66 results on '"Karen E. Sheppard"'
Search Results
2. CDK4/6 Inhibition Promotes Antitumor Immunity through the Induction of T-cell Memory
- Author
-
Matteo Costacurta, Edwin D. Hawkins, Ricky W. Johnstone, Grant A. McArthur, Jane Oliaro, Paul J Neeson, Paul A. Beavis, Stephin J. Vervoort, Joe Jiang Zhu, Deborah Meyran, Conor J. Kearney, Emily J. Lelliott, Ian A. Parish, Pilar M. Dominguez, Laura Kirby, Nicole M. Haynes, Lydia Lim, Luciano G. Martelotto, Kelly M Ramsbottom, Magnus Zethoven, Isabella Y. Kong, Karen E. Sheppard, Jarrod J. Sandow, and Izabela Todorovski
- Subjects
0301 basic medicine ,Pyridines ,medicine.medical_treatment ,T cell ,Antineoplastic Agents ,Breast Neoplasms ,Piperazines ,Memory T Cells ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,Cell Line, Tumor ,PD-L1 ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,biology ,business.industry ,Cyclin-Dependent Kinase 4 ,Cancer ,Cyclin-Dependent Kinase 6 ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Chimeric antigen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,CDK4/6 Inhibition ,business - Abstract
Pharmacologic inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6) are an approved treatment for hormone receptor–positive breast cancer and are currently under evaluation across hundreds of clinical trials for other cancer types. The clinical success of these inhibitors is largely attributed to well-defined tumor-intrinsic cytostatic mechanisms, whereas their emerging role as immunomodulatory agents is less understood. Using integrated epigenomic, transcriptomic, and proteomic analyses, we demonstrated a novel action of CDK4/6 inhibitors in promoting the phenotypic and functional acquisition of immunologic T-cell memory. Short-term priming with a CDK4/6 inhibitor promoted long-term endogenous antitumor T-cell immunity in mice, enhanced the persistence and therapeutic efficacy of chimeric antigen receptor T cells, and induced a retinoblastoma-dependent T-cell phenotype supportive of favorable responses to immune checkpoint blockade in patients with melanoma. Together, these mechanistic insights significantly broaden the prospective utility of CDK4/6 inhibitors as clinical tools to boost antitumor T-cell immunity. Significance: Immunologic memory is critical for sustained antitumor immunity. Our discovery that CDK4/6 inhibitors drive T-cell memory fate commitment sheds new light on their clinical activity, which is essential for the design of clinical trial protocols incorporating these agents, particularly in combination with immunotherapy, for the treatment of cancer. This article is highlighted in the In This Issue feature, p. 2355
- Published
- 2021
- Full Text
- View/download PDF
3. Is resistance to targeted therapy in cancer inevitable?
- Author
-
Karen E. Sheppard, Grant A. McArthur, and Lorey K. Smith
- Subjects
Cancer Research ,Resistance (ecology) ,business.industry ,medicine.medical_treatment ,Neural crest ,Cancer ,medicine.disease ,Targeted therapy ,Oncology ,Neural Crest ,Cancer cell ,Cancer research ,Humans ,Medicine ,business ,Melanoma - Abstract
Resistance to targeted therapies is a major challenge in cancer care and occurs via genetic and non-genetic mechanisms. In this issue of Cancer Cell, Marin-Bejar et al. demonstrate that melanomas recurrently select genetic or non-genetic resistance trajectories and that targeting neural crest stem cell-like cells prevents non-genetic, but not genetic, resistance.
- Published
- 2021
- Full Text
- View/download PDF
4. CX-5461 activates the DNA damage response and demonstrates therapeutic efficacy in high-grade serous ovarian cancer
- Author
-
Carleen Cullinane, Olga Kondrashova, Grant A. McArthur, Els M.J.J. Berns, Ross D. Hannan, Katherine M. Hannan, Jessica E. Ahern, Jian Kang, Elaine Sanij, Jinbae Son, Elizabeth Lieschke, John Soong, Keefe T. Chan, Natalie Brajanovski, Anna S Trigos, Karen E. Sheppard, Clare L. Scott, Matthew Wakefield, Linda Mileshkin, Jiachen Xuan, Purba Nag, Richard B. Pearson, Kum Kum Khanna, Andrew J. Deans, Daniel Frank, Shunfei Yan, Gretchen Poortinga, Sarah Ellis, and Medical Oncology
- Subjects
0301 basic medicine ,endocrine system diseases ,Molecular biology ,General Physics and Astronomy ,Mice, SCID ,Poly (ADP-Ribose) Polymerase Inhibitor ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Mice, Inbred NOD ,RNA Polymerase I ,Transcription (biology) ,Medicine ,Enzyme Inhibitors ,Homologous Recombination ,lcsh:Science ,Mice, Knockout ,Ovarian Neoplasms ,Multidisciplinary ,Cancer therapeutic resistance ,030220 oncology & carcinogenesis ,Heterografts ,Female ,DNA Replication ,Cell biology ,DNA damage ,Science ,Poly(ADP-ribose) Polymerase Inhibitors ,Models, Biological ,Article ,General Biochemistry, Genetics and Molecular Biology ,Replication fork protection ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Cell Line, Tumor ,Animals ,Humans ,Benzothiazoles ,Naphthyridines ,Rucaparib ,business.industry ,DNA replication ,General Chemistry ,Cystadenocarcinoma, Serous ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,Cancer research ,lcsh:Q ,Transcriptome ,business ,Homologous recombination ,DNA ,DNA Damage - Abstract
Acquired resistance to PARP inhibitors (PARPi) is a major challenge for the clinical management of high grade serous ovarian cancer (HGSOC). Here, we demonstrate CX-5461, the first-in-class inhibitor of RNA polymerase I transcription of ribosomal RNA genes (rDNA), induces replication stress and activates the DNA damage response. CX-5461 co-operates with PARPi in exacerbating replication stress and enhances therapeutic efficacy against homologous recombination (HR) DNA repair-deficient HGSOC-patient-derived xenograft (PDX) in vivo. We demonstrate CX-5461 has a different sensitivity spectrum to PARPi involving MRE11-dependent degradation of replication forks. Importantly, CX-5461 exhibits in vivo single agent efficacy in a HGSOC-PDX with reduced sensitivity to PARPi by overcoming replication fork protection. Further, we identify CX-5461-sensitivity gene expression signatures in primary and relapsed HGSOC. We propose CX-5461 is a promising therapy in combination with PARPi in HR-deficient HGSOC and also as a single agent for the treatment of relapsed disease., Acquired resistance limits the efficacy of PARP inhibitors (PARPi) in high grade serous ovarian cancer (HGSOC). Here, the authors show that inhibition of RNA polymerase I transcription using CX-5461 increases the therapeutic efficacy of PARPi and overcomes PARPi resistance in PDX models of HGSOC.
- Published
- 2020
- Full Text
- View/download PDF
5. Metabolic Plasticity in Melanoma Progression and Response to Oncogene Targeted Therapies
- Author
-
Grant A. McArthur, Karen E. Sheppard, Arwa Alkaraki, and Lorey K. Smith
- Subjects
Cancer Research ,Oncogene ,business.industry ,Melanoma ,medicine.medical_treatment ,Metabolic reprogramming ,Disease progression ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Review ,medicine.disease ,Bioinformatics ,targeted therapy ,Targeted therapy ,Metastasis ,resistance ,Oncology ,plasticity ,Metabolome ,melanoma ,Medicine ,metastasis ,Treatment resistance ,business ,metabolism ,RC254-282 - Abstract
Simple Summary Targeted anti-cancer therapies have revolutionised melanoma patient care; however, cures remain uncommon due to acquired drug resistance that results in disease relapse. Recent insights from the clinic and experimental settings have identified a key role for metabolic plasticity, defined as the flexibility to utilise different nutrients and process them in different ways, in both disease progression and response to targeted therapies. Here, we discuss how this plasticity creates a moving target with important implications for identifying new combination therapies. Abstract Resistance to therapy continues to be a barrier to curative treatments in melanoma. Recent insights from the clinic and experimental settings have highlighted a range of non-genetic adaptive mechanisms that contribute to therapy resistance and disease relapse, including transcriptional, post-transcriptional and metabolic reprogramming. A growing body of evidence highlights the inherent plasticity of melanoma metabolism, evidenced by reversible metabolome alterations and flexibility in fuel usage that occur during metastasis and response to anti-cancer therapies. Here, we discuss how the inherent metabolic plasticity of melanoma cells facilitates both disease progression and acquisition of anti-cancer therapy resistance. In particular, we discuss in detail the different metabolic changes that occur during the three major phases of the targeted therapy response—the early response, drug tolerance and acquired resistance. We also discuss how non-genetic programs, including transcription and translation, control this process. The prevalence and diverse array of these non-genetic resistance mechanisms poses a new challenge to the field that requires innovative strategies to monitor and counteract these adaptive processes in the quest to prevent therapy resistance.
- Published
- 2021
6. PRMT5: An Emerging Target for Pancreatic Adenocarcinoma
- Author
-
Grant A. McArthur, Karen E. Sheppard, Michael K. C. Lee, and Sean M. Grimmond
- Subjects
Cancer Research ,endocrine system diseases ,protein methyltransferase ,Review ,medicine.disease_cause ,Therapeutic approach ,alternative splicing ,Stroma ,FBXW7 ,Pancreatic cancer ,medicine ,pancreatic adenocarcinoma ,RC254-282 ,biology ,business.industry ,Protein arginine methyltransferase 5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,GSK3β ,medicine.disease ,digestive system diseases ,Ubiquitin ligase ,c-Myc ,Oncology ,biology.protein ,Cancer research ,Adenocarcinoma ,PRMT5 ,Carcinogenesis ,business - Abstract
Simple Summary The burden of pancreatic ductal adenocarcinoma (PDAC) increases with rising incidence, yet 5-year overall survival remains poor at 17%. Routine comprehensive genomic profiling of PDAC only finds 2.5% of patients who may benefit and receive matched targeted therapy. Protein arginine methyltransferase 5 (PRMT5) as an anti-cancer target has gained significant interest in recent years and high levels of PRMT5 protein are associated with worse survival outcomes across multiple cancer types. Inhibition of PRMT5 in pre-clinical models can lead to cancer growth inhibition. However, PRMT5 is involved in multiple cellular processes, thus determining its mechanism of action is challenging. While past reviews on PRMT5 have focused on its role in diverse cellular processes and past research studies have focused mainly on haematological malignancies and glioblastoma, this review provides an overview of the possible biological mechanisms of action of PRMT5 inhibition and its potential as a treatment in pancreatic cancer. Abstract The overall survival of pancreatic ductal adenocarcinoma (PDAC) remains poor and its incidence is rising. Targetable mutations in PDAC are rare, thus novel therapeutic approaches are needed. Protein arginine methyltransferase 5 (PRMT5) overexpression is associated with worse survival and inhibition of PRMT5 results in decreased cancer growth across multiple cancers, including PDAC. Emerging evidence also suggests that altered RNA processing is a driver in PDAC tumorigenesis and creates a partial dependency on this process. PRMT5 inhibition induces altered splicing and this vulnerability can be exploited as a novel therapeutic approach. Three possible biological pathways underpinning the action of PRMT5 inhibitors are discussed; c-Myc regulation appears central to its action in the PDAC setting. Whilst homozygous MTAP deletion and symmetrical dimethylation levels are associated with increased sensitivity to PRMT5 inhibition, neither measure robustly predicts its growth inhibitory response. The immunomodulatory effect of PRMT5 inhibitors on the tumour microenvironment will also be discussed, based on emerging evidence that PDAC stroma has a significant bearing on disease behaviour and response to therapy. Lastly, with the above caveats in mind, current knowledge gaps and the implications and rationales for PRMT5 inhibitor development in PDAC will be explored.
- Published
- 2021
7. Melanoma brain metastases that progress on BRAF-MEK inhibitors demonstrate resistance to ipilimumab-nivolumab that is associated with the Innate PD-1 Resistance Signature (IPRES)
- Author
-
David L Kok, Belinda Lee, Ramyar Molania, Alison Weppler, Shahneen Sandhu, Grant A. McArthur, Benjamin Solomon, Kortnye Smith, Han Xian Aw Yeang, Tony Papenfuss, Ismael A. Vergara, Peter Lau, Paul J Neeson, Amir Iravani, Karen E. Sheppard, Christopher Angel, Breon Feran, Arian Lasocki, Kate Drummond, Damien Kee, Richard J. Young, Prachi Bhave, and Lorey K. Smith
- Subjects
Oncology ,Adult ,Male ,Proto-Oncogene Proteins B-raf ,Cancer Research ,medicine.medical_specialty ,Immunology ,Programmed Cell Death 1 Receptor ,Ipilimumab ,Drug resistance ,central nervous system neoplasms ,Young Adult ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,melanoma ,Immunology and Allergy ,Humans ,tumor microenvironment ,Protein Kinase Inhibitors ,RC254-282 ,Aged ,Pharmacology ,Trametinib ,Mitogen-Activated Protein Kinase Kinases ,Clinical/Translational Cancer Immunotherapy ,business.industry ,Brain Neoplasms ,Melanoma ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dabrafenib ,Gene signature ,Middle Aged ,medicine.disease ,Nivolumab ,Molecular Medicine ,Female ,immunotherapy ,business ,medicine.drug - Abstract
BackgroundMelanoma brain metastases (MBMs) are a challenging clinical problem with high morbidity and mortality. Although first-line dabrafenib–trametinib and ipilimumab–nivolumab have similar intracranial response rates (50%–55%), central nervous system (CNS) resistance to BRAF-MEK inhibitors (BRAF-MEKi) usually occurs around 6 months, and durable responses are only seen with combination immunotherapy. We sought to investigate the utility of ipilimumab–nivolumab after MBM progression on BRAF-MEKi and identify mechanisms of resistance.MethodsPatients who received first-line ipilimumab–nivolumab for MBMs or second/third line ipilimumab–nivolumab for intracranial metastases with BRAFV600 mutations with prior progression on BRAF-MEKi and MRI brain staging from March 1, 2015 to June 30, 2018 were included. Modified intracranial RECIST was used to assess response. Formalin-fixed paraffin-embedded samples of BRAFV600 mutant MBMs that were naïve to systemic treatment (n=18) or excised after progression on BRAF-MEKi (n=14) underwent whole transcriptome sequencing. Comparative analyses of MBMs naïve to systemic treatment versus BRAF-MEKi progression were performed.ResultsTwenty-five and 30 patients who received first and second/third line ipilimumab–nivolumab, were included respectively. Median sum of MBM diameters was 13 and 20.5 mm for the first and second/third line ipilimumab–nivolumab groups, respectively. Intracranial response rate was 75.0% (12/16), and median progression-free survival (PFS) was 41.6 months for first-line ipilimumab–nivolumab. Efficacy of second/third line ipilimumab-nivolumab after BRAF-MEKi progression was poor with an intracranial response rate of 4.8% (1/21) and median PFS of 1.3 months. Given the poor activity of ipilimumab–nivolumab after BRAF-MEKi MBM progression, we performed whole transcriptome sequencing to identify mechanisms of drug resistance. We identified a set of 178 differentially expressed genes (DEGs) between naïve and MBMs with progression on BRAF-MEKi treatment (p value ConclusionsSecond-line ipilimumab–nivolumab for MBMs after BRAF-MEKi progression has poor activity. MBMs that are resistant to BRAF-MEKi that also conferred resistance to second-line ipilimumab–nivolumab showed enrichment of the IPRES gene signature.
- Published
- 2021
8. Regulation of PRMT5–MDM4 axis is critical in the response to CDK4/6 inhibitors in melanoma
- Author
-
Ygal Haupt, Prerana Ghosh, Carleen Cullinane, Natalie Brajanovski, Gretchen Poortinga, Ian P. Street, Emily J. Lelliott, Laura Kirby, Keefe T. Chan, Mark G. Devlin, Richard B. Pearson, Elaine Sanij, Shatha AbuHammad, Lorey K. Smith, Michael A. Davies, Anthony T. Papenfuss, Alison Slater, Kerry Ardley, Sue Haupt, Karen E. Sheppard, Peter Lau, David J. Curtis, Hendrik Falk, Stupple Paul Anthony, Huiqin Chen, Grant A. McArthur, Margarete Kleinschmidt, Zoe Bacolas, Teresa Ward, Claire Martin, Aparna D. Rao, and Ismael A. Vergara
- Subjects
p53 ,0301 basic medicine ,Medical Sciences ,CDK4 ,Palbociclib ,MDM4 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,neoplasms ,Methylosome ,Multidisciplinary ,biology ,Oncogene ,Chemistry ,Cyclin-dependent kinase 4 ,Melanoma ,Protein arginine methyltransferase 5 ,acquired resistance ,Cyclin-dependent kinase 2 ,Biological Sciences ,medicine.disease ,3. Good health ,030104 developmental biology ,PNAS Plus ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,PRMT5 ,Cyclin-dependent kinase 6 - Abstract
Significance Targeting CDK4/6 shows great promise in the treatment of many solid cancers, including melanoma. This study has uncovered a mechanism of action of CDK4/6 inhibitors in regulating the MDM4 oncogene and the tumor suppressor, p53. In melanoma, palbociclib activates p53 via modulating PRMT5-dependent alternate MDM4 pre-mRNA splicing, which results in a robust decrease in MDM4 protein expression. Loss of palbociclib regulation of the PRMT5–MDM4 axis leads to drug resistance. Dual inhibition of CDK4/6 and PRMT5 potently suppresses melanoma tumor growth and is well tolerated in vivo. Our findings not only have immediate implications for the advancement of CDK4/6 inhibition for treating melanoma, but also more generally offer insights into CDK4/6 mechanism of action., Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors are an established treatment in estrogen receptor-positive breast cancer and are currently in clinical development in melanoma, a tumor that exhibits high rates of CDK4 activation. We analyzed melanoma cells with acquired resistance to the CDK4/6 inhibitor palbociclib and demonstrate that the activity of PRMT5, a protein arginine methyltransferase and indirect target of CDK4, is essential for CDK4/6 inhibitor sensitivity. By indirectly suppressing PRMT5 activity, palbociclib alters the pre-mRNA splicing of MDM4, a negative regulator of p53, leading to decreased MDM4 protein expression and subsequent p53 activation. In turn, p53 induces p21, leading to inhibition of CDK2, the main kinase substituting for CDK4/6 and a key driver of resistance to palbociclib. Loss of the ability of palbociclib to regulate the PRMT5–MDM4 axis leads to resistance. Importantly, combining palbociclib with the PRMT5 inhibitor GSK3326595 enhances the efficacy of palbociclib in treating naive and resistant models and also delays the emergence of resistance. Our studies have uncovered a mechanism of action of CDK4/6 inhibitors in regulating the MDM4 oncogene and the tumor suppressor, p53. Furthermore, we have established that palbociclib inhibition of the PRMT5–MDM4 axis is essential for robust melanoma cell sensitivity and provide preclinical evidence that coinhibition of CDK4/6 and PRMT5 is an effective and well-tolerated therapeutic strategy. Overall, our data provide a strong rationale for further investigation of novel combinations of CDK4/6 and PRMT5 inhibitors, not only in melanoma but other tumor types, including breast, pancreatic, and esophageal carcinoma.
- Published
- 2019
- Full Text
- View/download PDF
9. First-in-Human RNA Polymerase I Transcription Inhibitor CX-5461 in Patients with Advanced Hematologic Cancers: Results of a Phase I Dose-Escalation Study
- Author
-
Nadine Hein, Ella R. Thompson, Richard B. Pearson, Natalie Brajanovski, Ross D. Hannan, Amit Khot, Simon J. Harrison, Grant A. McArthur, Piers Blombery, Elaine Sanij, Donald P. Cameron, Gretchen Poortinga, Karen E. Sheppard, John K.C. Lim, John Soong, Andrew Fellowes, Kylee H Maclachlan, and Emma Link
- Subjects
Adult ,Male ,0301 basic medicine ,Transcription, Genetic ,Nucleolus ,Ribosome biogenesis ,Antineoplastic Agents ,DNA, Ribosomal ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,RNA Polymerase I ,Transcription (biology) ,RNA polymerase I ,Humans ,Medicine ,Benzothiazoles ,Naphthyridines ,Adverse effect ,Anaplastic large-cell lymphoma ,Aged ,Neoplasm Staging ,business.industry ,Middle Aged ,medicine.disease ,Lymphoma ,030104 developmental biology ,Oncology ,Hematologic Neoplasms ,030220 oncology & carcinogenesis ,Toxicity ,Cancer research ,Female ,Neoplasm Grading ,business - Abstract
RNA polymerase I (Pol I) transcription of ribosomal RNA genes (rDNA) is tightly regulated downstream of oncogenic pathways, and its dysregulation is a common feature in cancer. We evaluated CX-5461, the first-in-class selective rDNA transcription inhibitor, in a first-in-human, phase I dose-escalation study in advanced hematologic cancers. Administration of CX-5461 intravenously once every 3 weeks to 5 cohorts determined an MTD of 170 mg/m2, with a predictable pharmacokinetic profile. The dose-limiting toxicity was palmar–plantar erythrodysesthesia; photosensitivity was a dose-independent adverse event (AE), manageable by preventive measures. CX-5461 induced rapid on-target inhibition of rDNA transcription, with p53 activation detected in tumor cells from one patient achieving a clinical response. One patient with anaplastic large cell lymphoma attained a prolonged partial response and 5 patients with myeloma and diffuse large B-cell lymphoma achieved stable disease as best response. CX-5461 is safe at doses associated with clinical benefit and dermatologic AEs are manageable. Significance: CX-5461 is a first-in-class selective inhibitor of rDNA transcription. This first-in-human study establishes the feasibility of targeting this process, demonstrating single-agent antitumor activity against advanced hematologic cancers with predictable pharmacokinetics and a safety profile allowing prolonged dosing. Consistent with preclinical data, antitumor activity was observed in TP53 wild-type and mutant malignancies. This article is highlighted in the In This Issue feature, p. 983
- Published
- 2019
- Full Text
- View/download PDF
10. Genome-wide RNAi screen for genes regulating glycolytic response to vemurafenib in BRAFV600 melanoma cells
- Author
-
Lorey K. Smith, Kaylene J. Simpson, Grant A. McArthur, Tiffany Parmenter, Karen E. Sheppard, Cathryn M. Gould, and Piyush B. Madhamshettiwar
- Subjects
Statistics and Probability ,Proto-Oncogene Proteins B-raf ,Data Descriptor ,Gene regulatory network ,Antineoplastic Agents ,Library and Information Sciences ,Biology ,Genome ,Education ,03 medical and health sciences ,0302 clinical medicine ,Targeted therapies ,RNA interference ,Cell Line, Tumor ,medicine ,Humans ,Glycolysis ,lcsh:Science ,Vemurafenib ,Gene ,neoplasms ,Melanoma ,030304 developmental biology ,0303 health sciences ,medicine.disease ,Cancer metabolism ,Computer Science Applications ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:Q ,RNA Interference ,Statistics, Probability and Uncertainty ,Information Systems ,medicine.drug - Abstract
Identification of mechanisms underlying sensitivity and response to targeted therapies, such as the BRAF inhibitor vemurafenib, is critical in order to improve efficacy of these therapies in the clinic and delay onset of resistance. Glycolysis has emerged as a key feature of the BRAF inhibitor response in melanoma cells, and importantly, the metabolic response to vemurafenib in melanoma patients can predict patient outcome. Here, we present a multiparameter genome-wide siRNA screening dataset of genes that when depleted improve the viability and glycolytic response to vemurafenib in BRAFV600 mutated melanoma cells. These datasets are suitable for analysis of genes involved in cell viability and glycolysis in steady state conditions and following treatment with vemurafenib, as well as computational approaches to identify gene regulatory networks that mediate response to BRAF inhibition in melanoma., Measurement(s) cell viability • glycolytic process Technology Type(s) immunofluorescence microscopy assay • absorbance Factor Type(s) drug treatment • gene silencing Sample Characteristic - Organism Homo sapiens Machine-accessible metadata file describing the reported data: 10.6084/m9.figshare.12885593
- Published
- 2020
11. Combined BRAF, MEK, and CDK4/6 Inhibition Depletes Intratumoral Immune-Potentiating Myeloid Populations in Melanoma
- Author
-
Grant A. McArthur, Riyaben P. Patel, Carleen Cullinane, Claire Martin, Anthony T. Papenfuss, Amanda J Oliver, Kelly M Ramsbottom, Emily J. Lelliott, Nicole M. Haynes, Stefano Mangiola, Peter Lau, Lydia Lim, Jane Oliaro, Laura Kirby, Karen E. Sheppard, Magnus Zethoven, Luciano G. Martelotto, and Alison Slater
- Subjects
0301 basic medicine ,Male ,Proto-Oncogene Proteins B-raf ,Cancer Research ,Myeloid ,Skin Neoplasms ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Mice, Transgenic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,neoplasms ,Melanoma ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,business.industry ,Cancer ,Cyclin-Dependent Kinase 4 ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Immunogenic cell death ,CDK4/6 Inhibition ,business - Abstract
Combined inhibition of BRAF, MEK, and CDK4/6 is currently under evaluation in clinical trials for patients with melanoma harboring a BRAFV600 mutation. While this triple therapy has potent tumor-intrinsic effects, the impact of this combination on antitumor immunity remains unexplored. Here, using a syngeneic BrafV600ECdkn2a−/−Pten−/− melanoma model, we demonstrated that triple therapy promoted durable tumor control through tumor-intrinsic mechanisms and promoted immunogenic cell death and T-cell infiltration. Despite this, tumors treated with triple therapy were unresponsive to immune checkpoint blockade (ICB). Flow cytometric and single-cell RNA sequencing analyses of tumor-infiltrating immune populations revealed that triple therapy markedly depleted proinflammatory macrophages and cross-priming CD103+ dendritic cells, the absence of which correlated with poor overall survival and clinical responses to ICB in patients with melanoma. Indeed, immune populations isolated from tumors of mice treated with triple therapy failed to stimulate T-cell responses ex vivo. While combined BRAF, MEK, and CDK4/6 inhibition demonstrates favorable tumor-intrinsic activity, these data suggest that collateral effects on tumor-infiltrating myeloid populations may impact antitumor immunity. These findings have important implications for the design of combination strategies and clinical trials that incorporate BRAF, MEK, and CDK4/6 inhibition with immunotherapy for the treatment of patients with melanoma.
- Published
- 2020
12. Targeting RNA Polymerase I transcription synergises with TOP1 inhibition in potentiating the DNA damage response in high-grade serous ovarian cancer
- Author
-
Ross D. Hannan, Elaine Sanij, Jian Kang, Carleen Cullinane, Piyush B. Madhamshettiwar, Kaylene J. Simpson, Richard B. Pearson, Karen E. Sheppard, Sarah Ellis, Shunfei Yan, Keefe T. Chan, and Katherine M. Hannan
- Subjects
biology ,DNA repair ,DNA damage ,Poly ADP ribose polymerase ,Topoisomerase ,Cancer research ,biology.protein ,medicine ,Topotecan ,Ataxia telangiectasia and Rad3 related ,Replication protein A ,Polymerase ,medicine.drug - Abstract
Limited effective therapeutic options are available for patients with recurrent high-grade serous carcinoma (HGSC), the most common histological subtype accounting for the majority of ovarian cancer deaths. We have shown efficacy in poly-ADP ribose polymerase (PARP) inhibitor-resistant HGSC for the RNA Polymerase I (Pol I) transcription inhibitor CX-5461 through its ability to activate a nucleolar-associated DNA damage response (DDR). Here, we screen the protein-coding genome to identify potential targets whose inhibition enhances the efficacy of CX-5461. We identify a network of cooperating inhibitory interactions, including components of homologous recombination (HR) DNA repair and DNA topoisomerase 1 (TOP1). We highlight that CX-5461 combined with topotecan, a TOP1 inhibitor used as salvage therapy in HGSC, induces robust cell cycle arrest and cell death in a panel of HR-proficient HGSC cell lines. The combination potentiates a nucleolar-associated DDR via recruitment of phosphorylated replication protein A (RPA) and ataxia telangiectasia and Rad3 related protein (ATR). CX-5461 plus low-dose topotecan cooperate to potently inhibit xenograft tumour growth, indicating the potential for this strategy to improve salvage therapeutic regimens to treat HGSC.
- Published
- 2019
- Full Text
- View/download PDF
13. CDK4/6 inhibition in cancer: the cell cycle splicing connection
- Author
-
Karen E. Sheppard and Shatha AbuHammad
- Subjects
0301 basic medicine ,Cancer Research ,CDK4 ,endocrine system diseases ,CDK6 ,03 medical and health sciences ,MDM4 ,0302 clinical medicine ,medicine ,Author’s Views ,melanoma ,TP53 ,neoplasms ,biology ,Oncogene ,p21 ,Kinase ,Melanoma ,Protein arginine methyltransferase 5 ,acquired resistance ,Cancer ,Cell cycle ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,pre-mRNA splicing ,biology.protein ,Cancer research ,Molecular Medicine ,PRMT5 ,Cyclin-dependent kinase 6 ,CDK4/6 Inhibition - Abstract
Cyclin-dependent kinase −4 and −6 (CDK4/6) inhibitors are currently being assessed in clinical trials for the treatment of many cancers including melanoma. While investigating the mechanisms of CDK4/6 inhibitor resistance in melanoma, we uncovered a mechanism of action of these inhibitors in regulating the expression of both the mouse double minute 4 (MDM4) oncogene and tumor protein p53 (TP53).
- Published
- 2019
14. Adaptive post-transcriptional reprogramming of metabolism limits response to targeted therapy in BRAFV600 melanoma
- Author
-
Grant A. McArthur, Margarete Kleinschmidt, David L Goode, Jian Kang, Anna S Trigos, Vihandha O. Wickramasinghe, Peter Lau, Rodney J. Hicks, Karen E. Sheppard, Teresa Ward, Lorey K. Smith, Tony Tiganis, Richard B. Pearson, Carleen Cullinane, Kaylene J. Simpson, Emily J. Lelliott, Aparna D. Rao, Tiffany Parmenter, Claire Martin, Ola Larsson, Eric P Kusnadi, and Julie Lorent
- Subjects
0303 health sciences ,Combination therapy ,business.industry ,Kinase ,medicine.medical_treatment ,Melanoma ,Cancer ,medicine.disease ,3. Good health ,Targeted therapy ,Gene expression profiling ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Gene expression ,Cancer research ,Medicine ,MRNA transport ,business ,neoplasms ,030304 developmental biology - Abstract
Despite the success of therapies targeting oncogenes in cancer, clinical outcomes are limited by a residual disease that results in relapse. This residual disease is characterized by drug-induced adaptation, that in melanoma includes altered metabolism. Here, we examined how targeted therapy reprograms metabolism in BRAF-mutant melanoma cells using a genome-wide RNAi screen and global gene expression profiling. This systematic approach revealed post-transcriptional regulation of metabolism following BRAF inhibition, involving selective mRNA transport and translation. As proof of concept we demonstrate the RNA binding kinase UHMK1 interacts with mRNAs that encode metabolic proteins and selectively controls their transport and translation during adaptation to BRAF targeted therapy. Inactivation of UHMK1 improves metabolic response to BRAF targeted therapy and delays resistance to BRAF and MEK combination therapy in vivo. Our data support a model wherein post-transcriptional gene expression pathways regulate metabolic adaptation underpinning targeted therapy response and suggest inactivation of these pathways may delay disease relapse.
- Published
- 2019
- Full Text
- View/download PDF
15. Inhibition of RNA Polymerase I Transcription Activates Targeted DNA Damage Response and Enhances the Efficacy of PARP Inhibitors in High-Grade Serous Ovarian Cancer
- Author
-
Purba Nag, Els M.J.J. Berns, Anna S Trigos, Sarah Ellis, Ross D. Hannan, Matthew Wakefield, Jian Kang, Karen E. Sheppard, Richard B. Pearson, Gretchen Poortinga, Clare L. Scott, Jinbae Son, Jiachen Xuan, Grant A. McArthur, John Soong, Carleen Cullinane, Jessica E. Ahern, Elaine Sanij, Linda Mileshkin, Daniel Frank, Shunfei Yan, Keefe T. Chan, Olga Kondrashova, Elizabeth Lieschke, Katherine M. Hannan, and Kum Kum Khanna
- Subjects
0303 health sciences ,business.industry ,DNA damage ,Poly ADP ribose polymerase ,medicine.disease ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Transcription (biology) ,030220 oncology & carcinogenesis ,Gene expression ,Cancer research ,medicine ,RNA polymerase I ,Ovarian cancer ,business ,Homologous recombination ,DNA ,030304 developmental biology - Abstract
High-grade serous ovarian cancer (HGSOC) accounts for the majority of ovarian cancer and has a dismal prognosis. PARP inhibitors (PARPi) have revolutionized disease management of patients with homologous recombination (HR) DNA repair-deficient HGSOC. However, acquired resistance to PARPi by complex mechanisms including HR restoration and stabilisation of replication forks is a major challenge in the clinic. Here, we demonstrate CX-5461, an inhibitor of RNA polymerase I transcription of ribosomal RNA genes (rDNA), induces replication stress at rDNA leading to activation of DNA damage response and DNA damage involving MRE11-dependent degradation of replication forks. CX-5461 cooperates with PARPi in exacerbating DNA damage and enhances synthetic lethal interactions of PARPi with HR deficiency in HGSOC-patient-derived xenograft (PDX)in vivo. We demonstrate CX-5461 has a different sensitivity spectrum to PARPi and destabilises replication forks irrespective of HR pathway status, overcoming two well-known mechanisms of resistance to PARPi. Importantly, CX-5461 exhibits single agent efficacy in PARPi-resistant HGSOC-PDX. Further, we identify CX-5461-sensitivity gene expression signatures in primary and relapsed HGSOC. Therefore, CX-5461 is a promising therapy alone and in combination therapy with PARPi in HR-deficient HGSOC. CX-5461 is also an exciting treatment option for patients with relapsed HGSOC tumors that have poor clinical outcome.
- Published
- 2019
- Full Text
- View/download PDF
16. A novel immunogenic mouse model of melanoma for the preclinical assessment of combination targeted and immune-based therapy
- Author
-
Laura Kirby, Shatha AbuHammad, Richard J. Young, Peter Lau, Grant A. McArthur, Claire Martin, Katrina Meeth, Karen E. Sheppard, Jessica Michie, Kelly M Ramsbottom, Nicole M. Haynes, Rachael Walker, Fernando Souza-Fonseca-Guimaraes, Jane Oliaro, Emily J. Lelliott, Carleen Cullinane, and Alison Slater
- Subjects
Male ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Skin Neoplasms ,Immunogen ,MAP Kinase Signaling System ,Ovalbumin ,medicine.medical_treatment ,lcsh:Medicine ,Mice, Transgenic ,Ipilimumab ,Pembrolizumab ,Article ,Targeted therapy ,Mice ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Immune system ,Costimulatory and Inhibitory T-Cell Receptors ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,lcsh:Science ,Melanoma ,Protein Kinase Inhibitors ,Cyclin-Dependent Kinase Inhibitor p16 ,Mitogen-Activated Protein Kinase Kinases ,Multidisciplinary ,business.industry ,lcsh:R ,PTEN Phosphohydrolase ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,lcsh:Q ,Drug Screening Assays, Antitumor ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Both targeted therapy and immunotherapy have been used successfully to treat melanoma, but the development of resistance and poor response rates to the individual therapies has limited their success. Designing rational combinations of targeted therapy and immunotherapy may overcome these obstacles, but requires assessment in preclinical models with the capacity to respond to both therapeutic classes. Herein, we describe the development and characterization of a novel, immunogenic variant of the BrafV600ECdkn2a−/−Pten−/− YUMM1.1 tumor model that expresses the immunogen, ovalbumin (YOVAL1.1). We demonstrate that, unlike parental tumors, YOVAL1.1 tumors are immunogenic in vivo and can be controlled by immunotherapy. Importantly, YOVAL1.1 tumors are sensitive to targeted inhibitors of BRAFV600E and MEK, responding in a manner consistent with human BRAFV600E melanoma. The YOVAL1.1 melanoma model is transplantable, immunogenic and sensitive to clinical therapies, making it a valuable platform to guide strategic development of combined targeted therapy and immunotherapy approaches in BRAFV600E melanoma.
- Published
- 2019
- Full Text
- View/download PDF
17. CDK4/6 Inhibition Reprograms Mitochondrial Metabolism in BRAFV600 Melanoma via a p53 Dependent Pathway
- Author
-
Shatha AbuHammad, Lorey K. Smith, Grant A. McArthur, Nancy T. Santiappillai, Karen E. Sheppard, Laura Kirby, and Alison Slater
- Subjects
0301 basic medicine ,Cancer Research ,CDK4 ,endocrine system diseases ,medicine.medical_treatment ,lcsh:RC254-282 ,BRAF ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,melanoma ,medicine ,neoplasms ,Beta oxidation ,Chemistry ,Kinase ,MEK inhibitor ,Melanoma ,Cell cycle ,targeted therapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Cell metabolism ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,CDK4/6 Inhibition ,metabolism - Abstract
Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors are being tested in numerous clinical trials and are currently employed successfully in the clinic for the treatment of breast cancers. Understanding their mechanism of action and interaction with other therapies is vital in their clinical development. CDK4/6 regulate the cell cycle via phosphorylation and inhibition of the tumour suppressor RB, and in addition can phosphorylate many cellular proteins and modulate numerous cellular functions including cell metabolism. Metabolic reprogramming is observed in melanoma following standard-of-care BRAF/MEK inhibition and is involved in both therapeutic response and resistance. In preclinical models, CDK4/6 inhibitors overcome BRAF/MEK inhibitor resistance, leading to sustained tumour regression, however, the metabolic response to this combination has not been explored. Here, we investigate how CDK4/6 inhibition reprograms metabolism and if this alters metabolic reprogramming observed upon BRAF/MEK inhibition. Although CDK4/6 inhibition has no substantial effect on the metabolic phenotype following BRAF/MEK targeted therapy in melanoma, CDK4/6 inhibition alone significantly enhances mitochondrial metabolism. The increase in mitochondrial metabolism in melanoma cells following CDK4/6 inhibition is fuelled in part by both glutamine metabolism and fatty acid oxidation pathways and is partially dependent on p53. Collectively, our findings identify new p53-dependent metabolic vulnerabilities that may be targeted to improve response to CDK4/6 inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
18. 1079MO Progression of BRAF mutant CNS metastases are associated with a transcriptional network bearing similarities with the innate PD-1 resistant signature (IPRES)
- Author
-
Damien Kee, Grant A. McArthur, Ramyar Molania, Christopher Angel, Kortnye Smith, Shahneen Sandhu, H.X. Aw Yeang, Karen E. Sheppard, B. Feran, Paul J Neeson, Ismael A. Vergara, Prachi Bhave, Lorey K. Smith, Arian Lasocki, Alison Weppler, David L Kok, Katharine J. Drummond, T. Papenfuss, Belinda Lee, and Peter Lau
- Subjects
Bearing (mechanical) ,Oncology ,law ,business.industry ,Mutant ,Cancer research ,Medicine ,Hematology ,Signature (topology) ,business ,law.invention - Published
- 2020
- Full Text
- View/download PDF
19. Abstract PR13: Inhibition of RNA polymerase I transcription activates targeted DNA damage response and enhances the efficacy of PARP inhibitors in high-grade serous ovarian cancer
- Author
-
Anna S Trigos, Gretchen Poortinga, Ross D. Hannan, Katherine M. Hannan, Jessica A Ahern, Matthew Wakefield, Elizabeth Lieschke, Natalie Brajanovski, Shunfei Yan, Karen E. Sheppard, Elaine Sanij, Keefe T. Chan, Kum Kum Khanna, Carleen Cullinane, Sarah Ellis, Jiachen Xuan, Grant A. McArthur, Richard B. Pearson, John Soong, Jinbae Son, Linda Mileshkin, Olga Kondrashova, Els M.J.J. Berns, and Clare L. Scott
- Subjects
Cancer Research ,DNA damage ,business.industry ,Poly ADP ribose polymerase ,02 engineering and technology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,medicine.disease ,01 natural sciences ,0104 chemical sciences ,chemistry.chemical_compound ,Oncology ,chemistry ,Transcription (biology) ,Gene expression ,medicine ,Cancer research ,RNA polymerase I ,0210 nano-technology ,Ovarian cancer ,Homologous recombination ,business ,DNA - Abstract
Introduction: PARP inhibitors (PARPi) have revolutionized disease management of patients with homologous recombination (HR) DNA repair-deficient high-grade serous ovarian cancer (HGSOC). However, acquired resistance to PARPi is a major challenge in the clinic. The specific inhibitor of RNA polymerase I (Pol I) transcription of ribosomal RNA genes (rDNA) has demonstrated single-agent antitumor activity in p53 wild-type and p53-mutant hematologic malignancies (first-in-human trial, dose escalation study of CX-5461 at Peter MacCallum Cancer Centre) (Khot et al., Cancer Discov 2019). CX-5461 has also been reported to exhibit synthetic lethality with BRCA1/2 deficiency through stabilization of G-quadruplex DNA (GQ) structures. Here, we investigate the efficacy of CX-5461 in treating HGSOC. Experimental Design: The mechanisms by which CX-5461 induces DNA damage response (DDR) and displays synthetic lethality in HR-deficient HGSOC cells are explored. We present in vivo data of mice bearing two functionally and genomically profiled HGSOC-patient-derived xenograft (PDX)s treated with CX-5461 and olaparib, alone and in combination. We also investigate CX-5461-sensitivity gene expression signatures in primary and relapsed HGSOC. Results: Utilizing ovarian cancer cell lines, we demonstrate that sensitivity to CX-5461 is associated with “BRCA1 mutation” and “MYC targets” gene expression signatures. In addition, sensitivity to CX-5461 is associated with high basal rates of Pol I transcription. Importantly, we demonstrate a novel mechanism for CX-5461 synthetic lethal interaction with HR deficiency mediated through the induction of replication stress at rDNA repeats. Our data reveal CX-5461-mediated DDR in HR-deficient cells does not involve stabilization of GQ structures as previously proposed. On the contrary, we show definitively that CX-5461 inhibits Pol I recruitment leading to rDNA chromatin defects including stabilization of R-loops, single-stranded DNA, and replication stress at the rDNA. Mechanistically, we demonstrate CX-5461 leads to replication-dependent DNA damage involving MRE11-dependent degradation of replication forks. Importantly, CX-5461 has a different sensitivity spectrum to olaparib and cooperates with PARPi in exacerbating replication stress, leading to enhanced therapeutic efficacy in HR-deficient HGSOC-PDX in vivo compared to single-agent treatment of both drugs. Further, CX-5461 exhibits single-agent efficacy in olaparib-resistant HGSOC-PDX overcoming PARPi-resistance mechanisms involving fork protection. Importantly, we identify CX-5461-sensitivity gene expression signatures in primary and relapsed HGSOC. Conclusions: CX-5461 is a promising therapy alone and in combination therapy with PARPi in HR-deficient HGSOC. CX-5461 also has exciting potential as a treatment option for patients with relapsed HGSOC tumors that have high MYC activity and poor clinical outcome; these patients currently have very limited effective treatment options. This abstract is also being presented as Poster A71. Citation Format: Elaine Sanij, Katherine Hannan, Jiachen Xuan, Shunfei Yan, Jessica A. Ahern, Anna S. Trigos, Natalie Brajanovski, Jinbae Son, Keefe T. Chan, Olga Kondrashova, Elizabeth Lieschke, Matthew J. Wakefield, Sarah Ellis, Carleen Cullinane, Gretchen Poortinga, Kum Kum Khanna, Linda Mileshkin, Grant A. McArthur, John Soong, Els M. Berns, Ross D. Hannan, Clare L. Scott, Karen E. Sheppard, Richard B. Pearson. Inhibition of RNA polymerase I transcription activates targeted DNA damage response and enhances the efficacy of PARP inhibitors in high-grade serous ovarian cancer [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research; 2019 Sep 13-16, 2019; Atlanta, GA. Philadelphia (PA): AACR; Clin Cancer Res 2020;26(13_Suppl):Abstract nr PR13.
- Published
- 2020
- Full Text
- View/download PDF
20. Obesity and the Impact on Cutaneous Melanoma: Friend or Foe?
- Author
-
Grant A. McArthur, Lorey K. Smith, Emily J. Lelliott, Shaghayegh Arabi, and Karen E. Sheppard
- Subjects
0301 basic medicine ,Oncology ,obesity ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Review ,lcsh:RC254-282 ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,melanoma ,Medicine ,Risk factor ,business.industry ,Melanoma ,Cancer ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,targeted therapy ,medicine.disease ,Obesity ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,immunotherapy ,business ,Body mass index - Abstract
Excess body weight has been identified as a risk factor for many types of cancers, and for the majority of cancers, it is associated with poor outcomes. In contrast, there are cancers in which obesity is associated with favorable outcomes and this has been termed the “obesity paradox”. In melanoma, the connection between obesity and the increased incidence is not as strong as for other cancer types with some but not all studies showing an association. However, several recent studies have indicated that increased body mass index (BMI) improves survival outcomes in targeted and immune therapy treated melanoma patients. The mechanisms underlying how obesity leads to changes in therapeutic outcomes are not completely understood. This review discusses the current evidence implicating obesity in melanoma progression and patient response to targeted and immunotherapy, and discusses potential mechanisms underpinning these associations.
- Published
- 2020
- Full Text
- View/download PDF
21. Abstract A061: Targeting PRMT5 enhances the response to CDK4/6 inhibitors in multiple cancer types
- Author
-
Grant A. McArthur, Karen E. Sheppard, and Shatha AbuHammad
- Subjects
Cancer Research ,Cell signaling ,Cell cycle checkpoint ,Cell growth ,Protein arginine methyltransferase 5 ,Cancer ,Palbociclib ,Biology ,medicine.disease ,Small hairpin RNA ,Oncology ,Cancer research ,medicine ,Mitotic catastrophe - Abstract
CDK4 is a central regulator of cell cycle progression and is frequently hyper-activated by various genomic changes in numerous cancer types. CDK4/6 inhibitors such as palbociclib are approved for treatment of HR-positive breast cancer and has shown efficacy across many cancers. However, CDK4/6 inhibitors are best used in combination therapy rather than as a monotherapy. Finding novel combinations that can enhance the efficacy of CDK4/6 inhibitors and overcome or delay the emergence of resistance may provide new treatment strategies for patients.PRMT5 is an emerging target for the treatment of cancer, with the newly developed inhibitor GSK3326595 currently in phase 1/2 clinical trials.PRMT5 is a type II protein methyl-transferase that primarily catalyses the formation of symmetric di-methylarginine modifications. PRMT5 methylates the arginine residue on Histones 2A, 3 and 4 and this is considered an important epigenetic regulatory mechanism. PRMT5 can also methylate a number of non-histone proteins that govern important cellular processes including, cell signalling, RNA nucleo-cytoplasmic transport, pre-RNA processing, and ribosome biogenesis. The combined inhibition of CDK4 and PRMT5 can be seen as a promising strategy for the treatment of many cancers. A high throughput drug screen with a boutique library of targeted therapeutics was performed in order to identify potential targets whose inhibition enhances palbociclib efficacy. The effectiveness of combination treatment was evaluated by measuring cell proliferation clonogenicity, and cell death. To understand the mechanism/s of response to CDK4 and PRMT5 inhibitors, we utilized RPPA and RNA sequencing assays. In addition to this, western blotting, PCR and real-time PCR were also used to study both gene and protein expression levels as well as to validate specific targets. Furthermore, gene depletion via shRNA was performed to validate the response to these treatments. Dual inhibition of CDK4/6 and PRMT5 resulted in a robust and more durable response than the single agents in breast cancer, oesophageal, pancreatic and melanoma cell lines. The response to this combination was observed in both p53-wild type and mutant cell lines. High throughput proteomic and genomic analysis revealed that Inhibition of CDK4/6 and/or PRMT5 acts in p53-dependent and independent mechanisms. In p53 wild type, the combination of palbociclib and GSKGSK3326595 resulted in a permenant state of cell cycle arrest, whereas in p53-mutant cells the combinaiton caused mitotic catastrophe and induced cell death. In conclusion, Inhibition of PRMT5 enhances the response to CDK4/6 inhibitors and the combination of palbociclib and GSK3326595 is a potentially promising treatment strategy for various cancers. Citation Format: Shatha AbuHammad, Grant A McArthur, Karen Sheppard. Targeting PRMT5 enhances the response to CDK4/6 inhibitors in multiple cancer types [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A061. doi:10.1158/1535-7163.TARG-19-A061
- Published
- 2019
- Full Text
- View/download PDF
22. Palbociclib synergizes with BRAF and MEK inhibitors in treatment naïve melanoma but not after the development of BRAF inhibitor resistance
- Author
-
Gretchen Poortinga, Carleen Cullinane, Rachael Walker, Ross D. Hannan, Shatha AbuHammad, Richard J. Young, Richard B. Pearson, Grant A. McArthur, Natalie Brajanovski, Rodney J. Hicks, Karen E. Sheppard, Kelly Waldeck, Elaine Sanij, Laura Kirby, Donald P. Cameron, Claire Martin, and Emily J. Lelliott
- Subjects
0301 basic medicine ,Proto-Oncogene Proteins B-raf ,Cancer Research ,Indoles ,endocrine system diseases ,Pyridines ,Mice, SCID ,Palbociclib ,Piperazines ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Vemurafenib ,neoplasms ,Melanoma ,Protein Kinase Inhibitors ,Sulfonamides ,biology ,Cyclin-dependent kinase 4 ,business.industry ,MEK inhibitor ,Cancer ,Cyclin-Dependent Kinase 4 ,Dabrafenib ,Drug Synergism ,Cyclin-Dependent Kinase 6 ,medicine.disease ,MAP Kinase Kinase Kinases ,Xenograft Model Antitumor Assays ,digestive system diseases ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Cyclin-dependent kinase 6 ,business ,medicine.drug - Abstract
Increased CDK4 activity occurs in the majority of melanomas and CDK4/6 inhibitors in combination with BRAF and MEK inhibitors are currently in clinical trials for the treatment of melanoma. We hypothesize that the timing of the addition of CDK4/6 inhibitors to the current BRAF and MEK inhibitor regime will impact on the efficacy of this triplet drug combination. The efficacy of BRAF, MEK and CDK4/6 inhibitors as single agents and in combination was assessed in human BRAF mutant cell lines that were treatment naive, BRAF inhibitor tolerant or had acquired resistance to BRAF inhibitors. Xenograft studies were then performed to test the in vivo efficacy of the BRAF and CDK4/6 inhibitor combination. Melanoma cells that had developed early reversible tolerance or acquired resistance to BRAF inhibition remained sensitive to palbociclib. In drug-tolerant cells, the efficacy of the combination of palbociclib with BRAF and/or MEK inhibitors was equivalent to single agent palbociclib. Similarly, acquired BRAF inhibitor resistance cells lost efficacy to the palbociclib and BRAF combination. In contrast, upfront treatment of melanoma cells with palbociclib in combination with BRAF and/or MEK inhibitors induced either cell death or senescence and was superior to a BRAF plus MEK inhibitor combination. In vivo palbociclib plus BRAF inhibitor induced rapid and sustained tumor regression without the development of therapy resistance. In summary, upfront dual targeting of CDK4/6 and mutant BRAF signaling enables tumor cells to evade resistance to monotherapy and is required for robust and sustained tumor regression. Melanoma patients whose tumors have acquired resistance to BRAF inhibition are less likely to have favorable responses to subsequent treatment with the triplet combination of BRAF, MEK and CDK4/6 inhibitors.
- Published
- 2017
23. Response of BRAF-Mutant Melanoma to BRAF Inhibition Is Mediated by a Network of Transcriptional Regulators of Glycolysis
- Author
-
Richard B. Pearson, Kathryn M. Kinross, Grant A. McArthur, Margarete Kleinschmidt, Aparna Rao, Richard A. Scolyer, Carleen Cullinane, Gulietta M. Pupo, Willy Hugo, Simon T. Bond, Mohan R. Kaadige, Roger S. Lo, Ricky W. Johnstone, Sean L. McGee, Helen Rizos, Rodney J. Hicks, Antoni Ribas, Jason Li, Georgina V. Long, Tiffany Parmenter, Karen E. Sheppard, and Donald E. Ayer
- Subjects
Proto-Oncogene Proteins B-raf ,Programmed cell death ,Indoles ,endocrine system diseases ,MAP Kinase Signaling System ,Pyridines ,Oncology and Carcinogenesis ,Mutant ,Drug Resistance ,Biology ,Article ,Piperazines ,Cell Line ,Clinical Research ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Glycolysis ,skin and connective tissue diseases ,Vemurafenib ,Melanoma ,Protein Kinase Inhibitors ,neoplasms ,Transcription factor ,Cancer ,Sulfonamides ,Tumor ,HEK 293 cells ,medicine.disease ,digestive system diseases ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,Oncology ,5.1 Pharmaceuticals ,Drug Resistance, Neoplasm ,Cell culture ,Cancer research ,Neoplasm ,Development of treatments and therapeutic interventions ,Transcription Factors ,medicine.drug - Abstract
Deregulated glucose metabolism fulfills the energetic and biosynthetic requirements for tumor growth driven by oncogenes. Because inhibition of oncogenic BRAF causes profound reductions in glucose uptake and a strong clinical benefit in BRAF-mutant melanoma, we examined the role of energy metabolism in responses to BRAF inhibition. We observed pronounced and consistent decreases in glycolytic activity in BRAF-mutant melanoma cells. Moreover, we identified a network of BRAF-regulated transcription factors that control glycolysis in melanoma cells. Remarkably, this network of transcription factors, including hypoxia-inducible factor-1α, MYC, and MONDOA (MLXIP), drives glycolysis downstream of BRAFV600, is critical for responses to BRAF inhibition, and is modulated by BRAF inhibition in clinical melanoma specimens. Furthermore, we show that concurrent inhibition of BRAF and glycolysis induces cell death in BRAF inhibitor (BRAFi)–resistant melanoma cells. Thus, we provide a proof-of-principle for treatment of melanoma with combinations of BRAFis and glycolysis inhibitors. Significance: BRAFis suppress glycolysis and provide strong clinical benefit in BRAFV600 melanoma. We show that BRAF inhibition suppresses glycolysis via a network of transcription factors that are critical for complete BRAFi responses. Furthermore, we provide evidence for the clinical potential of therapies that combine BRAFis with glycolysis inhibitors. Cancer Discov; 4(4); 423–33. ©2014 AACR. See related commentary by Haq, p. 390 This article is highlighted in the In This Issue feature, p. 377
- Published
- 2014
- Full Text
- View/download PDF
24. Loss ofCDKN2Aexpression is a frequent event in primary invasive melanoma and correlates with sensitivity to the CDK4/6 inhibitor PD0332991 in melanoma cell lines
- Author
-
Ken Dutton-Regester, Laura Kirby, Alexander Dobrovic, Kelly Waldeck, Karen E. Sheppard, Grant A. McArthur, Richard B. Pearson, Hongdo Do, Stephen B. Fox, Donald P. Cameron, Claire Martin, James G. Christensen, Wendy Liu, Nicholas Jene, Carleen Cullinane, Richard J. Young, Catherine Mitchell, Sophia Randolph, Nicholas K. Hayward, and Jung Hock Foo
- Subjects
Adult ,Male ,endocrine system diseases ,Pyridines ,Dermatology ,Biology ,Piperazines ,General Biochemistry, Genetics and Molecular Biology ,CDKN2A ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,Neoplasm Invasiveness ,Copy-number variation ,Melanoma ,Protein Kinase Inhibitors ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,Aged ,Aged, 80 and over ,Regulation of gene expression ,Genetics ,medicine.diagnostic_test ,Cyclin-Dependent Kinase 4 ,Cancer ,Cyclin-Dependent Kinase 6 ,Middle Aged ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Oncology ,Cutaneous melanoma ,Cancer research ,biology.protein ,Female ,Cyclin-dependent kinase 6 ,Fluorescence in situ hybridization - Abstract
We have investigated the potential for the p16-cyclin D-CDK4/6-retinoblastoma protein pathway to be exploited as a therapeutic target in melanoma. In a cohort of 143 patients with primary invasive melanoma, we used fluorescence in situ hybridization to detect gene copy number variations (CNVs) in CDK4, CCND1, and CDKN2A and immunohistochemistry to determine protein expression. CNVs were common in melanoma, with gain of CDK4 or CCND1 in 37 and 18% of cases, respectively, and hemizygous or homozygous loss of CDKN2A in 56%. Three-quarters of all patients demonstrated a CNV in at least one of the three genes. The combination of CCND1 gain with either a gain of CDK4 and/or loss of CDKN2A was associated with poorer melanoma-specific survival. In 47 melanoma cell lines homozygous loss, methylation or mutation of CDKN2A gene or loss of protein (p16(INK) (4A) ) predicted sensitivity to the CDK4/6 inhibitor PD0332991, while RB1 loss predicted resistance.
- Published
- 2014
- Full Text
- View/download PDF
25. The Cell-Cycle Regulator CDK4: An Emerging Therapeutic Target in Melanoma
- Author
-
Grant A. McArthur and Karen E. Sheppard
- Subjects
Cancer Research ,endocrine system diseases ,Cell ,Antineoplastic Agents ,CDKN2A ,medicine ,Animals ,Humans ,Melanoma ,Protein Kinase Inhibitors ,neoplasms ,integumentary system ,biology ,Cyclin-dependent kinase 4 ,Cell Cycle ,Retinoblastoma protein ,Cyclin-Dependent Kinase 4 ,Cell cycle ,medicine.disease ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,Oncology ,Apoptosis ,Immunology ,biology.protein ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Signal Transduction - Abstract
The recent clinical success of targeted therapies in melanoma directed at the oncogene BRAF validates the concept of targeting oncogenes. The p16-cyclin D-CDK4/6-retinoblastoma protein pathway (CDK4 pathway) is dysregulated in 90% of melanomas, and is, therefore, an obvious therapeutic target for this disease. The main outcome of CDK4 activation is the phosphorylation and, thus, inhibition of the retinoblastoma protein leading to G1–S cell-cycle transition. In addition, CDK4 directly phosphorylates other proteins that promote cell-cycle progression and inhibit both cell senescence and apoptosis. In preclinical studies, the response to CDK4 inhibition correlates with genomic changes that increase CDK4 activity, most notably where the tumor suppressor CDKN2A (p16INK4A) is deleted. A central question is whether melanomas with activating events in the CDK4 pathway have become “addicted” to this signaling pathway, in which case inhibition of CDK4 would not simply induce cell-cycle arrest but induce cell death and tumor regression. Recently, a number of selective CDK4/6 inhibitors have entered clinical trials, and these compounds are showing great promise in that they are well tolerated and show clinical benefit. This review discusses the CDK4 pathway, its dysregulation in melanoma, the consequences of CDK4 pathway inhibition, and potential novel combinational strategies for the treatment of melanoma. Clin Cancer Res; 19(19); 5320–8. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
26. Functional Analysis of Genes in Regions Commonly Amplified in High-Grade Serous and Endometrioid Ovarian Cancer
- Author
-
Ian G. Campbell, Sally J. Davis, Karen E. Sheppard, Kaylene J. Simpson, Richard B. Pearson, and Kylie L. Gorringe
- Subjects
Cancer Research ,DNA Copy Number Variations ,Protein Serine-Threonine Kinases ,Biology ,Bioinformatics ,Polymorphism, Single Nucleotide ,Breast cancer ,Cell Line, Tumor ,Gene duplication ,medicine ,Humans ,RNA, Small Interfering ,Gene ,In Situ Hybridization, Fluorescence ,Adaptor Proteins, Signal Transducing ,Neoplasm Staging ,Oligonucleotide Array Sequence Analysis ,Ovarian Neoplasms ,Intracellular Signaling Peptides and Proteins ,Cancer ,Protein-Tyrosine Kinases ,Amplicon ,Prognosis ,medicine.disease ,Primary tumor ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,Serous fluid ,p21-Activated Kinases ,Oncology ,Cancer research ,Female ,Ovarian cancer ,Carcinoma, Endometrioid - Abstract
Purpose: Ovarian cancer has the highest mortality rate of all the gynecologic malignancies and is responsible for approximately 140,000 deaths annually worldwide. Copy number amplification is frequently associated with the activation of oncogenic drivers in this tumor type, but their cytogenetic complexity and heterogeneity has made it difficult to determine which gene(s) within an amplicon represent(s) the genuine oncogenic driver. We sought to identify amplicon targets by conducting a comprehensive functional analysis of genes located in the regions of amplification in high-grade serous and endometrioid ovarian tumors. Experimental Design: High-throughput siRNA screening technology was used to systematically assess all genes within regions commonly amplified in high-grade serous and endometrioid cancer. We describe the results from a boutique siRNA screen of 272 genes in a panel of 18 ovarian cell lines. Hits identified by the functional viability screen were further interrogated in primary tumor cohorts to determine the clinical outcomes associated with amplification and gene overexpression. Results: We identified a number of genes as critical for cellular viability when amplified, including URI1, PAK4, GAB2, and DYRK1B. Integration of primary tumor gene expression and outcome data provided further evidence for the therapeutic use of such genes, particularly URI1 and GAB2, which were significantly associated with survival in 2 independent tumor cohorts. Conclusion: By taking this integrative approach to target discovery, we have streamlined the translation of high-resolution genomic data into preclinical in vitro studies, resulting in the identification of a number of genes that may be specifically targeted for the treatment of advanced ovarian tumors. Clin Cancer Res; 19(6); 1411–21. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
27. Desmoglein 2 promotes vasculogenic mimicry in melanoma and is associated with poor clinical outcome
- Author
-
Grant A. McArthur, Erwin Foster-Smith, Clare G Fedele, Zahied M. Johan, Brenton W Ebert, Lih Yin Tan, Mark Shackleton, Claudine S. Bonder, Lisa M. Ebert, Pacman Szeto, Karen E. Sheppard, Chris P Mintoff, You Fang Zhang, Andrew Ruszkiewicz, Michael P. Brown, Tan, Lih Yin, Mintoff, Chris, Johan, M Zahied, Ebert, Brenton W, Brown, Michael P, Bonder, Claudine S, and Ebert, Lisa M
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Angiogenesis ,desmoglein 2 ,Diagnosis, Differential ,03 medical and health sciences ,Cell Line, Tumor ,Cell Adhesion ,medicine ,melanoma ,Humans ,Vasculogenic mimicry ,Cell adhesion ,vasculogenic mimicry ,Tube formation ,Matrigel ,Desmoglein 2 ,Neovascularization, Pathologic ,Sequence Analysis, RNA ,Cadherin ,business.industry ,Melanoma ,TCGA ,medicine.disease ,3. Good health ,030104 developmental biology ,cadherin ,Oncology ,Tumor progression ,Cancer research ,Melanocytes ,business ,Research Paper - Abstract
Tumors can develop a blood supply not only by promoting angiogenesis but also by forming vessel-like structures directly from tumor cells, known as vasculogenic mimicry (VM). Understanding mechanisms that regulate VM is important, as these might be exploitable to inhibit tumor progression. Here, we reveal the adhesion molecule desmoglein 2 (DSG2) as a novel mediator of VM in melanoma. Analysis of patient-derived melanoma cell lines and tumor tissues, and interrogation of The Cancer Genome Atlas (TCGA) data, revealed that DSG2 is frequently overexpressed in primary and metastatic melanomas compared to normal melanocytes. Notably, this overexpression was associated with poor clinical outcome. DSG2+ melanoma cells self-organized into tube-like structures on Matrigel, indicative of VM activity, which was inhibited by DSG2 knockdown or treatment with a DSG2-blocking peptide. Mechanistic studies revealed that DSG2 regulates adhesion and cell-cell interactions during tube formation, but does not control melanoma cell viability, proliferation or motility. Finally, analysis of patient tumors revealed a correlation between DSG2 expression, VM network density and expression of VM-associated genes. These studies identify DSG2 as a key regulator of VM activity in human melanoma and suggest this molecule might be therapeutically targeted to reduce tumor blood supply and metastatic spread. Refereed/Peer-reviewed
- Published
- 2016
28. LRP1B deletion in high-grade serous ovarian cancers is associated with acquired chemotherapy resistance to liposomal doxorubicin
- Author
-
Sian Fereday, Peter Van Loo, Joy Hendley, Carleen Cullinane, Anna deFazio, Michael A. Quinn, Viola Heinzelmann-Schwarz, Michael Friedlander, Michael S. Anglesio, Peter J. Campbell, Joshy George, Dariush Etemadmoghadam, Karen E. Sheppard, Elizabeth L. Christie, Richard B. Pearson, Laura Galletta, Prue A. Cowin, Elizabeth Loehrer, David D.L. Bowtell, Orla McNally, Leanne Bowes, Paul R. Harnett, Sarah Ftouni, and Epidemiology
- Subjects
Cancer Research ,medicine.medical_treatment ,Gene Dosage ,Down-Regulation ,Gene Expression ,Drug resistance ,Cell Growth Processes ,Biology ,Metastasis ,SDG 3 - Good Health and Well-being ,Cell Line, Tumor ,Carcinoma ,medicine ,Humans ,Doxorubicin ,RNA, Small Interfering ,Lung cancer ,Aged ,Chromosome Aberrations ,Ovarian Neoplasms ,Chemotherapy ,Chromosomes, Human, X ,Chromosomes, Human, Pair 10 ,DNA, Neoplasm ,Middle Aged ,medicine.disease ,Cystadenocarcinoma, Serous ,Serous fluid ,Oncology ,Receptors, LDL ,Drug Resistance, Neoplasm ,Chromosomes, Human, Pair 2 ,Gene Knockdown Techniques ,Cancer research ,Female ,Neoplasm Grading ,Ovarian cancer ,Gene Deletion ,medicine.drug - Abstract
High-grade serous cancer (HGSC), the most common subtype of ovarian cancer, often becomes resistant to chemotherapy, leading to poor patient outcomes. Intratumoral heterogeneity occurs in nearly all solid cancers, including ovarian cancer, contributing to the development of resistance mechanisms. In this study, we examined the spatial and temporal genomic variation in HGSC using high-resolution single-nucleotide polymorphism arrays. Multiple metastatic lesions from individual patients were analyzed along with 22 paired pretreatment and posttreatment samples. We documented regions of differential DNA copy number between multiple tumor biopsies that correlated with altered expression of genes involved in cell polarity and adhesion. In the paired primary and relapse cohort, we observed a greater degree of genomic change in tumors from patients that were initially sensitive to chemotherapy and had longer progression-free interval compared with tumors from patients that were resistant to primary chemotherapy. Notably, deletion or downregulation of the lipid transporter LRP1B emerged as a significant correlate of acquired resistance in our analysis. Functional studies showed that reducing LRP1B expression was sufficient to reduce the sensitivity of HGSC cell lines to liposomal doxorubicin, but not to doxorubicin, whereas LRP1B overexpression was sufficient to increase sensitivity to liposomal doxorubicin. Together, our findings underscore the large degree of variation in DNA copy number in spatially and temporally separated tumors in HGSC patients, and they define LRP1B as a potential contributor to the emergence of chemotherapy resistance in these patients. Cancer Res; 72(16); 4060–73. ©2012 AACR.
- Published
- 2012
- Full Text
- View/download PDF
29. Enhanced GAB2 Expression Is Associated with Improved Survival in High-Grade Serous Ovarian Cancer and Sensitivity to PI3K Inhibition
- Author
-
Maria P. Intermaggio, Richard B. Pearson, Kaylene J. Simpson, Anna H. Wu, Usha Menon, Jacobus Pfisterer, Andreas du Bois, David G. Huntsman, Aleksandra Gentry-Maharaj, Sian Fereday, Ian G. Campbell, Nadia Traficante, Jacek Gronwald, David D.L. Bowtell, Kylie L. Gorringe, Jan Lubinski, Joshy George, Karen E. Sheppard, Malcolm C. Pike, Stefan Kommoss, Celeste Leigh Pearce, Susan J. Ramus, Felix Hilpert, Michael S. Anglesio, and Sally J. Davis
- Subjects
Adult ,Cancer Research ,DNA Copy Number Variations ,Cell Survival ,Pyridones ,Kaplan-Meier Estimate ,Biology ,Disease-Free Survival ,Metastasis ,Ovarian tumor ,Young Adult ,Ovarian carcinoma ,Cell Line, Tumor ,medicine ,Carcinoma ,Humans ,Gene Regulatory Networks ,Adaptor Proteins, Signal Transducing ,Aged ,Phosphoinositide-3 Kinase Inhibitors ,Aged, 80 and over ,Ovarian Neoplasms ,Oncogene ,Gene Expression Profiling ,Gene Amplification ,Middle Aged ,medicine.disease ,Cystadenocarcinoma, Serous ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Serous fluid ,Pyrimidines ,Oncology ,Multivariate Analysis ,Cancer research ,Female ,Neoplasm Grading ,Ovarian cancer - Abstract
Identification of genomic alterations defining ovarian carcinoma subtypes may aid the stratification of patients to receive targeted therapies. We characterized high-grade serous ovarian carcinoma (HGSC) for the association of amplified and overexpressed genes with clinical outcome using gene expression data from 499 HGSC patients in the Ovarian Tumor Tissue Analysis cohort for 11 copy number amplified genes: ATP13A4, BMP8B, CACNA1C, CCNE1, DYRK1B, GAB2, PAK4, RAD21, TPX2, ZFP36, and URI. The Australian Ovarian Cancer Study and The Cancer Genome Atlas datasets were also used to assess the correlation between gene expression, patient survival, and tumor classification. In a multivariate analysis, high GAB2 expression was associated with improved overall and progression-free survival (P = 0.03 and 0.02), whereas high BMP8B and ATP13A4 were associated with improved progression-free survival (P = 0.004 and P = 0.02). GAB2 overexpression and copy number gain were enriched in the AOCS C4 subgroup. High GAB2 expression correlated with enhanced sensitivity in vitro to the dual PI3K/mTOR inhibitor PF-04691502 and could be used as a genomic marker for identifying patients who will respond to treatments inhibiting PI3K signaling. Mol Cancer Ther; 14(6); 1495–503. ©2015 AACR.
- Published
- 2015
30. Cross talk between corticosteroids and alpha-adrenergic signalling augments cardiomyocyte hypertrophy: A possible role for SGK1
- Author
-
Kerrie Lister, Anna Jenkins, Karen E. Sheppard, Ross D. Hannan, and Dominic J. Autelitano
- Subjects
medicine.medical_specialty ,Transcription, Genetic ,Physiology ,medicine.drug_class ,Cardiomegaly ,Protein Serine-Threonine Kinases ,Biology ,Immediate-Early Proteins ,Muscle hypertrophy ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Adrenergic Agents ,Receptors, Glucocorticoid ,Mineralocorticoid receptor ,Glucocorticoid receptor ,Adrenal Cortex Hormones ,Corticosterone ,Physiology (medical) ,Internal medicine ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,medicine ,Animals ,Myocytes, Cardiac ,Receptor ,Aldosterone ,Cells, Cultured ,Rats ,Receptors, Mineralocorticoid ,Endocrinology ,Animals, Newborn ,Microscopy, Fluorescence ,chemistry ,Mineralocorticoid ,Signal transduction ,Cardiology and Cardiovascular Medicine ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,Signal Transduction ,medicine.drug - Abstract
Objective Mineralocorticoids and glucocorticoids have been implicated in the pathogenesis of cardiac diseases; however, both in vivo and in vitro studies indicate that changes in the cellular milieu of either the cardiomyocyte and/or cells of the vasculature is required for corticosteroid signalling to be pathological. The aim of the current study was to directly address whether signalling pathways that are activated during myocyte hypertrophy alter corticosteroid signalling and thus enable these steroids to significantly impact on the hypertrophic response. Methods Neonatal rat ventricular cardiomyocytes were treated with phenylephrine or phorbol ester for 48 h to induce myocyte hypertrophy. Following treatment, the expression of glucocorticoid receptor, mineralocorticoid receptor, and 11β-hydroxysteroid dehydrogenase were determined by ribonuclease protection assay. In addition, the activity of 11β-hydroxysteroid dehydrogenase and the ability of glucocorticoid and mineralocorticoid receptors to induce serum- and glucocorticoid-induced kinase 1 (SGK1) gene transcription were assessed. Corticosteroid effects on phenylephrine and phorbol ester-induced hypertrophy were determined by measuring atrial natriuretic peptide (ANP) mRNA expression, protein synthesis, or induction of rDNA transcription. Results Incubation of cardiomyocytes with phenylephrine and phorbol ester for 48 h led to a hypertrophic response with an associated 8- to 12-fold increase in ANP mRNA and 2-fold increase in rDNA transcription. Cardiomyocyte hypertrophy led to a significant 2-fold increase in glucocorticoid receptor and mineralocorticoid receptor expression that resulted in enhanced receptor signaling as judged via the ability of corticosterone and aldosterone to induce SGK1 gene transcription. 11β-Hydroxysteroid dehydrogenase2 was not detected in normal or hypertrophied cardiomyocytes, and 11β-hydroxysteroid dehydrogenase exclusively demonstrated reductase activity, converting the inactive 11-ketometabolite back to active glucocorticoid. 11β-Hydroxysteroid dehydrogenase1 expression and reductase activity were increased with phorbol ester-induced hypertrophy but not phenylephrine-induced hypertrophy. In basal cardiomyocytes, either aldosterone or corticosterone induced only a minor increase in ANP mRNA and protein synthesis; however, in cardiomyocytes primed with phenylephrine, both corticosteroids significantly potentiated phenylephrine-mediated effects via activation of the glucocorticoid receptor. Conclusion In the present study we demonstrate that significant cross talk exists in the cardiomyocyte between corticosteroid receptor-activated pathways and both protein kinase C and alpha-adrenergic signalling. Cellular changes associated with the hypertrophic response promote corticosteroid signalling and allow for corticosteroid-mediated potentiation of alpha-adrenergic receptor signalling. Such augmentation of cardiomyocyte hypertrophy may in part explain the role that corticosteroid hormones play in the pathophysiological progression of heart disease.
- Published
- 2006
- Full Text
- View/download PDF
31. Regression of pressure overload-induced left ventricular hypertrophy in mice
- Author
-
Xiao-Lei Moore, Anthony M. Dart, Karen E. Sheppard, Xinheng Feng, Helen Kiriazis, Xiao-Jun Du, and Xiao-Ming Gao
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Calcium-Transporting ATPases ,Left ventricular hypertrophy ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,Muscle hypertrophy ,Mice ,Physiology (medical) ,Internal medicine ,Pressure ,Ventricular Pressure ,Animals ,Medicine ,cardiovascular diseases ,DNA Primers ,Pressure overload ,Ventricular Remodeling ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Aortic constriction ,medicine.disease ,Matrix Metalloproteinases ,Regression ,Surgery ,Mice, Inbred C57BL ,Disease Models, Animal ,Echocardiography ,Circulatory system ,Time course ,Cardiology ,Female ,Hypertrophy, Left Ventricular ,Collagen ,Cardiology and Cardiovascular Medicine ,business ,Atrial Natriuretic Factor - Abstract
As a prelude to investigating the mechanism of regression of pressure overload-induced left ventricular (LV) hypertrophy (LVH), we studied the time course for the development and subsequent regression of LVH as well as accompanying alterations in cardiac function, histology, and gene expression. Mice were subjected to aortic banding for 4 or 8 wk to establish LVH, and regression was initiated by release of aortic banding for 6 wk. Progressive increase in LV mass and gradual chamber dilatation and dysfunction occurred after aortic banding. LVH was also associated with myocyte enlargement, interstitial fibrosis, and enhanced expression of atrial natriuretic peptide, collagen I, collagen III, and matrix metalloproteinase-2 but suppressed expression of α-myosin heavy chain and sarcoplasmic reticulum Ca2+-ATPase. Aortic debanding completely or partially reversed LVH, chamber dilatation and dysfunction, myocyte size, interstitial fibrosis, and gene expression pattern, each with a distinct time course. The extent of LVH regression was dependent on the duration of pressure overload, evidenced by the fact that restoration of LV structure and function was complete in animals subjected to 4 wk of aortic banding but incomplete in animals subjected to 8 wk of aortic banding. In conclusion, LVH regression comprises a variety of morphological, functional, and genetic components that show distinct time courses. A longer period of pressure overload is associated with a slower rate of LVH regression.
- Published
- 2005
- Full Text
- View/download PDF
32. UTP Transactivates Epidermal Growth Factor Receptors and Promotes Cardiomyocyte Hypertrophy Despite Inhibiting Transcription of the Hypertrophic Marker Gene, Atrial Natriuretic Peptide
- Author
-
Elizabeth A. Woodcock, James B. Morris, Karen E. Sheppard, Tam M. Pham, and Bronywn Kenney
- Subjects
Transcriptional Activation ,medicine.medical_specialty ,Cardiomegaly ,Uridine Triphosphate ,Biology ,Biochemistry ,Rats, Sprague-Dawley ,Atrial natriuretic peptide ,Epidermal growth factor ,Internal medicine ,Serum response factor ,medicine ,Animals ,Myocytes, Cardiac ,Promoter Regions, Genetic ,Receptor ,Enhancer ,Molecular Biology ,Purinergic receptor ,Promoter ,Cell Biology ,Rats ,ErbB Receptors ,Endocrinology ,Myocardin ,cardiovascular system ,Atrial Natriuretic Factor ,hormones, hormone substitutes, and hormone antagonists - Abstract
In neonatal rat ventricular myocytes, activation of receptors that couple to the G(q) family of heterotrimeric G proteins causes hypertrophic growth, together with expression of "hypertrophic marker" genes, such as atrial natriuretic peptide (ANP) and myosin light chain 2 (MLC2). As reported previously for other G(q)-coupled receptors, stimulation of alpha(1)-adrenergic receptors with phenylephrine (50 microM) caused phosphorylation of epidermal growth factor (EGF) receptors as well as activation of ERK1/2, cellular growth, and ANP transcription. These responses depended on EGF receptor activation. In marked contrast, stimulation of G(q)-coupled purinergic receptors with UTP caused EGF receptor phosphorylation, ERK1/2 activation, and cellular growth but minimal increases in ANP transcription. UTP inhibited phenylephrine-dependent transcription from ANP and MLC2 promoters but not transcription from myoglobin promoters or from AP-1 elements. Myocardin is a muscle-specific transcription enhancer that activates transcription from ANP and MLC2 promoters but not myoglobin promoters or AP-1 elements. UTP inhibited ANP and MLC2 responses to overexpressed myocardin but did not inhibit responses to c-Jun, GATA4, or serum response factor, all of which are active in nonmuscle cells. Thus, UTP inhibits transcriptional responses to phenylephrine only at cardiac-specific promoters, and this may involve the muscle-specific transcription enhancer, myocardin. These studies show that EGF receptor activation is necessary but not sufficient for ANP and MLC2 responses to activation of G(q)-coupled receptors in ventricular myocytes, because inhibitory mechanisms can oppose such stimulation. ANP is a compensatory and protective factor in cardiac hypertrophy, and mechanisms that reduce its generation need to be defined.
- Published
- 2004
- Full Text
- View/download PDF
33. Abstract A24: A genome-wide RNAi screen identifies synthetic lethality of CX-5461 with homologous recombination repair deficiency in ovarian cancer
- Author
-
Shunfei Yan, Ross D. Hannan, Kaylene J. Simpson, Keefe T. Chan, Richard B. Pearson, Elaine Sanij, Katherine M. Hannan, and Karen E. Sheppard
- Subjects
Cancer Research ,DNA repair ,Cell growth ,Ribosome biogenesis ,Synthetic lethality ,mTORC1 ,Biology ,medicine.disease ,Molecular biology ,Oncology ,Cancer research ,medicine ,Ovarian cancer ,Homologous recombination ,Molecular Biology ,Gene - Abstract
Cancer is characterized by deregulated cell growth and proliferation, both of which are associated with hyperactivation of ribosome biogenesis. Inhibition of ribosome biogenesis using CX-5461, a specific inhibitor of RNA polymerase I-dependent transcription, has shown therapeutic efficacy in a MYC driven B-cell lymphoma mouse model, which is enhanced when used in combination with the mTORC1 inhibitor Everolimus. However, the therapeutic potential of CX-5461 in solid cancers is yet to be determined. Our preliminary data utilizing a panel of 36 ovarian cancer (OVCA) cell lines suggest that acute CX-5461 treatment results in cell cycle arrest and does not induce apoptosis. We hypothesize that the identification of genes that can be targeted to cooperate with CX-5461 will define novel drug combinations for the improved treatment of OVCA. Therefore, we performed a genome-wide RNAi screen to identify synthetic lethal genes with CX-5461 in the high-grade serous ovarian cancer (HGSOC) cell line OVCAR4. Pathway enrichment analysis of the candidate hits showed significant enrichment in the homologous recombination DNA repair (HR) pathway. Synergy with CX-5461 was validated in multiple HGSOC cell lines by both genetic and pharmacological inhibition of HR pathway components. We are currently investigating the mechanism of this synergy and will further assess efficacy in vivo. As HR deficiency is observed in 20% of OVCA patients, we suggest that future application of our studies will lead to new therapeutic options to improve the survival of this cohort of patients. Citation Format: Shunfei Yan, Keefe T. Chan, Kaylene J. Simpson, Elaine Sanij, Karen E. Sheppard, Katherine M. Hannan, Ross D. Hannan, Richard B. Pearson. A genome-wide RNAi screen identifies synthetic lethality of CX-5461 with homologous recombination repair deficiency in ovarian cancer [abstract]. In: Proceedings of the AACR Special Conference on DNA Repair: Tumor Development and Therapeutic Response; 2016 Nov 2-5; Montreal, QC, Canada. Philadelphia (PA): AACR; Mol Cancer Res 2017;15(4_Suppl):Abstract nr A24.
- Published
- 2017
- Full Text
- View/download PDF
34. Whole exome sequencing identifies a recurrent RQCD1 P131L mutation in cutaneous melanoma
- Author
-
Stephen Q. Wong, Claire Martin, Alexander Dobrovic, Jonathan Cebon, Grant A. McArthur, Victoria Mar, John W Kelly, Jason Li, Andreas Behren, Rory Wolfe, Katherine Woods, and Karen E. Sheppard
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Oncology ,Male ,Models, Molecular ,Proto-Oncogene Proteins B-raf ,medicine.medical_specialty ,Skin Neoplasms ,Population ,Molecular Sequence Data ,Mutation, Missense ,Biology ,GTP Phosphohydrolases ,HLA Antigens ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Exome ,Amino Acid Sequence ,Neoplasm Metastasis ,Lentigo maligna melanoma ,education ,Melanoma ,Exome sequencing ,Genetics ,education.field_of_study ,Base Sequence ,Sequence Homology, Amino Acid ,Cancer ,Membrane Proteins ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Protein Structure, Tertiary ,Logistic Models ,Cutaneous melanoma ,Multivariate Analysis ,Female ,exome sequencing ,V600E ,Protein Binding ,Transcription Factors ,Research Paper ,RQCD1 - Abstract
// Stephen Q. Wong 1 , Andreas Behren 2, 3 , Victoria J. Mar 4, 5 , Katherine Woods 2, 3 , Jason Li 1 , Claire Martin 1 , Karen E. Sheppard 1, 6 , Rory Wolfe 5 , John Kelly 4 , Jonathan Cebon 2, 3 , Alexander Dobrovic 2, 3, 7 , Grant A. McArthur 1, 7 1 Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia 2 Ludwig Institute for Cancer Research, Olivia Newton-John Cancer and Wellness Centre Heidelberg, Victoria, Australia 3 School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia 4 Victorian Melanoma Service, Alfred Hospital, Prahran, Victoria, Australia 5 Department of Epidemiology and Preventive Medicine, Monash University, Clayton, Victoria, Australia 6 Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia 7 Department of Pathology, University of Melbourne, Parkville, Victoria, Australia Correspondence to: Stephen Q. Wong, e-mail: stephen.wong@petermac.org Keywords: Melanoma, RQCD1, exome sequencing Received: June 28, 2014 Accepted: November 11, 2014 Published: December 06, 2014 ABSTRACT Melanoma is often caused by mutations due to exposure to ultraviolet radiation. This study reports a recurrent somatic C > T change causing a P131L mutation in the RQCD1 (Required for Cell Differentiation1 Homolog) gene identified through whole exome sequencing of 20 metastatic melanomas. Screening in 715 additional primary melanomas revealed a prevalence of ~4%. This represents the first reported recurrent mutation in a member of the CCR4-NOT complex in cancer. Compared to tumors without the mutation, the P131L mutant positive tumors were associated with increased thickness ( p = 0.02), head and neck ( p = 0.009) and upper limb ( p = 0.03) location, lentigo maligna melanoma subtype ( p = 0.02) and BRAF V600K ( p = 0.04) but not V600E or NRAS codon 61 mutations. There was no association with nodal disease ( p = 0.3). Mutually exclusive mutations of other members of the CCR4-NOT complex were found in ~20% of the TCGA melanoma dataset suggesting the complex may play an important role in melanoma biology. Mutant RQCD1 was predicted to bind strongly to HLA-A0201 and HLA-Cw3 MHC1 complexes. From thirteen patients with mutant RQCD1 , an anti-tumor CD8 + T cell response was observed from a single patient’s peripheral blood mononuclear cell population stimulated with mutated peptide compared to wildtype indicating a neoantigen may be formed.
- Published
- 2014
35. Corticosteroid receptors and 11β-hydroxysteroid dehydrogenase isoforms in rat intestinal epithelia
- Author
-
Kevin X. Z. Li, Karen E. Sheppard, and Dominic J. Autelitano
- Subjects
Male ,Receptors, Steroid ,medicine.medical_specialty ,Physiology ,Carbenoxolone ,Ileum ,Biology ,Rats, Sprague-Dawley ,Jejunum ,Receptors, Glucocorticoid ,Glucocorticoid receptor ,Mineralocorticoid receptor ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,RNA, Messenger ,Intestinal Mucosa ,Receptor ,Hepatology ,Hydroxysteroid Dehydrogenases ,Gastroenterology ,Small intestine ,Rats ,Isoenzymes ,Receptors, Mineralocorticoid ,medicine.anatomical_structure ,Endocrinology ,11-beta-Hydroxysteroid Dehydrogenases ,Glucocorticoid ,medicine.drug - Abstract
To evaluate the potential roles that both receptors and enzymes play in corticosteroid regulation of intestinal function, we have determined glucocorticoid receptor (GR), mineralocorticoid receptor (MR), and 11β-hydroxysteroid dehydrogenase (11β-HSD) expression in intestinal epithelial cells. GR and MR mRNA and receptor binding were ubiquitously expressed in epithelial cells, with receptor levels higher in ileum and colon than jejunum and duodenum. RNase protection analysis showed that 11β-HSD1 was not expressed in intestinal epithelial cells, and enzyme activity studies detected no 11-reductase activity. 11β-HSD2 mRNA and protein were demonstrated in ileal and colonic epithelia; both MR and GR binding increased when enzyme activity was inhibited with carbenoxolone. Duodenal and jejunal epithelial cells showed very little 11β-HSD2 mRNA and undetectable 11β-HSD2 protein; despite minor (
- Published
- 1999
- Full Text
- View/download PDF
36. The type I and type II 11β-hydroxysteroid dehydrogenase enzymes
- Author
-
Hironobu Sasano, Robin E. Smith, K X Z Li, Zygmunt S. Krozowski, Tim J Cole, V.R. Obeyesekere, C Coulter, Karen E. Sheppard, K Koyama, and Alicia N Stein-Oakley
- Subjects
medicine.medical_specialty ,Protein Conformation ,Placenta ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Biology ,Kidney ,Biochemistry ,Isozyme ,chemistry.chemical_compound ,Receptors, Glucocorticoid ,Endocrinology ,Glucocorticoid receptor ,Mineralocorticoid receptor ,Corticosterone ,Neoplasms ,Internal medicine ,medicine ,Animals ,Humans ,Hydroxysteroid dehydrogenase ,Receptor ,Molecular Biology ,Cofactor binding ,Hydroxysteroid Dehydrogenases ,Brain ,Cell Biology ,Isoenzymes ,Arterioles ,chemistry ,Molecular Medicine ,11-beta-Hydroxysteroid Dehydrogenases ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,medicine.drug - Abstract
Local tissue concentrations of glucocorticoids are modulated by the enzyme 11 β -hydroxysteroid dehydrogenase which interconverts cortisol and the inactive glucocorticoid cortisone in man, and corticosterone and 11-dehydrocorticosterone in rodents. The type I isoform (11 β -HSD1) is a bidirectional enzyme but acts predominantly as a oxidoreductase to form the active glucocorticoids cortisol or corticosterone, while the type II enzyme (11 β -HSD2) acts unidirectionally producing inactive 11-keto metabolites. There are no known clinical conditions associated with 11 β -HSD1 deficiency, but gene deletion experiments in the mouse indicate that this enzyme is important both for the maintenance of normal serum glucocorticoid levels, and in the activation of key hepatic gluconeogenic enzymes. Other important sites of action include omental fat, the ovary, brain and vasculature. Congenital defects in the 11 β -HSD2 enzyme have been shown to account for the syndrome of apparent mineralocorticoid excess (AME), a low renin severe form of hypertension resulting from the overstimulation of the non-selective mineralocorticoid receptor by cortisol in the distal tubule of the kidney. Inactivation of the 11 β -HSD2 gene in mice results in a phenotype with similar features to AME. In addition, these mice show high neonatal mortality associated with marked colonic distention, and remarkable hypertrophy and hyperplasia of the distal tubule epithelia. 11 β -HSD2 also plays an important role in decreasing the exposure of the fetus to the high levels of maternal glucocorticoids. Recent work suggests a role for 11 β -HSD2 in non-mineralocorticoid target tissues where it would modulate glucocorticoid access to the glucocorticoid receptor, in invasive breast cancer and as a mechanism providing ligand for the putative 11-dehydrocorticosterone receptor. While previous homologies between members of the SCAD superfamily have been of the order of 20–30% phylogenetic analysis of a new branch of retinol dehydrogenases indicates identities of >60% and overlapping substrate specificities. The availability of crystal structures of family members has allowed the mapping of conserved 11 β -HSD domains A–D to a cleft in the protein structure (cofactor binding domain), two parallel β -sheets, and an α -helix (active site), respectively.
- Published
- 1999
- Full Text
- View/download PDF
37. Steroid specificity of the putative DHB receptor: evidence that the receptor is not 11βHSD
- Author
-
Karen Khoo, Karen E. Sheppard, Zygmunt S. Krozowski, and Kevin X. Z. Li
- Subjects
Male ,medicine.medical_specialty ,Colon ,Physiology ,Endocrinology, Diabetes and Metabolism ,CHO Cells ,Spironolactone ,Biology ,Transfection ,Substrate Specificity ,Rats, Sprague-Dawley ,Glucocorticoid receptor ,Mineralocorticoid receptor ,Cricetinae ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Intestinal Mucosa ,Binding site ,Receptor ,Cells, Cultured ,Cell Nucleus ,Chinese hamster ovary cell ,Hydroxysteroid Dehydrogenases ,Recombinant Proteins ,Rats ,Endocrinology ,Nuclear receptor ,Biochemistry ,Culture Media, Conditioned ,Nuclear receptor coactivator 3 ,Carbenoxolone ,11-beta-Hydroxysteroid Dehydrogenases ,Estrogen-related receptor gamma ,Corticosterone - Abstract
Recently, we identified a novel putative nuclear receptor in colonic crypt cells distinct from both mineralocorticoid receptor and glucocorticoid receptor, with high affinity for 11-dehydrocorticosterone (11-DHB) (33). In the present study, competitive nuclear binding assays demonstrated that this site has a unique steroid binding specificity that distinguishes it from other steroid receptors. Western blot analysis showed the presence of 11β-hydroxysteroid dehydrogenase-2 (11βHSD2) but not 11βHSD1in colonic crypt cells and showed that 11βHSD2was present in the nuclear pellet. Differences in steroid specificity between the putative DHB receptor and inhibition of 11βHSD activity indicate that binding is not to the enzyme. Furthermore, modified Chinese hamster ovary cells transfected with the 11βHSD2gene express nuclear 11βHSD2but not a nuclear DHB binding site. In conclusion, these data support the existence of a novel nuclear DHB receptor in rat colon that is distinct from the classic steroid receptors and from both 11βHSD1and 11βHSD2.
- Published
- 1998
- Full Text
- View/download PDF
38. Synergistic inhibition of ovarian cancer cell growth by combining selective PI3K/mTOR and RAS/ERK pathway inhibitors
- Author
-
David D.L. Bowtell, Pui Ng, Ross D. Hannan, Richard B. Pearson, Meaghan Wall, Ricky W. Johnstone, Donald P. Cameron, Kathryn M Kinross, Grant A. McArthur, Margarete Kleinschmidt, Katherine M. Hannan, Amelia Neilsen, Frances Barber, James G. Christensen, Karen E. Sheppard, Rodney J. Hicks, Wayne A. Phillips, Carleen Cullinane, Jung Hock Foo, Joanna C. Chan, and Jason Ellul
- Subjects
MAPK/ERK pathway ,Cancer Research ,Pyridones ,Immunoblotting ,MAP Kinase Kinase 2 ,MAP Kinase Kinase 1 ,Mice, Nude ,Phosphatidylinositol 3-Kinases ,Anti-apoptotic Ras signalling cascade ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Extracellular Signal-Regulated MAP Kinases ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Ovarian Neoplasms ,Mice, Inbred BALB C ,business.industry ,Kinase ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,TOR Serine-Threonine Kinases ,Diphenylamine ,Drug Synergism ,medicine.disease ,Xenograft Model Antitumor Assays ,Cell biology ,Gene Expression Regulation, Neoplastic ,Pyrimidines ,Oncology ,Drug Resistance, Neoplasm ,Benzamides ,Cancer research ,ras Proteins ,Female ,Signal transduction ,Ovarian cancer ,business ,Signal Transduction - Abstract
Ovarian cancer is the major cause of death from gynaecological malignancy with a 5year survival of only ∼30% due to resistance to platinum and paclitaxel-based first line therapy. Dysregulation of the phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) and RAS/extracellular signal-regulated kinase (ERK) pathways is common in ovarian cancer, providing potential new targets for 2nd line therapy.We determined the inhibition of proliferation of an extensive panel of ovarian cancer cell lines, encompassing all the major histotypes, by the dual PI3K/mTOR inhibitor PF-04691502 and a MEK inhibitor, PD-0325901. In addition, we analysed global gene expression, mutation status of key PI3K/mTOR and RAS/ERK pathway members and pathway activation to identify predictors of drug response.PF-04691502 inhibits proliferation of the majority of cell lines with potencies that correlate with the extent of pathway inhibition. Resistant cell lines were characterised by activation of the RAS/ERK pathway as indicated by differential gene expression profiles and pathway activity analysis. PD-0325901 suppressed growth of a subset of cell lines that were characterised by high basal RAS/ERK signalling. Strikingly, using PF-04691502 and PD-0325901 in combination resulted in synergistic growth inhibition in 5/6 of PF-04691502 resistant cell lines and two cell lines resistant to both single agents showed robust synergistic growth arrest. Xenograft studies confirm the utility of combination therapy to synergistically inhibit tumour growth of PF-04691502-resistant tumours in vivo.These studies identify dual targeted inhibitors of PI3K/mTOR in combination with inhibitors of RAS/ERK signalling as a potentially effective new approach to treating ovarian cancer.
- Published
- 2013
39. Glucocorticoid receptor expression is down-regulated by Lp(a) lipoprotein in vascular smooth muscle cells
- Author
-
D D Sviridov, A. Sato, John W. Funder, M. J. Fullerton, and Karen E. Sheppard
- Subjects
medicine.medical_specialty ,Time Factors ,Vascular smooth muscle ,Down-Regulation ,Estrogen receptor ,Aorta, Thoracic ,Lipoproteins, VLDL ,Muscle, Smooth, Vascular ,Rats, Sprague-Dawley ,Receptors, Glucocorticoid ,Endocrinology ,Glucocorticoid receptor ,Downregulation and upregulation ,Internal medicine ,medicine ,Animals ,Humans ,RNA, Messenger ,Mammary Arteries ,Receptor ,Glucocorticoids ,Cells, Cultured ,Aged ,Cell Nucleus ,biology ,Estrogen receptor binding ,Lipoprotein(a) ,Middle Aged ,musculoskeletal system ,Rats ,Lipoproteins, LDL ,Receptors, Estrogen ,cardiovascular system ,biology.protein ,Female ,Lipoproteins, HDL ,Cell Division ,Lipoprotein - Abstract
Glucocorticoids have been reported to protect against atherosclerosis and have been used clinically as protective therapy for restenosis after balloon angioplasty. Recently, Lp(a) lipoprotein [Lp(a)] levels have been suggested to be an independent risk factor for atherosclerosis, although its mechanisms of action are still uncertain. To clarify this atherogenic mechanism of Lp(a), we investigated the effects of Lp(a) on glucocorticoid receptor (GR) expression in human vascular smooth muscle cells (SMC). Levels of nuclear GR in SMC began to decrease after 12-h incubation with Lp(a), to 55 +/- 8% of the control value at 48 h; binding affinity did not change. Lp(a) had no effect on estrogen receptor binding in SMC. Moreover, low, very low, and high density lipoproteins had no effect on GR binding in SMC. The effects of Lp(a) on nuclear GR in rat SMC were very similar to those in human SMC; in contrast, Lp(a) did not alter GR or estrogen receptor levels in rat endothelial cells. GR messenger RNA levels in SMC decreased after 1-h treatment with Lp(a) to 23% of the control value after 12 h. Further, the antiproliferative effect of glucocorticoids on SMC was blunted by exposure to Lp(a). We conclude that Lp(a) down-regulates GR gene expression, resulting in a decreased number of GR in SMC. These findings suggest the possibility of a novel atherogenic mechanism of Lp(a) via inhibition of a protective action of glucocorticoids on SMC.
- Published
- 1995
- Full Text
- View/download PDF
40. Abstract 2826: Sustained melanoma regression is achieved with continuous palbociclib and PLX4720 treatment but not with intermittent or sequential dosing
- Author
-
Todd VanArsdale, Grant A. McArthur, Claire Martin, Kelly Waldeck, Richard A. Young, Carleen Cullinane, Laura Kirby, Nicole M. Haynes, and Karen E. Sheppard
- Subjects
MAPK/ERK pathway ,Senescence ,Cancer Research ,Programmed cell death ,biology ,business.industry ,Melanoma ,Cyclin D ,Palbociclib ,medicine.disease ,Oncology ,Immunology ,Cancer research ,medicine ,biology.protein ,Phosphorylation ,Dosing ,business - Abstract
In melanoma the development of resistance to BRAF inhibitors limits clinical responses. Thus the search for novel single agent and combination therapies as well as sequencing strategies that overcome or delay the emergence of resistance is needed. The p16-CDK4- cyclinD-RB1 pathway (CDK4 pathway) is deregulated in approximately 90% of melanomas and most melanoma cell lines are sensitive to palbociclib a specific CDK4/6 inhibitor. We hypothesized that dual targeting the MAPK/ERK and CDK4 pathways would lead to robust inhibition of the CDK4/Cyclin D complex and consequently induce greater tumor regression than single agent treatment. Proliferation and colony formation assays demonstrated that sequential and intermittent treatment with PLX4720 and palbociclib was not as effective as continuous combinational dosing. Only the combination of PLX4720 and palbociclib overcame the development of resistance leading to sustained inhibition of proliferation, cell death and induction of senescence. In A375 and HT144 human tumor xenograft models, palbociclib and PLX4720 initially induced tumor regression and tumor stasis, respectively, however resistance eventually developed to both agents. In contrast, the combination treatment induced rapid and sustained tumor regression. Biomarker studies indicate resistance to single agent PLX4720 was due to reactivation MAPK/ERK pathway and resistance to palbociclib to partial restoration of phosphorylated RB1. Tumor regression in the combination treatment was associated with the infiltration of leukocytes including activated natural killer cells indicating that these cells were involved in tumor clearance. These data demonstrate that dual targeting CDK4/6 and mutant BRAF evades resistance to single agents and leads to sustained tumor regression Citation Format: Karen E. Sheppard, Carleen Cullinane, Claire Martin, Laura Kirby, Nicole Haynes, Kelly Waldeck, Richard Young, Todd VanArsdale, Grant McArthur. Sustained melanoma regression is achieved with continuous palbociclib and PLX4720 treatment but not with intermittent or sequential dosing. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2826.
- Published
- 2016
- Full Text
- View/download PDF
41. Targeting PI3 kinase/AKT/mTOR signaling in cancer
- Author
-
Kathryn M. Kinross, Wayne A. Phillips, Benjamin Solomon, Karen E. Sheppard, and Richard B. Pearson
- Subjects
Cancer Research ,Angiogenesis ,Phosphatidylinositol 3-Kinases ,Pharmacology ,Biology ,Models, Biological ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Protein kinase B ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Feedback, Physiological ,Kinase ,TOR Serine-Threonine Kinases ,Cancer ,Receptor Cross-Talk ,medicine.disease ,Cancer research ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The phosphatidylinositol 3 kinase (PI3K) pathway is one of the major pathways modulating cell growth, proliferation, metabolism, survival, and angiogenesis. Hyperactivation of this pathway is one of the most frequent occurrences in human cancer and is thus an obvious target for treatment of this disease. Currently there are 26 novel compounds targeting the PI3K pathway being assessed in more than 150 cancer-related clinical trials. Although this pathway is involved in many vital biologic functions, data emanating from these clinical trials indicate that these drugs are well tolerated. This review outlines the interaction of the PI3K pathway with other signaling cascades, highlights mechanisms involved in hyperactivation, discusses current therapeutics in cancer-related clinical trials that target this pathway, and, based on preclinical data, discusses possible leads on patient selection and combinational therapy, including targeting multiple components of the associated signaling network.
- Published
- 2012
42. Calcium and protein kinase C regulation of the glucocorticoid receptor in mouse corticotrope tumor cells
- Author
-
Karen E. Sheppard
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,chemistry.chemical_element ,Cycloheximide ,Calcium ,Biology ,Biochemistry ,Dexamethasone ,Calcium in biology ,Mice ,chemistry.chemical_compound ,Receptors, Glucocorticoid ,Endocrinology ,Glucocorticoid receptor ,Internal medicine ,Tumor Cells, Cultured ,medicine ,Animals ,Pituitary Neoplasms ,RNA, Messenger ,Molecular Biology ,Calcimycin ,Protein Kinase C ,Protein kinase C ,Voltage-dependent calcium channel ,Cell Biology ,3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester ,Enzyme Activation ,Kinetics ,Cytosol ,chemistry ,Phorbol ,Tetradecanoylphorbol Acetate ,Molecular Medicine - Abstract
The effect of increasing intracellular free calcium and activating protein kinase C on glucocorticoid receptor (GR) expression was investigated in AtT-20 cells, a mouse corticotrope tumor cell line. Treatment of AtT-20 cells with the calcium ionophore A23187 induced a rapid time- and dose-dependent decrease in [ 3 H]dexamethasone ([ 3 H]DEX) binding when measured in intact cells. Binding fell to 16% of control following 3 h of treatment with 10 μM A23187. In contrast, A23187 did not acutely effect steady state levels of GR mRNA, although levels fell to 76±1% of control after 8–15 h of treatment. Scatchard analysis of A23187 treated cultures demonstrated a decrease in GR binding capacity but no change in affinity for [ 3 H]DEX. Acute inhibition of protein synthesis with cycloheximide had no effect on [ 3 H]DEX binding, suggesting that the calcium-dependent decrease was not simply due to inhibition of GR protein synthesis. In contrast to the A23187 induced decrease in [ 3 H]DEX binding in intact cells, when binding was measured in cytosol extract from A23187 treated cultures there was no decrease. These data suggest that the A23187 induced decrease in GR binding in whole cells is not due to a decrease in GR protein but reversible conversion of the receptor to a non-binding form. Inducing calcium influx only through L-type voltage-dependent calcium channels with BAY K8644 also decreased [ 3 H]DEX binding at AtT-20 cells, though the effect was less than that induced by A23187. Although activation of protein kinase C with phorbol ester transiently increases intracellular free calcium in AtT-20 cells, when cells were treated for 0.5 to 22 h with phorbol 12-myristate 13-acetate, there was no acute fall in [ 3 H]DEX binding, and only a small decrease following 16 h of treatment. These data demonstrate that sustained increases in intracellular calcium in corticotropes can induce a rapid and marked decrease in GR binding. The mechanism is post-translational and involves the reversible conversion of the receptor to a non-binding form. In addition, the cellular milieu is clearly important in conferring non-binding status on GR since once the cell is disrupted GR binding is restored.
- Published
- 1994
- Full Text
- View/download PDF
43. Glucocorticoid Receptor Function in Rat Pituitary Intermediate Lobe is Inhibited by an Endogenous Protein
- Author
-
James R. Roberts, Dominic J. Autelitano, Mariann Blum, and Karen E. Sheppard
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Pituitary gland ,Pro-Opiomelanocortin ,Endocrinology, Diabetes and Metabolism ,Melanotroph ,Dexamethasone ,Rats, Sprague-Dawley ,Cellular and Molecular Neuroscience ,Receptors, Glucocorticoid ,Endocrinology ,Glucocorticoid receptor ,Proopiomelanocortin ,Anterior pituitary ,Pituitary Gland, Anterior ,Internal medicine ,Gene expression ,medicine ,Animals ,Androstanols ,In Situ Hybridization ,Cell Nucleus ,biology ,Endocrine and Autonomic Systems ,Rats ,medicine.anatomical_structure ,Pituitary Gland ,biology.protein ,Autoradiography ,RNA ,Corticotropic cell ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,medicine.drug - Abstract
Though glucocorticoids inhibit proopiomelanocortin (POMC) gene expression and POMC-derived peptide release from corticotroph cells of the anterior pituitary, the regulation of this gene by glucocorticoids is less clear in the melanotroph cell of the pituitary intermediate lobe. To examine the difference between glucocorticoid sensitivity of the anterior lobe (AL) and intermediate lobe (IL) of the pituitary we have examined glucocorticoid receptor (GR) status of these two tissues and the role glucocorticoids play in regulating IL POMC gene expression. The rate of in vivo GR gene transcription, measured by nuclear run-on assay was consistently higher in the pituitary neurointermediate lobe (NIL) compared with the AL of the same animals. On a concentration basis, cytoplasmic GR mRNA in the NIL was similar to that found in the AL, and GR binding using [3H]dexamethasone (DEX) as ligand demonstrated similar concentrations of specific [3H]DEX binding in acutely isolated AL and NIL tissues. The specific Type II corticosteroid receptor ligand RU28362 displaced [3H]DEX binding to levels equivalent to non-specific binding, thus indicating that DEX was binding only to Type II corticosteroid receptors. To assess the direct action of glucocorticoids on POMC gene expression, NIL cells were cultured for 7 days and then treated with DEX. One hour DEX treatment of NIL primary cultures had no effect on levels of POMC heteronuclear RNA levels; in contrast, DEX induced a rapid and potent inhibition of POMC heteronuclear RNA levels in cells treated with the protein synthesis inhibitor puromycin.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1993
- Full Text
- View/download PDF
44. Differential Regulation of Type II Corticosteroid Receptor Messenger Ribonucleic Acid Expression in the Rat Anterior Pituitary and Hippocampus*
- Author
-
James L. Roberts, Karen E. Sheppard, and Mariann Blum
- Subjects
endocrine system ,medicine.medical_specialty ,Pituitary gland ,Pro-Opiomelanocortin ,Corticotropin-Releasing Hormone ,medicine.medical_treatment ,Biology ,Hippocampus ,Dexamethasone ,chemistry.chemical_compound ,Receptors, Glucocorticoid ,Endocrinology ,Anterior pituitary ,Pituitary Gland, Anterior ,Corticosterone ,Internal medicine ,polycyclic compounds ,medicine ,Animals ,RNA, Messenger ,Adrenalectomy ,Single-Strand Specific DNA and RNA Endonucleases ,Nucleic Acid Hybridization ,Rats, Inbred Strains ,Nuclease protection assay ,Rats ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Hypothalamus ,Solution hybridization ,Female ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,medicine.drug - Abstract
The present study was designed to characterize the regulation of the type II corticosteroid receptor (GR) mRNA in two tissues involved in the control of the hypothalamic-pituitary-adrenal axis. We have used a solution hybridization/S1 nuclease protection assay to quantitate GR mRNA levels in the rat hippocampus and anterior pituitary after CRF, dexamethasone (DEX), or corticosterone (CORT) treatment. In general, hippocampal GR mRNA levels increased after removal of endogenous corticosteroids by surgical adrenalectomy and decreased in response to glucocorticoid treatment. More specifically, in the hippocampus 1) GR mRNA expression was decreased when adrenalectomized (ADX) animals were replaced with a relatively low dose of CORT, but not with a low dose of DEX; 2) acutely, CRF was more effective than DEX in decreasing the levels of GR mRNA in intact animals; however, under the same paradigm in ADX animals, DEX decreased the level of GR mRNA, whereas CRF was ineffective; and 3) in contrast to the decrease in GR mRNA levels observed after acute and low doses of glucocorticoid treatment, chronic treatment with either DEX or CORT did not change the level of hippocampal GR mRNA. These results suggest that in the hippocampus the decrease in GR mRNA expression after CRF treatment is probably via the release of glucocorticoids, and that this tissue is more sensitive to endogenous glucocorticoids than DEX. Anterior pituitary GR mRNA was differentially regulated compared with that in the hippocampus. In marked contrast to Gr mRNA in the hippocampus, ADX did not alter anterior pituitary GR mRNA expression, and glucocorticoid treatment led to an increase in GR mRNA levels. In the anterior pituitary 1) glucocorticoid treatment led to an increase in GR mRNA expression, when replaced with a relatively low dose of DEX, but not when replaced with a low dose of CORT; 2) acutely, neither CRF nor DEX altered levels of GR mRNA in intact animals; however, under the same paradigm DEX increased levels in ADX animals; and 3) chronic DEX or CORT treatment of intact animals elevated levels of anterior pituitary GR mRNA. In summary, these data have demonstrated tissue-specific regulation of GR mRNA in the hippocampus and anterior pituitary, which is dependent on both the dose and length of treatment and, in addition, on the glucocorticoid itself.
- Published
- 1990
- Full Text
- View/download PDF
45. Abstract 2687: Receptor tyrosine kinases can mediate compensatory signaling and phenotype-switching associated with resistance to BRAF inhibitors
- Author
-
Petranel T. Ferrao, Zhentao Li, Rachel Ramsdale, Robert N. Jorissen, Karen E. Sheppard, Grant A. McArthur, and Amardeep S. Dhillon
- Subjects
MAPK/ERK pathway ,Cancer Research ,medicine.medical_specialty ,biology ,Melanoma ,Microphthalmia-associated transcription factor ,medicine.disease ,Phenotype ,Receptor tyrosine kinase ,Endocrinology ,Oncology ,Internal medicine ,SPRY2 ,medicine ,biology.protein ,Cancer research ,Protein kinase B ,Insulin-like growth factor 1 receptor - Abstract
BRAF/ERK is the most frequently activated oncogenic signaling pathway in metastatic melanoma. Although clinical studies have demonstrated initial efficacy of BRAF inhibitors (BRAFi), the therapeutic responses are short-lived in many patients due to the acquisition of resistance. Inhibition of mutant BRAF in melanoma has been reported to induce epithelial-mesenchymal transition (EMT)-like phenotype switching. One mechanism of phenotype-switching associated with resistance to BRAFi is down-regulation of MITF and increased levels of WNT5A. Across a panel of 22 BRAFV600E melanoma lines, those intrinsically resistant to BRAFi were found to express significantly higher levels of mesenchymal markers whereas the sensitive lines exhibited more epithelial-like characteristics. The intrinsically resistant lines featured high expression of multiple receptor tyrosine kinases (RTKs) including EGFR, but low levels of other RTKs such as IGFR. RTKs are known able to mediate melanoma phenotype-switching and can confer BRAFi resistance via reactivation of ERK signaling. Hence, we further assessed the link between specific RTKs and drug resistance, ERK signaling and phenotype-switching. In A375 and other drug sensitive cell lines, specific RTKs constitutively expressed and activated with ligands, differed in their ability to confer a proliferative advantage during drug treatment, which was associated with variation in signaling. Activated EGFR was able to drive compensatory ERK signaling whereas activated IGFR strongly activated AKT signaling. In the absence of RTK expression, BRAFi-treated A375 cells displayed an EMT-like phenotype switch. Drug adaptation induced resistance to BRAFi, a mesenchymal-like phenotype and increased migration in A375 cells, which were reversible by removal of the drug. During this period, we found reversible hyper-activation of ERK signaling due to relief of feedback regulation by BRAFi, and an altered composition of the AP-1 complex with down-regulated expression of FRA-1. Interestingly this was associated with increased MITF and SLUG in A375 cells. Expression and activation of EGFR in A375 cells treated with BRAFi maintained ERK signaling, expression of feedback regulator Sprouty2 (SPRY2) and FRA-1. Enforced expression of SPRY2 was able to reverse BRAFi resistance induced by EGFR signaling. EGFR activation also suppressed BRAFi induction of MITF, expression of EMT-inducers, including SLUG and ZEB2, with cells retaining the morphology similar to the drug-naive state. In contrast, activated IGFR did not alter the drug-induced effects in A375. Our study reveals that RTKs differ in their ability to confer proliferation signaling and the EMT-like phenotype-switching, both having strong implications in the development of resistance to BRAFi in melanoma. Citation Format: Zhentao Li, Rachel Ramsdale, Robert Jorissen, Karen Sheppard, Amardeep Dhillon, Grant McArthur, Petranel Ferrao. Receptor tyrosine kinases can mediate compensatory signaling and phenotype-switching associated with resistance to BRAF inhibitors. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2687. doi:10.1158/1538-7445.AM2015-2687
- Published
- 2015
- Full Text
- View/download PDF
46. Abstract 3089: CDKN2A and p53 status predicts response to CDK4/6 inhibition in melanoma
- Author
-
Laura Kirby, Karen E. Sheppard, Grant A. McArthur, Stephen Q. Wong, and Claire Martin
- Subjects
Cancer Research ,education.field_of_study ,Cell cycle checkpoint ,Melanoma ,Population ,Cancer ,Biology ,medicine.disease ,Cyclin D1 ,Oncology ,CDKN2A ,Immunology ,medicine ,Cancer research ,CDK4/6 Inhibition ,education ,neoplasms ,CDK inhibitor - Abstract
The p16-CyclinD1-CDK4-RB pathway regulates G1-S cell cycle transition. This pathway is deregulated in 90% of melanomas, through amplification or increased expression of CDK4 or Cyclin D1 (CCND1), or reduced expression of the CDK inhibitor p16INK4A (CDKN2A). To explore if CDK4 may be a viable target for the treatment of human melanoma we screened a panel of melanoma cell lines with the highly selective CDK4/6 inhibitor PD-0332991 (PD-991). A majority of cell lines were found to be sensitive to PD-991, although there was a distinct population of resistant cell lines. In sensitive melanoma cell lines, treatment with PD-991 resulted in loss of hyperphosphorylated RB and a concomitant G0/G1 cell cycle arrest. Treated cells displayed an enlarged flattened morphology and an increase in SA-β-galactosidase staining compared to vehicle controls, indicative of cellular senescence. RB-null cell lines were resistant to CDK4/6 inhibition, and did not display cell cycle arrest or senescence. Expression and mutational analysis was performed to identify additional genomic predictors of sensitivity and resistance. Loss of CDKN2A/p16 correlated with sensitivity to PD-991 (p Conclusions: These preliminary data identify RB, p16 and p53 as predictors of response to PD-991, and support further evaluation of CDK4/6 inhibitors in melanoma. Citation Format: Claire A. Martin, Laura Kirby, Stephen Wong, Grant A. McArthur, Karen E. Sheppard. CDKN2A and p53 status predicts response to CDK4/6 inhibition in melanoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3089. doi:10.1158/1538-7445.AM2015-3089
- Published
- 2015
- Full Text
- View/download PDF
47. Abstract B05: Targeting BRAF and CDK4 in BRAF mutant melanoma induces sustained tumor regression
- Author
-
Grant A. McArthur, Karen E. Sheppard, Cullinane Carleen, Richard J. Young, Kelly Waldeck, Sophia Randolph, Todd VanArsdale, Claire Martin, and Laura Kirby
- Subjects
MAPK/ERK pathway ,Cancer Research ,biology ,business.industry ,Melanoma ,medicine.medical_treatment ,Cyclin D ,Cancer ,Palbociclib ,medicine.disease ,Oncogene Addiction ,Targeted therapy ,Cyclin D1 ,Oncology ,Immunology ,medicine ,Cancer research ,biology.protein ,business ,neoplasms - Abstract
Background: The p16-cyclinD-CDK4-RB1 pathway (CDK4 pathway) is deregulated in 90% of melanomas. The main outcome of CDK4 activation is the phosphorylation and thus, inhibition of RB1 leading to G1–S cell-cycle transition. In addition, CDK4 directly phosphorylates other proteins that promote cell-cycle progression and inhibit both cell senescence and apoptosis. The MAPK/ERK pathway is deregulated in 90% of melanoma and enhances CDK4 pathway signaling through increasing cyclin D1 expression. We have previously demonstrated in a panel of melanoma cell lines that the majority of cell lines were sensitive to a CDK4/6 inhibitor (Palbociclib) and consistent with the concept of CDK4 pathway-dependent oncogene addiction, cell lines with loss of functional p16INK4A were significantly more sensitive to Palbociclib than cells with wild-type p16INK4A. We hypothesized that dual targeting the MAPK/ERK and CDK4 pathways would lead to robust inhibition of the CDK4/Cyclin D complex and consequently induce greater tumor regression than single agent treatment. Results: In vitro, PLX4720, Palbociclib or their combination inhibited BRAF mutant melanoma cell line proliferation and clonogenic survival, however, only with the combination treatment group was a rapid sustained inhibition achieved. Enlarged flattened cells and a significant increase in SA-β-galactosidase staining indicated that the dual treatment resulted in robust cellular senescence. In a BRAFV600E mutant A375 xenograft model, Palbociclib and PLX4720 initially induced tumor regression and tumor stasis respectively, but resistance eventually developed. In contrast, in the combination treatment, tumors regressed and sustained inhibition of tumor growth was maintained. Preliminary biomarker studies indicate resistance to PLX4720 was due to reactivation MAPK/ERK pathway and resistance to Palbociclib to partial restoration of phosphorylated RB1. Conclusions: Combination therapy of MAPK/ERK pathway inhibitors with CDK4 inhibitors is likely to be an effective treatment for BRAF mutant melanoma and in addition delay resistance to single agent targeted therapy. Citation Format: Karen E. Sheppard, Cullinane Carleen, Laura Kirby, Kelly Waldeck, Richard Young, Claire Martin, Todd VanArsdale, Sophia Randolph, Grant A. McArthur. Targeting BRAF and CDK4 in BRAF mutant melanoma induces sustained tumor regression. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Melanoma: From Biology to Therapy; Sep 20-23, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(14 Suppl):Abstract nr B05.
- Published
- 2015
- Full Text
- View/download PDF
48. Corticosteroid Receptors, 11β-Hydroxysteroid Dehydrogenase, and the Heart
- Author
-
Karen E. Sheppard
- Subjects
medicine.medical_specialty ,medicine.drug_class ,Adrenal cortex ,Biology ,Endocrinology ,Glucocorticoid receptor ,medicine.anatomical_structure ,Mineralocorticoid ,Internal medicine ,medicine ,Signal transduction ,Receptor ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,Intracellular ,medicine.drug ,Hormone - Abstract
Mineralocorticoid and glucocorticoid hormones are known as corticosteroid hormones and are synthesized mainly in the adrenal cortex; however, more recently the enzymes involved in their synthesis have been found in a variety of cells and tissues, including the heart. The effects of these hormones are mediated via both cytoplasmic mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs), which act as ligand-inducible transcription factors. In addition, rapid, nongenomically mediated effects of these steroids can occur that may be via novel corticosteroid receptors. The lipophilic nature of these hormones allows them to pass freely through the cell membrane, although the intracellular concentration of mineralocorticoids and glucocorticoids is dependent on several cellular factors. The main regulators of intracellular glucocorticoid levels are 11 beta-hydroxysteroid dehydrogenase (11 beta HSD) isoforms. 11 beta HSD1 acts predominantly as a reductase in vivo, facilitating glucocorticoid action by converting circulating receptor-inactive 11-ketoglucocorticoids to active glucocorticoids. In contrast, 11 beta HSD 2 acts exclusively as an 11 beta-dehydrogenase and decreases intracellular glucocorticoids by converting them to their receptor-inactive 11-ketometabolites. Furthermore, P-glycoproteins, by actively pumping steroids out of cells, can selectively decrease steroids and local steroid synthesis can increase steroid concentrations. Receptor concentration, receptor modification, and receptor-protein interactions can also significantly impact on the corticosteroid response. This review details the receptors and possible mechanisms involved in both mediating and modulating corticosteroid responses. In addition, direct effects of corticosteroids on the heart are described including a discussion of the corticosteroid receptors and the mechanisms involved in mediating their effects.
- Published
- 2003
- Full Text
- View/download PDF
49. Abstract 2718: Targeting ribosome biogenesis with CX5461 as a potential treatment for melanoma and ovarian cancer
- Author
-
Ross D. Hannan, Grant A. McArthur, Karen E. Sheppard, Richard B. Pearson, Jason Ellul, Katherine M. Hannan, Sean O'Brien, Denis Drygin, Natalie Brajanovski, and Jessica Ahearn
- Subjects
Cancer Research ,education.field_of_study ,Cell growth ,Melanoma ,Population ,Cell ,Ribosome biogenesis ,Biology ,medicine.disease ,Molecular biology ,medicine.anatomical_structure ,Oncology ,Cancer cell ,Cancer research ,medicine ,Ovarian cancer ,education ,PI3K/AKT/mTOR pathway - Abstract
Ribosome biogenesis provides the molecular machinery required for protein synthesis, thus it dictates the ability of a cell to grow and proliferate. We have recently demonstrated that inhibiting ribosome biogenesis with a small molecule that inhibits RNA Polymerase (POL) I driven transcription (CX5461) selectively killed B-lymphoma cells in vivo while maintaining a viable wild-type B cell population (Bywater et al., Cancer Cell 22: p51, 2012). The therapeutic effect was mediated via nucleolar disruption and activation of p53-dependent apoptotic signalling. Although this response was dependent on p53, cell lines from solid tumors with mutant p53 also respond to CX5461 (Drygin et al., Cancer Res 71: p1418, 2011). Dysregulation of both the PI3K and RAS/ERK pathways are common in ovarian cancer and BRAF is mutated and activated in over 50% of melanomas; a cancer that also commonly expresses wild type-p53. Given that both these pathways are critical regulators of ribosome biogenesis, we hypothesized that targeting ribosome biogenesis may be a new and potent approach to treating these cancers. To determine the efficacy of CX5461 a panel of ovarian cancer and melanoma cell lines were treated with CX5461 and cell proliferation assessed. The majority of cell lines were sensitive to CX5461 and differential gene expression between resistant and sensitive ovarian cancer cell lines revealed sensitivity was associated with signatures of increased MYC signalling and BRCA1 and 2 mutation. Gene expression and assessment of rDNA transcription suggested that resistance in both ovarian cancer and melanoma cell lines was associated with an inability of CX5461 to inhibit POL1 driven transcription. These studies indicate that targeting ribosome biogenesis and function may provide a new therapeutic option for treating ovarian cancer and melanoma. Citation Format: Karen E. Sheppard, Natalie Brajanovski, Katherine M. Hannan, Jessica Ahearn, Jason Ellul, Denis Drygin, Sean O'Brien, Grant McArthur, Ross D. Hannan, Richard B. Pearson. Targeting ribosome biogenesis with CX5461 as a potential treatment for melanoma and ovarian cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2718. doi:10.1158/1538-7445.AM2014-2718
- Published
- 2014
- Full Text
- View/download PDF
50. 11Beta-hydroxysteroid dehydrogenase 1 transforms 11-dehydrocorticosterone into transcriptionally active glucocorticoid in neonatal rat heart
- Author
-
Karen E. Sheppard and Dominic J. Autelitano
- Subjects
Transcriptional Activation ,medicine.medical_specialty ,Biology ,Protein Serine-Threonine Kinases ,Immediate-Early Proteins ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Endocrinology ,Receptors, Glucocorticoid ,Corticosterone ,Internal medicine ,Gene expression ,medicine ,Myocyte ,Animals ,RNA, Messenger ,Receptor ,Cells, Cultured ,Aldosterone ,Kinase ,Myocardium ,Hydroxysteroid Dehydrogenases ,Nuclear Proteins ,Fibroblasts ,Rats ,Receptors, Mineralocorticoid ,chemistry ,Animals, Newborn ,11-beta-Hydroxysteroid Dehydrogenases ,Glucocorticoid ,Homeostasis ,medicine.drug - Abstract
The ability of cells to directly respond to glucocorticoids and aldosterone is a function of GR and MR expression, and coexpression of 11β-hydroxysteroid dehydrogenases (11βHSDs), which convert glucocorticoids and their 11-ketometabolites into either receptor inactive or active derivatives. The aim of the present study was to determine the cellular expression of GR, MR, 11βHSD1, and 11βHSD2 in neonatal rat heart and determine the role these enzymes play in modulating glucocorticoid and aldosterone action. Ribonuclease protection analysis and steroid binding assays showed that GR is expressed in both cardiac myocytes and fibroblasts, whereas MR is expressed only in myocytes. 11βHSD2 was not detected in cardiac cells, but 11βHSD1 was expressed at high levels in both cardiac myocytes and fibroblasts. Enzyme activity studies demonstrated that 11βHSD1 acted as a reductase only, converting biologically inactive 11-dehydrocorticosterone to corticosterone, which then stimulated serum and glucocorticoid-induced kinase gene transcription via GR. In both cardiac myocytes and fibroblasts, aldosterone stimulated serum and glucocorticoid-induced kinase gene expression exclusively via GR, but not MR, indicating that aldosterone can have glucocorticoid-like actions in heart. The ability of cardiac cells to use both circulating corticosterone and 11-dehydrocorticosterone as a source of glucocorticoid suggests that the heart is under tonic glucocorticoid control, implying that glucocorticoids play important homeostatic roles in the heart.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.