94 results on '"Dougall WC"'
Search Results
2. RANK-ligand (RANKL) expression in young breast cancer patients and during pregnancy
- Author
-
Azim, HA, Peccatori, FA, Brohee, S, Branstetter, D, Loi, S, Viale, G, Piccart, M, Dougall, WC, Pruneri, G, Sotiriou, C, Azim, HA, Peccatori, FA, Brohee, S, Branstetter, D, Loi, S, Viale, G, Piccart, M, Dougall, WC, Pruneri, G, and Sotiriou, C
- Abstract
INTRODUCTION: RANKL is important in mammary gland development during pregnancy and mediates the initiation and progression of progesterone-induced breast cancer. No clinical data are available on the effect of pregnancy on RANK/RANKL expression in young breast cancer patients. METHODS: We used our previously published dataset of 65 pregnant and 130 matched young breast cancer patients with full clinical, pathological, and survival information. 85% of patients had available transcriptomic data as well. RANK/RANKL expression by immunohistochemistry using H-score on the primary tumor and adjacent normal tissue was performed. We examined the difference in expression of RANK/RANKL between pregnant and non-pregnant patients and their association with clinicopathological features and prognosis. We also evaluated genes and pathways associated with RANK/RANKL expression on primary tumors. RESULTS: RANKL but not RANK expression was more prevalent in the pregnant group, both on the tumor and adjacent normal tissue, independent of other clinicopathological factors (both P <0.001). 18.7% of pregnant and 5.3% of non-pregnant patients had tumors showing ≥10% of cells with 3+ RANKL expression. RANKL expression was significantly higher in progesterone receptor-positive, and luminal A-like tumors, with negative correlation with Ki-67 (all P <0.001). On the contrary, RANK expression was higher in triple negative tumors (P <0.001). Using false discovery rate <0.05, 151 and 1,207 genes were significantly correlated with tumor-expressed RANKL and RANK expression by immunohistochemistry, respectively. High RANKL expression within primary tumor was associated with pathways related to mammary gland development, bone resorption, T-cell proliferation and regulation of chemotaxis, while RANK expression was associated with immune response and proliferation pathways. At a median follow-up of 65 months, neither RANK nor RANKL expression within tumor was associated with disease free survival in pr
- Published
- 2015
3. Abstract P1-08-25: RANK expression is prognostic and predictive in primary breast carcinoma: Analysis of samples from the GeparTrio study
- Author
-
Pfitzner, BM, primary, Branstetter, D, additional, Mergler, B, additional, Loibl, S, additional, Denkert, C, additional, Fasching, PA, additional, Dougall, WC, additional, and von Minckwitz, G, additional
- Published
- 2013
- Full Text
- View/download PDF
4. Abstract P5-03-02: Expression of RANK and RANK ligand (RANKL) in breast carcinoma and distinct breast epithelial cells from BRCA1 mutation carriers
- Author
-
Lindeman, GJ, primary, Visvader, JE, additional, Vaillant, F, additional, Mann, GB, additional, Soriano, R, additional, Branstetter, D, additional, and Dougall, WC, additional
- Published
- 2013
- Full Text
- View/download PDF
5. P3-01-14: RANK and RANK Ligand (RANKL) Expression in Invasive Breast Carcinoma and Human Breast Cancer Cell Lines.
- Author
-
Jacob, A, primary, Branstetter, D, additional, Rohrbach, K, additional, Winters, A, additional, Erwert, R, additional, Allred, S, additional, Jones, J, additional, Miller, R, additional, Tometsko, M, additional, Blake, M, additional, and Dougall, WC, additional
- Published
- 2011
- Full Text
- View/download PDF
6. P2-01-02: Pharmacological Inhibition of RANKL Attenuates Mammary Tumor Development and Lung Metastases in the MMTV-neu Transgenic Spontaneous Tumor Model.
- Author
-
Branstetter, D, primary, Jacob, A, additional, Soriano, R, additional, Allred, S, additional, Jones, J, additional, Miller, R, additional, and Dougall, WC, additional
- Published
- 2011
- Full Text
- View/download PDF
7. P3-04-05: Expression Profiles of RANK and RANKL mRNA and Protein in the Mammary Gland of Female Cynomolgus Monkeys after Long-Term Treatment with Different Menopausal Hormone Therapies.
- Author
-
Branstetter, D, primary, Wood, CE, additional, Rohrbach, K, additional, Borgerink, H, additional, and Dougall, WC, additional
- Published
- 2011
- Full Text
- View/download PDF
8. Correlation of RANK and RANKL with mammographic density in primary breast cancer patients.
- Author
-
Wunderle M, Heindl F, Behrens AS, Häberle L, Hack CC, Heusinger K, Huebner H, Gass P, Ruebner M, Schulz-Wendtland R, Erber R, Hartmann A, Beckmann MW, Dougall WC, Press MF, Fasching PA, and Emons J
- Subjects
- Humans, Female, Middle Aged, Aged, Adult, Case-Control Studies, Immunohistochemistry, Tissue Array Analysis, Breast diagnostic imaging, Breast pathology, Breast metabolism, RANK Ligand metabolism, RANK Ligand analysis, Receptor Activator of Nuclear Factor-kappa B metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Breast Neoplasms diagnostic imaging, Breast Density
- Abstract
Purpose: The receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL) have been shown to promote proliferation of the breast and breast carcinogenesis. The objective of this analysis was to investigate whether tumor-specific RANK and RANKL expression in patients with primary breast cancer is associated with high percentage mammographic density (PMD), which is a known breast cancer risk factor., Methods: Immunohistochemical staining of RANK and RANKL was performed in tissue microarrays (TMAs) from primary breast cancer samples of the Bavarian Breast Cancer Cases and Controls (BBCC) study. For RANK and RANKL expression, histochemical scores (H scores) with a cut-off value of > 0 vs 0 were established. PMD was measured in the contralateral, non-diseased breast. Linear regression models with PMD as outcome were calculated using common predictors of PMD (age at breast cancer diagnosis, body mass index (BMI) and parity) and RANK and RANKL H scores. Additionally, Spearman rank correlations (ρ) between PMD and RANK and RANKL H score were performed., Results: In the final cohort of 412 patients, breast cancer-specific RANK and RANKL expression was not associated with PMD (P = 0.68). There was no correlation between PMD and RANK H score (Spearman's ρ = 0.01, P = 0.87) or RANKL H score (Spearman's ρ = 0.04, P = 0.41). RANK expression was highest in triple-negative tumors, followed by HER2-positive, luminal B-like and luminal A-like tumors, while no subtype-specific expression of RANKL was found., Conclusion: Results do not provide evidence for an association of RANK and RANKL expression in primary breast cancer with PMD., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Author Correction: The NK cell granule protein NKG7 regulates cytotoxic granule exocytosis and inflammation.
- Author
-
Ng SS, De Labastida Rivera F, Yan J, Corvino D, Das I, Zhang P, Kuns R, Chauhan SB, Hou J, Li XY, Frame TCM, McEnroe BA, Moore E, Na J, Engel JA, Soon MSF, Singh B, Kueh AJ, Herold MJ, Montes de Oca M, Singh SS, Bunn PT, Aguilera AR, Casey M, Braun M, Ghazanfari N, Wani S, Wang Y, Amante FH, Edwards CL, Haque A, Dougall WC, Singh OP, Baxter AG, Teng MWL, Loukas A, Daly NL, Cloonan N, Degli-Esposti MA, Uzonna J, Heath WR, Bald T, Tey SK, Nakamura K, Hill GR, Kumar R, Sundar S, Smyth MJ, and Engwerda CR
- Published
- 2024
- Full Text
- View/download PDF
10. RANK and RANKL Expression in Tumors of Patients with Early Breast Cancer.
- Author
-
Behrens A, Wurmthaler L, Heindl F, Gass P, Häberle L, Volz B, Hack CC, Emons J, Erber R, Hartmann A, Beckmann MW, Ruebner M, Dougall WC, Press MF, Fasching PA, and Huebner H
- Abstract
Introduction: The receptor activator of nuclear factor-κB (RANK) pathway was associated with the pathogenesis of breast cancer. Several studies attempted to link the RANK/RANKL pathway to prognosis; however, with inconsistent outcomes. We aimed to further contribute to the knowledge about RANK/RANKL as prognostic factors in breast cancer. Within this study, protein expression of RANK and its ligand, RANKL, in the tumor tissue was analyzed in association with disease-free survival (DFS) and overall survival (OS) in a study cohort of patients with early breast cancer., Patients and Methods: 607 samples of female primary and early breast cancer patients from the Bavarian Breast Cancer Cases and Controls Study were analyzed to correlate the RANK and RANKL expression with DFS and OS. Therefore, expression was quantified using immunohistochemical staining of a tissue microarray. H-scores were determined with the cut-off value of 8.5 for RANK and 0 for RANKL expression, respectively., Results: RANK and RANKL immunohistochemistry were assessed by H-score. Both biomarkers did not correlate (ρ = -0.04). According to molecular subtypes, triple-negative tumors and HER2-positive tumors showed a higher number of RANK-positive tumors (H-score ≥ 8.5), however, no subtype-specific expression of RANKL could be detected. Higher RANKL expression tended to correlate with a better prognosis. However, RANK and RANKL expression could not be identified as statistically significant prognostic factors within the study cohort., Conclusions: Tumor-specific RANK and RANKL expressions are not applicable as prognostic factors for DFS and OS, but might be associated with subtype-specific breast cancer progression., Competing Interests: Conflict of Interest P.A.F. reports personal fees from Novartis, grants from BioNTech, personal fees from Pfizer, personal fees from Daiichi Sankyo, personal fees from AstraZeneca, personal fees from Eisai, personal fees from Merck Sharp & Dohme, grants from Cepheid, personal fees from Lilly, personal fees from Pierre Fabre, personal fees from SeaGen, personal fees from Roche, personal fees from Hexal, personal fees from Agendia, personal fees from Gilead. P.G. has received honoraria from Novartis, MSD, and AstraZeneca. J.E. has received honoraria from Eisai, Pfizer, and Novartis. A.H. has received honoraria for lectures or consulting/advisory boards for Abbvie, Agilent, AstraZeneca, Biocartis, BMS, Boehringer Ingelheim, Cepheid, Diaceutics, Gilead, Illumina, Ipsen, Janssen, Lilly, Merck, MSD, Nanostring, Novartis, Pfizer, Qiagen, QUIP GmbH, Roche, Sanofi, 3DHistotech and other research support from AstraZeneca, Biocartis, Cepheid, Gilead, Illumina, Janssen, Nanostring, Novartis, Owkin, Qiagen, QUIP GmbH, Roche, Sanofi. M.F.P. declares advisory board membership for Biocartis, Cepheid, Lilly USA; reports a consultant role for AstraZeneca, Eli Lilly & Company, Merck, Novartis, and Zymeworks; reports ownership interest in TORL Biotherapeutics; reports fee-for-service agreements from 1200 Pharma, Ambrx, TORL Biotherapeutics, TRIO, TRIO-US, and Zymeworks. The remaining authors declare that they do not have a conflict of interest., (The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).)
- Published
- 2023
- Full Text
- View/download PDF
11. NMDAR antagonists suppress tumor progression by regulating tumor-associated macrophages.
- Author
-
Yuan D, Hu J, Ju X, Putz EM, Zheng S, Koda S, Sun G, Deng X, Xu Z, Nie W, Zhao Y, Li X, Dougall WC, Shao S, Chen Y, Tang R, Zheng K, and Yan J
- Subjects
- Humans, Tumor-Associated Macrophages, Neoplastic Processes, Memantine, Tumor Microenvironment, Liver Neoplasms, Sarcoma
- Abstract
Neurotransmitter receptors are increasingly recognized to play important roles in anti-tumor immunity. The expression of the ion channel N-methyl-D-aspartate receptor (NMDAR) on macrophages was reported, but the role of NMDAR on macrophages in the tumor microenvironment (TME) remains unknown. Here, we show that the activation of NMDAR triggered calcium influx and reactive oxygen species production, which fueled immunosuppressive activities in tumor-associated macrophages (TAMs) in the hepatocellular sarcoma and fibrosarcoma tumor settings. NMDAR antagonists, MK-801, memantine, and magnesium, effectively suppressed these processes in TAMs. Single-cell RNA sequencing analysis revealed that blocking NMDAR functionally and metabolically altered TAM phenotypes, such that they could better promote T cell- and Natural killer (NK) cell-mediated anti-tumor immunity. Treatment with NMDAR antagonists in combination with anti-PD-1 antibody led to the elimination of the majority of established preclinical liver tumors. Thus, our study uncovered an unknown role for NMDAR in regulating macrophages in the TME of hepatocellular sarcoma and provided a rationale for targeting NMDAR for tumor immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
12. Inhibition of CD39 unleashes macrophage antibody-dependent cellular phagocytosis against B-cell lymphoma.
- Author
-
Casey M, Segawa K, Law SC, Sabdia MB, Nowlan B, Salik B, Lee C, Winterford C, Pearson S, Madore J, Dougall WC, Gandhi MK, and Nakamura K
- Subjects
- Humans, Rituximab pharmacology, Rituximab therapeutic use, Adenosine metabolism, Phagocytosis, Macrophages, Lymphoma, Large B-Cell, Diffuse pathology
- Abstract
Redirection of tumor-associated macrophages to eliminate tumor cells holds great promise for overcoming therapeutic resistance to rituximab and other antibody drugs. Here, we determined the expression of ectonucleotidases CD39 and CD73 in diffuse large B-cell lymphoma (DLBCL), and examined the impact of extracellular ATP (eATP) metabolism on macrophage-mediated anti-lymphoma immunity. Immunostaining of tissue microarray samples showed that CD39 (the ecto-enzyme for eATP hydrolysis) was highly expressed in tumors with the non-germinal center B-cell-like (non-GCB) subtype, and to a lesser extent tumors with the GCB subtype. By contrast, the expression of CD73 (the ecto-enzyme for adenosine generation) was undetectable in tumor cells. Pharmacological blockade of CD39 prevented eATP degradation and enhanced engulfment of antibody-coated lymphoma cells by macrophages in a P2X7 receptor-dependent manner, indicating that eATP fueled antibody-dependent cellular phagocytosis (ADCP) activity. Importantly, inhibition of CD39 augmented in vivo anti-lymphoma effects by therapeutic antibodies including rituximab and daratumumab. Furthermore, the addition of a CD39 inhibitor to anti-CD20 and anti-CD47 combination therapy significantly improved survival in a disseminated model of aggressive B-cell lymphoma, supporting the benefit of dual targeting CD39-mediated eATP hydrolysis and CD47-mediated "don't eat me" signal. Together, preventing eATP degradation may be a potential approach to unleash macrophage-mediated anti-lymphoma immunity., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
13. CD155 loss enhances tumor suppression via combined host and tumor-intrinsic mechanisms.
- Author
-
Li XY, Das I, Lepletier A, Addala V, Bald T, Stannard K, Barkauskas D, Liu J, Aguilera AR, Takeda K, Braun M, Nakamura K, Jacquelin S, Lane SW, Teng MW, Dougall WC, and Smyth MJ
- Published
- 2022
- Full Text
- View/download PDF
14. CD155 on Tumor Cells Drives Resistance to Immunotherapy by Inducing the Degradation of the Activating Receptor CD226 in CD8 + T Cells.
- Author
-
Braun M, Aguilera AR, Sundarrajan A, Corvino D, Stannard K, Krumeich S, Das I, Lima LG, Meza Guzman LG, Li K, Li R, Salim N, Jorge MV, Ham S, Kelly G, Vari F, Lepletier A, Raghavendra A, Pearson S, Madore J, Jacquelin S, Effern M, Quine B, Koufariotis LT, Casey M, Nakamura K, Seo EY, Hölzel M, Geyer M, Kristiansen G, Taheri T, Ahern E, Hughes BGM, Wilmott JS, Long GV, Scolyer RA, Batstone MD, Landsberg J, Dietrich D, Pop OT, Flatz L, Dougall WC, Veillette A, Nicholson SE, Möller A, Johnston RJ, Martinet L, Smyth MJ, and Bald T
- Subjects
- Animals, Cell Line, Cell Line, Tumor, HEK293 Cells, Humans, Immune Checkpoint Inhibitors immunology, Immunotherapy methods, Jurkat Cells, Lymphocytes, Tumor-Infiltrating immunology, Male, Melanoma immunology, Mice, Mice, Inbred C57BL, Antigens, Differentiation, T-Lymphocyte immunology, CD8-Positive T-Lymphocytes immunology, Receptors, Virus immunology
- Abstract
The activating receptor CD226 is expressed on lymphocytes, monocytes, and platelets and promotes anti-tumor immunity in pre-clinical models. Here, we examined the role of CD226 in the function of tumor-infiltrating lymphocytes (TILs) and resistance to immunotherapy. In murine tumors, a large proportion of CD8
+ TILs had decreased surface expression of CD226 and exhibited features of dysfunction, whereas CD226hi TILs were highly functional. This correlation was seen also in TILs isolated from HNSCC patients. Mutation of CD226 at tyrosine 319 (Y319) led to increased CD226 surface expression, enhanced anti-tumor immunity and improved efficacy of immune checkpoint blockade (ICB). Mechanistically, tumor-derived CD155, the ligand for CD226, initiated phosphorylation of Y319 by Src kinases, thereby enabling ubiquitination of CD226 by CBL-B, internalization, and proteasomal degradation. In pre-treatment samples from melanoma patients, CD226+ CD8+ T cells correlated with improved progression-free survival following ICB. Our findings argue for the development of therapies aimed at maintaining the expression of CD226., Competing Interests: Declaration of Interests M.J.S. has research agreements with Bristol Myers Squibb (BMS) and Tizona Therapeutics and is on the Scientific Advisory Board of Tizona Therapeutics and Compass Therapeutics. T.B. has research agreements with BMS. B.G.M.H. is a consultant advisor to BMS, Merck Sharp and Dohme, Roche, AstraZenca, Pfizer, Eisai, and Takeda and has research agreements with Amgen. G.V.L. is a consultant advisor to Aduro, Amgen, Mass-Array, BMS, Merck MSD, Novartis, Roche, OncoSec Medical, Sandoz, and Pierre Fabre. A.M. is a consultant advisor for BMS, Merck MSD, Novartis, Roche, and Pierre Fabre. W.C.D. declares a scientific research agreement with BMS, consulting agreements with Omeros Corp. and Cascadia Drug Development Group, and receipt of speaker’s honoraria from Amgen. A.V. has research agreements with BMS. R.A.S. has received fees for professional services from Novartis Pharma AG, MSD Sharp & Dohme (Australia), NeraCare, AMGEN Inc., BMS, Novartis Pharmaceuticals Australia Pty. Limited, Myriad Genetics GmbH, GlaxoSmithKline Australia. L.F. reported grants from the Swiss National Science Foundation, Swiss Cancer League, Hookipa Pharma, Krebsliga Schweiz, and Novartis Foundation as well as an advisory role for Novartis and BMS. S.E.N. has a research agreement with Servier. M.B., M.J.S., and T.B. have registered a patent for the use of CD226 in cancer immunotherapies (Australian Provisional Patent Application No. 2019900621)., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
15. The NK cell granule protein NKG7 regulates cytotoxic granule exocytosis and inflammation.
- Author
-
Ng SS, De Labastida Rivera F, Yan J, Corvino D, Das I, Zhang P, Kuns R, Chauhan SB, Hou J, Li XY, Frame TCM, McEnroe BA, Moore E, Na J, Engel JA, Soon MSF, Singh B, Kueh AJ, Herold MJ, Montes de Oca M, Singh SS, Bunn PT, Aguilera AR, Casey M, Braun M, Ghazanfari N, Wani S, Wang Y, Amante FH, Edwards CL, Haque A, Dougall WC, Singh OP, Baxter AG, Teng MWL, Loukas A, Daly NL, Cloonan N, Degli-Esposti MA, Uzonna J, Heath WR, Bald T, Tey SK, Nakamura K, Hill GR, Kumar R, Sundar S, Smyth MJ, and Engwerda CR
- Subjects
- Animals, Cells, Cultured, Cytotoxicity, Immunologic, Disease Models, Animal, Exocytosis, Humans, Membrane Proteins genetics, Mice, Mice, Inbred C57BL, Secretory Vesicles metabolism, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Inflammation immunology, Killer Cells, Natural immunology, Leishmania donovani physiology, Leishmaniasis, Visceral immunology, Malaria immunology, Membrane Proteins metabolism, Plasmodium physiology
- Abstract
Immune-modulating therapies have revolutionized the treatment of chronic diseases, particularly cancer. However, their success is restricted and there is a need to identify new therapeutic targets. Here, we show that natural killer cell granule protein 7 (NKG7) is a regulator of lymphocyte granule exocytosis and downstream inflammation in a broad range of diseases. NKG7 expressed by CD4
+ and CD8+ T cells played key roles in promoting inflammation during visceral leishmaniasis and malaria-two important parasitic diseases. Additionally, NKG7 expressed by natural killer cells was critical for controlling cancer initiation, growth and metastasis. NKG7 function in natural killer and CD8+ T cells was linked with their ability to regulate the translocation of CD107a to the cell surface and kill cellular targets, while NKG7 also had a major impact on CD4+ T cell activation following infection. Thus, we report a novel therapeutic target expressed on a range of immune cells with functions in different immune responses.- Published
- 2020
- Full Text
- View/download PDF
16. Tumor CD155 Expression Is Associated with Resistance to Anti-PD1 Immunotherapy in Metastatic Melanoma.
- Author
-
Lepletier A, Madore J, O'Donnell JS, Johnston RL, Li XY, McDonald E, Ahern E, Kuchel A, Eastgate M, Pearson SA, Mallardo D, Ascierto PA, Massi D, Merelli B, Mandala M, Wilmott JS, Menzies AM, Leduc C, Stagg J, Routy B, Long GV, Scolyer RA, Bald T, Waddell N, Dougall WC, Teng MWL, and Smyth MJ
- Subjects
- Aged, Biopsy, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Female, Humans, Immune Checkpoint Inhibitors therapeutic use, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Male, Melanoma genetics, Melanoma mortality, Melanoma secondary, Middle Aged, Programmed Cell Death 1 Receptor antagonists & inhibitors, Progression-Free Survival, Prospective Studies, RNA-Seq, Response Evaluation Criteria in Solid Tumors, Retrospective Studies, Skin pathology, Skin Neoplasms genetics, Skin Neoplasms mortality, Skin Neoplasms pathology, Gene Expression Regulation, Neoplastic immunology, Immune Checkpoint Inhibitors pharmacology, Melanoma drug therapy, Receptors, Virus genetics, Skin Neoplasms drug therapy
- Abstract
Purpose: Resistance to anti-PD1-based immune checkpoint blockade (ICB) remains a problem for the treatment of metastatic melanoma. Tumor cells as well as host myeloid cells can express the immune checkpoint ligand CD155 to regulate immune cell function. However, the effect of tumor CD155 on the immune context of human melanoma has not been well described. This observational study characterizes tumor CD155 ligand expression by metastatic melanoma tumors and correlates results with differences in immune cell features and response to ICB., Experimental Design: Pretreatment tumor specimens, from 155 patients with metastatic melanoma treated with ICB and from 50 patients treated with BRAF/MEK-directed targeted therapy, were assessed for CD155 expression by IHC. Intratumor T-cell features were analyzed using multiplex-immunohistofluorescence for CD8, PD1, and SOX10. Correlations were made between CD155 tumor level and bulk tumor RNA sequencing results, as well as clinical RECIST response and progression-free survival., Results: High pretreatment CD155 tumor levels correlated with high parenchymal PD1
+ CD8+ /CD8+ T-cell ratios (PD1tR ) and poor response to anti-PD1 therapy. In PDL1 negative tumors, high CD155 tumor expression was associated with patients who had poor response to combination anti-PD1/CTLA4 therapy., Conclusions: Our findings are the first to suggest that tumor CD155 supports an increase in the fraction of PD1+ CD8+ T cells in anti-PD1 refractory melanoma tumors and, further, that targeting the CD155 pathway might improve response to anti-PD1 therapy for patients with metastatic melanoma., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
17. Pharmacodynamics of Pre-Operative PD1 checkpoint blockade and receptor activator of NFkB ligand (RANKL) inhibition in non-small cell lung cancer (NSCLC): study protocol for a multicentre, open-label, phase 1B/2, translational trial (POPCORN).
- Author
-
Ahern E, Cubitt A, Ballard E, Teng MWL, Dougall WC, Smyth MJ, Godbolt D, Naidoo R, Goldrick A, and Hughes BGM
- Subjects
- Adult, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung pathology, Chemotherapy, Adjuvant, Clinical Trials, Phase I as Topic, Clinical Trials, Phase II as Topic, Denosumab pharmacology, Denosumab therapeutic use, Female, Humans, Lung drug effects, Lung pathology, Lung surgery, Lung Neoplasms immunology, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Margins of Excision, Multicenter Studies as Topic, Neoplasm Staging, Nivolumab pharmacology, Nivolumab therapeutic use, Pneumonectomy, Programmed Cell Death 1 Receptor antagonists & inhibitors, Progression-Free Survival, RANK Ligand antagonists & inhibitors, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Non-Small-Cell Lung therapy, Lung Neoplasms therapy, Neoadjuvant Therapy methods
- Abstract
Background: Neoadjuvant immunotherapy targeting immune checkpoint programmed death-1 (PD-1) is under investigation in various tumour settings including non-small-cell lung cancer (NSCLC). Preclinical models demonstrate the superior power of the immunotherapy provided in a neoadjuvant (pre-operative) compared with an adjuvant (post-operative) setting to eradicate metastatic disease and induce long-lasting antigen-specific immunity. Novel effective immunotherapy combinations are widely sought in the oncology field, targeting non-redundant mechanisms of immune evasion. A promising combination partner with anti-PD1 in NSCLC is denosumab, a monoclonal antibody blocking receptor activator of NF-κB ligand (RANKL). In preclinical cancer models and in a large retrospective case series in NSCLC, anti-cancer activity has been reported for the combination of immune checkpoint inhibition (ICI) and denosumab. Furthermore, clinical trials of ICI and denosumab are underway in advanced melanoma and clear-cell renal cell carcinoma. However, the mechanism of action of combination anti-PD1 and anti-RANKL is poorly defined., Methods: This open-label multicentre trial will randomise by minimisation 30 patients with resectable stage IA (primary > 2 cm) to IIIA NSCLC to a neoadjuvant treatment regime of either two doses of nivolumab (3 mg/kg every 2 weeks) or two doses of nivolumab (same regimen) plus denosumab (120 mg every 2 weeks, following nivolumab). Each treatment arm is of equal size and will be approximately balanced with respect to histology (squamous vs. non-squamous) and clinical stage (I-II vs. IIIA). All patients will receive surgery for their tumour 2 weeks after the final dose of neoadjuvant therapy. The primary outcome will be translational research to define the tumour-immune correlates of combination therapy compared with monotherapy. Key secondary outcomes will include a comparison of rates of the following between each arm: toxicity, response (pathological and radiological), and microscopically complete resection., Discussion: The POPCORN study provides a unique platform for translational research to determine the mechanism of action of a novel proposed combination immunotherapy for cancer., Trial Registration: Prospectively registered on Australian New Zealand Clinical Trials Registry (ACTRN12618001121257) on 06/07/2018.
- Published
- 2019
- Full Text
- View/download PDF
18. Dual targeting of RANKL and PD-1 with a bispecific antibody improves anti-tumor immunity.
- Author
-
Dougall WC, Roman Aguilera A, and Smyth MJ
- Abstract
Objectives: The addition of RANKL/RANK blockade to immune checkpoint inhibitors (ICIs) such as anti-PD-1/PD-L1 and anti-CTLA4 antibodies is associated with increased anti-tumor immunity in mice. Recent retrospective clinical studies in patients with advanced melanoma and lung cancer suggest the addition of anti-RANKL antibody to ICI increases the overall response rate relative to ICI treatment alone. Based on this rationale, we developed a novel bispecific antibody (BsAb) co-targeting RANKL and PD-1., Methods: We characterized target binding and functional activity of the anti-RANKL/PD-1 BsAb in cell-based assays. Anti-tumor activity was confirmed in experimental lung metastasis models and in mice with established subcutaneously transplanted tumors., Results: The anti-RANKL/PD-1 BsAb retained binding to both RANKL and PD-1 and blocked the interaction with respective counter-structures RANK and PD-L1. The inhibitory effect of anti-RANKL/PD-1 BsAb was confirmed by demonstrating a complete block of RANKL-dependent osteoclast formation. Monotherapy activity of anti-RANKL/PD-1 BsAb was observed in anti-PD-1 resistant tumors and, when combined with anti-CTLA-4 mAb, increased anti-tumor responses. An equivalent or superior anti-tumor response was observed with the anti-RANKL/PD-1 BsAb compared with the combination of parental anti-RANKL plus anti-PD-1 antibodies depending upon the tumor model., Discussion: Mechanistically, the anti-tumor activity of anti-RANKL/PD-1 BsAb required CD8
+ T cells, host PD-1 and IFNγ. Targeting RANKL and PD-1 simultaneously within the tumor microenvironment (TME) improved anti-tumor efficacy compared with combination of two separate mAbs., Conclusion: In summary, the bispecific anti-RANKL/PD-1 antibody demonstrates potent tumor growth inhibition in settings of ICI resistance and represents a novel modality for clinical development in advanced cancer., (© 2019 The Authors. Clinical & Translational Immunology published by John Wiley & Sons Australia, Ltd on behalf of Australian and New Zealand Society for Immunology Inc.)- Published
- 2019
- Full Text
- View/download PDF
19. CD96 Is an Immune Checkpoint That Regulates CD8 + T-cell Antitumor Function.
- Author
-
Mittal D, Lepletier A, Madore J, Aguilera AR, Stannard K, Blake SJ, Whitehall VLJ, Liu C, Bettington ML, Takeda K, Long GV, Scolyer RA, Lan R, Siemers N, Korman A, Teng MWL, Johnston RJ, Dougall WC, and Smyth MJ
- Subjects
- Adoptive Transfer, Animals, Antigens, CD genetics, Cell Line, Tumor, Humans, Male, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Neoplasms therapy, Antigens, CD immunology, CD8-Positive T-Lymphocytes immunology, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms immunology
- Abstract
CD96 is a novel target for cancer immunotherapy shown to regulate NK cell effector function and metastasis. Here, we demonstrated that blocking CD96 suppressed primary tumor growth in a number of experimental mouse tumor models in a CD8
+ T cell-dependent manner. DNAM-1/CD226, Batf3, IL12p35, and IFNγ were also critical, and CD96-deficient CD8+ T cells promoted greater tumor control than CD96-sufficient CD8+ T cells. The antitumor activity of anti-CD96 therapy was independent of Fc-mediated effector function and was more effective in dual combination with blockade of a number of immune checkpoints, including PD-1, PD-L1, TIGIT, and CTLA-4. We consistently observed coexpression of PD-1 with CD96 on CD8+ T lymphocytes in tumor-infiltrating leukocytes both in mouse and human cancers using mRNA analysis, flow cytometry, and multiplex IHF. The combination of anti-CD96 with anti-PD-1 increased the percentage of IFNγ-expressing CD8+ T lymphocytes. Addition of anti-CD96 to anti-PD-1 and anti-TIGIT resulted in superior antitumor responses, regardless of the ability of the anti-TIGIT isotype to engage FcR. The optimal triple combination was also dependent upon CD8+ T cells and IFNγ. Overall, these data demonstrate that CD96 is an immune checkpoint on CD8+ T cells and that blocking CD96 in combination with other immune-checkpoint inhibitors is a strategy to enhance T-cell activity and suppress tumor growth., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
20. The immune checkpoint CD96 defines a distinct lymphocyte phenotype and is highly expressed on tumor-infiltrating T cells.
- Author
-
Lepletier A, Lutzky VP, Mittal D, Stannard K, Watkins TS, Ratnatunga CN, Smith C, McGuire HM, Kemp RA, Mukhopadhyay P, Waddell N, Smyth MJ, Dougall WC, and Miles JJ
- Subjects
- Antigens, CD biosynthesis, Humans, Immunophenotyping, Lymphocyte Activation, Lymphocyte Subsets immunology, Lymphocytes, Tumor-Infiltrating metabolism, Neoplasm Metastasis immunology, Neoplasms immunology, Neoplasms pathology, T-Lymphocytes metabolism, Transcriptome, Antigens, CD immunology, Lymphocytes, Tumor-Infiltrating immunology, T-Lymphocytes immunology
- Abstract
CD96 has recently been shown to be a potent immune checkpoint molecule in mice, but a similar role in humans is not known. In this study, we provide a detailed map of CD96 expression across human lymphocyte lineages, the kinetics of CD96 regulation on T-cell activation and co-expression with other conventional and emerging immune checkpoint molecules. We show that CD96 is predominantly expressed by T cells and has a unique lymphocyte expression profile. CD96
high T cells exhibited distinct effector functions on activation. Of note, CD96 expression was highly correlated with T-cell markers in primary and metastatic human tumors and was elevated on antigen-experienced T cells and tumor-infiltrating lymphocytes. Collectively, these data demonstrate that CD96 may be a promising immune checkpoint to enhance T-cell function against human cancer and infectious disease., (© 2018 Australasian Society for Immunology Inc.)- Published
- 2019
- Full Text
- View/download PDF
21. Receptor activator of NF-κB (RANK)-mediated induction of metastatic spread and association with poor prognosis in renal cell carcinoma.
- Author
-
Steven A, Leisz S, Fussek S, Nowroozizadeh B, Huang J, Branstetter D, Dougall WC, Burchardt M, Belldegrun AS, Seliger B, Pantuck A, and Kroeger N
- Subjects
- Adult, Aged, Carcinoma, Renal Cell metabolism, Cell Line, Tumor, Female, Humans, Kidney Neoplasms metabolism, Male, Middle Aged, Neoplasm Invasiveness pathology, Prognosis, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology, RANK Ligand metabolism, Receptor Activator of Nuclear Factor-kappa B metabolism
- Abstract
Background: Inhibition of the receptor activator of NF-κB ligand (RANKL) has become a standard of care supportive treatment to prevent skeletal related events in cancer patients. Moreover, RANKL inhibition has been implicated with better survival outcome in lung cancer, while RANKL expression induces tumor progression and metastatic spread in vivo in breast cancer. Whether RANK/RANKL may have an impact on the pathogenesis of clear cell renal cell carcinoma (ccRCC) is currently unknown., Patients and Methods: A retrospective tissue micro array (TMA)-study was carried out determining the expression of RANK/RANKL in primary tumors of 306 ccRCC patients. Additionally, 24 ccRCC cell lines were employed for in vitro analyses of the RANK/RANKL axis including cell proliferation, migration and anchorage independent growth., Results: RANK (+) vs. RANK (-) tumors had both worse cancer specific survival (CSS) (6.3 vs. 1.3 years; p < 0.001) and recurrence free survival (RFS) (9.9 vs. 5.8 years; p < 0.001). RANK (+) (HR 2.21; p < 0.001) was an independent prognostic factor for CSS and RFS (HR 4.98; p < 0.001). RANKL treatment resulted in increased proliferation, soft agar growth, and colony formation of RANK (+) RCC cell lines, which could be reversed by treatment with an NF-κB inhibitor and with a combination of osteoprotegrin and RANKL in vitro., Conclusions: RANK is expressed in ccRCC tissue, correlates with clinicopathological features, survival outcome, and when stimulated with RANKL can induce ccRCC progression in vitro. Consequently, RANKL inhibition combined with standard of care treatment may be a promising approach to improve ccRCC patient's survival., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
22. Roles of the RANKL-RANK axis in antitumour immunity - implications for therapy.
- Author
-
Ahern E, Smyth MJ, Dougall WC, and Teng MWL
- Subjects
- Carcinoma, Renal Cell immunology, Denosumab therapeutic use, Humans, Melanoma immunology, Osteogenesis immunology, RANK Ligand antagonists & inhibitors, Randomized Controlled Trials as Topic, Receptor Activator of Nuclear Factor-kappa B antagonists & inhibitors, Tumor Microenvironment immunology, Carcinoma, Renal Cell drug therapy, Melanoma drug therapy, RANK Ligand immunology, Receptor Activator of Nuclear Factor-kappa B immunology
- Abstract
Recognizing that the transformative effects of immunotherapy are currently limited to a minority of patients with cancer, research efforts are increasingly focused on expanding and enhancing clinical responses by combining immunotherapies; the repurposing of existing drugs is an attractive approach, given their well-characterized safety and pharmacokinetic profiles. Receptor activator of nuclear factor-κB (RANK) and the RANK ligand (RANKL) were initially described in the context of T cell-dendritic cell interactions; however, the discovery of an obligate role of RANK signalling in osteoclastogenesis led to the development of the anti-RANKL antibody denosumab for antiresorptive indications, including bone metastases. Randomized clinical trials and post-marketing surveillance studies have established the acceptable safety profile of denosumab. More recently, several case reports involving patients with advanced-stage melanoma have described remarkable responses following concurrent treatment with denosumab and immune-checkpoint inhibitors. Randomized trials assessing similar combinations in patients with melanoma or renal cell carcinoma are now underway. Herein, we discuss the hallmark clinical trials of denosumab in light of possible immunological effects of this agent. We highlight the role of immune cells as sources of RANK and RANKL in the tumour microenvironment and review data on RANKL inhibition in mouse models of cancer. Finally, we describe hypothetical immune-related mechanisms of action, which could be assessed in clinical trials of immune-checkpoint inhibitors and denosumab in patients with cancer.
- Published
- 2018
- Full Text
- View/download PDF
23. TIGIT immune checkpoint blockade restores CD8 + T-cell immunity against multiple myeloma.
- Author
-
Guillerey C, Harjunpää H, Carrié N, Kassem S, Teo T, Miles K, Krumeich S, Weulersse M, Cuisinier M, Stannard K, Yu Y, Minnie SA, Hill GR, Dougall WC, Avet-Loiseau H, Teng MWL, Nakamura K, Martinet L, and Smyth MJ
- Subjects
- Animals, CD8-Positive T-Lymphocytes drug effects, Cells, Cultured, Humans, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Multiple Myeloma etiology, Multiple Myeloma pathology, Programmed Cell Death 1 Receptor immunology, Receptors, Immunologic metabolism, Receptors, Immunologic physiology, Antibodies, Monoclonal pharmacology, CD8-Positive T-Lymphocytes immunology, Multiple Myeloma prevention & control, Programmed Cell Death 1 Receptor antagonists & inhibitors, Receptors, Immunologic antagonists & inhibitors
- Abstract
Immune-based therapies hold promise for the treatment of multiple myeloma (MM), but so far, immune checkpoint blockade targeting programmed cell death protein 1 has not proven effective as single agent in this disease. T-cell immunoglobulin and ITIM domains (TIGIT) is another immune checkpoint receptor known to negatively regulate T-cell functions. In this study, we investigated the therapeutic potential of TIGIT blockade to unleash immune responses against MM. We observed that, in both mice and humans, MM progression was associated with high levels of TIGIT expression on CD8
+ T cells. TIGIT+ CD8+ T cells from MM patients exhibited a dysfunctional phenotype characterized by decreased proliferation and inability to produce cytokines in response to anti-CD3/CD28/CD2 or myeloma antigen stimulation. Moreover, when challenged with Vk*MYC mouse MM cells, TIGIT-deficient mice showed decreased serum monoclonal immunoglobulin protein levels associated with reduced tumor burden and prolonged survival, indicating that TIGIT limits antimyeloma immune responses. Importantly, blocking TIGIT using monoclonal antibodies increased the effector function of MM patient CD8+ T cells and suppressed MM development. Altogether our data provide evidence for an immune-inhibitory role of TIGIT in MM and support the development of TIGIT-blocking strategies for the treatment of MM patients., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
24. An observational study of concomitant immunotherapies and denosumab in patients with advanced melanoma or lung cancer.
- Author
-
Liede A, Hernandez RK, Wade SW, Bo R, Nussbaum NC, Ahern E, Dougall WC, and Smyth MJ
- Abstract
After a case report of profound clinical response in a melanoma patient following treatment with an immune checkpoint inhibitor (ICI) and RANK-ligand inhibitor denosumab, we identified similar patients from electronic health records (EHR) and described patient characteristics and outcomes. This 2017 observational study used Flatiron Health's EHR database from ~255 US cancer clinics. Included were advanced melanoma or non-small-cell lung cancer (NSCLC) patients who received denosumab within 30 days of CTLA-4 (ipilimumab) or PD1 (pembrolizumab, nivolumab) inhibitors start with a minimum of 6 months of follow up. Real-world tumor response (rwTR) was analyzed for scans available up to 30 days after concomitant therapy. Preclinical experiments evaluated sequencing of ICI, denosumab vs monotherapy or control. Melanoma (n = 66) patients received concomitant denosumab/ICI for a mean 4.0 months, 3.1 months for NSCLC (n = 241). Two-thirds of patients had best rwTR evaluable (complete [CR], partial response [PR], stable disease [SD], or disease progression [PD]). Longer mean duration of concomitant exposure was associated with overall response rate (ORR; CR+PR) in melanoma (p = 0.0172), NSCLC (p < .0001), and combined cohorts (p < .0001). The disease control rate (ORR plus SD) was 56% amongst melanoma patients and 58% amongst NSCLC patients. Longer concomitant therapy was associated with increased overall survival, primarily in NSCLC (p < .0001). Preclinical data suggest that ICI initiated before or at same time as denosumab was optimal. Results provide proof-of-concept that rwTR is associated with concomitant denosumab/ICI. Crude survival analyses supported the association of concomitant therapy and improved outcomes outside of clinical trials and warrant comparative study.
- Published
- 2018
- Full Text
- View/download PDF
25. CD155 loss enhances tumor suppression via combined host and tumor-intrinsic mechanisms.
- Author
-
Li XY, Das I, Lepletier A, Addala V, Bald T, Stannard K, Barkauskas D, Liu J, Aguilera AR, Takeda K, Braun M, Nakamura K, Jacquelin S, Lane SW, Teng MW, Dougall WC, and Smyth MJ
- Subjects
- Animals, B7-H1 Antigen genetics, B7-H1 Antigen immunology, CD8-Positive T-Lymphocytes pathology, CTLA-4 Antigen genetics, CTLA-4 Antigen immunology, Cell Line, Tumor, Killer Cells, Natural pathology, Mice, Mice, Inbred BALB C, Mice, Knockout, Neoplasm Proteins immunology, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Receptors, Virus immunology, CD8-Positive T-Lymphocytes immunology, Immunity, Cellular, Killer Cells, Natural immunology, Neoplasm Proteins deficiency, Neoplasms, Experimental immunology, Receptors, Virus deficiency
- Abstract
Critical immune-suppressive pathways beyond programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) require greater attention. Nectins and nectin-like molecules might be promising targets for immunotherapy, since they play critical roles in cell proliferation and migration and exert immunomodulatory functions in pathophysiological conditions. Here, we show CD155 expression in both malignant cells and tumor-infiltrating myeloid cells in humans and mice. Cd155-/- mice displayed reduced tumor growth and metastasis via DNAM-1 upregulation and enhanced effector function of CD8+ T and NK cells, respectively. CD155-deleted tumor cells also displayed slower tumor growth and reduced metastases, demonstrating the importance of a tumor-intrinsic role of CD155. CD155 absence on host and tumor cells exerted an even greater inhibition of tumor growth and metastasis. Blockade of PD-1 or both PD-1 and CTLA4 was more effective in settings in which CD155 was limiting, suggesting the clinical potential of cotargeting PD-L1 and CD155 function.
- Published
- 2018
- Full Text
- View/download PDF
26. Deficiency of host CD96 and PD-1 or TIGIT enhances tumor immunity without significantly compromising immune homeostasis.
- Author
-
Harjunpää H, Blake SJ, Ahern E, Allen S, Liu J, Yan J, Lutzky V, Takeda K, Aguilera AR, Guillerey C, Mittal D, Li XY, Dougall WC, Smyth MJ, and Teng MWL
- Abstract
Multiple non-redundant immunosuppressive pathways co-exist in the tumor microenvironment and their co-targeting can increase clinical responses. Indeed, concurrent blockade of CTLA-4 and PD-1 in patients with advanced melanoma increased clinical responses over monotherapy alone although the frequency and severity of immune related adverse events (irAEs) also increased. Nevertheless, a substantial number of patients still display an innate resistance phenotype and are unresponsive to current approved immunotherapies even when utilized in combination. In this study, we generated Pdcd1
- / - CD96- / - and Tigit- / - CD96- /- mice to investigate how loss of CD96 in combination with PD-1 or TIGIT impacts on immune homeostasis and hence the potential of inducing immune related toxicities following co-targeting of these pairs of receptors. The ability of Pdcd1- / - CD96- / - and Tigit- / - CD96- /- mice to suppress primary tumor growth was also assessed using the MC38 colon carcinoma and SM1WT1 BRAF-mutated melanoma tumor models. Both Pdcd1- / - CD96- / - or Tigit- / - CD96- /- mice displayed no overt perturbations in immune homeostasis over what was previously reported with Pdcd1- / - or Tigit- / - mice even when aged for 22 months. Interestingly, increased suppression of subcutaneous tumor growth and complete responses was seen in Pdcd1- / - CD96- / - mice compared to Pdcd1- / - or CD96- / - mice depending upon the tumor model. In contrast, in these models, growth suppression in Tigit- / - CD96- / - were similar to Tigit- / - or CD96- / - . This enhanced anti-tumor efficacy of Pdcd1- / - CD96- / - appeared to be due to favorable changes in the ratio of CD8+ T cells to T regulatory cells or CD11b+ GR-1hi myeloid cells in the tumor microenvironment. Co-targeting CD96 and PD-1 may increase anti-tumor immunity over targeting PD-1 alone and potentially not induce serious immune-related toxicities and thus appears a promising strategy for clinical development.- Published
- 2018
- Full Text
- View/download PDF
27. RANKL blockade improves efficacy of PD1-PD-L1 blockade or dual PD1-PD-L1 and CTLA4 blockade in mouse models of cancer.
- Author
-
Ahern E, Harjunpää H, O'Donnell JS, Allen S, Dougall WC, Teng MWL, and Smyth MJ
- Abstract
Receptor activator of NF-κB ligand (RANKL) and its receptor RANK, are members of the tumor necrosis factor and receptor superfamilies, respectively. Antibodies targeting RANKL have recently been evaluated in combination with anti-CTLA4 in case reports of human melanoma and mouse models of cancer. However, the efficacy of anti-RANKL in combination with antibodies targeting other immune checkpoint receptors such as PD1 has not been reported. In this study, we demonstrated that blockade of RANKL improves anti-metastatic activity of antibodies targeting PD1/PD-L1 and improves subcutaneous growth suppression in mouse models of melanoma, prostate and colon cancer. Suppression of experimental lung metastasis following combination anti-RANKL with anti-PD1 requires NK cells and IFN-γ, whereas subcutaneous tumor growth suppression with this combination therapy is attenuated in the absence of T cells and IFN-γ. Furthermore, addition of anti-RANKL to anti-PD1 and anti-CTLA4 resulted in superior anti-tumor responses, irrespective of the ability of anti-CTLA4 isotype to engage activating FcR, and concurrent or delayed RANKL blockade was most effective. Early-during-treatment assessment reveals this triple combination therapy compared to dual anti-PD1 and anti-CTLA4 combination therapy further increased the proportion of tumor-infiltrating CD4
+ and CD8+ T cells that can produce both IFN-γ and TNF. Finally, RANKL expression appears to identify tumor-specific CD8+ T cells expressing higher levels of PD1 which can be modulated by anti-PD1. These data set the scene for clinical evaluation of denosumab use in patients receiving contemporary immune checkpoint blockade.- Published
- 2018
- Full Text
- View/download PDF
28. CD96 targeted antibodies need not block CD96-CD155 interactions to promote NK cell anti-metastatic activity.
- Author
-
Roman Aguilera A, Lutzky VP, Mittal D, Li XY, Stannard K, Takeda K, Bernhardt G, Teng MWL, Dougall WC, and Smyth MJ
- Abstract
CD96 is a transmembrane glycoprotein Ig superfamily receptor, expressed on various T cell subsets and NK cells, that interacts with nectin and nectin-like proteins, including CD155/polio virus receptor (PVR). Here, we have compared three rat anti-mouse CD96 mAbs, including two that block CD96-CD155 (3.3 and 6A6) and one that does not block CD96-CD155 (8B10). Using flow cytometry, we demonstrated that both mAbs 3.3 and 6A6 bind to the first Ig domain of mouse CD96 and compete with CD155 binding, while mAb 8B10 binds to the second Ig domain and does not block CD155. While Fc isotype was irrelevant concerning the anti-metastatic activity of 3.3 mAb, in four different experimental metastases models and one spontaneous metastasis model, the relative order of anti-metastatic potency was 6A6 > 3.3 > 8B10. The metastatic burden control of all of the anti-CD96 clones was highly dependent on NK cells and IFN-γ. Consistent with its inability to block CD96-CD155 interactions, 8B10 retained anti-metastatic activity in CD155-deficient mice, whereas 3.3 and 6A6 lost potency in CD155-deficient mice. Furthermore, 8B10 retained most of its anti-metastatic activity in IL-12p35-deficient mice whereas the activity of 3.3 and 6A6 were partially lost. All three mAbs were inactive in CD226-deficient mice. Altogether, these data demonstrate anti-CD96 need not block CD96-CD155 interactions (ie. immune checkpoint blockade) to promote NK cell anti-metastatic activity.
- Published
- 2018
- Full Text
- View/download PDF
29. Co-administration of RANKL and CTLA4 Antibodies Enhances Lymphocyte-Mediated Antitumor Immunity in Mice.
- Author
-
Ahern E, Harjunpää H, Barkauskas D, Allen S, Takeda K, Yagita H, Wyld D, Dougall WC, Teng MWL, and Smyth MJ
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal immunology, Antigens, CD genetics, Antigens, CD immunology, CD8-Positive T-Lymphocytes immunology, CTLA-4 Antigen genetics, Cytokines genetics, Dendritic Cells immunology, Gene Expression Regulation, Neoplastic immunology, Humans, Lymphocytes immunology, Lymphocytes, Tumor-Infiltrating immunology, Melanoma, Experimental genetics, Melanoma, Experimental immunology, Mice, RANK Ligand genetics, Receptor Activator of Nuclear Factor-kappa B immunology, T-Lymphocytes, Regulatory immunology, CTLA-4 Antigen immunology, Immunotherapy, Melanoma, Experimental therapy, RANK Ligand immunology, Receptor Activator of Nuclear Factor-kappa B genetics
- Abstract
Purpose: Novel partners for established immune checkpoint inhibitors in the treatment of cancer are needed to address the problems of primary and acquired resistance. The efficacy of combination RANKL and CTLA4 blockade in antitumor immunity has been suggested by recent case reports in melanoma. Here, we provide a rationale for this combination in mouse models of cancer. Experimental Design: The efficacy and mechanism of a combination of RANKL and CTLA4 blockade was examined by tumor-infiltrating lymphocyte analysis, tumor growth, and metastasis using a variety of neutralizing antibodies and gene-targeted mice. Results: RANKL blockade improved the efficacy of anti-CTLA4 mAbs against solid tumors and experimental metastases, with regulatory T-cell (Treg)-depleting anti-CTLA4 mAbs of the mouse IgG2a isotype showing greatest combinatorial activity. The optimal combination depended on the presence of activating Fc receptors and lymphocytes (NK cells for metastatic disease and predominantly CD8
+ T cells for subcutaneous tumor control), whereas anti-RANKL alone did not require FcR. The significantly higher T-cell infiltration into solid tumors post anti-RANKL and anti-CTLA4 was accompanied by increased T-cell effector function (cytokine polyfunctionality), and anti-RANKL activity occurred independently of Treg depletion. The majority of RANKL expression in tumors was on T cells whereas RANK-expressing cells were mostly tumor-associated macrophages (TAM), with some expression also observed on dendritic cells (DC) and myeloid-derived suppressor cells (MDSC). Conclusions: These results provide a rationale for the further investigation of RANKL-RANK interactions in tumor immunity and a basis for development of translational markers of interest in human clinical trials. Clin Cancer Res; 23(19); 5789-801. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
30. Poor prognosis of patients with triple-negative breast cancer can be stratified by RANK and RANKL dual expression.
- Author
-
Reyes ME, Fujii T, Branstetter D, Krishnamurthy S, Masuda H, Wang X, Reuben JM, Woodward WA, Edwards BJ, Hortobagyi GN, Tripathy D, Dougall WC, Eckhardt BL, and Ueno NT
- Subjects
- Adult, Aged, Biomarkers, Tumor genetics, Disease-Free Survival, Female, Gene Expression Regulation, Neoplastic, Humans, Kaplan-Meier Estimate, Middle Aged, Triple Negative Breast Neoplasms classification, Triple Negative Breast Neoplasms pathology, Prognosis, RANK Ligand genetics, Receptor Activator of Nuclear Factor-kappa B genetics, Triple Negative Breast Neoplasms genetics
- Abstract
Purpose: As clinical studies have correlated RANK expression levels with survival in breast cancer, and that RANK signaling is dependent on its cognate ligand RANKL, we hypothesized that dual protein expression further stratifies the poor outcome in TNBC., Methods: RANK mRNA and protein expression was evaluated in TNBC using genomic databases, cell lines and in a tissue microarray of curated primary tumor samples derived from 87 patients with TNBC. RANK expression was evaluated either by Mann-Whitney U test on log-normalized gene expression data or by Student's t test on FACS data. Analysis of RANK and RANKL immunostaining was calculated by H-score, and correlations to clinical factors performed using χ
2 or Fisher's exact test. Associations with RFS and OS were assessed using univariate and multivariate Cox proportional hazard models. Survival estimates were generated using the Kaplan-Meier method., Results: In three distinct datasets spanning 684 samples, RANK mRNA expression was higher in primary tumors derived from TNBC patients than from those with other molecular subtypes (P < 0.01). Cell surface-localized RANK protein was consistently higher in TNBC cell lines (P = 0.037). In clinical samples, TNBC patients that expressed both RANK and RANKL proteins had significantly worse RFS (P = 0.0032) and OS (P = 0.004) than patients with RANK-positive, RANKL-negative tumors. RANKL was an independent, poor prognostic factor for RFS (P = 0.04) and OS (P = 0.01) in multivariate analysis in samples that expressed both RANK and RANKL., Conclusions: RANK and RANKL co-expression is associated with poor RFS and OS in patients with TNBC.- Published
- 2017
- Full Text
- View/download PDF
31. Distribution of RANK and RANK Ligand in Normal Human Tissues as Determined by an Optimized Immunohistochemical Method.
- Author
-
Taylor CR, Branstetter D, Manna E, Dougall WC, Bussiere J, and Johnson CW
- Subjects
- Animals, Humans, Immunohistochemistry standards, Mice, RANK Ligand chemistry, Receptor Activator of Nuclear Factor-kappa B chemistry, Tissue Distribution, Antibodies metabolism, Immunohistochemistry methods, RANK Ligand metabolism, Receptor Activator of Nuclear Factor-kappa B metabolism
- Abstract
The expression and tissue distribution of RANK (Receptor Activator of Nuclear Factor κ B) and RANK Ligand (RANKL) are of critical interest in relation to efficacy and safety of antibodies against RANK or RANKL that are approved or under consideration as potential therapeutic agents. Data from the literature using protein or mRNA analyses of rodent and human tissues or immunohistochemical (IHC) studies with a variety of antibodies and methods have provided some background of the distribution of RANK and RANKL but have yielded inconsistent findings. The present study reports the generation of carefully validated antibodies to RANK and RANKL and the development of an optimized IHC method, with confirmatory data from 2 well-validated alternative protocols that were developed and performed in separate laboratories at USC and at Amgen. Tissue expression of RANK and RANKL is reported for the optimized IHC assay.
- Published
- 2017
- Full Text
- View/download PDF
32. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy.
- Author
-
Dougall WC, Kurtulus S, Smyth MJ, and Anderson AC
- Subjects
- Animals, Antigens, CD immunology, Antigens, Differentiation, T-Lymphocyte metabolism, CTLA-4 Antigen immunology, CTLA-4 Antigen metabolism, Humans, Lymphocyte Activation, Neoplasms immunology, Programmed Cell Death 1 Receptor immunology, Programmed Cell Death 1 Receptor metabolism, Receptors, Immunologic immunology, Signal Transduction, Tumor Escape, Antibodies, Monoclonal therapeutic use, Antigens, CD metabolism, Immunotherapy methods, Killer Cells, Natural immunology, Neoplasms therapy, Receptors, Immunologic metabolism, T-Lymphocytes immunology
- Abstract
While therapies targeting the co-inhibitory or immune checkpoint receptors PD-1 and CTLA-4 have shown remarkable success in many cancers, not all patients benefit from these therapies. This has catalyzed enormous interest in the targeting of other immune checkpoint receptors. In this regard, TIGIT and CD96 have recently entered the limelight as novel immune checkpoint receptor targets. TIGIT and CD96 together with the co-stimulatory receptor CD226 form a pathway that is analogous to the CD28/CTLA-4 pathway, in which shared ligands and differential receptor:ligand affinities fine-tune the immune response. Although the roles of TIGIT and CD96 as immune checkpoint receptors in T cell and natural killer cell biology are just beginning to be uncovered, accumulating data support the targeting of these receptors for improving anti-tumor immune responses. A clear understanding of the immune cell populations regulated by TIGIT and CD96 is key to the design of immunotherapies that target these receptors in combination with other existing immune checkpoint blockade therapies., (© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
33. Molecular Pathways: Targeting CD96 and TIGIT for Cancer Immunotherapy.
- Author
-
Blake SJ, Dougall WC, Miles JJ, Teng MW, and Smyth MJ
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal therapeutic use, Humans, Immunotherapy methods, Killer Cells, Natural drug effects, Killer Cells, Natural immunology, Neoplasms metabolism, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory immunology, Antigens, CD metabolism, Neoplasms immunology, Neoplasms therapy, Receptors, Immunologic metabolism
- Abstract
The receptors CD96 and TIGIT are expressed on the surface of T and natural killer (NK) cells, and recent studies suggest both play important inhibitory roles in immune function. CD96 has been shown to modulate immune cell activity in mice, with Cd96
- / - mice displaying hypersensitive NK-cell responses to immune challenge and significant tumor resistance. TIGIT overexpression has been shown to reduce NK-cell-mediated cytotoxicity. TIGIT is also upregulated on T cells during cancer and chronic viral infection, with expression associated with effector T-cell exhaustion and increased regulatory T-cell suppression. The counterbalance between the putative inhibitory CD96 and TIGIT receptors and the activating receptor, CD226, offers unique strategies for immuno-oncology drug development. Blocking CD96 or TIGIT with mAbs has been shown to improve tumor control in mice, in particular when used in combination with PD-1/PD-L1 blockade. These results have highlighted these pathways as promising new targets for immune modulation. This review will examine the rationale behind targeting CD96 and TIGIT, and discuss the potential approaches in translating these preclinical findings into novel clinical agents. Clin Cancer Res; 22(21); 5183-8. ©2016 AACR., (©2016 American Association for Cancer Research.)- Published
- 2016
- Full Text
- View/download PDF
34. RANK Signaling Blockade Reduces Breast Cancer Recurrence by Inducing Tumor Cell Differentiation.
- Author
-
Yoldi G, Pellegrini P, Trinidad EM, Cordero A, Gomez-Miragaya J, Serra-Musach J, Dougall WC, Muñoz P, Pujana MA, Planelles L, and González-Suárez E
- Subjects
- Animals, Apoptosis drug effects, Ataxin-1 analysis, Cell Differentiation drug effects, Docetaxel, Female, Humans, Lung Neoplasms prevention & control, Lung Neoplasms secondary, Mammary Neoplasms, Experimental pathology, Mammary Tumor Virus, Mouse, Mice, Mice, Inbred C57BL, Neoplastic Stem Cells drug effects, RANK Ligand antagonists & inhibitors, RANK Ligand pharmacology, Receptor Activator of Nuclear Factor-kappa B physiology, Taxoids pharmacology, Transcription Factor AP-2 physiology, Mammary Neoplasms, Experimental prevention & control, Neoplasm Recurrence, Local prevention & control, Receptor Activator of Nuclear Factor-kappa B antagonists & inhibitors, Signal Transduction physiology
- Abstract
RANK expression is associated with poor prognosis in breast cancer even though its therapeutic potential remains unknown. RANKL and its receptor RANK are downstream effectors of the progesterone signaling pathway. However, RANK expression is enriched in hormone receptor negative adenocarcinomas, suggesting additional roles for RANK signaling beyond its hormone-dependent function. Here, to explore the role of RANK signaling once tumors have developed, we use the mouse mammary tumor virus-Polyoma Middle T (MMTV-PyMT), which mimics RANK and RANKL expression patterns seen in human breast adenocarcinomas. Complementary genetic and pharmacologic approaches demonstrate that therapeutic inhibition of RANK signaling drastically reduces the cancer stem cell pool, decreases tumor and metastasis initiation, and enhances sensitivity to chemotherapy. Mechanistically, genome-wide expression analyses show that anti-RANKL therapy promotes lactogenic differentiation of tumor cells. Moreover, RANK signaling in tumor cells negatively regulates the expression of Ap2 transcription factors, and enhances the Wnt agonist Rspo1 and the Sca1-population, enriched in tumor-initiating cells. In addition, we found that expression of TFAP2B and the RANK inhibitor, OPG, in human breast cancer correlate and are associated with relapse-free tumors. These results support the use of RANKL inhibitors to reduce recurrence and metastasis in breast cancer patients based on its ability to induce tumor cell differentiation. Cancer Res; 76(19); 5857-69. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
35. RANK ligand as a potential target for breast cancer prevention in BRCA1-mutation carriers.
- Author
-
Nolan E, Vaillant F, Branstetter D, Pal B, Giner G, Whitehead L, Lok SW, Mann GB, Rohrbach K, Huang LY, Soriano R, Smyth GK, Dougall WC, Visvader JE, and Lindeman GJ
- Subjects
- Animals, Bone Density Conservation Agents therapeutic use, Breast metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, Carcinogenesis genetics, DNA Repair, Denosumab therapeutic use, Disease Models, Animal, Female, Flow Cytometry, Heterozygote, Humans, Immunohistochemistry, Mice, Molecular Targeted Therapy, Mutation, Neoplasm Transplantation, Organoids metabolism, Pilocarpine analogs & derivatives, Prophylactic Mastectomy, RANK Ligand metabolism, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells metabolism, Tumor Suppressor Proteins, Xenograft Model Antitumor Assays, BRCA1 Protein genetics, Bone Density Conservation Agents pharmacology, Breast drug effects, Breast Neoplasms prevention & control, Carcinogenesis drug effects, Cell Proliferation drug effects, Denosumab pharmacology, Organoids drug effects, RANK Ligand antagonists & inhibitors, Receptor Activator of Nuclear Factor-kappa B metabolism, Stem Cells drug effects
- Abstract
Individuals who have mutations in the breast-cancer-susceptibility gene BRCA1 (hereafter referred to as BRCA1-mutation carriers) frequently undergo prophylactic mastectomy to minimize their risk of breast cancer. The identification of an effective prevention therapy therefore remains a 'holy grail' for the field. Precancerous BRCA1(mut/+) tissue harbors an aberrant population of luminal progenitor cells, and deregulated progesterone signaling has been implicated in BRCA1-associated oncogenesis. Coupled with the findings that tumor necrosis factor superfamily member 11 (TNFSF11; also known as RANKL) is a key paracrine effector of progesterone signaling and that RANKL and its receptor TNFRSF11A (also known as RANK) contribute to mammary tumorigenesis, we investigated a role for this pathway in the pre-neoplastic phase of BRCA1-mutation carriers. We identified two subsets of luminal progenitors (RANK(+) and RANK(-)) in histologically normal tissue of BRCA1-mutation carriers and showed that RANK(+) cells are highly proliferative, have grossly aberrant DNA repair and bear a molecular signature similar to that of basal-like breast cancer. These data suggest that RANK(+) and not RANK(-) progenitors are a key target population in these women. Inhibition of RANKL signaling by treatment with denosumab in three-dimensional breast organoids derived from pre-neoplastic BRCA1(mut/+) tissue attenuated progesterone-induced proliferation. Notably, proliferation was markedly reduced in breast biopsies from BRCA1-mutation carriers who were treated with denosumab. Furthermore, inhibition of RANKL in a Brca1-deficient mouse model substantially curtailed mammary tumorigenesis. Taken together, these findings identify a targetable pathway in a putative cell-of-origin population in BRCA1-mutation carriers and implicate RANKL blockade as a promising strategy in the prevention of breast cancer.
- Published
- 2016
- Full Text
- View/download PDF
36. Rankl Impairs Lactogenic Differentiation Through Inhibition of the Prolactin/Stat5 Pathway at Midgestation.
- Author
-
Cordero A, Pellegrini P, Sanz-Moreno A, Trinidad EM, Serra-Musach J, Deshpande C, Dougall WC, Pujana MA, and González-Suárez E
- Subjects
- Animals, Cell Proliferation genetics, DNA-Binding Proteins biosynthesis, Female, Gene Expression Regulation, Developmental, Janus Kinase 2 biosynthesis, Janus Kinase 2 genetics, Lactation genetics, Mammary Glands, Animal growth & development, Mammary Glands, Animal metabolism, Mice, Mice, Knockout, Pregnancy, Progesterone genetics, Progesterone metabolism, Prolactin metabolism, RANK Ligand biosynthesis, STAT5 Transcription Factor biosynthesis, Signal Transduction, Transcription Factors biosynthesis, Cell Differentiation genetics, DNA-Binding Proteins genetics, Prolactin genetics, RANK Ligand genetics, STAT5 Transcription Factor genetics, Transcription Factors genetics
- Abstract
Prolactin and progesterone both orchestrate the proliferation and differentiation of the mammary gland during gestation. Differentiation of milk secreting alveoli depends on the presence of prolactin receptor, the downstream Jak2-Stat5 pathway and the transcription factor Elf5. A strict regulation of Rank signaling is essential for the differentiation of the mammary gland and in particular for alveolar commitment. Impaired alveologenesis and lactation failure are observed in both, knockout and Rank overexpressing mice; however, the underlying molecular mechanism responsible for these phenotypes remains largely unknown. Using genome-wide expression analyses and functional studies, we show here that Rankl (RL) exposure leads to impaired secretory differentiation of alveolar cells not only in MMTV-RANK but also in wild-type (WT) mammary acini. Conversely, pharmacological blockage of Rank signaling at midgestation in WT mice leads to precocious and exacerbated lactogenesis. Mechanistically, RL negatively regulates Stat5 phosphorylation and Elf5 expression at the onset of lactogenesis. Continuous RL exposure leads to the expansion of basal and bipotent cells in WT and MMTV-RANK acini. Overall, we demonstrate that enhanced Rank signaling impairs secretory differentiation during pregnancy by inhibition of the prolactin/p-Stat5 pathway., (© 2015 AlphaMed Press.)
- Published
- 2016
- Full Text
- View/download PDF
37. Osteoprotegerin (OPG), The Endogenous Inhibitor of Receptor Activator of NF-κB Ligand (RANKL), is Dysregulated in BRCA Mutation Carriers.
- Author
-
Widschwendter M, Burnell M, Fraser L, Rosenthal AN, Philpott S, Reisel D, Dubeau L, Cline M, Pan Y, Yi PC, Gareth Evans D, Jacobs IJ, Menon U, Wood CE, and Dougall WC
- Subjects
- Animals, Case-Control Studies, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Female, Gene Expression Regulation, Neoplastic, Germ-Line Mutation, Hereditary Breast and Ovarian Cancer Syndrome blood, Hereditary Breast and Ovarian Cancer Syndrome genetics, Hereditary Breast and Ovarian Cancer Syndrome metabolism, Hormones blood, Humans, Macaca fascicularis, Osteoprotegerin blood, Patient Outcome Assessment, Postmenopause, RANK Ligand blood, Risk Factors, Genes, BRCA1, Genes, BRCA2, Heterozygote, Mutation, Osteoprotegerin metabolism, RANK Ligand metabolism
- Abstract
Breast cancer development in BRCA1/2 mutation carriers is a net consequence of cell-autonomous and cell nonautonomous factors which may serve as excellent targets for cancer prevention. In light of our previous data we sought to investigate the consequences of the BRCA-mutation carrier state on RANKL/osteoprotegerin (OPG) signalling. We analysed serum levels of RANKL, OPG, RANKL/OPG complex, oestradiol (E2), and progesterone (P) during menstrual cycle progression in 391 BRCA1/2-mutation carriers and 782 noncarriers. These studies were complemented by analyses of RANKL and OPG in the serum and mammary tissues of female cynomolgus macaques (n = 88) and serum RANKL and OPG in postmenopausal women (n = 150). BRCA-mutation carriers had lower mean values of free serum OPG in particular in BRCA1-mutation carriers (p = 0.018) compared with controls. Among BRCA1/2 mutation carriers, lower OPG levels were associated with germline mutation locations known to confer an increased breast cancer risk (p = 0.003). P is associated with low OPG levels in serum and tissue, particularly in BRCA-mutation carriers (rho = - 0.216; p = 0.002). Serum OPG levels were inversely correlated (rho = - 0.545, p < 0.001) with mammary epithelial proliferation measured by Ki67 expression and increased (p = 0.01) in postmenopause. The P-RANKL/OPG system is dysregulated in BRCA-mutation carriers. These and previously published data provide a strong rationale for further investigation of antiprogestogens or an anti-RANKL antibody such as denosumab for breast cancer prevention.
- Published
- 2015
- Full Text
- View/download PDF
38. RANK and RANK ligand expression in primary human osteosarcoma.
- Author
-
Branstetter D, Rohrbach K, Huang LY, Soriano R, Tometsko M, Blake M, Jacob AP, and Dougall WC
- Abstract
Receptor activator of nuclear factor kappa-B ligand (RANKL) is an essential mediator of osteoclast formation, function and survival. In patients with solid tumor metastasis to the bone, targeting the bone microenvironment by inhibition of RANKL using denosumab, a fully human monoclonal antibody (mAb) specific to RANKL, has been demonstrated to prevent tumor-induced osteolysis and subsequent skeletal complications. Recently, a prominent functional role for the RANKL pathway has emerged in the primary bone tumor giant cell tumor of bone (GCTB). Expression of both RANKL and RANK is extremely high in GCTB tumors and denosumab treatment was associated with tumor regression and reduced tumor-associated bone lysis in GCTB patients. In order to address the potential role of the RANKL pathway in another primary bone tumor, this study assessed human RANKL and RANK expression in human primary osteosarcoma (OS) using specific mAbs, validated and optimized for immunohistochemistry (IHC) or flow cytometry. Our results demonstrate RANKL expression was observed in the tumor element in 68% of human OS using IHC. However, the staining intensity was relatively low and only 37% (29/79) of samples exhibited≥10% RANKL positive tumor cells. RANK expression was not observed in OS tumor cells. In contrast, RANK expression was clearly observed in other cells within OS samples, including the myeloid osteoclast precursor compartment, osteoclasts and in giant osteoclast cells. The intensity and frequency of RANKL and RANK staining in OS samples were substantially less than that observed in GCTB samples. The observation that RANKL is expressed in OS cells themselves suggests that these tumors may mediate an osteoclastic response, and anti-RANKL therapy may potentially be protective against bone pathologies in OS. However, the absence of RANK expression in primary human OS cells suggests that any autocrine RANKL/RANK signaling in human OS tumor cells is not operative, and anti-RANKL therapy would not directly affect the tumor.
- Published
- 2015
- Full Text
- View/download PDF
39. Aberrant Activation of the RANK Signaling Receptor Induces Murine Salivary Gland Tumors.
- Author
-
Szwarc MM, Kommagani R, Jacob AP, Dougall WC, Ittmann MM, and Lydon JP
- Subjects
- Animals, Carcinogenesis, Cell Proliferation, Mice, Mice, Transgenic, Salivary Gland Neoplasms metabolism, Salivary Gland Neoplasms pathology, Signal Transduction, RANK Ligand metabolism, Receptor Activator of Nuclear Factor-kappa B metabolism, Salivary Gland Neoplasms etiology
- Abstract
Unlike cancers of related exocrine tissues such as the mammary and prostate gland, diagnosis and treatment of aggressive salivary gland malignancies have not markedly advanced in decades. Effective clinical management of malignant salivary gland cancers is undercut by our limited knowledge concerning the key molecular signals that underpin the etiopathogenesis of this rare and heterogeneous head and neck cancer. Without knowledge of the critical signals that drive salivary gland tumorigenesis, tumor vulnerabilities cannot be exploited that allow for targeted molecular therapies. This knowledge insufficiency is further exacerbated by a paucity of preclinical mouse models (as compared to other cancer fields) with which to both study salivary gland pathobiology and test novel intervention strategies. Using a mouse transgenic approach, we demonstrate that deregulation of the Receptor Activator of NFkB Ligand (RANKL)/RANK signaling axis results in rapid tumor development in all three major salivary glands. In line with its established role in other exocrine gland cancers (i.e., breast cancer), the RANKL/RANK signaling axis elicits an aggressive salivary gland tumor phenotype both at the histologic and molecular level. Despite the ability of this cytokine signaling axis to drive advanced stage disease within a short latency period, early blockade of RANKL/RANK signaling markedly attenuates the development of malignant salivary gland neoplasms. Together, our findings have uncovered a tumorigenic role for RANKL/RANK in the salivary gland and suggest that targeting this pathway may represent a novel therapeutic intervention approach in the prevention and/or treatment of this understudied head and neck cancer.
- Published
- 2015
- Full Text
- View/download PDF
40. RANK-ligand (RANKL) expression in young breast cancer patients and during pregnancy.
- Author
-
Azim HA Jr, Peccatori FA, Brohée S, Branstetter D, Loi S, Viale G, Piccart M, Dougall WC, Pruneri G, and Sotiriou C
- Subjects
- Adult, Age Factors, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms metabolism, Breast Neoplasms mortality, Breast Neoplasms pathology, Disease-Free Survival, Female, Gene Expression Profiling, Humans, Immunohistochemistry, Middle Aged, Neoplasm Grading, Neoplasm Metastasis, Pregnancy, Prognosis, RANK Ligand metabolism, Receptor Activator of Nuclear Factor-kappa B genetics, Receptor Activator of Nuclear Factor-kappa B metabolism, Signal Transduction, Tumor Burden, Breast Neoplasms genetics, Gene Expression Regulation, Neoplastic, RANK Ligand genetics
- Abstract
Introduction: RANKL is important in mammary gland development during pregnancy and mediates the initiation and progression of progesterone-induced breast cancer. No clinical data are available on the effect of pregnancy on RANK/RANKL expression in young breast cancer patients., Methods: We used our previously published dataset of 65 pregnant and 130 matched young breast cancer patients with full clinical, pathological, and survival information. 85% of patients had available transcriptomic data as well. RANK/RANKL expression by immunohistochemistry using H-score on the primary tumor and adjacent normal tissue was performed. We examined the difference in expression of RANK/RANKL between pregnant and non-pregnant patients and their association with clinicopathological features and prognosis. We also evaluated genes and pathways associated with RANK/RANKL expression on primary tumors., Results: RANKL but not RANK expression was more prevalent in the pregnant group, both on the tumor and adjacent normal tissue, independent of other clinicopathological factors (both P <0.001). 18.7% of pregnant and 5.3% of non-pregnant patients had tumors showing ≥10% of cells with 3+ RANKL expression. RANKL expression was significantly higher in progesterone receptor-positive, and luminal A-like tumors, with negative correlation with Ki-67 (all P <0.001). On the contrary, RANK expression was higher in triple negative tumors (P <0.001). Using false discovery rate <0.05, 151 and 1,207 genes were significantly correlated with tumor-expressed RANKL and RANK expression by immunohistochemistry, respectively. High RANKL expression within primary tumor was associated with pathways related to mammary gland development, bone resorption, T-cell proliferation and regulation of chemotaxis, while RANK expression was associated with immune response and proliferation pathways. At a median follow-up of 65 months, neither RANK nor RANKL expression within tumor was associated with disease free survival in pregnant or non-pregnant group., Conclusions: Pregnancy increases RANKL expression both in normal breast and primary tumors. These results could guide further development of RANKL-targeted therapy.
- Published
- 2015
- Full Text
- View/download PDF
41. RANKL expression in normal and malignant breast tissue responds to progesterone and is up-regulated during the luteal phase.
- Author
-
Hu H, Wang J, Gupta A, Shidfar A, Branstetter D, Lee O, Ivancic D, Sullivan M, Chatterton RT Jr, Dougall WC, and Khan SA
- Subjects
- Adult, Aged, Biopsy, Fine-Needle, Breast Neoplasms pathology, Carrier Proteins biosynthesis, Estradiol blood, Female, Gene Expression Regulation, Neoplastic, Humans, Iodide Peroxidase biosynthesis, Luteal Phase genetics, Mammary Glands, Human metabolism, Mammary Glands, Human pathology, Menstrual Cycle metabolism, Middle Aged, Primary Cell Culture, RANK Ligand blood, RANK Ligand genetics, Iodothyronine Deiodinase Type II, Breast Neoplasms genetics, Carcinogenesis, Progesterone blood, RANK Ligand biosynthesis
- Abstract
The receptor activator of nuclear factor-κB ligand (RANKL) acts as a paracrine factor in progesterone-induced mammary epithelial proliferation and tumorigenesis. This evidence comes mainly from mouse models. Our aim was to examine whether RANKL expression in human normal and malignant breast is under the control of progesterone throughout the menstrual cycle. Breast epithelial samples were obtained by random fine needle aspiration (rFNA) of the contralateral unaffected breasts (CUB) of 18 breast cancer patients, with simultaneous serum hormone measurements. Genes correlated with serum progesterone levels were identified through Illumina microarray analysis. Validation was performed using qRT-PCR in rFNA samples from CUB of an additional 53 women and using immunohistochemistry in tissue microarrays of 61 breast cancer samples. Expression of RANKL, DIO2, and MYBPC1 was correlated with serum progesterone in CUB, and was significantly higher in luteal phase. RANKL and MYBPC1 mRNA expression were highly correlated between CUB and matched tumor samples. RANKL protein expression was also significantly increased in the luteal phase and highly correlated with serum progesterone levels in cancer samples, especially in hormone receptor positive tumors. The regulatory effects of progesterone on the expression of RANKL, DIO2, and MYBPC1 were confirmed in three-dimensional cultures of normal breast organoids. In normal breast and in breast cancer, RANKL mRNA and protein expression fluctuate with serum progesterone with highest levels in the luteal phase, suggesting that RANKL is a modulator of progesterone signaling in normal and malignant breast tissue and a potential biomarker of progesterone action and blockade.
- Published
- 2014
- Full Text
- View/download PDF
42. RANK expression as a prognostic and predictive marker in breast cancer.
- Author
-
Pfitzner BM, Branstetter D, Loibl S, Denkert C, Lederer B, Schmitt WD, Dombrowski F, Werner M, Rüdiger T, Dougall WC, and von Minckwitz G
- Subjects
- Breast Neoplasms mortality, Disease-Free Survival, Female, Humans, Middle Aged, Neoadjuvant Therapy, Predictive Value of Tests, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Breast Neoplasms pathology, RANK Ligand metabolism, Receptor Activator of Nuclear Factor-kappa B metabolism
- Abstract
RANK ligand (RANKL) is crucial for the development of mouse mammary glands during pregnancy. RANKL functions as a major paracrine effector of the mitogenic action of progesterone in mammary epithelium via its receptor RANK and has a role in expansion and regenerative potential of mammary stem cells. Pharmacologic inhibition of RANKL attenuates the development of mammary carcinoma and inhibits metastatic progression in multiple mouse models. Primary breast carcinoma samples from the neoadjuvant GeparTrio study were analyzed to correlate the expression of human RANK and RANKL with pathological complete response (pCR), disease-free (DFS), and overall (OS) survival. Pre-treatment FFPE core biopsies (n = 601) were analyzed for percentage and intensity of immunohistochemical RANK and RANKL expression. Antibodies against human RANK (N-1H8; Amgen) and human RANKL (M366; Amgen) were used. RANK protein was expressed in 160 (27 %) patients. Increased RANK expression was observed in 14.5 % of patients and correlated with high tumor grade (p < 0.023) and negative hormone receptor (HR) status (p < 0.001). Patients with high RANK expression showed a higher pCR rate (23.0 % vs. 12.6 %, p = 0.010), shorter DFS (p = 0.038), and OS (p = 0.011). However, prognostic and predictive information was not an independent parameter. Only 6 % of samples expressed RANKL, which was not correlated with any clinical features. Higher RANK expression in the primary tumor is associated with a higher sensitivity to chemotherapy, but also a higher risk of relapse and death. Our study provides a basis for further exploration of the antitumor activity of clinical antibodies against RANKL.
- Published
- 2014
- Full Text
- View/download PDF
43. Targeting RANKL in metastasis.
- Author
-
Dougall WC, Holen I, and González Suárez E
- Abstract
Acting through its cognate receptor, receptor activator of nuclear factor-κB (RANK), RANK ligand (RANKL) is an essential mediator of osteoclast function and survival. Preclinical data have now firmly established that blockade of tumor-induced osteoclastogenesis by RANKL inhibition will not only protect against bone destruction but will also inhibit the progression of established bone metastases and delay the formation of de novo bone metastases in cancer models. In patients with bone metastases, skeletal complications are driven by increased osteoclastic activity and may result in pathological fractures, spinal cord compression and the need for radiotherapy to the bone or orthopedic surgery (collectively known as skeletal-related events (SREs)). Denosumab, a fully human monoclonal antibody against RANKL, has been demonstrated to prevent or delay SREs in patients with solid tumors that have metastasized to bone. In addition to its central role in tumor-induced osteolysis, bone destruction and skeletal tumor progression, there is emerging evidence for direct pro-metastatic effects of RANKL, independent of osteoclasts. For example, RANKL also stimulates metastasis via activity on RANK-expressing cancer cells, resulting in increased invasion and migration. Pharmacological inhibition of RANKL may also reduce bone and lung metastasis through blockade of the direct action of RANKL on metastatic cells. This review describes these distinct but potentially overlapping mechanisms by which RANKL may promote metastases.
- Published
- 2014
- Full Text
- View/download PDF
44. RANKL inhibition blocks osteolytic lesions and reduces skeletal tumor burden in models of non-small-cell lung cancer bone metastases.
- Author
-
Miller RE, Jones JC, Tometsko M, Blake ML, and Dougall WC
- Subjects
- Animals, Anticoagulants pharmacology, Bone Neoplasms metabolism, Bone Neoplasms secondary, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Female, Humans, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mice, Mice, Nude, Osteoprotegerin immunology, Survival Rate, Tumor Burden, Tumor Cells, Cultured, Bone Neoplasms prevention & control, Carcinoma, Non-Small-Cell Lung prevention & control, Enoxaparin pharmacology, Lung Neoplasms prevention & control, Osteolysis drug therapy, Osteoprotegerin metabolism, RANK Ligand antagonists & inhibitors
- Abstract
Introduction: Bone metastasis is a serious complication in patients with lung cancer, occurring in up to 40% of patients. Tumor cell-mediated osteolysis occurs ultimately through induction of RANK ligand (RANKL) within the bone stroma although this hypothesis has not been tested extensively in the setting of non-small-cell lung cancer (NSCLC). By using two novel NSCLC bone metastasis mouse models, we examined the effects of RANKL inhibition on osteolysis and tumor progression., Methods: We treated mice bearing skeletal NSCLC tumors with osteoprotegerin-Fc (OPG-Fc) to assess whether osteoclast inhibition through RANKL inhibition would affect bone metastases at early or late stages of bone colonization. Progression of skeletal tumor was determined by radiography, longitudinal bioluminescent imaging, and histological analyses., Results: OPG-Fc reduced development and progression of radiographically evident osteolytic lesions and also significantly reduced skeletal tumor progression in both NSCLC bone metastasis models. In the H1299 human NSCLC bone metastasis model, OPG-Fc plus docetaxel in combination resulted in significantly greater inhibition of skeletal tumor growth compared with either single agent alone. The observed ability of RANKL inhibition to reduce NSCLC osteolytic bone destruction or skeletal tumor burden was associated with decreases in tumor-associated osteoclasts., Conclusions: These results demonstrate that RANKL is required for the development of tumor-induced osteolytic bone destruction caused by NSCLC cells in vivo. RANKL inhibition also reduced skeletal tumor burden, presumably through the indirect mechanism of blocking tumor-induced osteoclastogenesis and resultant production of growth factors and calcium from the bone microenvironment. RANKL inhibition also provided an additive benefit to docetaxel treatment by augmenting the reduction of tumor burden.
- Published
- 2014
- Full Text
- View/download PDF
45. RANK expression on breast cancer cells promotes skeletal metastasis.
- Author
-
Blake ML, Tometsko M, Miller R, Jones JC, and Dougall WC
- Subjects
- Breast Neoplasms pathology, Cell Line, Tumor, Female, Humans, Bone Neoplasms secondary, Breast Neoplasms metabolism, RANK Ligand metabolism
- Abstract
RANK ligand (RANKL), acting through its cognate receptor RANK, is a key factor for bone remodeling and metastasis by regulating the differentiation, survival and activation of osteoclasts. RANKL is also crucial for the development of mouse mammary glands during pregnancy and has been recently linked to the etiology of breast cancer via its direct activity on RANK-expressing normal or transformed breast epithelial cells, leading to increased mitogenesis, enhanced regenerative potential of mammary stem cells, and increased invasion and migration. We demonstrate that higher RANK expression in MDA-MB-231 breast cancer cells (MDA-231-RANK cells) is sufficient to confer a significantly greater metastatic growth rate in the bone compared with MDA-MB-231 cells which do not express high levels of RANK. Blockade of osteoclastic bone resorption, achieved with treatment by either RANKL inhibition or zoledronic acid, did reduce skeletal tumor progression of MDA-231-RANK cells suggesting that the vicious cycle contributes to metastatic growth. However, RANKL inhibition reduced skeletal growth of MDA-231-RANK tumors to a significantly greater extent than zoledronic acid, indicating that skeletal growth of RANK-positive tumors is also driven by direct RANKL effects. RANKL stimulated the expression of multiple genes associated with cell invasive behavior, including several matrix metalloproteinases and other genes previously defined as part of a bone metastasis gene signature. These data indicate that RANKL provokes breast cancer bone metastases via two distinct, but potentially overlapping mechanisms: stimulation of tumor-associated osteoclastogenesis and stimulation of RANK-expressing tumor cells.
- Published
- 2014
- Full Text
- View/download PDF
46. Constitutive activation of RANK disrupts mammary cell fate leading to tumorigenesis.
- Author
-
Pellegrini P, Cordero A, Gallego MI, Dougall WC, Muñoz P, Pujana MA, and Gonzalez-Suarez E
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma pathology, Aging pathology, Animals, Breast Neoplasms genetics, Breast Neoplasms pathology, Carcinogenesis genetics, Cell Compartmentation, Cell Differentiation, Cell Shape, Epithelium pathology, Female, Gene Expression Regulation, Neoplastic, Humans, Keratins metabolism, Mammary Glands, Animal metabolism, Mammary Glands, Animal pathology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Mammary Tumor Virus, Mouse physiology, Mice, Models, Biological, Parity, Precancerous Conditions genetics, Precancerous Conditions pathology, Pregnancy, Receptor Activator of Nuclear Factor-kappa B genetics, Stem Cells metabolism, Carcinogenesis pathology, Cell Lineage, Receptor Activator of Nuclear Factor-kappa B metabolism
- Abstract
Receptor Activator of NF-kappa B (RANK) pathway controls mammary gland development in mice but its role in mammary stem cell fate remains undefined. We show that constitutive RANK signaling expands luminal and basal mammary compartments including mammary stem and luminal progenitor cell pools and interferes with the generation of CD61+ and Sca1+ luminal cells and Elf5 expression. Impaired mammary cell commitment upon RANK overexpression leads to the accumulation of progenitors including K14+K8+ bipotent cells and the formation of heterogeneous tumors containing hyperplastic basal, luminal, and progenitor cells. RANK expression increases in wild-type mammary epithelia with age and parity, and spontaneous preneoplastic lesions express RANK and accumulate K14+K8+ cells. In human breast tumors, high RANK expression levels are also associated with altered mammary differentiation. These results suggest that increased RANK signaling interferes with mammary cell commitment, contributing to breast carcinogenesis., (© AlphaMed Press.)
- Published
- 2013
- Full Text
- View/download PDF
47. Progestin effects on cell proliferation pathways in the postmenopausal mammary gland.
- Author
-
Wood CE, Branstetter D, Jacob AP, Cline JM, Register TC, Rohrbach K, Huang LY, Borgerink H, and Dougall WC
- Subjects
- Animals, Biomarkers metabolism, Cell Proliferation drug effects, Cluster Analysis, Epithelial Cells metabolism, Estrogen Receptor Modulators pharmacology, Estrogens metabolism, Estrogens pharmacology, Female, Gene Expression Profiling, Hormone Replacement Therapy, Humans, Immunohistochemistry, Macaca fascicularis, Mammary Glands, Animal drug effects, Norpregnenes pharmacology, Progestins pharmacology, RANK Ligand genetics, RANK Ligand metabolism, Receptor Activator of Nuclear Factor-kappa B genetics, Receptor Activator of Nuclear Factor-kappa B metabolism, Receptors, Estrogen metabolism, Mammary Glands, Animal metabolism, Postmenopause, Progestins metabolism, Signal Transduction drug effects
- Abstract
Introduction: Menopausal hormone therapies vary widely in their effects on breast cancer risk, and the mechanisms underlying these differences are unclear. The primary goals of this study were to characterize the mammary gland transcriptional profile of estrogen + progestin therapy in comparison with estrogen-alone or tibolone and investigate pathways of cell proliferation in a postmenopausal primate model., Methods: Ovariectomized female cynomolgus macaque monkeys were randomized into the following groups: placebo (Con), oral conjugated equine estrogens (CEE), CEE with medroxyprogesterone acetate (MPA) (CEE + MPA), and tibolone given at a low or high dose (Lo or Hi Tib). All study treatment doses represented human clinical dose equivalents and were administered in the diet over a period of 2 years., Results: Treatment with CEE + MPA had the greatest effect on global mRNA profiles and markers of mammary gland proliferation compared to CEE or tibolone treatment. Changes in the transcriptional patterns resulting from the addition of MPA to CEE were related to increased growth factors and decreased estrogen receptor (ER) signaling. Specific genes induced by CEE + MPA treatment included key members of prolactin receptor (PRLR)/signal transducer and activator of transcription 5 (STAT5), epidermal growth factor receptor (EGFR), and receptor activator of nuclear factor kappa B (RANK)/receptor activator of nuclear factor kappa B ligand (RANKL) pathways that were highly associated with breast tissue proliferation. In contrast, tibolone did not affect breast tissue proliferation but did elicit a mixed pattern of ER agonist activity., Conclusion: Our findings indicate that estrogen + progestin therapy results in a distinct molecular profile compared to estrogen-alone or tibolone therapy, including upregulation of key growth factor targets associated with mammary carcinogenesis in mouse models. These changes may contribute to the promotional effects of estrogen + progestin therapy on breast cancer risk.
- Published
- 2013
- Full Text
- View/download PDF
48. RANKL inhibition combined with tamoxifen treatment increases anti-tumor efficacy and prevents tumor-induced bone destruction in an estrogen receptor-positive breast cancer bone metastasis model.
- Author
-
Canon J, Bryant R, Roudier M, Branstetter DG, and Dougall WC
- Subjects
- Animals, Apoptosis drug effects, Bone Density Conservation Agents pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Disease Models, Animal, Female, Humans, MCF-7 Cells, Mice, Mice, Nude, Osteoclasts drug effects, Osteoclasts pathology, Osteoprotegerin pharmacology, RANK Ligand metabolism, Receptors, Estrogen metabolism, Selective Estrogen Receptor Modulators pharmacology, Tumor Microenvironment, Bone Neoplasms drug therapy, Bone Neoplasms secondary, Breast Neoplasms pathology, RANK Ligand antagonists & inhibitors, Tamoxifen pharmacology
- Abstract
Tumor cells in bone can induce the activation of osteoclasts, which mediate bone resorption and release of growth factors and calcium from the bone matrix, resulting in a cycle of tumor growth and bone breakdown. Targeting the bone microenvironment by the inhibition of RANKL, an essential mediator of osteoclast function, not only prevents tumor-induced osteolysis but also decreases skeletal tumor burden in preclinical models. The inhibition of skeletal tumor progression after the inhibition of osteoclasts is via interruption of the "vicious cycle" of tumor/bone interactions. The majority of breast cancer patients at risk for bone metastases harbor estrogen receptor-positive (ER+) tumors. We developed a mouse model for ER+ breast cancer bone metastasis and evaluated the effect of RANKL inhibition on tumor-induced osteolysis and skeletal tumor growth both alone and in combination with tamoxifen. Luciferase-labeled MCF-7 cells (MCF-7Luc) formed metastatic foci in the hind limbs following intracardiac injection and caused mixed osteolytic/osteoblastic lesions. RANKL inhibition by OPG-Fc treatment blocked osteoclast activity and prevented tumor-induced osteolysis, as well as caused a marked decrease in skeletal tumor burden. Tamoxifen as a single agent reduced MCF-7Luc tumor growth in the hind limbs. In a combination experiment, OPG-Fc plus tamoxifen resulted in significantly greater tumor growth inhibition than either single agent alone. Histologic analysis revealed a decrease in the proliferation of tumor cells by both single agents, which was enhanced in the combination treatment. Upon treatment with OPG-Fc alone or in combination with tamoxifen, there was a complete absence of osteolytic lesions, demonstrating the ability of RANKL inhibition to prevent skeletal related morbidity in an ER+ model. The combination approach of targeting osteoclasts and the bone microenvironment by RANKL inhibition and the tumor directly via hormonal therapy may provide additional benefit to reducing skeletal tumor progression in ER+ breast cancer patients.
- Published
- 2012
- Full Text
- View/download PDF
49. The development of denosumab for the treatment of diseases of bone loss and cancer-induced bone destruction.
- Author
-
Goessl C, Katz L, Dougall WC, Kostenuik PJ, Zoog HB, Braun A, Dansey R, and Wagman RB
- Subjects
- Animals, Antineoplastic Agents, Hormonal adverse effects, Bone Neoplasms epidemiology, Bone Neoplasms immunology, Bone Resorption epidemiology, Bone Resorption etiology, Bone Resorption immunology, Denosumab, Humans, Osteoporosis epidemiology, Osteoporosis etiology, Osteoporosis immunology, RANK Ligand adverse effects, Treatment Outcome, Antibodies, Monoclonal, Humanized therapeutic use, Bone Neoplasms drug therapy, Drug Discovery, RANK Ligand antagonists & inhibitors
- Abstract
Denosumab is a fully human monoclonal antibody against RANK ligand (RANKL), an essential cytokine for the formation, function, and survival of osteoclasts. The role of excessive RANKL as a contributor to conditions characterized by bone loss or bone destruction has been well studied. With its novel mechanism of action, denosumab offers a significant advance in the treatment of postmenopausal osteoporosis; bone loss associated with hormone ablation therapy in women with breast cancer and men with prostate cancer; and the prevention of skeletal-related events in patients with bone metastases from solid tumors by offering clinical benefit to these patients in need., (© 2012 New York Academy of Sciences.)
- Published
- 2012
- Full Text
- View/download PDF
50. RANK induces epithelial-mesenchymal transition and stemness in human mammary epithelial cells and promotes tumorigenesis and metastasis.
- Author
-
Palafox M, Ferrer I, Pellegrini P, Vila S, Hernandez-Ortega S, Urruticoechea A, Climent F, Soler MT, Muñoz P, Viñals F, Tometsko M, Branstetter D, Dougall WC, and González-Suárez E
- Subjects
- Animals, BRCA1 Protein physiology, Breast Neoplasms pathology, CD24 Antigen analysis, Cell Line, Tumor, Cell Movement, Humans, Hyaluronan Receptors analysis, Mice, Mice, SCID, Neoplasm Invasiveness, Neoplasm Metastasis, RANK Ligand analysis, Receptor Activator of Nuclear Factor-kappa B analysis, Breast Neoplasms etiology, Cell Transformation, Neoplastic, Epithelial-Mesenchymal Transition, Receptor Activator of Nuclear Factor-kappa B physiology
- Abstract
Paracrine signaling through receptor activator of NF-κB (RANK) pathway mediates the expansion of mammary epithelia that occurs during pregnancy, and activation of RANK pathway promotes mammary tumorigenesis in mice. In this study we extend these previous data to human cells and show that the RANK pathway promotes the development of mammary stem cells and breast cancer. Overexpression of RANK (FL-RANK) in a panel of tumoral and normal human mammary cells induces the expression of breast cancer stem and basal/stem cell markers. High levels of RANK in untransformed MCF10A cells induce changes associated with both stemness and transformation, including mammary gland reconstitution, epithelial-mesenchymal transition (EMT), increased migration, and anchorage-independent growth. In addition, spheroids of RANK overexpressing MCF10A cells display disrupted acinar formation, impair growth arrest and polarization, and luminal filling. RANK overexpression in tumor cells with nonfunctional BRCA1 enhances invasiveness in acinar cultures and increases tumorigenesis and metastasis in immunodeficient mice. High levels of RANK were found in human primary breast adenocarcinomas that lack expression of the hormone receptors, estrogen and progesterone, and in tumors with high pathologic grade and proliferation index; high RANK/RANKL expression was significantly associated with metastatic tumors. Together, our findings show that RANK promotes tumor initiation, progression, and metastasis in human mammary epithelial cells by increasing the population of CD44(+)CD24(-) cells, inducing stemness and EMT. These results suggest that RANK expression in primary breast cancer associates with poor prognosis., (©2012 AACR)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.