according to Sims et al.43 (a*) IgD⫹ Memory B cells are
`IgDHiCD38LoCD27⫹. (b*) Post-GC memory B cells are IgDLoCD38LoCD27⫹. (c*) Pre–germinal center (GC) B cells are IgDHiCD38HiCD27⫹. (d*) Plasmablast or plasma cell–like
`(PB/PC) B cells are IgDLoCD38HiCD27⫹.
`
`SAN EX 1025, Page 05
`
`

`

`3870
`
`SARANTOPOULOS et al
`
`BLOOD, 16 APRIL 2009 䡠 VOLUME 113, NUMBER 16
`
`Downloaded from https://ashpublications.org/blood/article-pdf/113/16/3865/1307974/zh801609003865.pdf by NEW YORK UNIVERSITY user on 20 January 2020
`
`Figure 3. BAFF promotes CD27ⴙ pre-GC and PB/PC subsets in cGVHD. (A) In vivo correlation between soluble BAFF and numbers of CD27⫹ BCR-activated B-cell subsets
`is shown for patients with active cGVHD (top row) and patients with inactive cGVHD (bottom row). The total number of each CD27⫹ B-cell subset (Table 2) and the BAFF level
`(ng/mL) were natural log–transformed. (B) BCMA, TACI, and BAFF-R expression on pre-GC and PB/PC cells from a patient with active cGVHD compared with healthy control.
`Data are representative of 2 independent experiments conducted using 2 cGVHD patients and 2 healthy donors.
`
`the lowest proportion of CD27⫹ B cells (Table 1). Numbers of each
`CD27⫹ B-cell subset were lower than those found in healthy
`individuals with one notable exception. Importantly, active cGVHD
`patients had significantly higher than normal numbers of pre-GC
`cells (2.5 ⫻ 106/L vs 0.89 ⫻ 106/L, respectively, P ⫽ .03) and
`higher frequency of pre-GC B cells in patients with active cGVHD
`compared with healthy individuals (41.3% vs 4.7%, P ⫽ .02).
`Pre-GC cells have not previously been detected in healthy individu-
`als.42,43 In the current study, we detected circulating cells with a
`pre-GC phenotype in 3 (of 33 tested) healthy individuals. However,
`the pre-GC cells in these 3 healthy individuals were unlike cGVHD
`samples tested in that they did not coexpress the follicular marker
`CD10 (data not shown). Thus, we identified a distinct and
`potentially pathologic pre-GC population, similar to circulating
`pre-GC cells identified by other investigators in SLE patients, in
`patients with cGVHD.44,45
`
`Increasing BAFF levels correlate with increasing numbers of
`activated pre-GC and PB/PC cells in cGVHD
`
`To assess whether high BAFF levels were associated with in-
`creased numbers of any circulating B-cell subsets in vivo, we
`examined possible correlations between the BAFF level and total
`B-cell number in each subset for each patient within each
`post-HSCT group. Figure 3A depicts the significant correlations
`found between cell numbers and high BAFF levels in patients with
`active or inactive cGVHD. Increased BAFF levels in patients with
`inactive disease correlated inversely with post-GC memory subset
`numbers (r ⫽ ⫺0.57, P ⫽ .005), possibly reflecting reconstitution
`and turnover of memory B cells despite high BAFF levels in the
`group of patients with clinically inactive cGVHD. Only patients
`
`with active disease had a significant positive correlation between
`BAFF and PB/PC cell number (r ⫽ 0.52, P ⫽ .01). Active and
`inactive cGVHD patients had positive correlations between only
`BAFF and number of CD27⫹ pre-GC cells (r ⫽ 0.5 and r ⫽ 0.49,
`respectively; P ⫽ .02 each), suggesting that BAFF preferentially
`affected these 2 B-cell populations in vivo. Consistent with an
`activated state, BAFF-R expression was relatively low, whereas
`TACI and BCMA expression was increased47 on pre-GC cells in
`cGVHD (Figure 3B). PB/PC cells also expressed higher BCMA
`levels in cGVHD (Figure 3B). Taken together, these results suggest
`that high BAFF in cGVHD patients promotes survival and
`activation of pre-GC and PB/PC cells.
`We then examined qualitative differences in CD27⫹ popula-
`tions in patients with and without cGVHD. Human CD27⫹ B cells
`are antigen experienced and include memory B cells and antibody-
`secreting cells.41 To determine whether CD27⫹ B cells in patients
`with active cGVHD were functionally activated antibody-secreting
`cells in vivo, we measured IgG production of freshly purified
`CD27⫹ B cells from these patients compared with CD27⫹ B cells
`from patients without cGVHD and from healthy donors. As shown
`in Figure 4A, ex vivo–purified CD27⫹ B cells from patients with
`active and inactive cGVHD produce IgG without requiring in vitro
`antigen or BCR stimulation. Constitutive IgG secretion by CD27⫹
`B cells from patients without cGVHD or from healthy donors did
`not occur. Increasing in vitro BAFF concentration also did not
`result in IgG production by CD27⫹ B cells (data not shown). High
`IgG secretion by the PB/PC population occurred without additional
`BAFF (Figure 4B). Although constitutive IgM production by
`pre-GC B cells was found (data not shown), these cells produced
`IgG after addition of BAFF in vitro (Figure 4B). Except for PB/PC
`
`Figure 4. Constitutive production of IgG by CD27ⴙ B cells
`and B-cell subsets in patients with cGVHD. (A) Spontaneous
`IgG production by purified peripheral blood CD27⫹ B cells from
`post-HSCT patients or healthy individuals. (B) IgG production by
`cGVHD patient B-cell subsets after 48 hours with or without
`additional BAFF in vitro. Error bars indicate SD (⫾ mean).
`
`SAN EX 1025, Page 06
`
`

`

`BLOOD, 16 APRIL 2009 䡠 VOLUME 113, NUMBER 16
`
`ALTERED B-CELL HOMEOSTASIS IN CHRONIC GVHD
`
`3871
`
`Downloaded from https://ashpublications.org/blood/article-pdf/113/16/3865/1307974/zh801609003865.pdf by NEW YORK UNIVERSITY user on 20 January 2020
`
`Figure 5. Delayed reconstitution of B cells and increased proportion of CD27ⴙ B cells in patients who develop cGVHD. CD19⫹ B-cell number (left y-axis) and BAFF
`levels (right y-axis) were measured every 3 months after HSCT in 3 patient groups. (A) Eight patients who did not develop cGVHD by 12 months. (B) Seventeen patients who
`developed cGVHD during this period. (C) Eight patients who underwent autologous transplantation after myeloablative conditioning. Numbers of patients measured at ea

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