Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Eur J Cancer ; 198: 113506, 2024 Feb.
Article En | MEDLINE | ID: mdl-38184928

BACKGROUND: Immune checkpoint inhibitors are frequently associated with the development of immunotherapy-related adverse events (irAEs). The exact etiology, including the role of environmental factors, remains incompletely understood. METHODS: We analyzed the records of 394 melanoma patients from three centers (northern and southern hemisphere). Patients had received at least one cycle of anti-PD-1/anti-CTLA-4 with a minimum follow-up of 3 months. We study the distribution and time to irAEs onset throughout the calendar year. RESULTS: 764 irAEs were recorded; the most frequent were skin rash (35%), hepatitis (32%) and colitis (30%). The irAEs incidence was the highest in autumn and winter, and the ratio for the 'number of irAEs' per 'therapies commenced' was the highest in winter and lowest in summer (2.4 and 1.7, respectively). Season-specific patterns in the time of irAEs onset were observed for pneumonitis (shorter time to onset in autumn, p = 0.025), hepatitis (shorter time to onset in spring, p = 0.016) and sarcoid-like immune reaction (shorter time to onset in autumn, p = 0.041). Season-specific patterns for early-onset irAEs were observed for hepatitis (spring, p = 0.023) and nephritis (summer, p = 0.017). Early-onset pneumonitis was more frequent in autumn-winter (p = 0.008) and early-onset nephritis in spring-summer (p = 0.004). CONCLUSIONS: Environmental factors that are associated with particular seasons may contribute to the development of certain irAEs and suggest the potential effect of environmental triggers. The identification of these factors may enhance preventive and therapeutic strategies to reduce the morbidity of irAEs.


Hepatitis , Immune Checkpoint Inhibitors , Immunotherapy , Melanoma , Nephritis , Pneumonia , Humans , Antibodies, Monoclonal/therapeutic use , Hepatitis/etiology , Immunotherapy/adverse effects , Ipilimumab/adverse effects , Melanoma/drug therapy , Nephritis/complications , Nephritis/drug therapy , Pneumonia/etiology , Seasons , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use
3.
J Exp Clin Cancer Res ; 42(1): 92, 2023 Apr 19.
Article En | MEDLINE | ID: mdl-37072838

BACKGROUND: One of the key limitations of targeted cancer therapies is the rapid onset of therapy resistance. Taking BRAF-mutant melanoma as paradigm, we previously identified the lipogenic regulator SREBP-1 as a central mediator of resistance to MAPK-targeted therapy. Reasoning that lipogenesis-mediated alterations in membrane lipid poly-unsaturation lie at the basis of therapy resistance, we targeted fatty acid synthase (FASN) as key player in this pathway to evoke an exquisite vulnerability to clinical inducers of reactive oxygen species (ROS), thereby rationalizing a novel clinically actionable combination therapy to overcome therapy resistance. METHODS: Using gene expression analysis and mass spectrometry-based lipidomics of BRAF-mutant melanoma cell lines, melanoma PDX and clinical data sets, we explored the association of FASN expression with membrane lipid poly-unsaturation and therapy-resistance. Next, we treated therapy-resistant models with a preclinical FASN inhibitor TVB-3664 and a panel of ROS inducers and performed ROS analysis, lipid peroxidation tests and real-time cell proliferation assays. Finally, we explored the combination of MAPK inhibitors, TVB-3664 and arsenic trioxide (ATO, as a clinically used ROS-inducer) in Mel006 BRAF mutant PDX as a gold model of therapy resistance and assessed the effect on tumor growth, survival and systemic toxicity. RESULTS: We found that FASN expression is consistently increased upon the onset of therapy resistance in clinical melanoma samples, in cell lines and in Mel006 PDX and is associated with decreased lipid poly-unsaturation. Forcing lipid poly-unsaturation in therapy-resistant models by combining MAPK inhibition with FASN inhibition attenuated cell proliferation and rendered cells exquisitely sensitive to a host of ROS inducers. In particular, the triple combination of MAPK inhibition, FASN inhibition, and the clinical ROS-inducing compound ATO dramatically increased survival of Mel006 PDX models from 15 to 72% with no associated signs of toxicity. CONCLUSIONS: We conclude that under MAPK inhibition the direct pharmacological inhibition of FASN evokes an exquisite vulnerability to inducers of ROS by increasing membrane lipid poly-unsaturation. The exploitation of this vulnerability by combining MAPK and/or FASN inhibitors with inducers of ROS greatly delays the onset of therapy resistance and increases survival. Our work identifies a clinically actionable combinatorial treatment for therapy-resistant cancer.


Melanoma , Proto-Oncogene Proteins B-raf , Humans , Reactive Oxygen Species/metabolism , Proto-Oncogene Proteins B-raf/genetics , Membrane Lipids/pharmacology , Membrane Lipids/therapeutic use , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm
4.
Cancer Discov ; 13(6): 1364-1385, 2023 06 02.
Article En | MEDLINE | ID: mdl-36977461

Understanding the evolutionary pathways to metastasis and resistance to immune-checkpoint inhibitors (ICI) in melanoma is critical for improving outcomes. Here, we present the most comprehensive intrapatient metastatic melanoma dataset assembled to date as part of the Posthumous Evaluation of Advanced Cancer Environment (PEACE) research autopsy program, including 222 exome sequencing, 493 panel-sequenced, 161 RNA sequencing, and 22 single-cell whole-genome sequencing samples from 14 ICI-treated patients. We observed frequent whole-genome doubling and widespread loss of heterozygosity, often involving antigen-presentation machinery. We found KIT extrachromosomal DNA may have contributed to the lack of response to KIT inhibitors of a KIT-driven melanoma. At the lesion-level, MYC amplifications were enriched in ICI nonresponders. Single-cell sequencing revealed polyclonal seeding of metastases originating from clones with different ploidy in one patient. Finally, we observed that brain metastases that diverged early in molecular evolution emerge late in disease. Overall, our study illustrates the diverse evolutionary landscape of advanced melanoma. SIGNIFICANCE: Despite treatment advances, melanoma remains a deadly disease at stage IV. Through research autopsy and dense sampling of metastases combined with extensive multiomic profiling, our study elucidates the many mechanisms that melanomas use to evade treatment and the immune system, whether through mutations, widespread copy-number alterations, or extrachromosomal DNA. See related commentary by Shain, p. 1294. This article is highlighted in the In This Issue feature, p. 1275.


Brain Neoplasms , Melanoma , Humans , Melanoma/pathology , Mutation , Evolution, Molecular , DNA
5.
Cell Rep Med ; 3(10): 100781, 2022 10 18.
Article En | MEDLINE | ID: mdl-36240755

Patients with blood cancer continue to have a greater risk of inadequate immune responses following three COVID-19 vaccine doses and risk of severe COVID-19 disease. In the context of the CAPTURE study (NCT03226886), we report immune responses in 80 patients with blood cancer who received a fourth dose of BNT162b2. We measured neutralizing antibody titers (NAbTs) using a live virus microneutralization assay against wild-type (WT), Delta, and Omicron BA.1 and BA.2 and T cell responses against WT and Omicron BA.1 using an activation-induced marker (AIM) assay. The proportion of patients with detectable NAb titers and T cell responses after the fourth vaccine dose increased compared with that after the third vaccine dose. Patients who received B cell-depleting therapies within the 12 months before vaccination have the greatest risk of not having detectable NAbT. In addition, we report immune responses in 57 patients with breakthrough infections after vaccination.


COVID-19 Vaccines , COVID-19 , Neoplasms , Humans , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , Clinical Studies as Topic , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Immunity , SARS-CoV-2
6.
Oncol Res Treat ; 45(10): 618-621, 2022.
Article En | MEDLINE | ID: mdl-35882208

INTRODUCTION: Patients with metastatic soft-tissue sarcoma generally have a poor prognosis. Although chemotherapy is associated with improved overall survival in these patients, long-term benefit is rare. CASE REPORT: We report on 2 patients with metastatic soft-tissue sarcoma who achieved a complete response to chemotherapy, without any local therapy, and had no signs of disease progression more than a decade after discontinuing treatment. CONCLUSION: These observations highlight the potential benefit of chemotherapy in a disease that is often considered to be poorly responsive to this treatment. These observations support further efforts to identify factors to guide patient selection and treatment optimization.


Neoplasms, Second Primary , Sarcoma , Soft Tissue Neoplasms , Antineoplastic Combined Chemotherapy Protocols , Humans , Prognosis , Soft Tissue Neoplasms/pathology
7.
Eur J Cancer ; 169: 210-222, 2022 07.
Article En | MEDLINE | ID: mdl-35644725

PURPOSE: Guidelines addressing melanoma in-transit metastasis (ITM) recommend immune checkpoint inhibitors (ICI) as a first-line treatment option, despite the fact that there are no efficacy data available from prospective trials for exclusively ITM disease. The study aims to analyze the outcome of patients with ITM treated with ICI based on data from a large cohort of patients treated at international referral clinics. METHODS: A multicenter retrospective cohort study of patients treated between January 2015 and December 2020 from Australia, Europe, and the USA, evaluating treatment with ICI for ITM with or without nodal involvement (AJCC8 N1c, N2c, and N3c) and without distant disease (M0). Treatment was with PD-1 inhibitor (nivolumab or pembrolizumab) and/or CTLA-4 inhibitor (ipilimumab). The response was evaluated according to the RECIST criteria modified for cutaneous lesions. RESULTS: A total of 287 patients from 21 institutions in eight countries were included. Immunotherapy was first-line treatment in 64 (22%) patients. PD-1 or CTLA-4 inhibitor monotherapy was given in 233 (81%) and 23 (8%) patients, respectively, while 31 (11%) received both in combination. The overall response rate was 56%, complete response (CR) rate was 36%, and progressive disease (PD) rate was 32%. Median PFS was ten months (95% CI 7.4-12.6 months) with a one-, two-, and five-year PFS rate of 48%, 33%, and 18%, respectively. Median MSS was not reached, and the one-, two-, and five-year MSS rates were 95%, 83%, and 71%, respectively. CONCLUSION: Systemic immunotherapy is an effective treatment for melanoma ITM. Future studies should evaluate the role of systemic immunotherapy in the context of multimodality therapy, including locoregional treatments such as surgery, intralesional therapy, and regional therapies.


Immune Checkpoint Inhibitors , Melanoma , Humans , Immune Checkpoint Inhibitors/therapeutic use , Ipilimumab/therapeutic use , Melanoma/pathology , Prospective Studies , Retrospective Studies
8.
Cancer Res ; 82(10): 1849-1857, 2022 05 16.
Article En | MEDLINE | ID: mdl-35476646

Cancer is an evolutionary process that is characterized by the emergence of multiple genetically distinct populations or clones within the primary tumor. Intratumor heterogeneity provides a substrate for the selection of adaptive clones, such as those that lead to metastasis. Comparative molecular studies of primary tumors and metastases have identified distinct genomic features associated with the development of metastases. In this review, we discuss how these insights could inform clinical decision-making and uncover rational antimetastasis treatment strategies.


Neoplasms , Evolution, Molecular , Genetic Heterogeneity , Humans , Mutation , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology
11.
Cancer Cell ; 40(2): 114-116, 2022 02 14.
Article En | MEDLINE | ID: mdl-34968417
12.
Cancer Cell ; 39(8): 1135-1149.e8, 2021 08 09.
Article En | MEDLINE | ID: mdl-34143978

Therapy resistance arises from heterogeneous drug-tolerant persister cells or minimal residual disease (MRD) through genetic and nongenetic mechanisms. A key question is whether specific molecular features of the MRD ecosystem determine which of these two distinct trajectories will eventually prevail. We show that, in melanoma exposed to mitogen-activated protein kinase therapeutics, emergence of a transient neural crest stem cell (NCSC) population in MRD concurs with the development of nongenetic resistance. This increase relies on a glial cell line-derived neurotrophic factor-dependent signaling cascade, which activates the AKT survival pathway in a focal adhesion kinase (FAK)-dependent manner. Ablation of the NCSC population through FAK inhibition delays relapse in patient-derived tumor xenografts. Strikingly, all tumors that ultimately escape this treatment exhibit resistance-conferring genetic alterations and increased sensitivity to extracellular signal-regulated kinase inhibition. These findings identify an approach that abrogates the nongenetic resistance trajectory in melanoma and demonstrate that the cellular composition of MRD deterministically imposes distinct drug resistance evolutionary paths.


Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Melanoma/drug therapy , Melanoma/genetics , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Imidazoles/pharmacology , Melanoma/pathology , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasm Recurrence, Local/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neural Crest/pathology , Oximes/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Pyridones/pharmacology , Pyrimidinones/pharmacology , Xenograft Model Antitumor Assays
13.
J Immunother Cancer ; 9(1)2021 01.
Article En | MEDLINE | ID: mdl-33468556

BACKGROUND: Patients with cancer who are infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are more likely to develop severe illness and die compared with those without cancer. The impact of immune checkpoint inhibition (ICI) on the severity of COVID-19 illness is unknown. The aim of this study was to investigate whether ICI confers an additional risk for severe COVID-19 in patients with cancer. METHODS: We analyzed data from 110 patients with laboratory-confirmed SARS-CoV-2 while on treatment with ICI without chemotherapy in 19 hospitals in North America, Europe and Australia. The primary objective was to describe the clinical course and to identify factors associated with hospital and intensive care (ICU) admission and mortality. FINDINGS: Thirty-five (32%) patients were admitted to hospital and 18 (16%) died. All patients who died had advanced cancer, and only four were admitted to ICU. COVID-19 was the primary cause of death in 8 (7%) patients. Factors independently associated with an increased risk for hospital admission were ECOG ≥2 (OR 39.25, 95% CI 4.17 to 369.2, p=0.0013), treatment with combination ICI (OR 5.68, 95% CI 1.58 to 20.36, p=0.0273) and presence of COVID-19 symptoms (OR 5.30, 95% CI 1.57 to 17.89, p=0.0073). Seventy-six (73%) patients interrupted ICI due to SARS-CoV-2 infection, 43 (57%) of whom had resumed at data cut-off. INTERPRETATION: COVID-19-related mortality in the ICI-treated population does not appear to be higher than previously published mortality rates for patients with cancer. Inpatient mortality of patients with cancer treated with ICI was high in comparison with previously reported rates for hospitalized patients with cancer and was due to COVID-19 in almost half of the cases. We identified factors associated with adverse outcomes in ICI-treated patients with COVID-19.


COVID-19/epidemiology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , COVID-19/immunology , COVID-19/virology , Cohort Studies , Female , Humans , Immune Checkpoint Inhibitors/adverse effects , Male , Middle Aged , Neoplasms/immunology , Retrospective Studies , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification
14.
Nat Commun ; 10(1): 2723, 2019 06 20.
Article En | MEDLINE | ID: mdl-31222014

Non-genetic drug resistance is increasingly recognised in various cancers. Molecular insights into this process are lacking and it is unknown whether stable non-genetic resistance can be overcome. Using single cell RNA-sequencing of paired drug naïve and resistant AML patient samples and cellular barcoding in a unique mouse model of non-genetic resistance, here we demonstrate that transcriptional plasticity drives stable epigenetic resistance. With a CRISPR-Cas9 screen we identify regulators of enhancer function as important modulators of the resistant cell state. We show that inhibition of Lsd1 (Kdm1a) is able to overcome stable epigenetic resistance by facilitating the binding of the pioneer factor, Pu.1 and cofactor, Irf8, to nucleate new enhancers that regulate the expression of key survival genes. This enhancer switching results in the re-distribution of transcriptional co-activators, including Brd4, and provides the opportunity to disable their activity and overcome epigenetic resistance. Together these findings highlight key principles to help counteract non-genetic drug resistance.


Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Trans-Activators/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Epigenesis, Genetic/drug effects , Female , HEK293 Cells , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA , Single-Cell Analysis , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
15.
Nat Commun ; 9(1): 4775, 2018 11 14.
Article En | MEDLINE | ID: mdl-30429474

BRAF inhibitors target the BRAF-V600E/K mutated kinase, the driver mutation found in 50% of cutaneous melanoma. They give unprecedented anti-tumor responses but acquisition of resistance ultimately limits their clinical benefit. The master regulators driving the expression of resistance-genes remain poorly understood. Here, we demonstrate that the Aryl hydrocarbon Receptor (AhR) transcription factor is constitutively activated in a subset of melanoma cells, promoting the dedifferentiation of melanoma cells and the expression of BRAFi-resistance genes. Typically, under BRAFi pressure, death of BRAFi-sensitive cells leads to an enrichment of a small subpopulation of AhR-activated and BRAFi-persister cells, responsible for relapse. Also, differentiated and BRAFi-sensitive cells can be redirected towards an AhR-dependent resistant program using AhR agonists. We thus identify Resveratrol, a clinically compatible AhR-antagonist that abrogates deleterious AhR sustained-activation. Combined with BRAFi, Resveratrol reduces the number of BRAFi-resistant cells and delays tumor growth. We thus propose AhR-impairment as a strategy to overcome melanoma resistance.


Drug Resistance, Neoplasm/genetics , Melanoma/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Receptors, Aryl Hydrocarbon/genetics , Skin Neoplasms/genetics , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Imidazoles/pharmacology , MCF-7 Cells , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, SCID , Molecular Docking Simulation , Mutation , Oximes/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Resveratrol/pharmacology , Resveratrol/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Transcription Factors , Tumor Burden/drug effects , Vemurafenib/therapeutic use , Xenograft Model Antitumor Assays
16.
Cell ; 174(4): 843-855.e19, 2018 08 09.
Article En | MEDLINE | ID: mdl-30017245

Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.


Gene Expression Regulation, Neoplastic/drug effects , Melanoma/drug therapy , Neoplasm, Residual/drug therapy , Neoplastic Stem Cells/drug effects , Neural Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Retinoid X Receptor gamma/antagonists & inhibitors , Animals , Biomarkers, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Male , Melanoma/metabolism , Melanoma/pathology , Mice, SCID , Mutation , Neoplasm, Residual/metabolism , Neoplasm, Residual/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Nat Commun ; 9(1): 2500, 2018 06 27.
Article En | MEDLINE | ID: mdl-29950559

Whereas significant anti-tumor responses are observed in most BRAFV600E-mutant melanoma patients exposed to MAPK-targeting agents, resistance almost invariably develops. Here, we show that in therapy-responsive cells BRAF inhibition induces downregulation of the processing of Sterol Regulator Element Binding (SREBP-1) and thereby lipogenesis. Irrespective of the escape mechanism, therapy-resistant cells invariably restore this process to promote lipid saturation and protect melanoma from ROS-induced damage and lipid peroxidation. Importantly, pharmacological SREBP-1 inhibition sensitizes BRAFV600E-mutant therapy-resistant melanoma to BRAFV600E inhibitors both in vitro and in a pre-clinical PDX in vivo model. Together, these data indicate that targeting SREBP-1-induced lipogenesis may offer a new avenue to overcome acquisition of resistance to BRAF-targeted therapy. This work also provides evidence that targeting vulnerabilities downstream of oncogenic signaling offers new possibilities in overcoming resistance to targeted therapies.


Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Lipogenesis/genetics , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Lipogenesis/drug effects , Male , Melanocytes , Melanoma/genetics , Mice , Mice, Nude , Mice, SCID , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Pyridines/pharmacology , Signal Transduction/genetics , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/genetics , Thiazoles/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays
18.
Neuro Oncol ; 20(6): 818-825, 2018 05 18.
Article En | MEDLINE | ID: mdl-29409029

Background: Neurofibromatosis 1 (NF1) leads to the development of benign and malignant peripheral nerve sheath tumors (MPNST). MPNST have been described to develop in preexisting benign plexiform neurofibromas (PN) and have a poor prognosis. Atypical neurofibromas (ANF) were recently described as precursor lesions for MPNST, making early detection and management of ANF a possible strategy to prevent MPNST. We aimed to clinically characterize ANF and identify management approaches. Methods: We analyzed clinical, imaging, and pathology findings of all patients with NF1 and ANF at 3 institutions. Results: Sixty-three patients had 76 ANF (32M/31F; median age 27.1 y). On MRI, most ANF appeared as distinct nodular lesions and were 18F-fluorodeoxyglucose (FDG) avid. Forty-six ANF were associated with pain, 19 with motor weakness, 45 were palpable or visible, and 13 had no clinical signs. Completely resected ANF (N = 57) have not recurred (median follow-up, 4.1 y; range, 0-14 y). Four ANF transformed into MPNST and 17 patients had a history of MPNST in a different location than was their ANF. Conclusions: Growth of distinct nodular lesions, pain, and FDG-PET avidity should raise concern for ANF in NF1. Patients with ANF are at greater risk for development of MPNST. Complete resection of ANF may prevent development of MPNST.


Magnetic Resonance Imaging/methods , Neurofibroma/pathology , Neurofibromatosis 1/pathology , Neurofibrosarcoma/pathology , Positron-Emission Tomography/methods , Tomography, X-Ray Computed/methods , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Neurofibroma/complications , Neurofibroma/diagnostic imaging , Neurofibromatosis 1/diagnostic imaging , Neurofibromatosis 1/etiology , Neurofibrosarcoma/diagnostic imaging , Neurofibrosarcoma/etiology , Prognosis , Young Adult
19.
Nat Cell Biol ; 19(11): 1348-1357, 2017 Nov.
Article En | MEDLINE | ID: mdl-28991221

Competition among RNAs to bind miRNA is proposed to influence biological systems. However, the role of this competition in disease onset is unclear. Here, we report that TYRP1 mRNA, in addition to encoding tyrosinase-related protein 1 (TYRP1), indirectly promotes cell proliferation by sequestering miR-16 on non-canonical miRNA response elements. Consequently, the sequestered miR-16 is no longer able to repress its mRNA targets, such as RAB17, which is involved in melanoma cell proliferation and tumour growth. Restoration of miR-16 tumour-suppressor function can be achieved in vitro by silencing TYRP1 or increasing miR-16 expression. Importantly, TYRP1-dependent miR-16 sequestration can also be overcome in vivo by using small oligonucleotides that mask miR-16-binding sites on TYRP1 mRNA. Together, our findings assign a pathogenic non-coding function to TYRP1 mRNA and highlight miRNA displacement as a promising targeted therapeutic approach for melanoma.


Cell Proliferation/genetics , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins/genetics , Oxidoreductases/genetics , RNA, Messenger/genetics , Animals , Binding Sites/genetics , Cell Line, Tumor , Female , Humans , Mice , MicroRNAs/genetics
20.
Melanoma Res ; 27(5): 507-510, 2017 10.
Article En | MEDLINE | ID: mdl-28800030

Concurrent BRAF-MEK inhibition improves clinical outcomes in patients with advanced BRAF V600E/K-mutant melanoma. There is currently less evidence for the efficacy of this treatment in patients with rare BRAF non-V600E/K genotypes. We report on two patients with rare BRAF exon 15 mutations - BRAF A598_T599insV and V600_K601delinsE - obtaining clinical benefit and a radiological response to inhibitors directed against the mitogen-activated protein kinase pathway. This highlights the importance of using tests that detect both V600E/K and non-V600E/K BRAF mutations to keep open the possibility of treatment with targeted therapy in patients with uncommon, yet potentially actionable, BRAF exon 15 mutations.


Exons , Melanoma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Disease-Free Survival , Humans , Male , Melanoma/enzymology , Melanoma/pathology , Skin Neoplasms/enzymology , Skin Neoplasms/pathology
...