Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Chem Biol ; 17(9): 954-963, 2021 09.
Article in English | MEDLINE | ID: mdl-33972797

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-myc/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
2.
Mol Cancer ; 21(1): 126, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35689207

ABSTRACT

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.


Subject(s)
Neuroblastoma , Precision Medicine , Anaplastic Lymphoma Kinase/genetics , Cell Line, Tumor , Child , Humans , Mutation , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
3.
Blood ; 136(10): 1155-1160, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32573723

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.


Subject(s)
Arginine/metabolism , Argininosuccinate Synthase/metabolism , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Neuroblastoma/therapy , Ornithine Carbamoyltransferase/metabolism , T-Lymphocytes/transplantation , Animals , Apoptosis , Argininosuccinate Synthase/genetics , Cell Proliferation , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Metabolic Engineering/methods , Mice , Mice, Nude , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Ornithine Carbamoyltransferase/genetics , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Brain ; 141(5): 1300-1319, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29490009

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-ß 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-ß 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-ß/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.


Subject(s)
Carrier Proteins/metabolism , Cerebellar Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/physiology , Medulloblastoma/metabolism , Neoplasm Metastasis/physiopathology , PTEN Phosphohydrolase/metabolism , Adolescent , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Infant , Male , Medulloblastoma/pathology , Mice , Mice, Inbred BALB C , Models, Molecular , Neoplasm Metastasis/genetics , PTEN Phosphohydrolase/genetics , Phosphoric Monoester Hydrolases , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
5.
Proc Natl Acad Sci U S A ; 113(48): 13726-13731, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27837025

ABSTRACT

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.


Subject(s)
Aurora Kinase A/chemistry , N-Myc Proto-Oncogene Protein/chemistry , Neoplasms/drug therapy , SKP Cullin F-Box Protein Ligases/chemistry , Aurora Kinase A/genetics , Azepines/pharmacology , Binding Sites , Catalytic Domain/drug effects , Crystallography, X-Ray , Humans , N-Myc Proto-Oncogene Protein/genetics , Neoplasms/genetics , Neoplasms/pathology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Polyubiquitin/chemistry , Polyubiquitin/genetics , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , SKP Cullin F-Box Protein Ligases/genetics
6.
Genes Dev ; 24(10): 1059-72, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20478998

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.


Subject(s)
Gene Expression Regulation, Neoplastic , Medulloblastoma/physiopathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Cell Cycle/physiology , Cellular Senescence/physiology , Cerebellum/metabolism , Down-Regulation , Gene Expression Profiling , Genomic Instability , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/pathology , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics
7.
Lancet Oncol ; 18(7): e394-e404, 2017 07.
Article in English | MEDLINE | ID: mdl-28677575

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Legislation, Drug , Neoplasms/drug therapy , Precision Medicine , Adolescent , Antineoplastic Agents/pharmacology , Biological Products/therapeutic use , Child , Child, Preschool , Drug Discovery/legislation & jurisprudence , Europe , Humans , Infant , Infant, Newborn
8.
Br J Cancer ; 117(6): 791-800, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28787429

ABSTRACT

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.


Subject(s)
Magnetic Resonance Imaging/methods , Magnets , Neuroblastoma/diagnostic imaging , Stomach Neoplasms/diagnostic imaging , Anaplastic Lymphoma Kinase , Anilides/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Contrast Media , Cyclophosphamide/therapeutic use , Disease Models, Animal , Female , Magnetic Resonance Imaging/instrumentation , Male , Mice , Mice, Transgenic , Mutation , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Phantoms, Imaging , Phenotype , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/genetics , Signal-To-Noise Ratio , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Burden/drug effects
9.
JCO Precis Oncol ; 8: e2300117, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38207228

ABSTRACT

Greater collaboration needed to realize potential of molecular profiling initiatives for pediatric cancers.


Subject(s)
Neoplasms , Humans , Child , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine
10.
Cell Genom ; 4(2): 100483, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38359786

ABSTRACT

The MRC National Mouse Genetics Network (NMGN) has been established in the UK to bring together researchers from academia and industry across the country from a wide range of disease areas and research backgrounds to rapidly facilitate clinical translation of mouse research findings and foster an environment of interdisciplinary learning.


Subject(s)
Industry , Animals , Mice
11.
Nat Commun ; 15(1): 3745, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702304

ABSTRACT

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.


Subject(s)
Cell Differentiation , DNA Copy Number Variations , N-Myc Proto-Oncogene Protein , Neural Crest , Neuroblastoma , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neural Crest/metabolism , Neural Crest/pathology , Female , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Chromosome Aberrations , Human Embryonic Stem Cells/metabolism , Transcriptome , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
12.
Cancer Cell ; 42(2): 283-300.e8, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38181797

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.


Subject(s)
B7-H1 Antigen , Neuroblastoma , Humans , Child , Neoplasm Recurrence, Local , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Receptors, Immunologic/genetics , Immunotherapy , Sequence Analysis, RNA
13.
J Clin Oncol ; 42(10): 1135-1145, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38190578

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.


Subject(s)
Neuroblastoma , Topotecan , Child , Humans , Infant , Child, Preschool , Adolescent , Young Adult , Adult , Temozolomide/therapeutic use , Irinotecan/therapeutic use , Topotecan/adverse effects , Bevacizumab/adverse effects , Dacarbazine/adverse effects , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neuroblastoma/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects
14.
Radiology ; 266(1): 130-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23169794

ABSTRACT

PURPOSE: To evaluate noninvasive and clinically translatable magnetic resonance (MR) imaging biomarkers of therapeutic response in the TH-MYCN transgenic mouse model of aggressive, MYCN-amplified neuroblastoma. MATERIALS AND METHODS: All experiments were performed in accordance with the local ethical review panel and the UK Home Office Animals Scientific Procedures Act 1986 and with the UK National Cancer Research Institute guidelines for the welfare of animals in cancer research. Multiparametric MR imaging was performed of abdominal tumors found in the TH-MYCN model. T2-weighted MR imaging, quantitation of native relaxation times T1 and T2, the relaxation rate R2*, and dynamic contrast-enhanced MR imaging were used to monitor tumor response to cyclophosphamide (25 mg/kg), the vascular disrupting agent ZD6126 (200 mg/kg), or the antiangiogenic agent cediranib (6 mg/kg, daily). Any significant changes in the measured parameters, and in the magnitude of the changes after treatment between treated and control cohorts, were identified by using Student two-tailed paired and unpaired t test, respectively, with a 5% level of significance. RESULTS: Treatment with cyclophosphamide or cediranib induced a 54% or 20% reduction in tumor volume at 48 hours, respectively (P < .005 and P < .005, respectively; P < .005 and P < .005 versus control, respectively). Treatment with ZD6126 induced a 45% reduction in mean tumor volume 24 hours after treatment (P < .005; P < .005 versus control). The antitumor activity of cyclophosphamide, cediranib, and ZD6126 was consistently associated with a decrease in tumor T1 (P < .005, P < .005, and P < .005, respectively; P < .005, P < .005, and P < .005 versus control, respectively) and with a correlation between therapy-induced changes in native T1 and changes in tumor volume (r = 0.56; P < .005). Tumor response to cediranib was also associated with a decrease in the dynamic contrast-enhanced MR imaging-derived volume transfer constant (P = .07; P < .05 versus control) and enhancing fraction (P < .05; P < .01 versus control), and an increase in R2* (P < .005; P < .05 versus control). CONCLUSION: The T1 relaxation time is a robust noninvasive imaging biomarker of response to therapy in tumors in TH-MYCN mice, which emulate high-risk neuroblastoma in children. T1 measurements can be readily implemented on clinical MR systems and should be investigated in translational clinical trials of new targeted therapies for pediatric neuroblastoma. SUPPLEMENTAL MATERIAL: http://radiology.rsna.org/lookup/suppl/doi:10.1148/radiol.12120128/-/DC1.


Subject(s)
Antineoplastic Agents/therapeutic use , Disease Models, Animal , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Animals , Biomarkers , Humans , Magnetic Resonance Imaging , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Prognosis , Proto-Oncogene Proteins/genetics , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
15.
Semin Cancer Biol ; 21(4): 245-55, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21958944

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Neuroblastoma/genetics , Animals , Child , Genetic Engineering/methods , Genetic Engineering/trends , Humans , Mice , Neuroblastoma/pathology , Neuroblastoma/therapy , Pathology, Molecular/methods , Pathology, Molecular/trends
16.
Dev Cell ; 58(19): 1967-1982.e8, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37734383

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.


Subject(s)
Neuroblastoma , Piperazines , Pyridines , Tretinoin , Animals , Mice , Humans , Cell Line, Tumor , Cell Differentiation , Tretinoin/pharmacology , Neuroblastoma/drug therapy , Adrenergic Agents/therapeutic use
17.
Clin Cancer Res ; 29(7): 1317-1331, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36602782

ABSTRACT

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.


Subject(s)
Lung Neoplasms , Neuroblastoma , Mice , Animals , Humans , Anaplastic Lymphoma Kinase/genetics , Aminopyridines/therapeutic use , Lactams, Macrocyclic/pharmacology , Lactams, Macrocyclic/therapeutic use , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy
18.
Nat Med ; 29(5): 1092-1102, 2023 05.
Article in English | MEDLINE | ID: mdl-37012551

ABSTRACT

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 .


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Neuroblastoma , Adult , Humans , 3-Iodobenzylguanidine/therapeutic use , Aminopyridines/therapeutic use , Anaplastic Lymphoma Kinase/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Lactams, Macrocyclic/adverse effects , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Protein Kinase Inhibitors/therapeutic use , Child , Infant , Child, Preschool , Adolescent
19.
Front Pediatr ; 10: 957944, 2022.
Article in English | MEDLINE | ID: mdl-36467471

ABSTRACT

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.

20.
Cancer Gene Ther ; 29(7): 940-950, 2022 07.
Article in English | MEDLINE | ID: mdl-34522028

ABSTRACT

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.


Subject(s)
Biguanides , Neuroblastoma , Biguanides/therapeutic use , Humans , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/therapeutic use , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/therapeutic use , Polyamines/metabolism , Polyamines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL