170 results on '"Mayoh C"'
Search Results
2. A novel MYB::PAIP1 oncogenic fusion in pediatric blastic plasmacytoid dendritic cell neoplasm (BPDCN) is dependent on BCL2 expression and is sensitive to venetoclax
- Author
-
Kosasih, HJ, Healey, G, Brennan, MS, Bjelosevic, S, Sadras, T, Jalud, FB, Ibnat, T, Ng, AP, Mayoh, C, Mao, J, Tax, G, Ludlow, LEA, Johnstone, RW, Herold, MJ, Khaw, SL, de Bock, CE, Ekert, PG, Kosasih, HJ, Healey, G, Brennan, MS, Bjelosevic, S, Sadras, T, Jalud, FB, Ibnat, T, Ng, AP, Mayoh, C, Mao, J, Tax, G, Ludlow, LEA, Johnstone, RW, Herold, MJ, Khaw, SL, de Bock, CE, and Ekert, PG
- Published
- 2024
3. The transcriptional co-repressor Runx1t1 is essential for MYCN-driven neuroblastoma tumorigenesis.
- Author
-
Murray, JE, Valli, E, Milazzo, G, Mayoh, C, Gifford, AJ, Fletcher, JI, Xue, C, Jayatilleke, N, Salehzadeh, F, Gamble, LD, Rouaen, JRC, Carter, DR, Forgham, H, Sekyere, EO, Keating, J, Eden, G, Allan, S, Alfred, S, Kusuma, FK, Clark, A, Webber, H, Russell, AJ, de Weck, A, Kile, BT, Santulli, M, De Rosa, P, Fleuren, EDG, Gao, W, Wilkinson-White, L, Low, JKK, Mackay, JP, Marshall, GM, Hilton, DJ, Giorgi, FM, Koster, J, Perini, G, Haber, M, Norris, MD, Murray, JE, Valli, E, Milazzo, G, Mayoh, C, Gifford, AJ, Fletcher, JI, Xue, C, Jayatilleke, N, Salehzadeh, F, Gamble, LD, Rouaen, JRC, Carter, DR, Forgham, H, Sekyere, EO, Keating, J, Eden, G, Allan, S, Alfred, S, Kusuma, FK, Clark, A, Webber, H, Russell, AJ, de Weck, A, Kile, BT, Santulli, M, De Rosa, P, Fleuren, EDG, Gao, W, Wilkinson-White, L, Low, JKK, Mackay, JP, Marshall, GM, Hilton, DJ, Giorgi, FM, Koster, J, Perini, G, Haber, M, and Norris, MD
- Abstract
MYCN oncogene amplification is frequently observed in aggressive childhood neuroblastoma. Using an unbiased large-scale mutagenesis screen in neuroblastoma-prone transgenic mice, we identify a single germline point mutation in the transcriptional corepressor Runx1t1, which abolishes MYCN-driven tumorigenesis. This loss-of-function mutation disrupts a highly conserved zinc finger domain within Runx1t1. Deletion of one Runx1t1 allele in an independent Runx1t1 knockout mouse model is also sufficient to prevent MYCN-driven neuroblastoma development, and reverse ganglia hyperplasia, a known pre-requisite for tumorigenesis. Silencing RUNX1T1 in human neuroblastoma cells decreases colony formation in vitro, and inhibits tumor growth in vivo. Moreover, RUNX1T1 knockdown inhibits the viability of PAX3-FOXO1 fusion-driven rhabdomyosarcoma and MYC-driven small cell lung cancer cells. Despite the role of Runx1t1 in MYCN-driven tumorigenesis neither gene directly regulates the other. We show RUNX1T1 forms part of a transcriptional LSD1-CoREST3-HDAC repressive complex recruited by HAND2 to enhancer regions to regulate chromatin accessibility and cell-fate pathway genes.
- Published
- 2024
4. MYCN and SNRPD3 cooperate to maintain a balance of alternative splicing events that drives neuroblastoma progression.
- Author
-
Salib, A, Jayatilleke, N, Seneviratne, JA, Mayoh, C, De Preter, K, Speleman, F, Cheung, BB, Carter, DR, Marshall, GM, Salib, A, Jayatilleke, N, Seneviratne, JA, Mayoh, C, De Preter, K, Speleman, F, Cheung, BB, Carter, DR, and Marshall, GM
- Abstract
Many of the pro-tumorigenic functions of the oncogene MYCN are attributed to its regulation of global gene expression programs. Alternative splicing is another important regulator of gene expression and has been implicated in neuroblastoma development, however, the molecular mechanisms remain unknown. We found that MYCN up-regulated the expression of the core spliceosomal protein, SNRPD3, in models of neuroblastoma initiation and progression. High mRNA expression of SNRPD3 in human neuroblastoma tissues was a strong, independent prognostic factor for poor patient outcome. Repression of SNRPD3 expression correlated with loss of colony formation in vitro and reduced tumorigenicity in vivo. The effect of SNRPD3 on cell viability was in part dependent on MYCN as an oncogenic co-factor. RNA-sequencing revealed a global increase in the number of genes being differentially spliced when MYCN was overexpressed. Surprisingly, depletion of SNRPD3 in the presence of overexpressed MYCN further increased differential splicing, particularly of cell cycle regulators, such as BIRC5 and CDK10. MYCN directly bound SNRPD3, and the protein arginine methyltransferase, PRMT5, consequently increasing SNRPD3 methylation. Indeed, the PRMT5 inhibitor, JNJ-64619178, reduced cell viability and SNRPD3 methylation in neuroblastoma cells with high SNRPD3 and MYCN expression. Our findings demonstrate a functional relationship between MYCN and SNRPD3, which maintains the fidelity of MYCN-driven alternative splicing in the narrow range required for neuroblastoma cell growth. SNRPD3 methylation and its protein-protein interface with MYCN represent novel therapeutic targets. Hypothetical model for SNRPD3 as a co-factor for MYCN oncogenesis. SNRPD3 and MYCN participate in a regulatory loop to balance splicing fidelity in neuroblastoma cells. First MYCN transactivates SNRPD3 to lead to high-level expression. Second, SNRPD3 and MYCN form a protein complex involving PRMT5. Third, this leads to balanced alt
- Published
- 2024
5. Inhibitors of the Oncogenic PA2G4-MYCN Protein-Protein Interface
- Author
-
Massudi, H, Luo, J-S, Holien, JK, Gadde, S, Krishan, S, Herath, M, Koach, J, Stevenson, BW, Gorman, MA, Venkat, P, Mayoh, C, Luo, X-Q, Parker, MW, Cheung, BB, Marshall, GM, Massudi, H, Luo, J-S, Holien, JK, Gadde, S, Krishan, S, Herath, M, Koach, J, Stevenson, BW, Gorman, MA, Venkat, P, Mayoh, C, Luo, X-Q, Parker, MW, Cheung, BB, and Marshall, GM
- Abstract
MYCN is a major oncogenic driver for neuroblastoma tumorigenesis, yet there are no direct MYCN inhibitors. We have previously identified PA2G4 as a direct protein-binding partner of MYCN and drive neuroblastoma tumorigenesis. A small molecule known to bind PA2G4, WS6, significantly decreased tumorigenicity in TH-MYCN neuroblastoma mice, along with the inhibition of PA2G4 and MYCN interactions. Here, we identified a number of novel WS6 analogues, with 80% structural similarity, and used surface plasmon resonance assays to determine their binding affinity. Analogues #5333 and #5338 showed direct binding towards human recombinant PA2G4. Importantly, #5333 and #5338 demonstrated a 70-fold lower toxicity for normal human myofibroblasts compared to WS6. Structure-activity relationship analysis showed that a 2,3 dimethylphenol was the most suitable substituent at the R1 position. Replacing the trifluoromethyl group on the phenyl ring at the R2 position, with a bromine or hydrogen atom, increased the difference between efficacy against neuroblastoma cells and normal myofibroblast toxicity. The WS6 analogues inhibited neuroblastoma cell phenotype in vitro, in part through effects on apoptosis, while their anti-cancer effects required both PA2G4 and MYCN expression. Collectively, chemical inhibition of PA2G4-MYCN binding by WS6 analogues represents a first-in-class drug discovery which may have implications for other MYCN-driven cancers.
- Published
- 2023
6. Inhibition of mitochondrial translocase SLC25A5 and histone deacetylation is an effective combination therapy in neuroblastoma.
- Author
-
Seneviratne, JA, Carter, DR, Mittra, R, Gifford, A, Kim, PY, Luo, J-S, Mayoh, C, Salib, A, Rahmanto, AS, Murray, J, Cheng, NC, Nagy, Z, Wang, Q, Kleynhans, A, Tan, O, Sutton, SK, Xue, C, Chung, SA, Zhang, Y, Sun, C, Zhang, L, Haber, M, Norris, MD, Fletcher, JI, Liu, T, Dilda, PJ, Hogg, PJ, Cheung, BB, Marshall, GM, Seneviratne, JA, Carter, DR, Mittra, R, Gifford, A, Kim, PY, Luo, J-S, Mayoh, C, Salib, A, Rahmanto, AS, Murray, J, Cheng, NC, Nagy, Z, Wang, Q, Kleynhans, A, Tan, O, Sutton, SK, Xue, C, Chung, SA, Zhang, Y, Sun, C, Zhang, L, Haber, M, Norris, MD, Fletcher, JI, Liu, T, Dilda, PJ, Hogg, PJ, Cheung, BB, and Marshall, GM
- Abstract
The mitochondrion is a gatekeeper of apoptotic processes, and mediates drug resistance to several chemotherapy agents used to treat cancer. Neuroblastoma is a common solid cancer in young children with poor clinical outcomes following conventional chemotherapy. We sought druggable mitochondrial protein targets in neuroblastoma cells. Among mitochondria-associated gene targets, we found that high expression of the mitochondrial adenine nucleotide translocase 2 (SLC25A5/ANT2), was a strong predictor of poor neuroblastoma patient prognosis and contributed to a more malignant phenotype in pre-clinical models. Inhibiting this transporter with PENAO reduced cell viability in a panel of neuroblastoma cell lines in a TP53-status-dependant manner. We identified the histone deacetylase inhibitor, suberanilohydroxamic acid (SAHA), as the most effective drug in clinical use against mutant TP53 neuroblastoma cells. SAHA and PENAO synergistically reduced cell viability, and induced apoptosis, in neuroblastoma cells independent of TP53-status. The SAHA and PENAO drug combination significantly delayed tumour progression in pre-clinical neuroblastoma mouse models, suggesting that these clinically advanced inhibitors may be effective in treating the disease.
- Published
- 2023
7. A novel transcriptional signature identifies T-cell infiltration in high-risk paediatric cancer.
- Author
-
Mayoh, C, Gifford, AJ, Terry, R, Lau, LMS, Wong, M, Rao, P, Shai-Hee, T, Saletta, F, Khuong-Quang, D-A, Qin, V, Mateos, MK, Meyran, D, Miller, KE, Yuksel, A, Mould, EVA, Bowen-James, R, Govender, D, Senapati, A, Zhukova, N, Omer, N, Dholaria, H, Alvaro, F, Tapp, H, Diamond, Y, Pozza, LD, Moore, AS, Nicholls, W, Gottardo, NG, McCowage, G, Hansford, JR, Khaw, S-L, Wood, PJ, Catchpoole, D, Cottrell, CE, Mardis, ER, Marshall, GM, Tyrrell, V, Haber, M, Ziegler, DS, Vittorio, O, Trapani, JA, Cowley, MJ, Neeson, PJ, Ekert, PG, Mayoh, C, Gifford, AJ, Terry, R, Lau, LMS, Wong, M, Rao, P, Shai-Hee, T, Saletta, F, Khuong-Quang, D-A, Qin, V, Mateos, MK, Meyran, D, Miller, KE, Yuksel, A, Mould, EVA, Bowen-James, R, Govender, D, Senapati, A, Zhukova, N, Omer, N, Dholaria, H, Alvaro, F, Tapp, H, Diamond, Y, Pozza, LD, Moore, AS, Nicholls, W, Gottardo, NG, McCowage, G, Hansford, JR, Khaw, S-L, Wood, PJ, Catchpoole, D, Cottrell, CE, Mardis, ER, Marshall, GM, Tyrrell, V, Haber, M, Ziegler, DS, Vittorio, O, Trapani, JA, Cowley, MJ, Neeson, PJ, and Ekert, PG
- Abstract
BACKGROUND: Molecular profiling of the tumour immune microenvironment (TIME) has enabled the rational choice of immunotherapies in some adult cancers. In contrast, the TIME of paediatric cancers is relatively unexplored. We speculated that a more refined appreciation of the TIME in childhood cancers, rather than a reliance on commonly used biomarkers such as tumour mutation burden (TMB), neoantigen load and PD-L1 expression, is an essential prerequisite for improved immunotherapies in childhood solid cancers. METHODS: We combined immunohistochemistry (IHC) with RNA sequencing and whole-genome sequencing across a diverse spectrum of high-risk paediatric cancers to develop an alternative, expression-based signature associated with CD8+ T-cell infiltration of the TIME. Furthermore, we explored transcriptional features of immune archetypes and T-cell receptor sequencing diversity, assessed the relationship between CD8+ and CD4+ abundance by IHC and deconvolution predictions and assessed the common adult biomarkers such as neoantigen load and TMB. RESULTS: A novel 15-gene immune signature, Immune Paediatric Signature Score (IPASS), was identified. Using this signature, we estimate up to 31% of high-risk cancers harbour infiltrating T-cells. In addition, we showed that PD-L1 protein expression is poorly correlated with PD-L1 RNA expression and TMB and neoantigen load are not predictive of T-cell infiltration in paediatrics. Furthermore, deconvolution algorithms are only weakly correlated with IHC measurements of T-cells. CONCLUSIONS: Our data provides new insights into the variable immune-suppressive mechanisms dampening responses in paediatric solid cancers. Effective immune-based interventions in high-risk paediatric cancer will require individualised analysis of the TIME.
- Published
- 2023
8. In vitro and in vivo drug screens of tumor cells identify novel therapies for high-risk child cancer
- Author
-
Lau, LMS, Mayoh, C, Xie, J, Barahona, P, MacKenzie, KL, Wong, M, Kamili, A, Tsoli, M, Failes, TW, Kumar, A, Mould, EVA, Gifford, A, Chow, S-O, Pinese, M, Fletcher, J, Arndt, GM, Khuong-Quang, D-A, Wadham, C, Eden, G, Trebilcock, P, Joshi, S, Alfred, S, Gopalakrishnan, A, Khan, A, Wade, DG, Strong, PA, Manouvrier, E, Morgan, LT, Cadiz, R, Ung, C, Thomas, DM, Tucker, KM, Warby, M, McCowage, GB, Dalla-Pozza, L, Byrne, JA, Saletta, F, Fellowes, A, Fox, SB, Norris, MD, Tyrrell, V, Trahair, TN, Lock, RB, Cowley, MJ, Ekert, PG, Haber, M, Ziegler, DS, Marshall, GM, Lau, LMS, Mayoh, C, Xie, J, Barahona, P, MacKenzie, KL, Wong, M, Kamili, A, Tsoli, M, Failes, TW, Kumar, A, Mould, EVA, Gifford, A, Chow, S-O, Pinese, M, Fletcher, J, Arndt, GM, Khuong-Quang, D-A, Wadham, C, Eden, G, Trebilcock, P, Joshi, S, Alfred, S, Gopalakrishnan, A, Khan, A, Wade, DG, Strong, PA, Manouvrier, E, Morgan, LT, Cadiz, R, Ung, C, Thomas, DM, Tucker, KM, Warby, M, McCowage, GB, Dalla-Pozza, L, Byrne, JA, Saletta, F, Fellowes, A, Fox, SB, Norris, MD, Tyrrell, V, Trahair, TN, Lock, RB, Cowley, MJ, Ekert, PG, Haber, M, Ziegler, DS, and Marshall, GM
- Abstract
Biomarkers which better match anticancer drugs with cancer driver genes hold the promise of improved clinical responses and cure rates. We developed a precision medicine platform of rapid high-throughput drug screening (HTS) and patient-derived xenografting (PDX) of primary tumor tissue, and evaluated its potential for treatment identification among 56 consecutively enrolled high-risk pediatric cancer patients, compared with conventional molecular genomics and transcriptomics. Drug hits were seen in the majority of HTS and PDX screens, which identified therapeutic options for 10 patients for whom no targetable molecular lesions could be found. Screens also provided orthogonal proof of drug efficacy suggested by molecular analyses and negative results for some molecular findings. We identified treatment options across the whole testing platform for 70% of patients. Only molecular therapeutic recommendations were provided to treating oncologists and led to a change in therapy in 53% of patients, of whom 29% had clinical benefit. These data indicate that in vitro and in vivo drug screening of tumor cells could increase therapeutic options and improve clinical outcomes for high-risk pediatric cancer patients.
- Published
- 2022
9. Whole-genome sequencing facilitates patient-specific quantitative PCR-based minimal residual disease monitoring in acute lymphoblastic leukaemia, neuroblastoma and Ewing sarcoma
- Author
-
Subhash, VV, Huang, L, Kamili, A, Wong, M, Chen, D, Venn, NC, Atkinson, C, Mayoh, C, Venkat, P, Tyrrell, V, Marshall, GM, Cowley, MJ, Ekert, PG, Norris, MD, Haber, M, Henderson, MJ, Sutton, R, Fletcher, J, Trahair, TN, Subhash, VV, Huang, L, Kamili, A, Wong, M, Chen, D, Venn, NC, Atkinson, C, Mayoh, C, Venkat, P, Tyrrell, V, Marshall, GM, Cowley, MJ, Ekert, PG, Norris, MD, Haber, M, Henderson, MJ, Sutton, R, Fletcher, J, and Trahair, TN
- Abstract
BACKGROUND: Minimal residual disease (MRD) measurement is a cornerstone of contemporary acute lymphoblastic leukaemia (ALL) treatment. The presence of immunoglobulin (Ig) and T cell receptor (TCR) gene recombinations in leukaemic clones allows widespread use of patient-specific, DNA-based MRD assays. In contrast, paediatric solid tumour MRD remains experimental and has focussed on generic assays targeting tumour-specific messenger RNA, methylated DNA or microRNA. METHODS: We examined the feasibility of using whole-genome sequencing (WGS) data to design tumour-specific polymerase chain reaction (PCR)-based MRD tests (WGS-MRD) in 18 children with high-risk relapsed cancer, including ALL, high-risk neuroblastoma (HR-NB) and Ewing sarcoma (EWS) (n = 6 each). RESULTS: Sensitive WGS-MRD assays were generated for each patient and allowed quantitation of 1 tumour cell per 10-4 (0.01%)-10-5 (0.001%) mononuclear cells. In ALL, WGS-MRD and Ig/TCR-MRD were highly concordant. WGS-MRD assays also showed good concordance between quantitative PCR and droplet digital PCR formats. In serial clinical samples, WGS-MRD correlated with disease course. In solid tumours, WGS-MRD assays were more sensitive than RNA-MRD assays. CONCLUSIONS: WGS facilitated the development of patient-specific MRD tests in ALL, HR-NB and EWS with potential clinical utility in monitoring treatment response. WGS data could be used to design patient-specific MRD assays in a broad range of tumours.
- Published
- 2022
10. Suppression of the ABCA1 Cholesterol Transporter Impairs the Growth and Migration of Epithelial Ovarian Cancer
- Author
-
Gao, J, Jung, M, Williams, RT, Hui, D, Russell, AJ, Naim, AJ, Kamili, A, Clifton, M, Bongers, A, Mayoh, C, Ho, G, Scott, CL, Jessup, W, Haber, M, Norris, MD, Henderson, MJ, Gao, J, Jung, M, Williams, RT, Hui, D, Russell, AJ, Naim, AJ, Kamili, A, Clifton, M, Bongers, A, Mayoh, C, Ho, G, Scott, CL, Jessup, W, Haber, M, Norris, MD, and Henderson, MJ
- Abstract
BACKGROUND: Epithelial ovarian cancer (EOC) is the most lethal gynaecological malignancy with over 80% of cases already disseminated at diagnosis and facing a dismal five-year survival rate of 35%. EOC cells often spread to the greater omentum where they take-up cholesterol. Excessive amounts of cholesterol can be cytocidal, suggesting that cholesterol efflux through transporters may be important to maintain homeostasis, and this may explain the observation that high expression of the ATP-binding cassette A1 (ABCA1) cholesterol transporter has been associated with poor outcome in EOC patients. METHODS: ABCA1 expression was silenced in EOC cells to investigate the effect of inhibiting cholesterol efflux on EOC biology through growth and migration assays, three-dimensional spheroid culture and cholesterol quantification. RESULTS: ABCA1 suppression significantly reduced the growth, motility and colony formation of EOC cell lines as well as the size of EOC spheroids, whilst stimulating expression of ABCA1 reversed these effects. In serous EOC cells, ABCA1 suppression induced accumulation of cholesterol. Lowering cholesterol levels using methyl-B-cyclodextrin rescued the effect of ABCA1 suppression, restoring EOC growth. Furthermore, we identified FDA-approved agents that induced cholesterol accumulation and elicited cytocidal effects in EOC cells. CONCLUSIONS: Our data demonstrate the importance of ABCA1 in maintaining cholesterol balance and malignant properties in EOC cells, highlighting its potential as a therapeutic target for this disease.
- Published
- 2022
11. Glutamine addiction promotes glucose oxidation in triple-negative breast cancer
- Author
-
Quek, L-E, van Geldermalsen, M, Guan, YF, Wahi, K, Mayoh, C, Balaban, S, Pang, A, Wang, Q, Cowley, MJ, Brown, KK, Turner, N, Hoy, AJ, Holst, J, Quek, L-E, van Geldermalsen, M, Guan, YF, Wahi, K, Mayoh, C, Balaban, S, Pang, A, Wang, Q, Cowley, MJ, Brown, KK, Turner, N, Hoy, AJ, and Holst, J
- Abstract
Glutamine is a conditionally essential nutrient for many cancer cells, but it remains unclear how consuming glutamine in excess of growth requirements confers greater fitness to glutamine-addicted cancers. By contrasting two breast cancer subtypes with distinct glutamine dependencies, we show that glutamine-indispensable triple-negative breast cancer (TNBC) cells rely on a non-canonical glutamine-to-glutamate overflow, with glutamine carbon routed once through the TCA cycle. Importantly, this single-pass glutaminolysis increases TCA cycle fluxes and replenishes TCA cycle intermediates in TNBC cells, a process that achieves net oxidation of glucose but not glutamine. The coupling of glucose and glutamine catabolism appears hard-wired via a distinct TNBC gene expression profile biased to strip and then sequester glutamine nitrogen, but hampers the ability of TNBC cells to oxidise glucose when glutamine is limiting. Our results provide a new understanding of how metabolically rigid TNBC cells are sensitive to glutamine deprivation and a way to select vulnerable TNBC subtypes that may be responsive to metabolic-targeted therapies.
- Published
- 2022
12. Advances in CAR T cell immunotherapy for paediatric brain tumours
- Author
-
Rao, P, Furst, L, Meyran, D, Mayoh, C, Neeson, PJ, Terry, R, Khuong-Quang, D-A, Mantamadiotis, T, Ekert, PG, Rao, P, Furst, L, Meyran, D, Mayoh, C, Neeson, PJ, Terry, R, Khuong-Quang, D-A, Mantamadiotis, T, and Ekert, PG
- Abstract
Brain tumours are the most common solid tumour in children and the leading cause of cancer related death in children. Current treatments include surgery, chemotherapy and radiotherapy. The need for aggressive treatment means many survivors are left with permanent severe disability, physical, intellectual and social. Recent progress in immunotherapy, including genetically engineered T cells with chimeric antigen receptors (CARs) for treating cancer, may provide new avenues to improved outcomes for patients with paediatric brain cancer. In this review we discuss advances in CAR T cell immunotherapy, the major CAR T cell targets that are in clinical and pre-clinical development with a focus on paediatric brain tumours, the paediatric brain tumour microenvironment and strategies used to improve CAR T cell therapy for paediatric tumours.
- Published
- 2022
13. Acute central nervous system toxicity during treatment of pediatric acute lymphoblastic leukemia:phenotypes, risk factors and genotypes
- Author
-
Anastasopoulou, S. (Stavroula), Bodil Als-NielsenNielsen, R. L. (Rikke Linnemann), Als-Nielsen, B. (Bodil), Banerjee, J. (Joanna), Eriksson, M. A. (Mats A.), Helenius, M. (Marianne), Heyman, M. M. (Mats M.), Johannsdottir, I. M. (Inga Maria), Jonsson, O. G. (Olafur Gisli), MacGregor, S. (Stuart), Mateos, M. K. (Marion K.), Mayoh, C. (Chelsea), Mikkel, S. (Sirje), Myrberg, I. H. (Ida Hed), Niinimäki, R. (Riitta), Schmiegelow, K. (Kjeld), Taskinen, M. (Mervi), Vaitkeviciene, G. (Goda), Warnqvist, A. (Anna), Wolthers, B. (Benjamin), Harila-Saari, A. (Arja), Ranta, S. (Susanna), Anastasopoulou, S. (Stavroula), Bodil Als-NielsenNielsen, R. L. (Rikke Linnemann), Als-Nielsen, B. (Bodil), Banerjee, J. (Joanna), Eriksson, M. A. (Mats A.), Helenius, M. (Marianne), Heyman, M. M. (Mats M.), Johannsdottir, I. M. (Inga Maria), Jonsson, O. G. (Olafur Gisli), MacGregor, S. (Stuart), Mateos, M. K. (Marion K.), Mayoh, C. (Chelsea), Mikkel, S. (Sirje), Myrberg, I. H. (Ida Hed), Niinimäki, R. (Riitta), Schmiegelow, K. (Kjeld), Taskinen, M. (Mervi), Vaitkeviciene, G. (Goda), Warnqvist, A. (Anna), Wolthers, B. (Benjamin), Harila-Saari, A. (Arja), and Ranta, S. (Susanna)
- Abstract
Central nervous system (CNS) toxicity is common at diagnosis and during treatment of pediatric acute lymphoblastic leukemia (ALL). We studied CNS toxicity in 1,464 children aged 1.0–17.9 years, diagnosed with ALL and treated according to the Nordic Society of Pediatric Hematology and Oncology ALL2008 protocol. Genome-wide association studies, and a candidate single-nucleotide polymorphism (SNP; n=19) study were performed in 1,166 patients. Findings were validated in an independent Australian cohort of children with ALL (n=797) in whom two phenotypes were evaluated: diverse CNS toxicities (n=103) and methotrexate-related CNS toxicity (n=48). In total, 135/1,464 (9.2%) patients experienced CNS toxicity for a cumulative incidence of 8.7% (95% confidence interval: 7.31–10.20) at 12 months from diagnosis. Patients aged ≥10 years had a higher risk of CNS toxicity than had younger patients (16.3% vs. 7.4%; P<0.001). The most common CNS toxicities were posterior reversible encephalopathy syndrome (n=52, 43 with seizures), sinus venous thrombosis (n=28, 9 with seizures), and isolated seizures (n=16). The most significant SNP identified by the genome-wide association studies did not reach genomic significance (lowest P-value: 1.11x10-6), but several were annotated in genes regulating neuronal functions. In candidate SNP analysis, ATXN1 rs68082256, related to epilepsy, was associated with seizures in patients <10 years (P=0.01). ATXN1 rs68082256 was validated in the Australian cohort with diverse CNS toxicities (P=0.04). The role of ATXN1 as well as the novel SNP in neurotoxicity in pediatric ALL should be further explored.
- Published
- 2022
14. Methotrexate-related central neurotoxicity: clinical characteristics, risk factors and genome-wide association study in children treated for acute lymphoblastic leukemia.
- Author
-
Mateos, MK, Marshall, GM, Barbaro, PM, Quinn, MCJ, George, C, Mayoh, C, Sutton, R, Revesz, T, Giles, JE, Barbaric, D, Alvaro, F, Mechinaud, F, Catchpoole, D, Lawson, JA, Chenevix-Trench, G, MacGregor, S, Kotecha, RS, Dalla-Pozza, L, Trahair, TN, Mateos, MK, Marshall, GM, Barbaro, PM, Quinn, MCJ, George, C, Mayoh, C, Sutton, R, Revesz, T, Giles, JE, Barbaric, D, Alvaro, F, Mechinaud, F, Catchpoole, D, Lawson, JA, Chenevix-Trench, G, MacGregor, S, Kotecha, RS, Dalla-Pozza, L, and Trahair, TN
- Abstract
Symptomatic methotrexate-related central neurotoxicity (MTX neurotoxicity) is a severe toxicity experienced during acute lymphoblastic leukemia (ALL) therapy with potential long-term neurologic complications. Risk factors and long-term outcomes require further study. We conducted a systematic, retrospective review of 1,251 consecutive Australian children enrolled on Berlin-Frankfurt-Münster or Children's Oncology Group-based protocols between 1998-2013. Clinical risk predictors for MTX neurotoxicity were analyzed using regression. A genome-wide association study (GWAS) was performed on 48 cases and 537 controls. The incidence of MTX neurotoxicity was 7.6% (n=95 of 1,251), at a median of 4 months from ALL diagnosis and 8 days after intravenous or intrathecal MTX. Grade 3 elevation of serum aspartate aminotransferase (P=0.005, odds ratio 2.31 [range, 1.28-4.16]) in induction/consolidation was associated with MTX neurotoxicity, after accounting for the only established risk factor, age ≥10 years. Cumulative incidence of CNS relapse was increased in children where intrathecal MTX was omitted following symptomatic MTX neurotoxicity (n=48) compared to where intrathecal MTX was continued throughout therapy (n=1,174) (P=0.047). Five-year central nervous system relapse-free survival was 89.2 4.6% when intrathecal MTX was ceased compared to 95.4 0.6% when intrathecal MTX was continued. Recurrence of MTX neurotoxicity was low (12.9%) for patients whose intrathecal MTX was continued after their first episode. The GWAS identified single-nucletide polymorphism associated with MTX neurotoxicity near genes regulating neuronal growth, neuronal differentiation and cytoskeletal organization (P<1x10-6). In conclusion, increased serum aspartate aminotransferase and age ≥10 years at diagnosis were independent risk factors for MTX neurotoxicity. Our data do not support cessation of intrathecal MTX after a first MTX neurotoxicity event.
- Published
- 2022
15. Combination efficacy of ruxolitinib with standard-of-care drugs in CRLF2-rearranged Ph-like acute lymphoblastic leukemia
- Author
-
Bӧhm, JW, Sia, KCS, Jones, C, Evans, K, Mariana, A, Pang, I, Failes, T, Zhong, L, Mayoh, C, Landman, R, Collins, R, Erickson, SW, Arndt, G, Raftery, MJ, Wilkins, MR, Norris, MD, Haber, M, Marshall, GM, Lock, RB, Bӧhm, JW, Sia, KCS, Jones, C, Evans, K, Mariana, A, Pang, I, Failes, T, Zhong, L, Mayoh, C, Landman, R, Collins, R, Erickson, SW, Arndt, G, Raftery, MJ, Wilkins, MR, Norris, MD, Haber, M, Marshall, GM, and Lock, RB
- Abstract
Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a high-risk ALL subtype with high rates of relapse and poor patient outcome. Activating mutations affecting components of the JAK-STAT signaling pathway occur in the majority of Ph-like ALL cases. The use of JAK inhibitors represents a potential treatment option for Ph-like ALL, although we and others have shown that CRLF2-rearranged Ph-like ALL responds poorly to single-agent JAK inhibitors in the preclinical setting. Therefore, the aim of this study was to identify effective combination treatments against CRLF2-rearranged Ph-like ALL, and to elucidate the underlying mechanisms of synergy. We carried out a series of high-throughput combination drug screenings and found that ruxolitinib exerted synergy with standard-of-care drugs used in the treatment of ALL. In addition, we investigated the molecular effects of ruxolitinib on Ph-like ALL by combining mass spectrometry phosphoproteomics with gene expression analysis. Based on these findings, we conducted preclinical in vivo drug testing and demonstrated that ruxolitinib enhanced the in vivo efficacy of an induction-type regimen consisting of vincristine, dexamethasone, and l-asparaginase in 2/3 CRLF2-rearranged Ph-like ALL xenografts. Overall, our findings support evaluating the addition of ruxolitinib to conventional induction regimens for the treatment of CRLF2-rearranged Ph-like ALL.
- Published
- 2021
16. Whole-genome sequencing facilitates patient-specific quantitative PCR-based minimal residual disease monitoring in acute lymphoblastic leukaemia, neuroblastoma and Ewing sarcoma
- Author
-
Subhash, VV, Huang, L, Kamili, A, Wong, M, Chen, D, Venn, NC, Atkinson, C, Mayoh, C, Venkat, P, Tyrrell, V, Marshall, GM, Cowley, MJ, Ekert, PG, Norris, MD, Haber, M, Henderson, MJ, Sutton, R, Fletcher, JI, Trahair, TN, Subhash, VV, Huang, L, Kamili, A, Wong, M, Chen, D, Venn, NC, Atkinson, C, Mayoh, C, Venkat, P, Tyrrell, V, Marshall, GM, Cowley, MJ, Ekert, PG, Norris, MD, Haber, M, Henderson, MJ, Sutton, R, Fletcher, JI, and Trahair, TN
- Abstract
Background: Minimal residual disease (MRD) measurement is a cornerstone of contemporary acute lymphoblastic leukaemia (ALL) treatment. The presence of immunoglobulin (Ig) and T cell receptor (TCR) gene recombinations in leukaemic clones allows widespread use of patient-specific, DNA-based MRD assays. In contrast, paediatric solid tumour MRD remains experimental and has focussed on generic assays targeting tumour-specific messenger RNA, methylated DNA or microRNA. Methods: We examined the feasibility of using whole-genome sequencing (WGS) data to design tumour-specific polymerase chain reaction (PCR)-based MRD tests (WGS-MRD) in 18 children with high-risk relapsed cancer, including ALL, high-risk neuroblastoma (HR-NB) and Ewing sarcoma (EWS) (n = 6 each). Results: Sensitive WGS-MRD assays were generated for each patient and allowed quantitation of 1 tumour cell per 10−4 (0.01%)–10–5 (0.001%) mononuclear cells. In ALL, WGS-MRD and Ig/TCR-MRD were highly concordant. WGS-MRD assays also showed good concordance between quantitative PCR and droplet digital PCR formats. In serial clinical samples, WGS-MRD correlated with disease course. In solid tumours, WGS-MRD assays were more sensitive than RNA-MRD assays. Conclusions: WGS facilitated the development of patient-specific MRD tests in ALL, HR-NB and EWS with potential clinical utility in monitoring treatment response. WGS data could be used to design patient-specific MRD assays in a broad range of tumours.
- Published
- 2021
17. The important role of routine cytopathology in pediatric precision oncology
- Author
-
Xie, J, Kumar, A, Dolman, MEM, Mayoh, C, Khuong-Quang, DA, Cadiz, R, Wong-Erasmus, M, Mould, EVA, Grebert-Wade, D, Barahona, P, Kamili, A, Tsoli, M, Failes, TW, Chow, SO, Bhatia, K, Marshall, GM, Ziegler, DS, Haber, M, Lock, RB, Tyrrell, V, Lau, L, Athanasatos, P, Gifford, AJ, Xie, J, Kumar, A, Dolman, MEM, Mayoh, C, Khuong-Quang, DA, Cadiz, R, Wong-Erasmus, M, Mould, EVA, Grebert-Wade, D, Barahona, P, Kamili, A, Tsoli, M, Failes, TW, Chow, SO, Bhatia, K, Marshall, GM, Ziegler, DS, Haber, M, Lock, RB, Tyrrell, V, Lau, L, Athanasatos, P, and Gifford, AJ
- Abstract
BACKGROUND: The development of high-throughput drug screening (HTS) using primary cultures provides a promising, clinically translatable approach to tailoring treatment strategies for patients with cancer. However, this has been challenging for solid tumors because of often limited amounts of tissue available. In most cases, in vitro expansion is required before HTS, which may lead to overgrowth and contamination by non-neoplastic cells. METHODS: In this study, hematoxylin and eosin staining and immunohistochemical staining were performed on 129 cytopathology cases from 95 patients. These cytopathology cases comprised cell block preparations derived from primary tumor specimens or patient-derived xenografts as part of a pediatric precision oncology trial. Cytopathology cases were compared with the morphology and immunohistochemical staining profile of the original tumor. Cases were reported as tumor cells present, equivocal, or tumor cells absent. The HTS results from cytopathologically validated cultures were incorporated into a multidisciplinary tumor board report issued to the treating clinician to guide clinical decision making. RESULTS: On cytopathologic examination, tumor cells were present in 77 of 129 cases (60%) and were absent in 38 of 129 cases (29%), whereas 14 of 129 cases (11%) were equivocal. Cultures that contained tumor cells resembled the tumors from which they were derived. CONCLUSIONS: Cytopathologic examination of tumor cell block preparations is feasible and provides detailed morphologic characterization. Cytopathologic examination is essential for ensuring that samples submitted for HTS contain representative tumor cells and that in vitro drug sensitivity data are clinically translatable.
- Published
- 2021
18. Dual targeting of chromatin stability by the curaxin CBL0137 and histone deacetylase inhibitor panobinostat shows significant preclinical efficacy in neuroblastoma
- Author
-
Xiao, L, Somers, K, Murray, J, Pandher, R, Karsa, M, Ronca, E, Bongers, A, Terry, R, Ehteda, A, Gamble, LD, Issaeva, N, Leonova, KI, O’Connor, A, Mayoh, C, Venkat, P, Quek, H, Brand, J, Kusuma, FK, Pettitt, JA, Mosmann, E, Kearns, A, Eden, G, Alfred, S, Allan, S, Zhai, L, Kamili, A, Gifford, AJ, Carter, DR, Henderson, MJ, Fletcher, JI, Marshall, G, Johnstone, RW, Cesare, AJ, Ziegler, DS, Gudkov, AV, Gurova, KV, Norris, MD, Haber, M, Xiao, L, Somers, K, Murray, J, Pandher, R, Karsa, M, Ronca, E, Bongers, A, Terry, R, Ehteda, A, Gamble, LD, Issaeva, N, Leonova, KI, O’Connor, A, Mayoh, C, Venkat, P, Quek, H, Brand, J, Kusuma, FK, Pettitt, JA, Mosmann, E, Kearns, A, Eden, G, Alfred, S, Allan, S, Zhai, L, Kamili, A, Gifford, AJ, Carter, DR, Henderson, MJ, Fletcher, JI, Marshall, G, Johnstone, RW, Cesare, AJ, Ziegler, DS, Gudkov, AV, Gurova, KV, Norris, MD, and Haber, M
- Abstract
Purpose: We investigated whether targeting chromatin stability through a combination of the curaxin CBL0137 with the histone deacetylase (HDAC) inhibitor, panobinostat, constitutes an effective multimodal treatment for high-risk neuroblastoma. Experimental Design: The effects of the drug combination on cancer growth were examined in vitro and in animal models of MYCN-amplified neuroblastoma. The molecular mechanisms of action were analyzed by multiple techniques including whole transcriptome profiling, immune deconvolution analysis, immunofluorescence, flow cytometry, pulsed-field gel electrophoresis, assays to assess cell growth and apoptosis, and a range of cell-based reporter systems to examine histone eviction, heterochromatin transcription, and chromatin compaction. Results: The combination of CBL0137 and panobinostat enhanced nucleosome destabilization, induced an IFN response, inhibited DNA damage repair, and synergistically suppressed cancer cell growth. Similar synergistic effects were observed when combining CBL0137 with other HDAC inhibitors. The CBL0137/panobinostat combination significantly delayed cancer progression in xenograft models of poor outcome high-risk neuroblastoma. Complete tumor regression was achieved in the transgenic Th-MYCN neuroblastoma model which was accompanied by induction of a type I IFN and immune response. Tumor transplantation experiments further confirmed that the presence of a competent adaptive immune system component allowed the exploitation of the full potential of the drug combination. Conclusions: The combination of CBL0137 and panobinostat is effective and well-tolerated in preclinical models of aggressive high-risk neuroblastoma, warranting further preclinical and clinical investigation in other pediatric cancers. On the basis of its potential to boost IFN and immune responses in cancer models, the drug combination holds promising potential for addition to immunotherapies.
- Published
- 2021
19. A novel combination therapy targeting ubiquitin-specific protease 5 in MYCN-driven neuroblastoma
- Author
-
Cheung, BB, Kleynhans, A, Mittra, R, Kim, PY, Holien, JK, Nagy, Z, Ciampa, OC, Seneviratne, JA, Mayoh, C, Raipuria, M, Gadde, S, Massudi, H, Wong, IPL, Tan, O, Gong, A, Suryano, A, Diakiw, SM, Liu, B, Arndt, GM, Liu, T, Kumar, N, Sangfelt, O, Zhu, S, Norris, MD, Haber, M, Carter, DR, Parker, MW, Marshall, GM, Cheung, BB, Kleynhans, A, Mittra, R, Kim, PY, Holien, JK, Nagy, Z, Ciampa, OC, Seneviratne, JA, Mayoh, C, Raipuria, M, Gadde, S, Massudi, H, Wong, IPL, Tan, O, Gong, A, Suryano, A, Diakiw, SM, Liu, B, Arndt, GM, Liu, T, Kumar, N, Sangfelt, O, Zhu, S, Norris, MD, Haber, M, Carter, DR, Parker, MW, and Marshall, GM
- Abstract
Histone deacetylase (HDAC) inhibitors are effective in MYCN-driven cancers, because of a unique need for HDAC recruitment by the MYCN oncogenic signal. However, HDAC inhibitors are much more effective in combination with other anti-cancer agents. To identify novel compounds which act synergistically with HDAC inhibitor, such as suberanoyl hydroxamic acid (SAHA), we performed a cell-based, high-throughput drug screen of 10,560 small molecule compounds from a drug-like diversity library and identified a small molecule compound (SE486-11) which synergistically enhanced the cytotoxic effects of SAHA. Effects of drug combinations on cell viability, proliferation, apoptosis and colony forming were assessed in a panel of neuroblastoma cell lines. Treatment with SAHA and SE486-11 increased MYCN ubiquitination and degradation, and markedly inhibited tumorigenesis in neuroblastoma xenografts, and, MYCN transgenic zebrafish and mice. The combination reduced ubiquitin-specific protease 5 (USP5) levels and increased unanchored polyubiquitin chains. Overexpression of USP5 rescued neuroblastoma cells from the cytopathic effects of the combination and reduced unanchored polyubiquitin, suggesting USP5 is a therapeutic target of the combination. SAHA and SE486-11 directly bound to USP5 and the drug combination exhibited a 100-fold higher binding to USP5 than individual drugs alone in microscale thermophoresis assays. MYCN bound to the USP5 promoter and induced USP5 gene expression suggesting that USP5 and MYCN expression created a forward positive feedback loop in neuroblastoma cells. Thus, USP5 acts as an oncogenic cofactor with MYCN in neuroblastoma and the novel combination of HDAC inhibitor with SE486-11 represents a novel therapeutic approach for the treatment of MYCN-driven neuroblastoma.
- Published
- 2021
20. Dual targeting of polyamine synthesis and uptake in diffuse intrinsic pontine gliomas
- Author
-
Khan, A, Gamble, LD, Upton, DH, Ung, C, Yu, DMT, Ehteda, A, Pandher, R, Mayoh, C, Hébert, S, Jabado, N, Kleinman, CL, Burns, MR, Norris, MD, Haber, M, Tsoli, M, Ziegler, DS, Khan, A, Gamble, LD, Upton, DH, Ung, C, Yu, DMT, Ehteda, A, Pandher, R, Mayoh, C, Hébert, S, Jabado, N, Kleinman, CL, Burns, MR, Norris, MD, Haber, M, Tsoli, M, and Ziegler, DS
- Abstract
Diffuse intrinsic pontine glioma (DIPG) is an incurable malignant childhood brain tumor, with no active systemic therapies and a 5-year survival of less than 1%. Polyamines are small organic polycations that are essential for DNA replication, translation and cell proliferation. Ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme in polyamine synthesis, is irreversibly inhibited by difluoromethylornithine (DFMO). Herein we show that polyamine synthesis is upregulated in DIPG, leading to sensitivity to DFMO. DIPG cells compensate for ODC1 inhibition by upregulation of the polyamine transporter SLC3A2. Treatment with the polyamine transporter inhibitor AMXT 1501 reduces uptake of polyamines in DIPG cells, and co-administration of AMXT 1501 and DFMO leads to potent in vitro activity, and significant extension of survival in three aggressive DIPG orthotopic animal models. Collectively, these results demonstrate the potential of dual targeting of polyamine synthesis and uptake as a therapeutic strategy for incurable DIPG.
- Published
- 2021
21. An ALYREF-MYCN coactivator complex drives neuroblastoma tumorigenesis through effects on USP3 and MYCN stability
- Author
-
Nagy, Z, Seneviratne, JA, Kanikevich, M, Chang, W, Mayoh, C, Venkat, P, Du, Y, Jiang, C, Salib, A, Koach, J, Carter, DR, Mittra, R, Liu, T, Parker, MW, Cheung, BB, Marshall, GM, Nagy, Z, Seneviratne, JA, Kanikevich, M, Chang, W, Mayoh, C, Venkat, P, Du, Y, Jiang, C, Salib, A, Koach, J, Carter, DR, Mittra, R, Liu, T, Parker, MW, Cheung, BB, and Marshall, GM
- Abstract
To achieve the very high oncoprotein levels required to drive the malignant state cancer cells utilise the ubiquitin proteasome system to upregulate transcription factor levels. Here our analyses identify ALYREF, expressed from the most common genetic copy number variation in neuroblastoma, chromosome 17q21-ter gain as a key regulator of MYCN protein turnover. We show strong co-operativity between ALYREF and MYCN from transgenic models of neuroblastoma in vitro and in vivo. The two proteins form a nuclear coactivator complex which stimulates transcription of the ubiquitin specific peptidase 3, USP3. We show that increased USP3 levels reduce K-48- and K-63-linked ubiquitination of MYCN, thus driving up MYCN protein stability. In the MYCN-ALYREF-USP3 signal, ALYREF is required for MYCN effects on the malignant phenotype and that of USP3 on MYCN stability. This data defines a MYCN oncoprotein dependency state which provides a rationale for future pharmacological studies.
- Published
- 2021
22. Dual targeting of the epigenome via FACT complex and histone deacetylase is a potent treatment strategy for DIPG
- Author
-
Ehteda, A, Simon, S, Franshaw, L, Giorgi, FM, Liu, J, Joshi, S, Rouaen, JRC, Pang, CNI, Pandher, R, Mayoh, C, Tang, Y, Khan, A, Ung, C, Tolhurst, O, Kankean, A, Hayden, E, Lehmann, R, Shen, S, Gopalakrishnan, A, Trebilcock, P, Gurova, K, Gudkov, AV, Norris, MD, Haber, M, Vittorio, O, Tsoli, M, Ziegler, DS, Ehteda, A, Simon, S, Franshaw, L, Giorgi, FM, Liu, J, Joshi, S, Rouaen, JRC, Pang, CNI, Pandher, R, Mayoh, C, Tang, Y, Khan, A, Ung, C, Tolhurst, O, Kankean, A, Hayden, E, Lehmann, R, Shen, S, Gopalakrishnan, A, Trebilcock, P, Gurova, K, Gudkov, AV, Norris, MD, Haber, M, Vittorio, O, Tsoli, M, and Ziegler, DS
- Abstract
Diffuse intrinsic pontine glioma (DIPG) is an aggressive and incurable childhood brain tumor for which new treatments are needed. CBL0137 is an anti-cancer compound developed from quinacrine that targets facilitates chromatin transcription (FACT), a chromatin remodeling complex involved in transcription, replication, and DNA repair. We show that CBL0137 displays profound cytotoxic activity against a panel of patient-derived DIPG cultures by restoring tumor suppressor TP53 and Rb activity. Moreover, in an orthotopic model of DIPG, treatment with CBL0137 significantly extends animal survival. The FACT subunit SPT16 is found to directly interact with H3.3K27M, and treatment with CBL0137 restores both histone H3 acetylation and trimethylation. Combined treatment of CBL0137 with the histone deacetylase inhibitor panobinostat leads to inhibition of the Rb/E2F1 pathway and induction of apoptosis. The combination of CBL0137 and panobinostat significantly prolongs the survival of mice bearing DIPG orthografts, suggesting a potential treatment strategy for DIPG.
- Published
- 2021
23. Targeted therapy of TERT-rearranged neuroblastoma with BET bromodomain inhibitor and proteasome inhibitor combination therapy
- Author
-
Chen, J, Nelson, C, Wong, M, Tee, AE, Liu, PY, La, T, Fletcher, JI, Kamili, A, Mayoh, C, Bartenhagen, C, Trahair, TN, Xu, N, Jayatilleke, N, Peng, H, Atmadibrata, B, Cheung, BB, Lan, Q, Bryan, TM, Mestdagh, P, Vandesompele, J, Combaret, V, Boeva, V, Wang, JY, Janoueix-Lerosey, I, Cowley, MJ, MacKenzie, KL, Dolnikov, A, Li, J, Polly, P, Marshall, GM, Reddel, RR, Norris, MD, Haber, M, Fischer, M, Zhang, XD, Pickett, HA, Liu, T, Chen, J, Nelson, C, Wong, M, Tee, AE, Liu, PY, La, T, Fletcher, JI, Kamili, A, Mayoh, C, Bartenhagen, C, Trahair, TN, Xu, N, Jayatilleke, N, Peng, H, Atmadibrata, B, Cheung, BB, Lan, Q, Bryan, TM, Mestdagh, P, Vandesompele, J, Combaret, V, Boeva, V, Wang, JY, Janoueix-Lerosey, I, Cowley, MJ, MacKenzie, KL, Dolnikov, A, Li, J, Polly, P, Marshall, GM, Reddel, RR, Norris, MD, Haber, M, Fischer, M, Zhang, XD, Pickett, HA, and Liu, T
- Abstract
Purpose: TERT gene rearrangement with transcriptional superenhancers leads to TERT overexpression and neuroblastoma. No targeted therapy is available for clinical trials in patients with TERT-rearranged neuroblastoma. Experimental Design: Anticancer agents exerting the best synergistic anticancer effects with BET bromodomain inhibitors were identified by screening an FDA-approved oncology drug library. The synergistic effects of the BET bromodomain inhibitor OTX015 and the proteasome inhibitor carfilzomib were examined by immunoblot and flow cytometry analysis. The anticancer efficacy of OTX015 and carfilzomib combination therapy was investigated in mice xenografted with TERT-rearranged neuroblastoma cell lines or patient-derived xenograft (PDX) tumor cells, and the role of TERT reduction in the anticancer efficacy was examined through rescue experiments in mice. Results: The BET bromodomain protein BRD4 promoted TERT-rearranged neuroblastoma cell proliferation through upregulating TERT expression. Screening of an approved oncology drug library identified the proteasome inhibitor carfilzomib as the agent exerting the best synergistic anticancer effects with BET bromodomain inhibitors including OTX015. OTX015 and carfilzomib synergistically reduced TERT protein expression, induced endoplasmic reticulum stress, and induced TERT-rearranged neuroblastoma cell apoptosis which was blocked by TERT overexpression and endoplasmic reticulum stress antagonists. In mice xenografted with TERT-rearranged neuroblastoma cell lines or PDX tumor cells, OTX015 and carfilzomib synergistically blocked TERT expression, induced tumor cell apoptosis, suppressed tumor progression, and improved mouse survival, which was largely reversed by forced TERT overexpression. Conclusions: OTX015 and carfilzomib combination therapy is likely to be translated into the first clinical trial of a targeted therapy in patients with TERT-rearranged neuroblastoma.
- Published
- 2021
24. Whole genome, transcriptome and methylome profiling enhances actionable target discovery in high-risk pediatric cancer.
- Author
-
Lau L.M.S., Baber J., Priestley P., Dolman M.E.M., Fleuren E.D.G., Gauthier M.-E., Mould E.V.A., Gayevskiy V., Gifford A.J., Grebert-Wade D., Mayoh C., Bolanos N.A., Khuong-Quang D.-A., Pinese M., Kumar A., Barahona P., Wilkie E.E., Sullivan P., Bowen-James R., Syed M., Martincorena I., Abascal F., Sherstyuk A., Strong P.A., Manouvrier E., Warby M., Thomas D.M., Kirk J., Tucker K., O'Brien T., Alvaro F., McCowage G.B., Dalla-Pozza L., Gottardo N.G., Tapp H., Wood P., Khaw S.-L., Hansford J.R., Moore A.S., Norris M.D., Trahair T.N., Lock R.B., Tyrrell V., Haber M., Marshall G.M., Ziegler D.S., Ekert P.G., Cowley M.J., Wong M., Lau L.M.S., Baber J., Priestley P., Dolman M.E.M., Fleuren E.D.G., Gauthier M.-E., Mould E.V.A., Gayevskiy V., Gifford A.J., Grebert-Wade D., Mayoh C., Bolanos N.A., Khuong-Quang D.-A., Pinese M., Kumar A., Barahona P., Wilkie E.E., Sullivan P., Bowen-James R., Syed M., Martincorena I., Abascal F., Sherstyuk A., Strong P.A., Manouvrier E., Warby M., Thomas D.M., Kirk J., Tucker K., O'Brien T., Alvaro F., McCowage G.B., Dalla-Pozza L., Gottardo N.G., Tapp H., Wood P., Khaw S.-L., Hansford J.R., Moore A.S., Norris M.D., Trahair T.N., Lock R.B., Tyrrell V., Haber M., Marshall G.M., Ziegler D.S., Ekert P.G., Cowley M.J., and Wong M.
- Abstract
The Zero Childhood Cancer Program is a precision medicine program to benefit children with poor-outcome, rare, relapsed or refractory cancer. Using tumor and germline whole genome sequencing (WGS) and RNA sequencing (RNAseq) across 252 tumors from high-risk pediatric patients with cancer, we identified 968 reportable molecular aberrations (39.9% in WGS and RNAseq, 35.1% in WGS only and 25.0% in RNAseq only). Of these patients, 93.7% had at least one germline or somatic aberration, 71.4% had therapeutic targets and 5.2% had a change in diagnosis. WGS identified pathogenic cancer-predisposing variants in 16.2% of patients. In 76 central nervous system tumors, methylome analysis confirmed diagnosis in 71.1% of patients and contributed to a change of diagnosis in two patients (2.6%). To date, 43 patients have received a recommended therapy, 38 of whom could be evaluated, with 31% showing objective evidence of clinical benefit. Comprehensive molecular profiling resolved the molecular basis of virtually all high-risk cancers, leading to clinical benefit in some patients.Copyright © 2020, The Author(s), under exclusive licence to Springer Nature America, Inc.
- Published
- 2021
25. Precision Medicine for High-Risk Paediatric Cancers - Molecular Tumour Board Recommendations and Treatment Responses (Zero Childhood Cancer Program).
- Author
-
Lau L., Khuong-Quang D.-A., Nagabushan S., Senapati A., Barahona P., Ajuyah P., Altekoester A.-K., Wong M., Mayoh C., Mould E., Fuentes-Bolanos N., Zhukova N., Cowley M., Ekert P., Tyrrell V., Trahair T., Haber M., Marshall G., Ziegler D., Lau L., Khuong-Quang D.-A., Nagabushan S., Senapati A., Barahona P., Ajuyah P., Altekoester A.-K., Wong M., Mayoh C., Mould E., Fuentes-Bolanos N., Zhukova N., Cowley M., Ekert P., Tyrrell V., Trahair T., Haber M., Marshall G., and Ziegler D.
- Abstract
Background and Aims: Advances in our knowledge of the molecular landscape of cancer has led to the development of targeted therapy and personalised anti-cancer treatment. The PRISM trial (ZERO Childhood Cancer Program) evaluates the benefits of a comprehensive precision medicine approach for high-risk paediatric cancer (survival <30%). Method(s): WGS/RNA-seq/methylation results were discussed in a national Molecular Tumour Board (MTB) with 5 tiers of therapeutic recommendations: Tier 1and 2, clinical evidence in same and different cancer type, respectively; Tier 3 and 4, preclinical evidence in same and different cancer type, respectively; Tier 5, consensus opinion. Result(s): 386 patients (43% diagnosis and 57% relapse/refractory) were enrolled over 3 years, with median time to MTB of 6 weeks. A total of 500 recommendations (21% Tier 1, 38% Tier 2, 32% Tier 3, 14% Tier 4 and 5% Tier 5) were made in 261 (67%) patients (median: 1 recommendation/patient; range 1 - 6). 85% were single-agent recommendations and 15% were combination treatments. The PI3K/mTOR, MAPK and cell cycle/cyclin pathway were the 3 most frequently targeted pathways. 87/261 (33%) patients received MTB recommended treatment (median start time 5.3 weeks). Of the first 43 patients who received MTB recommended treatment, 35 could be evaluated for response: 11% complete response (CR), 20% partial response (PR), 40% stable disease (SD) and 29% progressive disease (PD). The duration of SD was >=24 weeks in 9/14 patients. The clinical benefit rate (CR, PR and SD >24 weeks) was 57%. The proportion of tier 1 and 2 recommendations was similar across all response groups. The CR+PR, SD and PD group consisted of 45%, 50% and 50% of tier 1 and 2 recommendations, respectively. Conclusion(s): The ZERO platform provided personalised therapeutic options for 67% of patients and clinical benefit was observed in majority who received the recommended treatment.
- Published
- 2021
26. Efficacy of MEK inhibition in a recurrent malignant peripheral nerve sheath tumor
- Author
-
Nagabushan, S, Lau, LMS, Barahona, P, Wong, M, Sherstyuk, A, Marshall, GM, Tyrrell, V, Wegner, EA, Ekert, PG, Cowley, MJ, Mayoh, C, Trahair, TN, Crowe, P, Anazodo, A, Ziegler, DS, Nagabushan, S, Lau, LMS, Barahona, P, Wong, M, Sherstyuk, A, Marshall, GM, Tyrrell, V, Wegner, EA, Ekert, PG, Cowley, MJ, Mayoh, C, Trahair, TN, Crowe, P, Anazodo, A, and Ziegler, DS
- Abstract
The prognosis of recurrent malignant peripheral nerve sheath tumors (MPNST) is dismal, with surgical resection being the only definitive salvage therapy. Treatment with chemoradiation approaches has not significantly improved patient outcomes. Similarly, trials of therapies targeting MPNST genomic drivers have thus far been unsuccessful. Improved understanding of the molecular pathogenesis of MPNST indicates frequent activation of the mitogen-activated protein kinase (MAPK) cell signaling pathway. MEK inhibitors have shown activity in preclinical studies; however, their clinical efficacy has not been reported to date. We describe here a case of sustained complete response to MEK inhibition in an adolescent patient with a recurrent metastatic MPNST with multiple alterations in the MAPK pathway, guided by a precision oncology approach.
- Published
- 2021
27. The unexplored immune landscape of high-risk pediatric cancers.
- Author
-
Mayoh, C, Terry, RL, Wong, M, Lau, LM, Khuong-Quang, DA, Mateos, MK, Tyrrell, V, Haber, M, Ziegler, DS, Cowley, MJ, Trapani, JA, Neeson, PJ, Ekert, PG, Mayoh, C, Terry, RL, Wong, M, Lau, LM, Khuong-Quang, DA, Mateos, MK, Tyrrell, V, Haber, M, Ziegler, DS, Cowley, MJ, Trapani, JA, Neeson, PJ, and Ekert, PG
- Abstract
In adult cancer, immune signatures such as the T cell-inflamed gene expression profile (GEP) have been developed to predict which patients are likely to respond to immune checkpoint inhibitors (ICIs) beyond high tumor mutation burden (TMB) and PD-L1 expression. The GEP infers T cell infiltration and activation in the tumor microenvironment (TME) from transcriptomic data. However, it is not known whether tools such as GEP are applicable in pediatric cancer, as the TME in childhood cancers is largely unexplored and response to ICIs are rare. We have undertaken an integrated analysis of the pediatric TME using RNA-sequencing (RNA-seq) and immunohistochemistry (IHC). Our goal is to identify patients with T cell-inflamed or “hot” tumors who may benefit from ICIs. Through Australia's ZERO childhood cancer precision medicine program we performed RNA-seq on 347 high-risk pediatric cancers (estimated <30% chance of survival) and performed IHC for CD4, CD8, CD45 and PD-L1 on 112 matching samples. Using both informatic assessments and IHC as independent measures of immune infiltration, we mapped the immune landscape of the TME across a broad range of high-risk pediatric cancers. As RNA-seq is increasingly used in the analysis of patient tumors, we investigated numerous molecular correlates of immune infiltration, tailored specifically to pediatric patients. RNA-seq was used to generate the GEP and map expression profiles of immune checkpoint genes, and deconvolution algorithms were used to extract the immune cell composition for every tumor. The correlation analysis between IHC, deconvolution of cell mixture composition and GEP were assessed, including PD-L1 protein and mRNA expression. We observed significant correlation between PD-L1 protein and mRNA expression and a weak correlation of CD8+ T cells with GEP. Deconvoluted TME estimates were most tightly correlated with the presence of T cell infiltrates (CD4 and CD8) with IHC. TMB and tumor purity estimates w
- Published
- 2021
28. Chimeric Antigen Receptor T cell Therapy and the Immunosuppressive Tumor Microenvironment in Pediatric Sarcoma
- Author
-
Terry, RL, Meyran, D, Fleuren, EDG, Mayoh, C, Zhu, J, Omer, N, Ziegler, DS, Haber, M, Darcy, PK, Trapani, JA, Neeson, PJ, Ekert, PG, Terry, RL, Meyran, D, Fleuren, EDG, Mayoh, C, Zhu, J, Omer, N, Ziegler, DS, Haber, M, Darcy, PK, Trapani, JA, Neeson, PJ, and Ekert, PG
- Abstract
Sarcomas are a diverse group of bone and soft tissue tumors that account for over 10% of childhood cancers. Outcomes are particularly poor for children with refractory, relapsed, or metastatic disease. Chimeric antigen receptor T (CAR T) cells are an exciting form of adoptive cell therapy that potentially offers new hope for these children. In early trials, promising outcomes have been achieved in some pediatric patients with sarcoma. However, many children do not derive benefit despite significant expression of the targeted tumor antigen. The success of CAR T cell therapy in sarcomas and other solid tumors is limited by the immunosuppressive tumor microenvironment (TME). In this review, we provide an update of the CAR T cell therapies that are currently being tested in pediatric sarcoma clinical trials, including those targeting tumors that express HER2, NY-ESO, GD2, EGFR, GPC3, B7-H3, and MAGE-A4. We also outline promising new CAR T cells that are in pre-clinical development. Finally, we discuss strategies that are being used to overcome tumor-mediated immunosuppression in solid tumors; these strategies have the potential to improve clinical outcomes of CAR T cell therapy for children with sarcoma.
- Published
- 2021
29. Targeted Therapy of TERT-Rearranged Neuroblastoma with BET Bromodomain Inhibitor and Proteasome Inhibitor Combination Therapy
- Author
-
Chen, J, Nelson, C, Wong, M, Tee, AE, Liu, PY, La, T, Fletcher, JI, Kamili, A, Mayoh, C, Bartenhagen, C, Trahair, TN, Xu, N, Jayatilleke, N, Peng, H, Atmadibrata, B, Cheung, BB, Lan, Q, Bryan, TM, Mestdagh, P, Vandesompele, J, Combaret, V, Boeva, V, Wang, JY, Janoueix-Lerosey, I, Cowley, MJ, MacKenzie, KL, Dolnikov, A, Li, J, Polly, P, Marshall, GM, Reddel, RR, Norris, MD, Haber, M, Fischer, M, Zhang, XD, Pickett, HA, and Liu, T
- Subjects
1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
PurposeTERT gene rearrangement with transcriptional superenhancers leads to TERT overexpression and neuroblastoma. No targeted therapy is available for clinical trials in patients with TERT-rearranged neuroblastoma.Experimental designAnticancer agents exerting the best synergistic anticancer effects with BET bromodomain inhibitors were identified by screening an FDA-approved oncology drug library. The synergistic effects of the BET bromodomain inhibitor OTX015 and the proteasome inhibitor carfilzomib were examined by immunoblot and flow cytometry analysis. The anticancer efficacy of OTX015 and carfilzomib combination therapy was investigated in mice xenografted with TERT-rearranged neuroblastoma cell lines or patient-derived xenograft (PDX) tumor cells, and the role of TERT reduction in the anticancer efficacy was examined through rescue experiments in mice.ResultsThe BET bromodomain protein BRD4 promoted TERT-rearranged neuroblastoma cell proliferation through upregulating TERT expression. Screening of an approved oncology drug library identified the proteasome inhibitor carfilzomib as the agent exerting the best synergistic anticancer effects with BET bromodomain inhibitors including OTX015. OTX015 and carfilzomib synergistically reduced TERT protein expression, induced endoplasmic reticulum stress, and induced TERT-rearranged neuroblastoma cell apoptosis which was blocked by TERT overexpression and endoplasmic reticulum stress antagonists. In mice xenografted with TERT-rearranged neuroblastoma cell lines or PDX tumor cells, OTX015 and carfilzomib synergistically blocked TERT expression, induced tumor cell apoptosis, suppressed tumor progression, and improved mouse survival, which was largely reversed by forced TERT overexpression.ConclusionsOTX015 and carfilzomib combination therapy is likely to be translated into the first clinical trial of a targeted therapy in patients with TERT-rearranged neuroblastoma.
- Published
- 2020
30. Genome-wide association meta-analysis of single-nucleotide polymorphisms and symptomatic venous thromboembolism during therapy for acute lymphoblastic leukemia and lymphoma in caucasian children
- Author
-
Mateos, MK, Tulstrup, M, Quinn, MCJ, Tuckuviene, R, Marshall, GM, Gupta, R, Mayoh, C, Wolthers, BO, Barbaro, PM, Ruud, E, Sutton, R, Huttunen, P, Revesz, T, Trakymiene, SS, Barbaric, D, Tedgård, U, Giles, JE, Alvaro, F, Jonsson, OG, Mechinaud, F, Saks, K, Catchpoole, D, Kotecha, RS, Dalla-Pozza, L, Chenevix-Trench, G, Trahair, TN, Macgregor, S, Schmiegelow, K, Mateos, MK, Tulstrup, M, Quinn, MCJ, Tuckuviene, R, Marshall, GM, Gupta, R, Mayoh, C, Wolthers, BO, Barbaro, PM, Ruud, E, Sutton, R, Huttunen, P, Revesz, T, Trakymiene, SS, Barbaric, D, Tedgård, U, Giles, JE, Alvaro, F, Jonsson, OG, Mechinaud, F, Saks, K, Catchpoole, D, Kotecha, RS, Dalla-Pozza, L, Chenevix-Trench, G, Trahair, TN, Macgregor, S, and Schmiegelow, K
- Abstract
Symptomatic venous thromboembolism (VTE) occurs in five percent of children treated for acute lymphoblastic leukemia (ALL), but whether a genetic predisposition exists across different ALL treatment regimens has not been well studied. Methods: We undertook a genome-wide association study (GWAS) meta-analysis for VTE in consecutively treated children in the Nordic/Baltic acute lymphoblastic leukemia 2008 (ALL2008) cohort and the Australian Evaluation of Risk of ALL Treatment-Related Side-Effects (ERASE) cohort. A total of 92 cases and 1481 controls of European ancestry were included. Results: No SNPs reached genome-wide significance (p < 5 × 10−8) in either cohort. Among the top 34 single-nucleotide polymorphisms (SNPs) (p < 1 × 10−6), two loci had concordant effects in both cohorts: ALOX15B (rs1804772) (MAF: 1%; p = 3.95 × 10−7) that influences arachidonic acid metabolism and thus platelet aggregation, and KALRN (rs570684) (MAF: 1%; p = 4.34 × 10−7) that has been previously associated with risk of ischemic stroke, atherosclerosis, and early-onset coronary artery disease. Conclusion: This represents the largest GWAS meta-analysis conducted to date associating SNPs to VTE in children and adolescents treated on childhood ALL protocols. Validation of these findings is needed and may then lead to patient stratification for VTE preventive interventions. As VTE hemostasis involves multiple pathways, a more powerful GWAS is needed to detect combination of variants associated with VTE.
- Published
- 2020
31. Accelerating development of high-risk neuroblastoma patient-derived xenograft models for preclinical testing and personalised therapy
- Author
-
Kamili, A, Gifford, AJ, Li, N, Mayoh, C, Chow, S-O, Failes, TW, Eden, GL, Cadiz, R, Xie, J, Lukeis, RE, Norris, MD, Haber, M, McCowage, GB, Arndt, GM, Trahair, TN, Fletcher, JI, Kamili, A, Gifford, AJ, Li, N, Mayoh, C, Chow, S-O, Failes, TW, Eden, GL, Cadiz, R, Xie, J, Lukeis, RE, Norris, MD, Haber, M, McCowage, GB, Arndt, GM, Trahair, TN, and Fletcher, JI
- Published
- 2020
32. Whole genome, transcriptome and methylome profiling enhances actionable target discovery in high-risk pediatric cancer
- Author
-
Wong, M, Mayoh, C, Lau, LMS, Khuong-Quang, DA, Pinese, M, Kumar, A, Barahona, P, Wilkie, EE, Sullivan, P, Bowen-James, R, Syed, M, Martincorena, I, Abascal, F, Sherstyuk, A, Bolanos, NA, Baber, J, Priestley, P, Dolman, MEM, Fleuren, EDG, Gauthier, ME, Mould, EVA, Gayevskiy, V, Gifford, AJ, Grebert-Wade, D, Strong, PA, Manouvrier, E, Warby, M, Thomas, DM, Kirk, J, Tucker, K, O’Brien, T, Alvaro, F, McCowage, GB, Dalla-Pozza, L, Gottardo, NG, Tapp, H, Wood, P, Khaw, SL, Hansford, JR, Moore, AS, Norris, MD, Trahair, TN, Lock, RB, Tyrrell, V, Haber, M, Marshall, GM, Ziegler, DS, Ekert, PG, Cowley, MJ, Wong, M, Mayoh, C, Lau, LMS, Khuong-Quang, DA, Pinese, M, Kumar, A, Barahona, P, Wilkie, EE, Sullivan, P, Bowen-James, R, Syed, M, Martincorena, I, Abascal, F, Sherstyuk, A, Bolanos, NA, Baber, J, Priestley, P, Dolman, MEM, Fleuren, EDG, Gauthier, ME, Mould, EVA, Gayevskiy, V, Gifford, AJ, Grebert-Wade, D, Strong, PA, Manouvrier, E, Warby, M, Thomas, DM, Kirk, J, Tucker, K, O’Brien, T, Alvaro, F, McCowage, GB, Dalla-Pozza, L, Gottardo, NG, Tapp, H, Wood, P, Khaw, SL, Hansford, JR, Moore, AS, Norris, MD, Trahair, TN, Lock, RB, Tyrrell, V, Haber, M, Marshall, GM, Ziegler, DS, Ekert, PG, and Cowley, MJ
- Abstract
The Zero Childhood Cancer Program is a precision medicine program to benefit children with poor-outcome, rare, relapsed or refractory cancer. Using tumor and germline whole genome sequencing (WGS) and RNA sequencing (RNAseq) across 252 tumors from high-risk pediatric patients with cancer, we identified 968 reportable molecular aberrations (39.9% in WGS and RNAseq, 35.1% in WGS only and 25.0% in RNAseq only). Of these patients, 93.7% had at least one germline or somatic aberration, 71.4% had therapeutic targets and 5.2% had a change in diagnosis. WGS identified pathogenic cancer-predisposing variants in 16.2% of patients. In 76 central nervous system tumors, methylome analysis confirmed diagnosis in 71.1% of patients and contributed to a change of diagnosis in two patients (2.6%). To date, 43 patients have received a recommended therapy, 38 of whom could be evaluated, with 31% showing objective evidence of clinical benefit. Comprehensive molecular profiling resolved the molecular basis of virtually all high-risk cancers, leading to clinical benefit in some patients.
- Published
- 2020
33. Examining treatment responses of diagnostic marrow in murine xenografts to predict relapse in children with acute lymphoblastic leukaemia
- Author
-
Alruwetei, AM, Bendak, K, Yadav, BD, Carol, H, Evans, K, Mayoh, C, Sutton, R, Marshall, GM, Lock, RB, Alruwetei, AM, Bendak, K, Yadav, BD, Carol, H, Evans, K, Mayoh, C, Sutton, R, Marshall, GM, and Lock, RB
- Abstract
Background: While current chemotherapy has increased cure rates for children with acute lymphoblastic leukaemia (ALL), the largest number of relapsing patients are still stratified as medium risk (MR) at diagnosis (50–60%). This highlights an opportunity to develop improved relapse-prediction models for MR patients. We hypothesised that bone marrow from MR patients who eventually relapsed would regrow faster in a patient-derived xenograft (PDX) model after induction chemotherapy than samples from patients in long-term remission. Methods: Diagnostic bone marrow aspirates from 30 paediatric MR-ALL patients (19 who relapsed, 11 who experienced remission) were inoculated into immune-deficient (NSG) mice and subsequently treated with either control or an induction-type regimen of vincristine, dexamethasone, and L-asparaginase (VXL). Engraftment was monitored by enumeration of the proportion of human CD45+ cells (%huCD45+) in the murine peripheral blood, and events were defined a priori as the time to reach 1% huCD45+, 25% huCD45+ (TT25%) or clinical manifestations of leukaemia (TTL). Results: The TT25% value significantly predicted MR patient relapse. Mutational profiles of PDXs matched their tumours of origin, with a clonal shift towards relapse observed in one set of VXL-treated PDXs. Conclusions: In conclusion, establishing PDXs at diagnosis and subsequently applying chemotherapy has the potential to improve relapse prediction in paediatric MR-ALL.
- Published
- 2020
34. A novel orthotopic patient-derived xenograft model of radiation-induced glioma following medulloblastoma
- Author
-
Whitehouse, JP, Howlett, M, Hii, H, Mayoh, C, Wong, M, Barahona, P, Ajuyah, P, White, CL, Buntine, MK, Dyke, JM, Lee, S, Valvi, S, Stanley, J, Andradas, C, Carline, B, Kuchibhotla, M, Ekert, PG, Cowley, MJ, Gottardo, NG, Endersby, R, Whitehouse, JP, Howlett, M, Hii, H, Mayoh, C, Wong, M, Barahona, P, Ajuyah, P, White, CL, Buntine, MK, Dyke, JM, Lee, S, Valvi, S, Stanley, J, Andradas, C, Carline, B, Kuchibhotla, M, Ekert, PG, Cowley, MJ, Gottardo, NG, and Endersby, R
- Abstract
Radiation-induced glioma (RIG) is a highly aggressive brain cancer arising as a consequence of radiation therapy. We report a case of RIG that arose in the brain stem following treatment for paediatric medulloblastoma, and the development and characterisation of a matched orthotopic patient-derived xenograft (PDX) model (TK-RIG915). Patient and PDX tumours were analysed using DNA methylation profiling, whole genome sequencing (WGS) and RNA sequencing. While initially thought to be a diffuse intrinsic pontine glioma (DIPG) based on disease location, results from methylation profiling and WGS were not consistent with this diagnosis. Furthermore, clustering analyses based on RNA expression suggested the tumours were distinct from primary DIPG. Additional gene expression analysis demonstrated concordance with a published RIG expression profile. Multiple genetic alterations that enhance PI3K/AKT and Ras/Raf/MEK/ERK signalling were discovered in TK-RIG915 including an activating mutation in PIK3CA, upregulation of PDGFRA and AKT2, inactivating mutations in NF1, and a gain-of-function mutation in PTPN11. Additionally, deletion of CDKN2A/B, increased IDH1 expression, and decreased ARID1A expression were observed. Detection of phosphorylated S6, 4EBP1 and ERK via immunohistochemistry confirmed PI3K pathway and ERK activation. Here, we report one of the first PDX models for RIG, which recapitulates the patient disease and is molecularly distinct from primary brain stem glioma. Genetic interrogation of this model has enabled the identification of potential therapeutic vulnerabilities in this currently incurable disease.
- Published
- 2020
35. Recurrent SPECC1L–NTRK fusions in pediatric sarcoma and brain tumors
- Author
-
Khuong-Quang, DA, Brown, LM, Wong, M, Mayoh, C, Sexton-Oates, A, Kumar, A, Pinese, M, Nagabushan, S, Lau, L, Ludlow, LE, Gifford, AJ, Rodriguez, M, Desai, J, Fox, SB, Haber, M, Ziegler, DS, Hansford, JR, Marshall, GM, Cowley, MJ, Ekert, PG, Khuong-Quang, DA, Brown, LM, Wong, M, Mayoh, C, Sexton-Oates, A, Kumar, A, Pinese, M, Nagabushan, S, Lau, L, Ludlow, LE, Gifford, AJ, Rodriguez, M, Desai, J, Fox, SB, Haber, M, Ziegler, DS, Hansford, JR, Marshall, GM, Cowley, MJ, and Ekert, PG
- Abstract
The identification of rearrangements driving expression of neurotrophic receptor tyrosine kinase (NTRK) family kinases in tumors has become critically important because of the availability of effective, specific inhibitor drugs. Whole-genome sequencing (WGS) combined with RNA sequencing (RNA-seq) can identify novel and recurrent expressed fusions. Here we describe three SPECC1L–NTRK fusions identified in two pediatric central nervous system cancers and an extracranial solid tumor using WGS and RNA-seq. These fusions arose either through a simple balanced rearrangement or in the context of a complex chromoplexy event. We cloned the SPECC1L–NTRK2 fusion directly from a patient sample and showed that enforced expression of this fusion is sufficient to promote cytokine-independent survival and proliferation. Cells transformed by SPECC1L–NTRK2 expression are sensitive to a TRK inhibitor drug. We report here that SPECC1L–NTRK fusions can arise in a range of pediatric cancers. Although WGS and RNA-seq are not required to detect NTRK fusions, these techniques may be of benefit when NTRK fusions are not suspected on clinical grounds or not identified by other methods.
- Published
- 2020
36. Reversal of glucocorticoid resistance in paediatric acute lymphoblastic leukaemia is dependent on restoring BIM expression
- Author
-
Toscan, CE, Jing, D, Mayoh, C, Lock, RB, Toscan, CE, Jing, D, Mayoh, C, and Lock, RB
- Abstract
Background: Acute lymphoblastic leukaemia (ALL) is the most common paediatric malignancy. Glucocorticoids form a critical component of chemotherapy regimens and resistance to glucocorticoid therapy is predictive of poor outcome. We have previously shown that glucocorticoid resistance is associated with upregulation of the oncogene C-MYC and failure to induce the proapoptotic gene BIM. Methods: A high-throughput screening (HTS) campaign was carried out to identify glucocorticoid sensitisers against an ALL xenograft derived from a glucocorticoid-resistant paediatric patient. Gene expression analysis was carried out using Illumina microarrays. Efficacy, messenger RNA and protein analysis were carried out by Resazurin assay, reverse transcription-PCR and immunoblotting, respectively. Results: A novel glucocorticoid sensitiser, 2-((4,5-dihydro-1H-imidazol-2-yl)thio)-N-isopropyl-N-phenylacetamide (GCS-3), was identified from the HTS campaign. The sensitising effect was specific to glucocorticoids and synergy was observed in a range of dexamethasone-resistant and dexamethasone-sensitive xenografts representative of B-ALL, T-ALL and Philadelphia chromosome-positive ALL. GCS-3 in combination with dexamethasone downregulated C-MYC and significantly upregulated BIM expression in a glucocorticoid-resistant ALL xenograft. The GCS-3/dexamethasone combination significantly increased binding of the glucocorticoid receptor to a novel BIM enhancer, which is associated with glucocorticoid sensitivity. Conclusions: This study describes the potential of the novel glucocorticoid sensitiser, GCS-3, as a biological tool to interrogate glucocorticoid action and resistance.
- Published
- 2020
37. Targeting TSLP-induced tyrosine kinase signaling pathways in CRLF2-Rearranged Ph-like ALL
- Author
-
Sia, KCS, Zhong, L, Mayoh, C, Norris, MD, Haber, M, Marshall, GM, Raftery, MJ, Lock, RB, Sia, KCS, Zhong, L, Mayoh, C, Norris, MD, Haber, M, Marshall, GM, Raftery, MJ, and Lock, RB
- Abstract
Philadelphia (Ph)-like acute lymphoblastic leukemia (ALL) is characterized by aberrant activation of signaling pathways and high risk of relapse. Approximately 50% of Ph-like ALL cases overexpress cytokine receptor-like factor 2 (CRLF2) associated with gene rearrangement. Activated by its ligand thymic stromal lymphopoietin (TSLP), CRLF2 signaling is critical for the development, proliferation, and survival of normal lymphocytes. To examine activation of tyrosine kinases regulated by TSLP/CRLF2, phosphotyrosine (P-Tyr) profiling coupled with stable isotope labeling of amino acids in cell culture (SILAC) was conducted using two CRLF2-rearranged (CRLF2r) Ph-like ALL cell lines stimulated with TSLP. As a result, increased P-Tyr was detected in previously reported TSLP-activated tyrosine kinases and substrates, including JAK1, JAK2, STAT5, and ERK1/2. Interestingly, TSLP also increased P-Tyr of insulin growth factor 1 receptor (IGF1R) and fibroblast growth factor receptor 1 (FGFR1), both of which can be targeted with small-molecule inhibitors. Fixed-ratio combination cytotoxicity assays using the tyrosine kinase inhibitors BMS-754807 and ponatinib that target IGF1R and FGFR1, respectively, revealed strong synergy against both cell line and patient-derived xenograft (PDX) models of CRLF2r Ph-like ALL. Further analyses also indicated off-target effects of ponatinib in the synergy, and novel association of the Ras-associated protein-1 (Rap1) signaling pathway with TSLP signaling in CRLF2r Ph-like ALL. When tested in vivo, the BMS-754807/ponatinib combination exerted minimal efficacy against 2 Ph-like ALL PDXs, associated with low achievable plasma drug concentrations. Although this study identified potential new targets in CRLF2r Ph-like ALL, it also highlights that in vivo validation of synergistic drug interactions is essential. Implication: Quantitative phosphotyrosine profiling identified potential therapeutic targets for high-risk CRLF2-rearranged Ph-like ALL.
- Published
- 2020
38. Intratumoral copper modulates PD-L1 expression and influences tumor immune evasion
- Author
-
Voli, F, Valli, E, Lerra, L, Kimpton, K, Saletta, F, Giorgi, FM, Mercatelli, D, Rouaen, JRC, Shen, S, Murray, JE, Ahmed-Cox, A, Cirillo, G, Mayoh, C, Beavis, PA, Haber, M, Trapani, JA, Kavallaris, M, Vittorio, O, Voli, F, Valli, E, Lerra, L, Kimpton, K, Saletta, F, Giorgi, FM, Mercatelli, D, Rouaen, JRC, Shen, S, Murray, JE, Ahmed-Cox, A, Cirillo, G, Mayoh, C, Beavis, PA, Haber, M, Trapani, JA, Kavallaris, M, and Vittorio, O
- Abstract
Therapeutic checkpoint antibodies blocking programmed death receptor 1/programmed death ligand 1 (PD-L1) signaling have radically improved clinical outcomes in cancer. However, the regulation of PD-L1 expression on tumor cells is still poorly understood. Here we show that intratumoral copper levels influence PD-L1 expression in cancer cells. Deep analysis of the The Cancer Genome Atlas database and tissue microarrays showed strong correlation between the major copper influx transporter copper transporter 1 (CTR-1) and PD-L1 expression across many cancers but not in corresponding normal tissues. Copper supplementation enhanced PD-L1 expression at mRNA and protein levels in cancer cells and RNA sequencing revealed that copper regulates key signaling pathways mediating PD-L1-driven cancer immune evasion. Conversely, copper chelators inhibited phosphorylation of STAT3 and EGFR and promoted ubiquitin-mediated degradation of PD-L1. Copper-chelating drugs also significantly increased the number of tumor-infiltrating CD8þ T and natural killer cells, slowed tumor growth, and improved mouse survival. Overall, this study reveals an important role for copper in regulating PD-L1 and suggests that anticancer immunotherapy might be enhanced by pharmacologically reducing intratumor copper levels.
- Published
- 2020
39. Effective targeting of NAMPT in patient-derived xenograft models of high-risk pediatric acute lymphoblastic leukemia
- Author
-
Somers, K, Evans, K, Cheung, L, Karsa, M, Pritchard, T, Kosciolek, A, Bongers, A, El-Ayoubi, A, Forgham, H, Middlemiss, S, Mayoh, C, Jones, L, Gupta, M, Kees, UR, Chernova, O, Korotchkina, L, Gudkov, AV, Erickson, SW, Teicher, B, Smith, MA, Norris, MD, Haber, M, Lock, RB, Henderson, MJ, Somers, K, Evans, K, Cheung, L, Karsa, M, Pritchard, T, Kosciolek, A, Bongers, A, El-Ayoubi, A, Forgham, H, Middlemiss, S, Mayoh, C, Jones, L, Gupta, M, Kees, UR, Chernova, O, Korotchkina, L, Gudkov, AV, Erickson, SW, Teicher, B, Smith, MA, Norris, MD, Haber, M, Lock, RB, and Henderson, MJ
- Abstract
The prognosis for children diagnosed with high-risk acute lymphoblastic leukemia (ALL) remains suboptimal, and more potent and less toxic treatments are urgently needed. We investigated the efficacy of a novel nicotinamide phosphoribosyltransferase inhibitor, OT-82, against a panel of patient-derived xenografts (PDXs) established from high-risk and poor outcome pediatric ALL cases. OT-82 was well-tolerated and demonstrated impressive single agent in vivo efficacy, achieving significant leukemia growth delay in 95% (20/21) and disease regression in 86% (18/21) of PDXs. In addition, OT-82 enhanced the efficacy of the established drugs cytarabine and dasatinib and, as a single agent, showed similar efficacy as an induction-type regimen combining three drugs used to treat pediatric ALL. OT-82 exerted its antileukemic action by depleting NAD+ and ATP, inhibiting the NAD+-requiring DNA damage repair enzyme PARP-1, increasing mitochondrial ROS levels and inducing DNA damage, culminating in apoptosis induction. OT-82 sensitivity was associated with the occurrence of mutations in major DNA damage response genes, while OT-82 resistance was characterized by high expression levels of CD38. In conclusion, our study provides evidence that OT-82, as a single agent, and in combination with established drugs, is a promising new therapeutic strategy for a broad spectrum of high-risk pediatric ALL for which improved therapies are urgently needed.
- Published
- 2020
40. Efficacy of combined CDK9/BET inhibition in preclinical models of MLL-rearranged acute leukemia
- Author
-
McCalmont, H, Li, KL, Jones, L, Toubia, J, Bray, SC, Casolari, DA, Mayoh, C, Samaraweera, SE, Lewis, ID, Prinjha, RK, Smithers, N, Wang, S, Lock, RB, D’Andrea, RJ, McCalmont, H, Li, KL, Jones, L, Toubia, J, Bray, SC, Casolari, DA, Mayoh, C, Samaraweera, SE, Lewis, ID, Prinjha, RK, Smithers, N, Wang, S, Lock, RB, and D’Andrea, RJ
- Abstract
Cyclin-dependent kinase 9 and bromodomain and extraterminal inhibitors are synergistic in MLL-rearranged leukemia.Multiple AML driver genes are downregulated by the combined therapy suggesting broad applicability for this subtype.
- Published
- 2020
41. Recurrent SPECC1L-NTRK fusions in pediatric sarcoma and brain tumors
- Author
-
Khuong-Quang, D-A, Brown, LM, Wong, M, Mayoh, C, Sexton-Oates, A, Kumar, A, Pinese, M, Nagabushan, S, Lau, L, Ludlow, LE, Gifford, AJ, Rodriguez, M, Desai, J, Fox, SB, Haber, M, Ziegler, DS, Hansford, JR, Marshall, GM, Cowley, MJ, Ekert, PG, Khuong-Quang, D-A, Brown, LM, Wong, M, Mayoh, C, Sexton-Oates, A, Kumar, A, Pinese, M, Nagabushan, S, Lau, L, Ludlow, LE, Gifford, AJ, Rodriguez, M, Desai, J, Fox, SB, Haber, M, Ziegler, DS, Hansford, JR, Marshall, GM, Cowley, MJ, and Ekert, PG
- Abstract
The identification of rearrangements driving expression of neurotrophic receptor tyrosine kinase (NTRK) family kinases in tumors has become critically important because of the availability of effective, specific inhibitor drugs. Whole-genome sequencing (WGS) combined with RNA sequencing (RNA-seq) can identify novel and recurrent expressed fusions. Here we describe three SPECC1L-NTRK fusions identified in two pediatric central nervous system cancers and an extracranial solid tumor using WGS and RNA-seq. These fusions arose either through a simple balanced rearrangement or in the context of a complex chromoplexy event. We cloned the SPECC1L-NTRK2 fusion directly from a patient sample and showed that enforced expression of this fusion is sufficient to promote cytokine-independent survival and proliferation. Cells transformed by SPECC1L-NTRK2 expression are sensitive to a TRK inhibitor drug. We report here that SPECC1L-NTRK fusions can arise in a range of pediatric cancers. Although WGS and RNA-seq are not required to detect NTRK fusions, these techniques may be of benefit when NTRK fusions are not suspected on clinical grounds or not identified by other methods.
- Published
- 2020
42. PRECISION MEDICINE FOR PAEDIATRIC HIGH-GRADE DIFFUSE MIDLINE GLIOMAS - RESULTS FROM THE ZERO CHILDHOOD CANCER COMPREHENSIVE PRECISION MEDICINE PROGRAM
- Author
-
Dong-Anh, K-Q, Nagabushan, S, Manoharan, N, Arndt, G, Barahona, P, Cowley, MJ, Ekert, PG, Failes, T, Bolanos, NF, Gauthier, M, Gifford, AJ, Haber, M, Kumar, A, Lock, RB, Marshall, GM, Mayoh, C, Mould, E, Norris, MD, Gopalakrishnan, A, Omer, N, Trebilcock, P, Trahair, TN, Tsoli, M, Tucker, K, Wong, M, Tyrrell, V, Lau, L, Ziegler, DS, Dong-Anh, K-Q, Nagabushan, S, Manoharan, N, Arndt, G, Barahona, P, Cowley, MJ, Ekert, PG, Failes, T, Bolanos, NF, Gauthier, M, Gifford, AJ, Haber, M, Kumar, A, Lock, RB, Marshall, GM, Mayoh, C, Mould, E, Norris, MD, Gopalakrishnan, A, Omer, N, Trebilcock, P, Trahair, TN, Tsoli, M, Tucker, K, Wong, M, Tyrrell, V, Lau, L, and Ziegler, DS
- Abstract
The Australian Zero Childhood Cancer (ZERO) program aims to assess the feasibility of a comprehensive precision medicine approach to improve outcomes for patients with an expected survival <30%. ZERO combines molecular profiling (whole genome sequencing, whole transcriptome sequencing, DNA methylation profiling) with in vitro high-throughput drug screening (HTS) and patient-derived xenograft drug efficacy testing. We report on the cohort of patients with midline high-grade glioma (HGG), including H3-K27M DMG, enrolled on the pilot study (TARGET) and on the ongoing ZERO clinical trial (PRISM). We identified 48 patients with midline HGG. Fresh or cryopreserved samples were submitted in 37 cases and cell culture was attempted in 30/37 cases with 45% success rate. The most commonly mutated genes/pathways identified by molecular profiling include H3-K27M mutations, DNA repair pathway, and PI3K/mTOR pathway. Two targetable fusions (NTRK and FGFR1) were reported. Five patients with germline alterations were identified. Thirty-five (72%) patients received a therapeutic recommendation from the ZERO molecular tumour board and the main recommended therapies were mTOR inhibitors, PARP inhibitors or tyrosine kinase inhibitors. HTS added evidence for the recommended therapy (n=3) or identified novel potential therapy (n=1). Out of the 35 patients, 16 received a recommended drug. Response to treatment was complete response for five months (n=1), partial response for nine months (n=1), stable disease (n=4), and progressive disease (n=10). These results highlight the feasibility of the ZERO platform and the value of fresh biopsy, necessary for pre-clinical drug testing. Targetable alterations were identified leading to clinical benefit in six patients.
- Published
- 2020
43. NEW GENES AND DISEASES / NGS & RELATED TECHNIQUES
- Author
-
Sullivan, P., primary, Mayoh, C., additional, Wong-Erasmus, M., additional, Gayevskiy, V., additional, Beecroft, S., additional, Pinese, M., additional, Oates, E., additional, and Cowley, M., additional
- Published
- 2020
- Full Text
- View/download PDF
44. Heterozygous loss of keratinocyte TRIM16 expression increases melanocytic cell lesions and lymph node metastasis
- Author
-
Sutton, SK, Cheung, BB, Massudi, H, Tan, O, Koach, J, Mayoh, C, Carter, DR, and Marshall, GM
- Subjects
Keratinocytes ,Mice, Knockout ,Skin Neoplasms ,Loss of Heterozygosity ,Gene Expression Regulation, Neoplastic ,Mice ,Cell Transformation, Neoplastic ,Lymphatic Metastasis ,Carcinoma, Squamous Cell ,Melanocytes ,Animals ,Oncology & Carcinogenesis ,Lymph Nodes ,Carrier Proteins ,neoplasms ,Melanoma ,Cells, Cultured ,Skin - Abstract
© 2019, The Author(s). Purpose: The tripartite motif (TRIM)16 acts as a tumour suppressor in both squamous cell carcinoma (SCC) and melanoma. TRIM16 is known to be secreted by keratinocytes, but no studies have been reported yet to assess the relationship between TRIM16 keratinocyte expression and melanoma development. Methods: To study the role of TRIM16 in skin cancer development, we developed a keratinocyte TRIM16-specific knockout mouse model, and used the classical two-stage skin carcinogenesis challenge method, to assess the loss of keratinocyte TRIM16 on both papilloma, SCC and melanoma development in the skin after topical carcinogen treatment. Results: Heterozygous, but not homozygous, TRIM16 knockout mice exhibited an accelerated development of skin papillomas and melanomas, larger melanoma lesions and an increased potential for lymph node metastasis. Conclusion: This study provides the first evidence that keratinocyte loss of the putative melanoma tumour suppressor protein, TRIM16, enhances melanomagenesis. Our data also suggest that TRIM16 expression in keratinocytes is involved in cross talk between keratinocytes and melanocytes, and has a role in melanoma tumorigenesis.
- Published
- 2019
45. A novel small molecule that kills a subset of MLL-rearranged leukemia cells by inducing mitochondrial dysfunction
- Author
-
Somers, K, Wen, VW, Middlemiss, SMC, Osborne, B, Forgham, H, Jung, MS, Karsa, M, Clifton, M, Bongers, A, Gao, J, Mayoh, C, Raoufi-Rad, N, Kusnadi, EP, Hannan, KM, Scott, DA, Kwek, A, Liu, B, Flemming, C, Chudakova, DA, Pandher, R, Failes, TW, Lim, J, Angeli, A, Osterman, AL, Imamura, T, Kees, UR, Supuran, CT, Pearson, RB, Hannan, RD, Davis, TP, McCarroll, J, Kavallaris, M, Turner, N, Gudkov, AV, Haber, M, Norris, MD, Henderson, MJ, Somers, K, Wen, VW, Middlemiss, SMC, Osborne, B, Forgham, H, Jung, MS, Karsa, M, Clifton, M, Bongers, A, Gao, J, Mayoh, C, Raoufi-Rad, N, Kusnadi, EP, Hannan, KM, Scott, DA, Kwek, A, Liu, B, Flemming, C, Chudakova, DA, Pandher, R, Failes, TW, Lim, J, Angeli, A, Osterman, AL, Imamura, T, Kees, UR, Supuran, CT, Pearson, RB, Hannan, RD, Davis, TP, McCarroll, J, Kavallaris, M, Turner, N, Gudkov, AV, Haber, M, Norris, MD, and Henderson, MJ
- Abstract
Survival rates for pediatric patients suffering from mixed lineage leukemia (MLL)-rearranged leukemia remain below 50% and more targeted, less toxic therapies are urgently needed. A screening method optimized to discover cytotoxic compounds selective for MLL-rearranged leukemia identified CCI-006 as a novel inhibitor of MLL-rearranged and CALM-AF10 translocated leukemias that share common leukemogenic pathways. CCI-006 inhibited mitochondrial respiration and induced mitochondrial membrane depolarization and apoptosis in a subset (7/11, 64%) of MLL-rearranged leukemia cell lines within a few hours of treatment. The unresponsive MLL-rearranged leukemia cells did not undergo mitochondrial membrane depolarization or apoptosis despite a similar attenuation of mitochondrial respiration by the compound. In comparison to the sensitive cells, the unresponsive MLL-rearranged leukemia cells were characterized by a more glycolytic metabolic phenotype, exemplified by a more pronounced sensitivity to glycolysis inhibitors and elevated HIF1α expression. Silencing of HIF1α expression sensitized an intrinsically unresponsive MLL-rearranged leukemia cell to CCI-006, indicating that this pathway plays a role in determining sensitivity to the compound. In addition, unresponsive MLL-rearranged leukemia cells expressed increased levels of MEIS1, an important leukemogenic MLL target gene that plays a role in regulating metabolic phenotype through HIF1α. MEIS1 expression was also variable in a pediatric MLL-rearranged ALL patient dataset, highlighting the existence of a previously undescribed metabolic variability in MLL-rearranged leukemia that may contribute to the heterogeneity of the disease. This study thus identified a novel small molecule that rapidly kills MLL-rearranged leukemia cells by targeting a metabolic vulnerability in a subset of low HIF1α/low MEIS1-expressing MLL-rearranged leukemia cells.
- Published
- 2019
46. Inhibition of polyamine synthesis and uptake reduces tumor progression and prolongs survival in mouse models of neuroblastoma
- Author
-
Gamble, LD, Purgato, S, Murray, J, Xiao, L, Yu, DMT, Hanssen, KM, Giorgi, FM, Carter, DR, Gifford, AJ, Valli, E, Milazzo, G, Kamili, A, Mayoh, C, Liu, B, Eden, G, Sarraf, S, Allan, S, Giacomo, SD, Flemming, CL, Russell, AJ, Cheung, BB, Oberthuer, A, London, WB, Fischer, M, Trahair, TN, Fletcher, JI, Marshall, GM, Ziegler, DS, Hogarty, MD, Burns, MR, Perini, G, Norris, MD, Haber, M, Gamble, LD, Purgato, S, Murray, J, Xiao, L, Yu, DMT, Hanssen, KM, Giorgi, FM, Carter, DR, Gifford, AJ, Valli, E, Milazzo, G, Kamili, A, Mayoh, C, Liu, B, Eden, G, Sarraf, S, Allan, S, Giacomo, SD, Flemming, CL, Russell, AJ, Cheung, BB, Oberthuer, A, London, WB, Fischer, M, Trahair, TN, Fletcher, JI, Marshall, GM, Ziegler, DS, Hogarty, MD, Burns, MR, Perini, G, Norris, MD, and Haber, M
- Abstract
Amplification of the MYCN oncogene is associated with an aggressive phenotype and poor outcome in childhood neuroblastoma. Polyamines are highly regulated essential cations that are frequently elevated in cancer cells, and the rate-limiting enzyme in polyamine synthesis, ornithine decarboxylase 1 (ODC1), is a direct transcriptional target of MYCN. Treatment of neuroblastoma cells with the ODC1 inhibitor difluoromethylornithine (DFMO), although a promising therapeutic strategy, is only partially effective at impeding neuroblastoma cell growth due to activation of compensatory mechanisms resulting in increased polyamine uptake from the surrounding microenvironment. In this study, we identified solute carrier family 3 member 2 (SLC3A2) as the key transporter involved in polyamine uptake in neuroblastoma. Knockdown of SLC3A2 in neuroblastoma cells reduced the uptake of the radiolabeled polyamine spermidine, and DFMO treatment increased SLC3A2 protein. In addition, MYCN directly increased polyamine synthesis and promoted neuroblastoma cell proliferation by regulating SLC3A2 and other regulatory components of the polyamine pathway. Inhibiting polyamine uptake with the small-molecule drug AMXT 1501, in combination with DFMO, prevented or delayed tumor development in neuroblastoma-prone mice and extended survival in rodent models of established tumors. Our findings suggest that combining AMXT 1501 and DFMO with standard chemotherapy might be an effective strategy for treating neuroblastoma.
- Published
- 2019
47. International experience in the development of patient-derived xenograft models of diffuse intrinsic pontine glioma
- Author
-
Tsoli, M, Shen, H, Mayoh, C, Franshaw, L, Ehteda, A, Upton, D, Carvalho, D, Vinci, M, Meel, MH, van Vuurden, D, Plessier, A, Castel, D, Drissi, R, Farrell, M, Cryan, J, Crimmins, D, Caird, J, Pears, J, Francis, S, Ludlow, LEA, Carai, A, Mastronuzzi, A, Liu, B, Hansford, J, Gottardo, N, Hassall, T, Kirby, M, Fouladi, M, Hawkins, C, Monje, M, Grill, J, Jones, C, Hulleman, E, Ziegler, DS, Tsoli, M, Shen, H, Mayoh, C, Franshaw, L, Ehteda, A, Upton, D, Carvalho, D, Vinci, M, Meel, MH, van Vuurden, D, Plessier, A, Castel, D, Drissi, R, Farrell, M, Cryan, J, Crimmins, D, Caird, J, Pears, J, Francis, S, Ludlow, LEA, Carai, A, Mastronuzzi, A, Liu, B, Hansford, J, Gottardo, N, Hassall, T, Kirby, M, Fouladi, M, Hawkins, C, Monje, M, Grill, J, Jones, C, Hulleman, E, and Ziegler, DS
- Abstract
Purpose: Diffuse intrinsic pontine glioma is the most aggressive form of high grade glioma in children with no effective therapies. There have been no improvements in survival in part due poor understanding of underlying biology, and lack of representative in vitro and in vivo models. Recently, it has been found feasible to use both biopsy and autopsy tumors to generate cultures and xenograft models. Methods: To further model development, we evaluated the collective international experience from 8 collaborating centers to develop DIPG pre-clinical models from patient-derived autopsies and biopsies. Univariate and multivariate analysis was performed to determine key factors associated with the success of in vitro and in vivo PDX development. Results: In vitro cultures were successfully established from 57% of samples (84.2% of biopsies and 38.2% of autopsies). Samples transferred in DMEM media were more likely to establish successful culture than those transported in Hibernate A. In vitro cultures were more successful from biopsies (84.2%) compared with autopsies (38.2%) and as monolayer on laminin-coated plates than as neurospheres. Primary cultures successfully established from autopsy samples were more likely to engraft in animal models than cultures established from biopsies (86.7% vs. 47.4%). Collectively, tumor engraftment was more successful when DIPG samples were directly implanted in mice (68%), rather than after culturing (40.7%). Conclusion: This multi-center study provides valuable information on the success rate of establishing patient-derived pre-clinical models of DIPG. The results can lead to further optimization of DIPG model development and ultimately assist in the investigation of new therapies for this aggressive pediatric brain tumor.
- Published
- 2019
48. Reversal of glucocorticoid resistance in pediatric acute lymphoblastic leukemia is dependent on restoring BIM expression
- Author
-
Lock, RB, Toscan, CE, Jing, D, Mayoh, C, Lock, RB, Toscan, CE, Jing, D, and Mayoh, C
- Published
- 2019
49. Drugging MYCN oncogenic signaling through the MYCN-PA2G4 binding interface
- Author
-
Koach, J, Holien, JK, Massudi, H, Carter, DR, Ciampa, OC, Herath, M, Lim, T, Seneviratne, JA, Milazzo, G, Murray, JE, McCarroll, JA, Liu, B, Mayoh, C, Keenan, B, Stevenson, BW, Gorman, MA, Bell, JL, Doughty, L, Hüttelmaier, S, Oberthuer, A, Fischer, M, Gifford, AJ, Liu, T, Zhang, X, Zhu, S, Gustafson, WC, Haber, M, Norris, MD, Fletcher, JI, Perini, G, Parker, MW, Cheung, BB, Marshall, GM, Koach, J, Holien, JK, Massudi, H, Carter, DR, Ciampa, OC, Herath, M, Lim, T, Seneviratne, JA, Milazzo, G, Murray, JE, McCarroll, JA, Liu, B, Mayoh, C, Keenan, B, Stevenson, BW, Gorman, MA, Bell, JL, Doughty, L, Hüttelmaier, S, Oberthuer, A, Fischer, M, Gifford, AJ, Liu, T, Zhang, X, Zhu, S, Gustafson, WC, Haber, M, Norris, MD, Fletcher, JI, Perini, G, Parker, MW, Cheung, BB, and Marshall, GM
- Abstract
MYCN is a major driver for the childhood cancer, neuroblastoma, however, there are no inhibitors of this target. Enhanced MYCN protein stability is a key component of MYCN oncogenesis and is maintained by multiple feedforward expression loops involving MYCN transactivation target genes. Here, we reveal the oncogenic role of a novel MYCN target and binding protein, proliferationassociated 2AG4 (PA2G4). Chromatin immunoprecipitation studies demonstrated that MYCN occupies the PA2G4 gene promoter, stimulating transcription. Direct binding of PA2G4 to MYCN protein blocked proteolysis of MYCN and enhanced colony formation in a MYCNdependent manner. Using molecular modeling, surface plasmon resonance, and mutagenesis studies, we mapped the MYCN-PA2G4 interaction site to a 14 amino acid MYCN sequence and a surface crevice of PA2G4. Competitive chemical inhibition of the MYCN-PA2G4 protein-protein interface had potent inhibitory effects on neuroblastoma tumorigenesis in vivo. Treated tumors showed reduced levels of both MYCN and PA2G4. Our findings demonstrate a critical role for PA2G4 as a cofactor in MYCN-driven neuroblastoma and highlight competitive inhibition of the PA2G4-MYCN protein binding as a novel therapeutic strategy in the disease.
- Published
- 2019
50. The long noncoding RNA lncNB1 promotes tumorigenesis by interacting with ribosomal protein RPL35
- Author
-
Liu, PY, Tee, AE, Milazzo, G, Hannan, KM, Maag, J, Mondal, S, Atmadibrata, B, Bartonicek, N, Peng, H, Ho, N, Mayoh, C, Ciaccio, R, Sun, Y, Henderson, MJ, Gao, J, Everaert, C, Hulme, AJ, Wong, M, Lan, Q, Cheung, BB, Shi, L, Wang, JY, Simon, T, Fischer, M, Zhang, XD, Marshall, GM, Norris, MD, Haber, M, Vandesompele, J, Li, J, Mestdagh, P, Hannan, RD, Dinger, ME, Perini, G, Liu, T, Liu, PY, Tee, AE, Milazzo, G, Hannan, KM, Maag, J, Mondal, S, Atmadibrata, B, Bartonicek, N, Peng, H, Ho, N, Mayoh, C, Ciaccio, R, Sun, Y, Henderson, MJ, Gao, J, Everaert, C, Hulme, AJ, Wong, M, Lan, Q, Cheung, BB, Shi, L, Wang, JY, Simon, T, Fischer, M, Zhang, XD, Marshall, GM, Norris, MD, Haber, M, Vandesompele, J, Li, J, Mestdagh, P, Hannan, RD, Dinger, ME, Perini, G, and Liu, T
- Abstract
The majority of patients with neuroblastoma due to MYCN oncogene amplification and consequent N-Myc oncoprotein over-expression die of the disease. Here our analyses of RNA sequencing data identify the long noncoding RNA lncNB1 as one of the transcripts most over-expressed in MYCN-amplified, compared with MYCN-non-amplified, human neuroblastoma cells and also the most over-expressed in neuroblastoma compared with all other cancers. lncNB1 binds to the ribosomal protein RPL35 to enhance E2F1 protein synthesis, leading to DEPDC1B gene transcription. The GTPase-activating protein DEPDC1B induces ERK protein phosphorylation and N-Myc protein stabilization. Importantly, lncNB1 knockdown abolishes neuroblastoma cell clonogenic capacity in vitro and leads to neuroblastoma tumor regression in mice, while high levels of lncNB1 and RPL35 in human neuroblastoma tissues predict poor patient prognosis. This study therefore identifies lncNB1 and its binding protein RPL35 as key factors for promoting E2F1 protein synthesis, N-Myc protein stability and N-Myc-driven oncogenesis, and as therapeutic targets.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.