Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nat Commun ; 15(1): 3422, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653965

ABSTRACT

Targeting Anaplastic lymphoma kinase (ALK) is a promising therapeutic strategy for aberrant ALK-expressing malignancies including neuroblastoma, but resistance to ALK tyrosine kinase inhibitors (ALK TKI) is a distinct possibility necessitating drug combination therapeutic approaches. Using high-throughput, genome-wide CRISPR-Cas9 knockout screens, we identify miR-1304-5p loss as a desensitizer to ALK TKIs in aberrant ALK-expressing neuroblastoma; inhibition of miR-1304-5p decreases, while mimics of this miRNA increase the sensitivity of neuroblastoma cells to ALK TKIs. We show that miR-1304-5p targets NRAS, decreasing cell viability via induction of apoptosis. It follows that the farnesyltransferase inhibitor (FTI) lonafarnib in addition to ALK TKIs act synergistically in neuroblastoma, inducing apoptosis in vitro. In particular, on combined treatment of neuroblastoma patient derived xenografts with an FTI and an ALK TKI complete regression of tumour growth is observed although tumours rapidly regrow on cessation of therapy. Overall, our data suggests that combined use of ALK TKIs and FTIs, constitutes a therapeutic approach to treat high risk neuroblastoma although prolonged therapy is likely required to prevent relapse.


Subject(s)
Anaplastic Lymphoma Kinase , Dibenzocycloheptenes , Farnesyltranstransferase , GTP Phosphohydrolases , MicroRNAs , Neuroblastoma , Piperidines , Protein Kinase Inhibitors , Pyridines , Animals , Female , Humans , Mice , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Gene Expression Regulation, Neoplastic/drug effects , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Xenograft Model Antitumor Assays
2.
Cell Rep Med ; 5(3): 101472, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508140

ABSTRACT

Anaplastic large cell lymphoma (ALCL) is an aggressive, CD30+ T cell lymphoma of children and adults. ALK fusion transcripts or mutations in the JAK-STAT pathway are observed in most ALCL tumors, but the mechanisms underlying tumorigenesis are not fully understood. Here, we show that dysregulated STAT3 in ALCL cooccupies enhancers with master transcription factors BATF3, IRF4, and IKZF1 to form a core regulatory circuit that establishes and maintains the malignant cell state in ALCL. Critical downstream targets of this network in ALCL cells include the protooncogene MYC, which requires active STAT3 to facilitate high levels of MYC transcription. The core autoregulatory transcriptional circuitry activity is reinforced by MYC binding to the enhancer regions associated with STAT3 and each of the core regulatory transcription factors. Thus, activation of STAT3 provides the crucial link between aberrant tyrosine kinase signaling and the core transcriptional machinery that drives tumorigenesis and creates therapeutic vulnerabilities in ALCL.


Subject(s)
Lymphoma, Large-Cell, Anaplastic , Signal Transduction , Adult , Child , Humans , Signal Transduction/genetics , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Cell Transformation, Neoplastic , Carcinogenesis/genetics , STAT3 Transcription Factor/genetics
3.
Br J Haematol ; 202(5): 985-994, 2023 09.
Article in English | MEDLINE | ID: mdl-37357529

ABSTRACT

Anaplastic large-cell lymphoma (ALCL) is a T-cell malignancy predominantly driven by the oncogenic anaplastic lymphoma kinase (ALK), accounting for approximately 15% of all paediatric non-Hodgkin lymphoma. Patients with central nervous system (CNS) relapse are particularly difficult to treat with a 3-year overall survival of 49% and a median survival of 23.5 months. The second-generation ALK inhibitor brigatinib shows superior penetration of the blood-brain barrier unlike the first-generation drug crizotinib and has shown promising results in ALK+ non-small-cell lung cancer. However, the benefits of brigatinib in treating aggressive paediatric ALK+ ALCL are largely unknown. We established a patient-derived xenograft (PDX) resource from ALK+ ALCL patients at or before CNS relapse serving as models to facilitate the development of future therapies. We show in vivo that brigatinib is effective in inducing the remission of PDX models of crizotinib-resistant (ALK C1156Y, TP53 loss) ALCL and furthermore that it is superior to crizotinib as a second-line approach to the treatment of a standard chemotherapy relapsed/refractory ALCL PDX pointing to brigatinib as a future therapeutic option.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Lymphoma, Large-Cell, Anaplastic , Child , Humans , Anaplastic Lymphoma Kinase , Crizotinib/pharmacology , Crizotinib/therapeutic use , Receptor Protein-Tyrosine Kinases/therapeutic use , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/pathology , Heterografts , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use
4.
Br J Haematol ; 192(2): 354-365, 2021 01.
Article in English | MEDLINE | ID: mdl-32880915

ABSTRACT

Burkitt lymphoma (BL) accounts for almost two-thirds of all B-cell non-Hodgkin lymphoma (B-NHL) in children and adolescents and is characterised by a MYC translocation and rapid cell turnover. Intensive chemotherapeutic regimens have been developed in recent decades, including the lymphomes malins B (LMB) protocol, which have resulted in a survival rate in excess of 90%. Recent clinical trials have focused on immunochemotherapy, with the addition of rituximab to chemotherapeutic backbones, showing encouraging results. Despite these advances, relapse and refractory disease occurs in up to 10% of patients and salvage options for these carry a dismal prognosis. Efforts to better understand the molecular and functional characteristics driving relapse and refractory disease may help improve this prognosis. This study has established a paediatric BL patient-derived xenograft (PDX) resource which captures and maintains tumour heterogeneity, may be used to better characterise tumours and identify cell populations responsible for therapy resistance.


Subject(s)
Burkitt Lymphoma/pathology , Animals , Burkitt Lymphoma/genetics , Burkitt Lymphoma/therapy , Child , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Heterografts/pathology , Humans , Male , Mice , Neoplasm Transplantation , Tumor Cells, Cultured
5.
Haematologica ; 106(6): 1693-1704, 2021 06 01.
Article in English | MEDLINE | ID: mdl-32327503

ABSTRACT

Patients diagnosed with Anaplastic Large Cell Lymphoma (ALCL) are still treated with toxic multi-agent chemotherapy and as many as 25-50% of patients relapse. To understand disease pathology and to uncover novel targets for therapy, Whole-Exome Sequencing (WES) of Anaplastic Lymphoma Kinase (ALK)+ ALCL was performed as well as Gene-Set Enrichment Analysis. This revealed that the T-cell receptor (TCR) and Notch pathways were the most enriched in mutations. In particular, variant T349P of NOTCH1, which confers a growth advantage to cells in which it is expressed, was detected in 12% of ALK+ and ALK- ALCL patient samples. Furthermore, we demonstrate that NPM-ALK promotes NOTCH1 expression through binding of STAT3 upstream of NOTCH1. Moreover, inhibition of NOTCH1 with γ-secretase inhibitors (GSIs) or silencing by shRNA leads to apoptosis; co-treatment in vitro with the ALK inhibitor Crizotinib led to additive/synergistic anti-tumour activity suggesting this may be an appropriate combination therapy for future use in the circumvention of ALK inhibitor resistance. Indeed, Crizotinib-resistant and sensitive ALCL were equally sensitive to GSIs. In conclusion, we show a variant in the extracellular domain of NOTCH1 that provides a growth advantage to cells and confirm the suitability of the Notch pathway as a second-line druggable target in ALK+ ALCL.


Subject(s)
Lymphoma, Large-Cell, Anaplastic , Cell Line, Tumor , Humans , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/genetics , Mutation , Neoplasm Recurrence, Local , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Notch1/genetics , Exome Sequencing
6.
Blood ; 136(14): 1657-1669, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32573700

ABSTRACT

Anaplastic large cell lymphoma (ALCL) is a T-cell malignancy predominantly driven by a hyperactive anaplastic lymphoma kinase (ALK) fusion protein. ALK inhibitors, such as crizotinib, provide alternatives to standard chemotherapy with reduced toxicity and side effects. Children with lymphomas driven by nucleophosmin 1 (NPM1)-ALK fusion proteins achieved an objective response rate to ALK inhibition therapy of 54% to 90% in clinical trials; however, a subset of patients progressed within the first 3 months of treatment. The mechanism for the development of ALK inhibitor resistance is unknown. Through genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) activation and knockout screens in ALCL cell lines, combined with RNA sequencing data derived from ALK inhibitor-relapsed patient tumors, we show that resistance to ALK inhibition by crizotinib in ALCL can be driven by aberrant upregulation of interleukin 10 receptor subunit alpha (IL10RA). Elevated IL10RA expression rewires the STAT3 signaling pathway, bypassing otherwise critical phosphorylation by NPM1-ALK. IL-10RA expression does not correlate with response to standard chemotherapy in pediatric patients, suggesting that a combination of crizotinib and chemotherapy could prevent ALK inhibitor resistance-specific relapse.


Subject(s)
Antineoplastic Agents/pharmacology , Crizotinib/pharmacology , Drug Resistance, Neoplasm/genetics , Interleukin-10 Receptor alpha Subunit/genetics , Lymphoma, Large-Cell, Anaplastic/genetics , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Antineoplastic Agents/therapeutic use , CRISPR-Cas Systems , Cell Line , Crizotinib/therapeutic use , Dose-Response Relationship, Drug , Gene Editing , Gene Expression , Humans , Immunohistochemistry , Interleukin-10 Receptor alpha Subunit/metabolism , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Models, Biological , Nucleophosmin , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
7.
Nat Commun ; 10(1): 5428, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31780656

ABSTRACT

Resistance to anaplastic lymphoma kinase (ALK)-targeted therapy in ALK-positive non-small cell lung cancer has been reported, with the majority of acquired resistance mechanisms relying on bypass signaling. To proactively identify resistance mechanisms in ALK-positive neuroblastoma (NB), we herein employ genome-wide CRISPR activation screens of NB cell lines treated with brigatinib or ceritinib, identifying PIM1 as a putative resistance gene, whose high expression is associated with high-risk disease and poor survival. Knockdown of PIM1 sensitizes cells of differing MYCN status to ALK inhibitors, and in patient-derived xenografts of high-risk NB harboring ALK mutations, the combination of the ALK inhibitor ceritinib and PIM1 inhibitor AZD1208 shows significantly enhanced anti-tumor efficacy relative to single agents. These data confirm that PIM1 overexpression decreases sensitivity to ALK inhibitors in NB, and suggests that combined front-line inhibition of ALK and PIM1 is a viable strategy for the treatment of ALK-positive NB independent of MYCN status.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Neuroblastoma/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Anaplastic Lymphoma Kinase/genetics , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Mice , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/drug therapy , Organophosphorus Compounds/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Sulfones/pharmacology , Sulfones/therapeutic use , Thiazolidines/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...