Bojko, J.
Kollareddy, M.
Szemes, M.
Bellamy, J.
Poon, E.
Moukachar, A.
Legge, D.
Vincent, E.E.
Jones, N.
Malik, S.
Greenhough, A.
Paterson, A.
Park, J.H.
Gallacher, K.
Chesler, L.
Malik, K.
(2024). Spliceosomal vulnerability of MYCN-amplified neuroblastoma is contingent on PRMT5-mediated regulation of epitranscriptomic and metabolomic pathways. Cancer lett,
Vol.604,
p. 217263.
show abstract
Approximately 50 % of poor prognosis neuroblastomas arise due to MYCN over-expression. We previously demonstrated that MYCN and PRMT5 proteins interact and PRMT5 knockdown led to apoptosis of MYCN-amplified (MNA) neuroblastoma. Here we evaluate the highly selective first-in-class PRMT5 inhibitor GSK3203591 and its in vivo analogue GSK3326593 as targeted therapeutics for MNA neuroblastoma. Cell-line analyses show MYCN-dependent growth inhibition and apoptosis, with approximately 200-fold greater sensitivity of MNA neuroblastoma lines. RNA sequencing of three MNA neuroblastoma lines treated with GSK3203591 reveal deregulated MYCN transcriptional programmes and altered mRNA splicing, converging on key regulatory pathways such as DNA damage response, epitranscriptomics and cellular metabolism. Stable isotope labelling experiments in the same cell lines demonstrate that glutamine metabolism is impeded following GSK3203591 treatment, linking with disruption of the MLX/Mondo nutrient sensors via intron retention of MLX mRNA. Interestingly, glutaminase (GLS) protein decreases after GSK3203591 treatment despite unchanged transcript levels. We demonstrate that the RNA methyltransferase METTL3 and cognate reader YTHDF3 proteins are lowered following their mRNAs undergoing GSK3203591-induced splicing alterations, indicating epitranscriptomic regulation of GLS; accordingly, we observe decreases of GLS mRNA m6A methylation following GSK3203591 treatment, and decreased GLS protein following YTHDF3 knockdown. In vivo efficacy of GSK3326593 is confirmed by increased survival of Th-MYCN mice, with drug treatment triggering splicing events and protein decreases consistent with in vitro data. Together our study demonstrates the PRMT5-dependent spliceosomal vulnerability of MNA neuroblastoma and identifies the epitranscriptome and glutamine metabolism as critical determinants of this sensitivity..
Moreno, L.
Weston, R.
Owens, C.
Valteau-Couanet, D.
Gambart, M.
Castel, V.
Zwaan, C.M.
Nysom, K.
Gerber, N.
Castellano, A.
Laureys, G.
Ladenstein, R.
Rössler, J.
Makin, G.
Murphy, D.
Morland, B.
Vaidya, S.
Thebaud, E.
van Eijkelenburg, N.
Tweddle, D.A.
Barone, G.
Tandonnet, J.
Corradini, N.
Chastagner, P.
Paillard, C.
Bautista, F.J.
Gallego Melcon, S.
De Wilde, B.
Marshall, L.
Gray, J.
Burchill, S.A.
Schleiermacher, G.
Chesler, L.
Peet, A.
Leach, M.O.
McHugh, K.
Hayes, R.
Jerome, N.
Caron, H.
Laidler, J.
Fenwick, N.
Holt, G.
Moroz, V.
Kearns, P.
Gates, S.
Pearson, A.D.
Wheatley, K.
Innovative Therapies for Children with Cancer (ITCC) and European Association for Neuroblastoma Research (SIOPEN),
(2024). Bevacizumab, Irinotecan, or Topotecan Added to Temozolomide for Children With Relapsed and Refractory Neuroblastoma: Results of the ITCC-SIOPEN BEACON-Neuroblastoma Trial. J clin oncol,
Vol.42
(10),
pp. 1135-1145.
show abstract
PURPOSE: Outcomes for children with relapsed and refractory high-risk neuroblastoma (RR-HRNB) remain dismal. The BEACON Neuroblastoma trial (EudraCT 2012-000072-42) evaluated three backbone chemotherapy regimens and the addition of the antiangiogenic agent bevacizumab (B). MATERIALS AND METHODS: Patients age 1-21 years with RR-HRNB with adequate organ function and performance status were randomly assigned in a 3 × 2 factorial design to temozolomide (T), irinotecan-temozolomide (IT), or topotecan-temozolomide (TTo) with or without B. The primary end point was best overall response (complete or partial) rate (ORR) during the first six courses, by RECIST or International Neuroblastoma Response Criteria for patients with measurable or evaluable disease, respectively. Safety, progression-free survival (PFS), and overall survival (OS) time were secondary end points. RESULTS: One hundred sixty patients with RR-HRNB were included. For B random assignment (n = 160), the ORR was 26% (95% CI, 17 to 37) with B and 18% (95% CI, 10 to 28) without B (risk ratio [RR], 1.52 [95% CI, 0.83 to 2.77]; P = .17). Adjusted hazard ratio for PFS and OS were 0.89 (95% CI, 0.63 to 1.27) and 1.01 (95% CI, 0.70 to 1.45), respectively. For irinotecan ([I]; n = 121) and topotecan (n = 60) random assignments, RRs for ORR were 0.94 and 1.22, respectively. A potential interaction between I and B was identified. For patients in the bevacizumab-irinotecan-temozolomide (BIT) arm, the ORR was 23% (95% CI, 10 to 42), and the 1-year PFS estimate was 0.67 (95% CI, 0.47 to 0.80). CONCLUSION: The addition of B met protocol-defined success criteria for ORR and appeared to improve PFS. Within this phase II trial, BIT showed signals of antitumor activity with acceptable tolerability. Future trials will confirm these results in the chemoimmunotherapy era..
Saldana-Guerrero, I.M.
Montano-Gutierrez, L.F.
Boswell, K.
Hafemeister, C.
Poon, E.
Shaw, L.E.
Stavish, D.
Lea, R.A.
Wernig-Zorc, S.
Bozsaky, E.
Fetahu, I.S.
Zoescher, P.
Pötschger, U.
Bernkopf, M.
Wenninger-Weinzierl, A.
Sturtzel, C.
Souilhol, C.
Tarelli, S.
Shoeb, M.R.
Bozatzi, P.
Rados, M.
Guarini, M.
Buri, M.C.
Weninger, W.
Putz, E.M.
Huang, M.
Ladenstein, R.
Andrews, P.W.
Barbaric, I.
Cresswell, G.D.
Bryant, H.E.
Distel, M.
Chesler, L.
Taschner-Mandl, S.
Farlik, M.
Tsakiridis, A.
Halbritter, F.
(2024). A human neural crest model reveals the developmental impact of neuroblastoma-associated chromosomal aberrations. Nat commun,
Vol.15
(1),
p. 3745.
show abstract
full text
Early childhood tumours arise from transformed embryonic cells, which often carry large copy number alterations (CNA). However, it remains unclear how CNAs contribute to embryonic tumourigenesis due to a lack of suitable models. Here we employ female human embryonic stem cell (hESC) differentiation and single-cell transcriptome and epigenome analysis to assess the effects of chromosome 17q/1q gains, which are prevalent in the embryonal tumour neuroblastoma (NB). We show that CNAs impair the specification of trunk neural crest (NC) cells and their sympathoadrenal derivatives, the putative cells-of-origin of NB. This effect is exacerbated upon overexpression of MYCN, whose amplification co-occurs with CNAs in NB. Moreover, CNAs potentiate the pro-tumourigenic effects of MYCN and mutant NC cells resemble NB cells in tumours. These changes correlate with a stepwise aberration of developmental transcription factor networks. Together, our results sketch a mechanistic framework for the CNA-driven initiation of embryonal tumours..
Wienke, J.
Visser, L.L.
Kholosy, W.M.
Keller, K.M.
Barisa, M.
Poon, E.
Munnings-Tomes, S.
Himsworth, C.
Calton, E.
Rodriguez, A.
Bernardi, R.
van den Ham, F.
van Hooff, S.R.
Matser, Y.A.
Tas, M.L.
Langenberg, K.P.
Lijnzaad, P.
Borst, A.L.
Zappa, E.
Bergsma, F.J.
Strijker, J.G.
Verhoeven, B.M.
Mei, S.
Kramdi, A.
Restuadi, R.
Sanchez-Bernabeu, A.
Cornel, A.M.
Holstege, F.C.
Gray, J.C.
Tytgat, G.A.
Scheijde-Vermeulen, M.A.
Wijnen, M.H.
Dierselhuis, M.P.
Straathof, K.
Behjati, S.
Wu, W.
Heck, A.J.
Koster, J.
Nierkens, S.
Janoueix-Lerosey, I.
de Krijger, R.R.
Baryawno, N.
Chesler, L.
Anderson, J.
Caron, H.N.
Margaritis, T.
van Noesel, M.M.
Molenaar, J.J.
(2024). Integrative analysis of neuroblastoma by single-cell RNA sequencing identifies the NECTIN2-TIGIT axis as a target for immunotherapy. Cancer cell,
Vol.42
(2),
pp. 283-300.e8.
show abstract
Pediatric patients with high-risk neuroblastoma have poor survival rates and urgently need more effective treatment options with less side effects. Since novel and improved immunotherapies may fill this need, we dissect the immunoregulatory interactions in neuroblastoma by single-cell RNA-sequencing of 24 tumors (10 pre- and 14 post-chemotherapy, including 5 pairs) to identify strategies for optimizing immunotherapy efficacy. Neuroblastomas are infiltrated by natural killer (NK), T and B cells, and immunosuppressive myeloid populations. NK cells show reduced cytotoxicity and T cells have a dysfunctional profile. Interaction analysis reveals a vast immunoregulatory network and identifies NECTIN2-TIGIT as a crucial immune checkpoint. Combined blockade of TIGIT and PD-L1 significantly reduces neuroblastoma growth, with complete responses (CR) in vivo. Moreover, addition of TIGIT+PD-L1 blockade to standard relapse treatment in a chemotherapy-resistant Th-ALKF1174L/MYCN 129/SvJ syngeneic model induces CR. In conclusion, our integrative analysis provides promising targets and a rationale for immunotherapeutic combination strategies..
Halliwell, E.
Vitali, A.
Muller, H.
Alonso-Ferrero, M.
Barisa, M.
Gavriil, A.
Piapi, A.
Leboreiro-Babe, C.
Gileadi, T.
Yeung, J.
Pataillot-Meakin, T.
Fisher, J.
Tucker, L.
Donovan, L.
Chesler, L.
Chester, K.
Anderson, J.
(2023). Targeting of low ALK antigen density neuroblastoma using AND logic-gate engineered CAR-T cells. Cytotherapy,
Vol.25
(1),
pp. 46-58.
show abstract
BACKGROUND AIMS: The targeting of solid cancers with chimeric antigen receptor (CAR) T cells faces many technological hurdles, including selection of optimal target antigens. Promising pre-clinical and clinical data of CAR T-cell activity have emerged from targeting surface antigens such as GD2 and B7H3 in childhood cancer neuroblastoma. Anaplastic lymphoma kinase (ALK) is expressed in a majority of neuroblastomas at low antigen density but is largely absent from healthy tissues. METHODS: To explore an alternate target antigen for neuroblastoma CAR T-cell therapy, the authors generated and screened a single-chain variable fragment library targeting ALK extracellular domain to make a panel of new anti-ALK CAR T-cell constructs. RESULTS: A lead novel CAR T-cell construct was capable of specific cytotoxicity against neuroblastoma cells expressing low levels of ALK, but with only weak cytokine and proliferative T-cell responses. To explore strategies for amplifying ALK CAR T cells, the authors generated a co-CAR approach in which T cells received signal 1 from a first-generation ALK construct and signal 2 from anti-B7H3 or GD2 chimeric co-stimulatory receptors. The co-CAR approach successfully demonstrated the ability to avoid targeting single-antigen-positive targets as a strategy for mitigating on-target off-tumor toxicity. CONCLUSIONS: These data provide further proof of concept for ALK as a neuroblastoma CAR T-cell target..
Tucker, E.R.
Jiménez, I.
Chen, L.
Bellini, A.
Gorrini, C.
Calton, E.
Gao, Q.
Che, H.
Poon, E.
Jamin, Y.
Martins Da Costa, B.
Barker, K.
Shrestha, S.
Hutchinson, J.C.
Dhariwal, S.
Goodman, A.
Del Nery, E.
Gestraud, P.
Bhalshankar, J.
Iddir, Y.
Saberi-Ansari, E.
Saint-Charles, A.
Geoerger, B.
Marques Da Costa, M.E.
Pierre-Eugène, C.
Janoueix-Lerosey, I.
Decaudin, D.
Nemati, F.
Carcaboso, A.M.
Surdez, D.
Delattre, O.
George, S.L.
Chesler, L.
Tweddle, D.A.
Schleiermacher, G.
(2023). Combination Therapies Targeting ALK-aberrant Neuroblastoma in Preclinical Models. Clin cancer res,
Vol.29
(7),
pp. 1317-1331.
show abstract
full text
PURPOSE: ALK-activating mutations are identified in approximately 10% of newly diagnosed neuroblastomas and ALK amplifications in a further 1%-2% of cases. Lorlatinib, a third-generation anaplastic lymphoma kinase (ALK) inhibitor, will soon be given alongside induction chemotherapy for children with ALK-aberrant neuroblastoma. However, resistance to single-agent treatment has been reported and therapies that improve the response duration are urgently required. We studied the preclinical combination of lorlatinib with chemotherapy, or with the MDM2 inhibitor, idasanutlin, as recent data have suggested that ALK inhibitor resistance can be overcome through activation of the p53-MDM2 pathway. EXPERIMENTAL DESIGN: We compared different ALK inhibitors in preclinical models prior to evaluating lorlatinib in combination with chemotherapy or idasanutlin. We developed a triple chemotherapy (CAV: cyclophosphamide, doxorubicin, and vincristine) in vivo dosing schedule and applied this to both neuroblastoma genetically engineered mouse models (GEMM) and patient-derived xenografts (PDX). RESULTS: Lorlatinib in combination with chemotherapy was synergistic in immunocompetent neuroblastoma GEMM. Significant growth inhibition in response to lorlatinib was only observed in the ALK-amplified PDX model with high ALK expression. In this PDX, lorlatinib combined with idasanutlin resulted in complete tumor regression and significantly delayed tumor regrowth. CONCLUSIONS: In our preclinical neuroblastoma models, high ALK expression was associated with lorlatinib response alone or in combination with either chemotherapy or idasanutlin. The synergy between MDM2 and ALK inhibition warrants further evaluation of this combination as a potential clinical approach for children with neuroblastoma..
Goldsmith, K.C.
Park, J.R.
Kayser, K.
Malvar, J.
Chi, Y.-.
Groshen, S.G.
Villablanca, J.G.
Krytska, K.
Lai, L.M.
Acharya, P.T.
Goodarzian, F.
Pawel, B.
Shimada, H.
Ghazarian, S.
States, L.
Marshall, L.
Chesler, L.
Granger, M.
Desai, A.V.
Mody, R.
Morgenstern, D.A.
Shusterman, S.
Macy, M.E.
Pinto, N.
Schleiermacher, G.
Vo, K.
Thurm, H.C.
Chen, J.
Liyanage, M.
Peltz, G.
Matthay, K.K.
Berko, E.R.
Maris, J.M.
Marachelian, A.
Mossé, Y.P.
(2023). Lorlatinib with or without chemotherapy in ALK-driven refractory/relapsed neuroblastoma: phase 1 trial results. Nat med,
Vol.29
(5),
pp. 1092-1102.
show abstract
full text
Neuroblastomas harbor ALK aberrations clinically resistant to crizotinib yet sensitive pre-clinically to the third-generation ALK inhibitor lorlatinib. We conducted a first-in-child study evaluating lorlatinib with and without chemotherapy in children and adults with relapsed or refractory ALK-driven neuroblastoma. The trial is ongoing, and we report here on three cohorts that have met pre-specified primary endpoints: lorlatinib as a single agent in children (12 months to <18 years); lorlatinib as a single agent in adults (≥18 years); and lorlatinib in combination with topotecan/cyclophosphamide in children (<18 years). Primary endpoints were safety, pharmacokinetics and recommended phase 2 dose (RP2D). Secondary endpoints were response rate and 123I-metaiodobenzylguanidine (MIBG) response. Lorlatinib was evaluated at 45-115 mg/m2/dose in children and 100-150 mg in adults. Common adverse events (AEs) were hypertriglyceridemia (90%), hypercholesterolemia (79%) and weight gain (87%). Neurobehavioral AEs occurred mainly in adults and resolved with dose hold/reduction. The RP2D of lorlatinib with and without chemotherapy in children was 115 mg/m2. The single-agent adult RP2D was 150 mg. The single-agent response rate (complete/partial/minor) for <18 years was 30%; for ≥18 years, 67%; and for chemotherapy combination in <18 years, 63%; and 13 of 27 (48%) responders achieved MIBG complete responses, supporting lorlatinib's rapid translation into active phase 3 trials for patients with newly diagnosed high-risk, ALK-driven neuroblastoma. ClinicalTrials.gov registration: NCT03107988 ..
Ferguson, K.M.
Gillen, S.L.
Chaytor, L.
Poon, E.
Marcos, D.
Gomez, R.L.
Woods, L.M.
Mykhaylechko, L.
Elfari, L.
Martins da Costa, B.
Jamin, Y.
Carroll, J.S.
Chesler, L.
Ali, F.R.
Philpott, A.
(2023). Palbociclib releases the latent differentiation capacity of neuroblastoma cells. Dev cell,
Vol.58
(19),
pp. 1967-1982.e8.
show abstract
Neuroblastoma is the most common extracranial solid tumor in infants, arising from developmentally stalled neural crest-derived cells. Driving tumor differentiation is a promising therapeutic approach for this devastating disease. Here, we show that the CDK4/6 inhibitor palbociclib not only inhibits proliferation but induces extensive neuronal differentiation of adrenergic neuroblastoma cells. Palbociclib-mediated differentiation is manifested by extensive phenotypic and transcriptional changes accompanied by the establishment of an epigenetic program driving expression of mature neuronal features. In vivo palbociclib significantly inhibits tumor growth in mouse neuroblastoma models. Furthermore, dual treatment with retinoic acid resets the oncogenic adrenergic core regulatory circuit of neuroblastoma cells, further suppresses proliferation, and can enhance differentiation, altering gene expression in ways that significantly correlate with improved patient survival. We therefore identify palbociclib as a therapeutic approach to dramatically enhance neuroblastoma differentiation efficacy that could be used in combination with retinoic acid to improve patient outcomes..
Sbirkov, Y.
Schenk, T.
Kwok, C.
Stengel, S.
Brown, R.
Brown, G.
Chesler, L.
Zelent, A.
Fuchter, M.J.
Petrie, K.
(2023). Dual inhibition of EZH2 and G9A/GLP histone methyltransferases by HKMTI-1-005 promotes differentiation of acute myeloid leukemia cells. Front cell dev biol,
Vol.11,
p. 1076458.
show abstract
full text
All-trans-retinoic acid (ATRA)-based differentiation therapy of acute promyelocytic leukemia (APL) represents one of the most clinically effective examples of precision medicine and the first example of targeted oncoprotein degradation. The success of ATRA in APL, however, remains to be translated to non-APL acute myeloid leukemia (AML). We previously showed that aberrant histone modifications, including histone H3 lysine 4 (H3K4) and lysine 27 (H3K27) methylation, were associated with this lack of response and that epigenetic therapy with small molecule inhibitors of the H3K4 demethylase LSD1/KDM1A could reprogram AML cells to respond to ATRA. Serving as the enzymatic component of Polycomb Repressive Complex 2, EZH2/KMT6A methyltransferase plays a critical role in normal hematopoiesis by affecting the balance between self-renewal and differentiation. The canonical function of EZH2 is methylation of H3K27, although important non-canonical roles have recently been described. EZH2 mutation or deregulated expression has been conclusively demonstrated in the pathogenesis of AML and response to treatment, thus making it an attractive therapeutic target. In this study, we therefore investigated whether inhibition of EZH2 might also improve the response of non-APL AML cells to ATRA-based therapy. We focused on GSK-343, a pyridone-containing S-adenosyl-L-methionine cofactor-competitive EZH2 inhibitor that is representative of its class, and HKMTI-1-005, a substrate-competitive dual inhibitor targeting EZH2 and the closely related G9A/GLP H3K9 methyltransferases. We found that treatment with HKMTI-1-005 phenocopied EZH2 knockdown and was more effective in inducing differentiation than GSK-343, despite the efficacy of GSK-343 in terms of abolishing H3K27 trimethylation. Furthermore, transcriptomic analysis revealed that in contrast to treatment with GSK-343, HKMTI-1-005 upregulated the expression of differentiation pathway genes with and without ATRA, while downregulating genes associated with a hematopoietic stem cell phenotype. These results pointed to a non-canonical role for EZH2, which was supported by the finding that EZH2 associates with the master regulator of myeloid differentiation, RARα, in an ATRA-dependent manner that was enhanced by HKMTI-1-005, possibly playing a role in co-regulator complex exchange during transcriptional activation. In summary, our results strongly suggest that addition of HKMTI-1-005 to ATRA is a new therapeutic approach against AML that warrants further investigation..
Stankunaite, R.
George, S.L.
Gallagher, L.
Jamal, S.
Shaikh, R.
Yuan, L.
Hughes, D.
Proszek, P.Z.
Carter, P.
Pietka, G.
Heide, T.
James, C.
Tari, H.
Lynn, C.
Jain, N.
Portela, L.R.
Rogers, T.
Vaidya, S.J.
Chisholm, J.C.
Carceller, F.
Szychot, E.
Mandeville, H.
Angelini, P.
Jesudason, A.B.
Jackson, M.
Marshall, L.V.
Gatz, S.A.
Anderson, J.
Sottoriva, A.
Chesler, L.
Hubank, M.
(2022). Circulating tumour DNA sequencing to determine therapeutic response and identify tumour heterogeneity in patients with paediatric solid tumours. Eur j cancer,
Vol.162,
pp. 209-220.
show abstract
full text
OBJECTIVE: Clinical diagnostic sequencing of circulating tumour DNA (ctDNA) is well advanced for adult patients, but application to paediatric cancer patients lags behind. METHODS: To address this, we have developed a clinically relevant (67 gene) NGS capture panel and accompanying workflow that enables sensitive and reliable detection of low-frequency genetic variants in cell-free DNA (cfDNA) from children with solid tumours. We combined gene panel sequencing with low pass whole-genome sequencing of the same library to inform on genome-wide copy number changes in the blood. RESULTS: Analytical validity was evaluated using control materials, and the method was found to be highly sensitive (0.96 for SNVs and 0.97 for INDEL), specific (0.82 for SNVs and 0.978 for INDEL), repeatable (>0.93 [95% CI: 0.89-0.95]) and reproducible (>0.87 [95% CI: 0.87-0.95]). Potential for clinical application was demonstrated in 39 childhood cancer patients with a spectrum of solid tumours in which the single nucleotide variants expected from tumour sequencing were detected in cfDNA in 94.4% (17/18) of cases with active extracranial disease. In 13 patients, where serial samples were available, we show a close correlation between events detected in cfDNA and treatment response, demonstrate that cfDNA analysis could be a useful tool to monitor disease progression, and show cfDNA sequencing has the potential to identify targetable variants that were not detected in tumour samples. CONCLUSIONS: This is the first pan-cancer DNA sequencing panel that we know to be optimised for cfDNA in children for blood-based molecular diagnostics in paediatric solid tumours..
Urban-Wójciuk, Z.
Graham, A.
Barker, K.
Kwok, C.
Sbirkov, Y.
Howell, L.
Campbell, J.
Woster, P.M.
Poon, E.
Petrie, K.
Chesler, L.
(2022). The biguanide polyamine analog verlindamycin promotes differentiation in neuroblastoma via induction of antizyme. Cancer gene ther,
Vol.29
(7),
pp. 940-950.
show abstract
full text
Deregulated polyamine biosynthesis is emerging as a common feature of neuroblastoma and drugs targeting this metabolic pathway such as DFMO are in clinical and preclinical development. The polyamine analog verlindamycin inhibits the polyamine biosynthesis pathway enzymes SMOX and PAOX, as well as the histone demethylase LSD1. Based on our previous research in acute myeloid leukemia (AML), we reasoned verlindamycin may also unblock neuroblastoma differentiation when combined with all-trans-retinoic acid (ATRA). Indeed, co-treatment with verlindamycin and ATRA strongly induced differentiation regardless of MYCN status, but in MYCN-expressing cells, protein levels were strongly diminished. This process was not transcriptionally regulated but was due to increased degradation of MYCN protein, at least in part via ubiquitin-independent, proteasome-dependent destruction. Here we report that verlindamycin effectively induces the expression of functional tumor suppressor-antizyme via ribosomal frameshifting. Consistent with previous results describing the function of antizyme, we found that verlindamycin treatment led to the selective targeting of ornithine decarboxylase (the rate-limiting enzyme for polyamine biosynthesis) as well as key oncoproteins, such as cyclin D and Aurora A kinase. Retinoid-based multimodal differentiation therapy is one of the few interventions that extends relapse-free survival in MYCN-associated high-risk neuroblastoma and these results point toward the potential use of verlindamycin in this regimen..
Berlak, M.
Tucker, E.
Dorel, M.
Winkler, A.
McGearey, A.
Rodriguez-Fos, E.
da Costa, B.M.
Barker, K.
Fyle, E.
Calton, E.
Eising, S.
Ober, K.
Hughes, D.
Koutroumanidou, E.
Carter, P.
Stankunaite, R.
Proszek, P.
Jain, N.
Rosswog, C.
Dorado-Garcia, H.
Molenaar, J.J.
Hubank, M.
Barone, G.
Anderson, J.
Lang, P.
Deubzer, H.E.
Künkele, A.
Fischer, M.
Eggert, A.
Kloft, C.
Henssen, A.G.
Boettcher, M.
Hertwig, F.
Blüthgen, N.
Chesler, L.
Schulte, J.H.
(2022). Mutations in ALK signaling pathways conferring resistance to ALK inhibitor treatment lead to collateral vulnerabilities in neuroblastoma cells. Mol cancer,
Vol.21
(1),
p. 126.
show abstract
full text
BACKGROUND: Development of resistance to targeted therapies has tempered initial optimism that precision oncology would improve poor outcomes for cancer patients. Resistance mechanisms, however, can also confer new resistance-specific vulnerabilities, termed collateral sensitivities. Here we investigated anaplastic lymphoma kinase (ALK) inhibitor resistance in neuroblastoma, a childhood cancer frequently affected by activating ALK alterations. METHODS: Genome-wide forward genetic CRISPR-Cas9 based screens were performed to identify genes associated with ALK inhibitor resistance in neuroblastoma cell lines. Furthermore, the neuroblastoma cell line NBLW-R was rendered resistant by continuous exposure to ALK inhibitors. Genes identified to be associated with ALK inhibitor resistance were further investigated by generating suitable cell line models. In addition, tumor and liquid biopsy samples of four patients with ALK-mutated neuroblastomas before ALK inhibitor treatment and during tumor progression under treatment were genomically profiled. RESULTS: Both genome-wide CRISPR-Cas9-based screens and preclinical spontaneous ALKi resistance models identified NF1 loss and activating NRASQ61K mutations to confer resistance to chemically diverse ALKi. Moreover, human neuroblastomas recurrently developed de novo loss of NF1 and activating RAS mutations after ALKi treatment, leading to therapy resistance. Pathway-specific perturbations confirmed that NF1 loss and activating RAS mutations lead to RAS-MAPK signaling even in the presence of ALKi. Intriguingly, NF1 loss rendered neuroblastoma cells hypersensitive to MEK inhibition. CONCLUSIONS: Our results provide a clinically relevant mechanistic model of ALKi resistance in neuroblastoma and highlight new clinically actionable collateral sensitivities in resistant cells..
Ruhen, O.
Lak, N.S.
Stutterheim, J.
Danielli, S.G.
Chicard, M.
Iddir, Y.
Saint-Charles, A.
Di Paolo, V.
Tombolan, L.
Gatz, S.A.
Aladowicz, E.
Proszek, P.
Jamal, S.
Stankunaite, R.
Hughes, D.
Carter, P.
Izquierdo, E.
Wasti, A.
Chisholm, J.C.
George, S.L.
Pace, E.
Chesler, L.
Aerts, I.
Pierron, G.
Zaidi, S.
Delattre, O.
Surdez, D.
Kelsey, A.
Hubank, M.
Bonvini, P.
Bisogno, G.
Di Giannatale, A.
Schleiermacher, G.
Schäfer, B.W.
Tytgat, G.A.
Shipley, J.
(2022). Molecular Characterization of Circulating Tumor DNA in Pediatric Rhabdomyosarcoma: A Feasibility Study. Jco precis oncol,
Vol.6,
p. e2100534.
show abstract
full text
PURPOSE: Rhabdomyosarcomas (RMS) are rare neoplasms affecting children and young adults. Efforts to improve patient survival have been undermined by a lack of suitable disease markers. Plasma circulating tumor DNA (ctDNA) has shown promise as a potential minimally invasive biomarker and monitoring tool in other cancers; however, it remains underexplored in RMS. We aimed to determine the feasibility of identifying and quantifying ctDNA in plasma as a marker of disease burden and/or treatment response using blood samples from RMS mouse models and patients. METHODS: We established mouse models of RMS and applied quantitative polymerase chain reaction (PCR) and droplet digital PCR (ddPCR) to detect ctDNA within the mouse plasma. Potential driver mutations, copy-number alterations, and DNA breakpoints associated with PAX3/7-FOXO1 gene fusions were identified in the RMS samples collected at diagnosis. Patient-matched plasma samples collected from 28 patients with RMS before, during, and after treatment were analyzed for the presence of ctDNA via ddPCR, panel sequencing, and/or whole-exome sequencing. RESULTS: Human tumor-derived DNA was detectable in plasma samples from mouse models of RMS and correlated with tumor burden. In patients, ctDNA was detected in 14/18 pretreatment plasma samples with ddPCR and 7/7 cases assessed by sequencing. Levels of ctDNA at diagnosis were significantly higher in patients with unfavorable tumor sites, positive nodal status, and metastasis. In patients with serial plasma samples (n = 18), fluctuations in ctDNA levels corresponded to treatment response. CONCLUSION: Comprehensive ctDNA analysis combining high sensitivity and throughput can identify key molecular drivers in RMS models and patients, suggesting potential as a minimally invasive biomarker. Preclinical assessment of treatments using mouse models and further patient testing through prospective clinical trials are now warranted..
Nijhuis, A.
Sikka, A.
Yogev, O.
Herendi, L.
Balcells, C.
Ma, Y.
Poon, E.
Eckold, C.
Valbuena, G.N.
Xu, Y.
Liu, Y.
da Costa, B.M.
Gruet, M.
Wickremesinghe, C.
Benito, A.
Kramer, H.
Montoya, A.
Carling, D.
Want, E.J.
Jamin, Y.
Chesler, L.
Keun, H.C.
(2022). Indisulam targets RNA splicing and metabolism to serve as a therapeutic strategy for high-risk neuroblastoma. Nat commun,
Vol.13
(1),
p. 1380.
show abstract
full text
Neuroblastoma is the most common paediatric solid tumour and prognosis remains poor for high-risk cases despite the use of multimodal treatment. Analysis of public drug sensitivity data showed neuroblastoma lines to be sensitive to indisulam, a molecular glue that selectively targets RNA splicing factor RBM39 for proteosomal degradation via DCAF15-E3-ubiquitin ligase. In neuroblastoma models, indisulam induces rapid loss of RBM39, accumulation of splicing errors and growth inhibition in a DCAF15-dependent manner. Integrative analysis of RNAseq and proteomics data highlight a distinct disruption to cell cycle and metabolism. Metabolic profiling demonstrates metabolome perturbations and mitochondrial dysfunction resulting from indisulam. Complete tumour regression without relapse was observed in both xenograft and the Th-MYCN transgenic model of neuroblastoma after indisulam treatment, with RBM39 loss, RNA splicing and metabolic changes confirmed in vivo. Our data show that dual-targeting of metabolism and RNA splicing with anticancer indisulam is a promising therapeutic approach for high-risk neuroblastoma..
Stankunaite, R.
Marshall, L.V.
Carceller, F.
Chesler, L.
Hubank, M.
George, S.L.
(2022). Liquid biopsy for children with central nervous system tumours: Clinical integration and technical considerations. Front pediatr,
Vol.10,
p. 957944.
show abstract
full text
Circulating cell-free DNA (cfDNA) analysis has the potential to revolutionise the care of patients with cancer and is already moving towards standard of care in some adult malignancies. Evidence for the utility of cfDNA analysis in paediatric cancer patients is also accumulating. In this review we discuss the limitations of blood-based assays in patients with brain tumours and describe the evidence supporting cerebrospinal fluid (CSF) cfDNA analysis. We make recommendations for CSF cfDNA processing to aid the standardisation and technical validation of future assays. We discuss the considerations for interpretation of cfDNA analysis and highlight promising future directions. Overall, cfDNA profiling shows great potential as an adjunct to the analysis of biopsy tissue in paediatric cancer patients, with the potential to provide a genetic molecular profile of the tumour when tissue biopsy is not feasible. However, to fully realise the potential of cfDNA analysis for children with brain tumours larger prospective studies incorporating serial CSF sampling are required..
Tsakaneli, A.
Corasolla Carregari, V.
Morini, M.
Eva, A.
Cangemi, G.
Chayka, O.
Makarov, E.
Poon, E.
Chesler, L.
Pieroni, L.
Larsen, M.
Palmisano, G.
Sala, A.
(2021). MYCN Regulates Metabolism Through Vesicular Transfer of Glycolytic Kinases. Open biology,
.
Roeschert, I.
Poon, E.
Henssen, A.G.
Garcia, H.D.
Gatti, M.
Giansanti, C.
Jamin, Y.
Ade, C.P.
Gallant, P.
Schülein-Völk, C.
Beli, P.
Richards, M.
Rosenfeldt, M.
Altmeyer, M.
Anderson, J.
Eggert, A.
Dobbelstein, M.
Bayliss, R.
Chesler, L.
Büchel, G.
Eilers, M.
(2021). Combined inhibition of Aurora-A and ATR kinase results in regression of MYCN-amplified neuroblastoma. Nat cancer,
Vol.2
(3),
pp. 312-326.
show abstract
full text
Amplification of MYCN is the driving oncogene in a subset of high-risk neuroblastoma. The MYCN protein and the Aurora-A kinase form a complex during S phase that stabilizes MYCN. Here we show that MYCN activates Aurora-A on chromatin, which phosphorylates histone H3 at serine 10 in S phase, promotes the deposition of histone H3.3 and suppresses R-loop formation. Inhibition of Aurora-A induces transcription-replication conflicts and activates the Ataxia telangiectasia and Rad3 related (ATR) kinase, which limits double-strand break accumulation upon Aurora-A inhibition. Combined inhibition of Aurora-A and ATR induces rampant tumor-specific apoptosis and tumor regression in mouse models of neuroblastoma, leading to permanent eradication in a subset of mice. The therapeutic efficacy is due to both tumor cell-intrinsic and immune cell-mediated mechanisms. We propose that targeting the ability of Aurora-A to resolve transcription-replication conflicts is an effective therapy for MYCN-driven neuroblastoma (141 words)..
Pearson, A.D.
DuBois, S.G.
Buenger, V.
Kieran, M.
Stegmaier, K.
Bandopadhayay, P.
Bennett, K.
Bourdeaut, F.
Brown, P.A.
Chesler, L.
Clymer, J.
Fox, E.
French, C.A.
Germovsek, E.
Giles, F.J.
Bender, J.G.
Hattersley, M.M.
Ludwinski, D.
Luptakova, K.
Maris, J.
McDonough, J.
Nikolova, Z.
Smith, M.
Tsiatis, A.C.
Vibhakar, R.
Weiner, S.
Yi, J.S.
Zheng, F.
Vassal, G.
(2021). Bromodomain and extra-terminal inhibitors-A consensus prioritisation after the Paediatric Strategy Forum for medicinal product development of epigenetic modifiers in children-ACCELERATE. Eur j cancer,
Vol.146,
pp. 115-124.
show abstract
full text
Based on biology and pre-clinical data, bromodomain and extra-terminal (BET) inhibitors have at least three potential roles in paediatric malignancies: NUT (nuclear protein in testis) carcinomas, MYC/MYCN-driven cancers and fusion-driven malignancies. However, there are now at least 10 BET inhibitors in development, with a limited relevant paediatric population in which to evaluate these medicinal products. Therefore, a meeting was convened with the specific aim to develop a consensus among relevant biopharmaceutical companies, academic researchers, as well as patient and family advocates, about the development of BET inhibitors, including prioritisation and their specific roles in children. Although BET inhibitors have been in clinical trials in adults since 2012, the first-in-child study (BMS-986158) only opened in 2019. In the future, when there is strong mechanistic rationale or pre-clinical activity of a class of medicinal product in paediatrics, early clinical evaluation with embedded correlative studies of a member of the class should be prioritised and rapidly executed in paediatric populations. There is a strong mechanistic and biological rationale to evaluate BET inhibitors in paediatrics, underpinned by substantial, but not universal, pre-clinical data. However, most pan-BET inhibitors have been challenging to administer in adults, since monotherapy results in only modest anti-tumour activity and provides a narrow therapeutic index due to thrombocytopenia. It was concluded that it is neither scientifically justified nor feasible to undertake simultaneously early clinical trials in paediatrics of all pan-BET inhibitors. However, there is a clinical need for global access to BET inhibitors for patients with NUT carcinoma, a very rare malignancy driven by bromodomain fusions, with proof of concept of clinical benefit in a subset of patients treated with BET inhibitors. Development and regulatory pathway in this indication should include children and adolescents as well as adults. Beyond NUT carcinoma, it was proposed that further clinical development of other pan-BET inhibitors in children should await the results of the first paediatric clinical trial of BMS-986158, unless there is compelling rationale based on the specific agent of interest. BDII-selective inhibitors, central nervous system-penetrant BET inhibitors (e.g. CC-90010), and those dual-targeting BET/p300 bromodomain are of particular interest and warrant further pre-clinical investigation. This meeting emphasised the value of a coordinated and integrated strategy to drug development in paediatric oncology. A multi-stakeholder approach with multiple companies developing a consensus with academic investigators early in the development of a class of compounds, and then engaging regulatory agencies would improve efficiency, productivity, conserve resources and maximise potential benefit for children with cancer..
Ingley, K.M.
Hughes, D.
Hubank, M.
Lindsay, D.
Plumb, A.
Cox, R.
Chesler, L.
Strauss, S.J.
(2021). Durable response to serial tyrosine kinase inhibitors (TKIs) in an adolescent with metastatic TFG-ROS1 fusion positive Inflammatory Myofibroblastic Tumor (IMT). Lung cancer,
Vol.158,
pp. 151-155.
show abstract
Here, we present the case of an adolescent with a rare metastatic Inflammatory myofibroblastic tumor (IMT) harboring a TFG-ROS1 fusion initially detected on tumor progression and retrospectively identified in the primary tumor after targeted RNA sequencing. The patient benefitted from sequential TKIs over a 5-year period with response to the third generation ALK/ROS inhibitor, lorlatinib leading to resection of the primary tumor. Detailed molecular analysis can identify targetable oncogenic kinase fusions that alters management in patients with unresectable disease and should be considered in all patients..
Dubiella, C.
Pinch, B.J.
Koikawa, K.
Zaidman, D.
Poon, E.
Manz, T.D.
Nabet, B.
He, S.
Resnick, E.
Rogel, A.
Langer, E.M.
Daniel, C.J.
Seo, H.-.
Chen, Y.
Adelmant, G.
Sharifzadeh, S.
Ficarro, S.B.
Jamin, Y.
Martins da Costa, B.
Zimmerman, M.W.
Lian, X.
Kibe, S.
Kozono, S.
Doctor, Z.M.
Browne, C.M.
Yang, A.
Stoler-Barak, L.
Shah, R.B.
Vangos, N.E.
Geffken, E.A.
Oren, R.
Koide, E.
Sidi, S.
Shulman, Z.
Wang, C.
Marto, J.A.
Dhe-Paganon, S.
Look, T.
Zhou, X.Z.
Lu, K.P.
Sears, R.C.
Chesler, L.
Gray, N.S.
London, N.
(2021). Sulfopin is a covalent inhibitor of Pin1 that blocks Myc-driven tumors in vivo. Nat chem biol,
Vol.17
(9),
pp. 954-963.
show abstract
full text
The peptidyl-prolyl isomerase, Pin1, is exploited in cancer to activate oncogenes and inactivate tumor suppressors. However, despite considerable efforts, Pin1 has remained an elusive drug target. Here, we screened an electrophilic fragment library to identify covalent inhibitors targeting Pin1's active site Cys113, leading to the development of Sulfopin, a nanomolar Pin1 inhibitor. Sulfopin is highly selective, as validated by two independent chemoproteomics methods, achieves potent cellular and in vivo target engagement and phenocopies Pin1 genetic knockout. Pin1 inhibition had only a modest effect on cancer cell line viability. Nevertheless, Sulfopin induced downregulation of c-Myc target genes, reduced tumor progression and conferred survival benefit in murine and zebrafish models of MYCN-driven neuroblastoma, and in a murine model of pancreatic cancer. Our results demonstrate that Sulfopin is a chemical probe suitable for assessment of Pin1-dependent pharmacology in cells and in vivo, and that Pin1 warrants further investigation as a potential cancer drug target..
Shern, J.F.
Selfe, J.
Izquierdo, E.
Patidar, R.
Chou, H.-.
Song, Y.K.
Yohe, M.E.
Sindiri, S.
Wei, J.
Wen, X.
Rudzinski, E.R.
Barkauskas, D.A.
Lo, T.
Hall, D.
Linardic, C.M.
Hughes, D.
Jamal, S.
Jenney, M.
Chisholm, J.
Brown, R.
Jones, K.
Hicks, B.
Angelini, P.
George, S.
Chesler, L.
Hubank, M.
Kelsey, A.
Gatz, S.A.
Skapek, S.X.
Hawkins, D.S.
Shipley, J.M.
Khan, J.
(2021). Genomic Classification and Clinical Outcome in Rhabdomyosarcoma: A Report From an International Consortium. J clin oncol,
Vol.39
(26),
pp. 2859-2871.
show abstract
full text
PURPOSE: Rhabdomyosarcoma is the most common soft tissue sarcoma of childhood. Despite aggressive therapy, the 5-year survival rate for patients with metastatic or recurrent disease remains poor, and beyond PAX-FOXO1 fusion status, no genomic markers are available for risk stratification. We present an international consortium study designed to determine the incidence of driver mutations and their association with clinical outcome. PATIENTS AND METHODS: Tumor samples collected from patients enrolled on Children's Oncology Group trials (1998-2017) and UK patients enrolled on malignant mesenchymal tumor and RMS2005 (1995-2016) trials were subjected to custom-capture sequencing. Mutations, indels, gene deletions, and amplifications were identified, and survival analysis was performed. RESULTS: DNA from 641 patients was suitable for analyses. A median of one mutation was found per tumor. In FOXO1 fusion-negative cases, mutation of any RAS pathway member was found in > 50% of cases, and 21% had no putative driver mutation identified. BCOR (15%), NF1 (15%), and TP53 (13%) mutations were found at a higher incidence than previously reported and TP53 mutations were associated with worse outcomes in both fusion-negative and FOXO1 fusion-positive cases. Interestingly, mutations in RAS isoforms predominated in infants < 1 year (64% of cases). Mutation of MYOD1 was associated with histologic patterns beyond those previously described, older age, head and neck primary site, and a dismal survival. Finally, we provide a searchable companion database (ClinOmics), containing all genomic variants, and clinical annotation including survival data. CONCLUSION: This is the largest genomic characterization of clinically annotated rhabdomyosarcoma tumors to date and provides prognostic genetic features that refine risk stratification and will be incorporated into prospective trials..
Tsakaneli, A.
Carregari, V.C.
Morini, M.
Eva, A.
Cangemi, G.
Chayka, O.
Makarov, E.
Bibbò, S.
Capone, E.
Sala, G.
De Laurenzi, V.
Poon, E.
Chesler, L.
Pieroni, L.
Larsen, M.R.
Palmisano, G.
Sala, A.
(2021). MYC regulates metabolism through vesicular transfer of glycolytic kinases. Open biol,
Vol.11
(12),
p. 210276.
show abstract
full text
Amplification of the proto-oncogene MYCN is a key molecular aberration in high-risk neuroblastoma and predictive of poor outcome in this childhood malignancy. We investigated the role of MYCN in regulating the protein cargo of extracellular vesicles (EVs) secreted by tumour cells that can be internalized by recipient cells with functional consequences. Using a switchable MYCN system coupled to mass spectrometry analysis, we found that MYCN regulates distinct sets of proteins in the EVs secreted by neuroblastoma cells. EVs produced by MYCN-expressing cells or isolated from neuroblastoma patients induced the Warburg effect, proliferation and c-MYC expression in target cells. Mechanistically, we linked the cancer-promoting activity of EVs to the glycolytic kinase pyruvate kinase M2 (PKM2) that was enriched in EVs secreted by MYC-expressing neuroblastoma cells. Importantly, the glycolytic enzymes PKM2 and hexokinase II were detected in the EVs circulating in the bloodstream of neuroblastoma patients, but not in those of non-cancer children. We conclude that MYC-activated cancers might spread oncogenic signals to remote body locations through EVs..
Bellini, A.
Pötschger, U.
Bernard, V.
Lapouble, E.
Baulande, S.
Ambros, P.F.
Auger, N.
Beiske, K.
Bernkopf, M.
Betts, D.R.
Bhalshankar, J.
Bown, N.
de Preter, K.
Clément, N.
Combaret, V.
Font de Mora, J.
George, S.L.
Jiménez, I.
Jeison, M.
Marques, B.
Martinsson, T.
Mazzocco, K.
Morini, M.
Mühlethaler-Mottet, A.
Noguera, R.
Pierron, G.
Rossing, M.
Taschner-Mandl, S.
Van Roy, N.
Vicha, A.
Chesler, L.
Balwierz, W.
Castel, V.
Elliott, M.
Kogner, P.
Laureys, G.
Luksch, R.
Malis, J.
Popovic-Beck, M.
Ash, S.
Delattre, O.
Valteau-Couanet, D.
Tweddle, D.A.
Ladenstein, R.
Schleiermacher, G.
(2021). Frequency and Prognostic Impact of ALK Amplifications and Mutations in the European Neuroblastoma Study Group (SIOPEN) High-Risk Neuroblastoma Trial (HR-NBL1). J clin oncol,
Vol.39
(30),
pp. 3377-3390.
show abstract
full text
PURPOSE: In neuroblastoma (NB), the ALK receptor tyrosine kinase can be constitutively activated through activating point mutations or genomic amplification. We studied ALK genetic alterations in high-risk (HR) patients on the HR-NBL1/SIOPEN trial to determine their frequency, correlation with clinical parameters, and prognostic impact. MATERIALS AND METHODS: Diagnostic tumor samples were available from 1,092 HR-NBL1/SIOPEN patients to determine ALK amplification status (n = 330), ALK mutational profile (n = 191), or both (n = 571). RESULTS: Genomic ALK amplification (ALKa) was detected in 4.5% of cases (41 out of 901), all except one with MYCN amplification (MNA). ALKa was associated with a significantly poorer overall survival (OS) (5-year OS: ALKa [n = 41] 28% [95% CI, 15 to 42]; no-ALKa [n = 860] 51% [95% CI, 47 to 54], [P < .001]), particularly in cases with metastatic disease. ALK mutations (ALKm) were detected at a clonal level (> 20% mutated allele fraction) in 10% of cases (76 out of 762) and at a subclonal level (mutated allele fraction 0.1%-20%) in 3.9% of patients (30 out of 762), with a strong correlation between the presence of ALKm and MNA (P < .001). Among 571 cases with known ALKa and ALKm status, a statistically significant difference in OS was observed between cases with ALKa or clonal ALKm versus subclonal ALKm or no ALK alterations (5-year OS: ALKa [n = 19], 26% [95% CI, 10 to 47], clonal ALKm [n = 65] 33% [95% CI, 21 to 44], subclonal ALKm (n = 22) 48% [95% CI, 26 to 67], and no alteration [n = 465], 51% [95% CI, 46 to 55], respectively; P = .001). Importantly, in a multivariate model, involvement of more than one metastatic compartment (hazard ratio [HR], 2.87; P < .001), ALKa (HR, 2.38; P = .004), and clonal ALKm (HR, 1.77; P = .001) were independent predictors of poor outcome. CONCLUSION: Genetic alterations of ALK (clonal mutations and amplifications) in HR-NB are independent predictors of poorer survival. These data provide a rationale for integration of ALK inhibitors in upfront treatment of HR-NB with ALK alterations..
Tucker, E.R.
George, S.
Angelini, P.
Bruna, A.
Chesler, L.
(2021). The Promise of Patient-Derived Preclinical Models to Accelerate the Implementation of Personalised Medicine for Children with Neuroblastoma. J pers med,
Vol.11
(4).
show abstract
full text
Patient-derived preclinical models are now a core component of cancer research and have the ability to drastically improve the predictive power of preclinical therapeutic studies. However, their development and maintenance can be challenging, time consuming, and expensive. For neuroblastoma, a developmental malignancy of the neural crest, it is possible to establish patient-derived models as xenografts in mice and zebrafish, and as spheroids and organoids in vitro. These varied approaches have contributed to comprehensive packages of preclinical evidence in support of new therapeutics for neuroblastoma. We discuss here the ethical and technical considerations for the creation of patient-derived models of neuroblastoma and how their use can be optimized for the study of tumour evolution and preclinical therapies. We also discuss how neuroblastoma patient-derived models might become avatars for personalised medicine for children with this devastating disease..
Shrestha, S.
Morcavallo, A.
Gorrini, C.
Chesler, L.
(2021). Biological Role of MYCN in Medulloblastoma: Novel Therapeutic Opportunities and Challenges Ahead. Front oncol,
Vol.11,
p. 694320.
show abstract
full text
The constitutive and dysregulated expression of the transcription factor MYCN has a central role in the pathogenesis of the paediatric brain tumour medulloblastoma, with an increased expression of this oncogene correlating with a worse prognosis. Consequently, the genomic and functional alterations of MYCN represent a major therapeutic target to attenuate tumour growth in medulloblastoma. This review will provide a comprehensive synopsis of the biological role of MYCN and its family components, their interaction with distinct signalling pathways, and the implications of this network in medulloblastoma development. We will then summarise the current toolbox for targeting MYCN and highlight novel therapeutic avenues that have the potential to results in better-tailored clinical treatments..
Turnock, S.
Turton, D.R.
Martins, C.D.
Chesler, L.
Wilson, T.C.
Gouverneur, V.
Smith, G.
Kramer-Marek, G.
(2020). 18F-meta-fluorobenzylguanidine (18F-mFBG) to monitor changes in norepinephrine transporter expression in response to therapeutic intervention in neuroblastoma models. Sci rep,
Vol.10
(1),
p. 20918.
show abstract
full text
Targeted radiotherapy with 131I-mIBG, a substrate of the human norepinephrine transporter (NET-1), shows promising responses in heavily pre-treated neuroblastoma (NB) patients. Combinatorial approaches that enhance 131I-mIBG tumour uptake are of substantial clinical interest but biomarkers of response are needed. Here, we investigate the potential of 18F-mFBG, a positron emission tomography (PET) analogue of the 123I-mIBG radiotracer, to quantify NET-1 expression levels in mouse models of NB following treatment with AZD2014, a dual mTOR inhibitor. The response to AZD2014 treatment was evaluated in MYCN amplified NB cell lines (Kelly and SK-N-BE(2)C) by Western blot (WB) and immunohistochemistry. PET quantification of 18F-mFBG uptake post-treatment in vivo was performed, and data correlated with NET-1 protein levels measured ex vivo. Following 72 h AZD2014 treatment, in vitro WB analysis indicated decreased mTOR signalling and enhanced NET-1 expression in both cell lines, and 18F-mFBG revealed a concentration-dependent increase in NET-1 function. AZD2014 treatment failed however to inhibit mTOR signalling in vivo and did not significantly modulate intratumoural NET-1 activity. Image analysis of 18F-mFBG PET data showed correlation to tumour NET-1 protein expression, while further studies are needed to elucidate whether NET-1 upregulation induced by blocking mTOR might be a useful adjunct to 131I-mIBG therapy..
Poon, E.
Liang, T.
Jamin, Y.
Walz, S.
Kwok, C.
Hakkert, A.
Barker, K.
Urban, Z.
Thway, K.
Zeid, R.
Hallsworth, A.
Box, G.
Ebus, M.E.
Licciardello, M.P.
Sbirkov, Y.
Lazaro, G.
Calton, E.
Costa, B.M.
Valenti, M.
De Haven Brandon, A.
Webber, H.
Tardif, N.
Almeida, G.S.
Christova, R.
Boysen, G.
Richards, M.W.
Barone, G.
Ford, A.
Bayliss, R.
Clarke, P.A.
De Bono, J.
Gray, N.S.
Blagg, J.
Robinson, S.P.
Eccles, S.A.
Zheleva, D.
Bradner, J.E.
Molenaar, J.
Vivanco, I.
Eilers, M.
Workman, P.
Lin, C.Y.
Chesler, L.
(2020). Orally bioavailable CDK9/2 inhibitor shows mechanism-based therapeutic potential in MYCN-driven neuroblastoma. J clin invest,
Vol.130
(11),
pp. 5875-5892.
show abstract
full text
The undruggable nature of oncogenic Myc transcription factors poses a therapeutic challenge in neuroblastoma, a pediatric cancer in which MYCN amplification is strongly associated with unfavorable outcome. Here, we show that CYC065 (fadraciclib), a clinical inhibitor of CDK9 and CDK2, selectively targeted MYCN-amplified neuroblastoma via multiple mechanisms. CDK9 - a component of the transcription elongation complex P-TEFb - bound to the MYCN-amplicon superenhancer, and its inhibition resulted in selective loss of nascent MYCN transcription. MYCN loss led to growth arrest, sensitizing cells for apoptosis following CDK2 inhibition. In MYCN-amplified neuroblastoma, MYCN invaded active enhancers, driving a transcriptionally encoded adrenergic gene expression program that was selectively reversed by CYC065. MYCN overexpression in mesenchymal neuroblastoma was sufficient to induce adrenergic identity and sensitize cells to CYC065. CYC065, used together with temozolomide, a reference therapy for relapsed neuroblastoma, caused long-term suppression of neuroblastoma growth in vivo, highlighting the clinical potential of CDK9/2 inhibition in the treatment of MYCN-amplified neuroblastoma..
Fultang, L.
Booth, S.
Yogev, O.
Martins da Costa, B.
Tubb, V.
Panetti, S.
Stavrou, V.
Scarpa, U.
Jankevics, A.
Lloyd, G.
Southam, A.
Lee, S.P.
Dunn, W.B.
Chesler, L.
Mussai, F.
De Santo, C.
(2020). Metabolic engineering against the arginine microenvironment enhances CAR-T cell proliferation and therapeutic activity. Blood,
Vol.136
(10),
pp. 1155-1160.
show abstract
Hematological and solid cancers catabolize the semiessential amino acid arginine to drive cell proliferation. However, the resulting low arginine microenvironment also impairs chimeric antigen receptor T cells (CAR-T) cell proliferation, limiting their efficacy in clinical trials against hematological and solid malignancies. T cells are susceptible to the low arginine microenvironment because of the low expression of the arginine resynthesis enzymes argininosuccinate synthase (ASS) and ornithine transcarbamylase (OTC). We demonstrate that T cells can be reengineered to express functional ASS or OTC enzymes, in concert with different chimeric antigen receptors. Enzyme modifications increase CAR-T cell proliferation, with no loss of CAR cytotoxicity or increased exhaustion. In vivo, enzyme-modified CAR-T cells lead to enhanced clearance of leukemia or solid tumor burden, providing the first metabolic modification to enhance CAR-T cell therapies..
George, S.L.
Parmar, V.
Lorenzi, F.
Marshall, L.V.
Jamin, Y.
Poon, E.
Angelini, P.
Chesler, L.
(2020). Novel therapeutic strategies targeting telomere maintenance mechanisms in high-risk neuroblastoma. J exp clin cancer res,
Vol.39
(1),
p. 78.
show abstract
full text
The majority of high-risk neuroblastomas can be divided into three distinct molecular subgroups defined by the presence of MYCN amplification, upstream TERT rearrangements or alternative lengthening of telomeres (ALT). The common defining feature of all three subgroups is altered telomere maintenance; MYCN amplification and upstream TERT rearrangements drive high levels of telomerase expression whereas ALT is a telomerase independent telomere maintenance mechanism. As all three telomere maintenance mechanisms are independently associated with poor outcomes, the development of strategies to selectively target either telomerase expressing or ALT cells holds great promise as a therapeutic approach that is applicable to the majority of children with aggressive disease.Here we summarise the biology of telomere maintenance and the molecular drivers of aggressive neuroblastoma before describing the most promising therapeutic strategies to target both telomerase expressing and ALT cancers. For telomerase-expressing neuroblastoma the most promising targeted agent to date is 6-thio-2'-deoxyguanosine, however clinical development of this agent is required. In osteosarcoma cell lines with ALT, selective sensitivity to ATR inhibition has been reported. However, we present data showing that in fact ALT neuroblastoma cells are more resistant to the clinical ATR inhibitor AZD6738 compared to other neuroblastoma subtypes. More recently a number of additional candidate compounds have been shown to show selectivity for ALT cancers, such as Tetra-Pt (bpy), a compound targeting the telomeric G-quadruplex and pifithrin-α, a putative p53 inhibitor. Further pre-clinical evaluation of these compounds in neuroblastoma models is warranted.In summary, telomere maintenance targeting strategies offer a significant opportunity to develop effective new therapies, applicable to a large proportion of children with high-risk neuroblastoma. In parallel to clinical development, more pre-clinical research specifically for neuroblastoma is urgently needed, if we are to improve survival for this common poor outcome tumour of childhood..
Moreno, L.
Barone, G.
DuBois, S.G.
Molenaar, J.
Fischer, M.
Schulte, J.
Eggert, A.
Schleiermacher, G.
Speleman, F.
Chesler, L.
Geoerger, B.
Hogarty, M.D.
Irwin, M.S.
Bird, N.
Blanchard, G.B.
Buckland, S.
Caron, H.
Davis, S.
De Wilde, B.
Deubzer, H.E.
Dolman, E.
Eilers, M.
George, R.E.
George, S.
Jaroslav, Š.
Maris, J.M.
Marshall, L.
Merchant, M.
Mortimer, P.
Owens, C.
Philpott, A.
Poon, E.
Shay, J.W.
Tonelli, R.
Valteau-Couanet, D.
Vassal, G.
Park, J.R.
Pearson, A.D.
(2020). Accelerating drug development for neuroblastoma: Summary of the Second Neuroblastoma Drug Development Strategy forum from Innovative Therapies for Children with Cancer and International Society of Paediatric Oncology Europe Neuroblastoma. Eur j cancer,
Vol.136,
pp. 52-68.
show abstract
full text
Only one class of targeted agents (anti-GD2 antibodies) has been incorporated into front-line therapy for neuroblastoma since the 1980s. The Neuroblastoma New Drug Development Strategy (NDDS) initiative commenced in 2012 to accelerate the development of new drugs for neuroblastoma. Advances have occurred, with eight of nine high-priority targets being evaluated in paediatric trials including anaplastic lymphoma kinase inhibitors being investigated in front-line, but significant challenges remain. This article reports the conclusions of the second NDDS forum, which expanded across the Atlantic to further develop the initiative. Pre-clinical and clinical data for 40 genetic targets and mechanisms of action were prioritised and drugs were identified for early-phase trials. Strategies to develop drugs targeting TERT, telomere maintenance, ATRX, alternative lengthening of telomeres (ALT), BRIP1 and RRM2 as well as direct targeting of MYCN are high priority and should be championed for drug discovery. Promising pre-clinical data suggest that targeting of ALT by ATM or PARP inhibition may be potential strategies. Drugs targeting CDK2/9, CDK7, ATR and telomere maintenance should enter paediatric clinical development rapidly. Optimising the response to anti-GD2 by combinations with chemotherapy, targeted agents and other immunological targets are crucial. Delivering this strategy in the face of small patient cohorts, genomically defined subpopulations and a large number of permutations of combination trials, demands even greater international collaboration. In conclusion, the NDDS provides an internationally agreed, biologically driven selection of prioritised genetic targets and drugs. Improvements in the strategy for conducting trials in neuroblastoma will accelerate bringing these new drugs more rapidly to front-line therapy..
George, S.L.
Lorenzi, F.
King, D.
Hartlieb, S.
Campbell, J.
Pemberton, H.
Toprak, U.H.
Barker, K.
Tall, J.
da Costa, B.M.
van den Boogaard, M.L.
Dolman, M.E.
Molenaar, J.J.
Bryant, H.E.
Westermann, F.
Lord, C.J.
Chesler, L.
(2020). Therapeutic vulnerabilities in the DNA damage response for the treatment of ATRX mutant neuroblastoma. Ebiomedicine,
Vol.59,
p. 102971.
show abstract
full text
BACKGROUND: In neuroblastoma, genetic alterations in ATRX, define a distinct poor outcome patient subgroup. Despite the need for new therapies, there is a lack of available models and a dearth of pre-clinical research. METHODS: To evaluate the impact of ATRX loss of function (LoF) in neuroblastoma, we utilized CRISPR-Cas9 gene editing to generate neuroblastoma cell lines isogenic for ATRX. We used these and other models to identify therapeutically exploitable synthetic lethal vulnerabilities associated with ATRX LoF. FINDINGS: In isogenic cell lines, we found that ATRX inactivation results in increased DNA damage, homologous recombination repair (HRR) defects and impaired replication fork processivity. In keeping with this, high-throughput compound screening showed selective sensitivity in ATRX mutant cells to multiple PARP inhibitors and the ATM inhibitor KU60019. ATRX mutant cells also showed selective sensitivity to the DNA damaging agents, sapacitabine and irinotecan. HRR deficiency was also seen in the ATRX deleted CHLA-90 cell line, and significant sensitivity demonstrated to olaparib/irinotecan combination therapy in all ATRX LoF models. In-vivo sensitivity to olaparib/irinotecan was seen in ATRX mutant but not wild-type xenografts. Finally, sustained responses to olaparib/irinotecan therapy were seen in an ATRX deleted neuroblastoma patient derived xenograft. INTERPRETATION: ATRX LoF results in specific DNA damage repair defects that can be therapeutically exploited. In ATRX LoF models, preclinical sensitivity is demonstrated to olaparib and irinotecan, a combination that can be rapidly translated into the clinic. FUNDING: This work was supported by Christopher's Smile, Neuroblastoma UK, Cancer Research UK, and the Royal Marsden Hospital NIHR BRC..
King, D.
Li, X.D.
Almeida, G.S.
Kwok, C.
Gravells, P.
Harrison, D.
Burke, S.
Hallsworth, A.
Jamin, Y.
George, S.
Robinson, S.P.
Lord, C.J.
Poon, E.
Yeomanson, D.
Chesler, L.
Bryant, H.E.
(2020). MYCN expression induces replication stress and sensitivity to PARP inhibition in neuroblastoma. Oncotarget,
Vol.11
(23),
pp. 2141-2159.
show abstract
full text
This study investigates the influence expression of the MYCN oncogene has on the DNA damage response, replication fork progression and sensitivity to PARP inhibition in neuroblastoma. In a panel of neuroblastoma cell lines, MYCN amplification or MYCN expression resulted in increased cell death in response to a range of PARP inhibitors (niraparib, veliparib, talazoparib and olaparib) compared to the response seen in non-expressing/amplified cells. MYCN expression slowed replication fork speed and increased replication fork stalling, an effect that was amplified by PARP inhibition or PARP1 depletion. Increased DNA damage seen was specifically induced in S-phase cells. Importantly, PARP inhibition caused a significant increase in the survival of mice bearing MYCN expressing tumours in a transgenic murine model of MYCN expressing neuroblastoma. Olaparib also sensitized MYCN expressing cells to camptothecin- and temozolomide-induced cell death to a greater degree than non-expressing cells. In summary, MYCN expression leads to increased replication stress in neuroblastoma cells. This effect is exaggerated by inhibition of PARP, resulting in S-phase specific DNA damage and ultimately increased tumour cell death. PARP inhibition alone or in combination with classical chemotherapeutics is therefore a potential therapeutic strategy for neuroblastoma and may be more effective in MYCN expressing tumours..
Caravagna, G.
Heide, T.
Williams, M.J.
Zapata, L.
Nichol, D.
Chkhaidze, K.
Cross, W.
Cresswell, G.D.
Werner, B.
Acar, A.
Chesler, L.
Barnes, C.P.
Sanguinetti, G.
Graham, T.A.
Sottoriva, A.
(2020). Subclonal reconstruction of tumors by using machine learning and population genetics. Nat genet,
Vol.52
(9),
pp. 898-907.
show abstract
full text
Most cancer genomic data are generated from bulk samples composed of mixtures of cancer subpopulations, as well as normal cells. Subclonal reconstruction methods based on machine learning aim to separate those subpopulations in a sample and infer their evolutionary history. However, current approaches are entirely data driven and agnostic to evolutionary theory. We demonstrate that systematic errors occur in the analysis if evolution is not accounted for, and this is exacerbated with multi-sampling of the same tumor. We present a novel approach for model-based tumor subclonal reconstruction, called MOBSTER, which combines machine learning with theoretical population genetics. Using public whole-genome sequencing data from 2,606 samples from different cohorts, new data and synthetic validation, we show that this method is more robust and accurate than current techniques in single-sample, multiregion and longitudinal data. This approach minimizes the confounding factors of nonevolutionary methods, thus leading to more accurate recovery of the evolutionary history of human cancers..
Zormpas-Petridis, K.
Poon, E.
Clarke, M.
Jerome, N.P.
Boult, J.K.
Blackledge, M.D.
Carceller, F.
Koers, A.
Barone, G.
Pearson, A.D.
Moreno, L.
Anderson, J.
Sebire, N.
McHugh, K.
Koh, D.-.
Chesler, L.
Yuan, Y.
Robinson, S.P.
Jamin, Y.
(2020). Noninvasive MRI Native T1 Mapping Detects Response to MYCN-targeted Therapies in the Th-MYCN Model of Neuroblastoma. Cancer res,
Vol.80
(16),
pp. 3424-3435.
show abstract
full text
Noninvasive early indicators of treatment response are crucial to the successful delivery of precision medicine in children with cancer. Neuroblastoma is a common solid tumor of young children that arises from anomalies in neural crest development. Therapeutic approaches aiming to destabilize MYCN protein, such as small-molecule inhibitors of Aurora A and mTOR, are currently being evaluated in early phase clinical trials in children with high-risk MYCN-driven disease, with limited ability to evaluate conventional pharmacodynamic biomarkers of response. T1 mapping is an MRI scan that measures the proton spin-lattice relaxation time T1. Using a multiparametric MRI-pathologic cross-correlative approach and computational pathology methodologies including a machine learning-based algorithm for the automatic detection and classification of neuroblasts, we show here that T1 mapping is sensitive to the rich histopathologic heterogeneity of neuroblastoma in the Th-MYCN transgenic model. Regions with high native T1 corresponded to regions dense in proliferative undifferentiated neuroblasts, whereas regions characterized by low T1 were rich in apoptotic or differentiating neuroblasts. Reductions in tumor-native T1 represented a sensitive biomarker of response to treatment-induced apoptosis with two MYCN-targeted small-molecule inhibitors, Aurora A kinase inhibitor alisertib (MLN8237) and mTOR inhibitor vistusertib (AZD2014). Overall, we demonstrate the potential of T1 mapping, a scan readily available on most clinical MRI scanners, to assess response to therapy and guide clinical trials for children with neuroblastoma. The study reinforces the potential role of MRI-based functional imaging in delivering precision medicine to children with neuroblastoma. SIGNIFICANCE: This study shows that MRI-based functional imaging can detect apoptotic responses to MYCN-targeted small-molecule inhibitors in a genetically engineered murine model of MYCN-driven neuroblastoma..
Franco-Luzón, L.
González-Murillo, Á.
Alcántara-Sánchez, C.
García-García, L.
Tabasi, M.
Huertas, A.L.
Chesler, L.
Ramírez, M.
(2020). Systemic oncolytic adenovirus delivered in mesenchymal carrier cells modulate tumor infiltrating immune cells and tumor microenvironment in mice with neuroblastoma. Oncotarget,
Vol.11
(4),
pp. 347-361.
show abstract
full text
Celyvir (autologous mesenchymal cells -MSCs- that carry an oncolytic adenovirus) is a new therapeutic strategy for metastatic tumors developed by our research group over the last decade. There are limitations for studying the immune effects of human oncolytic adenoviruses in murine models since these viruses do not replicate naturally in these animals. The use of xenografts in immunodeficient mice prevent assessing important clinical aspects of this therapy such as the antiadenoviral immune response or the possible intratumoral immune changes, both of tumor infiltrating leukocytes and of the microenvironment. In our strategy, the presence of MSCs in the medicinal product adds an extra level of complexity. We present here a murine model that overcomes many of these limitations. We found that carrier cells outcompeted intravenous administration of naked particles in delivering the oncolytic virus into the tumor masses. The protection that MSCs could provide to the oncolytic adenovirus did not preclude the development of an antiadenoviral immune response. However, the presence of circulating antiadenoviral antibodies did not prevent changes detected at the tumor masses: increased infiltration and changes in the quality of immune cells per unit of tumor volume, and a less protumoral and more inflammatory profile of the tumor microenvironment. We believe that the model described here will enable the study of crucial events related to the immune responses affecting both the medicinal product and the tumor..
Fultang, L.
Gamble, L.D.
Gneo, L.
Berry, A.M.
Egan, S.A.
De Bie, F.
Yogev, O.
Eden, G.L.
Booth, S.
Brownhill, S.
Vardon, A.
McConville, C.M.
Cheng, P.N.
Norris, M.D.
Etchevers, H.C.
Murray, J.
Ziegler, D.S.
Chesler, L.
Schmidt, R.
Burchill, S.A.
Haber, M.
De Santo, C.
Mussai, F.
(2019). Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma. Cancer res,
Vol.79
(3),
pp. 611-624.
show abstract
full text
Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)-dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1β and TNFα in a RAC-alpha serine/threonine-protein kinase (AKT)-dependent manner. IL1β and TNFα established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest-derived cells. Proteomic analysis revealed that enrichment of IL1β and TNFα in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited. SIGNIFICANCE: These findings illustrate that cross-talk between myeloid cells and tumor cells creates a metabolic regulatory loop that promotes neuroblastoma progression..
Zormpas-Petridis, K.
Jerome, N.P.
Blackledge, M.D.
Carceller, F.
Poon, E.
Clarke, M.
McErlean, C.M.
Barone, G.
Koers, A.
Vaidya, S.J.
Marshall, L.V.
Pearson, A.D.
Moreno, L.
Anderson, J.
Sebire, N.
McHugh, K.
Koh, D.-.
Yuan, Y.
Chesler, L.
Robinson, S.P.
Jamin, Y.
(2019). MRI Imaging of the Hemodynamic Vasculature of Neuroblastoma Predicts Response to Antiangiogenic Treatment. Cancer res,
Vol.79
(11),
pp. 2978-2991.
show abstract
full text
Childhood neuroblastoma is a hypervascular tumor of neural origin, for which antiangiogenic drugs are currently being evaluated; however, predictive biomarkers of treatment response, crucial for successful delivery of precision therapeutics, are lacking. We describe an MRI-pathologic cross-correlative approach using intrinsic susceptibility (IS) and susceptibility contrast (SC) MRI to noninvasively map the vascular phenotype in neuroblastoma Th-MYCN transgenic mice treated with the vascular endothelial growth factor receptor inhibitor cediranib. We showed that the transverse MRI relaxation rate R 2* (second-1) and fractional blood volume (fBV, %) were sensitive imaging biomarkers of hemorrhage and vascular density, respectively, and were also predictive biomarkers of response to cediranib. Comparison with MRI and pathology from patients with MYCN-amplified neuroblastoma confirmed the high degree to which the Th-MYCN model vascular phenotype recapitulated that of the clinical phenotype, thereby supporting further evaluation of IS- and SC-MRI in the clinic. This study reinforces the potential role of functional MRI in delivering precision medicine to children with neuroblastoma. SIGNIFICANCE: This study shows that functional MRI predicts response to vascular-targeted therapy in a genetically engineered murine model of neuroblastoma..
Carreno, G.
Boult, J.K.
Apps, J.
Gonzalez-Meljem, J.M.
Haston, S.
Guiho, R.
Stache, C.
Danielson, L.S.
Koers, A.
Smith, L.M.
Virasami, A.
Panousopoulos, L.
Buchfelder, M.
Jacques, T.S.
Chesler, L.
Robinson, S.P.
Martinez-Barbera, J.P.
(2019). SHH pathway inhibition is protumourigenic in adamantinomatous craniopharyngioma. Endocr relat cancer,
Vol.26
(3),
pp. 355-366.
show abstract
full text
Pharmacological inhibition of the sonic hedgehog (SHH) pathway can be beneficial against certain cancers but detrimental in others. Adamantinomatous craniopharyngioma (ACP) is a relevant pituitary tumour, affecting children and adults, that is associated with high morbidity and increased mortality in long-term follow-up. We have previously demonstrated overactivation of the SHH pathway in both human and mouse ACP. Here, we show that this activation is ligand dependent and induced by the expression of SHH protein in a small proportion of tumour cells. We investigate the functional relevance of SHH signalling in ACP through MRI-guided preclinical studies using an ACP mouse model. Treatment with vismodegib, a clinically approved SHH pathway inhibitor, results in a significant reduction in median survival due to premature development of highly proliferative and vascularised undifferentiated tumours. Reinforcing the mouse data, SHH pathway inhibition in human ACP leads to a significant increase in tumour cell proliferation both ex vivo, in explant cultures, and in vivo, in a patient-derived xenograft model. Together, our results demonstrate a protumourigenic effect of vismodegib-mediated SHH pathway inhibition in ACP..
Aldape, K.
Brindle, K.M.
Chesler, L.
Chopra, R.
Gajjar, A.
Gilbert, M.R.
Gottardo, N.
Gutmann, D.H.
Hargrave, D.
Holland, E.C.
Jones, D.T.
Joyce, J.A.
Kearns, P.
Kieran, M.W.
Mellinghoff, I.K.
Merchant, M.
Pfister, S.M.
Pollard, S.M.
Ramaswamy, V.
Rich, J.N.
Robinson, G.W.
Rowitch, D.H.
Sampson, J.H.
Taylor, M.D.
Workman, P.
Gilbertson, R.J.
(2019). Challenges to curing primary brain tumours. Nat rev clin oncol,
Vol.16
(8),
pp. 509-520.
show abstract
full text
Despite decades of research, brain tumours remain among the deadliest of all forms of cancer. The ability of these tumours to resist almost all conventional and novel treatments relates, in part, to the unique cell-intrinsic and microenvironmental properties of neural tissues. In an attempt to encourage progress in our understanding and ability to successfully treat patients with brain tumours, Cancer Research UK convened an international panel of clinicians and laboratory-based scientists to identify challenges that must be overcome if we are to cure all patients with a brain tumour. The seven key challenges summarized in this Position Paper are intended to serve as foci for future research and investment..
George, S.L.
Izquierdo, E.
Campbell, J.
Koutroumanidou, E.
Proszek, P.
Jamal, S.
Hughes, D.
Yuan, L.
Marshall, L.V.
Carceller, F.
Chisholm, J.C.
Vaidya, S.
Mandeville, H.
Angelini, P.
Wasti, A.
Bexelius, T.
Thway, K.
Gatz, S.A.
Clarke, M.
Al-Lazikani, B.
Barone, G.
Anderson, J.
Tweddle, D.A.
Gonzalez, D.
Walker, B.A.
Barton, J.
Depani, S.
Eze, J.
Ahmed, S.W.
Moreno, L.
Pearson, A.
Shipley, J.
Jones, C.
Hargrave, D.
Jacques, T.S.
Hubank, M.
Chesler, L.
(2019). A tailored molecular profiling programme for children with cancer to identify clinically actionable genetic alterations. Eur j cancer,
Vol.121,
pp. 224-235.
show abstract
full text
BACKGROUND: For children with cancer, the clinical integration of precision medicine to enable predictive biomarker-based therapeutic stratification is urgently needed. METHODS: We have developed a hybrid-capture next-generation sequencing (NGS) panel, specifically designed to detect genetic alterations in paediatric solid tumours, which gives reliable results from as little as 50 ng of DNA extracted from formalin-fixed paraffin-embedded (FFPE) tissue. In this study, we offered an NGS panel, with clinical reporting via a molecular tumour board for children with solid tumours. Furthermore, for a cohort of 12 patients, we used a circulating tumour DNA (ctDNA)-specific panel to sequence ctDNA from matched plasma samples and compared plasma and tumour findings. RESULTS: A total of 255 samples were submitted from 223 patients for the NGS panel. Using FFPE tissue, 82% of all submitted samples passed quality control for clinical reporting. At least one genetic alteration was detected in 70% of sequenced samples. The overall detection rate of clinically actionable alterations, defined by modified OncoKB criteria, for all sequenced samples was 51%. A total of 8 patients were sequenced at different stages of treatment. In 6 of these, there were differences in the genetic alterations detected between time points. Sequencing of matched ctDNA in a cohort of extracranial paediatric solid tumours also identified a high detection rate of somatic alterations in plasma. CONCLUSION: We demonstrate that tailored clinical molecular profiling of both tumour DNA and plasma-derived ctDNA is feasible for children with solid tumours. Furthermore, we show that a targeted NGS panel-based approach can identify actionable genetic alterations in a high proportion of patients..
Watts, E.
Heidenreich, D.
Tucker, E.
Raab, M.
Strebhardt, K.
Chesler, L.
Knapp, S.
Bellenie, B.
Hoelder, S.
(2019). Designing Dual Inhibitors of Anaplastic Lymphoma Kinase (ALK) and Bromodomain-4 (BRD4) by Tuning Kinase Selectivity. J med chem,
Vol.62
(5),
pp. 2618-2637.
show abstract
full text
Concomitant inhibition of anaplastic lymphoma kinase (ALK) and bromodomain-4 (BRD4) is a potential therapeutic strategy for targeting two key oncogenic drivers that co-segregate in a significant fraction of high-risk neuroblastoma patients, mutation of ALK and amplification of MYCN. Starting from known dual polo-like kinase (PLK)-1-BRD4 inhibitor BI-2536, we employed structure-based design to redesign this series toward compounds with a dual ALK-BRD4 profile. These efforts led to compound ( R)-2-((2-ethoxy-4-(1-methylpiperidin-4-yl)phenyl)amino)-7-ethyl-5-methyl-8-((4-methylthiophen-2-yl)methyl)-7,8-dihydropteridin-6(5 H)-one (16k) demonstrating improved ALK activity and significantly reduced PLK-1 activity, while maintaining BRD4 activity and overall kinome selectivity. We demonstrate the compounds' on-target engagement with ALK and BRD4 in cells as well as favorable broad kinase and bromodomain selectivity..
Yogev, O.
Almeida, G.S.
Barker, K.T.
George, S.L.
Kwok, C.
Campbell, J.
Zarowiecki, M.
Kleftogiannis, D.
Smith, L.M.
Hallsworth, A.
Berry, P.
Möcklinghoff, T.
Webber, H.T.
Danielson, L.S.
Buttery, B.
Calton, E.A.
da Costa, B.M.
Poon, E.
Jamin, Y.
Lise, S.
Veal, G.J.
Sebire, N.
Robinson, S.P.
Anderson, J.
Chesler, L.
(2019). In Vivo Modeling of Chemoresistant Neuroblastoma Provides New Insights into Chemorefractory Disease and Metastasis. Cancer res,
Vol.79
(20),
pp. 5382-5393.
show abstract
full text
Neuroblastoma is a pediatric cancer that is frequently metastatic and resistant to conventional treatment. In part, a lack of natively metastatic, chemoresistant in vivo models has limited our insight into the development of aggressive disease. The Th-MYCN genetically engineered mouse model develops rapidly progressive chemosensitive neuroblastoma and lacks clinically relevant metastases. To study tumor progression in a context more reflective of clinical therapy, we delivered multicycle treatment with cyclophosphamide to Th-MYCN mice, individualizing therapy using MRI, to generate the Th-MYCN CPM32 model. These mice developed chemoresistance and spontaneous bone marrow metastases. Tumors exhibited an altered immune microenvironment with increased stroma and tumor-associated fibroblasts. Analysis of copy number aberrations revealed genomic changes characteristic of human MYCN-amplified neuroblastoma, specifically copy number gains at mouse chromosome 11, syntenic with gains on human chromosome 17q. RNA sequencing revealed enriched expression of genes associated with 17q gain and upregulation of genes associated with high-risk neuroblastoma, such as the cell-cycle regulator cyclin B1-interacting protein 1 (Ccnb1ip1) and thymidine kinase (TK1). The antiapoptotic, prometastatic JAK-STAT3 pathway was activated in chemoresistant tumors, and treatment with the JAK1/JAK2 inhibitor CYT387 reduced progression of chemoresistant tumors and increased survival. Our results highlight that under treatment conditions that mimic chemotherapy in human patients, Th-MYCN mice develop genomic, microenvironmental, and clinical features reminiscent of human chemorefractory disease. The Th-MYCN CPM32 model therefore is a useful tool to dissect in detail mechanisms that drive metastasis and chemoresistance, and highlights dysregulation of signaling pathways such as JAK-STAT3 that could be targeted to improve treatment of aggressive disease. SIGNIFICANCE: An in vivo mouse model of high-risk treatment-resistant neuroblastoma exhibits changes in the tumor microenvironment, widespread metastases, and sensitivity to JAK1/2 inhibition..
Li, J.
Zormpas-Petridis, K.
Boult, J.K.
Reeves, E.L.
Heindl, A.
Vinci, M.
Lopes, F.
Cummings, C.
Springer, C.J.
Chesler, L.
Jones, C.
Bamber, J.C.
Yuan, Y.
Sinkus, R.
Jamin, Y.
Robinson, S.P.
(2019). Investigating the Contribution of Collagen to the Tumor Biomechanical Phenotype with Noninvasive Magnetic Resonance Elastography. Cancer res,
Vol.79
(22),
pp. 5874-5883.
show abstract
full text
Increased stiffness in the extracellular matrix (ECM) contributes to tumor progression and metastasis. Therefore, stromal modulating therapies and accompanying biomarkers are being developed to target ECM stiffness. Magnetic resonance (MR) elastography can noninvasively and quantitatively map the viscoelastic properties of tumors in vivo and thus has clear clinical applications. Herein, we used MR elastography, coupled with computational histopathology, to interrogate the contribution of collagen to the tumor biomechanical phenotype and to evaluate its sensitivity to collagenase-induced stromal modulation. Elasticity (G d) and viscosity (G l) were significantly greater for orthotopic BT-474 (G d = 5.9 ± 0.2 kPa, G l = 4.7 ± 0.2 kPa, n = 7) and luc-MDA-MB-231-LM2-4 (G d = 7.9 ± 0.4 kPa, G l = 6.0 ± 0.2 kPa, n = 6) breast cancer xenografts, and luc-PANC1 (G d = 6.9 ± 0.3 kPa, G l = 6.2 ± 0.2 kPa, n = 7) pancreatic cancer xenografts, compared with tumors associated with the nervous system, including GTML/Trp53KI/KI medulloblastoma (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 7), orthotopic luc-D-212-MG (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 7), luc-RG2 (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 5), and luc-U-87-MG (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 8) glioblastoma xenografts, intracranially propagated luc-MDA-MB-231-LM2-4 (G d = 3.7 ± 0.2 kPa, G l = 2.2 ± 0.1 kPa, n = 7) breast cancer xenografts, and Th-MYCN neuroblastomas (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 5). Positive correlations between both elasticity (r = 0.72, P < 0.0001) and viscosity (r = 0.78, P < 0.0001) were determined with collagen fraction, but not with cellular or vascular density. Treatment with collagenase significantly reduced G d (P = 0.002) and G l (P = 0.0006) in orthotopic breast tumors. Texture analysis of extracted images of picrosirius red staining revealed significant negative correlations of entropy with G d (r = -0.69, P < 0.0001) and G l (r = -0.76, P < 0.0001), and positive correlations of fractal dimension with G d (r = 0.75, P < 0.0001) and G l (r = 0.78, P < 0.0001). MR elastography can thus provide sensitive imaging biomarkers of tumor collagen deposition and its therapeutic modulation. SIGNIFICANCE: MR elastography enables noninvasive detection of tumor stiffness and will aid in the development of ECM-targeting therapies..
Tucker, E.R.
Poon, E.
Chesler, L.
(2019). Targeting MYCN and ALK in resistant and relapsing neuroblastoma. Cancer drug resist,
Vol.2
(3),
pp. 803-812.
show abstract
Neuroblastoma, a tumor of peripheral nerve, is the most common solid tumor of young children. In high-risk disease, which comprises approximately half of patients, death from chemotherapy-resistant, metastatic relapse is very frequent. Children who relapse exhibit clonal enrichment of two genomic alterations: high-level amplification of the MYCN oncogene, and kinase domain mutations of the anaplastic lymphoma kinase (ALK) gene. Overall survival in this patient cohort is less than 15% at 3 years, and there are few options for rationally targeted therapy. Neuroblastoma patients exhibit de novo resistance to many existing ALK inhibitors, and no clinical therapeutics to target MYCN have yet been developed. This review outlines the international efforts to uncover mechanisms of oncogenic action that are therapeutically targetable using small-molecule inhibitors. We describe a mechanistic interaction in which ALK upregulates MYCN transcription, and discuss clinical trials emerging to develop transcriptional inhibitors of MYCN, and to identify effective inhibitors of ALK in neuroblastoma patients..
Ferrucci, V.
de Antonellis, P.
Pennino, F.P.
Asadzadeh, F.
Virgilio, A.
Montanaro, D.
Galeone, A.
Boffa, I.
Pisano, I.
Scognamiglio, I.
Navas, L.
Diana, D.
Pedone, E.
Gargiulo, S.
Gramanzini, M.
Brunetti, A.
Danielson, L.
Carotenuto, M.
Liguori, L.
Verrico, A.
Quaglietta, L.
Errico, M.E.
Del Monaco, V.
D'Argenio, V.
Tirone, F.
Mastronuzzi, A.
Donofrio, V.
Giangaspero, F.
Picard, D.
Remke, M.
Garzia, L.
Daniels, C.
Delattre, O.
Swartling, F.J.
Weiss, W.A.
Salvatore, F.
Fattorusso, R.
Chesler, L.
Taylor, M.D.
Cinalli, G.
Zollo, M.
(2018). Metastatic group 3 medulloblastoma is driven by PRUNE1 targeting NME1-TGF-β-OTX2-SNAIL via PTEN inhibition. Brain,
Vol.141
(5),
pp. 1300-1319.
show abstract
Genetic modifications during development of paediatric groups 3 and 4 medulloblastoma are responsible for their highly metastatic properties and poor patient survival rates. PRUNE1 is highly expressed in metastatic medulloblastoma group 3, which is characterized by TGF-β signalling activation, c-MYC amplification, and OTX2 expression. We describe the process of activation of the PRUNE1 signalling pathway that includes its binding to NME1, TGF-β activation, OTX2 upregulation, SNAIL (SNAI1) upregulation, and PTEN inhibition. The newly identified small molecule pyrimido-pyrimidine derivative AA7.1 enhances PRUNE1 degradation, inhibits this activation network, and augments PTEN expression. Both AA7.1 and a competitive permeable peptide that impairs PRUNE1/NME1 complex formation, impair tumour growth and metastatic dissemination in orthotopic xenograft models with a metastatic medulloblastoma group 3 cell line (D425-Med cells). Using whole exome sequencing technology in metastatic medulloblastoma primary tumour cells, we also define 23 common 'non-synonymous homozygous' deleterious gene variants as part of the protein molecular network of relevance for metastatic processes. This PRUNE1/TGF-β/OTX2/PTEN axis, together with the medulloblastoma-driver mutations, is of relevance for future rational and targeted therapies for metastatic medulloblastoma group 3.10.1093/brain/awy039_video1awy039media15742053534001..
Zeid, R.
Lawlor, M.A.
Poon, E.
Reyes, J.M.
Fulciniti, M.
Lopez, M.A.
Scott, T.G.
Nabet, B.
Erb, M.A.
Winter, G.E.
Jacobson, Z.
Polaski, D.R.
Karlin, K.L.
Hirsch, R.A.
Munshi, N.P.
Westbrook, T.F.
Chesler, L.
Lin, C.Y.
Bradner, J.E.
(2018). Enhancer invasion shapes MYCN-dependent transcriptional amplification in neuroblastoma. Nat genet,
Vol.50
(4),
pp. 515-523.
show abstract
full text
Amplification of the locus encoding the oncogenic transcription factor MYCN is a defining feature of high-risk neuroblastoma. Here we present the first dynamic chromatin and transcriptional landscape of MYCN perturbation in neuroblastoma. At oncogenic levels, MYCN associates with E-box binding motifs in an affinity-dependent manner, binding to strong canonical E-boxes at promoters and invading abundant weaker non-canonical E-boxes clustered at enhancers. Loss of MYCN leads to a global reduction in transcription, which is most pronounced at MYCN target genes with the greatest enhancer occupancy. These highly occupied MYCN target genes show tissue-specific expression and are linked to poor patient survival. The activity of genes with MYCN-occupied enhancers is dependent on the tissue-specific transcription factor TWIST1, which co-occupies enhancers with MYCN and is required for MYCN-dependent proliferation. These data implicate tissue-specific enhancers in defining often highly tumor-specific 'MYC target gene signatures' and identify disruption of the MYCN enhancer regulatory axis as a promising therapeutic strategy in neuroblastoma..
Boult, J.K.
Apps, J.R.
Hölsken, A.
Hutchinson, J.C.
Carreno, G.
Danielson, L.S.
Smith, L.M.
Bäuerle, T.
Buslei, R.
Buchfelder, M.
Virasami, A.K.
Koers, A.
Arthurs, O.J.
Jacques, T.S.
Chesler, L.
Martinez-Barbera, J.P.
Robinson, S.P.
(2018). Preclinical transgenic and patient-derived xenograft models recapitulate the radiological features of human adamantinomatous craniopharyngioma. Brain pathol,
Vol.28
(4),
pp. 475-483.
show abstract
full text
To assess the clinical relevance of transgenic and patient-derived xenograft models of adamantinomatous craniopharyngioma (ACP) using serial magnetic resonance imaging (MRI) and high resolution post-mortem microcomputed tomography (μ-CT), with correlation with histology and human ACP imaging. The growth patterns and radiological features of tumors arising in Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ transgenic mice, and of patient-derived ACP xenografts implanted in the cerebral cortex, were monitored longitudinally in vivo with anatomical and functional MRI, and by ex vivo μ-CT at study end. Pathological correlates with hematoxylin and eosin stained sections were investigated. Early enlargement and heterogeneity of Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ mouse pituitaries was evident at initial imaging at 8 weeks, which was followed by enlargement of a solid tumor, and development of cysts and hemorrhage. Tumors demonstrated MRI features that recapitulated those of human ACP, specifically, T1 -weighted signal enhancement in the solid tumor component following Gd-DTPA administration, and in some animals, hyperintense cysts on FLAIR and T1 -weighted images. Ex vivo μ-CT correlated with MRI findings and identified smaller cysts, which were confirmed by histology. Characteristic histological features, including wet keratin and calcification, were visible on μ-CT and verified by histological sections of patient-derived ACP xenografts. The Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ transgenic mouse model and cerebral patient-derived ACP xenografts recapitulate a number of the key radiological features of the human disease and provide promising foundations for in vivo trials of novel therapeutics for the treatment of these tumors..
Gonzalez Malagon, S.G.
Lopez Muñoz, A.M.
Doro, D.
Bolger, T.G.
Poon, E.
Tucker, E.R.
Adel Al-Lami, H.
Krause, M.
Phiel, C.J.
Chesler, L.
Liu, K.J.
(2018). Glycogen synthase kinase 3 controls migration of the neural crest lineage in mouse and Xenopus. Nat commun,
Vol.9
(1),
p. 1126.
show abstract
full text
Neural crest migration is critical to its physiological function. Mechanisms controlling mammalian neural crest migration are comparatively unknown, due to difficulties accessing this cell population in vivo. Here we report requirements of glycogen synthase kinase 3 (GSK3) in regulating the neural crest in Xenopus and mouse models. We demonstrate that GSK3 is tyrosine phosphorylated (pY) in mouse neural crest cells and that loss of GSK3 leads to increased pFAK and misregulation of Rac1 and lamellipodin, key regulators of cell migration. Genetic reduction of GSK3 results in failure of migration. We find that pY-GSK3 phosphorylation depends on anaplastic lymphoma kinase (ALK), a protein associated with neuroblastoma. Consistent with this, neuroblastoma cells with increased ALK activity express high levels of pY-GSK3, and blockade of GSK3 or ALK can affect migration of these cells. Altogether, this work identifies a role for GSK3 in cell migration during neural crest development and cancer..
Almeida, G.S.
Panek, R.
Hallsworth, A.
Webber, H.
Papaevangelou, E.
Boult, J.K.
Jamin, Y.
Chesler, L.
Robinson, S.P.
(2017). Pre-clinical imaging of transgenic mouse models of neuroblastoma using a dedicated 3-element solenoid coil on a clinical 3T platform. Br j cancer,
Vol.117
(6),
pp. 791-800.
show abstract
full text
BACKGROUND: The use of clinical MRI scanners to conduct pre-clinical research facilitates comparisons with clinical studies. Here the utility and sensitivity of anatomical and functional MRI data/biomarkers acquired from transgenic mouse models of neuroblastoma using a dedicated radiofrequency (RF) coil on a clinical 3T scanner was evaluated. METHODS: Multiparametric MRI of transgenic mice bearing abdominal neuroblastomas was performed at 3T, and data cross-referenced to that acquired from the same mice on a pre-clinical 7T MRI system. T2-weighted imaging, quantitation of the native longitudinal relaxation time (T1) and the transverse relaxation rate (R2*), and dynamic contrast-enhanced (DCE)-MRI, was used to assess tumour volume, phenotype and response to cyclophosphamide or cabozantinib. RESULTS: Excellent T2-weighted image contrast enabled clear tumour delineation at 3T. Significant correlations of tumour volume (R=0.98, P<0.0001) and R2* (R=0.87, P<0.002) measured at 3 and 7T were established. Mice with neuroblastomas harbouring the anaplastic lymphoma kinase mutation exhibited a significantly slower R2* (P<0.001), consistent with impaired tumour perfusion. DCE-MRI was performed simultaneously on three transgenic mice, yielding estimates of Ktrans for each tumour (median Ktrans values of 0.202, 0.168 and 0.114 min-1). Cyclophosphamide elicited a significant reduction in both tumour burden (P<0.002) and native T1 (P<0.01), whereas cabozantinib induced significant (P<0.01) tumour growth delay. CONCLUSIONS: Simultaneous multiparametric MRI of multiple tumour-bearing animals using this coil arrangement at 3T can provide high efficiency/throughput for both phenotypic characterisation and evaluation of novel therapeutics, and facilitate the introduction of functional MRI biomarkers into aligned imaging-embedded clinical trials..
Pearson, A.D.
Pfister, S.M.
Baruchel, A.
Bourquin, J.-.
Casanova, M.
Chesler, L.
Doz, F.
Eggert, A.
Geoerger, B.
Jones, D.T.
Kearns, P.R.
Molenaar, J.J.
Morland, B.
Schleiermacher, G.
Schulte, J.H.
Vormoor, J.
Marshall, L.V.
Zwaan, C.M.
Vassal, G.
Executive and Biology Committees of the Innovative Therapies for Children with Cancer European Consortium,
(2017). From class waivers to precision medicine in paediatric oncology. Lancet oncol,
Vol.18
(7),
pp. e394-e404.
show abstract
full text
New drugs are crucially needed for children with cancer. The European Paediatric Regulation facilitates paediatric class waivers for drugs developed for diseases only occurring in adults. In this Review, we retrospectively searched oncology drugs that were class waivered between June, 2012, and June, 2015. 147 oncology class waivers were confirmed for 89 drugs. Mechanisms of action were then assessed as potential paediatric therapeutic targets by both a literature search and an expert review. 48 (54%) of the 89 class-waivered drugs had a mechanisms of action warranting paediatric development. Two (2%) class-waivered drugs were considered not relevant and 16 (18%) required further data. In light of these results, we propose five initiatives: an aggregated database of paediatric biological tumour drug targets; molecular profiling of all paediatric tumours at diagnosis and relapse; a joint academic-pharmaceutical industry preclinical platform to help analyse the activity of new drugs (Innovative Therapy for Children with Cancer Paediatric Preclinical Proof-of-Concept Platform); paediatric strategy forums; and the suppression of article 11b of the European Paediatric Regulation, which allows product-specific waivers on the grounds that the associated condition does not occur in children. These initiatives and a mechanism of action-based approach to drug development will accelerate the delivery of new therapeutic drugs for front-line therapy for those children who have unmet medical needs..
Tucker, E.R.
Tall, J.R.
Danielson, L.S.
Gowan, S.
Jamin, Y.
Robinson, S.P.
Banerji, U.
Chesler, L.
(2017). Immunoassays for the quantification of ALK and phosphorylated ALK support the evaluation of on-target ALK inhibitors in neuroblastoma. Mol oncol,
Vol.11
(8),
pp. 996-1006.
show abstract
full text
Targeted inhibition of anaplastic lymphoma kinase (ALK) is a successful approach for the treatment of many ALK-aberrant malignancies; however, the presence of resistant mutations necessitates both the development of more potent compounds and pharmacodynamic methods with which to determine their efficacy. We describe immunoassays designed to quantitate phosphorylation of ALK, and their use in preclinical models of neuroblastoma, a pediatric malignancy in which gain-of-function ALK mutations predict a poor overall outcome to conventional treatment. Validation of the immunoassays is presented using a panel of neuroblastoma cell lines and evidence of on-target ALK inhibition provided by treatment of a genetically engineered murine model of neuroblastoma with two clinical ALK inhibitors, crizotinib and ceritinib, highlighting the superior efficacy of ceritinib..
Moreno, L.
Caron, H.
Geoerger, B.
Eggert, A.
Schleiermacher, G.
Brock, P.
Valteau-Couanet, D.
Chesler, L.
Schulte, J.H.
De Preter, K.
Molenaar, J.
Schramm, A.
Eilers, M.
Van Maerken, T.
Johnsen, J.I.
Garrett, M.
George, S.L.
Tweddle, D.A.
Kogner, P.
Berthold, F.
Koster, J.
Barone, G.
Tucker, E.R.
Marshall, L.
Herold, R.
Sterba, J.
Norga, K.
Vassal, G.
Pearson, A.D.
(2017). Accelerating drug development for neuroblastoma - New Drug Development Strategy: an Innovative Therapies for Children with Cancer, European Network for Cancer Research in Children and Adolescents and International Society of Paediatric Oncology Europe Neuroblastoma project. Expert opin drug discov,
Vol.12
(8),
pp. 801-811.
show abstract
full text
Neuroblastoma, the commonest paediatric extra-cranial tumour, remains a leading cause of death from cancer in children. There is an urgent need to develop new drugs to improve cure rates and reduce long-term toxicity and to incorporate molecularly targeted therapies into treatment. Many potential drugs are becoming available, but have to be prioritised for clinical trials due to the relatively small numbers of patients. Areas covered: The current drug development model has been slow, associated with significant attrition, and few new drugs have been developed for neuroblastoma. The Neuroblastoma New Drug Development Strategy (NDDS) has: 1) established a group with expertise in drug development; 2) prioritised targets and drugs according to tumour biology (target expression, dependency, pre-clinical data; potential combinations; biomarkers), identifying as priority targets ALK, MEK, CDK4/6, MDM2, MYCN (druggable by BET bromodomain, aurora kinase, mTORC1/2) BIRC5 and checkpoint kinase 1; 3) promoted clinical trials with target-prioritised drugs. Drugs showing activity can be rapidly transitioned via parallel randomised trials into front-line studies. Expert opinion: The Neuroblastoma NDDS is based on the premise that optimal drug development is reliant on knowledge of tumour biology and prioritisation. This approach will accelerate neuroblastoma drug development and other poor prognosis childhood malignancies..
Büchel, G.
Carstensen, A.
Mak, K.-.
Roeschert, I.
Leen, E.
Sumara, O.
Hofstetter, J.
Herold, S.
Kalb, J.
Baluapuri, A.
Poon, E.
Kwok, C.
Chesler, L.
Maric, H.M.
Rickman, D.S.
Wolf, E.
Bayliss, R.
Walz, S.
Eilers, M.
(2017). Association with Aurora-A Controls N-MYC-Dependent Promoter Escape and Pause Release of RNA Polymerase II during the Cell Cycle. Cell rep,
Vol.21
(12),
pp. 3483-3497.
show abstract
full text
MYC proteins bind globally to active promoters and promote transcriptional elongation by RNA polymerase II (Pol II). To identify effector proteins that mediate this function, we performed mass spectrometry on N-MYC complexes in neuroblastoma cells. The analysis shows that N-MYC forms complexes with TFIIIC, TOP2A, and RAD21, a subunit of cohesin. N-MYC and TFIIIC bind to overlapping sites in thousands of Pol II promoters and intergenic regions. TFIIIC promotes association of RAD21 with N-MYC target sites and is required for N-MYC-dependent promoter escape and pause release of Pol II. Aurora-A competes with binding of TFIIIC and RAD21 to N-MYC in vitro and antagonizes association of TOP2A, TFIIIC, and RAD21 with N-MYC during S phase, blocking N-MYC-dependent release of Pol II from the promoter. Inhibition of Aurora-A in S phase restores RAD21 and TFIIIC binding to chromatin and partially restores N-MYC-dependent transcriptional elongation. We propose that complex formation with Aurora-A controls N-MYC function during the cell cycle..
Izquierdo, E.
Yuan, L.
George, S.
Hubank, M.
Jones, C.
Proszek, P.
Shipley, J.
Gatz, S.A.
Stinson, C.
Moore, A.S.
Clifford, S.C.
Hicks, D.
Lindsey, J.C.
Hill, R.M.
Jacques, T.S.
Chalker, J.
Thway, K.
O'Connor, S.
Marshall, L.
Moreno, L.
Pearson, A.
Chesler, L.
Walker, B.A.
De Castro, D.G.
(2017). Development of a targeted sequencing approach to identify prognostic, predictive and diagnostic markers in paediatric solid tumours. Oncotarget,
Vol.8
(67),
pp. 112036-112050.
show abstract
full text
The implementation of personalised medicine in childhood cancers has been limited by a lack of clinically validated multi-target sequencing approaches specific for paediatric solid tumours. In order to support innovative clinical trials in high-risk patients with unmet need, we have developed a clinically relevant targeted sequencing panel spanning 311 kb and comprising 78 genes involved in childhood cancers. A total of 132 samples were used for the validation of the panel, including Horizon Discovery cell blends (n=4), cell lines (n=15), formalin-fixed paraffin embedded (FFPE, n=83) and fresh frozen tissue (FF, n=30) patient samples. Cell blends containing known single nucleotide variants (SNVs, n=528) and small insertion-deletions (indels n=108) were used to define panel sensitivities of ≥98% for SNVs and ≥83% for indels [95% CI] and panel specificity of ≥98% [95% CI] for SNVs. FFPE samples performed comparably to FF samples (n=15 paired). Of 95 well-characterised genetic abnormalities in 33 clinical specimens and 13 cell lines (including SNVs, indels, amplifications, rearrangements and chromosome losses), 94 (98.9%) were detected by our approach. We have validated a robust and practical methodology to guide clinical management of children with solid tumours based on their molecular profiles. Our work demonstrates the value of targeted gene sequencing in the development of precision medicine strategies in paediatric oncology..
Guan, J.
Tucker, E.R.
Wan, H.
Chand, D.
Danielson, L.S.
Ruuth, K.
El Wakil, A.
Witek, B.
Jamin, Y.
Umapathy, G.
Robinson, S.P.
Johnson, T.W.
Smeal, T.
Martinsson, T.
Chesler, L.
Palmer, R.H.
Hallberg, B.
(2016). The ALK inhibitor PF-06463922 is effective as a single agent in neuroblastoma driven by expression of ALK and MYCN. Dis model mech,
Vol.9
(9),
pp. 941-952.
show abstract
full text
The first-in-class inhibitor of ALK, c-MET and ROS1, crizotinib (Xalkori), has shown remarkable clinical efficacy in treatment of ALK-positive non-small cell lung cancer. However, in neuroblastoma, activating mutations in the ALK kinase domain are typically refractory to crizotinib treatment, highlighting the need for more potent inhibitors. The next-generation ALK inhibitor PF-06463922 is predicted to exhibit increased affinity for ALK mutants prevalent in neuroblastoma. We examined PF-06463922 activity in ALK-driven neuroblastoma models in vitro and in vivo In vitro kinase assays and cell-based experiments examining ALK mutations of increasing potency show that PF-06463922 is an effective inhibitor of ALK with greater activity towards ALK neuroblastoma mutants. In contrast to crizotinib, single agent administration of PF-06463922 caused dramatic tumor inhibition in both subcutaneous and orthotopic xenografts as well as a mouse model of high-risk neuroblastoma driven by Th-ALK(F1174L)/MYCN Taken together, our results suggest PF-06463922 is a potent inhibitor of crizotinib-resistant ALK mutations, and highlights an important new treatment option for neuroblastoma patients..
Smith, J.R.
Moreno, L.
Heaton, S.P.
Chesler, L.
Pearson, A.D.
Garrett, M.D.
(2016). Novel pharmacodynamic biomarkers for MYCN protein and PI3K/AKT/mTOR pathway signaling in children with neuroblastoma. Mol oncol,
Vol.10
(4),
pp. 538-552.
show abstract
There is an urgent need for improved therapies for children with high-risk neuroblastoma where survival rates remain low. MYCN amplification is the most common genomic change associated with aggressive neuroblastoma and drugs targeting PI3K/AKT/mTOR, to activate MYCN oncoprotein degradation, are entering clinical evaluation. Our aim was to develop and validate pharmacodynamic (PD) biomarkers to evaluate both proof of mechanism and proof of concept for drugs that block PI3K/AKT/mTOR pathway activity in children with neuroblastoma. We have addressed the issue of limited access to tumor biopsies for quantitative detection of protein biomarkers by optimizing a three-color fluorescence activated cell sorting (FACS) method to purify CD45-/GD2+/CD56+ neuroblastoma cells from bone marrow. We then developed a novel quantitative measurement of MYCN protein in these isolated neuroblastoma cells, providing the potential to demonstrate proof of concept for drugs that inhibit PI3K/AKT/mTOR signaling in this disease. In addition we have established quantitative detection of three biomarkers for AKT pathway activity (phosphorylated and total AKT, GSK3β and P70S6K) in surrogate platelet-rich plasma (PRP) from pediatric patients. Together our new approach to neuroblastoma cell isolation for protein detection and suite of PD assays provides for the first time the opportunity for robust, quantitative measurement of protein-based PD biomarkers in this pediatric patient population. These will be ideal tools to support clinical evaluation of PI3K/AKT/mTOR pathway drugs and their ability to target MYCN oncoprotein in upcoming clinical trials in neuroblastoma..
Vaughan, L.
Clarke, P.A.
Barker, K.
Chanthery, Y.
Gustafson, C.W.
Tucker, E.
Renshaw, J.
Raynaud, F.
Li, X.
Burke, R.
Jamin, Y.
Robinson, S.P.
Pearson, A.
Maira, M.
Weiss, W.A.
Workman, P.
Chesler, L.
(2016). Inhibition of mTOR-kinase destabilizes MYCN and is a potential therapy for MYCN-dependent tumors. Oncotarget,
Vol.7
(36),
pp. 57525-57544.
show abstract
full text
MYC oncoproteins deliver a potent oncogenic stimulus in several human cancers, making them major targets for drug development, but efforts to deliver clinically practical therapeutics have not yet been realized. In childhood cancer, aberrant expression of MYC and MYCN genes delineates a group of aggressive tumours responsible for a major proportion of pediatric cancer deaths. We designed a chemical-genetic screen that identifies compounds capable of enhancing proteasomal elimination of MYCN oncoprotein. We isolated several classes of compound that selectively kill MYCN expressing cells and we focus on inhibitors of PI3K/mTOR pathway in this study. We show that PI3K/mTOR inhibitors selectively killed MYCN-expressing neuroblastoma tumor cells, and induced significant apoptosis of transgenic MYCN-driven neuroblastoma tumors concomitant with elimination of MYCN protein in vivo. Mechanistically, the ability of these compounds to degrade MYCN requires complete blockade of mTOR but not PI3 kinase activity and we highlight NVP-BEZ235 as a PI3K/mTOR inhibitor with an ideal activity profile. These data establish that MYCN expression is a marker indicative of likely clinical sensitivity to mTOR inhibition, and provide a rationale for the selection of clinical candidate MYCN-destabilizers likely to be useful for the treatment of MYCN-driven cancers..
Pearson, A.D.
Herold, R.
Rousseau, R.
Copland, C.
Bradley-Garelik, B.
Binner, D.
Capdeville, R.
Caron, H.
Carleer, J.
Chesler, L.
Geoerger, B.
Kearns, P.
Marshall, L.V.
Pfister, S.M.
Schleiermacher, G.
Skolnik, J.
Spadoni, C.
Sterba, J.
van den Berg, H.
Uttenreuther-Fischer, M.
Witt, O.
Norga, K.
Vassal, G.
Members of Working Group 1 of the Paediatric Platform of ACCELERATE,
(2016). Implementation of mechanism of action biology-driven early drug development for children with cancer. Eur j cancer,
Vol.62,
pp. 124-131.
show abstract
An urgent need remains for new paediatric oncology drugs to cure children who die from cancer and to reduce drug-related sequelae in survivors. In 2007, the European Paediatric Regulation came into law requiring industry to create paediatric drug (all types of medicinal products) development programmes alongside those for adults. Unfortunately, paediatric drug development is still largely centred on adult conditions and not a mechanism of action (MoA)-based model, even though this would be more logical for childhood tumours as these have much fewer non-synonymous coding mutations than adult malignancies. Recent large-scale sequencing by International Genome Consortium and Paediatric Cancer Genome Project has further shown that the genetic and epigenetic repertoire of driver mutations in specific childhood malignancies differs from more common adult-type malignancies. To bring about much needed change, a Paediatric Platform, ACCELERATE, was proposed in 2013 by the Cancer Drug Development Forum, Innovative Therapies for Children with Cancer, the European Network for Cancer Research in Children and Adolescents and the European Society for Paediatric Oncology. The Platform, comprising multiple stakeholders in paediatric oncology, has three working groups, one with responsibility for promoting and developing high-quality MoA-informed paediatric drug development programmes, including specific measures for adolescents. Key is the establishment of a freely accessible aggregated database of paediatric biological tumour drug targets to be aligned with an aggregated pipeline of drugs. This will enable prioritisation and conduct of early phase clinical paediatric trials to evaluate these drugs against promising therapeutic targets and to generate clinical paediatric efficacy and safety data in an accelerated time frame. Through this work, the Platform seeks to ensure that potentially effective drugs, where the MoA is known and thought to be relevant to paediatric malignancies, are evaluated in early phase clinical trials, and that this approach to generate pre-clinical and clinical data is systematically pursued by academia, sponsors, industry, and regulatory bodies to bring new paediatric oncology drugs to front-line therapy more rapidly..
Yogev, O.
Barker, K.
Sikka, A.
Almeida, G.S.
Hallsworth, A.
Smith, L.M.
Jamin, Y.
Ruddle, R.
Koers, A.
Webber, H.T.
Raynaud, F.I.
Popov, S.
Jones, C.
Petrie, K.
Robinson, S.P.
Keun, H.C.
Chesler, L.
(2016). p53 Loss in MYC-Driven Neuroblastoma Leads to Metabolic Adaptations Supporting Radioresistance. Cancer res,
Vol.76
(10),
pp. 3025-3035.
show abstract
full text
Neuroblastoma is the most common childhood extracranial solid tumor. In high-risk cases, many of which are characterized by amplification of MYCN, outcome remains poor. Mutations in the p53 (TP53) tumor suppressor are rare at diagnosis, but evidence suggests that p53 function is often impaired in relapsed, treatment-resistant disease. To address the role of p53 loss of function in the development and pathogenesis of high-risk neuroblastoma, we generated a MYCN-driven genetically engineered mouse model in which the tamoxifen-inducible p53ER(TAM) fusion protein was expressed from a knock-in allele (Th-MYCN/Trp53(KI)). We observed no significant differences in tumor-free survival between Th-MYCN mice heterozygous for Trp53(KI) (n = 188) and Th-MYCN mice with wild-type p53 (n = 101). Conversely, the survival of Th-MYCN/Trp53(KI/KI) mice lacking functional p53 (n = 60) was greatly reduced. We found that Th-MYCN/Trp53(KI/KI) tumors were resistant to ionizing radiation (IR), as expected. However, restoration of functional p53ER(TAM) reinstated sensitivity to IR in only 50% of Th-MYCN/Trp53(KI/KI) tumors, indicating the acquisition of additional resistance mechanisms. Gene expression and metabolic analyses indicated that the principal acquired mechanism of resistance to IR in the absence of functional p53 was metabolic adaptation in response to chronic oxidative stress. Tumors exhibited increased antioxidant metabolites and upregulation of glutathione S-transferase pathway genes, including Gstp1 and Gstz1, which are associated with poor outcome in human neuroblastoma. Accordingly, glutathione depletion by buthionine sulfoximine together with restoration of p53 activity resensitized tumors to IR. Our findings highlight the complex pathways operating in relapsed neuroblastomas and the need for combination therapies that target the diverse resistance mechanisms at play. Cancer Res; 76(10); 3025-35. ©2016 AACR..
Richards, M.W.
Burgess, S.G.
Poon, E.
Carstensen, A.
Eilers, M.
Chesler, L.
Bayliss, R.
(2016). Structural basis of N-Myc binding by Aurora-A and its destabilization by kinase inhibitors. Proc natl acad sci u s a,
Vol.113
(48),
pp. 13726-13731.
show abstract
full text
Myc family proteins promote cancer by inducing widespread changes in gene expression. Their rapid turnover by the ubiquitin-proteasome pathway is regulated through phosphorylation of Myc Box I and ubiquitination by the E3 ubiquitin ligase SCFFbxW7 However, N-Myc protein (the product of the MYCN oncogene) is stabilized in neuroblastoma by the protein kinase Aurora-A in a manner that is sensitive to certain Aurora-A-selective inhibitors. Here we identify a direct interaction between the catalytic domain of Aurora-A and a site flanking Myc Box I that also binds SCFFbxW7 We determined the crystal structure of the complex between Aurora-A and this region of N-Myc to 1.72-Å resolution. The structure indicates that the conformation of Aurora-A induced by compounds such as alisertib and CD532 is not compatible with the binding of N-Myc, explaining the activity of these compounds in neuroblastoma cells and providing a rational basis for the design of cancer therapeutics optimized for destabilization of the complex. We also propose a model for the stabilization mechanism in which binding to Aurora-A alters how N-Myc interacts with SCFFbxW7 to disfavor the generation of Lys48-linked polyubiquitin chains..
Mussai, F.
Egan, S.
Hunter, S.
Webber, H.
Fisher, J.
Wheat, R.
McConville, C.
Sbirkov, Y.
Wheeler, K.
Bendle, G.
Petrie, K.
Anderson, J.
Chesler, L.
De Santo, C.
(2015). Neuroblastoma Arginase Activity Creates an Immunosuppressive Microenvironment That Impairs Autologous and Engineered Immunity. Cancer res,
Vol.75
(15),
pp. 3043-3053.
show abstract
full text
Neuroblastoma is the most common extracranial solid tumor of childhood, and survival remains poor for patients with advanced disease. Novel immune therapies are currently in development, but clinical outcomes have not matched preclinical results. Here, we describe key mechanisms in which neuroblastoma inhibits the immune response. We show that murine and human neuroblastoma tumor cells suppress T-cell proliferation through increased arginase activity. Arginase II is the predominant isoform expressed and creates an arginine-deplete local and systemic microenvironment. Neuroblastoma arginase activity results in inhibition of myeloid cell activation and suppression of bone marrow CD34(+) progenitor proliferation. Finally, we demonstrate that the arginase activity of neuroblastoma impairs NY-ESO-1-specific T-cell receptor and GD2-specific chimeric antigen receptor-engineered T-cell proliferation and cytotoxicity. High arginase II expression correlates with poor survival for patients with neuroblastoma. The results support the hypothesis that neuroblastoma creates an arginase-dependent immunosuppressive microenvironment in both the tumor and blood that leads to impaired immunosurveillance and suboptimal efficacy of immunotherapeutic approaches..
Cheung, B.B.
Tan, O.
Koach, J.
Liu, B.
Shum, M.S.
Carter, D.R.
Sutton, S.
Po'uha, S.T.
Chesler, L.
Haber, M.
Norris, M.D.
Kavallaris, M.
Liu, T.
O'Neill, G.M.
Marshall, G.M.
(2015). Thymosin-β4 is a determinant of drug sensitivity for Fenretinide and Vorinostat combination therapy in neuroblastoma. Mol oncol,
Vol.9
(7),
pp. 1484-1500.
show abstract
Retinoids are an important component of neuroblastoma therapy at the stage of minimal residual disease, yet 40-50% of patients treated with 13-cis-retinoic acid (13-cis-RA) still relapse, indicating the need for more effective retinoid therapy. Vorinostat, or Suberoylanilide hydroxamic acid (SAHA), is a potent inhibitor of histone deacetylase (HDAC) classes I & II and has antitumor activity in vitro and in vivo. Fenretinide (4-HPR) is a synthetic retinoid which acts on cancer cells through both nuclear retinoid receptor and non-receptor mechanisms. In this study, we found that the combination of 4-HPR + SAHA exhibited potent cytotoxic effects on neuroblastoma cells, much more effective than 13-cis-RA + SAHA. The 4-HPR + SAHA combination induced caspase-dependent apoptosis through activation of caspase 3, reduced colony formation and cell migration in vitro, and tumorigenicity in vivo. The 4-HPR and SAHA combination significantly increased mRNA expression of thymosin-beta-4 (Tβ4) and decreased mRNA expression of retinoic acid receptor α (RARα). Importantly, the up-regulation of Tβ4 and down-regulation of RARα were both necessary for the 4-HPR + SAHA cytotoxic effect on neuroblastoma cells. Moreover, Tβ4 knockdown in neuroblastoma cells increased cell migration and blocked the effect of 4-HPR + SAHA on cell migration and focal adhesion formation. In primary human neuroblastoma tumor tissues, low expression of Tβ4 was associated with metastatic disease and predicted poor patient prognosis. Our findings demonstrate that Tβ4 is a novel therapeutic target in neuroblastoma, and that 4-HPR + SAHA is a potential therapy for the disease..
Hill, R.M.
Kuijper, S.
Lindsey, J.C.
Petrie, K.
Schwalbe, E.C.
Barker, K.
Boult, J.K.
Williamson, D.
Ahmad, Z.
Hallsworth, A.
Ryan, S.L.
Poon, E.
Robinson, S.P.
Ruddle, R.
Raynaud, F.I.
Howell, L.
Kwok, C.
Joshi, A.
Nicholson, S.L.
Crosier, S.
Ellison, D.W.
Wharton, S.B.
Robson, K.
Michalski, A.
Hargrave, D.
Jacques, T.S.
Pizer, B.
Bailey, S.
Swartling, F.J.
Weiss, W.A.
Chesler, L.
Clifford, S.C.
(2015). Combined MYC and P53 defects emerge at medulloblastoma relapse and define rapidly progressive, therapeutically targetable disease. Cancer cell,
Vol.27
(1),
pp. 72-84.
show abstract
full text
We undertook a comprehensive clinical and biological investigation of serial medulloblastoma biopsies obtained at diagnosis and relapse. Combined MYC family amplifications and P53 pathway defects commonly emerged at relapse, and all patients in this group died of rapidly progressive disease postrelapse. To study this interaction, we investigated a transgenic model of MYCN-driven medulloblastoma and found spontaneous development of Trp53 inactivating mutations. Abrogation of p53 function in this model produced aggressive tumors that mimicked characteristics of relapsed human tumors with combined P53-MYC dysfunction. Restoration of p53 activity and genetic and therapeutic suppression of MYCN all reduced tumor growth and prolonged survival. Our findings identify P53-MYC interactions at medulloblastoma relapse as biomarkers of clinically aggressive disease that may be targeted therapeutically..
Dolman, M.E.
Poon, E.
Ebus, M.E.
den Hartog, I.J.
van Noesel, C.J.
Jamin, Y.
Hallsworth, A.
Robinson, S.P.
Petrie, K.
Sparidans, R.W.
Kok, R.J.
Versteeg, R.
Caron, H.N.
Chesler, L.
Molenaar, J.J.
(2015). Cyclin-Dependent Kinase Inhibitor AT7519 as a Potential Drug for MYCN-Dependent Neuroblastoma. Clin cancer res,
Vol.21
(22),
pp. 5100-5109.
show abstract
full text
PURPOSE: MYCN-dependent neuroblastomas have low cure rates with current multimodal treatment regimens and novel therapeutic drugs are therefore urgently needed. In previous preclinical studies, we have shown that targeted inhibition of cyclin-dependent kinase 2 (CDK2) resulted in specific killing of MYCN-amplified neuroblastoma cells. This study describes the in vivo preclinical evaluation of the CDK inhibitor AT7519. EXPERIMENTAL DESIGN: Preclinical drug testing was performed using a panel of MYCN-amplified and MYCN single copy neuroblastoma cell lines and different MYCN-dependent mouse models of neuroblastoma. RESULTS: AT7519 killed MYCN-amplified neuroblastoma cell lines more potently than MYCN single copy cell lines with a median LC50 value of 1.7 compared to 8.1 μmol/L (P = 0.0053) and a significantly stronger induction of apoptosis. Preclinical studies in female NMRI homozygous (nu/nu) mice with neuroblastoma patient-derived MYCN-amplified AMC711T xenografts revealed dose-dependent growth inhibition, which correlated with intratumoral AT7519 levels. CDK2 target inhibition by AT7519 was confirmed by significant reductions in levels of phosphorylated retinoblastoma (p-Rb) and nucleophosmin (p-NPM). AT7519 treatment of Th-MYCN transgenic mice resulted in improved survival and clinically significant tumor regression (average tumor size reduction of 86% at day 7 after treatment initiation). The improved efficacy of AT7519 observed in Th-MYCN mice correlated with higher tumor exposure to the drug. CONCLUSIONS: This study strongly suggests that AT7519 is a promising drug for the treatment of high-risk neuroblastoma patients with MYCN amplification..
Tucker, E.R.
Danielson, L.S.
Innocenti, P.
Chesler, L.
(2015). Tackling Crizotinib Resistance: The Pathway from Drug Discovery to the Pediatric Clinic. Cancer res,
Vol.75
(14),
pp. 2770-2774.
show abstract
Neuroblastoma is a childhood malignancy that has not yet benefitted from the rapid progress in the development of small-molecule therapeutics for cancer. An opportunity to take advantage of pharmaceutical innovation in this area arose when the identification of ALK fusion proteins in non-small cell lung cancer (NSCLC) occurred in parallel to the discovery of point mutations of ALK in neuroblastomas. ALK is now known to be a marker of poor outcome in neuroblastoma, and therefore, urgent development of specific ALK inhibitors to treat this devastating disease is a necessity. However, the translation of small molecules from adult directly into pediatric practice has thus far been challenging, due to mutation-specific structural variances in the ALK kinase domain. We discuss how the most recent structural and biological characterizations of ALK are directing preclinical and clinical studies of ALK inhibitors for both NSCLC and neuroblastoma..
Ahmad, Z.
Jasnos, L.
Gil, V.
Howell, L.
Hallsworth, A.
Petrie, K.
Sawado, T.
Chesler, L.
(2015). Molecular and in vivo characterization of cancer-propagating cells derived from MYCN-dependent medulloblastoma. Plos one,
Vol.10
(3),
p. e0119834.
show abstract
Medulloblastoma (MB) is the most common malignant pediatric brain tumor. While the pathways that are deregulated in MB remain to be fully characterized, amplification and/or overexpression of the MYCN gene, which is has a critical role in cerebellar development as a regulator of neural progenitor cell fate, has been identified in several MB subgroups. Phenotypically, aberrant expression of MYCN is associated with the large-cell/anaplastic MB variant, which accounts for 5-15% of cases and is associated with aggressive disease and poor clinical outcome. To better understand the role of MYCN in MB in vitro and in vivo and to aid the development of MYCN-targeted therapeutics we established tumor-derived neurosphere cell lines from the GTML (Glt1-tTA/TRE-MYCN-Luc) genetically engineered mouse model. A fraction of GTML neurospheres were found to be growth factor independent, expressed CD133 (a marker of neural stem cells), failed to differentiate upon MYCN withdrawal and were highly tumorigenic when orthotopically implanted into the cerebellum. Principal component analyzes using single cell RNA assay data suggested that the clinical candidate aurora-A kinase inhibitor MLN8237 converts GTML neurospheres to resemble non-MYCN expressors. Correlating with this, MLN8237 significantly extended the survival of mice bearing GTML MB allografts. In summary, our results demonstrate that MYCN plays a critical role in expansion and survival of aggressive MB-propagating cells, and establish GTML neurospheres as an important resource for the development of novel therapeutic strategies..
Cage, T.A.
Chanthery, Y.
Chesler, L.
Grimmer, M.
Knight, Z.
Shokat, K.
Weiss, W.A.
Gustafson, W.C.
(2015). Downregulation of MYCN through PI3K Inhibition in Mouse Models of Pediatric Neural Cancer. Front oncol,
Vol.5,
p. 111.
show abstract
The MYCN proto-oncogene is associated with poor outcome across a broad range of pediatric tumors. While amplification of MYCN drives subsets of high-risk neuroblastoma and medulloblastoma, dysregulation of MYCN in medulloblastoma (in the absence of amplification) also contributes to pathogenesis. Since PI3K stabilizes MYCN, we have used inhibitors of PI3K to drive degradation. In this study, we show PI3K inhibitors by themselves induce cell cycle arrest, with modest induction of apoptosis. In screening inhibitors of PI3K against MYCN, we identified PIK-75 and its derivative, PW-12, inhibitors of both PI3K and of protein kinases, to be highly effective in destabilizing MYCN. To determine the effects of PW-12 treatment in vivo, we analyzed a genetically engineered mouse model for MYCN-driven neuroblastoma and a model of MYCN-driven medulloblastoma. PW-12 showed significant activity in both models, inducing vascular collapse and regression of medulloblastoma with prominent apoptosis in both models. These results demonstrate that inhibitors of lipid and protein kinases can drive apoptosis in MYCN-driven cancers and support the importance of MYCN as a therapeutic target..
Barone, G.
Tweddle, D.A.
Shohet, J.M.
Chesler, L.
Moreno, L.
Pearson, A.D.
Van Maerken, T.
(2014). MDM2-p53 interaction in paediatric solid tumours: preclinical rationale, biomarkers and resistance. Curr drug targets,
Vol.15
(1),
pp. 114-123.
show abstract
p53 is one of the main regulators of apoptosis, senescence, cell cycle arrest and DNA repair. The expression, function and stabilization of p53 are governed by a complex network of regulators including p14(ARF) and MDM2. MDM2 is the main negative regulator of p53 activity and stability. Unlike tumours in adults, which tend to overcome p53 regulation by p53 mutations, the paediatric tumours neuroblastoma and sarcoma frequently retain wild type p53. Nevertheless, in childhood cancer the p53 pathway is commonly impaired due to upstream MDM2-p14(ARF)-p53 network aberrations. In contrast, aberrations of the p53 downstream pathway are very rare. In cancer cells with intact p53 downstream function MDM2 inhibition, and subsequent rapid increases in nuclear p53 levels, potently "re-activate" dormant apoptotic pathways and rapidly induce apoptotic cell death. As a result MDM2-p53 interaction inhibitors, including cis-imidazolines analogs (Nutlins), are potentially very effective agents in neuroblastoma and sarcomas. Predictive biomarkers are important as a lack of p53 mutations appears to reliably predict response to these inhibitors. Tumours should be screened for p53 mutations in children considered for MDM2-p53 interaction inhibitors. In addition, it is essential that other predictive biomarkers are investigated. The serum concentration of macrophage inhibitory cytokine- 1 (MIC-1) may be a good pharmacodynamic biomarker based on recent findings. In conclusion, targeting the interaction between p53 and its main negative regulator MDM2 represents a major new therapeutic approach in poor prognosis paediatric malignancies without p53 mutations..
Chesler, L.
(2014). Paraneoplasia, cancer development and immunity: what are the connections?. Nature reviews cancer,
Vol.14
(7),
pp. 447-448.
Schnepp, R.
Hart, L.
Raman, P.
Danielson, L.
Gagliardi, M.
Kinsey, R.
Wyce, A.
Barbash, O.
Tummino, P.
Chesler, L.
Maris, J.
(2014). Defining the antitumor activity and sensitivity profiles of BET inhibitors in neuroblastoma. Cancer research,
Vol.74
(20).
Kim, P.Y.
Tan, O.
Diakiw, S.M.
Carter, D.
Sekerye, E.O.
Wasinger, V.C.
Liu, T.
Kavallaris, M.
Norris, M.D.
Haber, M.
Chesler, L.
Dolnikov, A.
Trahair, T.N.
Cheung, N.-.
Marshall, G.M.
Cheung, B.B.
(2014). Identification of plasma complement C3 as a potential biomarker for neuroblastoma using a quantitative proteomic approach. J proteomics,
Vol.96,
pp. 1-12.
show abstract
UNLABELLED: The majority of patients diagnosed with neuroblastoma present with aggressive disease. Improved detection of neuroblastoma cancer cells following initial therapy may help in stratifying patient outcome and monitoring for relapse. To identify potential plasma biomarkers, we utilised a liquid chromatography-tandem mass spectrometry-based proteomics approach to detect differentially-expressed proteins in serum from TH-MYCN mice. TH-MYCN mice carry multiple copies of the human MYCN oncogene in the germline and homozygous mice for the transgene develop neuroblastoma in a manner resembling the human disease. The abundance of plasma proteins was measured over the course of disease initiation and progression. A list of 86 candidate plasma biomarkers was generated. Pathway analysis identified significant association of these proteins with genes involved in the complement system. One candidate, complement C3 protein, was significantly enriched in the plasma of TH-MYCN(+/+) mice at both 4 and 6weeks of age, and was found to be elevated in a cohort of human neuroblastoma plasma samples, compared to healthy subjects. In conclusion, we have demonstrated the suitability of the TH-MYCN(+/+) mouse model of neuroblastoma for identification of novel disease biomarkers in humans, and have identified Complement C3 as a candidate plasma biomarker for measuring disease state in neuroblastoma patients. BIOLOGICAL SIGNIFICANCE: This study has utilised a unique murine model which develops neuroblastoma tumours that are biologically indistinguishable from human neuroblastoma. This animal model has effectively allowed the identification of plasma proteins which may serve as potential biomarkers of neuroblastoma. Furthermore, the label-free ion count quantitation technique which was used displays significant benefits as it is less labour intensive, feasible and accurate. We have been able to successfully validate this approach by confirming the differential abundance of two different plasma proteins. In addition, we have been able to confirm that the candidate biomarker Complement C3, is more abundant in the plasma of human neuroblastoma patient plasma samples when compared to healthy counterparts. Overall we have demonstrated that this approach can be potentially useful in the identification of biomarker candidates, and that further validation of the candidates may lead to the discovery of novel, clinically useful diagnostic tools in the detection of sub-clinical neuroblastoma..
Umapathy, G.
El Wakil, A.
Witek, B.
Chesler, L.
Danielson, L.
Deng, X.
Gray, N.S.
Johansson, M.
Kvarnbrink, S.
Ruuth, K.
Schönherr, C.
Palmer, R.H.
Hallberg, B.
(2014). The kinase ALK stimulates the kinase ERK5 to promote the expression of the oncogene MYCN in neuroblastoma. Sci signal,
Vol.7
(349),
p. ra102.
show abstract
Anaplastic lymphoma kinase (ALK) is an important molecular target in neuroblastoma. Although tyrosine kinase inhibitors abrogating ALK activity are currently in clinical use for the treatment of ALK-positive (ALK(+)) disease, monotherapy with ALK tyrosine kinase inhibitors may not be an adequate solution for ALK(+) neuroblastoma patients. Increased expression of the gene encoding the transcription factor MYCN is common in neuroblastomas and correlates with poor prognosis. We found that the kinase ERK5 [also known as big mitogen-activated protein kinase (MAPK) 1 (BMK1)] is activated by ALK through a pathway mediated by phosphoinositide 3-kinase (PI3K), AKT, MAPK kinase kinase 3 (MEKK3), and MAPK kinase 5 (MEK5). ALK-induced transcription of MYCN and stimulation of cell proliferation required ERK5. Pharmacological or RNA interference-mediated inhibition of ERK5 suppressed the proliferation of neuroblastoma cells in culture and enhanced the antitumor efficacy of the ALK inhibitor crizotinib in both cells and xenograft models. Together, our results indicate that ERK5 mediates ALK-induced transcription of MYCN and proliferation of neuroblastoma, suggesting that targeting both ERK5 and ALK may be beneficial in neuroblastoma patients..
Moore, N.F.
Azarova, A.M.
Bhatnagar, N.
Ross, K.N.
Drake, L.E.
Frumm, S.
Liu, Q.S.
Christie, A.L.
Sanda, T.
Chesler, L.
Kung, A.L.
Gray, N.S.
Stegmaier, K.
George, R.E.
(2014). Molecular rationale for the use of PI3K/AKT/mTOR pathway inhibitors in combination with crizotinib in ALK-mutated neuroblastoma. Oncotarget,
Vol.5
(18),
pp. 8737-8749.
show abstract
Mutations in the ALK tyrosine kinase receptor gene represent important therapeutic targets in neuroblastoma, yet their clinical translation has been challenging. The ALK(F1174L) mutation is sensitive to the ALK inhibitor crizotinib only at high doses and mediates acquired resistance to crizotinib in ALK-translocated cancers. We have shown that the combination of crizotinib and an inhibitor of downstream signaling induces a favorable response in transgenic mice bearing ALK(F1174L)/MYCN-positive neuroblastoma. Here, we investigated the molecular basis of this effect and assessed whether a similar strategy would be effective in ALK-mutated tumors lacking MYCN overexpression. We show that in ALK-mutated, MYCN-amplified neuroblastoma cells, crizotinib alone does not affect mTORC1 activity as indicated by persistent RPS6 phosphorylation. Combined treatment with crizotinib and an ATP-competitive mTOR inhibitor abrogated RPS6 phosphorylation, leading to reduced tumor growth and prolonged survival in ALK(F1174L)/MYCN-positive models compared to single agent treatment. By contrast, this combination, while inducing mTORC1 downregulation, caused reciprocal upregulation of PI3K activity in ALK-mutated cells expressing wild-type MYCN. Here, an inhibitor with potency against both mTOR and PI3K was more effective in promoting cytotoxicity when combined with crizotinib. Our findings should enable a more precise selection of molecularly targeted agents for patients with ALK-mutated tumors..
Jamin, Y.
Glass, L.
Hallsworth, A.
George, R.
Koh, D.-.
Pearson, A.D.
Chesler, L.
Robinson, S.P.
(2014). Intrinsic susceptibility MRI identifies tumors with ALKF1174L mutation in genetically-engineered murine models of high-risk neuroblastoma. Plos one,
Vol.9
(3),
p. e92886.
show abstract
full text
The early identification of children presenting ALK(F1174L)-mutated neuroblastoma, which are associated with resistance to the promising ALK inhibitor crizotinib and a marked poorer prognosis, has become a clinical priority. In comparing the radiology of the novel Th-ALK(F1174L)/Th-MYCN and the well-established Th-MYCN genetically-engineered murine models of neuroblastoma using MRI, we have identified a marked ALK(F1174L)-driven vascular phenotype. We demonstrate that quantitation of the transverse relaxation rate R2* (s(-1)) using intrinsic susceptibility-MRI under baseline conditions and during hyperoxia, can robustly discriminate this differential vascular phenotype, and identify MYCN-driven tumors harboring the ALK(F1174L) mutation with high specificity and selectivity. Intrinsic susceptibility-MRI could thus potentially provide a non-invasive and clinically-exploitable method to help identifying children with MYCN-driven neuroblastoma harboring the ALK(F1174L) mutation at the time of diagnosis..
Barone, G.
Anderson, J.
Pearson, A.D.
Petrie, K.
Chesler, L.
(2013). New strategies in neuroblastoma: Therapeutic targeting of MYCN and ALK. Clin cancer res,
Vol.19
(21),
pp. 5814-5821.
show abstract
Clinical outcome remains poor in patients with high-risk neuroblastoma, in which chemoresistant relapse is common following high-intensity conventional multimodal therapy. Novel treatment approaches are required. Although recent genomic profiling initiatives have not revealed a high frequency of mutations in any significant number of therapeutically targeted genes, two exceptions, amplification of the MYCN oncogene and somatically acquired tyrosine kinase domain point mutations in anaplastic lymphoma kinase (ALK), present exciting possibilities for targeted therapy. In contrast with the situation with ALK, in which a robust pipeline of pharmacologic agents is available from early clinical use in adult malignancy, therapeutic targeting of MYCN (and MYC oncoproteins in general) represents a significant medicinal chemistry challenge that has remained unsolved for two decades. We review the latest approaches envisioned for blockade of ALK activity in neuroblastoma, present a classification of potential approaches for therapeutic targeting of MYCN, and discuss how recent developments in targeting of MYC proteins seem to make therapeutic inhibition of MYCN a reality in the clinic..
Brockmann, M.
Poon, E.
Chesler, L.
Eilers, M.
(2013). Targeting the dependence of N-Myc on interaction with Aurora-A with small molecules. Molecular cancer therapeutics,
Vol.12
(5).
Brockmann, M.
Poon, E.
Berry, T.
Carstensen, A.
Deubzer, H.E.
Rycak, L.
Jamin, Y.
Thway, K.
Robinson, S.P.
Roels, F.
Witt, O.
Fischer, M.
Chesler, L.
Eilers, M.
(2013). Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer cell,
Vol.24
(1),
pp. 75-89.
show abstract
Amplification of MYCN is a driver mutation in a subset of human neuroendocrine tumors, including neuroblastoma. No small molecules that target N-Myc, the protein encoded by MYCN, are clinically available. N-Myc forms a complex with the Aurora-A kinase, which protects N-Myc from proteasomal degradation. Although stabilization of N-Myc does not require the catalytic activity of Aurora-A, we show here that two Aurora-A inhibitors, MLN8054 and MLN8237, disrupt the Aurora-A/N-Myc complex and promote degradation of N-Myc mediated by the Fbxw7 ubiquitin ligase. Disruption of the Aurora-A/N-Myc complex inhibits N-Myc-dependent transcription, correlating with tumor regression and prolonged survival in a mouse model of MYCN-driven neuroblastoma. We conclude that Aurora-A is an accessible target that makes destabilization of N-Myc a viable therapeutic strategy..
Lastowska, M.
Al-Afghani, H.
Al-Balool, H.H.
Sheth, H.
Mercer, E.
Coxhead, J.M.
Redfern, C.P.
Peters, H.
Burt, A.D.
Santibanez-Koref, M.
Bacon, C.M.
Chesler, L.
Rust, A.G.
Adams, D.J.
Williamson, D.
Clifford, S.C.
Jackson, M.S.
(2013). Identification of a neuronal transcription factor network involved in medulloblastoma development. Acta neuropathol commun,
Vol.1,
p. 35.
show abstract
BACKGROUND: Medulloblastomas, the most frequent malignant brain tumours affecting children, comprise at least 4 distinct clinicogenetic subgroups. Aberrant sonic hedgehog (SHH) signalling is observed in approximately 25% of tumours and defines one subgroup. Although alterations in SHH pathway genes (e.g. PTCH1, SUFU) are observed in many of these tumours, high throughput genomic analyses have identified few other recurring mutations. Here, we have mutagenised the Ptch+/- murine tumour model using the Sleeping Beauty transposon system to identify additional genes and pathways involved in SHH subgroup medulloblastoma development. RESULTS: Mutagenesis significantly increased medulloblastoma frequency and identified 17 candidate cancer genes, including orthologs of genes somatically mutated (PTEN, CREBBP) or associated with poor outcome (PTEN, MYT1L) in the human disease. Strikingly, these candidate genes were enriched for transcription factors (p=2x10-5), the majority of which (6/7; Crebbp, Myt1L, Nfia, Nfib, Tead1 and Tgif2) were linked within a single regulatory network enriched for genes associated with a differentiated neuronal phenotype. Furthermore, activity of this network varied significantly between the human subgroups, was associated with metastatic disease, and predicted poor survival specifically within the SHH subgroup of tumours. Igf2, previously implicated in medulloblastoma, was the most differentially expressed gene in murine tumours with network perturbation, and network activity in both mouse and human tumours was characterised by enrichment for multiple gene-sets indicating increased cell proliferation, IGF signalling, MYC target upregulation, and decreased neuronal differentiation. CONCLUSIONS: Collectively, our data support a model of medulloblastoma development in SB-mutagenised Ptch+/- mice which involves disruption of a novel transcription factor network leading to Igf2 upregulation, proliferation of GNPs, and tumour formation. Moreover, our results identify rational therapeutic targets for SHH subgroup tumours, alongside prognostic biomarkers for the identification of poor-risk SHH patients..
DuBois, S.G.
Chesler, L.
Groshen, S.
Hawkins, R.
Goodarzian, F.
Shimada, H.
Yanik, G.
Tagen, M.
Stewart, C.
Mosse, Y.P.
Maris, J.M.
Tsao-Wei, D.
Marachelian, A.
Villablanca, J.G.
Matthay, K.K.
(2012). Phase I study of vincristine, irinotecan, and ¹³¹I-metaiodobenzylguanidine for patients with relapsed or refractory neuroblastoma: a new approaches to neuroblastoma therapy trial. Clin cancer res,
Vol.18
(9),
pp. 2679-2686.
show abstract
PURPOSE: (131)I-metaiodobenzylguanidine (MIBG) is a targeted radiopharmaceutical with activity in patients with relapsed or refractory neuroblastoma. Irinotecan is a known radiosensitizer with activity in neuroblastoma. This phase I study aimed to determine the recommended phase 2 dose of MIBG together with fixed doses of vincristine and irinotecan. EXPERIMENTAL DESIGN: Patients 1 to 30 years old with relapsed or refractory neuroblastoma and MIBG-avid tumors were eligible. All patients had autologous hematopoietic stem cells (PBSC) available and met standard phase I organ function requirements. Irinotecan (20 mg/m(2)/dose IV) was given on days 0 to 4 and 7 to 11, with vincristine (1.5 mg/m(2) IV) on days 0 and 7. MIBG was given on day 1 following a 3 + 3 phase I dose escalation design starting at 8 mCi/kg MIBG. PBSCs were administered at dose level 8 mCi/kg for prolonged myelosuppression and for all patients at 12 mCi/kg or more. RESULTS: Twenty-four patients evaluable for dose escalation (median age, 6.7 years; range, 1.9-26.8 years) received 1 (n = 17), 2 (n = 5), or 3 (n = 2) cycles of therapy. Myelosuppression and diarrhea were the most common toxicities. Two of 6 patients at the 18 mCi/kg dose level had dose-limiting toxicity (DLT), including one with protocol-defined DLT with prolonged mild aspartate aminotransferase elevation. Eighteen mCi/kg was the recommended phase 2 dose. Six additional patients were treated at 18 mCi/kg, with one additional DLT. Responses (2 complete and 4 partial responses) occurred in 6 of 24 (25%) evaluable patients. CONCLUSIONS: MIBG is tolerable and active at 18 mCi/kg with standard doses of vincristine and irinotecan..
Berry, T.
Luther, W.
Bhatnagar, N.
Jamin, Y.
Poon, E.
Sanda, T.
Pei, D.
Sharma, B.
Vetharoy, W.R.
Hallsworth, A.
Ahmad, Z.
Barker, K.
Moreau, L.
Webber, H.
Wang, W.
Liu, Q.
Perez-Atayde, A.
Rodig, S.
Cheung, N.-.
Raynaud, F.
Hallberg, B.
Robinson, S.P.
Gray, N.S.
Pearson, A.D.
Eccles, S.A.
Chesler, L.
George, R.E.
(2012). The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer cell,
Vol.22
(1),
pp. 117-130.
show abstract
The ALK(F1174L) mutation is associated with intrinsic and acquired resistance to crizotinib and cosegregates with MYCN in neuroblastoma. In this study, we generated a mouse model overexpressing ALK(F1174L) in the neural crest. Compared to ALK(F1174L) and MYCN alone, co-expression of these two oncogenes led to the development of neuroblastomas with earlier onset, higher penetrance, and enhanced lethality. ALK(F1174L)/MYCN tumors exhibited increased MYCN dosage due to ALK(F1174L)-induced activation of the PI3K/AKT/mTOR and MAPK pathways, coupled with suppression of MYCN pro-apoptotic effects. Combined treatment with the ATP-competitive mTOR inhibitor Torin2 overcame the resistance of ALK(F1174L)/MYCN tumors to crizotinib. Our findings demonstrate a pathogenic role for ALK(F1174L) in neuroblastomas overexpressing MYCN and suggest a strategy for improving targeted therapy for ALK-positive neuroblastoma..
Walton, M.I.
Eve, P.D.
Hayes, A.
Valenti, M.R.
De Haven Brandon, A.K.
Box, G.
Hallsworth, A.
Smith, E.L.
Boxall, K.J.
Lainchbury, M.
Matthews, T.P.
Jamin, Y.
Robinson, S.P.
Aherne, G.W.
Reader, J.C.
Chesler, L.
Raynaud, F.I.
Eccles, S.A.
Collins, I.
Garrett, M.D.
(2012). CCT244747 is a novel potent and selective CHK1 inhibitor with oral efficacy alone and in combination with genotoxic anticancer drugs. Clin cancer res,
Vol.18
(20),
pp. 5650-5661.
show abstract
PURPOSE: Many tumors exhibit defective cell-cycle checkpoint control and increased replicative stress. CHK1 is critically involved in the DNA damage response and maintenance of replication fork stability. We have therefore discovered a novel potent, highly selective, orally active ATP-competitive CHK1 inhibitor, CCT244747, and present its preclinical pharmacology and therapeutic activity. EXPERIMENTAL DESIGN: Cellular CHK1 activity was assessed using an ELISA assay, and cytotoxicity a SRB assay. Biomarker modulation was measured using immunoblotting, and cell-cycle effects by flow cytometry analysis. Single-agent oral CCT244747 antitumor activity was evaluated in a MYCN-driven transgenic mouse model of neuroblastoma by MRI and in genotoxic combinations in human tumor xenografts by growth delay. RESULTS: CCT244747 inhibited cellular CHK1 activity (IC(50) 29-170 nmol/L), significantly enhanced the cytotoxicity of several anticancer drugs, and abrogated drug-induced S and G(2) arrest in multiple tumor cell lines. Biomarkers of CHK1 (pS296 CHK1) activity and cell-cycle inactivity (pY15 CDK1) were induced by genotoxics and inhibited by CCT244747 both in vitro and in vivo, producing enhanced DNA damage and apoptosis. Active tumor concentrations of CCT244747 were obtained following oral administration. The antitumor activity of both gemcitabine and irinotecan were significantly enhanced by CCT244747 in several human tumor xenografts, giving concomitant biomarker modulation indicative of CHK1 inhibition. CCT244747 also showed marked antitumor activity as a single agent in a MYCN-driven neuroblastoma. CONCLUSION: CCT244747 represents the first structural disclosure of a highly selective, orally active CHK1 inhibitor and warrants further evaluation alone or combined with genotoxic anticancer therapies..
Terrile, M.
Bryan, K.
Kuijper, S.
Chesler, L.
Stallings, R.L.
(2012). MiRNA expression profiling of the murine GTML medulloblastoma model reveals similarities with human tumors and identifies novel candidate miRNAs. Cancer research,
Vol.72.
Swartling, F.J.
Savov, V.
Persson, A.I.
Chen, J.
Hackett, C.S.
Northcott, P.A.
Grimmer, M.R.
Lau, J.
Chesler, L.
Perry, A.
Phillips, J.J.
Taylor, M.D.
Weiss, W.A.
(2012). Distinct neural stem cell populations give rise to disparate brain tumors in response to N-MYC. Cancer cell,
Vol.21
(5),
pp. 601-613.
show abstract
The proto-oncogene MYCN is mis-expressed in various types of human brain tumors. To clarify how developmental and regional differences influence transformation, we transduced wild-type or mutationally stabilized murine N-myc(T58A) into neural stem cells (NSCs) from perinatal murine cerebellum, brain stem, and forebrain. Transplantation of N-myc(WT) NSCs was insufficient for tumor formation. N-myc(T58A) cerebellar and brain stem NSCs generated medulloblastoma/primitive neuroectodermal tumors, whereas forebrain NSCs developed diffuse glioma. Expression analyses distinguished tumors generated from these different regions, with tumors from embryonic versus postnatal cerebellar NSCs demonstrating Sonic Hedgehog (SHH) dependence and SHH independence, respectively. These differences were regulated in part by the transcription factor SOX9, activated in the SHH subclass of human medulloblastoma. Our results demonstrate context-dependent transformation of NSCs in response to a common oncogenic signal..
Moreno, L.
Chesler, L.
Hargrave, D.
Eccles, S.A.
Pearson, A.D.
(2011). Preclinical drug development for childhood cancer. Expert opinion on drug discovery,
Vol.6
(1),
pp. 49-16.
Terrile, M.
Bryan, K.
Vaughan, L.
Hallsworth, A.
Webber, H.
Chesler, L.
Stallings, R.L.
(2011). miRNA Expression Profiling of the Murine TH-MYCN Neuroblastoma Model Reveals Similarities with Human Tumors and Identifies Novel Candidate MiRNAs. Plos one,
Vol.6
(12).
Chesler, L.
Weiss, W.A.
(2011). Genetically engineered murine models--contribution to our understanding of the genetics, molecular pathology and therapeutic targeting of neuroblastoma. Semin cancer biol,
Vol.21
(4),
pp. 245-255.
show abstract
full text
Genetically engineered mouse models (GEMM) have made major contributions to a molecular understanding of several adult cancers and these results are increasingly being translated into the pre-clinical setting where GEMM will very likely make a major impact on the development of targeted therapeutics in the near future. The relationship of pediatric cancers to altered developmental programs, and their genetic simplicity relative to adult cancers provides unique opportunities for the application of new advances in GEMM technology. In neuroblastoma the well-characterized TH-MYCN GEMM is increasingly used for a variety of molecular-genetic, developmental and pre-clinical therapeutics applications. We discuss: the present and historical application of GEMM to neuroblastoma research, future opportunities, and relevant targets suitable for new GEMM strategies in neuroblastoma. We review the potential of these models to contribute both to an understanding of the developmental nature of neuroblastoma and to improved therapy for this disease..
Faisal, A.
Vaughan, L.
Bavetsias, V.
Sun, C.
Atrash, B.
Avery, S.
Jamin, Y.
Robinson, S.P.
Workman, P.
Blagg, J.
Raynaud, F.I.
Eccles, S.A.
Chesler, L.
Linardopoulos, S.
(2011). The aurora kinase inhibitor CCT137690 downregulates MYCN and sensitizes MYCN-amplified neuroblastoma in vivo. Mol cancer ther,
Vol.10
(11),
pp. 2115-2123.
show abstract
Aurora kinases regulate key stages of mitosis including centrosome maturation, spindle assembly, chromosome segregation, and cytokinesis. Aurora A and B kinase overexpression has also been associated with various human cancers, and as such, they have been extensively studied as novel antimitotic drug targets. Here, we characterize the Aurora kinase inhibitor CCT137690, a highly selective, orally bioavailable imidazo[4,5-b]pyridine derivative that inhibits Aurora A and B kinases with low nanomolar IC(50) values in both biochemical and cellular assays and exhibits antiproliferative activity against a wide range of human solid tumor cell lines. CCT137690 efficiently inhibits histone H3 and transforming acidic coiled-coil 3 phosphorylation (Aurora B and Aurora A substrates, respectively) in HCT116 and HeLa cells. Continuous exposure of tumor cells to the inhibitor causes multipolar spindle formation, chromosome misalignment, polyploidy, and apoptosis. This is accompanied by p53/p21/BAX induction, thymidine kinase 1 downregulation, and PARP cleavage. Furthermore, CCT137690 treatment of MYCN-amplified neuroblastoma cell lines inhibits cell proliferation and decreases MYCN protein expression. Importantly, in a transgenic mouse model of neuroblastoma that overexpresses MYCN protein and is predisposed to spontaneous neuroblastoma formation, this compound significantly inhibits tumor growth. The potent preclinical activity of CCT137690 suggests that this inhibitor may benefit patients with MYCN-amplified neuroblastoma..
George, R.E.
Luther, W.
Ahmad, Z.
Cullis, E.R.
Webber, H.
Rodig, S.
Pearson, A.D.
Chesler, L.
(2011). The ALK-F1174L mutation accelerates MYCN-driven tumorigenesis in a murine transgenic neuroblastoma model. Cancer research,
Vol.71.
Swartling, F.J.
Persson, A.I.
Lau, J.
Northcott, P.A.
Grimmer, M.R.
Chesler, L.
Perry, A.
Phillips, J.J.
Taylor, M.D.
Weiss, W.A.
(2011). Interactions between N-myc and Sox9 promote medulloblastoma and determine cell fate decisions in cerebellar neural stem cells. Cancer research,
Vol.71.
DuBois, S.G.
Chesler, L.
Groshen, S.G.
Hawkins, R.
Goodarzian, F.
Yanik, G.A.
Stewart, C.F.
Mosse, Y.P.
Maris, J.M.
Villablanca, J.
Matthay, K.K.
(2011). Phase I study of vincristine, irinotecan, and (131)I-MIBG for patients with relapsed or refractory neuroblastoma: A New Approach to Neuroblastoma Therapy (NANT) study. J clin oncol,
Vol.29
(15_suppl),
p. 9513.
show abstract
9513 Background: (131)I-metaiodobenzylguanidine (MIBG) is a targeted radiopharmaceutical with activity in neuroblastoma. Irinotecan is a radiosensitizer with activity in neuroblastoma. This phase I study aimed to determine the recommended phase II dose (RP2D) of MIBG together with vincristine and irinotecan. METHODS: Patients 1-30 years old with relapsed or refractory MIBG-avid neuroblastoma were eligible. All patients had autologous hematopoietic stem cells (PBSCs) available and met standard phase 1 requirements. Prior vincristine and irinotecan, but not prior MIBG, were allowed. Patients received cefixime (8 mg/kg/day) on days -5 to +21. Irinotecan (20 mg/m(2)/dose IV) was given on days 0-4 and 7-11, with vincristine (1.5 mg/m(2)) on days 0 and 7. MIBG was given on day 1 following a 3+3 phase I design with dose levels of 8, 12, 15, and 18 mCi/kg, with 6 additional patients treated at the RP2D. PBSCs were administered to 1 patient at 8 mCi/kg for myelosuppression and on day 13 for all patients at > 12 mCi/kg. RESULTS: 24 evaluable patients (median age 6.7 years; range 1.9 - 24.8) received 33 courses. Myelosuppression and diarrhea were the most common toxicities. 70% of courses were associated with diarrhea (grade 3 in 6 courses). No dose limiting toxicities (DLTs) were seen at dose levels 8 and 12 mCi/kg. At 15 mCi/kg, 1 patient had DLT of grade 3 diarrhea. At 18 mCi/kg, 1 patient had DLT with hallucinations and hyponatremia. One patient had protocol-defined DLT with prolonged ALT elevation that was not deemed clinically dose limiting. 18 mCi/kg was declared the RP2D and 6 additional patients enrolled, with 1 DLT (ALT and AST elevation). 7 patients received 2-3 courses to cumulative doses of 24-36 mCi/kg without DLT. The objective response rate was 25% (95% CI: 11-46%), with 3 complete responses, 1 very good partial response, and 2 partial responses. The response rate among 12 patients treated with 18 mCi/kg was 25% (2 complete responses and 1 very good partial response). CONCLUSIONS: MIBG is tolerable and active at doses of 18 mCi/kg with vincristine and irinotecan. Further studies are planned to determine this regimen's role in frontline therapy for high risk patients..
DuBois, S.G.
Chesler, L.
Groshen, S.G.
Hawkins, R.
Goodarzian, F.
Yanik, G.A.
Stewart, C.F.
Mosse, Y.P.
Maris, J.M.
Villablanca, J.
Matthay, K.K.
(2011). Phase I study of vincristine, irinotecan, and I-131-MIBG for patients with relapsed or refractory neuroblastoma: A New Approach to Neuroblastoma Therapy (NANT) study. Journal of clinical oncology,
Vol.29
(15).
Jamin, Y.
Cullis, E.R.
Vaughan, L.
Koh, D.-.
Chesler, L.
Robinson, S.P.
(2010). Characterization of tumor progression and chemoresponse in a novel transgenic mouse model of neuroblastoma (TH-MYCN) using magnetic resonance imaging. Cancer research,
Vol.70.
Swartling, F.J.
Grimmer, M.R.
Hackett, C.S.
Northcott, P.A.
Fan, Q.-.
Goldenberg, D.D.
Lau, J.
Masic, S.
Nguyen, K.
Yakovenko, S.
Zhe, X.-.
Gilmer, H.C.
Collins, R.
Nagaoka, M.
Phillips, J.J.
Jenkins, R.B.
Tihan, T.
Vandenberg, S.R.
James, C.D.
Tanaka, K.
Taylor, M.D.
Weiss, W.A.
Chesler, L.
(2010). Pleiotropic role for MYCN in medulloblastoma. Genes dev,
Vol.24
(10),
pp. 1059-1072.
show abstract
Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Sonic Hedgehog (SHH) signaling drives a minority of MB, correlating with desmoplastic pathology and favorable outcome. The majority, however, arises independently of SHH and displays classic or large cell anaplastic (LCA) pathology and poor prognosis. To identify common signaling abnormalities, we profiled mRNA, demonstrating misexpression of MYCN in the majority of human MB and negligible expression in normal cerebella. We clarified a role in pathogenesis by targeting MYCN (and luciferase) to cerebella of transgenic mice. MYCN-driven MB showed either classic or LCA pathologies, with Shh signaling activated in approximately 5% of tumors, demonstrating that MYCN can drive MB independently of Shh. MB arose at high penetrance, consistent with a role for MYCN in initiation. Tumor burden correlated with bioluminescence, with rare metastatic spread to the leptomeninges, suggesting roles for MYCN in both progression and metastasis. Transient pharmacological down-regulation of MYCN led to both clearance and senescence of tumor cells, and improved survival. Targeted expression of MYCN thus contributes to initiation, progression, and maintenance of MB, suggesting a central role for MYCN in pathogenesis..
Weiss, W.
Grimmer, M.
Itsara, M.
Chanthery, Y.
Persson, A.
Chen, J.
Nguyen, K.
Yakovenko, S.
Mueller, S.
Goldenberg, D.
Kim, G.
Haas-Kogan, D.
Matthay, K.
Chesler, L.
(2009). Genetically engineered mouse models for neuroblastoma: applications to developmental therapeutics. Cancer research,
Vol.69.
Swartling, F.
Persson, A.
Grimmer, M.
Chesler, L.
Weiss, W.
(2009). Targeting notch and BMP pathways in MYCN-induced medulloblastoma spheres. Cancer research,
Vol.69.
Chesler, L.
Goldenberg, D.
Collins, R.
Grimmer, M.
Kim, G.E.
Tihan, T.
Nguyen, K.
Yakovenko, S.
Matthay, K.K.
Weiss, W.A.
(2008). Chemotherapy-Induced Apoptosis in a Transgenic Model of Neuroblastoma Proceeds Through p53 Induction. Neoplasia,
Vol.10
(11),
pp. 1268-9.
full text
Chesler, L.
Goldenberg, D.D.
Seales, I.T.
Satchi-Fainaro, R.
Grimmer, M.
Collins, R.
Struett, C.
Nguyen, K.N.
Kim, G.
Tihan, T.
Bao, Y.
Brekken, R.A.
Bergers, G.
Folkman, J.
Weiss, W.A.
(2007). Malignant progression and blockade of angiogenesis in a murine transgenic model of neuroblastoma. Cancer res,
Vol.67
(19),
pp. 9435-9442.
show abstract
Targeted expression of MYCN to the neural crest [under control of the rat tyrosine hydroxylase (TH) promoter] causes neuroblastoma in transgenic mice (TH-MYCN) and is a well-established model for this disease. Because high levels of MYCN are associated with enhanced tumor angiogenesis and poor clinical outcome in neuroblastoma, we serially characterized malignant progression, angiogenesis, and sensitivity to angiogenic blockade in tumors from these animals. Tumor cells were proliferative, secreted high levels of the angiogenic ligand vascular endothelial growth factor (VEGF), and recruited a complex vasculature expressing the angiogenic markers VEGF-R2, alpha-SMA, and matrix metalloproteinases MMP-2 and MMP-9, all of which are also expressed in human disease. Treatment of established murine tumors with the angiogenesis inhibitor TNP-470 caused near-complete ablation, with reduced proliferation, enhanced apoptosis, and vasculature disruption. Because TNP-470 has been associated with neurotoxicity, we tested the recently described water-soluble HPMA copolymer-TNP-470 conjugate (caplostatin), which showed comparable efficacy and was well tolerated without weight loss or neurotoxicity as measured by rotarod testing. This study highlights the importance of angiogenesis inhibition in a spontaneous murine tumor with native tumor-microenvironment interactions, validates the use of mice transgenic for TH-MYCN as a model for therapy in this common pediatric tumor, and supports further clinical development of caplostatin as an antiangiogenic therapy in childhood neuroblastoma..
Meyer, G.E.
Chesler, L.
Liu, D.
Gable, K.
Maddux, B.A.
Goldenberg, D.D.
Youngren, J.F.
Goldfine, I.D.
Weiss, W.A.
Matthay, K.K.
Rosenthal, S.M.
(2007). Nordihydroguaiaretic acid inhibits insulin-like growth factor signaling, growth, and survival in human neuroblastoma cells. J cell biochem,
Vol.102
(6),
pp. 1529-1541.
show abstract
full text
Neuroblastoma is a common pediatric malignancy that metastasizes to the liver, bone, and other organs. Children with metastatic disease have a less than 50% chance of survival with current treatments. Insulin-like growth factors (IGFs) stimulate neuroblastoma growth, survival, and motility, and are expressed by neuroblastoma cells and the tissues they invade. Thus, therapies that disrupt the effects of IGFs on neuroblastoma tumorigenesis may slow disease progression. We show that NVP-AEW541, a specific inhibitor of the IGF-I receptor (IGF-IR), potently inhibits neuroblastoma growth in vitro. Nordihydroguaiaretic acid (NDGA), a phenolic compound isolated from the creosote bush (Larrea divaricata), has anti-tumor properties against a number of malignancies, has been shown to inhibit the phosphorylation and activation of the IGF-IR in breast cancer cells, and is currently in Phase I trials for prostate cancer. In the present study in neuroblastoma, NDGA inhibits IGF-I-mediated activation of the IGF-IR and disrupts activation of ERK and Akt signaling pathways induced by IGF-I. NDGA inhibits growth of neuroblastoma cells and induces apoptosis at higher doses, causing IGF-I-resistant activation of caspase-3 and a large increase in the fraction of sub-G0 cells. In addition, NDGA inhibits the growth of xenografted human neuroblastoma tumors in nude mice. These results indicate that NDGA may be useful in the treatment of neuroblastoma and may function in part via disruption of IGF-IR signaling..
Chesler, L.
Schlieve, C.
Goldenberg, D.D.
Kenney, A.
Kim, G.
McMillan, A.
Matthay, K.K.
Rowitch, D.
Weiss, W.A.
(2006). Inhibition of phosphatidylinositol 3-kinase destabilizes Mycn protein and blocks malignant progression in neuroblastoma. Cancer res,
Vol.66
(16),
pp. 8139-8146.
show abstract
Amplification of MYCN occurs commonly in neuroblastoma. We report that phosphatidylinositol 3-kinase (PI3K) inhibition in murine neuroblastoma (driven by a tyrosine hydroxylase-MYCN transgene) led to decreased tumor mass and decreased levels of Mycn protein without affecting levels of MYCN mRNA. Consistent with these observations, PI3K inhibition in MYCN-amplified human neuroblastoma cell lines resulted in decreased levels of Mycn protein without affecting levels of MYCN mRNA and caused decreased proliferation and increased apoptosis. To clarify the importance of Mycn as a target of broad-spectrum PI3K inhibitors, we transduced wild-type N-myc and N-myc mutants lacking glycogen synthase kinase 3beta phosphorylation sites into human neuroblastoma cells with no endogenous expression of myc. In contrast to wild-type N-myc, the phosphorylation-defective mutant proteins were stabilized and were resistant to the antiproliferative effects of PI3K inhibition. Our results show the importance of Mycn as a therapeutic target in established tumors in vivo, offer a mechanistic rationale to test PI3K inhibitors in MYCN-amplified neuroblastoma, and represent a therapeutic approach applicable to a broad range of cancers in which transcription factors are stabilized through a PI3K-dependent mechanism..
Feusner, J.
Chesler, L.
Shen, V.
(2003). t-PA use in pediatric patients. J pediatr hematol oncol,
Vol.25
(7),
p. 589.
Chesler, L.
Feusner, J.H.
(2002). Use of tissue plasminogen activator (rt-PA) in young children with cancer and dysfunctional central venous catheters. J pediatr hematol oncol,
Vol.24
(8),
pp. 653-656.
show abstract
PURPOSE: To determine the efficacy and safety of low, nonescalating dose tissue plasminogen activator (rt-PA) in restoring the patency of occluded central venous access devices (CVCs) in children with cancer who weigh less than 30 kg. PATIENTS AND METHODS: A single-center review of the use of rt-PA (0.5 mg indwelling for 30 minutes in the CVC) was conducted in 42 cancer patients with large bore central venous access devices implanted over a 2-year period. All patients weighed less than 30 kg. None had been previously treated with a thrombolytic agent. The efficacy for restoring function to CVCs was measured and correlated with patient age, weight and CVC lumen size. Propensity to rethrombose following an initial occlusion and treatment was also determined. RESULTS: Of 235 doses of rt-PA administered in a 2-year period, 55 doses administered to 42 patients met the eligibility criteria as outlined. Twenty-nine patients (69%) had function restored with a single dose; 8 patients (19%) required 2 doses, and 5 patients (12%) failed 2 doses; for an overall success rate of 88%. No significant adverse events occurred. Of the 37 cleared CVCs, 14 (38%) reoccluded within 1 month. A higher proportion of patients initially treated with one rt-PA (71%) experienced another CVC dysfunction within 1 month, compared with 29% CVC dysfunction in those requiring >1 dose. CONCLUSIONS: This article describes the use of rt-PA (0.5 mg, without dose escalation) to lyse CVC-associated thrombi specifically in small children with cancer, a patient population in which it is particularly desirable to minimize the degree of fibrinolysis. One dose of 0.5 mg rt-PA, with an additional dose if necessary, is as safe and effective as previously reported escalating dose regimens for CVC clot lysis. There is no statistically significant correlation of treatment failure with patient age, weight, or catheter lumen size, and no significant propensity for rapid rethrombosis following a single dysfunction and treatment. Patients initially treated with a single dose of rt-PA appear to have more subsequent dysfunctions in the month after treatment, an observation that warrants further study..
Chesler, L.
Hwang, L.
Patton, W.
Heyman, M.B.
(2000). Henoch-Schönlein purpura with severe jejunitis and minimal skin lesions. J pediatr gastroenterol nutr,
Vol.30
(1),
pp. 92-95.
Vodovotz, Y.
Chesler, L.
Chong, H.
Kim, S.J.
Simpson, J.T.
DeGraff, W.
Cox, G.W.
Roberts, A.B.
Wink, D.A.
Barcellos-Hoff, M.H.
(1999). Regulation of transforming growth factor beta1 by nitric oxide. Cancer res,
Vol.59
(9),
pp. 2142-2149.
show abstract
Many tumor cells or their secreted products suppress the function of tumor-infiltrating macrophages. Tumor cells often produce abundant transforming growth factor beta1 (TGF-beta1), which in addition to other immunosuppressive actions suppresses the inducible isoform of NO synthase. TGF-beta1 is secreted in a latent form, which consists of TGF-beta1 noncovalently associated with latency-associated peptide (LAP) and which can be activated efficiently by exposure to reactive oxygen species. Coculture of the human lung adenocarcinoma cell line A549 and ANA-1 macrophages activated with IFN-gamma plus lipopolysaccharide resulted in increased synthesis and activation of latent TGF-beta1 protein by both A549 and ANA-1 cells, whereas unstimulated cultures of either cell type alone expressed only latent TGF-beta1. We investigated whether exposure of tumor cells to NO influences the production, activation, or activity of TGF-beta1.A549 human lung adenocarcinoma cells exposed to the chemical NO donor diethylamine-NONOate showed increased immunoreactivity of cell-associated latent and active TGF-beta1 in a time- and dose-dependent fashion at 24-48 h after treatment. Exposure of latent TGF-beta1 to solution sources of NO neither led to recombinant latent TGF-beta1 activation nor modified recombinant TGF-beta1 activity. A novel mechanism was observed, however: treatment of recombinant LAP with NO resulted in its nitrosylation and interfered with its ability to neutralize active TGF-beta1. These results provide the first evidence that nitrosative stress influences the regulation of TGF-beta1 and raise the possibility that NO production may augment TGF-beta1 activity by modifying a naturally occurring neutralizing peptide..
Vodovotz, Y.
Geiser, A.G.
Chesler, L.
Letterio, J.J.
Campbell, A.
Lucia, M.S.
Sporn, M.B.
Roberts, A.B.
(1996). Spontaneously increased production of nitric oxide and aberrant expression of the inducible nitric oxide synthase in vivo in the transforming growth factor beta 1 null mouse. J exp med,
Vol.183
(5),
pp. 2337-2342.
show abstract
Transforming growth factor beta 1 null mice (TGF-beta 1-/-) suffer from multifocal inflammation and die by 3-4 wk of age. In these mice, levels of nitric oxide (NO) reaction products in serum are elevated approximately fourfold over levels in controls, peaking at 15-17 d of life. Shortterm treatment of TGF-beta 1-/- mice with NG-monomethyl-L-arginine suppressed this elevated production of NO. Expression of inducible NO synthase (iNOS) mRNA and protein is increased in the kidney and heart of TGF-beta 1-/- mice. These findings demonstrate that TGF-beta 1 negatively regulates iNOS expression in vivo, as had been inferred from mechanistic studies on the control of iNOS expression by TGF-beta 1 in vitro..
Volpert, O.V.
Ward, W.F.
Lingen, M.W.
Chesler, L.
Solt, D.B.
Johnson, M.D.
Molteni, A.
Polverini, P.J.
Bouck, N.P.
(1996). Captopril inhibits angiogenesis and slows the growth of experimental tumors in rats. J clin invest,
Vol.98
(3),
pp. 671-679.
show abstract
Captopril, an inhibitor of angiotensin converting enzyme, is widely used clinically to manage hypertension and congestive heart failure. Here captopril is shown to be an inhibitor of angiogenesis able to block neovascularization induced in the rat cornea. Captopril acted directly and specifically on capillary endothelial cells, inhibiting their chemotaxis with a biphasic dose-response curve showing an initial decrease at clinically achievable doses under 10 microM and a further slow decline in the millimolar range. Captopril inhibition of endothelial cell migration was not mediated by angiotensin converting enzyme inhibition, but was suppressed by zinc. Direct inhibition by captopril of zinc-dependent endothelial cell-derived 72-and 92-kD metalloproteinases known to be essential for angiogenesis was also seen. When used systemically on rats captopril inhibited corneal neovascularization and showed the antitumor activity expected of an inhibitor of angiogenesis, decreasing the number of mitoses present in carcinogen-induced foci of preneoplastic liver cells and slowing the growth rate of an experimental fibrosarcoma whose cells were resistant to captopril in vitro. These data define this widely used drug as a new inhibitor of neovascularization and raise the possibility that patients on long term captopril therapy may derive unexpected benefits from its antiangiogenic activities..
Vodovotz, Y.
Lucia, M.S.
Flanders, K.C.
Chesler, L.
Xie, Q.W.
Smith, T.W.
Weidner, J.
Mumford, R.
Webber, R.
Nathan, C.
Roberts, A.B.
Lippa, C.F.
Sporn, M.B.
(1996). Inducible nitric oxide synthase in tangle-bearing neurons of patients with Alzheimer's disease. J exp med,
Vol.184
(4),
pp. 1425-1433.
show abstract
In Alzheimer's disease (AD), affected neurons accumulate beta amyloid protein, components of which can induce mouse microglia to express the high-output isoform of nitric oxide synthase (NOS2) in vitro. Products of NOS2 can be neurotoxic. In mice, NOS2 is normally suppressed by transforming growth factor beta 1 (TGF-beta 1). Expression of TGF-beta 1 is decreased in brains from AD patients, a situation that might be permissive for accumulation of NOS2. Accordingly, we investigated the expression of NOS2 in patients with AD, using three monospecific antibodies: a previously described polyclonal and two new monoclonal antibodies. Neurofibrillary tangle-bearing neurons and neuropil threads contained NOS2 in brains from each of 11 AD patients ranging in age from 47 to 81 years. NOS2 was undetectable in brains from 6 control subjects aged 23-72 years, but was expressed in small amounts in 3 control subjects aged 77-87 years. Thus, human neurons can express NOS2 in vivo. The high-output pathway of NO production may contribute to pathogenesis in AD..
Vodovotz, Y.
Letterio, J.J.
Geiser, A.G.
Chesler, L.
Roberts, A.B.
Sparrow, J.
(1996). Control of nitric oxide production by endogenous TGF-beta1 and systemic nitric oxide in retinal pigment epithelial cells and peritoneal macrophages. J leukoc biol,
Vol.60
(2),
pp. 261-270.
show abstract
Both in vivo and in vitro experiments demonstrate that transforming growth factor-beta1 (TGF-beta1) suppresses expression of the inducible form of nitric oxide synthase (iNOS). In this study, we examined the effects of exogenous and endogenous TGF-beta1 on retinal pigment epithelial (RPE) cells and resident peritoneal macrophages ex vivo using cells from TGF-beta1 null (TGF-beta1-/-) mice or age-matched wild-type (TGF-beta1+/+) or heterozygous (TGF-beta1+/-) littermates. RPE cells from both TGF-beta1-/- mice and TGF-beta1+/+ littermates produced NO and were immunocytochemically positive for iNOS protein only following treatment with interferon-gamma (IFN-gamma) and bacterial lipopolysaccharide (LPS); however, RPE cells from TGF-beta1-/- mice produced 40% more NO than cells from TGF-beta1+/+ mice. In contrast, resident peritoneal macrophages from both TGF-beta1+/+ and TGF-beta1-/- mice expressed iNOS protein without stimulation and in the absence of detectable production of NO. The expression of iNOS was increased by treatment with IFN-gamma, resulting in detectable levels of NO. Macrophages from TGF-beta1+/+ mice appeared to produce NO in a manner inversely proportional to the serum content of NO2- and NO3- of the mice from which the cells were obtained; no such correlation existed in TGF-beta1+/- or TGF-beta1-/- mice. Treatment of RPE cells or macrophages from both TGF-beta1+/+ and TGF-beta1-/- mice with exogenous TGF-beta1 decreased both iNOS protein and NO production. These findings demonstrate a novel role of endogenous TGF-beta1 in coupling systemic NO production to the production of NO by macrophages, and demonstrate that endogenous and exogenous TGF-beta1 can act differently to suppress NO production..
Cohn, S.L.
Look, A.T.
Joshi, V.V.
Holbrook, T.
Salwen, H.
Chagnovich, D.
Chesler, L.
Rowe, S.T.
Valentine, M.B.
Komuro, H.
(1995). Lack of correlation of N-myc gene amplification with prognosis in localized neuroblastoma: a Pediatric Oncology Group study. Cancer res,
Vol.55
(4),
pp. 721-726.
show abstract
Multiple copies of N-myc proto-oncogene are only rarely detected in localized neuroblastomas (NBs), and the prognostic relevance of amplification in this subset of patients is not clear. We analyzed a series of 850 children with NB admitted to a Pediatric Oncology Group NB Biology Study and identified six patients with localized NBs harboring N-myc gene amplification. Three patients whose tumors showed favorable histology by Shimada classification and low-risk histological features according to the Joshi classification have remained disease-free, whereas two of three patients with unfavorable histology tumors have developed recurrent disease. Although earlier studies have indicated that N-myc amplification is associated with diploid DNA content, flow cytometric analysis revealed that only two of the localized tumors contained stem lines with diploid DNA content. Loss of chromosome 1p was not detected by fluorescence in situ hybridization in the two tumors examined. N-myc protein was detected by immunohistochemical studies in four of the five NBs analyzed. However, N-myc protein was not visualized in one of the tumors with stroma-rich histology, and Western blot analysis revealed only low levels of N-myc protein expression in another NB with favorable histology. These studies indicate that the presence of N-myc amplification in localized NBs does not necessarily portend an adverse outcome. Furthermore, the biological features of this subset of N-myc-amplified NBs appear to differ from those of more advanced N-myc-amplified tumors..
Chesler, L.
Golde, D.W.
Bersch, N.
Johnson, M.D.
(1995). Metalloproteinase inhibition and erythroid potentiation are independent activities of tissue inhibitor of metalloproteinases-1. Blood,
Vol.86
(12),
pp. 4506-4515.
show abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1), the major physiological matrix metalloproteinase inhibitor and a potent antimetastatic factor, also stimulates the growth of erythroid progenitors (erythroid-potentiating activity). We analyzed the relationship between the growth factor activity and protease inhibition by preparing purified TIMP-1 "knockout" proteins lacking in vitro antiproteolytic activity. The growth-stimulatory effect of these N-terminal TIMP-1 point mutants, as tested in an in vitro assay using erythroid precursors (erythroid burst-forming units) was equal to that of unmutated TIMP-1. A fully antiproteolytic C-terminal TIMP-1 truncation also stimulated growth in the erythroid burst-forming unit assay. The results indicate that the influence of TIMP-1 on erythroid precursor growth is independent of its ability to inhibit metalloproteinases. TIMP-1 is analogous to proteins that have both proteolytic and growth factor activity, such as plasmin, thrombin, and urokinase. However, TIMP-1 is novel in this regard because it is a metalloproteinase inhibitor. We show that the antiproteolytic and growth factor activities of the TIMP-1 molecule are physically and functionally distinct..
Johnson, M.D.
Kim, H.R.
Chesler, L.
Tsao-Wu, G.
Bouck, N.
Polverini, P.J.
(1994). Inhibition of angiogenesis by tissue inhibitor of metalloproteinase. J cell physiol,
Vol.160
(1),
pp. 194-202.
show abstract
Matrix proteases play a critical role in cell invasion and migration, including the process of angiogenesis. The ability of specific factors to induce angiogenic responses correlates with their stimulation of matrix protease synthesis and release. Using an in vivo angiogenesis assay, the endothelial cell response to known angiogenic factors, basic fibroblast growth factor (bFGF) and adipocyte conditioned medium, was blocked by an inhibitor of matrix metalloproteinase activity, TIMP-1. The TIMP effect was mediated, at least in part, through the inhibition of endothelial cell migration, as determined by the ability of TIMP to block chemotaxis in a Boyden chamber assay. These results indicate that the inhibition of migration is a direct effect on the endothelial cells and does not require accessory cells. An additional observation was that the RNA levels for TIMP were significantly reduced in differentiated adipocytes, compared to undifferentiated F442A controls. Therefore, the acquisition of an angiogenic phenotype may involve not only the induction of positive factors, but also the suppression of angiogenesis inhibitors..
Jamin, Y.
Tucker, E.R.
Poon, E.S.
Popov, S.
Vaughan, L.
Boult, J.K.
Webber, H.
Hallsworth, A.
Baker, L.C.
Jones, C.
Koh, D.-.
Pearson, A.D.
Chesler, L.
Robinson, S.P.
Evaluation of clinically translatable magnetic resonance imaging biomarkers of therapeutic response in the TH-MYCN transgenic mouse model of neuroblastoma. Radiology,
.
full text