Sorafenib, but not sunitinib, affects function of dendritic cells and induction of primary immune responses by Madeleine M. Hipp, Norbert Hilf, Steffen.

Slides:



Advertisements
Similar presentations
An anti-CD19 antibody inhibits the interaction between P-glycoprotein (P-gp) and CD19, causes P-gp to translocate out of lipid rafts, and chemosensitizes.
Advertisements

Supplementary Figure S1.
Volume 132, Issue 3, Pages (March 2007)
Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activation by Juliette Mouriès, Gabriel Moron, Géraldine Schlecht,
Cheng-Ming Sun, Edith Deriaud, Claude Leclerc, Richard Lo-Man  Immunity 
Involvement of suppressors of cytokine signaling in toll-like receptor–mediated block of dendritic cell differentiation by Holger Bartz, Nicole M. Avalos,
Host-Derived CD8+ Dendritic Cells Protect Against Acute Graft-versus-Host Disease after Experimental Allogeneic Bone Marrow Transplantation  Michael Weber,
by JoAnn Castelli, Elaine K
Depletion of Alloreactive Donor T Lymphocytes by CD95-Mediated Activation-Induced Cell Death Retains Antileukemic, Antiviral, and Immunoregulatory T Cell.
Dendritic cell differentiation potential of mouse monocytes: monocytes represent immediate precursors of CD8- and CD8+ splenic dendritic cells by Beatriz.
by Rosemary E. Smith, Vanshree Patel, Sandra D. Seatter, Maureen R
Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor–κB and IκBα kinase in human multiple myeloma cells, leading to.
The TCL1 oncoprotein inhibits activation-induced cell death by impairing PKCθ and ERK pathways by Gilles Despouy, Marjorie Joiner, Emilie Le Toriellec,
The CXC-chemokine platelet factor 4 promotes monocyte survival and induces monocyte differentiation into macrophages by Barbara Scheuerer, Martin Ernst,
Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat-5 independent signaling pathway by Kevin M. Elias, Arian Laurence,
Enhancement of intracellular signaling associated with hematopoietic progenitor cell survival in response to SDF-1/CXCL12 in synergy with other cytokines.
Non-small Cell Lung Cancer Induces an Immunosuppressive Phenotype of Dendritic Cells in Tumor Microenvironment by Upregulating B7-H3  Thomas Schneider,
Megakaryocyte Growth and Development Factor-Induced Proliferation and Differentiation Are Regulated by the Mitogen-Activated Protein Kinase Pathway in.
by Juan C. Rodríguez-Alba, Miguel E
Following the Development of a CD4 T Cell Response In Vivo
Commonly used prophylactic vaccines as an alternative for synthetically produced TLR ligands to mature monocyte-derived dendritic cells by Gerty Schreibelt,
by Norman Nausch, Ioanna E
Ex vivo induction of multiple myeloma–specific cytotoxic T lymphocytes
Macrophages from C3-deficient mice have impaired potency to stimulate alloreactive T cells by Wuding Zhou, Hetal Patel, Ke Li, Qi Peng, Marie-Bernadette.
Interleukin-21 is a growth and survival factor for human myeloma cells
Prostaglandin E2 is a key factor for CCR7 surface expression and migration of monocyte-derived dendritic cells by Elke Scandella, Ying Men, Silke Gillessen,
Hyaluronate-Enhanced Hematopoiesis: Two Different Receptors Trigger the Release of Interleukin-1β and Interleukin-6 From Bone Marrow Macrophages by Sophia.
Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients by Devi.
Increased survival is a selective feature of human circulating antigen-induced plasma cells synthesizing high-affinity antibodies by Inés González-García,
The heat shock protein Gp96 binds to human neutrophils and monocytes and stimulates effector functions by Markus P. Radsak, Norbert Hilf, Harpreet Singh-Jasuja,
Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy by Fumihiko Tsushima, Sheng Yao, Tahiro Shin, Andrew Flies, Sarah.
Histone deacetylase inhibitors: a new class of immunosuppressors targeting a novel signal pathway essential for CD154 expression by Søren Skov, Klaus Rieneck,
Noncanonical NF-κB signaling in dendritic cells is required for indoleamine 2,3-dioxygenase (IDO) induction and immune regulation by Sander W. Tas, Margriet.
Functional human regulatory T cells fail to control autoimmune inflammation due to PKB/c-akt hyperactivation in effector cells by Ellen J. Wehrens, Gerdien.
Targeting the nuclear antigen 1 of Epstein-Barr virus to the human endocytic receptor DEC-205 stimulates protective T-cell responses by Cagan Gurer, Till.
FLT3 ligand administration after hematopoietic cell transplantation increases circulating dendritic cell precursors that can be activated by CpG oligodeoxynucleotides.
Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression by Giovanna Borsellino, Markus Kleinewietfeld,
CD134-Allodepletion Allows Selective Elimination of Alloreactive Human T Cells without Loss of Virus-Specific and Leukemia-Specific Effectors  Xupeng.
Ifosfamide impairs the allostimulatory capacity of human dendritic cells by intracellular glutathione depletion by Maria C. Kuppner, Anabel Scharner, Valeria.
by Bindu Varghese, Adam Widman, James Do, Behnaz Taidi, Debra K
CC chemokine ligand 20 partially controls adhesion of naive B cells to activated endothelial cells under shear stress by Anja Meissner, Olaf Zilles, Rosa.
TGF-β combined with M-CSF and IL-4 induces generation of immune inhibitory cord blood dendritic cells capable of enhancing cytokine-induced ex vivo expansion.
Human Keratinocytes Express Functional CD14 and Toll-Like Receptor 4
Tuning the volume of the immune response: strength and persistence of stimulation determine migration and cytokine secretion of dendritic cells by Thomas.
Volume 18, Issue 5, Pages (May 2003)
Akio Horiguchi, Mototsugu Oya, Ken Marumo, Masaru Murai 
Non-small Cell Lung Cancer Induces an Immunosuppressive Phenotype of Dendritic Cells in Tumor Microenvironment by Upregulating B7-H3  Thomas Schneider,
Volume 13, Issue 12, Pages (December 2015)
Volume 24, Issue 6, Pages (June 2006)
Volume 29, Issue 6, Pages (December 2008)
P38 Mitogen-Activated Protein Kinase Mediates Dual Role of Ultraviolet B Radiation in Induction of Maturation and Apoptosis of Monocyte-Derived Dendritic.
Volume 22, Issue 2, Pages (February 2005)
Volume 19, Issue 3, Pages (September 2003)
CD25 expression distinguishes functionally distinct alloreactive CD4+ CD134+ (OX40+) T-cell subsets in acute graft-versus-host disease  Philip R Streeter,
Volume 13, Issue 2, Pages (February 2006)
Volume 32, Issue 5, Pages (May 2010)
Fateh Ouaaz, Joseph Arron, Ye Zheng, Yongwon Choi, Amer A Beg  Immunity 
by Derek Hoi-Hang Ho, and Roger Hoi-Fung Wong
Opposing Effects of TGF-β and IL-15 Cytokines Control the Number of Short-Lived Effector CD8+ T Cells  Shomyseh Sanjabi, Munir M. Mosaheb, Richard A.
Human CD4+ T Lymphocytes with Remarkable Regulatory Functions on Dendritic Cells and Nickel-Specific Th1 Immune Responses  Andrea Cavani, Francesca Nasorri,
by Defne Bayik, Debra Tross, Lydia A
Human Beta-Defensin 3 Induces Maturation of Human Langerhans Cell–Like Dendritic Cells: An Antimicrobial Peptide that Functions as an Endogenous Adjuvant 
Volume 21, Issue 1, Pages (July 2004)
Volume 11, Issue 3, Pages (September 1999)
Volume 33, Issue 1, Pages (July 2010)
Volume 17, Issue 5, Pages (May 2009)
Notch 1 Signaling Regulates Peripheral T Cell Activation
ALT-803 stimulates proliferation and activation of human NK cells and T cells in vitro. ALT-803 stimulates proliferation and activation of human NK cells.
Pretreatment of donor T cells with a c-Rel antagonist does not impair GVT activity. Pretreatment of donor T cells with a c-Rel antagonist does not impair.
Rapamycin inhibits IL-4—induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo by Holger Hackstein, Timucin Taner,
Presentation transcript:

Sorafenib, but not sunitinib, affects function of dendritic cells and induction of primary immune responses by Madeleine M. Hipp, Norbert Hilf, Steffen Walter, Daniela Werth, Katharina M. Brauer, Markus P. Radsak, Toni Weinschenk, Harpreet Singh-Jasuja, and Peter Brossart Blood Volume 111(12):5610-5620 June 15, 2008 ©2008 by American Society of Hematology

Sorafenib lowers expression of cell surface molecules on DCs stimulated with LPS. DCs were generated by incubating adherent monocytes with GM-CSF and IL-4. Sorafenib lowers expression of cell surface molecules on DCs stimulated with LPS. DCs were generated by incubating adherent monocytes with GM-CSF and IL-4. On day 5 of culture, cells were exposed for 24 hours to sunitinib (A,B) or sorafenib at the indicated concentrations (C,D). LPS (100 ng/mL) was added as a stimulus to the culture media for the last 24 hours (B,D). In addition, to analyze the effects of sorafenib on the phenotype of mature DCs sorafenib was added to the culture medium after the activation of cells with LPS for 24 hours (E). The changes in phenotype of DCs by sorafenib treatment were analyzed by flow cytometry after staining with Abs against CD1a, CD80, CD86, CD83, DC-SIGN, or CCR7. Shaded histograms represent isotype control and open diagrams staining with the specific antibody. The numbers represent mean fluorescence intensity. One representative experiment of at least 3 is shown. Madeleine M. Hipp et al. Blood 2008;111:5610-5620 ©2008 by American Society of Hematology

LPS-induced DC migration toward CCL19/MIP-3β is impaired by sorafenib. LPS-induced DC migration toward CCL19/MIP-3β is impaired by sorafenib. DCs were generated by incubating adherent monocytes with GM-CSF and IL-4. On day 5 of culture, cells were exposed for 24 hours to sorafenib (A,B) or sunitinib (C) at the indicated concentrations. LPS (100 ng/mL) was added as a stimulus to the culture media for the last 24 hours. (A) Migration toward CCL19 was analyzed using transwell chambers. DCs (105) were seeded in the upper chamber in triplicates, and the number of migrated DCs was analyzed after 4 hours by counting gated DCs for one minute by FACS analysis (*P < .05, **P < .001). (B,C) Surface expression of CCR7 was analyzed by staining of DCs with FITC conjugated antibodies against CCR7 and measuring by FACS analysis. Numbers represent mean fluorescence intensity. Madeleine M. Hipp et al. Blood 2008;111:5610-5620 ©2008 by American Society of Hematology

Treatment with sorafenib affects FITC-dextran uptake by mature DCs Treatment with sorafenib affects FITC-dextran uptake by mature DCs. Monocytes were cultured with GM-CSF and IL-4 for 7 days. Treatment with sorafenib affects FITC-dextran uptake by mature DCs. Monocytes were cultured with GM-CSF and IL-4 for 7 days. On day 5 of culture, sorafenib was added at the indicated concentrations (A,B). LPS (100 ng/mL) was added as a maturation stimulus for the last 24 hours (B). A total of 105 cells were incubated with FITC-dextran for 1 hour at 37°C and 4°C. The cells were washed and analyzed by FACS analysis. Open histograms represent FITC-dextran treated cells; solid histograms cells incubated in the absence of FITC-dextran. Mean fluorescence intensity of DCs labeled with FITC-dextran is shown. Madeleine M. Hipp et al. Blood 2008;111:5610-5620 ©2008 by American Society of Hematology

Sorafenib reduces lymphocyte stimulation capacity of DCs Sorafenib reduces lymphocyte stimulation capacity of DCs. DCs were irradiated with 30 Gy and were incubated for 5 days with 105 allogenic PBMCs or CD3+ T cells. Sorafenib reduces lymphocyte stimulation capacity of DCs. DCs were irradiated with 30 Gy and were incubated for 5 days with 105 allogenic PBMCs or CD3+ T cells. Thymidine incorporation was measured by a 16-hour pulse with [3H]thymidine. PBMCs or T cells without DCs were included as a control. (A) DCs were pretreated with sorafenib 48 hours before harvesting and immature as well as LPS-stimulated DCs (103 DCs per well) were incubated with untreated PBMCs. (B) Untreated mature DCs (103 DCs per well) were incubated with MACS isolated CD3+ T cells pretreated with different concentrations of sorafenib as indicated in the figure. (C) Untreated mature DCs (103 DCs per well) were incubated with MACS isolated CD3+ T cells pretreated with different concentrations of sorafenib as indicated in the figure. Sorafenib was added to the cell culture medium in the same concentrations during MLR. (D) DCs were pretreated with sunitinib, stimulated with LPS, and incubated with untreated PBMCs (*P < .05, ** P < .001). (E) Induction of Her-2/neu specific primary CTL responses was analyzed using LPS activated DCs pretreated with sorafenib or sunitinib and pulsed with an HLA-A2 binding Her-2/neu-derived E75 peptide. DCs pulsed with the E75 peptide as well as the HLA-A2-positive and Her-2/neu-expressing cell line A-498 were used as targets. The HLA-A2-negative and Her-2/neu-positive cell line SKOV-3, the HLA-A2-negative and Her-2/neu-negative cell line K-562, and DCs pulsed with an irrelevant HIV-derived peptide were included as negative controls. Madeleine M. Hipp et al. Blood 2008;111:5610-5620 ©2008 by American Society of Hematology

Sorafenib affects intracellular signaling pathways. Sorafenib affects intracellular signaling pathways. LPS (100 ng/mL) was added to immature DCs 15 minutes (A, top figure) or 24 hours (A, bottom figure, B-E) before harvesting. Protein levels were detected by SDS-PAGE and Western blot. Ponceau S staining was performed to ensure equal loading of the gel. (A) Sorafenib affects the TLR pathway. MyD88 and TRAF6 levels were reduced by sorafenib treatment. (B) Sorafenib reduces nuclear localization of the NF-κB family members c-Rel, Rel-B, Rel-A, and of PU.1 and IRF-3 in mature cells and nuclear translocation of Rel-B, IRF-3, and PU.1 also in immature cells. Protein levels of IRF-8 in nuclei were not affected. (C) The inhibitory effects of sorafenib partly are mediated by MAPK cascade and the PI3K/AKT pathway. Sorafenib reduces phosphorylation states of p38, whereas sorafenib induces phosphorylation of ERK. (D) Addition of sunitinib to the culture medium has no effect on the signaling pathways in DCs. (E) Addition of sunitinib to the culture medium has no effect on nuclear translocation of c-Rel, Rel-B, Rel-A, PU.1, IRF-3, and IRF-8. Representative experiments of at least 4 are presented. Madeleine M. Hipp et al. Blood 2008;111:5610-5620 ©2008 by American Society of Hematology

Sorafenib, but not sunitinib, induces apoptosis of DCs Sorafenib, but not sunitinib, induces apoptosis of DCs. DCs generated by culturing adherent monocytes in the presence of GM-CSF and IL-4 for 7 days were incubated for 48 hours with sorafenib (A) or sunitinib (B). Sorafenib, but not sunitinib, induces apoptosis of DCs. DCs generated by culturing adherent monocytes in the presence of GM-CSF and IL-4 for 7 days were incubated for 48 hours with sorafenib (A) or sunitinib (B). Cells were harvested, washed, stained according to Nicoletti et al,25 and then analyzed by flow cytometry (*P < .05, **P < .001). Madeleine M. Hipp et al. Blood 2008;111:5610-5620 ©2008 by American Society of Hematology

Sorafenib reduces strongly, but reversible specific, CD8+ T-cell responses. Sorafenib reduces strongly, but reversible specific, CD8+ T-cell responses. C57BL/6 mice were pretreated for 2 weeks with the indicated dosage of tyrosine kinase inhibitors. Thereafter, animals were immunized twice in weekly interval with OVA257-264 and adjuvants under continued treatment, as described in “Methods.” Negative controls were immunized with VSV NP52-59 peptide. One week after the last immunization, mice were killed and spleen cells were analyzed ex vivo for peptide-specific CD8+ T cells by staining with anti-CD8-FITC, anti-CD3ϵ-PerCP, VSV NP52-59/H2-Kb-APC, and OVA257-264/H2-Kb-PE tetramers. (A) Staining for both tetramers gated on CD8+ CD3ϵ+ lymphocytes is shown for a representative negative (left) and positive (right) control sample. (B) Percentage of OVA257-264/H2-Kb tetramer-positive cells among CD8+ T cells for individual mice and means (lines) are shown. Treatment with 80 mg/kg body weight for 4 weeks resulted in severe toxicity. Therefore only 2 samples were evaluable, and this group was excluded from statistical analysis. (C) In addition, percentage of CD25+ cells among blood CD4+ cells was analyzed. Means of triplicates are shown and error bars indicate the standard deviations of means. Significance was tested by unpaired, heteroscedastic Student t test (*P < .05). (D) Reduced levels of CD25+ FoxP3+ T cells among CD4+ splenocytes of nonimmunized mice can also be observed after 14 days of daily sunitinib treatment. (E,F) Reversibility of the observed effects was assessed in an additional experiment with discontinued treatment 48 hours before first immunization. Mice had been fed daily for 2 weeks with sorafenib, sunitinib, or vehicle only in the indicated concentrations. Analysis was performed as described for panels B and C. Madeleine M. Hipp et al. Blood 2008;111:5610-5620 ©2008 by American Society of Hematology