GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2358-2358
    Abstract: Breast cancer remains the second leading cause of cancer-related deaths among women. In recent years, immunotherapy has been tremendously successful in some metastatic cancers such as melanoma. However, a majority of breast cancer patients do not benefit from existing immunotherapy treatments, leaving many with an unmet need. Although undoubtedly multifactorial, one major obstacle to anti-cancer therapies, is the highly immunosuppressive breast tumor microenvironment. This phenomenon is strongly maintained by myeloid immune cells and immunosuppressive regulatory T cells (Tregs), which hinder anti-tumor immunosurveillance and promote tumor progression. Thus, strategies to ‘re-educate’ myeloid cells to inhibit Tregs is a potentially promising anti-cancer strategy. Mining clinical data, we have found that elevated mRNA expression of the nuclear receptor, NR0B2 within breast tumors is associated with an increased time to recurrence. Single cell RNA-sequencing indicates that NR0B2 is expressed within the macrophage populations of normal breast tissue, and various dendritic cell (DC) types in PBMCs. Overexpression of NR0B2 or activation with a small molecule agonist in murine bone marrow derived macrophages (BMDMs) or DCs resulted in a dichotomous T cell expansion - away from Tregs. Conversely, Treg expansion increased when NR0B2 was knocked-down. Tumor growth was markedly increased in mice lacking myeloid specific NR0B2 expression. We further investigated the downstream targets of NR0B2 mediating this anti-tumor phenotype and identified that NLRP3 inflammasome-IL1β activity is a likely modulator in re-educating myeloid cell-Treg function. Importantly, a putative small molecule agonist decreased established metastatic lesions and increased the efficacy of αPD-L1. Subsequent medicinal chemistry was used to develop a novel NR0B2 agonist with strong anti-metastatic properties when used as a single agent in a preclinical mouse model. Collectively, our data implicates NR0B2 within myeloid cells as a modulator of Tregs, a cell population that has thus far been therapeutically intractable. Therefore, NR0B2 may prove to be a promising therapeutic target to reshape the tumor microenvironment and improve breast cancer immunotherapy. This work was supported by the Era of Hope Scholar Award from the Department of Defense Breast Cancer Research Program grant (BC200206), National Cancer Institute (R01CA234025), and NIH Chemistry-Biology Interface Training Grant (T32-GM136629). Citation Format: Hashni Epa Vidana Gamage, Sayyed Hamed Shahoei, Tiffany Nguyen, Rachel Farmer, Samuel Albright, Erin Weisser, Rafael O. Bautista, Claire P. Schane, Yu Wang, Adam Nelczyk, Liqian Ma, Srishti Tiwari, Anasuya Das Gupta, Shruti Bendre, Lionel Apetoh, Paul J. Hergenrother, Erik R. Nelson. NR0B2 re-educates myeloid cells within the tumor microenvironment: Potential novel strategy for breast cancer immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2358.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 4, No. Supplement_1 ( 2020-05-08)
    Abstract: Immune checkpoint blockade has had underwhelming responses in breast cancer, in part due to the highly immune suppressive microenvironment. As a result, breast cancer continues to be the second most common cancer-related mortality amongst women, providing strong rationale for the development of new therapeutic approaches. Elevated circulating cholesterol is a poor prognostic, while breast cancer patients taking cholesterol-lowering drugs display increased time to recurrence. We and others have previously demonstrated that cholesterol metabolites mediate these effects by promoting breast cancer growth and metastasis, in part by suppressing the immune system. Therefore, given the demonstrated importance of cholesterol and its metabolites in breast cancer pathophysiology and immunology, we hypothesized that proteins involved in the regulation of cholesterol homeostasis would influence cancer progression. Through informatics analysis of breast tumors, we found that elevated expression of Small Heterodimer Partner (SHP; NR0B2) was a favorable prognostic. Antigen presenting cells such as macrophages and dendritic cells were found to express SHP, and manipulation of SHP altered the expression of genes involved in cross-talk with T cells. Intriguingly, when activated T cells were co-cultured with macrophages overexpressing SHP, there was a decrease in the expansion of regulatory T cells (Tregs) and vice versa in the absence of SHP. Adoptive transfer studies confirmed that loss of SHP resulted in immune suppressive Tregs. We hypothesized that myeloid cell-expressed SHP would promote immune surveillance and tumor clearance. In support of this hypothesis, tumors in the MMTV-PyMT model of mammary cancer grew at an accelerated rate in SHP-knockout mice. Tumors from these mice had significantly more Tregs and fewer effector T cells. Furthermore, orthotopic mammary tumor grafts grew at an increased rate in mice lacking SHP expression in myeloid cells (SHPfl/fl;LysMCre), compared to controls. A small molecule agonist of SHP impaired primary and metastatic tumor growth, and significantly enhanced the efficacy of immune checkpoint blockade in murine models of mammary cancer. Therefore, SHP represents a potential target to decrease suppressive Tregs, thereby allowing for immune-clearance of tumors.
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2020
    detail.hit.zdb_id: 2881023-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 5, No. Supplement_1 ( 2021-05-03), p. A1019-A1019
    Abstract: Despite the development of various therapeutic strategies, breast cancer persists as the second leading cause of cancer-related death among women in the United States. While endocrine modulation and monoclonal antibody therapy have proved to be indispensable modes of intervention for hormone receptor (HR)-positive and HER-2 positive patients, the triple negative breast cancer (TNBC) patient population do not respond to these therapies. As TNBC is considered one of the most challenging subtypes of breast cancer to treat, there is a significant need for the development of targeted therapeutics. Due to their well-known amenability to small-molecule modulation, we investigated whether any nuclear receptors beyond those that are traditionally studied in breast cancer (e.g. ER, PR, and AR), may represent a novel target in the TNBC patient population. Analysis of clinical data revealed that expression of the orphan nuclear receptor TLX (NR2E1) was positively correlated with relapse-free survival, distant metastasis-free survival, and overall survival in both ER-negative and basal-like breast cancer patients. Therefore, we hypothesized that TLX could influence the pathophysiology of TNBC. To interrogate this hypothesis, we established TNBC cells with stable expression of TLX in order to identify direct regulatory targets, as well as the precise physiological mechanism(s) TLX may be regulating. To date, our work has revealed that TLX inhibits proliferation, slows migration, alters chemosensitivity, and impairs cell cycle progression in TNBC cells. In agreement with these findings, our work has also revealed that TLX is capable of modulating the expression of several genes that are known to regulate the processes of growth, migration, and cell cycle. Taken together, our early work supports our hypothesis, and provides valuable insight into the potential pro-survival function of TLX in TNBC. Ongoing work will continue to probe the mechanisms by which TLX impacts breast cancer biology, and establish whether the growth-inhibitory effects translate to in vivo models. As prior work has demonstrated that TLX’s transcriptional activity can be regulated by both synthetic and natural ligands, the results of our work would provide the foundational data necessary for the development of a TLX-based therapy for a patient population with limited therapeutic options and a poor prognostic outlook.
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2021
    detail.hit.zdb_id: 2881023-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Biomaterials Science, Royal Society of Chemistry (RSC), Vol. 9, No. 2 ( 2021), p. 506-518
    Type of Medium: Online Resource
    ISSN: 2047-4830 , 2047-4849
    Language: English
    Publisher: Royal Society of Chemistry (RSC)
    Publication Date: 2021
    detail.hit.zdb_id: 2693928-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. P6-05-01-P6-05-01
    Abstract: It has become clear that cholesterol metabolism and homeostasis play significant roles in the progression of breast cancer. Specifically, elevated circulating cholesterol is a poor prognostic, while patients taking cholesterol-lowering drugs such as statins display increased recurrence-free survival time. Preclinical and clinical work has established that in addition to cholesterol, various downstream metabolites play direct roles in promoting breast cancer growth and metastasis. Given the demonstrated importance of cholesterol and its metabolites in breast cancer pathophysiology, we hypothesized that proteins involved in the regulation of cholesterol homeostasis would play a role in cancer progression. Therefore, we performed an informatics screen to identify those regulatory proteins associated with breast cancer progression. We focused on nuclear receptors due to their well-defined ligand-binding pocket and thus their proclivity to drug intervention. Our screen revealed that increased expression of Small Heterodimer Partner (SHP; NR0B2) was associated with an increased time to recurrence. However, manipulation of SHP within breast cancer cells did not alter proliferation or migration, suggesting that its protective role is likely conveyed through the tumor microenvironment. Macrophages were found to express SHP, and manipulation of SHP within macrophages resulted in altered expression of molecules associated with antigen presentation. Considering the clinical data indicating a protective role for SHP, it was somewhat paradoxical that its loss within macrophages resulted in an increased expansion of T cells. Upon further investigation, we found that this expansion was skewed towards regulatory T cells (Tregs). On the other hand, overexpression of SHP resulted in decreased expansion of Tregs. The immune-suppressive activity of the resulting Tregs was confirmed in subsequent assays. While immune therapies have revolutionized the treatment of certain cancers, their utility in breast cancer has been limited, especially outside of triple-negative disease. It has been speculated that this may be due to the highly immune-suppressive activities of certain myeloid and T cell populations. Thus, reducing Treg infiltration or activity likely represents a rational way to enhance immune therapies. In this regard, SHP-knockout mice bred with the MMTV-PyMT model of mammary cancer displayed significantly enhanced tumor growth compared to SHP-replete mice. Likewise, orthotopic mammary tumor grafts grew at an increased rate in mice where SHP was selectively knocked out in cells of the myeloid lineage (SHPfl/fl;LysMCre), compared to controls. Importantly, treatment with a small molecule agonist of SHP significantly enhanced the efficacy of anti-PD-L1 therapy in blocking the growth of an orthotopically grafted tumor, as well as in a model of metastatic mammary cancer. Collectively, our data strongly support a role for SHP in reducing the progression of breast cancer by limiting Treg expansion, thereby facilitating an anti-cancer immune response. As this nuclear receptor is amenable to small molecule intervention, SHP may represent a unique way to enhance the efficacy of immune checkpoint blockade. This study was funded in part by awards to ERN from the DOD BCRP (BC171214) and NCI (R01CA234025), to LA from the French National Research Agency Lipstic Labex (ANR-11-LABX-0021), and a STEM Chateaubriand Fellowship to SHS from the Embassy of France in the United States. Citation Format: Erik R. Nelson, Sayyed Hamed Shahoei, Adam T Nelson, Madeline A Henn, Ashley E Mathews, Joy J Chen, Varsha Vembar, Liqian Ma, Lionel Apetoh. The small heterodimer partner in macrophages reduces expansion of regulatory T cells and enhances immune checkpoint inhibition in breast cancer [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P6-05-01.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    The Endocrine Society ; 2019
    In:  Endocrinology Vol. 160, No. 7 ( 2019-07-01), p. 1573-1589
    In: Endocrinology, The Endocrine Society, Vol. 160, No. 7 ( 2019-07-01), p. 1573-1589
    Abstract: The involvement of small heterodimer partner (SHP) in the inhibition of hepatic bile acid synthesis from cholesterol has been established. However, extrahepatic expression of SHP implies that SHP may have regulatory functions other than those in the liver. Here, we find that SHP mRNA expression is high in murine bone marrow cells, suggesting a physiological role within macrophages. Indeed, expression of SHP in macrophages decreases the transcriptional activity and nuclear localization of nuclear factor κB, whereas downregulation of SHP has the opposite effects. Expression of genes associated with macrophage–T cell crosstalk were altered by overexpression or downregulation of SHP. Intriguingly, increasing SHP expression in macrophages resulted in decreased T cell expansion, a hallmark of T cell activation, whereas knockdown of SHP resulted in increased expansion. Analyses of the expanded T cells revealed a dichotomous skewing between effector T cells and regulatory T cells (Tregs), with SHP overexpression reducing Tregs and downregulation of SHP increasing their expansion. The expanded Tregs were confirmed to be suppressive via adoptive transfers. IL-2 and TGF-β, known inducers of Treg differentiation, were found to be regulated by SHP. Furthermore, SHP occupancy at the promoter region of IL-2 was increased after macrophages were challenged with lipopolysaccharide. Neutralizing antibodies to IL-2 and TGF-β inhibited the expansion of Tregs mediated by downregulation of SHP. This study demonstrates that expression and activity of SHP within macrophages can alter T cell fate and identifies SHP as a potential therapeutic target for autoimmune diseases or solid cancers.
    Type of Medium: Online Resource
    ISSN: 1945-7170
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2019
    detail.hit.zdb_id: 2011695-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Advanced Materials Interfaces, Wiley, Vol. 5, No. 23 ( 2018-12)
    Type of Medium: Online Resource
    ISSN: 2196-7350 , 2196-7350
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2018
    detail.hit.zdb_id: 2750376-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 9, No. 2_Supplement ( 2021-02-01), p. PR006-PR006
    Abstract: Breast cancer remains one of the leading causes of cancer mortality in the US. Elevated cholesterol is a major risk factor for breast cancer onset and recurrence, while cholesterol-lowering drugs, such as statins, are associated with a good prognosis. Previous work in murine models showed that cholesterol increases breast cancer metastasis, and the pro-metastatic effects of cholesterol were due to its primary metabolite, 27HC. In our prior work, myeloid cells were found to be required for the pro-metastatic effects of 27HC, but their precise contribution remains unclear. Here we report that 27HC impairs T cell expansion and cytotoxic function through its actions on myeloid cells, including macrophages, in an LXR-dependent manner. Many oxysterols and LXR ligands had similar effects on T cell expansion. Moreover, their ability to induce the LXR target gene ABCA1 was associated with their effectiveness in impairing T cell expansion. Interestingly, the enzyme responsible for the synthesis of 27HC, CYP27A1, is highly expressed in myeloid cells, suggesting that 27HC may have important autocrine or paracrine functions in these cells, a hypothesis supported by our finding that breast cancer metastasis was reduced in mice with a myeloid specific knockout of CYP27A1. Pharmacologic inhibition of CYP27A1 reduced metastatic growth and improved the efficacy of checkpoint inhibitor, anti-PD-L1. RNA sequencing of 27HC-treated macrophages and GSEA analysis provide further mechanistic insight, revealing an enrichment of MYC and NOTCH signaling pathways, both of which are documented pathways involved in tumor-associated macrophages. Taken together, our work suggests that targeting the CYP27A1 axis in myeloid cells may present therapeutic benefits and improve the response rate to immune therapies in breast cancer. Delineating the link between LXR and the known mechanisms of tumor-associated macrophages is important to fully understand 27HC-driven cancer metastasis. This work was supported by grants to ERN from the National Cancer Institute of the National Institutes of Health (R01CA234025) and METAvivor. This abstract is also being presented as PO050. Citation Format: Liqian Ma, Lawrence Wang, Adam T. Nelson, Chaeyeon Han, Sisi He, Madeline A. Henn, Karan Menon, Joy J. Chen, Amy E. Baek, Anna Vardanyan, Sayyed Hamed Shahoei, Sunghee Park, David J. Shapiro, Som G. Nanjappa, Erik R. Nelson. 27-Hydroxycholesterol acts on myeloid immune cells to induce T cell dysfunction, promoting breast cancer progression [abstract]. In: Abstracts: AACR Virtual Special Conference: Tumor Immunology and Immunotherapy; 2020 Oct 19-20. Philadelphia (PA): AACR; Cancer Immunol Res 2021;9(2 Suppl):Abstract nr PR006.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 3_Supplement ( 2020-03-01), p. A93-A93
    Abstract: Breast cancer continues to be the second most common cancer-related mortality among women, providing strong rationale for the development of new therapeutic approaches. Cholesterol and its metabolism have been implicated in the progression of breast cancer. Specifically, elevated circulating cholesterol is a poor prognostic, while patients taking cholesterol-lowering drugs such as statins display increased recurrence-free survival time. In addition to cholesterol, various downstream metabolites play direct roles in promoting breast cancer growth and metastasis. Given the demonstrated importance of cholesterol and its metabolites in breast cancer pathophysiology, we hypothesized that proteins involved in the regulation of cholesterol homeostasis would play a role in cancer progression. A bioinformatics-based screen identified small heterodimer partner (SHP; NR0B2) as being associated with an increased time to recurrence. However, manipulation of this negative regulator of cholesterol metabolism within breast cancer cells did not alter proliferation or migration, suggesting that its protective role is likely conveyed through the tumor microenvironment. Macrophages were found to express SHP, and manipulation of SHP within macrophages resulted in altered expression of molecules associated with antigen presentation. Considering the clinical data indicating a protective role for SHP, it was somewhat paradoxical that its loss within macrophages resulted in an increased expansion of T cells. Upon further investigation, we found that this expansion was skewed towards regulatory T cells (Tregs). While immune therapies have revolutionized the treatment of certain cancers, their utility in breast cancer has been limited, especially outside of triple-negative disease. It has been postulated that this may be due to the highly immune-suppressive activities of certain myeloid and T-cell populations. Thus, reducing Treg infiltration or activity likely represents a rational way to enhance immune therapies. In this regard, SHP-knockout mice bred with the MMTV-PyMT model of mammary cancer displayed significantly enhanced tumor growth compared to SHP-replete mice. Likewise, orthotopic mammary tumor grafts grew at an increased rate in mice where SHP was selectively knocked out in cells of the myeloid lineage (SHPfl/fl;LysMCre), compared to controls. Importantly, treatment with a small-molecule agonist of SHP significantly enhanced the efficacy of anti-PD-L1 therapy in blocking the growth of an orthotopically grafted tumor, as well as in a model of metastatic mammary cancer. Collectively, our data highlight SHP as a modulator of Tregs, a cell population that has thus far been therapeutically intractable. By limiting Treg expansion and thus facilitating an anticancer immune response, SHP may represent a unique way to enhance the efficacy of immune checkpoint blockade. Funding: DOD BCRP BC171214 and NCI R01CA234025 (ERN), Lipstic Labex ANR-11-LABX-0021 (LA), Chateaubriand Fellowship (SHS). Citation Format: Sayyed Hamed Shahoei, Adam T. Nelson, Madeline A. Henn, Ashley E. Mathews, Joy J. Chen, Varsha Vembar, Liqian Ma, Lionel Apetoh, Erik R. Nelson. Macrophage-expressed small heterodimer partner impairs expansion of regulatory T cells and enhances immune checkpoint inhibition [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr A93.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 10, No. 1 ( 2022-01), p. e003459-
    Abstract: While stimulator of interferon genes (STING) activation in innate immune cells of the tumor microenvironment can result in CD8 T cell-dependent antitumor immunity, whether STING signaling affects CD4 T-cell responses remains elusive. Methods Here, we tested whether STING activation modulated the effector functions of CD4 T cells in vivo by analyzing tumor-infiltrating CD4 T cells and evaluating the contribution of the CD4 T cell-derived cytokines in the antitumor activity of the STING ligand 2′3′-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) in two mouse tumor models. We performed ex vivo experiments to assess the impact of STING activation on CD4 T-cell differentiation and investigate the underlying molecular mechanisms. Finally, we tested whether STING activation enhances T H 9 cell antitumor activity against mouse melanoma upon adoptive transfer. Results We found that activation of STING signaling cell-intrinsically enhances the differentiation and antitumor functions of T H 1 and T H 9 cells by increasing their respective production of interferon gamma (IFN-γ) and interleukin-9. IRF3 and type I interferon receptors (IFNARs) are required for the STING-driven enhancement of T H 1 cell differentiation. However, STING activation favors T H 9 cell differentiation independently of the IFNARs/IRF3 pathway but through mammalian target of rapamycin (mTOR) signaling, underscoring that STING activation differentially affects the fate of distinct CD4 T-cell subsets. The therapeutic effect of STING activation relies on T H 1 and T H 9-derived cytokines, and STING activation enhances the antitumor activity of T H 9 cells upon adoptive transfer. Conclusion Our results reveal the STING signaling pathway as a therapeutic target to boost CD4 T-cell effector functions and antitumor immunity.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2022
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...