Skip to main content
Erschienen in: Cancer Immunology, Immunotherapy 11/2023

02.08.2023 | Research

BTK inhibition potentiates anti-PD-L1 treatment in murine melanoma: potential role for MDSC modulation in immunotherapy

verfasst von: Steven H. Sun, Colin D. Angell, Himanshu Savardekar, Debasish Sundi, David Abood, Brooke Benner, Mallory J. DiVincenzo, Megan Duggan, Fouad Choueiry, Thomas Mace, Prashant Trikha, Gabriella Lapurga, Courtney Johnson, Erick J. Carlson, Catherine Chung, Blake R. Peterson, Lianbo Yu, Jing Zhao, Kari L. Kendra, William E. Carson III

Erschienen in: Cancer Immunology, Immunotherapy | Ausgabe 11/2023

Einloggen, um Zugang zu erhalten

Abstract

Myeloid-derived suppressor cells (MDSC) have been linked to loss of immune effector cell function through a variety of mechanisms such as the generation of reactive oxygen and nitrogen species and the production of inhibitory cytokines. Our group has shown that signaling through Bruton’s tyrosine kinase (BTK) is important for MDSC function. Ibrutinib is an orally administered targeted agent that inhibits BTK activation and is currently used for the treatment of B cell malignancies. Using a syngeneic murine model of melanoma, the effect of BTK inhibition with ibrutinib on the therapeutic response to systemic PD-L1 blockade was studied. BTK was expressed by murine MDSC and their activation was inhibited by ibrutinib. Ibrutinib was not directly cytotoxic to cancer cells in vitro, but it inhibited BTK activation in MDSC and reduced expression of inducible nitric oxide synthase (NOS2) and production of nitric oxide. Ibrutinib treatments decreased the levels of circulating MDSC in vivo and increased the therapeutic efficacy of anti-PD-L1 antibody treatment. Gene expression profiling showed that ibrutinib decreased Cybb (NOX2) signaling, and increased IL-17 signaling (upregulating downstream targets Mmp9, Ptgs2, and S100a8). These results suggest that further exploration of MDSC inhibition could enhance the immunotherapy of advanced melanoma.
Précis
Inhibition of Bruton’s tyrosine kinase, a key enzyme in myeloid cellular function, improves therapeutic response to an anti-PD-L1 antibody in an otherwise fairly resistant murine melanoma model.
Literatur
1.
Zurück zum Zitat Bissell MJ, Hines WC (2011) Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med 17(3):320–329PubMedPubMedCentralCrossRef Bissell MJ, Hines WC (2011) Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med 17(3):320–329PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Trikha P, Plews RL, Stiff A et al (2016) Targeting myeloid-derived suppressor cells using a novel adenosine monophosphate-activated protein kinase (AMPK) activator. Oncoimmunology 5(9):e1214787PubMedPubMedCentralCrossRef Trikha P, Plews RL, Stiff A et al (2016) Targeting myeloid-derived suppressor cells using a novel adenosine monophosphate-activated protein kinase (AMPK) activator. Oncoimmunology 5(9):e1214787PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Youn JI, Nagaraj S, Collazo M, Gabrilovich DI (2008) Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol 181(8):5791–5802PubMedCrossRef Youn JI, Nagaraj S, Collazo M, Gabrilovich DI (2008) Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol 181(8):5791–5802PubMedCrossRef
4.
Zurück zum Zitat Trikha P, Carson WE III (2014) Signaling pathways involved in MDSC regulation. Biochim Biophys Acta (BBA)-Rev Cancer. 1:55–65CrossRef Trikha P, Carson WE III (2014) Signaling pathways involved in MDSC regulation. Biochim Biophys Acta (BBA)-Rev Cancer. 1:55–65CrossRef
5.
Zurück zum Zitat Mandruzzato S, Solito S, Falisi E et al (2009) IL4Rα+ myeloid-derived suppressor cell expansion in cancer patients. J Immunol 182(10):6562–6568PubMedCrossRef Mandruzzato S, Solito S, Falisi E et al (2009) IL4Rα+ myeloid-derived suppressor cell expansion in cancer patients. J Immunol 182(10):6562–6568PubMedCrossRef
6.
7.
Zurück zum Zitat Markowitz J, Brooks TR, Duggan MC et al (2015) Patients with pancreatic adenocarcinoma exhibit elevated levels of myeloid-derived suppressor cells upon progression of disease. Cancer Immunol Immunother 64(2):149–159PubMedCrossRef Markowitz J, Brooks TR, Duggan MC et al (2015) Patients with pancreatic adenocarcinoma exhibit elevated levels of myeloid-derived suppressor cells upon progression of disease. Cancer Immunol Immunother 64(2):149–159PubMedCrossRef
8.
Zurück zum Zitat Zhang S, Ma X, Zhu C, Liu L, Wang G, Yuan X (2016) The role of myeloid-derived suppressor cells in patients with solid tumors: a meta-analysis. PLoS ONE 11(10):e0164514PubMedPubMedCentralCrossRef Zhang S, Ma X, Zhu C, Liu L, Wang G, Yuan X (2016) The role of myeloid-derived suppressor cells in patients with solid tumors: a meta-analysis. PLoS ONE 11(10):e0164514PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Gonda K, Shibata M, Ohtake T et al (2017) Myeloid-derived suppressor cells are increased and correlated with type 2 immune responses, malnutrition, inflammation, and poor prognosis in patients with breast cancer. Oncol Lett 14(2):1766–1774PubMedPubMedCentralCrossRef Gonda K, Shibata M, Ohtake T et al (2017) Myeloid-derived suppressor cells are increased and correlated with type 2 immune responses, malnutrition, inflammation, and poor prognosis in patients with breast cancer. Oncol Lett 14(2):1766–1774PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Parker KH, Beury DW, Ostrand-Rosenberg S (2015) Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv Cancer Res 128:95–139PubMedPubMedCentralCrossRef Parker KH, Beury DW, Ostrand-Rosenberg S (2015) Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv Cancer Res 128:95–139PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Khair DO, Bax HJ, Mele S et al (2019) Combining immune checkpoint inhibitors: established and emerging targets and strategies to improve outcomes in melanoma. Front Immunol 10:453PubMedPubMedCentralCrossRef Khair DO, Bax HJ, Mele S et al (2019) Combining immune checkpoint inhibitors: established and emerging targets and strategies to improve outcomes in melanoma. Front Immunol 10:453PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Hodi FS, O’day SJ, McDermott DF et al (2010) Improved survival with ipilimumab in patients with metastatic melanoma. New Eng J Med 363(8):711PubMedCrossRef Hodi FS, O’day SJ, McDermott DF et al (2010) Improved survival with ipilimumab in patients with metastatic melanoma. New Eng J Med 363(8):711PubMedCrossRef
14.
Zurück zum Zitat Topalian SL, Sznol M, McDermott DF et al (2014) Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32(10):1020PubMedPubMedCentralCrossRef Topalian SL, Sznol M, McDermott DF et al (2014) Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32(10):1020PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Robert C, Schachter J, Long GV et al (2015) Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 372(26):2521–2532PubMedCrossRef Robert C, Schachter J, Long GV et al (2015) Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 372(26):2521–2532PubMedCrossRef
16.
17.
Zurück zum Zitat Mueller H, Stadtmann A, Van Aken H et al (2010) Tyrosine kinase Btk regulates E-selectin–mediated integrin activation and neutrophil recruitment by controlling phospholipase C (PLC) γ2 and PI3Kγ pathways. Blood J Am Soc Hematol 115(15):3118–3127 Mueller H, Stadtmann A, Van Aken H et al (2010) Tyrosine kinase Btk regulates E-selectin–mediated integrin activation and neutrophil recruitment by controlling phospholipase C (PLC) γ2 and PI3Kγ pathways. Blood J Am Soc Hematol 115(15):3118–3127
18.
Zurück zum Zitat Lee KG, Xu S, Kang ZH et al (2012) Bruton’s tyrosine kinase phosphorylates Toll-like receptor 3 to initiate antiviral response. Proc Natl Acad Sci 109(15):5791–5796PubMedPubMedCentralCrossRef Lee KG, Xu S, Kang ZH et al (2012) Bruton’s tyrosine kinase phosphorylates Toll-like receptor 3 to initiate antiviral response. Proc Natl Acad Sci 109(15):5791–5796PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Bunt SK, Clements VK, Hanson EM, Sinha P, Ostrand-Rosenberg S (2009) Inflammation enhances myeloid-derived suppressor cell cross-talk by signaling through Toll-like receptor 4. J Leukoc Biol 85(6):996–1004PubMedPubMedCentralCrossRef Bunt SK, Clements VK, Hanson EM, Sinha P, Ostrand-Rosenberg S (2009) Inflammation enhances myeloid-derived suppressor cell cross-talk by signaling through Toll-like receptor 4. J Leukoc Biol 85(6):996–1004PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Pal Singh S, Dammeijer F, Hendriks RW (2018) Role of Bruton’s tyrosine kinase in B cells and malignancies. Mol Cancer 17(1):1–23CrossRef Pal Singh S, Dammeijer F, Hendriks RW (2018) Role of Bruton’s tyrosine kinase in B cells and malignancies. Mol Cancer 17(1):1–23CrossRef
21.
Zurück zum Zitat Stiff A, Trikha P, Wesolowski R et al (2016) Myeloid-derived suppressor cells express bruton’s tyrosine kinase and can be depleted in tumor-bearing hosts by ibrutinib treatment ibrutinib impairs MDSC generation and function. Cancer Res 76(8):2125–2136PubMedPubMedCentralCrossRef Stiff A, Trikha P, Wesolowski R et al (2016) Myeloid-derived suppressor cells express bruton’s tyrosine kinase and can be depleted in tumor-bearing hosts by ibrutinib treatment ibrutinib impairs MDSC generation and function. Cancer Res 76(8):2125–2136PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Herman SEM, Mustafa RZ, Gyamfi JA et al (2014) Ibrutinib inhibits BCR and NF-κB signaling and reduces tumor proliferation in tissue-resident cells of patients with CLL. Blood J Am Soc Hematol 123(21):3286–3295 Herman SEM, Mustafa RZ, Gyamfi JA et al (2014) Ibrutinib inhibits BCR and NF-κB signaling and reduces tumor proliferation in tissue-resident cells of patients with CLL. Blood J Am Soc Hematol 123(21):3286–3295
23.
24.
Zurück zum Zitat Chang BY, Huang MM, Francesco M et al (2011) The Bruton tyrosine kinase inhibitor PCI-32765 ameliorates autoimmune arthritis by inhibition of multiple effector cells. Arthritis Res Ther 13(4):1–15CrossRef Chang BY, Huang MM, Francesco M et al (2011) The Bruton tyrosine kinase inhibitor PCI-32765 ameliorates autoimmune arthritis by inhibition of multiple effector cells. Arthritis Res Ther 13(4):1–15CrossRef
25.
Zurück zum Zitat Apolloni E, Bronte V, Mazzoni A et al (2000) Immortalized myeloid suppressor cells trigger apoptosis in antigen-activated T lymphocytes. J Immunol 165(12):6723–6730PubMedCrossRef Apolloni E, Bronte V, Mazzoni A et al (2000) Immortalized myeloid suppressor cells trigger apoptosis in antigen-activated T lymphocytes. J Immunol 165(12):6723–6730PubMedCrossRef
26.
Zurück zum Zitat Mundy-Bosse BL, Young GS, Bauer T et al (2011) Distinct myeloid suppressor cell subsets correlate with plasma IL-6 and IL-10 and reduced interferon-alpha signaling in CD4+ T cells from patients with GI malignancy. Cancer Immunol Immunother 60(9):1269–1279PubMedPubMedCentralCrossRef Mundy-Bosse BL, Young GS, Bauer T et al (2011) Distinct myeloid suppressor cell subsets correlate with plasma IL-6 and IL-10 and reduced interferon-alpha signaling in CD4+ T cells from patients with GI malignancy. Cancer Immunol Immunother 60(9):1269–1279PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Zhu J, Inomata T, Fujimoto K et al (2021) Ex Vivo-induced bone marrow-derived myeloid suppressor cells prevent corneal allograft rejection in mice. Invest Ophthalmol Vis Sci 62(7):3PubMedPubMedCentralCrossRef Zhu J, Inomata T, Fujimoto K et al (2021) Ex Vivo-induced bone marrow-derived myeloid suppressor cells prevent corneal allograft rejection in mice. Invest Ophthalmol Vis Sci 62(7):3PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Liu X, Quan N (2015) Immune cell isolation from mouse femur bone marrow. Bio Protoc 5(20):e1631–e1631PubMed Liu X, Quan N (2015) Immune cell isolation from mouse femur bone marrow. Bio Protoc 5(20):e1631–e1631PubMed
30.
Zurück zum Zitat Rane D, Carlson EJ, Yin Y, Peterson BR. Fluorescent detection of peroxynitrite during antibody-dependent cellular phagocytosis. In: Methods in Enzymology. Vol 640. Elsevier; 2020:1–35. Rane D, Carlson EJ, Yin Y, Peterson BR. Fluorescent detection of peroxynitrite during antibody-dependent cellular phagocytosis. In: Methods in Enzymology. Vol 640. Elsevier; 2020:1–35.
31.
Zurück zum Zitat Knewtson KE, Rane D, Peterson BR (2018) Targeting fluorescent sensors to endoplasmic reticulum membranes enables detection of peroxynitrite during cellular phagocytosis. ACS Chem Biol 13(9):2595–2602PubMedPubMedCentralCrossRef Knewtson KE, Rane D, Peterson BR (2018) Targeting fluorescent sensors to endoplasmic reticulum membranes enables detection of peroxynitrite during cellular phagocytosis. ACS Chem Biol 13(9):2595–2602PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Corzo CA, Cotter MJ, Cheng P et al (2009) Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol 182(9):5693–5701PubMedCrossRef Corzo CA, Cotter MJ, Cheng P et al (2009) Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol 182(9):5693–5701PubMedCrossRef
33.
Zurück zum Zitat Stiff A, Trikha P, Mundy-Bosse B et al (2018) Nitric oxide production by myeloid-derived suppressor cells plays a role in impairing fc receptor-mediated natural killer cell function MDSC inhibit antibody therapy via nitric oxide production. Clin Cancer Res 24(8):1891–1904PubMedPubMedCentralCrossRef Stiff A, Trikha P, Mundy-Bosse B et al (2018) Nitric oxide production by myeloid-derived suppressor cells plays a role in impairing fc receptor-mediated natural killer cell function MDSC inhibit antibody therapy via nitric oxide production. Clin Cancer Res 24(8):1891–1904PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Serafini P, Borrello I, Bronte V. Myeloid suppressor cells in cancer: recruitment, phenotype, properties, and mechanisms of immune suppression. In: Seminars in Cancer Biology. Vol 16. Elsevier; 2006:53–65. Serafini P, Borrello I, Bronte V. Myeloid suppressor cells in cancer: recruitment, phenotype, properties, and mechanisms of immune suppression. In: Seminars in Cancer Biology. Vol 16. Elsevier; 2006:53–65.
35.
Zurück zum Zitat Schneider T, Sevko A, Heussel CP et al (2015) Serum inflammatory factors and circulating immunosuppressive cells are predictive markers for efficacy of radiofrequency ablation in non-small-cell lung cancer. Clin Exp Immunol 180(3):467–474PubMedPubMedCentralCrossRef Schneider T, Sevko A, Heussel CP et al (2015) Serum inflammatory factors and circulating immunosuppressive cells are predictive markers for efficacy of radiofrequency ablation in non-small-cell lung cancer. Clin Exp Immunol 180(3):467–474PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Wesolowski R, Markowitz J, Carson WE (2013) Myeloid derived suppressor cells–a new therapeutic target in the treatment of cancer. J Immunother Cancer 1(1):1–11CrossRef Wesolowski R, Markowitz J, Carson WE (2013) Myeloid derived suppressor cells–a new therapeutic target in the treatment of cancer. J Immunother Cancer 1(1):1–11CrossRef
37.
Zurück zum Zitat Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP (2007) Amino-biphosphonate–mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res 67(23):11438–11446PubMedPubMedCentralCrossRef Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP (2007) Amino-biphosphonate–mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res 67(23):11438–11446PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Srikrishna G (2012) S100A8 and S100A9: new insights into their roles in malignancy. J Innate Immun 4(1):31–40PubMedCrossRef Srikrishna G (2012) S100A8 and S100A9: new insights into their roles in malignancy. J Innate Immun 4(1):31–40PubMedCrossRef
39.
Zurück zum Zitat Bercusson A, Colley T, Shah A, Warris A, Armstrong-James D (2018) Ibrutinib blocks Btk-dependent NF-ĸB and NFAT responses in human macrophages during Aspergillus fumigatus phagocytosis. Blood J Am Soc Hematol 132(18):1985–1988 Bercusson A, Colley T, Shah A, Warris A, Armstrong-James D (2018) Ibrutinib blocks Btk-dependent NF-ĸB and NFAT responses in human macrophages during Aspergillus fumigatus phagocytosis. Blood J Am Soc Hematol 132(18):1985–1988
40.
Zurück zum Zitat Fraietta JA, Beckwith KA, Patel PR et al (2016) Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood J Am Soc Hematol 127(9):1117–1127 Fraietta JA, Beckwith KA, Patel PR et al (2016) Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood J Am Soc Hematol 127(9):1117–1127
41.
42.
Zurück zum Zitat Veglia F, Hashimoto A, Dweep H et al (2021) Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice. J Exp Med 218(4):1CrossRef Veglia F, Hashimoto A, Dweep H et al (2021) Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice. J Exp Med 218(4):1CrossRef
44.
Zurück zum Zitat Weber R, Fleming V, Hu X, et al. Myeloid-derived suppressor cells hinder the anti-cancer activity of immune checkpoint inhibitors. Front Immunol. 2018:1310. Weber R, Fleming V, Hu X, et al. Myeloid-derived suppressor cells hinder the anti-cancer activity of immune checkpoint inhibitors. Front Immunol. 2018:1310.
45.
Zurück zum Zitat Kim K, Skora AD, Li Z et al (2014) Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci 111(32):11774–11779PubMedPubMedCentralCrossRef Kim K, Skora AD, Li Z et al (2014) Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci 111(32):11774–11779PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Fleming V, Hu X, Weller C et al (2019) Melanoma extracellular vesicles generate immunosuppressive myeloid cells by upregulating PD-L1 via TLR4 signaling induction of immunosuppression by extracellular vesicles. Cancer Res 79(18):4715–4728PubMedCrossRef Fleming V, Hu X, Weller C et al (2019) Melanoma extracellular vesicles generate immunosuppressive myeloid cells by upregulating PD-L1 via TLR4 signaling induction of immunosuppression by extracellular vesicles. Cancer Res 79(18):4715–4728PubMedCrossRef
47.
48.
Zurück zum Zitat Holtzhausen A, Harris W, Ubil E et al (2019) TAM family receptor kinase inhibition reverses MDSC-mediated suppression and augments anti–PD-1 therapy in melanoma TAM RTK inhibition reverses MDSC suppression and augments PD-1. Cancer Immunol Res 7(10):1672–1686PubMedPubMedCentralCrossRef Holtzhausen A, Harris W, Ubil E et al (2019) TAM family receptor kinase inhibition reverses MDSC-mediated suppression and augments anti–PD-1 therapy in melanoma TAM RTK inhibition reverses MDSC suppression and augments PD-1. Cancer Immunol Res 7(10):1672–1686PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Grauers Wiktorin H, Nilsson MS, Kiffin R et al (2019) Histamine targets myeloid-derived suppressor cells and improves the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade. Cancer Immunol Immunother 68(2):163–174PubMedCrossRef Grauers Wiktorin H, Nilsson MS, Kiffin R et al (2019) Histamine targets myeloid-derived suppressor cells and improves the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade. Cancer Immunol Immunother 68(2):163–174PubMedCrossRef
50.
Zurück zum Zitat Highfill SL, Cui Y, Giles AJ et al (2014) Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci Transl Med. 6(237):ra67-237CrossRef Highfill SL, Cui Y, Giles AJ et al (2014) Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci Transl Med. 6(237):ra67-237CrossRef
51.
Zurück zum Zitat Zhou J, Liu M, Sun H et al (2018) Hepatoma-intrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut 67(5):931–944PubMedCrossRef Zhou J, Liu M, Sun H et al (2018) Hepatoma-intrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut 67(5):931–944PubMedCrossRef
52.
Zurück zum Zitat Benner B, Quiroga DM, Good L, et al. A pilot study of Bruton’s tyrosine kinase inhibitor ibrutinib alone and in combination with PD-1 inhibitor nivolumab in patients with metastatic solid tumors. 2020. Benner B, Quiroga DM, Good L, et al. A pilot study of Bruton’s tyrosine kinase inhibitor ibrutinib alone and in combination with PD-1 inhibitor nivolumab in patients with metastatic solid tumors. 2020.
53.
Zurück zum Zitat Young MRI. Th17 cells in protection from tumor or promotion of tumor progression. J Clin Cell Immunol. 2016;7(3). Young MRI. Th17 cells in protection from tumor or promotion of tumor progression. J Clin Cell Immunol. 2016;7(3).
54.
Zurück zum Zitat Wen L, Gong P, Liang C et al (2016) Interplay between myeloid-derived suppressor cells (MDSCs) and Th17 cells: foe or friend? Oncotarget 7(23):35490PubMedPubMedCentralCrossRef Wen L, Gong P, Liang C et al (2016) Interplay between myeloid-derived suppressor cells (MDSCs) and Th17 cells: foe or friend? Oncotarget 7(23):35490PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Ma M, Huang W, Kong D (2018) IL-17 inhibits the accumulation of myeloid-derived suppressor cells in breast cancer via activating STAT3. Int Immunopharmacol 59:148–156PubMedCrossRef Ma M, Huang W, Kong D (2018) IL-17 inhibits the accumulation of myeloid-derived suppressor cells in breast cancer via activating STAT3. Int Immunopharmacol 59:148–156PubMedCrossRef
56.
Zurück zum Zitat Li J, Liu J, Mao X, Tang Q, Lu H (2014) IL-7 receptor blockade inhibits IL-17-producing γδ cells and suppresses melanoma development. Inflammation 37(5):1444–1452PubMedCrossRef Li J, Liu J, Mao X, Tang Q, Lu H (2014) IL-7 receptor blockade inhibits IL-17-producing γδ cells and suppresses melanoma development. Inflammation 37(5):1444–1452PubMedCrossRef
57.
Metadaten
Titel
BTK inhibition potentiates anti-PD-L1 treatment in murine melanoma: potential role for MDSC modulation in immunotherapy
verfasst von
Steven H. Sun
Colin D. Angell
Himanshu Savardekar
Debasish Sundi
David Abood
Brooke Benner
Mallory J. DiVincenzo
Megan Duggan
Fouad Choueiry
Thomas Mace
Prashant Trikha
Gabriella Lapurga
Courtney Johnson
Erick J. Carlson
Catherine Chung
Blake R. Peterson
Lianbo Yu
Jing Zhao
Kari L. Kendra
William E. Carson III
Publikationsdatum
02.08.2023
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Immunology, Immunotherapy / Ausgabe 11/2023
Print ISSN: 0340-7004
Elektronische ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-023-03497-1

Weitere Artikel der Ausgabe 11/2023

Cancer Immunology, Immunotherapy 11/2023 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.