Craig T. Wallington-Beddoe
University of Sydney
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Craig T. Wallington-Beddoe.
Autophagy | 2011
Craig T. Wallington-Beddoe; John Hewson; Kenneth F. Bradstock; Linda J. Bendall
Acute lymphoblastic leukemia (ALL), the most common form of childhood cancer, usually responds to chemotherapy but patients who develop disease relapse have a poor prognosis. New agents to treat ALL are urgently required. FTY720 is an immunosuppressive drug that has promising in vitro activity in a number of malignancies, with the proposed mechanism being the reactivation of the protein serine/threonine phosphatase, PP2A. FTY720 reduced the proliferation and viability of Ph+ and Ph- ALL cell lines and patient samples with IC50 values for viability between 5.3 to 7.9 μM. Cell death was caspase-independent with negligible activation of caspase-3 and no inhibition of FTY720-induced cell death by the caspase inhibitor Z-VAD-FMK. The cytotoxic effects of FTY720 were independent of PP2A reactivation as determined by the lack of effect of the PP2A inhibitor okadaic acid. Features of autophagy, including autophagosomes, LC3II expression and increased autophagic flux, were induced by FTY720. However the phosphorylated form of FTY720 (FTY720-P) similarly induced autophagy but not cell death. This suggests that autophagy is prosurvival in this setting, a finding supported by protection from cell death induced by the cytotoxic agent vincristine. FTY720 also induced reactive oxygen species (ROS) and the antioxidant N-acetyl-cysteine (NAC) partially reversed the cytotoxic effects, demonstrating a role for ROS generation in the cell death mechanism. FTY720 is an active drug in vitro in ALL cell lines and patient samples. Evidence supports a caspase-independent mechanism of cell death with the occurrence of autophagy and necrosis.
Cancer Research | 2014
Craig T. Wallington-Beddoe; Jason A. Powell; Daochen Tong; Stuart M. Pitson; Kenneth F. Bradstock; Linda J. Bendall
Sphingosine kinase 2 (SK2) may have utility as a prognostic marker in inflammatory diseases such as cancer in which it has been rationalized as a candidate therapeutic target. Here, we show that SK2 has an oncogenic role in acute lymphoblastic leukemia (ALL) by influencing expression of MYC. Genetic ablation of SK2 impaired leukemia development in a mouse model of ALL and pharmacologic inhibition extended survival in mouse xenograft models of human disease. SK2 attenuation in both the settings reduced MYC expression in leukemic cells, with reduced levels of acetylated histone H3 within the MYC gene associated with reduced levels of MYC protein and expression of MYC-regulated genes. Our results demonstrated that SK2 regulates MYC, which has a pivotal role in hematologic malignancies, providing a preclinical proof of concept for this pathway as a broad-based therapeutic target in this setting.
PLOS ONE | 2012
Craig T. Wallington-Beddoe; Anthony S. Don; John Hewson; Qiao Qiao; Rachael A. Papa; Richard B. Lock; Kenneth F. Bradstock; Linda J. Bendall
Most patients with acute lymphoblastic leukemia (ALL) respond well to standard chemotherapy-based treatments. However a significant proportion of patients, particularly adult patients, relapse with the majority dying of leukemia. FTY720 is an immunosuppressive drug that was recently approved for the treatment of multiple sclerosis and is currently under pre-clinical investigation as a therapy for a number of hematological malignancies. Using human ALL xenografts in NOD/SCIDγc−/− mice, we show for the first time that three Ph+ human ALL xenografts responded to FTY720 with an 80±12% (p = 0.048) reduction in overall disease when treatment was commenced early. In contrast, treatment of mice with FTY720 did not result in reduced leukemia compared to controls using four separate human Ph− ALL xenografts. Although FTY720 reactivated PP2A in vitro, this reactivation was not required for death of Ph− ALL cells. The plasma levels of FTY720 achieved in the mice were in the high nanomolar range. However, the response seen in the Ph+ ALL xenografts when treatment was initiated early implies that in vivo efficacy may be obtained with substantially lower drug concentrations than those required in vitro. Our data suggest that while FTY720 may have potential as a treatment for Ph+ ALL it will not be a useful agent for the treatment of Ph− B-ALL.
Blood | 2017
Jason A. Powell; Alexander C. Lewis; Wenying Zhu; John Toubia; Melissa R. Pitman; Craig T. Wallington-Beddoe; Paul A.B. Moretti; Diana Iarossi; Saumya E. Samaraweera; Nik Cummings; Hayley S. Ramshaw; Daniel Thomas; Andrew Wei; Angel F. Lopez; Richard J. D'Andrea; Ian D. Lewis; Stuart M. Pitson
Acute myeloid leukemia (AML) is an aggressive malignancy where despite improvements in conventional chemotherapy and bone marrow transplantation, overall survival remains poor. Sphingosine kinase 1 (SPHK1) generates the bioactive lipid sphingosine 1-phosphate (S1P) and has established roles in tumor initiation, progression, and chemotherapy resistance in a wide range of cancers. The role and targeting of SPHK1 in primary AML, however, has not been previously investigated. Here we show that SPHK1 is overexpressed and constitutively activated in primary AML patient blasts but not in normal mononuclear cells. Subsequent targeting of SPHK1 induced caspase-dependent cell death in AML cell lines, primary AML patient blasts, and isolated AML patient leukemic progenitor/stem cells, with negligible effects on normal bone marrow CD34+ progenitors from healthy donors. Furthermore, administration of SPHK1 inhibitors to orthotopic AML patient-derived xenografts reduced tumor burden and prolonged overall survival without affecting murine hematopoiesis. SPHK1 inhibition was associated with reduced survival signaling from S1P receptor 2, resulting in selective downregulation of the prosurvival protein MCL1. Subsequent analysis showed that the combination of BH3 mimetics with either SPHK1 inhibition or S1P receptor 2 antagonism triggered synergistic AML cell death. These results support the notion that SPHK1 is a bona fide therapeutic target for the treatment of AML.
Biology of Blood and Marrow Transplantation | 2009
Craig T. Wallington-Beddoe; David Gottlieb; Fran Garvin; Vicki Antonenas; Mary Sartor
To predict platelet engraftment more accurately post autologous stem cell transplantation (SCT), we retrospectively analyzed the CD34(+)CD110(+) (CD110 or c-mpl, thrombopoietin receptor) content in the grafts of 70 patients undergoing transplantation for multiple myeloma (MM) with an in-house flow cytometric assay. We found that infusing at least 3.0 x 10(4) CD34(+)CD110(+) cells/kg clearly separated the cohort into those who achieved platelet engraftment before or after 21 days. This early megakaryocyte cell marker correlated more closely with early versus delayed platelet engraftment than CD34(+) measurements. Of the 70 patients, 4 required > or = 21 days to achieve platelet transfusion independence. Three of the 4 received a CD34(+)CD110(+) cell dose of <3.0 x 10(4) cells/kg, whereas 66 of 70 patients who received >3.0 x 10(4) CD34(+)CD110(+) cells/kg achieved platelet transfusion independence in <21 days (P < .001). Infusing >3.0 x 10(4) CD34(+)CD110(+) cells/kg was sensitive (100%) and specific (75%) for achieving platelet engraftment within 21 days. Patients with delayed platelet engraftment received a median of 2.28 x 10(4) CD34(+)CD110(+) cells/kg versus 12.1 x 10(4) cells/kg in those without this complication (P = .033). No effect was seen with neutrophil engraftment. Patients with early engraftment required a median of 1 platelet transfusion post transplant versus 2.5 in those with late engraftment (P = .009). A subthreshold absolute CD34(+)CD110(+) cell dose in the graft is a reliable predictor of delayed platelet engraftment, and could be used to guide the timing and number of peripheral blood stem cell (PBSC) collections for patients with MM undergoing an SCT.
British Journal of Haematology | 2013
Craig T. Wallington-Beddoe; Kenneth F. Bradstock; Linda J. Bendall
The sphingosine kinases (SphKs) have relatively recently been implicated in contributing to malignant cellular processes with particular interest in the oncogenic properties of SPHK1. Whilst SPHK1 has received considerable attention as a putative oncoprotein, SPHK2 has been much more difficult to study, with often conflicting data surrounding its role in cancer. Initial studies focused on non‐haemopoietic malignancies, however a growing body of literature on the role of sphingolipid metabolism in haemopoietic malignancies is now emerging. This review provides an overview of the current state of knowledge of the SphKs and the bioactive lipid sphingosine 1‐phosphate (S1P), the product of the reaction they catalyse. It then reviews the current literature regarding the roles of S1P and the SphKs in haemopoietic malignancies and discusses the compounds currently available that modulate sphingolipid metabolism and their potential and shortcomings as therapeutic agents for the treatment of haematological malignancies.
Oncotarget | 2017
Craig T. Wallington-Beddoe; Melissa K. Bennett; Kate Vandyke; Lorena Davies; Julia R. Zebol; Paul A.B. Moretti; Melissa R. Pitman; Duncan R. Hewett; Andrew C.W. Zannettino; Stuart M. Pitson
The proteasome inhibitor bortezomib has proven to be invaluable in the treatment of myeloma. By exploiting the inherent high immunoglobulin protein production of malignant plasma cells, bortezomib induces endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), resulting in myeloma cell death. In most cases, however, the disease remains incurable highlighting the need for new therapeutic targets. Sphingosine kinase 2 (SK2) has been proposed as one such therapeutic target for myeloma. Our observations that bortezomib and SK2 inhibitors independently elicited induction of ER stress and the UPR prompted us to examine potential synergy between these agents in myeloma. Targeting SK2 synergistically contributed to ER stress and UPR activation induced by bortezomib, as evidenced by activation of the IRE1 pathway and stress kinases JNK and p38MAPK, thereby resulting in potent synergistic myeloma apoptosis in vitro. The combination of bortezomib and SK2 inhibition also exhibited strong in vivo synergy and favourable effects on bone disease. Therefore, our studies suggest that perturbations of sphingolipid signalling can synergistically enhance the effects seen with proteasome inhibition, highlighting the potential for the combination of these two modes of increasing ER stress to be formally evaluated in clinical trials for the treatment of myeloma patients.
British Journal of Haematology | 2018
Craig T. Wallington-Beddoe; Magdalena Sobieraj-Teague; Bryone J. Kuss; Stuart M. Pitson
The number of novel therapies for the treatment of myeloma is rapidly increasing, as are the clinical trials evaluating them in combination with other novel and established therapies. Proteasome inhibitors, immunomodulatory agents and monoclonal antibodies are the most well known and studied classes of novel agents targeting myeloma, with histone deacetylase inhibitors, nuclear export inhibitors and several other approaches also being actively investigated. However, in parallel with the development and clinical use of these novel myeloma therapies is the emergence of novel mechanisms of resistance, many of which remain elusive, particularly for more recently developed agents. Whilst resistance mechanisms have been best studied for proteasome inhibitors, particularly bortezomib, class effects do not universally apply to all class members, and within‐class differences in efficacy, toxicity and resistance mechanisms have been observed. Although immunomodulatory agents share the common cellular target cereblon and thus resistance patterns relate to cereblon expression, the unique cell surface antigens to which monoclonal antibodies are directed means these agents frequently exhibit unique within‐class differences in clinical efficacy and resistance patterns. This review describes the major classes of novel therapies for myeloma, highlights the major clinical trials within each class and discusses known resistance mechanisms.
British Journal of Haematology | 2018
Craig T. Wallington-Beddoe; Vicki Xie; Daochen Tong; Jason A. Powell; Alexander C. Lewis; Lorena Davies; Stuart M. Pitson; Kenneth F. Bradstock; Linda J. Bendall
Craig T. Wallington‐Beddoe, Vicki Xie, Daochen Tong, Jason A. Powell, Alexander C. Lewis, Lorena Davies, Stuart M. Pitson, Kenneth F. Bradstock Linda J. Bendall
Cell death discovery | 2018
Alexander C. Lewis; Craig T. Wallington-Beddoe; Jason A. Powell; Stuart M. Pitson
AbstractConventional chemotherapy-based drug combinations have, until recently, been the backbone of most therapeutic strategies for cancer. In a time of emerging rationale drug development, targeted therapies are beginning to be added to traditional chemotherapeutics to synergistically enhance clinical responses. Of note, the importance of pro-apoptotic ceramide in mediating the anti-cancer effects of these therapies is becoming more apparent. Furthermore, reduced cellular ceramide in favour of pro-survival sphingolipids correlates with tumorigenesis and most importantly, drug resistance. Thus, agents that manipulate sphingolipid metabolism have been explored as potential anti-cancer agents and have recently demonstrated exciting potential to augment the efficacy of anti-cancer therapeutics. This review examines the biology underpinning these observations and the potential use of sphingolipid manipulating agents in the context of existing and emerging therapies for haematological malignancies.Facts• Efficacy of many chemotherapeutics and targeted therapies is dictated by cellular ceramide levels.• Oncogene activation skews sphingolipid metabolism to favour the production of pro-survival sphingolipids.• Inhibitors of enzymes involved in ceramide metabolism exhibit promise in the relapsed-refractory setting.• Anti-cancer activity of sphingosine kinase inhibitors provides several options for new drug combinations. Open Questions • What other clinically utilised drugs rely on increases in ceramide levels for their efficacy and can they be effectively partnered with other ceramide inducing agents?• How does ceramide modulate the Bcl-2 family proteins, Mcl-1 and Bcl-2?• Are sphingolipid enzyme inhibitors best suited in the frontline or relapsed-refractory setting?