Skip to main content
Erschienen in: Cancer Immunology, Immunotherapy 7/2019

28.05.2019 | Original Article

Impact of combination immunochemotherapies on progression of 4NQO-induced murine oral squamous cell carcinoma

verfasst von: Sonja Ludwig, Chang-Sook Hong, Beatrice M. Razzo, Kellsye P. L. Fabian, Manoj Chelvanambi, Stephan Lang, Walter J. Storkus, Theresa L. Whiteside

Erschienen in: Cancer Immunology, Immunotherapy | Ausgabe 7/2019

Einloggen, um Zugang zu erhalten

Abstract

Advanced oral squamous cell carcinomas (OSCC) have limited therapeutic options. Although immune therapies are emerging as a potentially effective alternative or adjunct to chemotherapies, the therapeutic efficacy of combination immune chemotherapies has yet to be determined. Using a 4-nitroquinolone-N-oxide (4NQO) orthotopic model of OSCC in immunocompetent mice, we evaluated the therapeutic efficacy of single- and combined-agent treatment with a poly-epitope tumor peptide vaccine, cisplatin and/or an A2AR inhibitor, ZM241385. The monotherapies or their combinations resulted in a partial inhibition of tumor growth and, in some cases, a significant but transient upregulation of systemic anti-tumor CD8+ T cell responses. These responses eroded in the face of expanding immunoregulatory cell populations at later stages of tumor progression. Our findings support the need for the further development of combinatorial therapeutic approaches that could more effectively silence dominant immune inhibitory pathways operating in OSCC and provide novel, more beneficial treatment options for this tumor.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
2.
Zurück zum Zitat Leemans CR, Braakhuis BJ, Brakenhoff RH (2011) The molecular biology of head and neck cancer. Nat Rev Cancer 11(1):9–22CrossRefPubMed Leemans CR, Braakhuis BJ, Brakenhoff RH (2011) The molecular biology of head and neck cancer. Nat Rev Cancer 11(1):9–22CrossRefPubMed
3.
Zurück zum Zitat Ferlay J et al (2015) Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386CrossRefPubMed Ferlay J et al (2015) Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386CrossRefPubMed
4.
Zurück zum Zitat Whiteside TL (2018) Head and neck carcinoma immunotherapy: facts and hopes. Clin Cancer Res 24(1):6–13CrossRefPubMed Whiteside TL (2018) Head and neck carcinoma immunotherapy: facts and hopes. Clin Cancer Res 24(1):6–13CrossRefPubMed
5.
Zurück zum Zitat Melero I et al (2018) Introducing a new series: immunotherapy facts and hopes. Clin Cancer Res 24(8):1773–1774CrossRefPubMed Melero I et al (2018) Introducing a new series: immunotherapy facts and hopes. Clin Cancer Res 24(8):1773–1774CrossRefPubMed
6.
Zurück zum Zitat Economopoulou P et al (2016) The promise of immunotherapy in head and neck squamous cell carcinoma. Ann Oncol 27(9):1675–1685CrossRefPubMed Economopoulou P et al (2016) The promise of immunotherapy in head and neck squamous cell carcinoma. Ann Oncol 27(9):1675–1685CrossRefPubMed
7.
Zurück zum Zitat Cekic C et al (2014) Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res 74(24):7250–7259CrossRefPubMedPubMedCentral Cekic C et al (2014) Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res 74(24):7250–7259CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Mandapathil M et al (2010) Adenosine and prostaglandin E2 cooperate in the suppression of immune responses mediated by adaptive regulatory T cells. J Biol Chem 285(36):27571–27580CrossRefPubMedPubMedCentral Mandapathil M et al (2010) Adenosine and prostaglandin E2 cooperate in the suppression of immune responses mediated by adaptive regulatory T cells. J Biol Chem 285(36):27571–27580CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Di Virgilio F, Adinolfi E (2017) Extracellular purines, purinergic receptors and tumor growth. Oncogene 36(3):293–303CrossRefPubMed Di Virgilio F, Adinolfi E (2017) Extracellular purines, purinergic receptors and tumor growth. Oncogene 36(3):293–303CrossRefPubMed
11.
Zurück zum Zitat Mediavilla-Varela M et al (2017) A novel antagonist of the immune checkpoint protein adenosine A2a receptor restores tumor-infiltrating lymphocyte activity in the context of the tumor microenvironment. Neoplasia 19(7):530–536CrossRefPubMedPubMedCentral Mediavilla-Varela M et al (2017) A novel antagonist of the immune checkpoint protein adenosine A2a receptor restores tumor-infiltrating lymphocyte activity in the context of the tumor microenvironment. Neoplasia 19(7):530–536CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Goetzl EJ, An S, Zeng L (1995) Specific suppression by prostaglandin E2 of activation-induced apoptosis of human CD4+CD8+ T lymphoblasts. J Immunol 154(3):1041–1047PubMed Goetzl EJ, An S, Zeng L (1995) Specific suppression by prostaglandin E2 of activation-induced apoptosis of human CD4+CD8+ T lymphoblasts. J Immunol 154(3):1041–1047PubMed
15.
Zurück zum Zitat Hutchinson L (2015) Immunotherapy: evading immune escape: synergy of COX and immune-checkpoint inhibitors. Nat Rev Clin Oncol 12(11):622PubMed Hutchinson L (2015) Immunotherapy: evading immune escape: synergy of COX and immune-checkpoint inhibitors. Nat Rev Clin Oncol 12(11):622PubMed
16.
Zurück zum Zitat Beavis PA et al (2015) Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced antitumor T-cell responses. Cancer Immunol Res 3(5):506–517CrossRefPubMed Beavis PA et al (2015) Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced antitumor T-cell responses. Cancer Immunol Res 3(5):506–517CrossRefPubMed
18.
Zurück zum Zitat Zhou G, Drake CG, Levitsky HI (2006) Amplification of tumor-specific regulatory T cells following therapeutic cancer vaccines. Blood 107(2):628–636CrossRefPubMedPubMedCentral Zhou G, Drake CG, Levitsky HI (2006) Amplification of tumor-specific regulatory T cells following therapeutic cancer vaccines. Blood 107(2):628–636CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Liu Z et al (2017) Intratumoral delivery of tumor antigen-loaded DC and tumor-primed CD4(+) T cells combined with agonist alpha-GITR mAb promotes durable CD8(+) T-cell-dependent antitumor immunity. Oncoimmunology 6(6):e1315487CrossRefPubMedPubMedCentral Liu Z et al (2017) Intratumoral delivery of tumor antigen-loaded DC and tumor-primed CD4(+) T cells combined with agonist alpha-GITR mAb promotes durable CD8(+) T-cell-dependent antitumor immunity. Oncoimmunology 6(6):e1315487CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Arab S et al (2017) Increased efficacy of a dendritic cell-based therapeutic cancer vaccine with adenosine receptor antagonist and CD73 inhibitor. Tumour Biol 39(3):1010428317695021CrossRefPubMed Arab S et al (2017) Increased efficacy of a dendritic cell-based therapeutic cancer vaccine with adenosine receptor antagonist and CD73 inhibitor. Tumour Biol 39(3):1010428317695021CrossRefPubMed
22.
Zurück zum Zitat Leone RD, Lo YC, Powell JD (2015) A2aR antagonists: next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J 13:265–272CrossRefPubMedPubMedCentral Leone RD, Lo YC, Powell JD (2015) A2aR antagonists: next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J 13:265–272CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Hawkins BL et al (1994) 4NQO carcinogenesis: a mouse model of oral cavity squamous cell carcinoma. Head Neck 16(5):424–432CrossRefPubMed Hawkins BL et al (1994) 4NQO carcinogenesis: a mouse model of oral cavity squamous cell carcinoma. Head Neck 16(5):424–432CrossRefPubMed
25.
Zurück zum Zitat Yang Z et al (2013) Comparable molecular alterations in 4-nitroquinoline 1-oxide-induced oral and esophageal cancer in mice and in human esophageal cancer, associated with poor prognosis of patients. Vivo 27(4):473–484 Yang Z et al (2013) Comparable molecular alterations in 4-nitroquinoline 1-oxide-induced oral and esophageal cancer in mice and in human esophageal cancer, associated with poor prognosis of patients. Vivo 27(4):473–484
26.
Zurück zum Zitat Tomayko MM, Reynolds CP (1989) Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol 24(3):148–154CrossRefPubMed Tomayko MM, Reynolds CP (1989) Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol 24(3):148–154CrossRefPubMed
27.
Zurück zum Zitat Qu Y et al (2010) Intralesional delivery of dendritic cells engineered to express T-bet promotes protective type 1 immunity and the normalization of the tumor microenvironment. J Immunol 185(5):2895–2902CrossRefPubMed Qu Y et al (2010) Intralesional delivery of dendritic cells engineered to express T-bet promotes protective type 1 immunity and the normalization of the tumor microenvironment. J Immunol 185(5):2895–2902CrossRefPubMed
29.
Zurück zum Zitat Son YI et al (2002) A novel bulk-culture method for generating mature dendritic cells from mouse bone marrow cells. J Immunol Methods 262(1–2):145–157CrossRefPubMed Son YI et al (2002) A novel bulk-culture method for generating mature dendritic cells from mouse bone marrow cells. J Immunol Methods 262(1–2):145–157CrossRefPubMed
30.
Zurück zum Zitat Zhao X et al (2012) Vaccines targeting tumor blood vessel antigens promote CD8(+) T cell-dependent tumor eradication or dormancy in HLA-A2 transgenic mice. J Immunol 188(4):1782–1788CrossRefPubMed Zhao X et al (2012) Vaccines targeting tumor blood vessel antigens promote CD8(+) T cell-dependent tumor eradication or dormancy in HLA-A2 transgenic mice. J Immunol 188(4):1782–1788CrossRefPubMed
31.
Zurück zum Zitat Liu Q et al (2018) BRAF peptide vaccine facilitates therapy of murine BRAF-mutant melanoma. Cancer Immunol Immunother 67(2):299–310CrossRefPubMed Liu Q et al (2018) BRAF peptide vaccine facilitates therapy of murine BRAF-mutant melanoma. Cancer Immunol Immunother 67(2):299–310CrossRefPubMed
32.
Zurück zum Zitat Hatano M et al (2004) Vaccination with EphA2-derived T cell-epitopes promotes immunity against both EphA2-expressing and EphA2-negative tumors. J Transl Med 2(1):40CrossRefPubMedPubMedCentral Hatano M et al (2004) Vaccination with EphA2-derived T cell-epitopes promotes immunity against both EphA2-expressing and EphA2-negative tumors. J Transl Med 2(1):40CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Umano Y et al (2001) Generation of cytotoxic T cell responses to an HLA-A24 restricted epitope peptide derived from wild-type p53. Br J Cancer 84(8):1052–1057CrossRefPubMedPubMedCentral Umano Y et al (2001) Generation of cytotoxic T cell responses to an HLA-A24 restricted epitope peptide derived from wild-type p53. Br J Cancer 84(8):1052–1057CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Lacabanne V et al (1996) A wild-type p53 cytotoxic T cell epitope is presented by mouse hepatocarcinoma cells. Eur J Immunol 26(11):2635–2639CrossRefPubMed Lacabanne V et al (1996) A wild-type p53 cytotoxic T cell epitope is presented by mouse hepatocarcinoma cells. Eur J Immunol 26(11):2635–2639CrossRefPubMed
35.
Zurück zum Zitat Hofmann UB et al (2009) Identification and characterization of survivin-derived H-2Kb-restricted CTL epitopes. Eur J Immunol 39(5):1419–1424CrossRefPubMed Hofmann UB et al (2009) Identification and characterization of survivin-derived H-2Kb-restricted CTL epitopes. Eur J Immunol 39(5):1419–1424CrossRefPubMed
36.
Zurück zum Zitat Dong Y et al (2006) Identification of H-2Db-specific CD8 + T-cell epitopes from mouse VEGFR2 that can inhibit angiogenesis and tumor growth. J Immunother 29(1):32–40CrossRefPubMed Dong Y et al (2006) Identification of H-2Db-specific CD8 + T-cell epitopes from mouse VEGFR2 that can inhibit angiogenesis and tumor growth. J Immunother 29(1):32–40CrossRefPubMed
37.
Zurück zum Zitat Waickman AT et al (2012) Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol Immunother 61(6):917–926CrossRefPubMed Waickman AT et al (2012) Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol Immunother 61(6):917–926CrossRefPubMed
38.
Zurück zum Zitat Fabian KP et al (2017) Therapeutic efficacy of combined vaccination against tumor pericyte-associated antigens DLK1 and DLK2 in mice. Oncoimmunology 6(3):e1290035CrossRefPubMedPubMedCentral Fabian KP et al (2017) Therapeutic efficacy of combined vaccination against tumor pericyte-associated antigens DLK1 and DLK2 in mice. Oncoimmunology 6(3):e1290035CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Theodoraki MN et al (2018) Clinical significance of PD-L1(+) exosomes in plasma of head and neck cancer patients. Clin Cancer Res 24(4):896–905CrossRefPubMed Theodoraki MN et al (2018) Clinical significance of PD-L1(+) exosomes in plasma of head and neck cancer patients. Clin Cancer Res 24(4):896–905CrossRefPubMed
40.
Zurück zum Zitat Chen WC et al (2017) Inflammation-induced myeloid-derived suppressor cells associated with squamous cell carcinoma of the head and neck. Head Neck 39(2):347–355CrossRefPubMed Chen WC et al (2017) Inflammation-induced myeloid-derived suppressor cells associated with squamous cell carcinoma of the head and neck. Head Neck 39(2):347–355CrossRefPubMed
41.
Zurück zum Zitat Chu M et al (2012) Myeloid-derived suppressor cells contribute to oral cancer progression in 4NQO-treated mice. Oral Dis 18(1):67–73CrossRefPubMed Chu M et al (2012) Myeloid-derived suppressor cells contribute to oral cancer progression in 4NQO-treated mice. Oral Dis 18(1):67–73CrossRefPubMed
42.
Zurück zum Zitat Fuse H et al (2016) Enhanced expression of PD-L1 in oral squamous cell carcinoma-derived CD11b(+)Gr-1(+) cells and its contribution to immunosuppressive activity. Oral Oncol 59:20–29CrossRefPubMed Fuse H et al (2016) Enhanced expression of PD-L1 in oral squamous cell carcinoma-derived CD11b(+)Gr-1(+) cells and its contribution to immunosuppressive activity. Oral Oncol 59:20–29CrossRefPubMed
43.
Zurück zum Zitat Schaefer C et al (2005) Characteristics of CD4 + CD25 + regulatory T cells in the peripheral circulation of patients with head and neck cancer. Br J Cancer 92(5):913–920CrossRefPubMedPubMedCentral Schaefer C et al (2005) Characteristics of CD4 + CD25 + regulatory T cells in the peripheral circulation of patients with head and neck cancer. Br J Cancer 92(5):913–920CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Mandapathil M et al (2009) Increased ectonucleotidase expression and activity in regulatory T cells of patients with head and neck cancer. Clin Cancer Res 15(20):6348–6357CrossRefPubMedPubMedCentral Mandapathil M et al (2009) Increased ectonucleotidase expression and activity in regulatory T cells of patients with head and neck cancer. Clin Cancer Res 15(20):6348–6357CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Jie HB et al (2013) Intratumoral regulatory T cells upregulate immunosuppressive molecules in head and neck cancer patients. Br J Cancer 109(10):2629–2635CrossRefPubMedPubMedCentral Jie HB et al (2013) Intratumoral regulatory T cells upregulate immunosuppressive molecules in head and neck cancer patients. Br J Cancer 109(10):2629–2635CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Theodoraki MN, Hoffmann TK, Whiteside TL (2018) Separation of plasma-derived exosomes into CD3((+)) and CD3((−)) fractions allows for association of immune cell and tumour cell markers with disease activity in HNSCC patients. Clin Exp Immunol 192(3):271–283CrossRefPubMed Theodoraki MN, Hoffmann TK, Whiteside TL (2018) Separation of plasma-derived exosomes into CD3((+)) and CD3((−)) fractions allows for association of immune cell and tumour cell markers with disease activity in HNSCC patients. Clin Exp Immunol 192(3):271–283CrossRefPubMed
47.
Zurück zum Zitat Liu JF et al (2018) Inhibition of JAK2/STAT3 reduces tumor-induced angiogenesis and myeloid-derived suppressor cells in head and neck cancer. Mol Carcinog 57(3):429–439CrossRefPubMed Liu JF et al (2018) Inhibition of JAK2/STAT3 reduces tumor-induced angiogenesis and myeloid-derived suppressor cells in head and neck cancer. Mol Carcinog 57(3):429–439CrossRefPubMed
49.
Zurück zum Zitat Schuler PJ et al (2014) Human CD4 + CD39 + regulatory T cells produce adenosine upon co-expression of surface CD73 or contact with CD73 + exosomes or CD73 + cells. Clin Exp Immunol 177(2):531–543CrossRefPubMedPubMedCentral Schuler PJ et al (2014) Human CD4 + CD39 + regulatory T cells produce adenosine upon co-expression of surface CD73 or contact with CD73 + exosomes or CD73 + cells. Clin Exp Immunol 177(2):531–543CrossRefPubMedPubMedCentral
Metadaten
Titel
Impact of combination immunochemotherapies on progression of 4NQO-induced murine oral squamous cell carcinoma
verfasst von
Sonja Ludwig
Chang-Sook Hong
Beatrice M. Razzo
Kellsye P. L. Fabian
Manoj Chelvanambi
Stephan Lang
Walter J. Storkus
Theresa L. Whiteside
Publikationsdatum
28.05.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Immunology, Immunotherapy / Ausgabe 7/2019
Print ISSN: 0340-7004
Elektronische ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-019-02348-2

Weitere Artikel der Ausgabe 7/2019

Cancer Immunology, Immunotherapy 7/2019 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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