Skip to main content
Top
Published in: Clinical and Translational Oncology 1/2021

Open Access 01-01-2021 | Zoledronic Acid | Research Article

Effect of Gemcitabine based chemotherapy on the immunogenicity of pancreatic tumour cells and T-cells

Authors: P. L. Smith, Y. Yogaratnam, M. Samad, S. Kasow, A. G. Dalgleish

Published in: Clinical and Translational Oncology | Issue 1/2021

Login to get access

Abstract

Purpose

Chemotherapy for advanced pancreatic cancer has limited efficacy due to the difficultly of treating established tumours and the evolution of tumour resistance. Chemotherapies for pancreatic cancer are typically studied for their cytotoxic properties rather than for their ability to increase the immunogenicity of pancreatic tumour cells. In this study Gemcitabine in combination with immune modulatory chemotherapies Oxaliplatin, zoledronic acid and pomalidomide was studied to determine how combination therapy alters the immunogenicity of pancreatic tumour cell lines and subsequent T-cell responses.

Methods

Pancreatic tumour cell lines were stimulated with the chemotherapeutic agents and markers of immune recognition were assessed. The effect of chemotherapeutic agents on DC function was measured using uptake of CFSE-stained PANC-1 cells, changes in markers of maturation and their ability to activate CD8+ T-cells. The effect of chemotherapeutic agents on T-cell priming prior to activation using anti-CD3 and anti-CD28 antibodies was determined by measuring IFN-γ expression and Annexin V staining using flow cytometry.

Results

These agents demonstrate both additive and inhibitory properties on a range of markers of immunogenicity. Gemcitabine was notable for its ability to induce the upregulation of human leukocyte antigen and checkpoints on pancreatic tumour cell lines whilst inhibiting T-cell activation. Pomalidomide demonstrated immune modulatory properties on dendritic cells and T-cells, even in the presence of gemcitabine.

Discussion

These data highlight the complex interactions of different agents in the modulation of tumour immunogenicity and immune cell activation and emphasise the complexity in rationally designing chemo immunogenic combinations for use with immunotherapy.
Literature
1.
go back to reference Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.CrossRef Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.CrossRef
2.
go back to reference Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.CrossRef Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.CrossRef
3.
go back to reference Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin. 2015;65:5–29.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin. 2015;65:5–29.CrossRef
4.
go back to reference Abrams TA, Meyer G, Meyerhardt JA, et al. (2017) Patterns of chemotherapy use in a US-based cohort of patients with metastatic pancreatic cancer. Oncologist. 2017;22(8):925–33.CrossRef Abrams TA, Meyer G, Meyerhardt JA, et al. (2017) Patterns of chemotherapy use in a US-based cohort of patients with metastatic pancreatic cancer. Oncologist. 2017;22(8):925–33.CrossRef
5.
go back to reference Smyth EN, Bapat B, Ball DE, André T, Kaye JA. Metastatic pancreatic adenocarcinoma treatment patterns, health care resource use, and outcomes in France and the United Kingdom between 2009 and 2012: a retrospective study. Clin Ther. 2015;37:1301–16.CrossRef Smyth EN, Bapat B, Ball DE, André T, Kaye JA. Metastatic pancreatic adenocarcinoma treatment patterns, health care resource use, and outcomes in France and the United Kingdom between 2009 and 2012: a retrospective study. Clin Ther. 2015;37:1301–16.CrossRef
6.
go back to reference Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarcinoma: a review of current treatment strategies and developing therapies. Cancer Sci. 2015;7(2):68–84. Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarcinoma: a review of current treatment strategies and developing therapies. Cancer Sci. 2015;7(2):68–84.
7.
go back to reference Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):165.CrossRef Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):165.CrossRef
8.
go back to reference Gong J, Hendifar A, Tuli R, et al. Combination systemic therapies with immune checkpoint inhibitors in pancreatic cancer: overcoming resistance to single-agent checkpoint blockade. Clin Transl Med. 2018;7(1):32.CrossRef Gong J, Hendifar A, Tuli R, et al. Combination systemic therapies with immune checkpoint inhibitors in pancreatic cancer: overcoming resistance to single-agent checkpoint blockade. Clin Transl Med. 2018;7(1):32.CrossRef
9.
go back to reference Lafaro KJ, Melstrom LG. The paradoxical web of pancreatic cancer tumor microenvironment. Am J Pathol. 2019;189(1):44–57.CrossRef Lafaro KJ, Melstrom LG. The paradoxical web of pancreatic cancer tumor microenvironment. Am J Pathol. 2019;189(1):44–57.CrossRef
10.
go back to reference Kamath SD, Kalyan A, Kircher S et al (2019) Ipilimumab and gemcitabine for advanced pancreatic cancer: a phase Ib study. Oncologist [published online ahead of print, 2019 Nov 18] Kamath SD, Kalyan A, Kircher S et al (2019) Ipilimumab and gemcitabine for advanced pancreatic cancer: a phase Ib study. Oncologist [published online ahead of print, 2019 Nov 18]
11.
go back to reference Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–65.CrossRef Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–65.CrossRef
12.
go back to reference Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33(8):828–33.CrossRef Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33(8):828–33.CrossRef
13.
go back to reference Aglietta M, Barone C, Sawyer MB, et al. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann Oncol. 2014;25(9):1750–5.CrossRef Aglietta M, Barone C, Sawyer MB, et al. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann Oncol. 2014;25(9):1750–5.CrossRef
14.
go back to reference Weiss GJ, Blaydorn L, Beck J, et al. Phase Ib/II study of gemcitabine, nab-paclitaxel, and pembrolizumab in metastatic pancreatic adenocarcinoma. Invest New Drugs. 2019;36(1):96–102.CrossRef Weiss GJ, Blaydorn L, Beck J, et al. Phase Ib/II study of gemcitabine, nab-paclitaxel, and pembrolizumab in metastatic pancreatic adenocarcinoma. Invest New Drugs. 2019;36(1):96–102.CrossRef
15.
go back to reference Wainberg ZA, Hochster HS, Kim EJ-H. Phase I study of nivolumab (Nivo) + nab-paclitaxel (nab-P) + gemcitabine (Gem) in advanced pancreatic cancer (APC). J Clin Oncol. 2019;37(4):298.CrossRef Wainberg ZA, Hochster HS, Kim EJ-H. Phase I study of nivolumab (Nivo) + nab-paclitaxel (nab-P) + gemcitabine (Gem) in advanced pancreatic cancer (APC). J Clin Oncol. 2019;37(4):298.CrossRef
16.
go back to reference Wang-Gillam A, Plambeck-Suess S, Goedegebuure P, et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Invest New Drugs. 2013;31(3):707–13.CrossRef Wang-Gillam A, Plambeck-Suess S, Goedegebuure P, et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Invest New Drugs. 2013;31(3):707–13.CrossRef
17.
go back to reference Gravett AM, Trautwein N, Stevanović S, Dalgleish AG, Copier J. GEM alters the proteasome composition and immunopeptidome of tumour cells. Oncoimmunology. 2018;7(6):e1438107.CrossRef Gravett AM, Trautwein N, Stevanović S, Dalgleish AG, Copier J. GEM alters the proteasome composition and immunopeptidome of tumour cells. Oncoimmunology. 2018;7(6):e1438107.CrossRef
18.
go back to reference Richardson PG, Mark TM, Lacy MQ. Pomalidomide: new immunomodulatory agent with potent antiproliferative effects. Crit Rev Oncol Hematol. 2013;88(Suppl 1):S36–S44.CrossRef Richardson PG, Mark TM, Lacy MQ. Pomalidomide: new immunomodulatory agent with potent antiproliferative effects. Crit Rev Oncol Hematol. 2013;88(Suppl 1):S36–S44.CrossRef
19.
go back to reference Corral LG, Haslett PA, Muller GW, et al. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-alpha. J Immunol. 1999;163(1):380–6.PubMed Corral LG, Haslett PA, Muller GW, et al. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-alpha. J Immunol. 1999;163(1):380–6.PubMed
20.
go back to reference Kalanxhi E, Meltzer S, Schou JV, et al. Systemic immune response induced by oxaliplatin-based neoadjuvant therapy favours survival without metastatic progression in high-risk rectal cancer. Br J Cancer. 2018;118(10):1322–8.CrossRef Kalanxhi E, Meltzer S, Schou JV, et al. Systemic immune response induced by oxaliplatin-based neoadjuvant therapy favours survival without metastatic progression in high-risk rectal cancer. Br J Cancer. 2018;118(10):1322–8.CrossRef
21.
go back to reference Liu H, Wang SH, Chen SC, Chen CY, Lo JL, Lin TM. Immune modulation of CD4+CD25+ regulatory T cells by ZAedronic acid. BMC Immunol. 2016;17(1):45.CrossRef Liu H, Wang SH, Chen SC, Chen CY, Lo JL, Lin TM. Immune modulation of CD4+CD25+ regulatory T cells by ZAedronic acid. BMC Immunol. 2016;17(1):45.CrossRef
22.
go back to reference Smith PL, Norgate KJ, Hegarty E, et al. Effect of the modification of p24 peptide antigen on dendritic cell uptake and T cell activation. Curr HIV Res. 2017;15(1):3–14.CrossRef Smith PL, Norgate KJ, Hegarty E, et al. Effect of the modification of p24 peptide antigen on dendritic cell uptake and T cell activation. Curr HIV Res. 2017;15(1):3–14.CrossRef
23.
go back to reference Fryer RA, Barlett B, Galustian C, Dalgleish AG. Mechanisms underlying gemcitabine resistance in pancreatic cancer and sensitisation by the iMiD™ lenalidomide. Anticancer Res. 2011;31(11):3747–56.PubMed Fryer RA, Barlett B, Galustian C, Dalgleish AG. Mechanisms underlying gemcitabine resistance in pancreatic cancer and sensitisation by the iMiD™ lenalidomide. Anticancer Res. 2011;31(11):3747–56.PubMed
24.
go back to reference Yamazaki T, Hannani D, Poirier-Colame V, et al. (2014) Defective immunogenic cell death of HMGB1-deficient tumors: compensatory therapy with TLR4 agonists. Cell Death Differ. 2019;21(1):69–78.CrossRef Yamazaki T, Hannani D, Poirier-Colame V, et al. (2014) Defective immunogenic cell death of HMGB1-deficient tumors: compensatory therapy with TLR4 agonists. Cell Death Differ. 2019;21(1):69–78.CrossRef
25.
go back to reference Zhang M, Yang J, Zhou J, et al. Prognostic values of CD38+CD101+PD1+CD8+ T cells in pancreatic cancer. Immunol Invest. 2019;48(5):466–79.CrossRef Zhang M, Yang J, Zhou J, et al. Prognostic values of CD38+CD101+PD1+CD8+ T cells in pancreatic cancer. Immunol Invest. 2019;48(5):466–79.CrossRef
26.
go back to reference Hudson WH, Gensheimer J, Hashimoto M, et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity. 2019;51(6):1043–1058.e4.CrossRef Hudson WH, Gensheimer J, Hashimoto M, et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity. 2019;51(6):1043–1058.e4.CrossRef
27.
go back to reference Verma V, Shrimali RK, Ahmad S, et al. PD-1 blockade in subprimed CD8 cells induces dysfunctional PD-1+CD38hi cells and anti-PD-1 resistance. Nat Immunol. 2019;20(9):1231–43.CrossRef Verma V, Shrimali RK, Ahmad S, et al. PD-1 blockade in subprimed CD8 cells induces dysfunctional PD-1+CD38hi cells and anti-PD-1 resistance. Nat Immunol. 2019;20(9):1231–43.CrossRef
28.
go back to reference Versteven M, Van den Bergh JMJ, Marcq E. Dendritic cells and programmed death-1 blockade: a joint venture to combat cancer. Front Immunol. 2018;1(9):394.CrossRef Versteven M, Van den Bergh JMJ, Marcq E. Dendritic cells and programmed death-1 blockade: a joint venture to combat cancer. Front Immunol. 2018;1(9):394.CrossRef
29.
go back to reference Kabacaoglu D, Ciecielski KJ, Ruess DA, et al. Immune checkpoint inhibition for pancreatic ductal adenocarcinoma: current limitations and future options. Front Immunol. 2018;9:1878.CrossRef Kabacaoglu D, Ciecielski KJ, Ruess DA, et al. Immune checkpoint inhibition for pancreatic ductal adenocarcinoma: current limitations and future options. Front Immunol. 2018;9:1878.CrossRef
30.
go back to reference Tesniere A, Schlemmer F, Boige V. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29(4):482–91.CrossRef Tesniere A, Schlemmer F, Boige V. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29(4):482–91.CrossRef
31.
go back to reference Matsumoto M, Takeda Y, Tatematsu M, Seya T. Toll-like receptor 3 signal in dendritic cells benefits cancer immunotherapy. Front Immunol. 2017;8:1897.CrossRef Matsumoto M, Takeda Y, Tatematsu M, Seya T. Toll-like receptor 3 signal in dendritic cells benefits cancer immunotherapy. Front Immunol. 2017;8:1897.CrossRef
32.
go back to reference Yamazaki T, Hannani D, Poirier-Colame V, et al. Defective immunogenic cell death of HMGB1-deficient tumors: compensatory therapy with TLR4 agonists. Cell Death Differ. 2014;21(1):69–78.CrossRef Yamazaki T, Hannani D, Poirier-Colame V, et al. Defective immunogenic cell death of HMGB1-deficient tumors: compensatory therapy with TLR4 agonists. Cell Death Differ. 2014;21(1):69–78.CrossRef
33.
go back to reference Fritz J, Karakhanova S, Brecht R, et al. In vitro immunomodulatory properties of gemcitabine alone and in combination with interferon-alpha. Immunol Lett. 2015;168(1):111–9.CrossRef Fritz J, Karakhanova S, Brecht R, et al. In vitro immunomodulatory properties of gemcitabine alone and in combination with interferon-alpha. Immunol Lett. 2015;168(1):111–9.CrossRef
34.
go back to reference Dalgleish AG, Stebbing J, Adamson DJ, et al. Randomised, open-label, phase II study of GEM with and without IMM-101 for advanced pancreatic cancer. Br J Cancer. 2016;115(7):789–96.CrossRef Dalgleish AG, Stebbing J, Adamson DJ, et al. Randomised, open-label, phase II study of GEM with and without IMM-101 for advanced pancreatic cancer. Br J Cancer. 2016;115(7):789–96.CrossRef
Metadata
Title
Effect of Gemcitabine based chemotherapy on the immunogenicity of pancreatic tumour cells and T-cells
Authors
P. L. Smith
Y. Yogaratnam
M. Samad
S. Kasow
A. G. Dalgleish
Publication date
01-01-2021
Publisher
Springer International Publishing
Published in
Clinical and Translational Oncology / Issue 1/2021
Print ISSN: 1699-048X
Electronic ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-020-02429-0

Other articles of this Issue 1/2021

Clinical and Translational Oncology 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine