137 results on '"Evdokiou, Andreas"'
Search Results
102. Depletion of Host CD122+ Cells Facilitates Widespread Skeletal Multiple Myeloma Engraftment in NOD/SCID Recipients
- Author
-
Freeman, Lisa M, primary, Petcu, Eugene, primary, Smith, Robert, primary, Salajegheh, Ali, primary, Diamond, Peter, primary, Zannettino, Andrew Christopher William, primary, Evdokiou, Andreas, primary, Luff, John, primary, Khalil, Dalia, primary, Vari, Frank, primary, Catley, Lawrence, primary, Hart, Derek, primary, and Vuckovic, Slavica, primary
- Published
- 2008
- Full Text
- View/download PDF
103. Does Apo2L/TRAIL play any physiologic role in osteoclastogenesis?
- Author
-
Labrinidis, Agatha, primary, Liapis, Vasilios, additional, Thai, Le M., additional, Atkins, Gerald J., additional, Vincent, Cristina, additional, Hay, Shelley, additional, Sims, Natalie A., additional, Zannettino, Andrew C. W., additional, Findlay, David M., additional, and Evdokiou, Andreas, additional
- Published
- 2008
- Full Text
- View/download PDF
104. Suberoylanilide hydroxamic acid (vorinostat) represses androgen receptor expression and acts synergistically with an androgen receptor antagonist to inhibit prostate cancer cell proliferation
- Author
-
Marrocco, Deborah L., primary, Tilley, Wayne D., additional, Bianco-Miotto, Tina, additional, Evdokiou, Andreas, additional, Scher, Howard I., additional, Rifkind, Richard A., additional, Marks, Paul A., additional, Richon, Victoria M., additional, and Butler, Lisa M., additional
- Published
- 2007
- Full Text
- View/download PDF
105. RANK Expression as a Cell Surface Marker of Human Osteoclast Precursors in Peripheral Blood, Bone Marrow, and Giant Cell Tumors of Bone
- Author
-
Atkins, Gerald J, primary, Kostakis, Panagiota, additional, Vincent, Cristina, additional, Farrugia, Amanda N, additional, Houchins, Jeffrey P, additional, Findlay, David M, additional, Evdokiou, Andreas, additional, and Zannettino, Andrew CW, additional
- Published
- 2006
- Full Text
- View/download PDF
106. Apo2l/Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand Prevents Breast Cancer–Induced Bone Destruction in a Mouse Model
- Author
-
Thai, Le Minh, primary, Labrinidis, Agatha, additional, Hay, Shelley, additional, Liapis, Vasilios, additional, Bouralexis, Steve, additional, Welldon, Katie, additional, Coventry, Brendon J., additional, Findlay, David M., additional, and Evdokiou, Andreas, additional
- Published
- 2006
- Full Text
- View/download PDF
107. The histone deacetylase inhibitor, suberoylanilide hydroxamic acid, overcomes resistance of human breast cancer cells to Apo2L/TRAIL
- Author
-
Butler, Lisa M., primary, Liapis, Vasilios, additional, Bouralexis, Stelios, additional, Welldon, Katie, additional, Hay, Shelley, additional, Thai, Le M., additional, Labrinidis, Agatha, additional, Tilley, Wayne D., additional, Findlay, David M., additional, and Evdokiou, Andreas, additional
- Published
- 2006
- Full Text
- View/download PDF
108. RANKL Expression Is Related to the Differentiation State of Human Osteoblasts
- Author
-
Atkins, Gerald J, primary, Kostakis, Panagiota, additional, Pan, Beiqing, additional, Farrugia, Amanda, additional, Gronthos, Stan, additional, Evdokiou, Andreas, additional, Harrison, Kate, additional, Findlay, David M, additional, and Zannettino, Andrew Cw, additional
- Published
- 2003
- Full Text
- View/download PDF
109. Chemotherapeutic agents sensitize osteogenic sarcoma cells, but not normal human bone cells, to apo2l/trail‐induced apoptosis
- Author
-
Evdokiou, Andreas, primary, Bouralexis, Stelios, additional, Atkins, Gerald J., additional, Chai, Fugui, additional, Hay, Shelley, additional, Clayer, Mark, additional, and Findlay, David M., additional
- Published
- 2002
- Full Text
- View/download PDF
110. Tumor-suppressive activity of the growth arrest-specific gene GAS1 in human tumor cell lines
- Author
-
Evdokiou, Andreas, primary and Cowled, Prudence A., additional
- Published
- 1998
- Full Text
- View/download PDF
111. Chromosomal Localization of the Human Urothelial “Tetraspan” Gene, UPK1B, to 3q13.3–q21 and Detection of aTaqI Polymorphism
- Author
-
Finch, Jennie L., primary, Webb, Graham C., additional, Evdokiou, Andreas, additional, and Cowled, Prudence A., additional
- Published
- 1997
- Full Text
- View/download PDF
112. Localization of the human growth arrest-specific gene ( GAS1 ) to chromosome bands 9q21.3-q22, a region frequently deleted in myeloid malignancies
- Author
-
Evdokiou, Andreas, primary, Webb, Graham C., additional, Peters, Gregory B., additional, Dobrovic, Alexander, additional, O'Keefe, Denise S., additional, Forbes, Ian J., additional, and Cowled, Prudence A., additional
- Published
- 1993
- Full Text
- View/download PDF
113. Investigation of Spontaneously Aborted Concepti for Microbial DNA: Investigation for Cytomegalovirus DNA Using Polymerase Chain Reaction
- Author
-
Putland, Rodney A., primary, Ford, Judith, additional, Korban, George, additional, Evdokiou, Andreas, additional, and Tremaine, Maureen, additional
- Published
- 1990
- Full Text
- View/download PDF
114. Expression of Osteoclast Differentiation Signals by Stromal Elements of Giant Cell Tumors.
- Author
-
Atkins, Gerald J., Haynes, David R., Graves, Stephen E., Evdokiou, Andreas, Hay, Shelley, Bouralexis, Stelios, and Findlay, David M.
- Published
- 2000
- Full Text
- View/download PDF
115. Tumor-suppressive activity of the growth arrest-specific gene <TOGGLE>GAS</TOGGLE>1 in human tumor cell lines
- Author
-
Evdokiou, Andreas and Cowled, Prudence A.
- Abstract
The GAS1 gene product induces growth arrest through a p53-dependent mechanism. To investigate whether GAS1 is a tumor suppressor gene, we transfected GAS1-negative human tumor cells with GAS1 plasmids and analyzed growth characteristics of stable transfectants. When a constitutively expressing GAS1 plasmid was transfected into A549 cells, no stable colonies expressing GAS1 were isolated. When A549 cells were transfected with a dexamethasone-inducible GAS1 plasmid, expression of GAS1 inhibited growth in vitro, and fewer slow-growing tumors arose in nude mice. GAS1 also inhibited proliferation of an HT1080 subline with wild-type (wt) p53 and normal MDM2. However, when the HT1080 subline HTD114 was transfected with the constitutive GAS1 plasmid, there was no reduction in colony number. GAS1-transfectant clones had unaltered growth in vitro, were morphologically unchanged and showed no difference in their ability to form tumors in nude mice. Although HTD114 cells contain wt p53, levels of MDM2 were elevated by 1015 fold. The HT1080-6TGc5 subline with mutant p53 and normal MDM2 was also refractory to GAS1. Our results show that GAS1 suppresses the growth and tumorigenicity of human tumor cells and overexpression of MDM2 or p53 mutation inhibits the GAS1-mediated growth-suppressing pathway. Int. J. Cancer 75:568577, 1998. © 1998 Wiley-Liss, Inc.
- Published
- 1998
- Full Text
- View/download PDF
116. Near-Infrared Photoimmunotherapy Using a Small Protein Mimetic for HER2-Overexpressing Breast Cancer.
- Author
-
Yamaguchi, Haruka, Pantarat, Namfon, Suzuki, Takamasa, and Evdokiou, Andreas
- Subjects
BREAST cancer ,CELL death ,CANCER cells ,BLOOD-brain barrier ,CELL survival ,MONOCLONAL antibodies ,NEAR infrared radiation - Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a new and promising cancer therapy based on a monoclonal antibody conjugated to a photosensitizer which is activated by near-infrared light irradiation, causing cell death. We investigated NIR-PIT using a small protein mimetic (6–7 kDa), Affibody molecules, instead of a monoclonal antibody for HER2-overexpressing cancer. Because of its small size, the Affibody has rapid clearance, high imaging contrast, and good tumor penetration. Due to the small size of the Affibodies, which can cross the blood–brain barrier, NIR-PIT using Affibodies has the potential to extend the target cancer of NIR-PIT, including brain metastases. In vitro, NIR-PIT using HER2 Affibody–IR700Dye conjugates induced the selective destruction of HER2-overexpressing breast cancer cells without damage to control cells having low level expression of HER2. HER2-overexpressing cancer cells showed necrotic cell death and their viability maintained at low levels, even 5 days after NIR-PIT. In contrast, treatment with high concentration of HER2 Affibody–IR700Dye conjugate alone or irradiation with high dose of NIR light alone was without effect on cell viability. Affibody and IR700Dye are currently used clinically, and therefore, we would expect the current formulation to be safely and quickly transitioned into clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
117. Depletion of Host CD122+Cells Facilitates Widespread Skeletal Multiple Myeloma Engraftment in NOD/SCID Recipients
- Author
-
Freeman, Lisa M, Petcu, Eugene, Smith, Robert, Salajegheh, Ali, Diamond, Peter, Zannettino, Andrew Christopher William, Evdokiou, Andreas, Luff, John, Khalil, Dalia, Vari, Frank, Catley, Lawrence, Hart, Derek, and Vuckovic, Slavica
- Abstract
To facilitate human multiple myeloma (MM) engraftment into NOD/SCID recipients, mice were depleted of CD122+cells (NK and myeloid cells) by antibody-mediated ablation prior to transplantation with the MM cell lines (RPMI8226, RPMI8226-TGL or U226). The MM engraftment, skeletal MM distribution, osteolysis, lambda chain paraprotein and associating disease symptoms in CD122+cell-depleted and CD122+cellreplete mice were compared. The CD122+cell-depleted mice engrafted at a significantly higher frequency with human CD38+, CD56+, PCA-1+and CD138+cells. In the CD122+cell-depleted mice, bioluminescence MM signal involved the whole mouse compared to limited imaging signal in the CD122+cell-replete mice. The majority 88%–100% of CD122+cell-depleted mice developed MM engraftment throughout the appendicular and axial skeletons with osteolysis and rare subcutaneous plasmacytomas (11% of mice). Serum paraprotein appeared earlier at 4–5 weeks post-transplant in CD122+cell-depleted mice and continued to increase during the 12–13 weeks of analysis. The majority, 92% of CD122+cell-depleted mice developed hind-limb paralysis and had a significantly shortened 45 day survival. Thus, depletion of CD122+cells reduced resistance to the human MM and produced a new MM xenograft-NOD/SCID model that recapitulates the clinical manifestations of MM and eliminates the major limitations associated with the published MM xenograft-mouse model.
- Published
- 2008
- Full Text
- View/download PDF
118. Plant-derived soybean peroxidase stimulates osteoblast collagen biosynthesis, matrix mineralization, and accelerates bone regeneration in a sheep model
- Author
-
Stan Gronthos, Romana Panagopoulos, Andreas Evdokiou, Alexandra J. Barker, Mark O. DeNichilo, Peter J. Anderson, Andrew C.W. Zannettino, Vasilios Panagopoulos, Agnes Arthur, Barker, Alexandra J, Arthur, Agnes, DeNichilo, Mark O., Panagopoulos, Romana, Gronthos, Stan, Anderson, Peter J., Zannettino, Andrew C.W., Evdokiou, Andreas, and Panagopoulos, Vasilios
- Subjects
Endocrinology, Diabetes and Metabolism ,Long bone ,Osteoinductive agent ,Bone healing ,Diseases of the musculoskeletal system ,soybean peroxidase ,Article ,Bone remodeling ,Extracellular matrix ,matrix mineralization ,Matrix mineralization ,medicine ,Orthopedics and Sports Medicine ,Bone regeneration ,osteoinductive agent ,Ossification ,Chemistry ,collagen biosynthesis ,Osteoblast ,Collagen biosynthesis ,Soybean peroxidase ,bone repair ,Cell biology ,medicine.anatomical_structure ,RC925-935 ,Intramembranous ossification ,Bone repair ,osteoblast ,medicine.symptom - Abstract
Bone defects arising from fractures or disease represent a significant problem for surgeons to manage and are a substantial economic burden on the healthcare economy. Recent advances in the development of biomaterial substitutes provides an attractive alternative to the current “gold standard” autologous bone grafting. Despite on-going research, we are yet to identify cost effective biocompatible, osteo-inductive factors that stimulate controlled, accelerated bone regeneration.We have recently reported that enzymes with peroxidase activity possess previously unrecognised roles in extracellular matrix biosynthesis, angiogenesis and osteoclastogenesis, which are essential processes in bone remodelling and repair. Here, we report for the first time, that plant-derived soybean peroxidase (SBP) possesses pro-osteogenic ability by promoting collagen I biosynthesis and matrix mineralization of human osteoblasts in vitro. Mechanistically, SBP regulates osteogenic genes responsible for inflammation, extracellular matrix remodelling and ossification, which are necessary for normal bone healing. Furthermore, SBP was shown to have osteo-inductive properties, that when combined with commercially available biphasic calcium phosphate (BCP) granules can accelerate bone repair in a critical size long bone defect ovine model. Micro-CT analysis showed that SBP when combined with commercially available biphasic calcium phosphate (BCP) granules significantly increased bone formation within the defects as early as 4 weeks compared to BCP alone. Histomorphometric assessment demonstrated accelerated bone formation prominent at the defect margins and surrounding individual BCP granules, with evidence of intramembranous ossification. These results highlight the capacity of SBP to be an effective regulator of osteoblastic function and may be beneficial as a new and cost effective osteo-inductive agent to accelerate repair of large bone defects. Refereed/Peer-reviewed
- Published
- 2021
119. Myeloma-Induced Alloreactive T Cells Arising in Myeloma-Infiltratecl Bones Include Double-Positive CD8+CD4+ T Cells: Evidence from Myeloma-Bearing Mouse Model.
- Author
-
Freeman, Lisa M., Lam, Alfred, Petcu, Eugene, Smith, Robert, Salajegheh, Ali, Diamond, Peter, Zannettino, Andrew, Evdokiou, Andreas, Luff, John, Pooi-Fong Wong, Khalil, Dalia, Waterhouse, Nigel, Vari, Frank, Rice, Alison M., Catley, Laurence, Hart, Derek N. J., and Vuckovic, Slavica
- Subjects
- *
MYELOMA proteins , *TUMOR proteins , *T cells , *IMMUNOGLOBULIN idiotypes , *LYMPHOCYTES - Abstract
The graft-versus-myeloma (GVM) effect represents a powerful form of immune attack exerted by alloreactive T cells against multiple myeloma cells, which leads to clinical responses in multiple myeloma transplant recipients. Whether myeloma cells are themselves able to induce alloreactive T cells capable of the GVM effect is not defined. Using adoptive transfer of T naive cells into myeloma-bearing mice (established by transplantation of human RPMI8226-TGL myeloma cells into CD122+ cell-depleted NOD/SCID hosts), we found that myeloma cells induced alloreactive T cells that suppressed myeloma growth and prolonged survival of T cell recipients. Myeloma-induced alloreactive T cells arising in the myeloma-infiltrated bones exerted cytotoxic activity against resident myeloma cells, but limited activity against control myeloma cells obtained from myeloma-bearing mice that did not receive T naive cells. These myeloma-induced alloreactive T cells were derived through multiple CD8+ T cell divisions and enriched in double-positive (DP) T cells coexpressing the CD8αα and CD4 coreceptors. MHC class I expression on myeloma cells and contact with T cells were required for CD8+ T cell divisions and DP-T cell development. DP-T cells present in myeloma-infiltrated bones contained a higher proportion of cells expressing cytotoxic mediators IFN-γ and/or perforin compared with single-positive CD8+ T cells, acquired the capacity to degranulate as measured by CD107 expression, and contributed to an elevated perforin level seen in the myeloma-infiltrated bones. These observations suggest that myeloma-induced alloreactive T cells arising in myeloma-infiltrated bones are enriched with DP-T cells equipped with cytotoxic effector functions that are likely to be involved in the GVM effect. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
120. Clodronate-liposome mediated macrophage depletion abrogates multiple myeloma tumor establishment in vivo
- Author
-
Khatora S. Opperman, Kate Vandyke, Elizabeth A. Coulter, Andreas Evdokiou, Krzysztof M. Mrozik, Kimberley C. Clark, N. Schwarz, Peter I. Croucher, Jacqueline E. Noll, Peter J. Psaltis, Duncan R. Hewett, Andrew C.W. Zannettino, Opperman, Khatora S, Vandyke, Kate, Clark, Kimberley C, Coulter, Elizabeth A, Hewett, Duncan R, Mrozik, Krzysztof M, Schwarz, Nisha, Evdokiou, Andreas, Croucher, Peter I, Psaltis, Peter J, Noll, Jacqueline E, and Zannettino, Andrew CW
- Subjects
0301 basic medicine ,Original article ,Cancer Research ,medicine.medical_treatment ,Cell Count ,Plasma cell ,Immunophenotyping ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,hemic and lymphatic diseases ,Tumor Microenvironment ,Animals ,Medicine ,Multiple myeloma ,Tumor microenvironment ,Osteoblasts ,Bone Density Conservation Agents ,business.industry ,Macrophages ,Growth factor ,Mesenchymal stem cell ,medicine.disease ,3. Good health ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Liposomes ,Cancer research ,Clodronic acid ,Disease Susceptibility ,Bone marrow ,Clodronic Acid ,Multiple Myeloma ,business ,Biomarkers ,medicine.drug - Abstract
Multiple myeloma is a fatal plasma cell malignancy that is reliant on the bone marrow microenvironment. The bone marrow is comprised of numerous cells of mesenchymal and hemopoietic origin. Of these, macrophages have been implicated to play a role in myeloma disease progression, angiogenesis, and drug resistance; however, the role of macrophages in myeloma disease establishment remains unknown. In this study, the antimyeloma efficacy of clodronate-liposome treatment, which globally and transiently depletes macrophages, was evaluated in the well-established C57BL/KaLwRijHsd murine model of myeloma. Our studies show, for the first time, that clodronate-liposome pretreatment abrogates myeloma tumor development in vivo. Clodronate-liposome administration resulted in depletion of CD169+ bone marrow–resident macrophages. Flow cytometric analysis revealed that clodronate-liposome pretreatment impaired myeloma plasma cell homing and retention within the bone marrow 24 hours postmyeloma plasma cell inoculation. This was attributed in part to decreased levels of macrophage-derived insulin-like growth factor 1. Moreover, a single dose of clodronate-liposome led to a significant reduction in myeloma tumor burden in KaLwRij mice with established disease. Collectively, these findings support a role for CD169-expressing bone marrow–resident macrophages in myeloma disease establishment and progression and demonstrate the potential of targeting macrophages as a therapy for myeloma patients. Refereed/Peer-reviewed
- Published
- 2019
121. Pharmacologic inhibition of bone resorption prevents cancer-induced osteolysis but enhances soft tissue metastasis in a mouse model of osteolytic breast cancer
- Author
-
Vasilios Liapis, Clara Bik-San Lau, Agatha Labrinidis, David M. Findlay, Vladimir Ponomarev, Gerald J. Atkins, Vasilios Panagopoulos, Andreas Evdokiou, Ke-Wang Luo, Wendy V. Ingman, Grace Gar-Lee Yue, Andrew C.W. Zannettino, Chun-Hay Ko, Irene Zinonos, Shelley Hay, Mark O. DeNichilo, Zinonos, Irene, Luo, Ke-Wang, Labrinidis, Agatha, Liapis, Vasilios, Hay, Shelley, Panagopoulos, Vasilios, DeNichilo, Mark, Ko, Chun-Hay, Yue, Grace Gar-Lee, Lau, Clara Bik-San, Ingman, Wendy, Ponomarev, Vladimir, Atkins, Gerald J, Findlay, David M, Zannettino, Andrew CW, and Evdokiou, Andreas
- Subjects
musculoskeletal diseases ,Cancer Research ,Pathology ,medicine.medical_specialty ,Osteolysis ,Mice, Nude ,Bone Neoplasms ,Breast Neoplasms ,Enzyme-Linked Immunosorbent Assay ,Soft Tissue Neoplasms ,Transfection ,Bone resorption ,antiresorptiveagents ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Osteoprotegerin ,Osteoclast ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Bone Resorption ,bisphosphonates ,bone metastasis ,030304 developmental biology ,0303 health sciences ,biology ,antiresorptive agents ,Bone metastasis ,X-Ray Microtomography ,Articles ,medicine.disease ,3. Good health ,Disease Models, Animal ,medicine.anatomical_structure ,Oncology ,osteoprotegerin ,RANKL ,030220 oncology & carcinogenesis ,biology.protein ,Female ,bone resorption - Abstract
Osteoprotegerin (OPG) is a secreted member of the TNF receptor superfamily, which binds to the receptor activator of nuclear factor κB ligand (RANKL) and inhibits osteoclast activity and bone resorption. Systemic administration of recombinant OPG was previously shown to inhibit tumor growth in bone and to prevent cancer-induced osteolysis. In this study, we examined the effect of OPG, when produced locally by breast cancer cells located within bone, using a mouse model of osteolytic breast cancer. MDA-MB-231-TXSA breast cancer cells, tagged with a luciferase reporter gene construct and engineered to overexpress full-length human OPG, were transplanted directly into the tibial marrow cavity of nude mice. Overexpression of OPG by breast cancer cells protected the bone from breast cancer-induced osteolysis and diminished intra-osseous tumor growth but had no effect on extra-skeletal tumor growth. This effect was associated with a significant reduction in the number of osteoclasts that lined the bone surface, resulting in a net increase in bone volume. Despite limiting breast cancer-mediated bone loss, OPG over-expression resulted in a significant increase in the incidence of pulmonary metastasis. Our results demonstrate that inhibition of osteoclastic bone resorption by OPG when secreted locally by tumors in bone may affect the behaviour of cancer cells within the bone microenvironment and their likelihood of spreading and establishing metastasis elsewhere in the body. Refereed/Peer-reviewed
- Published
- 2014
- Full Text
- View/download PDF
122. Nutlin-3a Efficacy in Sarcoma Predicted by Transcriptomic and Epigenetic Profiling
- Author
-
Robert J. Whitfield, Jayesh Desai, Michael P. Brown, Paul M. Neilsen, Gelareh Farshid, Andreas W. Schreiber, David F. Callen, Jim Manavis, Kathleen I. Pishas, Andreas Evdokiou, Michelle Perugini, Mark Clayer, David M. Lawrence, Susan J. Neuhaus, Rebecca C Haycox, Bronwen J. Mayo, Steve Chryssidis, Kristen Ho, David Thomas, Richard J D'Andrea, Pishas, Kathleen I, Neuhaus, Susan J, Clayer, Mark T, Schreiber, Andreas W, Lawrence, David M, Perugini, Michelle, Whitfield, Robert J, Farshid, Gelareh, Manavis, Jim, Chyrssidis, Steve, Mayo, Bronwen J, Haycox, Rebecca C, Ho, Kristen, Brown, Michael P, D'Andrea, Richard J, Evdokiou, Andreas, Thomas, David M, Desai, Jayesh, Callen, David F, and Neilsen, Paul M
- Subjects
Epigenomics ,Male ,Cancer Research ,Apoptosis ,Cell Cycle Proteins ,Piperazines ,Epigenesis, Genetic ,Transcriptome ,Cluster Analysis ,Aged, 80 and over ,Regulation of gene expression ,Tumor ,biology ,Imidazoles ,Nuclear Proteins ,Proto-Oncogene Proteins c-mdm2 ,Sarcoma ,Middle Aged ,Prognosis ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,Oncology ,DNA methylation ,Mdm2 ,Female ,Signal Transduction ,Adult ,Adolescent ,Antineoplastic Agents ,Polymorphism, Single Nucleotide ,Cell Line ,Young Adult ,Cell Line, Tumor ,medicine ,Humans ,Epigenetics ,Epigenesi ,neoplasms ,Aged ,Gene Expression Profiling ,Gene Amplification ,DNA Methylation ,medicine.disease ,Gene expression profiling ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Tumor Suppressor Protein p53 - Abstract
Nutlin-3a is a small-molecule antagonist of p53/MDM2 that is being explored as a treatment for sarcoma. In this study, we examined the molecular mechanisms underlying the sensitivity of sarcomas to Nutlin-3a. In an ex vivo tissue explant system, we found that TP53 pathway alterations (TP53 status, MDM2/MDM4 genomic amplification/mRNA overexpression, MDM2 SNP309, and TP53 SNP72) did not confer apoptotic or cytostatic responses in sarcoma tissue biopsies (n = 24). Unexpectedly, MDM2 status did not predict Nutlin-3a sensitivity. RNA sequencing revealed that the global transcriptomic profiles of these sarcomas provided a more robust prediction of apoptotic responses to Nutlin-3a. Expression profiling revealed a subset of TP53 target genes that were transactivated specifically in sarcomas that were highly sensitive to Nutlin-3a. Of these target genes, the GADD45A promoter region was shown to be hypermethylated in 82% of wild-type TP53 sarcomas that did not respond to Nutlin-3a, thereby providing mechanistic insight into the innate ability of sarcomas to resist apoptotic death following Nutlin-3a treatment. Collectively, our findings argue that the existing benchmark biomarker for MDM2 antagonist efficacy (MDM2 amplification) should not be used to predict outcome but rather global gene expression profiles and epigenetic status of sarcomas dictate their sensitivity to p53/MDM2 antagonists. Cancer Res; 74(3); 921–31. ©2013 AACR.
- Published
- 2014
- Full Text
- View/download PDF
123. Myeloma plasma cells alter the bone marrow microenvironment by stimulating the proliferation of mesenchymal stromal cells
- Author
-
Christine M. Tong, Katherine R. Pilkington, Stan Gronthos, Sharon A. Williams, L. B. To, Louise E. Purton, Jacqueline E. Noll, Hongsheng Wang, Julie M. Quach, Andreas Evdokiou, Andrew C.W. Zannettino, Noll, Jacqueline E, Williams, Sharon A, Tong, Christine M, Wang, Hongsheng, Quach, Julie M, Purton, Louise E, Pilkington, Katherine, To, Luen B, Evdokiou, Andreas, Gronthos, Stan, and Zannettino, Andrew CW
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,Plasma Cells ,Biology ,Plasma cell ,Monoclonal Gammopathy of Undetermined Significance ,Severity of Illness Index ,Immunophenotyping ,Mice ,Bone Marrow ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Tumor Stem Cell Assay ,Multiple myeloma ,Cell Proliferation ,bone marrow microenvironment ,Lymphokines ,mesenchymal stem cells ,Tumor microenvironment ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Osteoblast ,Articles ,Hematology ,medicine.disease ,multiple myeloma ,Disease Models, Animal ,medicine.anatomical_structure ,Antigens, Surface ,Cancer research ,Bone marrow ,Multiple Myeloma ,Monoclonal gammopathy of undetermined significance ,monoclonal gammopathy of undetermined significance - Abstract
Multiple myeloma is an incurable haematological cancer characterised by the clonal proliferation of malignant plasma cells within the bone marrow. Numerous studies suggest that the myeloma plasma cells occupy and alter the stromal tissue of the bone marrow as a means of enhancing their survival and growth. However, the nature and magnitude of the changes to the stromal cell tissue remain to be determined. In this study, we used mesenchymal stromal cell and osteoblast-related cell surface marker expression (STRO-1 and alkaline phosphatase, respectively) and flow cytometry to enumerate mesenchymal stromal cell and osteoblast numbers in bone marrow recovered from myeloma patients at the time of diagnosis. Using this approach, we identified an increase in the number of STRO-1 positive colony forming mesenchymal stromal cell and a concomitant decrease in alkaline phophatase osteoblasts. Notably, this increase in mesenchymal stromal cell numbers correlated closely with plasma cell burden at the time of diagnosis. Additionally, in comparison with the osteoblast population, the STRO-1+ mesenchymal stromal cell population was found to express higher levels of plasma cell- and osteoclast- activating factors, including RANKL and IL-6, providing a mechanism by which an increase in mesenchymal stromal cell may promote and aid the progression of myeloma. Importantly, these findings were faithfully replicated in the C57BL/KaLwRij murine model of myeloma, suggesting that this model may present a unique and clinically relevant system in which to identify and therapeutically modulate the bone microenvironment and in turn, alter the progression of myeloma disease. Refereed/Peer-reviewed
- Published
- 2013
- Full Text
- View/download PDF
124. Anticancer efficacy of the hypoxia-activated prodrug evofosfamide is enhanced in combination with proapoptotic receptor agonists against osteosarcoma
- Author
-
Christopher Difelice, David M. Findlay, Vasilios Panagopoulos, Vasilios Liapis, Andreas Evdokiou, Gerald J. Atkins, Aneta Zysk, Vladimir Ponomarev, Wendy V. Ingman, Andrew C.W. Zannettino, Irene Zinonos, Shelley Hay, Alexandra J. Shoubridge, Mark O. DeNichilo, Liapis, Vasilios, Zysk, Aneta, DeNichilo, Mark, Zinonos, Irene, Hay, Shelley, Panagopoulos, Vasilios, Shoubridge, Alexandra J, Difelice, Christopher, Ponomarev, Vladimir, Ingman, Wendy, Atkins, Gerald J, Findlay, David M, Zannettino, Andrew CW, and Evdokiou, Andreas
- Subjects
0301 basic medicine ,Cancer Research ,dulanermin ,Pharmacology ,TNF-Related Apoptosis-Inducing Ligand ,chemistry.chemical_compound ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Prodrugs ,Original Research ,drozitumab ,Osteosarcoma ,Antibodies, Monoclonal ,Drug Synergism ,Cell Hypoxia ,3. Good health ,Oncology ,Nitroimidazoles ,030220 oncology & carcinogenesis ,Phosphoramide Mustards ,medicine.symptom ,Cancer Prevention ,musculoskeletal diseases ,Cell Survival ,Bone Neoplasms ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,Cell Line, Tumor ,osteosarcoma ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,evofosfamide ,Drozitumab ,Cell Proliferation ,Dulanermin ,Evofosfamide ,Tumor hypoxia ,Cell growth ,business.industry ,hypoxia ,Hypoxia (medical) ,medicine.disease ,Xenograft Model Antitumor Assays ,Transplantation ,030104 developmental biology ,chemistry ,Cancer cell ,business - Abstract
Tumor hypoxia is a major cause of treatment failure for a variety of malignancies. However, hypoxia also leads to treatment opportunities as demonstrated by the development of compounds that target regions of hypoxia within tumors. Evofosfamide is a hypoxia-activated prodrug that is created by linking the hypoxia-seeking 2-nitroimidazole moiety to the cytotoxic bromo-isophosphoramide mustard (Br-IPM). When evofosfamide is delivered to hypoxic regions of tumors, the DNA cross-linking toxin, Br-IPM, is released leading to cell death. This study assessed the anticancer efficacy of evofosfamide in combination with the Proapoptotic Receptor Agonists (PARAs) dulanermin and drozitumab against human osteosarcoma in vitro and in an intratibial murine model of osteosarcoma. Under hypoxic conditions in vitro, evofosfamide cooperated with dulanermin and drozitumab, resulting in the potentiation of cytotoxicity to osteosarcoma cells. In contrast, under the same conditions, primary human osteoblasts were resistant to treatment. Animals transplanted with osteosarcoma cells directly into their tibiae developed mixed osteosclerotic/osteolytic bone lesions and consequently developed lung metastases 3 weeks post cancer cell transplantation. Tumor burden in the bone was reduced by evofosfamide treatment alone and in combination with drozitumab and prevented osteosarcoma-induced bone destruction while also reducing the growth of pulmonary metastases. These results suggest that evofosfamide may be an attractive therapeutic agent, with strong anticancer activity alone or in combination with either drozitumab or dulanermin against osteosarcoma. Refereed/Peer-reviewed
- Published
- 2017
125. Peroxidase enzymes inhibit osteoclast differentiation and bone resorption
- Author
-
Damien A. Leach, Gerald J. Atkins, Irene Zinonos, Mark O. DeNichilo, Wendy V. Ingman, Vasilios Liapis, Andrew C.W. Zannettino, Vasilios Panagopoulos, Andreas Evdokiou, David M. Findlay, Shelley Hay, Panagopoulos, Vasilios, Liapis, Vasilios, Zinonos, Irene, Hay, Shelley, Leach, Damien A, Ingman, Wendy, DeNichilo, Mark O, Atkins, Gerald J, Findlay, David M, Zannettino, Andrew CW, and Evdokiou, Andreas
- Subjects
0301 basic medicine ,musculoskeletal diseases ,Bone disease ,Osteoclasts ,030209 endocrinology & metabolism ,Biochemistry ,Bone resorption ,03 medical and health sciences ,Mice ,Endocrinology & Metabolism ,0302 clinical medicine ,Endocrinology ,Osteoclast ,medicine ,Animals ,Humans ,Bone Resorption ,Molecular Biology ,Peroxidase ,biology ,Chemistry ,Macrophage Colony-Stimulating Factor ,RANK Ligand ,Cell Differentiation ,Cell Biology ,medicine.disease ,Endocytosis ,Resorption ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,RAW 264.7 Cells ,RANKL ,Myeloperoxidase ,biology.protein ,Eosinophil peroxidase ,Signal Transduction - Abstract
Myeloperoxidase (MPO) and eosinophil peroxidase (EPO) are heme-containing enzymes, well known for their antimicrobial activity, are released in abundance by innate immune infiltrates at sites of inflammation and injury. We have discovered new and previously unrecognised roles for heme peroxidases in extracellular matrix biosynthesis, angiogenesis, and bone mineralisation, all of which play an essential role in skeletal integrity. In this study we used in vitro models of osteoclastogenesis to investigate the effects of heme peroxidase enzymes on osteoclast differentiation and bone resorbing activity, pertinent to skeletal development and remodelling. Receptor activator of nuclear factor kappa B-ligand (RANKL) stimulates the formation of tartate-resistant acid phosphatase (TRAP) positive multinucleated cells and increases bone resorption when cultured with human peripheral blood mononuclear cells (PBMCs) or the RAW264.7 murine monocytic cell line. When RANKL was added in combination with either MPO or EPO, a dose-dependent inhibition of osteoclast differentiation and bone resorption was observed. Notably, peroxidases had no effect on the bone resorbing activity of mature osteoclasts, suggesting that the inhibitory effect of the peroxidases was limited to osteoclast precursor cells. Mechanistically, we observed that osteoclast precursor cells readily internalize peroxidases, and inhibited the phosphorylation of JNK, p38 MAPK and ERK1/2, important signalling molecules central to osteoclastogenesis. Our findings suggest that peroxidase enzymes, like MPO and EPO, may play a fundamental role in inhibiting RANKL-induced osteoclast differentiation at inflammatory sites of bone fracture and injury. Therefore, peroxidase enzymes could be considered as potential therapeutic agents to treat osteolytic bone disease and aberrant bone resorption. Refereed/Peer-reviewed
- Published
- 2017
126. Adoptive transfer of ex vivo expanded Vγ9Vδ2 T cells in combination with zoledronic acid inhibits cancer growth and limits osteolysis in a murine model of osteolytic breast cancer
- Author
-
Vasilios Liapis, Wendy V. Ingman, Andreas Evdokiou, Mark O. DeNichilo, Irene Zinonos, Vasilios Panagopoulos, Andrew C.W. Zannettino, Vladimir Ponomarev, Gerald J. Atkins, David M. Findlay, Aneta Zysk, Shelley Hay, Zysk, Aneta, DeNichilo, Mark O, Panagopoulos, Vasilios, Zinonos, Irene, Liapis, Vasilios, Hay, Shelley, Ingman, Wendy, Ponomarev, Vladimir, Atkins, Gerald, Findlay, David, Zannettino, Andrew, and Evdokiou, Andreas
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,bisphosphonate ,Cancer Research ,Pathology ,Adoptive cell transfer ,Lung Neoplasms ,Time Factors ,medicine.medical_treatment ,Mice, SCID ,Osteolysis ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Zoledronic Acid ,Metastasis ,0302 clinical medicine ,Mice, Inbred NOD ,Cytotoxic T cell ,Bone Density Conservation Agents ,Diphosphonates ,Imidazoles ,Receptors, Antigen, T-Cell, gamma-delta ,medicine.anatomical_structure ,Phenotype ,Oncology ,tumour associated macrophage ,030220 oncology & carcinogenesis ,osteoclast ,Female ,immunotherapy ,medicine.drug ,medicine.medical_specialty ,T cell ,Antineoplastic Agents ,Bone Neoplasms ,Breast Neoplasms ,Article ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,medicine ,metastasis ,Animals ,Humans ,Cell Proliferation ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Zoledronic acid ,Cancer research ,business ,T-Lymphocytes, Cytotoxic - Abstract
Bone metastases occur in over 75% of patients with advanced breast cancer and are responsible for high levels of morbidity and mortality. In this study, ex vivo expanded cytotoxic Vγ9Vδ2T cells isolated from human peripheral blood were tested for their anti-cancer efficacy in combination with zoledronic acid (ZOL), using a mouse model of osteolytic breast cancer. In vitro, expanded γ9Vδ2 T cells were cytotoxic against a panel of human breast cancer cell lines, and ZOL pre-treatment further sensitised breast cancer cells to killing by Vγ9Vδ2 T cells. Vγ9Vδ2 T cells adoptively transferred into NOD/SCID mice localised to osteolytic breast cancer lesions in the bone, and multiple infusions of Vγ9Vδ2T cells reduced tumour growth in the bone. ZOL pre-treatment potentiated the anti-cancer efficacy of Vγ9Vδ2 T cells, with mice showing further reductions in tumour burden. Mice treated with the combination also had reduced tumour burden of secondary pulmonary metastases, and decreased bone degradation. Our data suggests that adoptive transfer of Vγ9Vδ2 T cell in combination with ZOL may prove an effective immunotherapeutic approach for the treatment of breast cancer bone metastases. National Breast Cancer FoundationNBCF-13-09 Refereed/Peer-reviewed
- Published
- 2016
127. Anticancer efficacy of Apo2L/TRAIL is retained in the presence of high and biologically active concentrations of osteoprotegerin in vivo
- Author
-
Agatha Labrinidis, Vladimir Ponomarev, Vasilios Liapis, Andreas Evdokiou, Irene Zinonos, Shelley Hay, David M. Findlay, Andrew C.W. Zannettino, Michelle Lee, Peter Diamond, Zinonos, Irene, Labrinidis, Agatha, Lee, Michelle, Liapis, Vasilios, Hay, Shelley, Ponomarev, Vladimir, Diamond, Peter, Findlay, David M, Zannettino, Andrew CW, and Evdokiou, Andreas
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,Osteolysis ,Endocrinology, Diabetes and Metabolism ,tumor necrosis factor (TNF) ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Bone Neoplasms ,Breast Neoplasms ,Transfection ,Article ,Bone resorption ,TNF-Related Apoptosis-Inducing Ligand ,Mice ,chemistry.chemical_compound ,Osteoprotegerin ,In vivo ,Cell Line, Tumor ,Internal medicine ,medicine ,cancer ,Animals ,Humans ,Orthopedics and Sports Medicine ,Tibia ,biology ,business.industry ,medicine.disease ,Xenograft Model Antitumor Assays ,Treatment Outcome ,Endocrinology ,Solubility ,chemistry ,Cytoprotection ,RANKL ,Cancer research ,biology.protein ,osteoprotegerin (OPG) ,Female ,Tumor necrosis factor alpha ,Growth inhibition ,business - Abstract
Osteoprotegerin (OPG) is a secreted member of the tumor necrosis factor (TNF) receptor superfamily that binds to the ligand for receptor activator of nuclear factor κB (RANKL) and inhibits bone resorption. OPG can also bind and inhibit the activity of the TNF-related apoptosis-inducing ligand (Apo2L/TRAIL), raising the possibility that the anticancer efficacy of soluble Apo2L/TRAIL may be abrogated in the bone microenvironment where OPG expression is high. In this study we used a murine model of breast cancer growth in bone to evaluate the efficacy of recombinant soluble Apo2L/TRAIL against intratibial tumors that were engineered to overexpress native full-length human OPG. In vitro, OPG-overexpressing breast cancer cells were protected from Apo2L/TRAIL-induced apoptosis, an effect that was reversed with the addition of soluble RANKL or neutralizing antibodies to OPG. In vivo, mice injected intratibially with cells containing the empty vector developed large osteolytic lesions. In contrast, OPG overexpression preserved the integrity of bone and prevented breast cancer–induced bone destruction. This effect was due primarily to the complete absence of osteoclasts in the tibias of mice inoculated with OPG-transfected cells, confirming the biologic activity of the transfected OPG in vivo. Despite the secretion of supraphysiologic levels of OPG, treatment with Apo2L/TRAIL resulted in strong growth inhibition of both empty vector and OPG-overexpressing intratibial tumors. While Apo2L/TRAIL-induced apoptosis may be abrogated in vitro by OPG overexpression, the in vivo anticancer efficacy of recombinant soluble Apo2L/TRAIL is retained in the bone microenvironment even in the presence of biologically active OPG at supraphysiologic concentrations. © 2011 American Society for Bone and Mineral Research.
- Published
- 2011
- Full Text
- View/download PDF
128. Uncovering a new role for peroxidase enzymes as drivers of angiogenesis
- Author
-
Wendy V. Ingman, Damien A. Leach, Andreas Evdokiou, Aneta Zysk, Shelley Hay, Vasilios Liapis, Vasilios Panagopoulos, Mark O. DeNichilo, Irene Zinonos, Panagopoulos, Vasilios, Zinonos, Irene, Leach, Damien A, Hay, Shelley J, Liapis, Vasilios, Zysk, Aneta, Ingman, Wendy V, DeNichilo, Mark O, and Evdokiou, Andreas
- Subjects
Eosinophil Peroxidase ,Angiogenesis ,Neovascularization, Physiologic ,Inflammation ,Biochemistry ,p38 Mitogen-Activated Protein Kinases ,Fibroblast migration ,eosinophil peroxidase ,angiogenesis ,Mice ,Cell Movement ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Enzyme Inhibitors ,Protein kinase B ,Cell Proliferation ,Peroxidase ,Mitogen-Activated Protein Kinase 1 ,Mice, Inbred BALB C ,Aniline Compounds ,Mitogen-Activated Protein Kinase 3 ,biology ,Cell Biology ,Endocytosis ,Cell biology ,myeloperoxidase ,Drug Combinations ,Gene Expression Regulation ,inflammation ,Myeloperoxidase ,Focal Adhesion Kinase 1 ,biology.protein ,Phosphorylation ,Biological Assay ,Female ,Proteoglycans ,Collagen ,Laminin ,medicine.symptom ,Eosinophil peroxidase ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Peroxidases are heme-containing enzymes released by activated immune cells at sites of inflammation. To-date their functional role in human health has mainly been limited to providing a mechanism for oxidative defence against invading bacteria and other pathogenic microorganisms. Our laboratory has recently identified a new functional role for peroxidase enzymes in stimulating fibroblast migration and collagen biosynthesis, offering a new insight into the causative association between inflammation and the pro-fibrogenic events that mediate tissue repair and regeneration. Peroxidases are found at elevated levels within and near blood vessels however, their direct involvement in angiogenesis has never been reported. Here we report for the first time that myeloperoxidase (MPO) and eosinophil peroxidase (EPO) are readily internalised by human umbilical vein endothelial cells (HUVEC) where they promote cellular proliferation, migration, invasion, and stimulate angiogenesis both in vitro and in vivo. These pro-angiogenic effects were attenuated using the specific peroxidase inhibitor 4-ABAH, indicating the enzyme's catalytic activity is essential in mediating this response. Mechanistically, we provide evidence that MPO and EPO regulate endothelial FAK, Akt, p38 MAPK, ERK1/2 phosphorylation and stabilisation of HIF-2α, culminating in transcriptional regulation of key angiogenesis pathways. These findings uncover for the first time an important and previously unsuspected role for peroxidases as drivers of angiogenesis, and suggest that peroxidase inhibitors may have therapeutic potential for the treatment of angiogenesis related diseases driven by inflammation. Refereed/Peer-reviewed
- Published
- 2015
129. Hypoxia-activated pro-drug TH-302 exhibits potent tumor suppressive activity and cooperates with chemotherapy against osteosarcoma
- Author
-
Shelley Hay, Vasilios Liapis, Andreas Evdokiou, Gerald J. Atkins, Andrew C.W. Zannettino, Vasilios Panagopoulos, Agatha Labrinidis, Irene Zinonos, David M. Findlay, Vladimir Ponomarev, Mark O. DeNichilo, Wendy V. Ingman, Liapis, Vasilios, Labrinidis, Agatha, Zinonos, Irene, Hay, Shelley, Ponomarev, Vladimir, Panagopoulos, Vasilios, DeNichilo, Mark O, Ingman, Wendy, Atkins, Gerald J, Findlay, David M, Zannettino, Andrew CW, and Evdokiou, Andreas
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Mice, Nude ,Apoptosis ,Bone Neoplasms ,chemotherapy ,Article ,Metastasis ,Mice ,osteosarcoma ,medicine ,Cytotoxic T cell ,metastasis ,Animals ,Humans ,Doxorubicin ,Prodrugs ,Neoplasm Metastasis ,TH-302 ,Chemotherapy ,Osteosarcoma ,Tumor hypoxia ,business.industry ,hypoxia ,Hypoxia (medical) ,medicine.disease ,In vitro ,Cell Hypoxia ,Oncology ,Nitroimidazoles ,Cancer research ,Female ,Phosphoramide Mustards ,medicine.symptom ,business ,medicine.drug - Abstract
Tumor hypoxia is a major cause of treatment failure for a variety of malignancies. However, tumor hypoxia also offers treatment opportunities, exemplified by the development compounds that target hypoxic regions within tumors. TH-302 is a pro-drug created by the conjugation of 2-nitroimidazole to bromo-isophosphoramide (Br-IPM). When TH-302 is delivered to regions of hypoxia, Br-IPM, the DNA cross linking toxin, is released. In this study we assessed the cytotoxic activity of TH-302 against osteosarcoma cells in vitro and evaluated its anticancer efficacy as a single agent, and in combination with doxorubicin, in an orthotopic mouse model of human osteosarcoma (OS). In vitro, TH-302 was potently cytotoxic to osteosarcoma cells selectively under hypoxic conditions, whereas primary normal human osteoblasts were protected. Animals transplanted with OS cells directly into their tibiae and left untreated developed mixed osteolytic/osteosclerotic bone lesions and subsequently developed lung metastases. TH-302 reduced tumor burden in bone and cooperated with doxorubicin to protect bone from osteosarcoma induced bone destruction, while it also reduced lung metastases. TH-302 may therefore be an attractive therapeutic agent with strong activity as a single agent and in combination with chemotherapy against OS. Refereed/Peer-reviewed
- Published
- 2014
130. Doxorubicin overcomes resistance to drozitumab by antagonizing inhibitor of apoptosis proteins (IAPs)
- Author
-
Zinonos, Irene, Labrinidis, Agatha, Liapis, Vasilios, Hay, Shelley, Panagopoulos, Vasilios, DeNichilo, Mark, Ponomarev, Vladimir, Ingman, Wendy, Atkins, Gerald J, Findlay, David M, Zannettino, Andrew CW, and Evdokiou, Andreas
- Subjects
drozitumab ,apo2L/TRAIL ,drug resistance ,breast cancer ,apoptosis ,chemotherapy - Abstract
Background/Aim: Drozitumab is a fully human agonistic monoclonal antibody that binds to death receptor DR5 and induces apoptosis. However, drozitumab resistance is a major obstacle limiting anticancer efficacy. Materials and Methods: We examined the potential for the chemotherapeutic agent doxorubicin to overcome resistance against drozitumab-resistant breast cancer cells both in vitro and in vivo. Results: Treatment with doxorubicin increased cell surface expression of DR5, reduced levels of Inhibitors of Apoptosis Proteins (cIAPs) and re-sensitised cells to drozitumab-induced apoptosis. Animals implanted with resistant breast cancer cells into the mammary fat pad and treated with a combination of drozitumab and doxorubicin showed inhibition of tumor growth and a substantial delay in tumor progression compared to untreated controls and mice treated with each agent alone. Conclusion: These results suggest that combination of drozitumab with chemotherapy and agents that modulate IAP levels could potentially be a useful strategy in the treatment of breast cancer. Refereed/Peer-reviewed
- Published
- 2014
131. Pre-activation of the p53 pathway through Nutlin-3a sensitises sarcomas to drozitumab therapy
- Author
-
Alaknanda Adwal, Paul M. Neilsen, David F. Callen, Andreas Evdokiou, Mark Clayer, Susan J. Neuhaus, Kathleen I. Pishas, Michael P. Brown, Pishas, Kathleen I, Neuhaus, Susan J, Clayer, Mark T, Adwal, Alaknanda, Brown, Michael P, Evdokiou, Andreas, Callen, David F, and Neilsen, Paul M
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Nutlin-3a ,sarcoma ,Combination therapy ,Apoptosis ,Biology ,Antibodies, Monoclonal, Humanized ,Piperazines ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,TP53 ,RNA, Messenger ,RNA, Small Interfering ,Drozitumab ,Gene knockdown ,Imidazoles ,Cancer ,Antibodies, Monoclonal ,Sarcoma ,General Medicine ,Cell cycle ,medicine.disease ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Monoclonal ,Cancer research ,RNA Interference ,Tumor Suppressor Protein p53 ,Ex vivo - Abstract
The present study evaluated the efficacy of drozitumab, a human monoclonal agonistic antibody directed against death receptor 5 (DR5), as a new therapeutic avenue for the targeted treatment of bone and soft-tissue sarcomas. The antitumour activity of drozitumab as a monotherapy or in combination with Nutlin-3a was evaluated in a panel of sarcoma cell lines in vitro and human sarcoma patient samples ex vivo. Knockdown experiments were used to investigate the central role of p53 as a regulator of drozitumab cytotoxicity. Pre-activation of the p53 pathway through Nutlin-3a upregulated DR5, subsequently sensitising sarcoma cell lines and human sarcoma specimens to the pro-apoptotic effects of drozitumab. Silencing of p53 strongly decreased DR5 mRNA expression resulting in abrogation of drozitumab-induced apoptosis. Our study provides the first pre-clinical evaluation of combination therapy using p53-activating agents with drozitumab to further sensitise sarcomas to the cytotoxic effects of DR5 antibody therapy. Refereed/Peer-reviewed
- Published
- 2013
132. Nutlin-3a is a potential therapeutic for Ewing sarcoma
- Author
-
Kathleen I. Pishas, Paul M. Neilsen, David F. Callen, Irene Zinonos, Michael P. Brown, Andreas Evdokiou, Fares Al-Ejeh, Raman Kumar, Pishas, Kathleen I, Al-Ejeh, Fares, Zinonos, Irene, Kumar, Raman, Evdokiou, Andreas, Brown, Michael P, Callen, David F, and Neilsen, Paul M
- Subjects
p53 ,Cancer Research ,Vincristine ,Nutlin-3a ,Cell cycle checkpoint ,medicine.medical_treatment ,Apoptosis ,Cell Cycle Proteins ,Sarcoma, Ewing ,Biology ,Piperazines ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,cancer ,Doxorubicin ,Molecular Targeted Therapy ,TP53 ,Etoposide ,Chemotherapy ,Imidazoles ,Cancer ,Nuclear Proteins ,Drug Synergism ,Proto-Oncogene Proteins c-mdm2 ,medicine.disease ,Genes, p53 ,Oncology ,Immunology ,biology.protein ,Cancer research ,Mdm2 ,Sarcoma ,Ewing sarcoma ,medicine.drug - Abstract
Purpose: Although mutations in the TP53 gene occur in half of all cancers, approximately 90% of Ewing sarcomas retain a functional wild-type p53. The low frequency of TP53 alterations in Ewing sarcoma makes this tumor type an ideal candidate for p53-targeted therapies. In this study, we have examined the molecular and cellular responses of cultured Ewing sarcoma cell lines following exposure to Nutlin-3a, a recently developed MDM2 antagonist. Experimental Design: The ability of Nutlin-3a to impart apoptosis or cell cycle arrest in a p53-dependent manner was determined in a comprehensive panel of Ewing sarcoma cell lines. The capacity of Nutlin-3a to augment the antitumor activity of MDM4 antagonists and cytotoxic agents currently used in the clinical treatment of Ewing sarcoma was also investigated. Results: Apoptosis was the primary response of wild-type p53 expressing Ewing sarcoma cell lines. The cytotoxicity of Nultin-3a was also synergistic with the chemotherapeutic agents, vincristine, actinomycin D, doxorubicin, and etoposide in a concentration-dependent manner. Significant MDM4 protein overexpression was observed in Ewing sarcoma cell lines of wild-type p53 status, providing a mechanism through which Ewing sarcomas can develop in the absence of TP53 alterations. This study provides the first evidence of synergism between targeted inhibition of MDM2 and MDM4. Conclusion: Our findings suggest that p53-dependent apoptosis is the primary cellular response of Ewing sarcoma cell lines following exposure to Nutlin-3a. Furthermore, Nutlin-3a can synergize with the current Ewing sarcoma chemotherapy protocols, suggesting p53 activation as a novel systemic therapeutic approach for this disease. Clin Cancer Res; 17(3); 494–504. ©2010 AACR.
- Published
- 2011
133. A role for the phosphatidylinositol 3-kinase--protein kinase C zeta--Sp1 pathway in the 1,25-dihydroxyvitamin D3 induction of the 25-hydroxyvitamin D3 24-hydroxylase gene in human kidney cells
- Author
-
Brian K. May, Xiu-Hui Gao, Antonio Ferrante, Prem P. Dwivedi, Charles S. T. Hii, Joseph Cheng-Ta Tan, Andreas Evdokiou, Howard A. Morris, Dwivedi, Prem, Gao, Xiu-Hui, Tan, Joseph Cheng-Ta, Evdokiou, Andreas, Ferrante, Antonio, Morris, Howard Arthur, May, Brian, and Hii, Charles
- Subjects
Sp1 Transcription Factor ,Morpholines ,CAAT box ,Biology ,Kidney ,Cell Line ,Phosphatidylinositol 3-Kinases ,Calcitriol ,Humans ,Phosphorylation ,Promoter Regions, Genetic ,Vitamin D3 24-Hydroxylase ,Protein Kinase C ,Phosphoinositide-3 Kinase Inhibitors ,Regulation of gene expression ,Kinase ,HEK 293 cells ,Promoter ,Cell Biology ,Molecular biology ,VDRE ,Chromones ,Mutation ,Steroid Hydroxylases ,Signal transduction - Abstract
The molecular mechanisms that underlie non-genomic induction of the 25-hydroxyvitamin D3 24-hydroxylase (CYP24) gene promoter by the steroid hormone, 1,25-Dihydroxyvitamin D3 (1,25D), are poorly understood. Although we have previously identified a functional inverted GC-box in the early promoter at − 113/−105 bp, it is not known whether this site is important for 1,25D induction of the promoter. Using transfected human embryonic kidney (HEK) 293 T cells, we now report the functional characterisation of the GC-box and that 1,25D induction of the promoter requires PI3-kinase, PKCζ and Sp1 but not Sp3. The data show that 1,25D rapidly stimulates PI3-kinase activity which is required for the activation of PKCζ and the phosphorylation of Sp1. The effects of the PI3-kinase inhibitor, LY294002, and a dominant negative PKCζ mutant on 1,25D induction of wild-type and a GC-box mutated CYP24 promoter constructs are consistent with the Sp1 site being the target of both kinases. However, these kinases are not required for basal expression of the CYP24 promoter. The data establish a novel non-genomic mechanism which couples 1,25D to the induction of CYP24 gene transcription via the PI3-kinase – PKCζ – Sp1 pathway acting through the GC-box.
- Published
- 2009
134. Pro-inflammatory cytokines TNF-related weak inducer of apoptosis (TWEAK) and TNFa induce the mitogen-activated protein kinase (MAPK)-dependent expression of sclerostin in human osteoblasts
- Author
-
Cristina Vincent, Gerald J. Atkins, Nicola L. Fazzalari, Timothy S. Zheng, Katie J. Welldon, David R. Haynes, Andreas Evdokiou, Asiri R. Wijenayaka, David M. Findlay, Vincent, Cristina, Findlay, David M, Welldon, Katie J, Wijenayaka, Asiri R, Zheng, Timothy S, Haynes, David R, Fazzalari, Nicola L, Evdokiou, Andreas, and Atkins, Gerald J
- Subjects
Genetic Markers ,MAPK/ERK pathway ,Transcription, Genetic ,Endocrinology, Diabetes and Metabolism ,Blotting, Western ,Immunology ,Medical Physiology ,TNF ,Fluorescent Antibody Technique ,sclerostin ,Biology ,Proinflammatory cytokine ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,Humans ,Orthopedics and Sports Medicine ,Cytokine TWEAK ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,DNA Primers ,TNF-related weak inducer of apoptosis ,Osteoblasts ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,Wnt signaling pathway ,Osteoblast ,3T3 Cells ,Flow Cytometry ,Recombinant Proteins ,RUNX2 ,medicine.anatomical_structure ,chemistry ,Osteocyte ,Bone Morphogenetic Proteins ,Tumor Necrosis Factors ,Cancer research ,osteoblast ,Sclerostin ,Inflammation Mediators ,Mitogen-Activated Protein Kinases ,SOST - Abstract
We have recently shown that TNF‐related weak inducer of apoptosis (TWEAK) is a mediator of inflammatory bone remodeling. The aim of this study was to investigate the role of TWEAK in modulating human osteoblast activity, and how TWEAK and TNFα might interact in this context. Recombinant TWEAK and TNF were both mitogenic for human primary osteoblasts (NHBC). TWEAK dose‐ and time‐dependently regulated the expression of the osteoblast transcription factors RUNX2 and osterix. TWEAK inhibited in vitro mineralization and downregulated the expression of osteogenesis‐associated genes. Significantly, TWEAK and TWEAK/TNF induced the expression of the osteoblast differentiation inhibitor and SOST gene product, sclerostin. Sclerostin induction was mitogen‐activated protein kinase (MAPK) dependent. The SOST mRNA levels induced by TWEAK were equivalent to or exceeded those seen in steady‐state human bone, and the TWEAK/TNF induction of SOST mRNA was recapitulated in fresh cancellous bone explants. TWEAK‐induced sclerostin expression was observed in immature osteoblastic cells, both in cycling (Ki67+) primary NHBC and in the cell lines MC3T3‐E1 and MG‐63, as well as in human osteocyte‐like cells and in the osteocyte cell line, MLO‐Y4. Treatment of NHBC with recombinant human sclerostin mimicked the effects of TWEAK to suppress RUNX2 and osteocalcin (OCN). TWEAK, TNF, and sclerostin treatment of NHBC similarly altered levels of phosphorylated and total GSK3β and active and total levels of β‐catenin, implying that the Wnt signaling pathway was affected by all three stimuli. Sclerostin also rapidly activated ERK‐1/2 MAPK signaling, indicating the involvement of additional signaling pathways. Together, our findings suggest that TWEAK, alone and with TNF, can regulate osteoblast function, at least in part by inducing sclerostin expression. Our results also suggest new roles and modes of action for sclerostin. Refereed/Peer-reviewed
- Published
- 2009
135. Apo2L/TRAIL inhibits tumor growth and bone destruction in a murine model of multiple myeloma
- Author
-
Gerald J. Atkins, Vasilios Liapis, Peter Diamond, David M. Findlay, Cristina Vincent, Agatha Labrinidis, Andrew C.W. Zannettino, Irene Zinonos, Shelley Hay, Sally Martin, Vladimir Ponomarev, Andreas Evdokiou, Natalie A. Sims, Labrinidis, Agatha, Diamond, Peter, Martin, Sally, Hay, Shelley, Liapis, Vasilios, Zinonos, Irene, Sims, Natalie A, Atkins, Gerald J, Vincent, Cristina, Ponomarev, Vladimir, Findlay, David M, Zannettino, Andrew CW, and Evdokiou, Andreas
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Osteolysis ,Medullary cavity ,Transplantation, Heterologous ,Clinical Sciences ,Oncology and Carcinogenesis ,Osteoclasts ,tumor progression ,Bone and Bones ,Article ,TNF-Related Apoptosis-Inducing Ligand ,in vivo study ,Mice ,Osteoprotegerin ,Cell Line, Tumor ,hemic and lymphatic diseases ,Animals ,Humans ,Bioluminescence imaging ,Medicine ,Multiple myeloma ,business.industry ,Cell Differentiation ,bioluminescence imaging ,medicine.disease ,Recombinant Proteins ,Transplantation ,multiple myeloma ,Disease Models, Animal ,Oncology ,Tumor progression ,osteoprotegerin ,Cancer cell ,Female ,Multiple Myeloma ,business ,Neoplasm Transplantation - Abstract
Purpose: Multiple myeloma is an incurable disease, for which the development of new therapeutic approaches is required. Here, we report on the efficacy of recombinant soluble Apo2L/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to inhibit tumor progression and bone destruction in a xenogeneic model of human multiple myeloma.Experimental Design: We established a mouse model of myeloma, in which Apo2L/TRAIL-sensitive RPMI-8226 or KMS-11 cells, tagged with a triple reporter gene construct (NES-HSV-TK/GFP/Luc), were transplanted directly into the tibial marrow cavity of nude mice. Tumor burden was monitored progressively by bioluminescence imaging and the development of myeloma-induced osteolysis was measured using high resolution in vivo micro-computed tomography.Results: Tumor burden increased progressively in the tibial marrow cavity of mice transplanted with Apo2L/TRAIL-sensitive RPMI-8226 or KMS-11 cells associated with extensive osteolysis directly in the area of cancer cell transplantation. Treatment of mice with recombinant soluble Apo2L/TRAIL reduced myeloma burden in the bone marrow cavity and significantly protected against myeloma-induced osteolysis. The protective effects of Apo2L/TRAIL treatment on bone were mediated by the direct apoptotic actions of Apo2L/TRAIL on myeloma cells within the bone microenvironment.Conclusions: This is the first in vivo study that investigates the efficacy of recombinant Apo2L/TRAIL on myeloma burden within the bone microenvironment and associated myeloma-induced bone destruction. Our findings that recombinant soluble Apo2L/TRAIL reduces myeloma burden within the bone microenvironment and protects the bone from myeloma-induced bone destruction argue against an inhibitory role of osteoprotegerin in Apo2L/TRAIL-induced apoptosis in vivo and highlight the need to clinically evaluate Apo2L/TRAIL in patients with multiple myeloma.
- Published
- 2009
136. RANK Expression as a cell surface marker of human osteoclast precursors in peripheral blood, bone marrow, and giant cell tumors of bone
- Author
-
Jeffrey P Houchins, Cristina Vincent, David M. Findlay, Panagiota Kostakis, Andreas Evdokiou, Gerald J. Atkins, Andrew C.W. Zannettino, Amanda N. Farrugia, Atkins, GJ, Kostakis, P, Vincent, Cristina, Farrugis, A, Houchins, J, Findlay, David, Evdokiou, Andreas, and Zannettino, ACW
- Subjects
Endocrinology, Diabetes and Metabolism ,Medical Physiology ,CD34 ,Lipopolysaccharide Receptors ,Osteoclasts ,Bone Marrow Cells ,Biology ,Peripheral blood mononuclear cell ,CD19 ,Monocytes ,Blood cell ,Osteoclast ,medicine ,Humans ,Orthopedics and Sports Medicine ,Cell Lineage ,Giant Cell Tumor of Bone ,B-Lymphocytes ,Membrane Glycoproteins ,Receptor Activator of Nuclear Factor-kappa B ,RANK Ligand ,Antibodies, Monoclonal ,Hematopoietic Stem Cells ,Molecular biology ,Recombinant Proteins ,Haematopoiesis ,medicine.anatomical_structure ,Immunology ,Antigens, Surface ,biology.protein ,Bone marrow ,Carrier Proteins ,Giant cell tumor ,Hematopoietic cells ,Monoclonal antibodies ,RANK - Abstract
RANK expression in vivo on hematopoietic subsets including pre-osteoclasts, identified by monoclonal antibodies, has not been described. We describe the lineages that express RANK in bone marrow, peripheral blood, and GCTs. We show that CD14+RANKhigh cells constitute a circulating pre-osteoclast pool. Introduction: The expression of RANK by subsets of hematopoietic cells has not been adequately studied in humans. While attributed to the monocytoid lineage, the phenotype of the pre-osteoclast (pre-OC) with respect to RANK expression in vivo remains unclear. We tested monoclonal antibodies (MAbs) raised against the extracellular domain of recombinant human RANK for reactivity with normal peripheral blood (PB) and bone marrow (BM) mononuclear cells (PBMNCs and BMMNCs, respectively). We also tested reactivity with giant cell tumor cells (GCT), a confirmed source of pre-OC and mature OCs. Materials and Methods: Human PBMNCs, BMMNCs, and GCT cells were analyzed for reactivity with anti-RANK MAbs by flow cytometry in combination with hematopoietic lineage restricted markers. GCTs were also analyzed by immunofluorescence. CD14+ monocytoid cells were sorted by fluorescence-activated cell sorting (FACS) based on their relative RANK expression and cultured under OC-forming conditions. Results: RANK+ cells were detected similarly by three independent anti-RANK MAbs. One MAb (80736) immunoprecipitated RANK–RANKL complexes from surface-biotinylated GCT lysates. Using dual-color flow cytometry, RANK was detected on CD14+ (monocytoid), CD19+ (B-lymphoid), CD56+ (NK cell), and glycophorin A+ erythroid progenitors. Minor populations of both CD3+ T lymphocytes and BM CD34+ hematopoietic progenitors also expressed cell surface RANK. In GCTs, RANK expression was identified on mononuclear CD45+CD14+αVβ3+c-Fms+ cells, likely to be committed pre-OC, and on multinucleated CD45+αVβ3+TRACP+ OCs. Importantly, sorted CD14+RANKhigh PBMNCs treated with recombinant RANKL and macrophage-colony stimulating factor (M-CSF) gave rise to approximately twice the number of osteoclasts than RANKmid or RANKlow cells. Conclusions: These results suggest that committed monocytoid RANK+ pre-OCs are represented in the marrow and circulate in the periphery, forming a pool of cells capable of responding rapidly to RANKL. The ability to reliably detect committed pre-OC in peripheral blood could have important clinical applications in the management of diseases characterized by abnormal osteoclastic activity.
- Published
- 2006
137. Doxorubicin overcomes resistance to drozitumab by antagonizing Inhibitor of Apoptosis Proteins (IAPs).
- Author
-
Zinonos I, Labrinidis A, Liapis V, Hay S, Panagopoulos V, Denichilo M, Ponomarev V, Ingman W, Atkins GJ, Findlay DM, Zannettino AC, and Evdokiou A
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Apoptosis drug effects, Cell Line, Tumor, Drug Synergism, Female, Humans, Mice, Mice, Nude, Xenograft Model Antitumor Assays, Antibiotics, Antineoplastic therapeutic use, Antibodies, Monoclonal therapeutic use, Breast Neoplasms drug therapy, Doxorubicin therapeutic use, Drug Resistance, Neoplasm drug effects, Inhibitor of Apoptosis Proteins antagonists & inhibitors, Receptors, TNF-Related Apoptosis-Inducing Ligand biosynthesis
- Abstract
Background/aim: Drozitumab is a fully human agonistic monoclonal antibody that binds to death receptor DR5 and induces apoptosis. However, drozitumab resistance is a major obstacle limiting anticancer efficacy., Materials and Methods: We examined the potential for the chemotherapeutic agent doxorubicin to overcome resistance against drozitumab-resistant breast cancer cells both in vitro and in vivo., Results: Treatment with doxorubicin increased cell surface expression of DR5, reduced levels of Inhibitors of Apoptosis Proteins (cIAPs) and re-sensitised cells to drozitumab-induced apoptosis. Animals implanted with resistant breast cancer cells into the mammary fat pad and treated with a combination of drozitumab and doxorubicin showed inhibition of tumor growth and a substantial delay in tumor progression compared to untreated controls and mice treated with each agent alone., Conclusion: These results suggest that combination of drozitumab with chemotherapy and agents that modulate IAP levels could potentially be a useful strategy in the treatment of breast cancer., (Copyright© 2014 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved.)
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.