Skip to main content
Top
Published in: Journal of Translational Medicine 1/2021

Open Access 01-12-2021 | Ionizing Radiation | Review

Immunobiology of cancer-associated fibroblasts in the context of radiotherapy

Authors: Turid Hellevik, Rodrigo Berzaghi, Kristin Lode, Ashraful Islam, Inigo Martinez-Zubiaurre

Published in: Journal of Translational Medicine | Issue 1/2021

Login to get access

Abstract

Radiotherapy (RT) still represents a mainstay of treatment in clinical oncology. Traditionally, the effectiveness of radiotherapy has been attributed to the killing potential of ionizing radiation (IR) over malignant cells, however, it has become clear that therapeutic efficacy of RT also involves activation of innate and adaptive anti-tumor immune responses. Therapeutic irradiation of the tumor microenvironment (TME) provokes profound cellular and biological reconfigurations which ultimately may influence immune recognition. As one of the major constituents of the TME, cancer-associated fibroblasts (CAFs) play central roles in cancer development at all stages and are recognized contributors of tumor immune evasion. While some studies argue that RT affects CAFs negatively through growth arrest and impaired motility, others claim that exposure of fibroblasts to RT promotes their conversion into a more activated phenotype. Nevertheless, despite the well-described immunoregulatory functions assigned to CAFs, little is known about the interplay between CAFs and immune cells in the context of RT. In this review, we go over current literature on the effects of radiation on CAFs and the influence that CAFs have on radiotherapy outcomes, and we summarize present knowledge on the transformed cellular crosstalk between CAFs and immune cells after radiation.
Literature
1.
go back to reference Chargari C, et al. Optimize and refine therapeutic index in radiation therapy: overview of a century. Cancer Treat Rev. 2016;45:58–67.PubMedCrossRef Chargari C, et al. Optimize and refine therapeutic index in radiation therapy: overview of a century. Cancer Treat Rev. 2016;45:58–67.PubMedCrossRef
2.
go back to reference Sharma RA, et al. Clinical development of new drug-radiotherapy combinations. Nat Rev Clin Oncol. 2016;13(10):627–42.PubMedCrossRef Sharma RA, et al. Clinical development of new drug-radiotherapy combinations. Nat Rev Clin Oncol. 2016;13(10):627–42.PubMedCrossRef
4.
5.
go back to reference Griffin RJ, et al. Understanding high-dose, ultra-high dose rate, and spatially fractionated radiation therapy. Int J Radiat Oncol Biol Phys. 2020;107(4):766–78.PubMedCrossRef Griffin RJ, et al. Understanding high-dose, ultra-high dose rate, and spatially fractionated radiation therapy. Int J Radiat Oncol Biol Phys. 2020;107(4):766–78.PubMedCrossRef
7.
10.
go back to reference Vanpouille-Box C, Formenti SC, Demaria S. Toward precision radiotherapy for use with immune checkpoint blockers. Clin Cancer Res. 2018;24(2):259–65.PubMedCrossRef Vanpouille-Box C, Formenti SC, Demaria S. Toward precision radiotherapy for use with immune checkpoint blockers. Clin Cancer Res. 2018;24(2):259–65.PubMedCrossRef
11.
go back to reference Arina A, Gutiontov SI, Weichselbaum RR. Radiotherapy and immunotherapy for cancer: from “systemic” to “multisite.” Clin Cancer Res. 2020;26(12):2777–82.PubMedCrossRef Arina A, Gutiontov SI, Weichselbaum RR. Radiotherapy and immunotherapy for cancer: from “systemic” to “multisite.” Clin Cancer Res. 2020;26(12):2777–82.PubMedCrossRef
13.
15.
go back to reference Rodriguez-Ruiz ME, et al. Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol. 2020;21(2):120–34.PubMedCrossRef Rodriguez-Ruiz ME, et al. Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol. 2020;21(2):120–34.PubMedCrossRef
17.
go back to reference Frey B, et al. Hypofractionated irradiation has immune stimulatory potential and induces a timely restricted infiltration of immune cells in colon cancer tumors. Front Immunol. 2017;8:231.PubMedPubMedCentralCrossRef Frey B, et al. Hypofractionated irradiation has immune stimulatory potential and induces a timely restricted infiltration of immune cells in colon cancer tumors. Front Immunol. 2017;8:231.PubMedPubMedCentralCrossRef
20.
go back to reference Dewan MZ, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009;15(17):5379–88.PubMedPubMedCentralCrossRef Dewan MZ, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009;15(17):5379–88.PubMedPubMedCentralCrossRef
21.
22.
go back to reference Gutiontov SI, et al. Cytoreduction and the optimization of immune checkpoint inhibition with radiation therapy. Int J Radiat Oncol Biol Phys. 2020;108(1):17–26.PubMedCrossRef Gutiontov SI, et al. Cytoreduction and the optimization of immune checkpoint inhibition with radiation therapy. Int J Radiat Oncol Biol Phys. 2020;108(1):17–26.PubMedCrossRef
23.
go back to reference Cremasco V, et al. FAP delineates heterogeneous and functionally divergent stromal cells in immune-excluded breast tumors. Cancer Immunol Res. 2018;6(12):1472–85.PubMedPubMedCentralCrossRef Cremasco V, et al. FAP delineates heterogeneous and functionally divergent stromal cells in immune-excluded breast tumors. Cancer Immunol Res. 2018;6(12):1472–85.PubMedPubMedCentralCrossRef
24.
25.
go back to reference Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18(2):99–115.PubMedCrossRef Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18(2):99–115.PubMedCrossRef
28.
go back to reference Erez N, et al. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010;17(2):135–47.PubMedCrossRef Erez N, et al. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010;17(2):135–47.PubMedCrossRef
30.
go back to reference Su S, et al. CD10(+)GPR77(+) cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell. 2018;172(4):841-856 e16.PubMedCrossRef Su S, et al. CD10(+)GPR77(+) cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell. 2018;172(4):841-856 e16.PubMedCrossRef
32.
33.
go back to reference Costa A, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33(3):463-479 e10.PubMedCrossRef Costa A, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33(3):463-479 e10.PubMedCrossRef
34.
go back to reference Huelsken J, Hanahan D. A subset of cancer-associated fibroblasts determines therapy resistance. Cell. 2018;172(4):643–4.PubMedCrossRef Huelsken J, Hanahan D. A subset of cancer-associated fibroblasts determines therapy resistance. Cell. 2018;172(4):643–4.PubMedCrossRef
35.
go back to reference Pereira BA, et al. CAF subpopulations: a new reservoir of stromal targets in pancreatic cancer. Trends Cancer. 2019;5(11):724–41.PubMedCrossRef Pereira BA, et al. CAF subpopulations: a new reservoir of stromal targets in pancreatic cancer. Trends Cancer. 2019;5(11):724–41.PubMedCrossRef
36.
go back to reference Bartoschek M, et al. Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat Commun. 2018;9(1):5150.PubMedPubMedCentralCrossRef Bartoschek M, et al. Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat Commun. 2018;9(1):5150.PubMedPubMedCentralCrossRef
37.
go back to reference Ozdemir BC, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719–34.PubMedPubMedCentralCrossRef Ozdemir BC, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719–34.PubMedPubMedCentralCrossRef
40.
go back to reference Kraman M, et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science. 2010;330(6005):827–30.PubMedCrossRef Kraman M, et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science. 2010;330(6005):827–30.PubMedCrossRef
41.
go back to reference Servais C, Erez N. From sentinel cells to inflammatory culprits: cancer-associated fibroblasts in tumour-related inflammation. J Pathol. 2013;229(2):198–207.PubMedCrossRef Servais C, Erez N. From sentinel cells to inflammatory culprits: cancer-associated fibroblasts in tumour-related inflammation. J Pathol. 2013;229(2):198–207.PubMedCrossRef
43.
go back to reference Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015;15(11):669–82.PubMedCrossRef Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015;15(11):669–82.PubMedCrossRef
44.
go back to reference Monteran L, Erez N. The dark side of fibroblasts: cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front Immunol. 2019;10:1835.PubMedPubMedCentralCrossRef Monteran L, Erez N. The dark side of fibroblasts: cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front Immunol. 2019;10:1835.PubMedPubMedCentralCrossRef
45.
go back to reference Elahi-Gedwillo KY, et al. Antifibrotic therapy disrupts stromal barriers and modulates the immune landscape in pancreatic ductal adenocarcinoma. Cancer Res. 2019;79(2):372–86.PubMedCrossRef Elahi-Gedwillo KY, et al. Antifibrotic therapy disrupts stromal barriers and modulates the immune landscape in pancreatic ductal adenocarcinoma. Cancer Res. 2019;79(2):372–86.PubMedCrossRef
46.
go back to reference Comito G, et al. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene. 2014;33(19):2423–31.PubMedCrossRef Comito G, et al. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene. 2014;33(19):2423–31.PubMedCrossRef
47.
go back to reference Takahashi H, et al. Cancer-associated fibroblasts promote an immunosuppressive microenvironment through the induction and accumulation of protumoral macrophages. Oncotarget. 2017;8(5):8633–47.PubMedCrossRef Takahashi H, et al. Cancer-associated fibroblasts promote an immunosuppressive microenvironment through the induction and accumulation of protumoral macrophages. Oncotarget. 2017;8(5):8633–47.PubMedCrossRef
48.
go back to reference Gok Yavuz B, et al. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1(+) TAMs. Sci Rep. 2019;9(1):3172.PubMedPubMedCentralCrossRef Gok Yavuz B, et al. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1(+) TAMs. Sci Rep. 2019;9(1):3172.PubMedPubMedCentralCrossRef
49.
go back to reference Orimo A, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48.PubMedCrossRef Orimo A, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48.PubMedCrossRef
50.
go back to reference Shani O, et al. Fibroblast-derived IL33 facilitates breast cancer metastasis by modifying the immune microenvironment and driving type 2 immunity. Cancer Res. 2020;80(23):5317–29.PubMedPubMedCentralCrossRef Shani O, et al. Fibroblast-derived IL33 facilitates breast cancer metastasis by modifying the immune microenvironment and driving type 2 immunity. Cancer Res. 2020;80(23):5317–29.PubMedPubMedCentralCrossRef
52.
go back to reference Cohen N, et al. Fibroblasts drive an immunosuppressive and growth-promoting microenvironment in breast cancer via secretion of Chitinase 3-like 1. Oncogene. 2017;36(31):4457–68.PubMedPubMedCentralCrossRef Cohen N, et al. Fibroblasts drive an immunosuppressive and growth-promoting microenvironment in breast cancer via secretion of Chitinase 3-like 1. Oncogene. 2017;36(31):4457–68.PubMedPubMedCentralCrossRef
54.
go back to reference Kumar V, et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell. 2017;32(5):654-668 e5.PubMedPubMedCentralCrossRef Kumar V, et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell. 2017;32(5):654-668 e5.PubMedPubMedCentralCrossRef
55.
go back to reference Cheng Y, et al. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 2018;9(4):422.PubMedPubMedCentralCrossRef Cheng Y, et al. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 2018;9(4):422.PubMedPubMedCentralCrossRef
56.
go back to reference Zhang J, et al. Tumor-educated neutrophils activate mesenchymal stem cells to promote gastric cancer growth and metastasis. Front Cell Dev Biol. 2020;8:788.PubMedPubMedCentralCrossRef Zhang J, et al. Tumor-educated neutrophils activate mesenchymal stem cells to promote gastric cancer growth and metastasis. Front Cell Dev Biol. 2020;8:788.PubMedPubMedCentralCrossRef
57.
go back to reference Cheng JT, et al. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis. 2016;5:e198.PubMedPubMedCentralCrossRef Cheng JT, et al. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis. 2016;5:e198.PubMedPubMedCentralCrossRef
58.
go back to reference Hsu YL, et al. Lung cancer-derived galectin-1 contributes to cancer associated fibroblast-mediated cancer progression and immune suppression through TDO2/kynurenine axis. Oncotarget. 2016;7(19):27584–98.PubMedPubMedCentralCrossRef Hsu YL, et al. Lung cancer-derived galectin-1 contributes to cancer associated fibroblast-mediated cancer progression and immune suppression through TDO2/kynurenine axis. Oncotarget. 2016;7(19):27584–98.PubMedPubMedCentralCrossRef
59.
go back to reference De Monte L, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208(3):469–78.PubMedPubMedCentralCrossRef De Monte L, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208(3):469–78.PubMedPubMedCentralCrossRef
60.
go back to reference Mace TA, et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 2013;73(10):3007–18.PubMedPubMedCentralCrossRef Mace TA, et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 2013;73(10):3007–18.PubMedPubMedCentralCrossRef
61.
go back to reference Deng Y, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2017;36(8):1090–101.PubMedCrossRef Deng Y, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2017;36(8):1090–101.PubMedCrossRef
62.
go back to reference Yang X, et al. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res. 2016;76(14):4124–35.PubMedCrossRef Yang X, et al. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res. 2016;76(14):4124–35.PubMedCrossRef
63.
go back to reference Huntington ND, Cursons J, Rautela J. The cancer-natural killer cell immunity cycle. Nat Rev Cancer. 2020;20(8):437–54.PubMedCrossRef Huntington ND, Cursons J, Rautela J. The cancer-natural killer cell immunity cycle. Nat Rev Cancer. 2020;20(8):437–54.PubMedCrossRef
64.
go back to reference Li T, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318(2):154–61.PubMedCrossRef Li T, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318(2):154–61.PubMedCrossRef
65.
go back to reference Li T, et al. Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med Oncol. 2013;30(3):663.PubMedCrossRef Li T, et al. Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med Oncol. 2013;30(3):663.PubMedCrossRef
66.
go back to reference Balsamo M, et al. Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc Natl Acad Sci USA. 2009;106(49):20847–52.PubMedPubMedCentralCrossRef Balsamo M, et al. Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc Natl Acad Sci USA. 2009;106(49):20847–52.PubMedPubMedCentralCrossRef
67.
go back to reference Ziani L, et al. Melanoma-associated fibroblasts decrease tumor cell susceptibility to NK cell-mediated killing through matrix-metalloproteinases secretion. Oncotarget. 2017;8(12):19780–94.PubMedPubMedCentralCrossRef Ziani L, et al. Melanoma-associated fibroblasts decrease tumor cell susceptibility to NK cell-mediated killing through matrix-metalloproteinases secretion. Oncotarget. 2017;8(12):19780–94.PubMedPubMedCentralCrossRef
68.
go back to reference Yang N, et al. Irradiated tumor fibroblasts avoid immune recognition and retain immunosuppressive functions over natural killer cells. Front Immunol. 2020;11:602530.PubMedCrossRef Yang N, et al. Irradiated tumor fibroblasts avoid immune recognition and retain immunosuppressive functions over natural killer cells. Front Immunol. 2020;11:602530.PubMedCrossRef
69.
70.
go back to reference Feig C, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 2013;110(50):20212–7.PubMedPubMedCentralCrossRef Feig C, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 2013;110(50):20212–7.PubMedPubMedCentralCrossRef
71.
go back to reference Curran TA, et al. IDO expressing fibroblasts promote the expansion of antigen specific regulatory T cells. Immunobiology. 2014;219(1):17–24.PubMedCrossRef Curran TA, et al. IDO expressing fibroblasts promote the expansion of antigen specific regulatory T cells. Immunobiology. 2014;219(1):17–24.PubMedCrossRef
72.
go back to reference Ino Y, et al. Arginase II expressed in cancer-associated fibroblasts indicates tissue hypoxia and predicts poor outcome in patients with pancreatic cancer. PLoS ONE. 2013;8(2):e55146.PubMedPubMedCentralCrossRef Ino Y, et al. Arginase II expressed in cancer-associated fibroblasts indicates tissue hypoxia and predicts poor outcome in patients with pancreatic cancer. PLoS ONE. 2013;8(2):e55146.PubMedPubMedCentralCrossRef
73.
go back to reference Sun K, et al. Oxidized ATM-mediated glycolysis enhancement in breast cancer-associated fibroblasts contributes to tumor invasion through lactate as metabolic coupling. EBioMedicine. 2019;41:370–83.PubMedPubMedCentralCrossRef Sun K, et al. Oxidized ATM-mediated glycolysis enhancement in breast cancer-associated fibroblasts contributes to tumor invasion through lactate as metabolic coupling. EBioMedicine. 2019;41:370–83.PubMedPubMedCentralCrossRef
74.
go back to reference Elyada E, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019;9(8):1102–23.PubMedPubMedCentralCrossRef Elyada E, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019;9(8):1102–23.PubMedPubMedCentralCrossRef
75.
go back to reference Liao D, et al. Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4T1 murine breast cancer model. PLoS ONE. 2009;4(11):e7965.PubMedPubMedCentralCrossRef Liao D, et al. Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4T1 murine breast cancer model. PLoS ONE. 2009;4(11):e7965.PubMedPubMedCentralCrossRef
76.
go back to reference Barnas JL, et al. Reciprocal functional modulation of the activation of T lymphocytes and fibroblasts derived from human solid tumors. J Immunol. 2010;185(5):2681–92.PubMedCrossRef Barnas JL, et al. Reciprocal functional modulation of the activation of T lymphocytes and fibroblasts derived from human solid tumors. J Immunol. 2010;185(5):2681–92.PubMedCrossRef
77.
go back to reference Pankova D, et al. Cancer-associated fibroblasts induce a collagen cross-link switch in tumor stroma. Mol Cancer Res. 2016;14(3):287–95.PubMedCrossRef Pankova D, et al. Cancer-associated fibroblasts induce a collagen cross-link switch in tumor stroma. Mol Cancer Res. 2016;14(3):287–95.PubMedCrossRef
80.
81.
go back to reference Salmon H, et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest. 2012;122(3):899–910.PubMedPubMedCentralCrossRef Salmon H, et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest. 2012;122(3):899–910.PubMedPubMedCentralCrossRef
84.
go back to reference Zaghdoudi S, et al. FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Mol Med. 2020;12(11):e12010.PubMedPubMedCentralCrossRef Zaghdoudi S, et al. FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Mol Med. 2020;12(11):e12010.PubMedPubMedCentralCrossRef
85.
86.
go back to reference Chen Y, et al. Type I collagen deletion in alphaSMA(+) myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell. 2021;39(4):548-565 e6.PubMedCrossRefPubMedCentral Chen Y, et al. Type I collagen deletion in alphaSMA(+) myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell. 2021;39(4):548-565 e6.PubMedCrossRefPubMedCentral
87.
go back to reference Jiang H, et al. Development of resistance to FAK inhibition in pancreatic cancer is linked to stromal depletion. Gut. 2020;69(1):122–32.PubMedCrossRef Jiang H, et al. Development of resistance to FAK inhibition in pancreatic cancer is linked to stromal depletion. Gut. 2020;69(1):122–32.PubMedCrossRef
88.
go back to reference Hayashi Y, et al. p53 functional deficiency in human colon cancer cells promotes fibroblast-mediated angiogenesis and tumor growth. Carcinogenesis. 2016;37(10):972–84.PubMedCrossRef Hayashi Y, et al. p53 functional deficiency in human colon cancer cells promotes fibroblast-mediated angiogenesis and tumor growth. Carcinogenesis. 2016;37(10):972–84.PubMedCrossRef
89.
go back to reference Bektas S, et al. CD24 and galectin-1 expressions in gastric adenocarcinoma and clinicopathologic significance. Pathol Oncol Res. 2010;16(4):569–77.PubMedCrossRef Bektas S, et al. CD24 and galectin-1 expressions in gastric adenocarcinoma and clinicopathologic significance. Pathol Oncol Res. 2010;16(4):569–77.PubMedCrossRef
90.
go back to reference Schoppmann SF, et al. Podoplanin expressing cancer associated fibroblasts are associated with unfavourable prognosis in adenocarcinoma of the esophagus. Clin Exp Metastasis. 2013;30(4):441–6.PubMedCrossRef Schoppmann SF, et al. Podoplanin expressing cancer associated fibroblasts are associated with unfavourable prognosis in adenocarcinoma of the esophagus. Clin Exp Metastasis. 2013;30(4):441–6.PubMedCrossRef
91.
go back to reference Yang J, et al. Vascular mimicry formation is promoted by paracrine TGF-beta and SDF1 of cancer-associated fibroblasts and inhibited by miR-101 in hepatocellular carcinoma. Cancer Lett. 2016;383(1):18–27.PubMedCrossRef Yang J, et al. Vascular mimicry formation is promoted by paracrine TGF-beta and SDF1 of cancer-associated fibroblasts and inhibited by miR-101 in hepatocellular carcinoma. Cancer Lett. 2016;383(1):18–27.PubMedCrossRef
92.
go back to reference Lederle W, et al. MMP13 as a stromal mediator in controlling persistent angiogenesis in skin carcinoma. Carcinogenesis. 2010;31(7):1175–84.PubMedCrossRef Lederle W, et al. MMP13 as a stromal mediator in controlling persistent angiogenesis in skin carcinoma. Carcinogenesis. 2010;31(7):1175–84.PubMedCrossRef
93.
go back to reference Noman MZ, et al. Hypoxia: a key player in antitumor immune response. A review in the theme: cellular responses to hypoxia. Am J Physiol Cell Physiol. 2015;309(9):C569-79.PubMedPubMedCentralCrossRef Noman MZ, et al. Hypoxia: a key player in antitumor immune response. A review in the theme: cellular responses to hypoxia. Am J Physiol Cell Physiol. 2015;309(9):C569-79.PubMedPubMedCentralCrossRef
94.
go back to reference Noman MZ, et al. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–90.PubMedPubMedCentralCrossRef Noman MZ, et al. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–90.PubMedPubMedCentralCrossRef
95.
go back to reference Barsoum IB, et al. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665–74.PubMedCrossRef Barsoum IB, et al. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665–74.PubMedCrossRef
97.
go back to reference Allard B, et al. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol. 2020;17(10):611–29.PubMedCrossRef Allard B, et al. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol. 2020;17(10):611–29.PubMedCrossRef
98.
go back to reference Hellevik T, et al. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat Oncol. 2012;7:59.PubMedPubMedCentralCrossRef Hellevik T, et al. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat Oncol. 2012;7:59.PubMedPubMedCentralCrossRef
99.
go back to reference Grinde MT, et al. Ionizing radiation abrogates the pro-tumorigenic capacity of cancer-associated fibroblasts co-implanted in xenografts. Sci Rep. 2017;7:46714.PubMedPubMedCentralCrossRef Grinde MT, et al. Ionizing radiation abrogates the pro-tumorigenic capacity of cancer-associated fibroblasts co-implanted in xenografts. Sci Rep. 2017;7:46714.PubMedPubMedCentralCrossRef
100.
go back to reference Papadopoulou A, Kletsas D. Human lung fibroblasts prematurely senescent after exposure to ionizing radiation enhance the growth of malignant lung epithelial cells in vitro and in vivo. Int J Oncol. 2011;39(4):989–99.PubMed Papadopoulou A, Kletsas D. Human lung fibroblasts prematurely senescent after exposure to ionizing radiation enhance the growth of malignant lung epithelial cells in vitro and in vivo. Int J Oncol. 2011;39(4):989–99.PubMed
101.
go back to reference Tommelein J, et al. Radiotherapy-activated cancer-associated fibroblasts promote tumor progression through paracrine IGF1R activation. Cancer Res. 2018;78(3):659–70.PubMedCrossRef Tommelein J, et al. Radiotherapy-activated cancer-associated fibroblasts promote tumor progression through paracrine IGF1R activation. Cancer Res. 2018;78(3):659–70.PubMedCrossRef
102.
go back to reference Rodningen OK, et al. Microarray analysis of the transcriptional response to single or multiple doses of ionizing radiation in human subcutaneous fibroblasts. Radiother Oncol. 2005;77(3):231–40.PubMedCrossRef Rodningen OK, et al. Microarray analysis of the transcriptional response to single or multiple doses of ionizing radiation in human subcutaneous fibroblasts. Radiother Oncol. 2005;77(3):231–40.PubMedCrossRef
103.
go back to reference Martinez-Zubiaurre I, et al. Tumorigenic responses of cancer-associated stromal fibrioblasts after ablative radiotherapy: a transcriptome-profiling study. J Cancer Ther. 2013;4(1):208–50.CrossRef Martinez-Zubiaurre I, et al. Tumorigenic responses of cancer-associated stromal fibrioblasts after ablative radiotherapy: a transcriptome-profiling study. J Cancer Ther. 2013;4(1):208–50.CrossRef
104.
go back to reference Hellevik T, et al. Changes in the secretory profile of NSCLC-associated fibroblasts after ablative radiotherapy: potential impact on angiogenesis and tumor growth. Transl Oncol. 2013;6(1):66–74.PubMedPubMedCentralCrossRef Hellevik T, et al. Changes in the secretory profile of NSCLC-associated fibroblasts after ablative radiotherapy: potential impact on angiogenesis and tumor growth. Transl Oncol. 2013;6(1):66–74.PubMedPubMedCentralCrossRef
106.
107.
go back to reference Kamochi N, et al. Irradiated fibroblast-induced bystander effects on invasive growth of squamous cell carcinoma under cancer-stromal cell interaction. Cancer Sci. 2008;99(12):2417–27.PubMedCrossRef Kamochi N, et al. Irradiated fibroblast-induced bystander effects on invasive growth of squamous cell carcinoma under cancer-stromal cell interaction. Cancer Sci. 2008;99(12):2417–27.PubMedCrossRef
108.
go back to reference Tsai KK, et al. Low-dose radiation-induced senescent stromal fibroblasts render nearby breast cancer cells radioresistant. Radiat Res. 2009;172(3):306–13.PubMedPubMedCentralCrossRef Tsai KK, et al. Low-dose radiation-induced senescent stromal fibroblasts render nearby breast cancer cells radioresistant. Radiat Res. 2009;172(3):306–13.PubMedPubMedCentralCrossRef
109.
go back to reference Patel ZS, et al. Ionizing radiation enhances esophageal epithelial cell migration and invasion through a paracrine mechanism involving stromal-derived hepatocyte growth factor. Radiat Res. 2012;177(2):200–8.PubMedCrossRef Patel ZS, et al. Ionizing radiation enhances esophageal epithelial cell migration and invasion through a paracrine mechanism involving stromal-derived hepatocyte growth factor. Radiat Res. 2012;177(2):200–8.PubMedCrossRef
110.
go back to reference Tsai KK, et al. Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Res. 2005;65(15):6734–44.PubMedCrossRef Tsai KK, et al. Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Res. 2005;65(15):6734–44.PubMedCrossRef
111.
go back to reference Barcellos-Hoff MH, Ravani SA. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 2000;60(5):1254–60.PubMed Barcellos-Hoff MH, Ravani SA. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 2000;60(5):1254–60.PubMed
113.
go back to reference Chu TY, et al. Crosstalk with cancer-associated fibroblasts increases the growth and radiation survival of cervical cancer cells. Radiat Res. 2014;181(5):540–7.PubMedCrossRef Chu TY, et al. Crosstalk with cancer-associated fibroblasts increases the growth and radiation survival of cervical cancer cells. Radiat Res. 2014;181(5):540–7.PubMedCrossRef
114.
go back to reference Zhang H, et al. CAF-secreted CXCL1 conferred radioresistance by regulating DNA damage response in a ROS-dependent manner in esophageal squamous cell carcinoma. Cell Death Dis. 2017;8(5):e2790.PubMedPubMedCentralCrossRef Zhang H, et al. CAF-secreted CXCL1 conferred radioresistance by regulating DNA damage response in a ROS-dependent manner in esophageal squamous cell carcinoma. Cell Death Dis. 2017;8(5):e2790.PubMedPubMedCentralCrossRef
116.
go back to reference Al-Assar O, et al. Contextual regulation of pancreatic cancer stem cell phenotype and radioresistance by pancreatic stellate cells. Radiother Oncol. 2014;111(2):243–51.PubMedCrossRef Al-Assar O, et al. Contextual regulation of pancreatic cancer stem cell phenotype and radioresistance by pancreatic stellate cells. Radiother Oncol. 2014;111(2):243–51.PubMedCrossRef
117.
go back to reference Bao CH, et al. Irradiated fibroblasts promote epithelial-mesenchymal transition and HDGF expression of esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 2015;458(2):441–7.PubMedCrossRef Bao CH, et al. Irradiated fibroblasts promote epithelial-mesenchymal transition and HDGF expression of esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 2015;458(2):441–7.PubMedCrossRef
118.
go back to reference Ohuchida K, et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 2004;64(9):3215–22.PubMedCrossRef Ohuchida K, et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 2004;64(9):3215–22.PubMedCrossRef
119.
go back to reference Li D, et al. Radiation promotes epithelial-to-mesenchymal transition and invasion of pancreatic cancer cell by activating carcinoma-associated fibroblasts. Am J Cancer Res. 2016;6(10):2192–206.PubMedPubMedCentral Li D, et al. Radiation promotes epithelial-to-mesenchymal transition and invasion of pancreatic cancer cell by activating carcinoma-associated fibroblasts. Am J Cancer Res. 2016;6(10):2192–206.PubMedPubMedCentral
120.
121.
go back to reference Arshad A, Deutsch E, Vozenin MC. Simultaneous irradiation of fibroblasts and carcinoma cells repress the secretion of soluble factors able to stimulate carcinoma cell migration. PLoS ONE. 2015;10(1):e0115447.PubMedPubMedCentralCrossRef Arshad A, Deutsch E, Vozenin MC. Simultaneous irradiation of fibroblasts and carcinoma cells repress the secretion of soluble factors able to stimulate carcinoma cell migration. PLoS ONE. 2015;10(1):e0115447.PubMedPubMedCentralCrossRef
122.
123.
go back to reference van Maaren MC, et al. 10 year survival after breast-conserving surgery plus radiotherapy compared with mastectomy in early breast cancer in the Netherlands: a population-based study. Lancet Oncol. 2016;17(8):1158–70.PubMedCrossRef van Maaren MC, et al. 10 year survival after breast-conserving surgery plus radiotherapy compared with mastectomy in early breast cancer in the Netherlands: a population-based study. Lancet Oncol. 2016;17(8):1158–70.PubMedCrossRef
124.
go back to reference Gorchs L, et al. Cancer-associated fibroblasts from lung tumors maintain their immunosuppressive abilities after high-dose irradiation. Front Oncol. 2015;5:87.PubMedPubMedCentralCrossRef Gorchs L, et al. Cancer-associated fibroblasts from lung tumors maintain their immunosuppressive abilities after high-dose irradiation. Front Oncol. 2015;5:87.PubMedPubMedCentralCrossRef
125.
go back to reference Berzaghi R, et al. Ionizing radiation curtails immunosuppressive effects from cancer-associated fibroblasts on dendritic cells. Front Immunol. 2021;12:662594.PubMedPubMedCentralCrossRef Berzaghi R, et al. Ionizing radiation curtails immunosuppressive effects from cancer-associated fibroblasts on dendritic cells. Front Immunol. 2021;12:662594.PubMedPubMedCentralCrossRef
126.
go back to reference Rubin P, et al. A perpetual cascade of cytokines postirradiation leads to pulmonary fibrosis. Int J Radiat Oncol Biol Phys. 1995;33(1):99–109.PubMedCrossRef Rubin P, et al. A perpetual cascade of cytokines postirradiation leads to pulmonary fibrosis. Int J Radiat Oncol Biol Phys. 1995;33(1):99–109.PubMedCrossRef
127.
go back to reference Finkelstein JN, et al. Early alterations in extracellular matrix and transforming growth factor beta gene expression in mouse lung indicative of late radiation fibrosis. Int J Radiat Oncol Biol Phys. 1994;28(3):621–31.PubMedCrossRef Finkelstein JN, et al. Early alterations in extracellular matrix and transforming growth factor beta gene expression in mouse lung indicative of late radiation fibrosis. Int J Radiat Oncol Biol Phys. 1994;28(3):621–31.PubMedCrossRef
128.
go back to reference Rube CE, et al. Dose-dependent induction of transforming growth factor beta (TGF-beta) in the lung tissue of fibrosis-prone mice after thoracic irradiation. Int J Radiat Oncol Biol Phys. 2000;47(4):1033–42.PubMedCrossRef Rube CE, et al. Dose-dependent induction of transforming growth factor beta (TGF-beta) in the lung tissue of fibrosis-prone mice after thoracic irradiation. Int J Radiat Oncol Biol Phys. 2000;47(4):1033–42.PubMedCrossRef
130.
go back to reference Mavrogonatou E, et al. Extracellular matrix alterations in senescent cells and their significance in tissue homeostasis. Matrix Biol. 2019;75–76:27–42.PubMedCrossRef Mavrogonatou E, et al. Extracellular matrix alterations in senescent cells and their significance in tissue homeostasis. Matrix Biol. 2019;75–76:27–42.PubMedCrossRef
Metadata
Title
Immunobiology of cancer-associated fibroblasts in the context of radiotherapy
Authors
Turid Hellevik
Rodrigo Berzaghi
Kristin Lode
Ashraful Islam
Inigo Martinez-Zubiaurre
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2021
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-021-03112-w

Other articles of this Issue 1/2021

Journal of Translational Medicine 1/2021 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.