throbber
IMMUNOBIOLOGY
`
`Programmed death 1 signaling on chronic myeloid leukemia–specific T cells
`results in T-cell exhaustion and disease progression
`Sabine Mumprecht,1 Christian Schu¨ rch,1 Juerg Schwaller,2 Max Solenthaler,3 and Adrian F. Ochsenbein1,4
`
`1Tumor Immunology, Department of Clinical Research, University of Berne, Berne; 2Department of Research, Childhood Leukemia, University of Basel, Basel;
`and 3Department of Hematology and Central Hematology Laboratory and 4Institute for Medical Oncology, Inselspital, Berne, Switzerland
`
`Chronic myeloid leukemia (CML) is a ma-
`lignant myeloproliferative disease with a
`characteristic chronic phase (cp) of sev-
`eral years before progression to blast
`crisis (bc). The immune system may con-
`tribute to disease control
`in CML. We
`analyzed leukemia-specific immune re-
`sponses in cpCML and bcCML in a retrovi-
`ral-induced murine CML model.
`In the
`presence of cpCML and bcCML express-
`ing the glycoprotein of lymphocytic cho-
`Introduction
`
`riomeningitis virus as a model leukemia
`antigen,
`leukemia-specific
`cytotoxic
`T lymphocytes (CTLs) became exhausted.
`They maintained only limited cytotoxic
`activity, and did not produce interferon-␥
`or tumor necrosis factor-␣ or expand
`after restimulation. CML-specific CTLs
`were characterized by high expression of
`programmed death 1 (PD-1), whereas CML
`cells expressed PD-ligand 1 (PD-L1).
`Blocking the PD-1/PD-L1 interaction by
`
`generating bcCML in PD-1–deficient mice
`or by repetitive administration of ␣PD-L1
`antibody prolonged survival. In addition,
`we found that PD-1 is up-regulated on
`CD8ⴙ T cells from CML patients. Taken
`together, our results suggest that block-
`ing the PD-1/PD-L1 interaction may re-
`store the function of CML-specific CTLs
`and may represent a novel therapeutic
`approach for CML.
`(Blood. 2009;114:
`1528-1536)
`
`Chronic myeloid leukemia (CML) is a clonal myeloproliferative
`disorder resulting from the neoplastic transformation of a hemato-
`poietic stem cell.1 The disease is bi- or triphasic, comprising a
`chronic, an accelerated, and a terminal blast phase in which the
`patients develop an acute leukemia of either myeloid (AML) or,
`less often, lymphoid (ALL) cell type. More than 90% of all CML
`cases are associated with the presence of the Philadelphia chromo-
`some, which results from a reciprocal
`translocation between
`chromosomes 9 and 22 forming the breakpoint cluster region/
`Abelson protein tyrosine kinase (BCR/ABL) fusion protein, a
`constitutively activated tyrosine kinase.2,3 Depending on the pre-
`cise breakpoints in the BCR gene, different forms of BCR/ABL
`fusion protein with different molecular weights can be generated
`(p190BCR/ABL, p210BCR/ABL, and p230BCR/ABL). CML pa-
`tients predominantly express p210BCR/ABL.1
`Currently, BCR/ABL-selective tyrosine kinase inhibitors are
`the standard treatment for CML. However, resistant clones often
`develop during treatment. At present, the only curative treatment
`for CML is allogeneic hematopoietic stem cell transplantation.4
`Several earlier studies suggested that the immune system plays
`an important role in the control of CML. CML cells are susceptible
`to lysis by CD8⫹ T cells5 and natural killer (NK) cells in vitro.6 For
`unknown reasons, CML is the most graft-versus-leukemia–
`sensitive leukemia.7 In addition, cytotoxic T lymphocytes (CTLs)
`directed against leukemia antigens are found in CML patients
`without hematopoietic stem cell transplantation, including CTLs
`specific for BCR/ABL, overexpressed self-proteins such as protein-
`ase-3, and Wilms tumor 1 protein.5,8 However, the physiologic
`relevance of these leukemia-specific CTL responses in the control
`of CML is unknown. The presence of CTL escape mechanisms
`
`during CML disease progression to blast crisis suggests that CTLs
`are involved in the control of the chronic phase of the disease.
`Documented escape mechanisms include the expression of Fas
`ligand during blast crisis,9 the deletion of high-avidity CTLs
`specific for a leukemia-associated self-antigen,10 and the develop-
`ment of functional blocks in the caspase activation pathway in
`AML cells.11
`Coinhibitory molecules are essential for the maintenance of
`T-cell homeostasis, self-tolerance, and tolerance to chronic infec-
`tions. Programmed death 1 (PD-1) is a member of the immunoglobu-
`lin (Ig) superfamily, which is inducibly expressed on T cells,
`B cells, and activated monocytes.12 To mediate inhibitory signals
`through PD-1, the binding of either of the 2 ligands to the receptor
`is necessary. PD-ligand 1 (PD-L1, B7-H1) is expressed constitu-
`tively on resting T cells, B cells, dendritic cells (DCs), macro-
`phages, mesenchymal stem cells, some parenchymal cells, and
`cultured bone marrow-derived mast cells. PD-L1 expression is
`further up-regulated after activation.13 PD-ligand 2 (PD-L2, B7-
`DC) is only inducibly expressed on DCs, macrophages, and
`cultured bone marrow-derived mast cells.13 Recently, PD-1 signal-
`ing has been identified as an important mechanism of antigen-
`specific T-cell dysfunction in chronic infections such as lympho-
`cytic choriomeningitis virus (LCMV) infection in mice14 or HIV15
`and hepatitis C virus (HCV)16 infections in humans. In addition, it
`has been shown that PD-1 leads to functional impairment of
`tumor-infiltrating T cells in solid tumors.17 In contrast, mechanisms
`determining the function of leukemia-specific CTLs have not yet
`been analyzed in detail.
`In the present study, we analyzed the immunosurveillance of
`chronic phase CML (cpCML) and blast crisis CML (bcCML) in a
`
`Submitted September 16, 2008; accepted April 25, 2009. Prepublished online
`as Blood First Edition paper, May 6, 2009; DOI 10.1182/blood-2008-09-
`179697.
`
`The publication costs of this article were defrayed in part by page charge
`payment. Therefore, and solely to indicate this fact, this article is hereby
`marked ‘‘advertisement’’ in accordance with 18 USC section 1734.
`
`An Inside Blood analysis of this article appears at the front of this issue.
`
`© 2009 by The American Society of Hematology
`
`1528
`
`BLOOD, 20 AUGUST 2009 䡠 VOLUME 114, NUMBER 8
`
`UPenn Ex. 2059
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`BLOOD, 20 AUGUST 2009 䡠 VOLUME 114, NUMBER 8
`
`EXHAUSTION OF CML-SPECIFIC T CELLS BY PD-1 IMMUNOBIOLOGY
`
`1529
`
`murine retroviral bone marrow transduction and transplantation
`model.18 We found that leukemia-specific CTLs were functionally
`exhausted and phenotypically characterized by high expression of
`PD-1, in both cpCML and bcCML. Blocking PD-1 signal transduc-
`tion by using PD-1–deficient recipient mice or by administration of
`␣PD-L1 antibody reversed CML-specific T-cell tolerance, and the
`time to disease progression was increased.
`
`Methods
`
`Mice
`
`C57BL/6 mice were purchased from Harlan. The p14 T-cell receptor (TCR)
`transgenic mice19 and H8 transgenic mice20 were obtained from the Institute
`for Laboratory Animals. CD45.1⫹ mice were obtained from C. Mueller
`(University of Berne). PD-1–deficient mice21 were provided by T. Honjo
`(Kyoto University). Animal experiments were performed with sex- and
`age-matched mice and approved by the Experimental Animal Committee of
`the Canton of Berne and performed according to Swiss laws for animal
`protection.
`
`Viruses, peptide, and retroviral vectors
`
`LCMV, strain WE, and Docile were provided by R. M. Zinkernagel
`(University Hospital, Zurich) and propagated, as described.22,23 The LCMV-
`GP, amino acids 33-41 (glycoprotein [gp]33, KAVYNFATM), was pur-
`chased from NeoMPS SA. The retroviral vectors pMSCV-p210 BCR/ABL-
`IRES-GFP, pMSCV-p210 BCR/ABL-pgk-neo, and pMSCV-NUP98/
`HOXA9-IRES-GFP (MSCV, mouse stem cell virus; IRES, internal ribosomal
`entry site; GFP, green fluorescent protein; neo, neomycin) were a gift from
`D. G. Gilliland (Havard Medical School), and the packaging vector pIK6
`was purchased from Cell Genesys.24-26
`
`51Cr-release assay
`
`Spleens were isolated and analyzed after 5 days of in vitro restimulation in a
`standard 51Cr-release assay, as described.27 The cytotoxicity assay of ex
`vivo isolated p14 T cells was performed as follows: 6 ⫻ 105 CD45.1⫹ cells,
`purified by magnetic cell sorting (MACS; Miltenyi Biotec), were either
`used directly or restimulated for 5 days with 3 ⫻ 105 irradiated gp33-
`pulsed naive C57BL/6 splenocytes in the presence of 50 U/mL murine
`interleukin (IL)-2.
`
`Cells and retroviral particle production
`
`Retroviral particles were generated by transient cotransfection of 293-
`T cells with the respective MSCV vector and pIK6, using Superfect
`transfection reagent (Qiagen), according to the manufacturer’s protocol.
`After 48 hours, virus-containing supernatant was harvested. For determina-
`tion of retroviral titers, BA/F3 cells were infected with different amounts of
`retroviral supernatant using polybrene transfection reagent (10 ␮g/mL;
`Sigma-Aldrich). After 48 hours, retroviral
`titers were determined by
`enumerating GFP-positive cells by flow cytometry. Alternatively, to assess
`virus titers of the vector containing a neocassette, BA/F3 cells were infected
`and then cultivated for 5 days in the presence or absence of 0.8 mg/mL
`G-418 sulfate (Gibco). Surviving cells were assessed with trypan blue
`staining.
`
`retroviral particles with polybrene (6.7 ␮g/mL) and 0.01 M HEPES (N-2-
`hydroxyethylpiperazine-N⬘-2-ethanesulfonic acid) through spin infection
`(90 minutes at 1258g, 30°C). A total of 105 transduced bone marrow cells
`was injected intravenously into previously irradiated (4.5, 6.5, or 9.5 Gy)
`syngeneic recipient mice (transduction efficiency: 24.0% ⫾ 9.8%).
`
`Antibodies and flow cytometry
`
`␣CD8-allophycocyanin (APC), ␣CD4-biotin, ␣B220-biotin, ␣I-Ab-major histo-
`compatibility complex (MHC) class II-biotin, ␣CD45.1-phycoerythrin (PE) and
`-APC, ␣CD11c-biotin, ␣ interferon (IFN)-␥-PE, ␣ tumor necrosis factor (TNF)-
`␣-PE, streptavidin-PE and APC, ␣NK1.1-biotin, ␣PD-1-biotin, ␣PD-L1-biotin,
`␣PD-L2-biotin, rat IgG2a isotype-biotin, and human ␣PD-1-PE were obtained
`from eBioscience. ␣GR-1-PE, ␣CD8-peridinin chlorophyll protein (PerCP)-
`Cy5.5, ␣CD4-PerCP-Cy5.5, ␣PD-1-PerCP-Cy5.5, ␣V␣2-biotin and PE, ␣Sca-1-
`APC, ␣MAC-1-PE-Cy7, ␣C-kit-PE-Cy7, mouse IgG1-PE isotype control,
`human ␣CD8-fluorescein isothiocyanate, and human ␣CD45-PerCP-Cy5.5 were
`obtained from BD Pharmingen. MHC class I (H-2Db) tetramer-PE complexed
`with gp33 or nuclear protein (np)396 was obtained from ProImmune and used
`according to the manufacturer’s protocol. Intracellular staining was performed, as
`described.28 Relative fluorescence intensities were measured on a BD LSRII flow
`cytometer (BD Biosciences) and analyzed using FlowJo software (TreeStar). The
`frequency of IFN-␥– and TNF-␣–producing CD8⫹ T cells was calculated as
`gp33-pulsed minus nonpulsed samples.
`
`Adoptive transfer experiments and reisolation of transferred
`CD8ⴙ T cells
`
`P14 ⫻ CD45.1 T cells were isolated and purified by MACS for CD8⫹V␣2⫹
`T cells. A total of 2.5 to 4 ⫻ 106 CD8⫹Va2⫹CD45.1⫹ cells was injected
`intravenously into CML mice, naive C57BL/6 control mice, C57BL/6 mice
`that were infected with 104 plaque-forming units (pfu) of LCMV-WE, and
`C57BL/6 mice chronically infected with 107 pfu of LCMV Docile (all
`recipient mice were CD45.1⫺). CML disease progress was monitored by
`fluorescence-activated cell sorter (FACS) analysis of blood, and on the days
`indicated, CD45.1⫹ T cells from CML mice, naive C57BL/6 mice, and
`C57BL/6 mice infected with LCMV were isolated by MACS.
`
`Proliferation assays
`
`For ex vivo proliferation of transferred p14 T cells, 2 ⫻ 105 CD45.1⫹ cells
`were restimulated for 4 days with 2 ⫻ 105 irradiated gp33-pulsed or
`nonpulsed naive C57BL/6 splenocytes. For in vitro proliferation, naive p14
`CD8⫹ T cells and p14 ⫻ PD-1⫺/⫺ CD8⫹ T cells (2 ⫻ 105) were restimu-
`lated with irradiated (50 Gy) FACS-sorted H8-cpCML cells (BCR/ABL-
`GFP⫹), FACS-sorted H8-bcCML cells (NUP98/HOXA9-GFP⫹), H8 spleno-
`cytes, or C57BL/6 splenocytes (0.4 ⫻ 105). 3H-thymidine incorporation
`assay was performed, as described.22 The proliferation index was calculated
`as the ratio between gp33-pulsed and nonpulsed samples (ex vivo prolifera-
`tion assay) and as the ratio between H8-CML cells or H8 splenocytes and
`C57BL/6 splenocytes used as stimulators (in vitro proliferation assay).
`
`Cytometric bead array assay
`
`A total of 1.5 ⫻ 105 CD45.1⫹ cells was restimulated for 18 hours with
`3 ⫻ 105 irradiated gp33-pulsed or nonpulsed naive C57BL/6 splenocytes.
`Cytokine production in supernatant was analyzed with a mouse T helper
`cell 1/2 cytokine cytometric bead array assay according to the manufactur-
`er’s protocol (BD Biosciences).
`
`CML model
`
`PD-L1 blockade
`
`Bone marrow donor mice were pretreated with 150 mg/kg 5-fluorouracil
`intraperitoneally (Sigma-Aldrich). After 6 days, the bone marrow was
`flushed out from femurs and tibias. Erythrocytes were removed and the
`bone marrow cells were incubated in transplant media (RPMI/10% fetal
`calf serum with recombinant murine IL-3 [6 ng/mL; BD Biosciences],
`recombinant murine stem cell factor [10 ng/mL; Biocoba], and recombinant
`human IL-6 [10 ng/mL; BD Biosciences]) for 24 hours. A total of 4 ⫻ 106
`cells was transfected twice on 2 consecutive days with the respective
`
`bcCML mice were treated intraperitoneally every third day with 200 ␮g of
`␣PD-L1 monoclonal antibody (clone 2G9; BioXCell) or IgG from rat
`serum (I8015; Sigma-Aldrich) starting on the day of transplantation.
`
`CD8 depletion
`
`bcCML mice were treated intraperitoneally on day 0 and day 2, and from
`then on weekly with 100 ␮g of␣CD8 monoclonal antibody (YTS 169.4).
`
`UPenn Ex. 2059
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`1530
`
`MUMPRECHT et al
`
`BLOOD, 20 AUGUST 2009 䡠 VOLUME 114, NUMBER 8
`
`Figure 1. Model of CML-like disease in mice. (A) Schema of
`retroviral bone marrow transduction and transplantation to irradi-
`ated recipient mice. (B) Donor bone marrow (CD45.2⫹) was
`transferred into CD45.1⫹ recipient mice (irradiated with 4.5 Gy).
`Twenty days later, PBMCs were stained for GR-1, CD8, CD4,
`B220, and NK1.1, and analyzed by flow cytometry. Representa-
`tive dot plots of 1 of 6 mice are shown. (C) Survival of cpCML (—,
`n ⫽ 12) and bcCML (哹, n ⫽ 18) mice is shown in Kaplan-Meier
`plots. Statistical comparison was performed with the log-rank test.
`(D) Phenotypical characterization of GFP⫹ leukemic cells in
`cpCML and bcCML mice. Cells were gated on granulocytes in
`control C57BL/6 mice and on transduced granulocyte populations
`in cpCML and bcCML mice. One representative staining of 5 is
`shown.
`
`The treatment depletes CD8⫹ T cells to below the detection limit of flow
`cytometry analysis (data not shown).
`
`CML patients
`
`Human blood sample collections were approved by the ethical committee of
`the Canton of Berne, Switzerland, and patients gave their written informed
`consent in accordance with the Declaration of Helsinki. The average age of
`healthy donors was 40.6 ⫾ 13.8 years (3 [female] and 5 [male]) and of
`cpCML patients 46.0 ⫾ 12.2 years (3 [female] and 5 [male]). All cpCML
`patients expressed p210BCR/ABL.
`
`Statistical analysis
`
`Data are presented as the mean plus or minus SEM. The significance of the
`differences in Kaplan-Meier survival curves was determined using the
`log-rank test (2-tailed). The significance between groups of human samples
`was determined by using unpaired Student
`test (2-tailed) and the
`t
`correlation with the Spearman correlation (2-tailed, ␣ ⫽.05).
`
`Results
`
`CML-like disease in immunocompetent mice
`
`It has been shown previously that BCR/ABL expression in bone
`marrow cells leads to cpCML in mice.29 Coexpression of NUP98/
`HOXA9 leads to accumulation of immature myeloid cells and
`progression to blast crisis.18 Therefore, we transduced bone mar-
`row from C57BL/6 mice with retroviral particles expressing
`p210BCR/ABL alone, or cotransduced bone marrow cells with
`p210BCR/ABL and NUP98/HOXA9 retroviral particles. Trans-
`duced bone marrow cells were adoptively transferred into irradi-
`ated syngeneic recipient mice (Figure 1A).
`In previous studies, recipient mice were lethally irradiated.2,29,30
`This completely prevented a leukemia-specific immune response
`by the host and made it impossible to study the immunosurveil-
`
`lance of CML. In this study, we titrated the irradiation dose and the
`number of retroviral particles NUP98/HOXA9 and BCR/ABL
`needed to generate cpCML and bcCML originating from donor
`bone marrow cells in a host with a largely intact immune system.
`To address whether immune cells for the control of the leukemia
`originate from recipient or donor bone marrow, transduced bone
`marrow cells were transferred into CD45.1⫹ recipient mice. CD8⫹
`and CD4⫹ T cells, B cells, and NK cells were found to originate
`from donor bone marrow (CD45.1⫺) when recipient mice were
`irradiated with 9.5 Gy (data not shown). In contrast, when recipient
`mice were irradiated with 4.5 Gy (Figure 1B) or 6.5 Gy (data not
`shown), immune cells were found to originate from the recipient
`mouse, whereas the leukemia cells (GR-1⫹) originated from the
`cotransduced donor bone marrow.
`Transduction of bone marrow with BCR/ABL retroviral par-
`ticles alone (105 U) or cotransduction with NUP98/HOXA9 (106 U)
`regularly induced cpCML or bcCML, respectively. In contrast,
`transduction with NUP98/HOXA9 retroviral particles alone did not
`induce leukemia up to 3 months after transplantation. bcCML
`rapidly progressed and animals died within 3 weeks, whereas
`cpCML mice lived up to 6 weeks (Figure 1C). Because the
`retroviral vectors BCR/ABL in cpCML and NUP98/HOXA9 in
`bcCML coexpressed the fluorescent dye GFP, the development of
`CML-like disease was followed with flow cytometry of peripheral
`blood (Figure 1D). The majority of granulocytes were GFP
`positive, indicating that these cells expressed BCR/ABL in cpCML
`or NUP98/HOXA9 in bcCML. In addition, the granulocyte popula-
`tion in the blood of cpCML and bcCML mice was increased to
`9 ⫻ 107 granulocytes/mL blood compared with the C57BL/6
`control (⬍ 2 ⫻ 106 granulocytes/mL blood; data not shown).
`Immature myeloid blast cells (MAC-1⫹, GR-1⫹, c-kit⫹) were
`detectable in bcCML. In contrast, all transduced GFP⫹ leukemic
`cells in cpCML phenotypically resembled mature granulocytes
`
`UPenn Ex. 2059
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`BLOOD, 20 AUGUST 2009 䡠 VOLUME 114, NUMBER 8
`
`EXHAUSTION OF CML-SPECIFIC T CELLS BY PD-1 IMMUNOBIOLOGY
`
`1531
`
`(MAC-1⫹, GR-1⫹, c-kit⫺) comparable with untransduced granulo-
`cytes in naive C57BL/6 mice (Figure 1D). Therefore, by reducing
`the dose of irradiation given to recipient mice (4.5 or 6.5 Gy) and
`by administering defined amounts of retroviral particles, it was
`possible to reproducibly induce cpCML and bcCML in recipient
`mice with a largely intact immune system.
`
`Leukemia-specific CTLs are not detectable in CML disease
`progression
`
`To analyze antigen-specific immune responses in vivo, bone
`marrow from H8 transgenic mice was transduced with the retrovi-
`ral particles. In this experimental setup, all leukemia cells ex-
`pressed the immunodominant CTL epitope gp33 of LCMV on
`MHC class I molecules as a model leukemia antigen (H8-CML
`mice). The leukemia-specific CTL response was analyzed in
`H8-CML mice at different time points after bone marrow transplan-
`tation. Transduction of bone marrow with 0.01 ⫻ 106 U of
`BCR/ABL or cotransduction with 0.01 ⫻ 106 U of BCR/ABL and
`0.1 ⫻ 106 U of NUP98/HOXA9 retroviral particles induced a
`transient increase of GFP-positive granulocytes in recipient mice,
`but by day 30 all leukemia cells were eliminated (data not shown).
`Tetramer staining revealed that recipient mice that eliminated CML
`developed a LCMV-gp33–specific CTL response (Figure 2A). In
`contrast, if bone marrow cells were transduced with 0.1 ⫻ 106 U of
`BCR/ABL alone or together with 106 U of NUP98/HOX-A9
`retroviral particles, H8-cpCML and H8-bcCML persisted and
`progressed to the death of the animals (disease kinetics comparable
`with Figure 1C). In H8-cpCML and H8-bcCML mice, LCMV-gp33–
`specific CTLs were not detectable by tetramer staining (Figure 2A)
`or by intracellular staining for IFN-␥ or TNF-␣ after in vitro
`restimulation with gp33 (Figure 2B). In addition, CTLs isolated
`from cpCML and bcCML mice were not able to lyse gp33
`peptide-pulsed target cells ex vivo after 5 days of in vitro
`restimulation (Figure 2C). Naive C57BL/6 mice and LCMV-
`immune mice that have been infected 8 weeks previously with
`200 pfu of LCMV-WE were used as controls (Figure 2A-C).
`To test whether gp33-specific CTLs can be activated in mice
`with CML-like disease, H8-cpCML, H8-bcCML mice, and control
`C57BL/6 mice were infected with 200 pfu of LCMV. Eight days
`later, the frequencies of CML-specific CTLs (gp33) and of CTLs
`specific for an unrelated viral epitope (np396) were analyzed by
`tetramer staining. In H8-cpCML and H8-bcCML mice, np396-
`specific CTLs, but not gp33-specific CTLs, were detectable (Figure
`2D). In contrast, naive C57BL/6 mice infected with LCMV
`mounted gp33- and np396-specific CTL responses. These results
`demonstrate that (a) leukemia cells were able to induce a specific
`CTL response, and (b) in the presence of CML-like disease with
`high leukocytes counts, leukemia antigen-specific CTLs were
`not present.
`
`CML-specific CTLs have residual effector functions
`
`To study CML-specific CTL responses in more detail, purified p14
`TCR transgenic CD8⫹ T cells (CD45.1⫹CD8⫹V␣2⫹) specific for
`LCMV-gp33 were adoptively transferred to H8-bcCML mice and
`to naive and LCMV-infected control mice. After transfer to
`H8-bcCML mice, p14 CD8⫹ T cells rapidly expanded in blood
`(Figure 3A) and spleen (Figure 3B). The specific CTLs reached a
`frequency of 25% of total
`lymphocytes 6 days after transfer.
`Thereafter, their number rapidly declined, reaching a stable fre-
`quency of approximately 2.8% (⫾ 1.9%), which is slightly above
`the frequency of transferred p14 CTLs in naive C57BL/6 control
`
`mice. In addition, the leukemia-specific TCR chain V␣2 was
`down-regulated on CML-specific CTLs in blood (mean V␣2⫹ p14
`42.1% ⫾ 25.8%;
`CD8⫹ T cells: CML,
`naive C57BL/6,
`92.3% ⫾ 3.5%; LCMV-infected, 79.2% ⫾ 16.8%; Figure 3C) and
`spleen (data not shown), indicating TCR ligation and CTL activa-
`tion. P14 CD8⫹ T cells transferred to C57BL/6 control mice did not
`proliferate or down-regulate the specific TCR, whereas p14 CD8⫹
`T cells transferred to LCMV-infected mice rapidly expanded. Due
`to the large number of transferred specific CTLs, LCMV-WE was
`rapidly eliminated and V␣2 expression was already reconstituted in
`most animals at day 3 after transfer (Figure 3A-C). The down-
`regulation of the TCR V␣2 did not correlate with the percentage of
`leukemic GFP⫹ granulocytes when analyzed in blood (Spearman
`correlation, P ⫽ .49, r ⫽ ⫺0.17; Figure 3D). We concluded that
`naive specific CTLs in H8-bcCML mice were initially activated
`after TCR ligation and expanded. However, the majority of these
`CML-specific CTLs were deleted, and only a small fraction
`persisted long-term.
`Memory CD8⫹ T cells in chronic infections with persisting
`antigen are either physically deleted or functional impaired, a
`process termed exhaustion.31 To analyze the degree of functional
`impairment of specific CD8⫹ T cells in the presence of H8-bcCML,
`adoptively transferred p14 CD8⫹ T cells were isolated from
`spleens of H8-bcCML mice and of naive and LCMV-infected
`control mice 6 days after transfer. P14 CTLs isolated from H8-
`bcCML mice produced hardly any proinflammatory cytokines such
`as IFN-␥, TNF-␣, or IL-2 after in vitro restimulation (Figure 4A).
`In contrast, p14 CTLs isolated from LCMV-infected mice pro-
`duced high amounts of IFN-␥ and TNF-␣ (Figure 4A). In addition,
`p14 CTLs isolated from H8-bcCML mice did not lyse peptide-
`pulsed target cells directly ex vivo (data not shown), and had only
`limited lytic capacity after 5 days of in vitro restimulation (Figure
`4B). Therefore, the cytokine profile and lytic function of LCMV-
`specific CD8⫹ T cells define effector CTLs, whereas the character-
`istic of CML-specific CD8⫹ T cells is consistent with partial
`exhaustion.
`To analyze whether the lytic function of the transferred CTLs is
`sufficient to influence the leukemia load, we followed the fre-
`quency of GFP⫹ granulocytes before and 6 days after the transfer
`of p14 CD8⫹ T cells. In most animals, the frequency of GFP⫹
`granulocytes was at least transiently reduced (Figure 4C). This
`indicates that the transferred naive p14 CD8⫹ T cells acquired
`effector function in vivo sufficient
`to transiently reduce the
`leukemia load.
`To assess the proliferative capacity of the isolated p14 CD8⫹
`T cells, 3H-thymidine incorporation was measured after in vitro
`restimulation. P14 CTLs from C57BL/6 control mice with a naive
`phenotype proliferated vigorously in contrast to effector p14 CTLs
`isolated from acute LCMV-infected mice (Figure 4A-B,D). P14
`CTLs isolated from H8-bcCML mice neither expanded efficiently
`in vitro (Figure 4D) nor exhibited effector functions (Figure 4A-B).
`Thus, CML-specific CTLs are partially exhausted with limited
`residual effector functions.
`
`Myeloid leukemia cells suppress T-cell proliferation through
`PD-1/PD-L1 interaction
`
`CTLA-4 and PD-1 are 2 well-known coinhibitory molecules that
`are of importance in the maintenance of peripheral tolerance.32
`Therefore, we analyzed their expression on total and on CML-
`specific CD8⫹ T cells. In H8-bcCML mice, PD-1 was highly
`expressed on the transferred CML-specific p14 CTLs in the blood
`(data not shown) and spleen (Figure 5A). In contrast, PD-1 was not
`
`UPenn Ex. 2059
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`1532
`
`MUMPRECHT et al
`
`BLOOD, 20 AUGUST 2009 䡠 VOLUME 114, NUMBER 8
`
`expressed on transferred p14 CD8⫹ T cells in C57BL/6 control
`mice, and was only expressed at low levels on p14 CTLs in acutely
`LCMV-WE–infected mice. However, as shown previously,14 PD-1
`expression was high on p14 CTLs in mice chronically infected with
`LCMV Docile. The expression of PD-1 on transferred p14 CTLs in
`H8-bcCML mice was substantially higher than in mice with
`persistent LCMV infection (Figure 5A). Surprisingly, a large
`fraction of total CD8⫹ T cells expressed PD-1 (93.5% ⫾ 1.8% in
`
`Figure 3. Adoptive transfer of leukemia-specific TCR transgenic CD8ⴙ T cells to
`H8-bcCML mice. Purified p14 CD8⫹ T cells (CD45.1⫹CD8⫹V␣2⫹) were adoptively
`transferred to H8-bcCML mice (F), to naive C57BL/6 control mice (⽧), and to
`C57BL/6 mice infected with 104 pfu of LCMV (f). (A-B) The expansion of the
`transferred p14 CD8⫹ T cells (CD45.1⫹) was analyzed in the blood (A) and spleen (B)
`by flow cytometry analysis. (C) The expression of the leukemia-specific TCR V␣2 on
`the transferred p14 CD8⫹ T cells (CD45.1⫹) was analyzed in blood 3 days after
`the TCR V␣2 was
`transfer by flow cytometry analysis. (D) The expression of
`compared with the percentage of leukemic granulocytes (GFP⫹) in the blood 3 days
`after transfer. Results are pooled from 5 independent experiments. Statistical
`comparison was made using the Spearman correlation.
`
`H8-CML mice; data not shown). In contrast, CTLA-4 was not
`expressed on transferred CML-specific CTLs in blood (data not
`shown) and spleen (Figure 5B) in H8-bcCML mice.
`The observation of high rates of PD-1 expression on total CD8⫹
`T cells prompted us to analyze the expression of PD-1 on non–
`leukemia-specific CTLs. Therefore, cpCML and bcCML from
`C57BL/6 bone marrow donors were induced in p14 recipient mice.
`In this experimental setup, 60% of CD8⫹ T cells express the TCR
`V␣2 and are non–leukemia specific. PD-1 was expressed on 3.4%
`(⫾ 2.3%) of p14 CD8⫹ T cells in cpCML and on 7.9% (⫾ 3.2%) in
`bcCML (Figure 5C). Therefore, PD-1 expression in cpCML and in
`bcCML is not restricted to leukemia-specific CTLs. However, the
`majority of leukemia-specific CTLs expressed PD-1 (Figure 5A).
`We next investigated the expression of both PD-Ls on granulo-
`cytes in various organs, such as bone marrow, spleen, inguinal and
`mesenterial lymph nodes, thymus, and peripheral blood. PD-L1
`was up-regulated on leukemic (GFP⫹) granulocytes in H8-cpCML
`and H8-bcCML in spleen (H8-cpCML, 38%; H8-bcCML, 100%)
`and bone marrow compared with the granulocyte population of
`naive C57BL/6 mice (Figure 5D). In contrast, PD-L2 was not
`expressed on leukemic cells in H8-cpCML and H8-bcCML mice
`(Figure 5D).
`
`Figure 2. Presence of leukemia-specific CTLs in CML. Bone marrow from H8 mice
`was transduced with BCR/ABL-GFP or cotransduced with NUP98/HOXA9-GFP and
`BCR/ABL-neo and injected into 4.5 Gy irradiated C57BL/6 mice. (A-B) The frequency
`of gp33-specific CTLs was analyzed by tetramer staining (A) and intracellular IFN-␥
`and TNF-␣ staining (B) in the blood of mice that eliminated H8-cpCML and
`H8-bcCML, in mice with progressive H8-cpCML and H8-bcCML, in naive C57BL/6,
`and in LCMV-immune mice. (C) Splenocytes of H8-cpCML and H8-bcCML mice were
`isolated 20 days after BMT and analyzed in a standard 51Cr-release assay (gp33-
`pulsed target cells [filled symbols]; nonpulsed target cells [open symbols]). CTL
`activity is given as mean ⫾ SEM of 4 CML mice (F), naive C57BL/6 (⽧), and
`LCMV-immune mice (f). (D) H8-cpCML and H8-bcCML mice and C57BL/6 controls
`were infected with 200 pfu of LCMV intravenously. Eight days after LCMV infection,
`the frequency of gp33-specific and np396-specific CTLs was analyzed in the blood
`and spleen by tetramer staining. One representative experiment of 2 is shown.
`
`UPenn Ex. 2059
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`BLOOD, 20 AUGUST 2009 䡠 VOLUME 114, NUMBER 8
`
`EXHAUSTION OF CML-SPECIFIC T CELLS BY PD-1 IMMUNOBIOLOGY
`
`1533
`
`Figure 4. Functional characterization of transferred TCR transgenic CD8ⴙ
`T cells. P14 CD8⫹ T cells were isolated 5 or 6 days after transfer from the spleens of
`H8-bcCML mice, C57BL/6 controls, and LCMV-infected mice. (A) The production of
`IFN-␥, TNF-␣, and IL-2 by p14 CD8⫹ T cells was determined in the supernatant with a
`cytometric bead array assay. Results are given as mean ⫾ SEM of 3 to 10 samples
`per group, pooled from 4 independent experiments. (B) Splenocytes were analyzed
`in a 51Cr-release assay (gp33-pulsed target cells [filled symbols]; nonpulsed target
`cells [open symbols]). CTL activity is given as mean ⫾ SEM of 4 H8-bcCML mice (F),
`C57BL/6 controls (⽧), and LCMV-infected mice (f). One representative experiment
`of 2 is shown. (C) FACS analysis of blood of H8-bcCML mice before and 6 days after
`adoptive transfer of p14 CD8⫹ T cells and control H8-bcCML mice. One representa-
`tive staining of 5 independent experiments is shown. (D) Isolated p14 CD8⫹ T cells
`were restimulated in vitro. 3H-thymidine incorporation of isolated p14 CD8⫹ T cells is
`shown as proliferation index (mean ⫾ SEM of 4-10 mice per group). Results are
`pooled from 3 independent experiments.
`
`To functionally analyze the role of PD-L1 expressed on
`leukemic cells, H8-cpCML and H8-bcCML cells were used as
`stimulators in a 3H-thymidine incorporation assay with either p14
`or p14 ⫻ PD-1⫺/⫺ CD8⫹ T cells as responders. H8-cpCML and
`H8-bcCML cells stimulated only a limited expansion of p14 CD8⫹
`T cells, whereas p14 CD8⫹ T cells proliferated efficiently when
`stimulated with splenocytes from naive H8 mice. In contrast,
`p14 ⫻ PD-1⫺/⫺ CD8⫹ T cells efficiently expanded when stimu-
`lated with H8-cpCML and H8-bcCML cells (Figure 5E).
`In summary, PD-1 is expressed on CML-specific CTLs and
`PD-L1 on leukemic cells with higher expression during bcCML
`than in cpCML.
`
`Prolonged survival of bcCML mice in the absence of PD-1
`signaling
`
`Our data to date using the LCMV-gp33 as model leukemia antigen
`suggested that PD-1 may inhibit leukemia-specific CTLs and lead
`to disease progression. LCMV-gp33 is a foreign antigen that is
`expressed in the H8 transgenic mice under a relatively strong
`promoter. Therefore, the model leukemia antigen used has many
`similarities to the junction peptides derived of BCR/ABL, which
`are similarly expressed under a strong promoter and are novel
`antigens without preexisting self-tolerance. Nevertheless, the H8-
`CML model might overestimate the contribution of CD8⫹ T cells
`and the role of PD-1 in CML-specific tolerance induction. To test
`the physiologic role of PD-1 in CML control, bcCML was induced
`in PD-1–deficient mice and C57BL/6 control mice using trans-
`
`Figure 5. PD-1 and PD-L expression in H8-cpCML and H8-bcCML mice. (A) PD-1
`expression on purified p14 CD8⫹ T cells (CD45.1⫹CD8⫹Va2⫹) isolated from the
`spleen 5 days after transfer. One representative histogram from 3 to 5 mice per group
`is shown. One representative experiment of 2 is shown. (B) CTLA-4 expression on
`purified p14 CD8⫹ T cells (CD45.1⫹CD8⫹V␣2⫹) isolated from the spleen 7 days after
`transfer. (C) cpCML and bcCML were generated from C57BL/6 donors in p14
`recipient mice. PD-1 expression on total CD8⫹ T cells and on unspecific CD8⫹ T cells
`(V␣2⫹) in the spleen was analyzed 20 days after bone marrow transplantation. One
`representative FACS plot of 3 is shown. (D) PD-L1 and PD-L2 expression in the
`spleen and bone marrow of H8-cpCML, H8-bcCML, and naive C57BL/6 mice. Cells
`were gated on GFP-positive gran

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