Skip to main content
Top
Published in: Breast Cancer Research 1/2022

Open Access 01-12-2022 | Breast Cancer | Research

Steroid receptor coactivator-3 inhibition generates breast cancer antitumor immune microenvironment

Authors: Sang Jun Han, Nuri Sung, Jin Wang, Bert W. O’Malley, David M. Lonard

Published in: Breast Cancer Research | Issue 1/2022

Login to get access

Abstract

Background

The tumor immune microenvironment (TIME) generated by cancer-infiltrating immune cells has a crucial role in promoting or suppressing breast cancer progression. However, whether the steroid receptor coactivator-3 (SRC-3) modulates TIME to progress breast cancer is unclear. Therefore, the present study evaluates whether SRC-3 generates a tumor-promoting TIME in breast tumors using a syngeneic immune-intact mouse model of breast cancer.

Methods

We employed E0771 and 4T1 breast cancer in immune-intact syngeneic female C57BL/6 and BALB/c mice, respectively. SI-2, a specific small-molecule inhibitor of SRC-3, was administered daily (2.5 mg/kg) to E0771 and 4T1 breast tumor-bearing immune-intact mice. In addition, SRC-3 knockdown (KD)-E0771 and SRC-3 KD-4T1 cells and their parental breast cancer cells were injected into their syngeneic immune-intact female mice versus immune-deficiency mice to validate that the host immune system is required for breast tumor suppression by SRC-3 KD in immune-intact mice. Furthermore, tumor-infiltrating immune cells (such as CD4+, CD8+, CD56+, and Foxp3+ cells) in E0771 and 4T1 breast cancers treated with SI-2 and in SRC-3 KD E0771 and 4T1 breast cancers were determined by immunohistochemistry. Additionally, cytokine levels in SI-2-treated and SRC-3 KD E0771 breast tumors and their control cancers were defined with a Mouse Cytokine Array.

Results

SRC-3 inhibition by SI-2 significantly suppressed the progression of breast cancer cells (E0771 and 4T1) into breast cancers in immune-intact syngeneic female mice. SRC-3 KD-E0771 and -4T1 breast cancer cells did not produce well-developed tumors in immune-intact syngeneic female mice compared to their parental cells, but SRC-3 KD breast cancers were well developed in immune-defective host mice. SRC-3 inhibition by SI-2 and SRC-3 KD effectively increased the numbers of cytotoxic immune cells, such as CD4+ and CD8+ T cells and CD56+ NK cells, and Interferon γ (Ifng) in breast cancers compared to vehicle. However, SI-2 treatment reduced the number of tumor-infiltrating CD4+/Foxp3+ regulatory T (Treg) cells compared to vehicle treatment. In addition, SRC-3 inhibition by SI-2 and SRC-3 KD increased C-X-C motif chemokine ligand 9 (Cxcl9) expression in breast cancer to recruit C-X-C motif chemokine receptor 3 (Cxcr3)-expressing cytotoxic immune cells into breast tumors.

Conclusions

SRC-3 is a critical immunomodulator in breast cancer, generating a protumor immune microenvironment. SRC-3 inhibition by SI-2 or SRC-3 KD activates the Cxcl9/Cxcr3 axis in breast tumors and enhances the antitumor immune microenvironment to suppress breast cancer progression.
Appendix
Available only for authorised users
Literature
1.
go back to reference Dasgupta S, Lonard DM, O’Malley BW. Nuclear receptor coactivators: master regulators of human health and disease. Annu Rev Med. 2014;65:279–92.PubMedCrossRef Dasgupta S, Lonard DM, O’Malley BW. Nuclear receptor coactivators: master regulators of human health and disease. Annu Rev Med. 2014;65:279–92.PubMedCrossRef
3.
go back to reference Lonard DM, O’Malley BW. Molecular pathways: targeting steroid receptor coactivators in cancer. Clin Cancer. 2016;22(22):5403–7.CrossRef Lonard DM, O’Malley BW. Molecular pathways: targeting steroid receptor coactivators in cancer. Clin Cancer. 2016;22(22):5403–7.CrossRef
4.
go back to reference Anzick SL, Kononen J, Walker RL, Azorsa DO, Tanner MM, Guan XY, Sauter G, Kallioniemi OP, Trent JM, Meltzer PS. AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science (New York, NY). 1997;277(5328):965–8.CrossRef Anzick SL, Kononen J, Walker RL, Azorsa DO, Tanner MM, Guan XY, Sauter G, Kallioniemi OP, Trent JM, Meltzer PS. AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science (New York, NY). 1997;277(5328):965–8.CrossRef
5.
go back to reference Bautista S, Vallès H, Walker RL, Anzick S, Zeillinger R, Meltzer P, Theillet C. In breast cancer, amplification of the steroid receptor coactivator gene AIB1 is correlated with estrogen and progesterone receptor positivity. Clin Cancer Res. 1998;4(12):2925–9.PubMed Bautista S, Vallès H, Walker RL, Anzick S, Zeillinger R, Meltzer P, Theillet C. In breast cancer, amplification of the steroid receptor coactivator gene AIB1 is correlated with estrogen and progesterone receptor positivity. Clin Cancer Res. 1998;4(12):2925–9.PubMed
6.
go back to reference Bouras T, Southey MC, Venter DJ. Overexpression of the steroid receptor coactivator AIB1 in breast cancer correlates with the absence of estrogen and progesterone receptors and positivity for p53 and HER2/neu. Can Res. 2001;61(3):903–7. Bouras T, Southey MC, Venter DJ. Overexpression of the steroid receptor coactivator AIB1 in breast cancer correlates with the absence of estrogen and progesterone receptors and positivity for p53 and HER2/neu. Can Res. 2001;61(3):903–7.
7.
go back to reference Glaeser M, Floetotto T, Hanstein B, Beckmann MW, Niederacher D. Gene amplification and expression of the steroid receptor coactivator SRC3 (AIB1) in sporadic breast and endometrial carcinomas. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme 2001; 33(3):121–6. Glaeser M, Floetotto T, Hanstein B, Beckmann MW, Niederacher D. Gene amplification and expression of the steroid receptor coactivator SRC3 (AIB1) in sporadic breast and endometrial carcinomas. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme 2001; 33(3):121–6.
8.
go back to reference Hudelist G, Czerwenka K, Kubista E, Marton E, Pischinger K, Singer CF. Expression of sex steroid receptors and their co-factors in normal and malignant breast tissue: AIB1 is a carcinoma-specific co-activator. Breast Cancer Res Treat. 2003;78(2):193–204.PubMedCrossRef Hudelist G, Czerwenka K, Kubista E, Marton E, Pischinger K, Singer CF. Expression of sex steroid receptors and their co-factors in normal and malignant breast tissue: AIB1 is a carcinoma-specific co-activator. Breast Cancer Res Treat. 2003;78(2):193–204.PubMedCrossRef
9.
go back to reference Zhao C, Yasui K, Lee CJ, Kurioka H, Hosokawa Y, Oka T, Inazawa J. Elevated expression levels of NCOA3, TOP1, and TFAP2C in breast tumors as predictors of poor prognosis. Cancer. 2003;98(1):18–23.PubMedCrossRef Zhao C, Yasui K, Lee CJ, Kurioka H, Hosokawa Y, Oka T, Inazawa J. Elevated expression levels of NCOA3, TOP1, and TFAP2C in breast tumors as predictors of poor prognosis. Cancer. 2003;98(1):18–23.PubMedCrossRef
10.
go back to reference Wang L, Lonard DM, O’Malley BW. The role of steroid receptor coactivators in hormone dependent cancers and their potential as therapeutic targets. Hormones Cancer. 2016;7(4):229–35.PubMedPubMedCentralCrossRef Wang L, Lonard DM, O’Malley BW. The role of steroid receptor coactivators in hormone dependent cancers and their potential as therapeutic targets. Hormones Cancer. 2016;7(4):229–35.PubMedPubMedCentralCrossRef
11.
go back to reference Song X, Chen J, Zhao M, Zhang C, Yu Y, Lonard DM, Chow DC, Palzkill T, Xu J, O’Malley BW, et al. Development of potent small-molecule inhibitors to drug the undruggable steroid receptor coactivator-3. Proc Natl Acad Sci USA. 2016;113(18):4970–5.PubMedPubMedCentralCrossRef Song X, Chen J, Zhao M, Zhang C, Yu Y, Lonard DM, Chow DC, Palzkill T, Xu J, O’Malley BW, et al. Development of potent small-molecule inhibitors to drug the undruggable steroid receptor coactivator-3. Proc Natl Acad Sci USA. 2016;113(18):4970–5.PubMedPubMedCentralCrossRef
12.
go back to reference Gonzalez H, Hagerling C, Werb Z. Roles of the immune system in cancer: from tumor initiation to metastatic progression. Genes Dev. 2018;32(19–20):1267–84.PubMedPubMedCentralCrossRef Gonzalez H, Hagerling C, Werb Z. Roles of the immune system in cancer: from tumor initiation to metastatic progression. Genes Dev. 2018;32(19–20):1267–84.PubMedPubMedCentralCrossRef
13.
go back to reference Zhang Q, Qin J, Zhong L, Gong L, Zhang B, Zhang Y, Gao WQ. CCL5-mediated Th2 immune polarization promotes metastasis in luminal breast cancer. Can Res. 2015;75(20):4312–21.CrossRef Zhang Q, Qin J, Zhong L, Gong L, Zhang B, Zhang Y, Gao WQ. CCL5-mediated Th2 immune polarization promotes metastasis in luminal breast cancer. Can Res. 2015;75(20):4312–21.CrossRef
14.
go back to reference Gobert M, Treilleux I, Bendriss-Vermare N, Bachelot T, Goddard-Leon S, Arfi V, Biota C, Doffin AC, Durand I, Olive D, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Can Res. 2009;69(5):2000–9.CrossRef Gobert M, Treilleux I, Bendriss-Vermare N, Bachelot T, Goddard-Leon S, Arfi V, Biota C, Doffin AC, Durand I, Olive D, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Can Res. 2009;69(5):2000–9.CrossRef
15.
go back to reference Hojo S, Koizumi K, Tsuneyama K, Arita Y, Cui Z, Shinohara K, Minami T, Hashimoto I, Nakayama T, Sakurai H, et al. High-level expression of chemokine CXCL16 by tumor cells correlates with a good prognosis and increased tumor-infiltrating lymphocytes in colorectal cancer. Can Res. 2007;67(10):4725–31.CrossRef Hojo S, Koizumi K, Tsuneyama K, Arita Y, Cui Z, Shinohara K, Minami T, Hashimoto I, Nakayama T, Sakurai H, et al. High-level expression of chemokine CXCL16 by tumor cells correlates with a good prognosis and increased tumor-infiltrating lymphocytes in colorectal cancer. Can Res. 2007;67(10):4725–31.CrossRef
16.
go back to reference Nikolai BC, Jain P, Cardenas DL, York B, Feng Q, McKenna NJ, Dasgupta S, Lonard DM, O’Malley BW. Steroid receptor coactivator 3 (SRC-3/AIB1) is enriched and functional in mouse and human Tregs. Sci Rep. 2021;11(1):3441.PubMedPubMedCentralCrossRef Nikolai BC, Jain P, Cardenas DL, York B, Feng Q, McKenna NJ, Dasgupta S, Lonard DM, O’Malley BW. Steroid receptor coactivator 3 (SRC-3/AIB1) is enriched and functional in mouse and human Tregs. Sci Rep. 2021;11(1):3441.PubMedPubMedCentralCrossRef
17.
go back to reference Zhang GL, Zhang Y, Cao KX, Wang XM. Orthotopic injection of breast cancer cells into the mice mammary fat pad. J. Visualized Exp.: JoVE 2019(143). Zhang GL, Zhang Y, Cao KX, Wang XM. Orthotopic injection of breast cancer cells into the mice mammary fat pad. J. Visualized Exp.: JoVE 2019(143).
18.
go back to reference Sugiura K, Stock CC. Studies in a tumor spectrum. I. Comparison of the action of methylbis (2-chloroethyl)amine and 3-bis(2-chloroethyl)aminomethyl-4-methoxymethyl -5-hydroxy-6-methylpyridine on the growth of a variety of mouse and rat tumors. Cancer 1952; 5(2):382–402. Sugiura K, Stock CC. Studies in a tumor spectrum. I. Comparison of the action of methylbis (2-chloroethyl)amine and 3-bis(2-chloroethyl)aminomethyl-4-methoxymethyl -5-hydroxy-6-methylpyridine on the growth of a variety of mouse and rat tumors. Cancer 1952; 5(2):382–402.
19.
go back to reference Johnson CH, Fisher TS, Hoang LT, Felding BH, Siuzdak G, O’Brien PJ. Luciferase does not alter metabolism in cancer cells. Metabolomics. 2014;10(3):354–60.PubMedPubMedCentralCrossRef Johnson CH, Fisher TS, Hoang LT, Felding BH, Siuzdak G, O’Brien PJ. Luciferase does not alter metabolism in cancer cells. Metabolomics. 2014;10(3):354–60.PubMedPubMedCentralCrossRef
20.
go back to reference Ewens A, Mihich E, Ehrke MJ. Distant metastasis from subcutaneously grown E0771 medullary breast adenocarcinoma. Anticancer Res. 2005;25(6b):3905–15.PubMed Ewens A, Mihich E, Ehrke MJ. Distant metastasis from subcutaneously grown E0771 medullary breast adenocarcinoma. Anticancer Res. 2005;25(6b):3905–15.PubMed
21.
go back to reference Popel AS. Immunoactivating the tumor microenvironment enhances immunotherapy as predicted by integrative computational model. Proc Natl Acad Sci USA. 2020;117(9):4447–9.PubMedPubMedCentralCrossRef Popel AS. Immunoactivating the tumor microenvironment enhances immunotherapy as predicted by integrative computational model. Proc Natl Acad Sci USA. 2020;117(9):4447–9.PubMedPubMedCentralCrossRef
22.
go back to reference Haabeth OA, Tveita AA, Fauskanger M, Schjesvold F, Lorvik KB, Hofgaard PO, Omholt H, Munthe LA, Dembic Z, Corthay A, et al. How do CD4(+) T cells detect and eliminate tumor cells that either lack or express MHC class II molecules? Front Immunol. 2014;5:174.PubMedPubMedCentralCrossRef Haabeth OA, Tveita AA, Fauskanger M, Schjesvold F, Lorvik KB, Hofgaard PO, Omholt H, Munthe LA, Dembic Z, Corthay A, et al. How do CD4(+) T cells detect and eliminate tumor cells that either lack or express MHC class II molecules? Front Immunol. 2014;5:174.PubMedPubMedCentralCrossRef
23.
go back to reference Farhood B, Najafi M, Mortezaee K. CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2019;234(6):8509–21.PubMedCrossRef Farhood B, Najafi M, Mortezaee K. CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2019;234(6):8509–21.PubMedCrossRef
24.
25.
go back to reference Van Acker HH, Van Acker ZP, Versteven M, Ponsaerts P, Pende D, Berneman ZN, Anguille S, Van Tendeloo VF, Smits EL. CD56 homodimerization and participation in anti-tumor immune effector cell functioning: a role for interleukin-15. Cancers 2019; 11(7). Van Acker HH, Van Acker ZP, Versteven M, Ponsaerts P, Pende D, Berneman ZN, Anguille S, Van Tendeloo VF, Smits EL. CD56 homodimerization and participation in anti-tumor immune effector cell functioning: a role for interleukin-15. Cancers 2019; 11(7).
26.
go back to reference Bhat P, Leggatt G, Waterhouse N, Frazer IH. Interferon-γ derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis. 2017;8(6):e2836–e2836.PubMedPubMedCentralCrossRef Bhat P, Leggatt G, Waterhouse N, Frazer IH. Interferon-γ derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis. 2017;8(6):e2836–e2836.PubMedPubMedCentralCrossRef
27.
go back to reference Hill JA, Feuerer M, Tash K, Haxhinasto S, Perez J, Melamed R, Mathis D, Benoist C. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity. 2007;27(5):786–800.PubMedCrossRef Hill JA, Feuerer M, Tash K, Haxhinasto S, Perez J, Melamed R, Mathis D, Benoist C. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity. 2007;27(5):786–800.PubMedCrossRef
28.
go back to reference Takenaka M, Seki N, Toh U, Hattori S, Kawahara A, Yamaguchi T, Koura K, Takahashi R, Otsuka H, Takahashi H, et al. FOXP3 expression in tumor cells and tumor-infiltrating lymphocytes is associated with breast cancer prognosis. Mol Clin Oncol. 2013;1(4):625–32.PubMedPubMedCentralCrossRef Takenaka M, Seki N, Toh U, Hattori S, Kawahara A, Yamaguchi T, Koura K, Takahashi R, Otsuka H, Takahashi H, et al. FOXP3 expression in tumor cells and tumor-infiltrating lymphocytes is associated with breast cancer prognosis. Mol Clin Oncol. 2013;1(4):625–32.PubMedPubMedCentralCrossRef
29.
go back to reference Yang S, Liu Y, Li MY, Ng CSH, Yang SL, Wang S, Zou C, Dong Y, Du J, Long X, et al. FOXP3 promotes tumor growth and metastasis by activating Wnt/β-catenin signaling pathway and EMT in non-small cell lung cancer. Mol Cancer. 2017;16(1):124.PubMedPubMedCentralCrossRef Yang S, Liu Y, Li MY, Ng CSH, Yang SL, Wang S, Zou C, Dong Y, Du J, Long X, et al. FOXP3 promotes tumor growth and metastasis by activating Wnt/β-catenin signaling pathway and EMT in non-small cell lung cancer. Mol Cancer. 2017;16(1):124.PubMedPubMedCentralCrossRef
30.
go back to reference Pulaski BA, Ostrand-Rosenberg S. Mouse 4T1 breast tumor model. Current protocols in immunology 2001, Chapter 20:Unit 20.22. Pulaski BA, Ostrand-Rosenberg S. Mouse 4T1 breast tumor model. Current protocols in immunology 2001, Chapter 20:Unit 20.22.
31.
go back to reference Gilad Y, Eliaz Y, Yu Y, Han SJ, O’Malley BW, Lonard DM. Drug-induced PD-L1 expression and cell stress response in breast cancer cells can be balanced by drug combination. Sci Rep. 2019;9(1):15099.PubMedPubMedCentralCrossRef Gilad Y, Eliaz Y, Yu Y, Han SJ, O’Malley BW, Lonard DM. Drug-induced PD-L1 expression and cell stress response in breast cancer cells can be balanced by drug combination. Sci Rep. 2019;9(1):15099.PubMedPubMedCentralCrossRef
32.
go back to reference Custer RP, Bosma GC, Bosma MJ. Severe combined immunodeficiency (SCID) in the mouse. Pathology, reconstitution, neoplasms. Am J Pathol 1985;120(3):464–77. Custer RP, Bosma GC, Bosma MJ. Severe combined immunodeficiency (SCID) in the mouse. Pathology, reconstitution, neoplasms. Am J Pathol 1985;120(3):464–77.
34.
go back to reference Zhuang Z, Ju HQ, Aguilar M, Gocho T, Li H, Iida T, Lee H, Fan X, Zhou H, Ling J, et al. IL1 receptor antagonist inhibits pancreatic cancer growth by abrogating NF-κB activation. Clin Cancer Res. 2016;22(6):1432–44.PubMedCrossRef Zhuang Z, Ju HQ, Aguilar M, Gocho T, Li H, Iida T, Lee H, Fan X, Zhou H, Ling J, et al. IL1 receptor antagonist inhibits pancreatic cancer growth by abrogating NF-κB activation. Clin Cancer Res. 2016;22(6):1432–44.PubMedCrossRef
35.
go back to reference Tokunaga R, Zhang W, Naseem M, Puccini A, Berger MD, Soni S, McSkane M, Baba H, Lenz HJ. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation: a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40–7.PubMedCrossRef Tokunaga R, Zhang W, Naseem M, Puccini A, Berger MD, Soni S, McSkane M, Baba H, Lenz HJ. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation: a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40–7.PubMedCrossRef
37.
go back to reference Park EJ, Kang YJ, Lee HK, Kim HS, Hong JT, Kim Y, Han S-B: Role of CXCR3 in natural killer cell migration to melanoma cells. J Immunol. 2019; 202(1 Supplement):51.17. Park EJ, Kang YJ, Lee HK, Kim HS, Hong JT, Kim Y, Han S-B: Role of CXCR3 in natural killer cell migration to melanoma cells. J Immunol. 2019; 202(1 Supplement):51.17.
38.
go back to reference Xu J, Wu RC, O’Malley BW. Normal and cancer-related functions of the p160 steroid receptor co-activator (SRC) family. Nat Rev Cancer. 2009;9(9):615–30.PubMedPubMedCentralCrossRef Xu J, Wu RC, O’Malley BW. Normal and cancer-related functions of the p160 steroid receptor co-activator (SRC) family. Nat Rev Cancer. 2009;9(9):615–30.PubMedPubMedCentralCrossRef
39.
go back to reference Shimabukuro-Vornhagen A, Gödel P, Subklewe M, Stemmler HJ, Schlößer HA, Schlaak M, Kochanek M, Böll B, von Bergwelt-Baildon MS. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56.PubMedPubMedCentralCrossRef Shimabukuro-Vornhagen A, Gödel P, Subklewe M, Stemmler HJ, Schlößer HA, Schlaak M, Kochanek M, Böll B, von Bergwelt-Baildon MS. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56.PubMedPubMedCentralCrossRef
40.
go back to reference O’Melia MJ, Manspeaker MP, Thomas SN. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol Immunotherapy: CII. 2021;70(8):2179–95.PubMedCrossRef O’Melia MJ, Manspeaker MP, Thomas SN. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol Immunotherapy: CII. 2021;70(8):2179–95.PubMedCrossRef
41.
go back to reference Gravekamp C, Leal B, Denny A, Bahar R, Lampkin S, Castro F, Kim SH, Moore D, Reddick R. In vivo responses to vaccination with Mage-b, GM-CSF and thioglycollate in a highly metastatic mouse breast tumor model, 4T1. Cancer Immunol Immunother: CII. 2008;57(7):1067–77.PubMedCrossRef Gravekamp C, Leal B, Denny A, Bahar R, Lampkin S, Castro F, Kim SH, Moore D, Reddick R. In vivo responses to vaccination with Mage-b, GM-CSF and thioglycollate in a highly metastatic mouse breast tumor model, 4T1. Cancer Immunol Immunother: CII. 2008;57(7):1067–77.PubMedCrossRef
42.
go back to reference Chavez KJ, Garimella SV, Lipkowitz S. Triple negative breast cancer cell lines: one tool in the search for better treatment of triple negative breast cancer. Breast Dis. 2010;32(1–2):35–48.PubMedPubMedCentral Chavez KJ, Garimella SV, Lipkowitz S. Triple negative breast cancer cell lines: one tool in the search for better treatment of triple negative breast cancer. Breast Dis. 2010;32(1–2):35–48.PubMedPubMedCentral
43.
go back to reference van der Hage JA, Mieog JS, van de Vijver MJ, van de Velde CJ. Efficacy of adjuvant chemotherapy according to hormone receptor status in young patients with breast cancer: a pooled analysis. Breast Cancer Res. 2007;9(5):R70.PubMedPubMedCentralCrossRef van der Hage JA, Mieog JS, van de Vijver MJ, van de Velde CJ. Efficacy of adjuvant chemotherapy according to hormone receptor status in young patients with breast cancer: a pooled analysis. Breast Cancer Res. 2007;9(5):R70.PubMedPubMedCentralCrossRef
45.
go back to reference Nikolova M, Lelievre JD, Carriere M, Bensussan A, Lévy Y. Regulatory T cells differentially modulate the maturation and apoptosis of human CD8+ T-cell subsets. Blood. 2009;113(19):4556–65.PubMedCrossRef Nikolova M, Lelievre JD, Carriere M, Bensussan A, Lévy Y. Regulatory T cells differentially modulate the maturation and apoptosis of human CD8+ T-cell subsets. Blood. 2009;113(19):4556–65.PubMedCrossRef
46.
go back to reference Littwitz-Salomon E, Malyshkina A, Schimmer S, Dittmer U. The cytotoxic activity of natural killer cells is suppressed by IL-10(+) regulatory T cells during acute retroviral infection. Front Immunol. 1947;2018:9. Littwitz-Salomon E, Malyshkina A, Schimmer S, Dittmer U. The cytotoxic activity of natural killer cells is suppressed by IL-10(+) regulatory T cells during acute retroviral infection. Front Immunol. 1947;2018:9.
47.
go back to reference Yang S, Liu Y, Li M-Y, Ng CSH, Yang S-l, Wang S, Zou C, Dong Y, Du J, Long X, et al. FOXP3 promotes tumor growth and metastasis by activating Wnt/β-catenin signaling pathway and EMT in non-small cell lung cancer. Mol Cancer 2017;16(1):124. Yang S, Liu Y, Li M-Y, Ng CSH, Yang S-l, Wang S, Zou C, Dong Y, Du J, Long X, et al. FOXP3 promotes tumor growth and metastasis by activating Wnt/β-catenin signaling pathway and EMT in non-small cell lung cancer. Mol Cancer 2017;16(1):124.
48.
go back to reference Hor JL, Whitney PG, Zaid A, Brooks AG, Heath WR, Mueller SN. Spatiotemporally distinct interactions with dendritic cell subsets facilitates CD4+ and CD8+ T cell activation to localized viral infection. Immunity. 2015;43(3):554–65.PubMedCrossRef Hor JL, Whitney PG, Zaid A, Brooks AG, Heath WR, Mueller SN. Spatiotemporally distinct interactions with dendritic cell subsets facilitates CD4+ and CD8+ T cell activation to localized viral infection. Immunity. 2015;43(3):554–65.PubMedCrossRef
49.
go back to reference Kennedy R, Celis E. Multiple roles for CD4+ T cells in anti-tumor immune responses. Immunol Rev. 2008;222:129–44.PubMedCrossRef Kennedy R, Celis E. Multiple roles for CD4+ T cells in anti-tumor immune responses. Immunol Rev. 2008;222:129–44.PubMedCrossRef
50.
go back to reference Krummel MF, Mahale JN, Uhl LFK, Hardison EA, Mujal AM, Mazet JM, Weber RJ, Gartner ZJ, Gérard A. Paracrine costimulation of IFN-γ signaling by integrins modulates CD8 T cell differentiation. Proc Natl Acad Sci USA. 2018;115(45):11585–90.PubMedPubMedCentralCrossRef Krummel MF, Mahale JN, Uhl LFK, Hardison EA, Mujal AM, Mazet JM, Weber RJ, Gartner ZJ, Gérard A. Paracrine costimulation of IFN-γ signaling by integrins modulates CD8 T cell differentiation. Proc Natl Acad Sci USA. 2018;115(45):11585–90.PubMedPubMedCentralCrossRef
51.
go back to reference Brehm MA, Daniels KA, Welsh RM: Rapid production of TNF-alpha following TCR engagement of naive CD8 T cells. J Immunol (Baltimore, Md : 1950) 2005;175(8):5043–9. Brehm MA, Daniels KA, Welsh RM: Rapid production of TNF-alpha following TCR engagement of naive CD8 T cells. J Immunol (Baltimore, Md : 1950) 2005;175(8):5043–9.
52.
go back to reference Harari A, Bellutti Enders F, Cellerai C, Bart PA, Pantaleo G. Distinct profiles of cytotoxic granules in memory CD8 T cells correlate with function, differentiation stage, and antigen exposure. J Virol. 2009;83(7):2862–71.PubMedPubMedCentralCrossRef Harari A, Bellutti Enders F, Cellerai C, Bart PA, Pantaleo G. Distinct profiles of cytotoxic granules in memory CD8 T cells correlate with function, differentiation stage, and antigen exposure. J Virol. 2009;83(7):2862–71.PubMedPubMedCentralCrossRef
54.
go back to reference Cohavy O, Targan SR: CD56 marks an effector T cell subset in the human intestine. J Immunol. (Baltimore, Md : 1950) 2007;178(9):5524–32. Cohavy O, Targan SR: CD56 marks an effector T cell subset in the human intestine. J Immunol. (Baltimore, Md : 1950) 2007;178(9):5524–32.
55.
go back to reference Riedel DJ, Sajadi MM, Armstrong CL, Cummings JS, Cairo C, Redfield RR, Pauza CD. Natural viral suppressors of HIV-1 have a unique capacity to maintain gammadelta T cells. AIDS (London, England). 2009;23(15):1955–64.CrossRef Riedel DJ, Sajadi MM, Armstrong CL, Cummings JS, Cairo C, Redfield RR, Pauza CD. Natural viral suppressors of HIV-1 have a unique capacity to maintain gammadelta T cells. AIDS (London, England). 2009;23(15):1955–64.CrossRef
56.
go back to reference Wang Y, Wang K, Han GC, Wang RX, Xiao H, Hou CM, Guo RF, Dou Y, Shen BF, Li Y, et al. Neutrophil infiltration favors colitis-associated tumorigenesis by activating the interleukin-1 (IL-1)/IL-6 axis. Mucosal Immunol. 2014;7(5):1106–15.PubMedCrossRef Wang Y, Wang K, Han GC, Wang RX, Xiao H, Hou CM, Guo RF, Dou Y, Shen BF, Li Y, et al. Neutrophil infiltration favors colitis-associated tumorigenesis by activating the interleukin-1 (IL-1)/IL-6 axis. Mucosal Immunol. 2014;7(5):1106–15.PubMedCrossRef
58.
go back to reference Arend WP, Malyak M, Guthridge CJ, Gabay C. Interleukin-1 receptor antagonist: role in biology. Annu Rev Immunol. 1998;16:27–55.PubMedCrossRef Arend WP, Malyak M, Guthridge CJ, Gabay C. Interleukin-1 receptor antagonist: role in biology. Annu Rev Immunol. 1998;16:27–55.PubMedCrossRef
Metadata
Title
Steroid receptor coactivator-3 inhibition generates breast cancer antitumor immune microenvironment
Authors
Sang Jun Han
Nuri Sung
Jin Wang
Bert W. O’Malley
David M. Lonard
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2022
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-022-01568-2

Other articles of this Issue 1/2022

Breast Cancer Research 1/2022 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