Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stuart J. Gallagher is active.

Publication


Featured researches published by Stuart J. Gallagher.


PLOS ONE | 2015

Inducible but not constitutive expression of PD-L1 in human melanoma cells is dependent on activation of NF-κB

Kavitha Gowrishankar; Dilini Gunatilake; Stuart J. Gallagher; Jessamy Tiffen; Helen Rizos; Peter Hersey

Monoclonal antibodies against immune checkpoint blockade have proven to be a major success in the treatment of melanoma. The programmed death receptor-1 ligand-1 (PD-L1) expression on melanoma cells is believed to have an inhibitory effect on T cell responses and to be an important escape mechanism from immune attack. Previous studies have shown that PD-L1 can be expressed constitutively or can be induced by IFN-γ secreted by infiltrating lymphocytes. In the present study we have investigated the mechanism underlying these two modes of PD-L1 expression in melanoma cells including cells that had acquired resistance to the BRAF inhibitor vemurafenib. PD-L1 expression was examined by flow cytometry and immunoblotting. Specific inhibitors and siRNA knockdown approaches were used to examine the roles of the RAF/ MEK, PI3K, NF-κB, STAT3 and AP1/ c-Jun pathways. IFN-γ inducible expression of PD-L1 was dependent on NF-κB as shown by inhibition with BMS-345541, an inhibitor of IκB and the BET protein inhibitor I-BET151, as well as by siRNA knockdown of NF-κB subunits. We were unable to implicate the BRAF/MEK pathway as major regulators in PD-L1 expression on vemurafenib resistant cells. Similarly the PI3K/AKT pathway and the transcription factors STAT3 and c-Jun had only minor roles in IFN-γ induced expression of PD-L1. The mechanism underlying constitutive expression remains unresolved. We suggest these results have significance in selection of treatments that can be used in combination with monoclonal antibodies against PD1, to enhance their effectiveness and to reduce inhibitory effects melanoma cells have against cytotoxic T cell activity.


Carcinogenesis | 2013

The BH3-mimetic ABT-737 sensitizes human melanoma cells to apoptosis induced by selective BRAF inhibitors but does not reverse acquired resistance

David Wroblewski; Branka Mijatov; Nethia Mohana-Kumaran; Fritz Lai; Stuart J. Gallagher; Nikolas K. Haass; Xu Dong Zhang; Peter Hersey

Although the introduction of selective v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitors has been a major advance in treatment of metastatic melanoma, approximately 50% of patients have limited responses including stabilization of disease or no response at all. This study aims to identify a novel means of overcoming resistance of melanoma to killing by BRAF inhibitors. We examined the influence of the BH3-mimetic ABT-737 on induction of apoptosis by the selective BRAF inhibitor PLX4720 in melanoma cells with or without BRAF V600E mutation. Included were cell lines established from four patients before and during treatment with selective BRAF inhibitors and 3D spheroids derived from these cell lines. Cell lines with no or low sensitivity to PLX4720 underwent synergistic increases and increased rates of apoptosis when combined with ABT-737. This degree of synergism was not seen in cell lines without BRAF V600E mutations. Apoptosis was mediated through the mitochondrial pathway and was due in part to upregulation of Bim as shown by inhibition of apoptosis following small interfering RNA knockdown of Bim. Similar effects were seen in cell lines established from patients prior to treatment but not in lines from patients clinically resistant to the selective BRAF inhibitors and in 3D spheroids derived from these cell lines. These results suggest that combination of selective BRAF inhibitors with ABT-737 or the related orally available compound ABT-263 may increase the degree and rate of responses in previously untreated patients with V600E melanoma but not in those with acquired resistance to these agents.


Pigment Cell & Melanoma Research | 2014

Control of NF‐kB activity in human melanoma by bromodomain and extra‐terminal protein inhibitor I‐BET151

Stuart J. Gallagher; Branka Mijatov; Dilini Gunatilake; Kavitha Gowrishankar; Jessamy Tiffen; Wilmott James; Lei Jin; Gulietta M. Pupo; Carleen Cullinane; Grant A. McArthur; Peter J. Tummino; Helen Rizos; Peter Hersey

The transcription factor NF‐kappaB (NF‐kB) is a key regulator of cytokine and chemokine production in melanoma and is responsible for symptoms such as anorexia, fatigue, and weight loss. In addition, NF‐kB is believed to contribute to progression of the disease by upregulation of cell cycle and anti‐apoptotic genes and to contribute to resistance against targeted therapies and immunotherapy. In this study, we have examined the ability of the bromodomain and extra‐terminal (BET) protein inhibitor I‐BET151 to inhibit NF‐kB in melanoma cells. We show that I‐BET151 is a potent, selective inhibitor of a number of NF‐kB target genes involved in induction of inflammation and cell cycle regulation and downregulates production of cytokines such as IL‐6 and IL‐8. SiRNA studies indicate that BRD2 is the main BET protein involved in regulation of NF‐kB and that I‐BET151 caused transcriptional downregulation of the NF‐kB subunit p105/p50. These results suggest that BET inhibitors may have an important role in treatment of melanoma where activation of NF‐kB may have a key pathogenic role.


Oncotarget | 2015

Targeting activating mutations of EZH2 leads to potent cell growth inhibition in human melanoma by derepression of tumor suppressor genes

Jessamy Tiffen; Dilini Gunatilake; Stuart J. Gallagher; Kavitha Gowrishankar; Anja Heinemann; Carleen Cullinane; Ken Dutton-Regester; Gulietta M. Pupo; Dario Strbenac; Jean Y. Yang; Jason Madore; Graham J. Mann; Nicholas K. Hayward; Grant A. McArthur; Fabian V. Filipp; Peter Hersey

The epigenetic modifier EZH2 is part of the polycomb repressive complex that suppresses gene expression via histone methylation. Activating mutations in EZH2 are found in a subset of melanoma that contributes to disease progression by inactivating tumor suppressor genes. In this study we have targeted EZH2 with a specific inhibitor (GSK126) or depleted EZH2 protein by stable shRNA knockdown. We show that inhibition of EZH2 has potent effects on the growth of both wild-type and EZH2 mutant human melanoma in vitro particularly in cell lines harboring the EZH2Y646 activating mutation. This was associated with cell cycle arrest, reduced proliferative capacity in both 2D and 3D culture systems, and induction of apoptosis. The latter was caspase independent and mediated by the release of apoptosis inducing factor (AIFM1) from mitochondria. Gene expression arrays showed that several well characterized tumor suppressor genes were reactivated by EZH2 inhibition. This included activating transcription factor 3 (ATF3) that was validated as an EZH2 target gene by ChIP-qPCR. These results emphasize a critical role for EZH2 in the proliferation and viability of melanoma and highlight the potential for targeted therapy against EZH2 in treatment of patients with melanoma.


Advances in pharmacology | 2012

Histone Deacetylases (HDACs) as Mediators of Resistance to Apoptosis in Melanoma and as Targets for Combination Therapy with Selective BRAF Inhibitors

Fritz Lai; Lei Jin; Stuart J. Gallagher; Branka Mijatov; Xu Dong Zhang; Peter Hersey

HDACs are viewed as enzymes used by cancer cells to inhibit tumor suppressor mechanisms. In particular, we discuss their role as suppressors of apoptosis in melanoma cells and as mediators of resistance to selective BRAF inhibitors. Synergistic increases in apoptosis are seen when pan-HDAC inhibitors are combined with selective BRAF inhibitors. Moreover, cell lines from patients with acquired resistance to Vemurafenib undergo PLX4720 induced apoptosis when combined with pan-HDAC inhibitors. The mechanisms of upregulation of HDACs and the mechanisms involved in HDACi reversal of resistance to apoptosis are as yet poorly understood.


Cell Cycle | 2005

Enforced expression of p14ARF induces p53-dependent cell cycle arrest but not apoptosis.

Stuart J. Gallagher; Richard F. Kefford; Helen Rizos

Expression of the p14ARF tumour suppressor is induced by hyperproliferative signals produced by RAS, MYC and other oncogenes. p14ARF quenches inappropriate mitogenic signaling by activating the p53 pathway, and the frequent loss of p14ARF in human cancer diminishes the duration and level of the p53 response. Consistent with this role, p14ARF accumulation can induce potent cell cycle arrest, but its role in promoting apoptosis has not been well established. Therefore we investigated the effects of p14ARF on the survival and growth of several human cell types. To avoid the toxicity associated with adenoviral-based vectors, we established inducible expression of p14ARF in p53-intact and p53-deficient human cell lines. As expected, transient and inducible expression of p14ARF induced rapid cell cycle arrest only in tumour cells expressing intact p53. Further, p14ARF expression did not promote apoptosis in primary human fibroblasts, or in any human tumour cell line tested, irrespective of p53 status. Instead, p14ARF expression sensitized cells to apoptosis in the presence of inhibitors of topoisomerase II (adriamycin) and transcription (DRB). Thus, loss of p14ARF would be an important step in the selection of apoptotic resistant tumour cells.


Pigment Cell & Melanoma Research | 2015

EZH2: an emerging role in melanoma biology and strategies for targeted therapy

Jessamy Tiffen; Stuart J. Gallagher; Peter Hersey

Histone modifications are increasingly being recognized as important epigenetic mechanisms that govern chromatin structure and gene expression. EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2), responsible for tri‐methylation of lysine 27 on histone 3 (H3K27me3) that leads to gene silencing. This highly conserved histone methyltransferase is found to be overexpressed in many different types of cancers including melanoma, where it is postulated to abnormally repress tumor suppressor genes. Somatic mutations have been identified in approximately 3% of melanomas, and activating mutations described within the catalytic SET domain of EZH2 confer its oncogenic activity. In the following review, we discuss the evidence that EZH2 is an important driver of melanoma progression and we summarize the progress of EZH2 inhibitors against this promising therapeutic target.


Neoplasia | 2016

Somatic Copy Number Amplification and Hyperactivating Somatic Mutations of EZH2 Correlate With DNA Methylation and Drive Epigenetic Silencing of Genes Involved in Tumor Suppression and Immune Responses in Melanoma

Jessamy Tiffen; Stephen Wilson; Stuart J. Gallagher; Peter Hersey; Fabian V. Filipp

The epigenetic modifier EZH2 is in the center of a repressive complex controlling differentiation of normal cells. In cancer EZH2 has been implicated in silencing tumor suppressor genes. Its role in melanoma as well as target genes affected by EZH2 are poorly understood. In view of this we have used an integrated systems biology approach to analyze 471 cases of skin cutaneous melanoma (SKCM) in The Cancer Genome Atlas (TCGA) for mutations and amplifications of EZH2. Identified changes in target genes were validated by interrogation of microarray data from melanoma cells treated with the EZH2 inhibitor GSK126. We found that EZH2 activation by mutations, gene amplification and increased transcription occurred in about 20% of the cohort. These alterations were associated with significant hypermethylation of DNA and significant downregulation of 11% of transcripts in patient RNASeq data. GSK126 treatment of melanoma lines containing EZH2 activation reversed such transcriptional repression in 98 candidate target genes. Gene enrichment analysis revealed genes associated with tumor suppression, cell differentiation, cell cycle inhibition and repression of metastases as well as antigen processing and presentation pathways. The identified changes in EZH2 were associated with an adverse prognosis in the TCGA dataset. These results suggest that inhibiting of EZH2 is a promising therapeutic avenue for a substantial fraction of melanoma patients.


Clinical Cancer Research | 2013

A Focus on PD-L1 in Human Melanoma

Peter Hersey; Stuart J. Gallagher

Treatment of metastatic melanoma with inhibitors of the BRAF V600 oncogene in melanoma has been limited by the development of resistance. Combining the BRAF inhibitors with immunotherapy may prolong the response, but will acquisition of resistance to BRAF inhibitors also make melanoma cells resistant to immunotherapy? Clin Cancer Res; 19(3); 514–6. ©2012 AACR.


Cell Cycle | 2010

Amino terminal hydrophobic import signals target the p14ARF tumor suppressor to the mitochondria

Mal Irvine; Suzanah Philipsz; Monika Frausto; Branka Mijato; Stuart J. Gallagher; Carina Fung; Therese M. Becker; Richard F. Kefford; Helen Rizos

The p14ARF tumour suppressor is frequently targeted for inactivation in many human cancers and in individuals predisposed to cutaneous melanoma. The functions of p14ARF are closely linked with its subcellular distribution. Nucleolar p14ARF dampens ribosome biosynthesis and nucleoplasmic forms of p14ARF activate the p53 pathway and induce cell cycle arrest. p14ARF can also be recruited to mitochondria where it interacts with many mitochondrial proteins, including Bcl-xL and p32 to induce cell death. It has been suggested that the movement of p14ARF to mitochondria requires its interaction with p32, but we now show that the ARF-p32 interaction is not necessary for the accumulation of p14ARF in mitochondria. Instead, highly hydrophobic domains within the amino-terminal half of p14ARF act as mitochondrial import sequences. We suggest that once this hydrophobic pocket is exposed, possibly in a stimulus-dependent manner, it accelerates the mitochondrial import of p14ARF. This allows the interaction of p14ARF with mitochondrial proteins, including p32 and enables p53-independent cell death.

Collaboration


Dive into the Stuart J. Gallagher's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dilini Gunatilake

Kolling Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lei Jin

University of Sydney

View shared research outputs
Top Co-Authors

Avatar

Carleen Cullinane

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Grant A. McArthur

Peter MacCallum Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge