Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Sci Adv ; 10(18): eadk4946, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38691597

ABSTRACT

This phase 2a trial investigated the efficacy of NFX-179 Topical Gel, a metabolically labile MEK inhibitor, in the treatment of cutaneous neurofibromas (cNFs) in neurofibromatosis type 1. Forty-eight participants were randomized to four treatment arms: NFX-179 Topical Gel 0.05%, 0.15%, and 0.5% or vehicle applied once daily to five target cNFs for 28 days. Treatment with NFX-179 Topical Gel resulted in a dose-dependent reduction in p-ERK levels in cNFs at day 28, with a 47% decrease in the 0.5% NFX-179 group compared to the vehicle (P = 0.0001). No local or systemic toxicities were observed during the treatment period, and systemic concentrations of NFX-179 remained below 1 ng/ml. In addition, 20% of cNFs treated with 0.5% NFX-179 Topical Gel showed a ≥50% reduction in volume compared to 6% in the vehicle group by ruler measurement with calculated volume (P = 0.021). Thus, NFX-179 Topical Gel demonstrated significant inhibition of MEK in cNF with excellent safety and potential therapeutic benefit.


Subject(s)
Neurofibromatosis 1 , Protein Kinase Inhibitors , Skin Neoplasms , Humans , Neurofibromatosis 1/drug therapy , Female , Male , Adult , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/adverse effects , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Neurofibroma/drug therapy , Neurofibroma/pathology , Neurofibroma/metabolism , Young Adult , Adolescent , Treatment Outcome , Administration, Topical , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism
2.
JCI Insight ; 9(3)2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38175707

ABSTRACT

Cutaneous neurofibromas (cNFs) are benign Schwann cell (SC) tumors arising from subepidermal glia. Individuals with neurofibromatosis type 1 (NF1) may develop thousands of cNFs, which greatly affect their quality of life. cNF growth is driven by the proliferation of NF1-/- SCs and their interaction with the NF1+/- microenvironment. We analyzed the crosstalk between human cNF-derived SCs and fibroblasts (FBs), identifying an expression signature specific to the SC-FB interaction. We validated the secretion of proteins involved in immune cell migration, suggesting a role of SC-FB crosstalk in immune cell recruitment. The signature also captured components of developmental signaling pathways, including the cAMP elevator G protein-coupled receptor 68 (GPR68). Activation of Gpr68 by ogerin in combination with the MEK inhibitor (MEKi) selumetinib reduced viability and induced differentiation and death of human cNF-derived primary SCs, a result corroborated using an induced pluripotent stem cell-derived 3D neurofibromasphere model. Similar results were obtained using other Gpr68 activators or cAMP analogs/adenylyl cyclase activators in combination with selumetinib. Interestingly, whereas primary SC cultures restarted their proliferation after treatment with selumetinib alone was stopped, the combination of ogerin-selumetinib elicited a permanent halt on SC expansion that persisted after drug removal. These results indicate that unbalancing the Ras and cAMP pathways by combining MEKi and cAMP elevators could be used as a potential treatment for cNFs.


Subject(s)
Neurofibroma , Neurofibromatosis 1 , Skin Neoplasms , Triazines , Humans , Quality of Life , Neurofibroma/drug therapy , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/pathology , Benzyl Alcohols , Skin Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment , Receptors, G-Protein-Coupled
3.
Transl Res ; 261: 16-27, 2023 11.
Article in English | MEDLINE | ID: mdl-37331503

ABSTRACT

Cutaneous neurofibromas (cNFs) are a hallmark of patients with the neurofibromatosis type 1 (NF1) genetic disorder. These benign nerve sheath tumors, which can amount to thousands, develop from puberty onward, often cause pain and are considered by patients to be the primary burden of the disease. Mutations of NF1, encoding a negative regulator of the RAS signaling pathway, in the Schwann cell (SCs) lineage are considered to be at the origin of cNFs. The mechanisms governing cNFs development are poorly understood, and therapeutics to reduce cNFs are missing, mainly due to the lack of appropriate animal models. To address this, we designed the Nf1-KO mouse model that develops cNFs. Using this model, we found that cNFs development is a singular event and goes through 3 successive stages: initiation, progression, and stabilization characterized by changes in the proliferative and MAPK activities of tumor SCs. We found that skin trauma accelerated the development of cNFs and further used this model to explore the efficacy of the MEK inhibitor binimetinib to cure these tumors. We showed that while topically delivered binimetinib has a selective and minor effect on mature cNFs, the same drug prevents their development over long periods.


Subject(s)
Neurofibroma , Neurofibromatosis 1 , Skin Neoplasms , Humans , Mice , Animals , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibroma/drug therapy , Neurofibroma/genetics , Benzimidazoles , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/prevention & control , Protein Kinase Inhibitors/pharmacology , Mitogen-Activated Protein Kinases
4.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37140985

ABSTRACT

Neurofibromatosis type 1 (NF1) is one of the most common tumor-predisposing genetic disorders. Neurofibromas are NF1-associated benign tumors. A hallmark feature of neurofibromas is an abundant collagen-rich extracellular matrix (ECM) that constitutes more than 50% of the tumor dry weight. However, little is known about the mechanism underlying ECM deposition during neurofibroma development and treatment response. We performed a systematic investigation of ECM enrichment during plexiform neurofibroma (pNF) development and identified basement membrane (BM) proteins, rather than major collagen isoforms, as the most upregulated ECM component. Following MEK inhibitor treatment, the ECM profile displayed an overall downregulation signature, suggesting ECM reduction as a therapeutic benefit of MEK inhibition. Through these proteomic studies, TGF-ß1 signaling was identified as playing a role in ECM dynamics. Indeed, TGF-ß1 overexpression promoted pNF progression in vivo. Furthermore, by integrating single-cell RNA sequencing, we found that immune cells including macrophages and T cells produce TGF-ß1 to induce Schwann cells to produce and deposit BM proteins for ECM remodeling. Following Nf1 loss, neoplastic Schwann cells further increased BM protein deposition in response to TGF-ß1. Our data delineate the regulation governing ECM dynamics in pNF and suggest that BM proteins could serve as biomarkers for disease diagnosis and treatment response.


Subject(s)
Neurofibroma , Neurofibromatosis 1 , Humans , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/complications , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Membrane Proteins/metabolism , Proteomics , Neurofibroma/drug therapy , Neurofibroma/genetics , Protein Kinase Inhibitors , Collagen/metabolism , Basement Membrane/metabolism , Basement Membrane/pathology , Extracellular Matrix/metabolism , Mitogen-Activated Protein Kinase Kinases , Schwann Cells/pathology
5.
J Pharmacol Exp Ther ; 385(2): 106-116, 2023 05.
Article in English | MEDLINE | ID: mdl-36849412

ABSTRACT

Individuals with neurofibromatosis type 1 develop rat sarcoma virus (RAS)-mitogen-activated protein kinase-mitogen-activated and extracellular signal-regulated kinase (RAS-MAPK-MEK)-driven nerve tumors called neurofibromas. Although MEK inhibitors transiently reduce volumes of most plexiform neurofibromas in mouse models and in neurofibromatosis type 1 (NF1) patients, therapies that increase the efficacy of MEK inhibitors are needed. BI-3406 is a small molecule that prevents Son of Sevenless (SOS)1 interaction with Kirsten rat sarcoma viral oncoprotein (KRAS)-GDP, interfering with the RAS-MAPK cascade upstream of MEK. Single agent SOS1 inhibition had no significant effect in the DhhCre;Nf1 fl/fl mouse model of plexiform neurofibroma, but pharmacokinetics (PK)-driven combination of selumetinib with BI-3406 significantly improved tumor parameters. Tumor volumes and neurofibroma cell proliferation, reduced by MEK inhibition, were further reduced by the combination. Neurofibromas are rich in ionized calcium binding adaptor molecule 1 (Iba1)+ macrophages; combination treatment resulted in small and round macrophages, with altered cytokine expression indicative of altered activation. The significant effects of MEK inhibitor plus SOS1 inhibition in this preclinical study suggest potential clinical benefit of dual targeting of the RAS-MAPK pathway in neurofibromas. SIGNIFICANCE STATEMENT: Interfering with the RAS-mitogen-activated protein kinase (RAS-MAPK) cascade upstream of mitogen activated protein kinase kinase (MEK), together with MEK inhibition, augment effects of MEK inhibition on neurofibroma volume and tumor macrophages in a preclinical model system. This study emphasizes the critical role of the RAS-MAPK pathway in controlling tumor cell proliferation and the tumor microenvironment in benign neurofibromas.


Subject(s)
Neurofibroma, Plexiform , Neurofibroma , Neurofibromatosis 1 , Animals , Mice , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases , Neurofibroma/drug therapy , Neurofibroma, Plexiform/drug therapy , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/therapeutic use , Tumor Microenvironment , SOS1 Protein/metabolism
6.
Photobiomodul Photomed Laser Surg ; 39(6): 411-417, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33470897

ABSTRACT

Background: Neurofibromatosis type 1 (NF1) has no current effective treatments beyond surgery. Topical photodynamic therapy (PDT) has the potential to provide a less invasive treatment modality. Objective: Based on murine data, we hypothesized PDT could be used for the treatment of cutaneous neurofibromas (cNF). Methods and results: We conducted a phase I trial to examine absorption and conversion of topical aminolevulinic acid (ALA) in cNF and determine safety in a dose escalation study. ALA or control vehicle was applied to neurofibromas through microneedle-assisted delivery (n = 4) and excised specimens were examined 24 h later for protoporphyrin IX fluorescence. Fluorescence was detected in the tumors at 304 ± 94 U/µm2, while adjacent paralesional normal skin and vehicle-treated tumors showed no fluorescence (p < 0.0001). Subsequently, neurofibromas (n = 27) were treated with ALA and irradiated with 633 nm red light 18 h later, at escalating dosages of 50 and 100 mJ/cm2. Maximum tolerable dose was established at 100 mJ/cm2. Light microscopy study of tumors biopsied 48 h after PDT (ALA n = 14 and vehicle n = 4) showed mixed inflammatory infiltrate in the ALA, but not in the vehicle-treated tumors or perilesional normal skin. TUNEL evaluation showed 42.5 ± 19.9 apoptotic cells per visual field for ALA-treated and 1.1 ± 1.4 for vehicle-treated tumors (p = 0.002). Conclusions: In the first reported clinical trial of PDT for NF1, PDT targeted neurofibromas specifically, and may offer a normal tissue-sparing treatment modality in the future. This study is registered at Clintrials.gov (NCT01682811).


Subject(s)
Neurofibroma , Photochemotherapy , Skin Neoplasms , Aminolevulinic Acid/therapeutic use , Animals , Lighting , Mice , Neurofibroma/drug therapy , Skin Neoplasms/drug therapy
7.
Cancer Res ; 80(23): 5367-5379, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33032988

ABSTRACT

Loss of the RAS GTPase-activating protein (RAS-GAP) NF1 drives aberrant activation of RAS/MEK/ERK signaling and other effector pathways in the majority of malignant peripheral nerve sheath tumors (MPNST). These dysregulated pathways represent potential targets for therapeutic intervention. However, studies of novel single agents including MEK inhibitors (MEKi) have demonstrated limited efficacy both preclinically and clinically, with little advancement in overall patient survival. By interrogation of kinome activity through an unbiased screen and targeted evaluation of the signaling response to MEK inhibition, we have identified global activation of upstream receptor tyrosine kinases (RTK) that converges on activation of RAS as a mechanism to limit sensitivity to MEK inhibition. As no direct inhibitors of pan-RAS were available, an inhibitor of the protein tyrosine phosphatase SHP2, a critical mediator of RAS signal transduction downstream of multiple RTK, represented an alternate strategy. The combination of MEKi plus SHP099 was superior to MEKi alone in models of NF1-MPNST, including those with acquired resistance to MEKi. Our findings have immediate translational implications and may inform future clinical trials for patients with MPNST harboring alterations in NF1. SIGNIFICANCE: Combined inhibition of MEK and SHP2 is effective in models of NF1-MPNST, both those naïve to and those resistant to MEKi, as well as in the MPNST precursor lesion plexiform neurofibroma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nerve Sheath Neoplasms/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Sheath Neoplasms/metabolism , Neurofibroma/drug therapy , Neurofibroma/pathology , Neurofibromin 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Pyridones/administration & dosage , Pyridones/pharmacology , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacology , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays , ras Proteins/metabolism
8.
Pediatr Dermatol ; 36(3): 360-361, 2019 May.
Article in English | MEDLINE | ID: mdl-30828862

ABSTRACT

A 14-year-old boy with neurofibromatosis type I (NF1) presented with a painful neurofibroma on his right palm. The lesion was treated with topical sirolimus, resulting in decreased size and pain and improvement in motor function of his hand. This case demonstrates the efficacy of topical sirolimus in the management of neurofibromas in NF1.


Subject(s)
Immunosuppressive Agents/therapeutic use , Neurofibroma/drug therapy , Neurofibromatosis 1/complications , Sirolimus/therapeutic use , Skin Neoplasms/drug therapy , Administration, Cutaneous , Adolescent , Humans , Male , Neurofibroma/etiology , Skin Neoplasms/etiology
9.
Neurology ; 91(2 Suppl 1): S21-S30, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29987132

ABSTRACT

OBJECTIVE: The only therapies currently available for cutaneous neurofibromas (cNF) are procedural. The goals of the Therapies Development Working Group were to (1) summarize currently available treatment options for cNF, (2) define key considerations for drug discovery and development generally, and specifically for cNF, and (3) outline recommendations for the successful development of medical therapies for cNF. METHODS: The subgroup reviewed published and unpublished data on procedural, drug/device, and medical treatment approaches utilized for cNFs via literature search. The team defined disease- and patient-specific factors to consider for therapies development in a series of consensus meetings. RESULTS: The team identified 5 approaches entailing procedural and drug/device methods currently under study. There have been 4 clinical studies exploring various interventional therapies, from which outcomes were highly variable. The team identified 4 key factors to prioritize during the development of products for the treatment for cNF: safety, anatomic distribution of cNF, numbers of tumors to be treated, and route of administration. CONCLUSIONS: The number, size, and distribution of cNF is highly variable among patients with NF1 and it is possible that different phenotypes will require different drug development paths. The nonfatal nature of the disease and relatively limited patient numbers suggest that for any product to have a higher likelihood of acceptance, it will have to (1) demonstrate an effect that is clinically meaningful, (2) have a safety profile conducive to long-term dosing, and (3) have a low manufacturing cost.


Subject(s)
Drug Development , Neurofibroma/drug therapy , Neurofibromatosis 1/complications , Skin Neoplasms/drug therapy , Drug Discovery , Humans , Neurofibroma/complications , Skin Neoplasms/complications
10.
Arch Med Res ; 48(6): 498-505, 2017 08.
Article in English | MEDLINE | ID: mdl-29198560

ABSTRACT

BACKGROUND AND AIMS: Neurofibromatosis type I (NF1) is one of the most common neurocutaneous syndromes characterized by development of adult neurofibromas which is mainly made up of Schwann cells. The disease is generally accepted to be caused by inactivation mutation of Nf1 gene. And Nf1 deficiency had been reported to lead to ROS overproduction and epithelial-mesenchymal transition (EMT) phenotype. This study was designed to investigate whether excessive ROS conferred to Nf1 deficiency-induced EMT in Schwann cells. METHODS: Colony formation, wound healing assay and transwell assay was used to evaluate the effects of stable Nf1 knockdown in SW10 Schwann cells. Western blot and ROS assay was conducted to explore the molecular mechanisms of Nf1 inactivation in tumorigenesis. Animal experiments were performed to assess the inhibitory effects of lipoamide, which is the neutral amide of α-lipoic acid and functions as a potent antioxidant to scavenge ROS, on Nf1-deficiency tumor growth in vivo. RESULTS: Nf1 knockdown enhanced the cellular capacities of proliferation, migration and invasion, promoted ROS generation, decreased the expression of epithelial surface marker E-cadherin, and up-regulated several EMT-associated molecules in Schwann cells. Moreover, lipoamide dose-dependently inhibited not only Nf1 deficiency-induced EMT but also spontaneous EMT. Furthermore, lipoamide markedly suppresses tumor growth in a mouse model of NF1-associated neurofibroma. CONCLUSIONS: Our results clearly reveal that ROS overproduction is responsible for Nf1 deficiency-induced EMT and plays a crucial role in NF1 tumor growth. The findings presented herein shed light on the potential of antioxidant therapy to prevent the progression of NF1-associated neurofibroma.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Genes, Neurofibromatosis 1 , Neurofibroma/drug therapy , Neurofibromatosis 1/drug therapy , Neurofibromin 1/deficiency , Reactive Oxygen Species/metabolism , Schwann Cells/drug effects , Thioctic Acid/analogs & derivatives , Animals , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Male , Mice , Mice, Nude , Thioctic Acid/pharmacology , Up-Regulation , Wound Healing/drug effects
11.
PLoS One ; 12(5): e0178316, 2017.
Article in English | MEDLINE | ID: mdl-28542306

ABSTRACT

Benign neurofibromas, the main phenotypic manifestations of the rare neurological disorder neurofibromatosis type 1, degenerate to malignant tumors associated to poor prognosis in about 10% of patients. Despite efforts in the field of (epi)genomics, the lack of prognostic biomarkers with which to predict disease evolution frustrates the adoption of appropriate early therapeutic measures. To identify potential biomarkers of malignant neurofibroma transformation, we integrated four human experimental studies and one for mouse, using a gene score-based meta-analysis method, from which we obtained a score-ranked signature of 579 genes. Genes with the highest absolute scores were classified as promising disease biomarkers. By grouping genes with similar neurofibromatosis-related profiles, we derived panels of potential biomarkers. The addition of promoter methylation data to gene profiles indicated a panel of genes probably silenced by hypermethylation. To identify possible therapeutic treatments, we used the gene signature to query drug expression databases. Trichostatin A and other histone deacetylase inhibitors, as well as cantharidin and tamoxifen, were retrieved as putative therapeutic means to reverse the aberrant regulation that drives to malignant cell proliferation and metastasis. This in silico prediction corroborated reported experimental results that suggested the inclusion of these compounds in clinical trials. This experimental validation supported the suitability of the meta-analysis method used to integrate several sources of public genomic information, and the reliability of the gene signature associated to the malignant evolution of neurofibromas to generate working hypotheses for prognostic and drug-responsive biomarkers or therapeutic measures, thus showing the potential of this in silico approach for biomarker discovery.


Subject(s)
Nerve Sheath Neoplasms/genetics , Neurofibroma/genetics , Animals , Biomarkers, Tumor/genetics , Cantharidin/pharmacology , Chromosome Mapping , Computer Simulation , CpG Islands , DNA Methylation , Drug Screening Assays, Antitumor , Gene Silencing , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/pathology , Neurofibroma/drug therapy , Neurofibroma/pathology , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Prognosis , Promoter Regions, Genetic , Tamoxifen/pharmacology , Transcriptome
12.
Sci Rep ; 7: 43315, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28256556

ABSTRACT

Neurofibromas are benign peripheral nerve tumors driven by NF1 loss in Schwann cells (SCs). Macrophages are abundant in neurofibromas, and macrophage targeted interventions may have therapeutic potential in these tumors. We generated gene expression data from fluorescence-activated cell sorted (FACS) SCs and macrophages from wild-type and mutant nerve and neurofibroma to identify candidate pathways involved in SC-macrophage cross-talk. While in 1-month-old Nf1 mutant nerve neither SCs nor macrophages significantly differed from their normal counterparts, both macrophages and SCs showed significantly altered cytokine gene expression in neurofibromas. Computationally reconstructed SC-macrophage molecular networks were enriched for inflammation-associated pathways. We verified that neurofibroma SC conditioned medium contains macrophage chemo-attractants including colony stimulation factor 1 (CSF1). Network analysis confirmed previously implicated pathways and predict novel paracrine and autocrine loops involving cytokines, chemokines, and growth factors. Network analysis also predicted a central role for decreased type-I interferon signaling. We validated type-I interferon expression in neurofibroma by protein profiling, and show that treatment of neurofibroma-bearing mice with polyethylene glycolyated (PEGylated) type-I interferon-α2b reduces the expression of many cytokines overexpressed in neurofibroma. These studies reveal numerous potential targetable interactions between Nf1 mutant SCs and macrophages for further analyses.


Subject(s)
Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Neurofibroma/genetics , Neurofibromin 1/genetics , Peripheral Nervous System Neoplasms/genetics , Schwann Cells/metabolism , Animals , Chemokines/genetics , Chemokines/metabolism , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Flow Cytometry , Gene Expression Profiling , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interferon alpha-2 , Interferon-alpha/pharmacology , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofibroma/drug therapy , Neurofibroma/metabolism , Neurofibroma/pathology , Neurofibromin 1/deficiency , Organ Specificity , Peripheral Nervous System/drug effects , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Peripheral Nervous System Neoplasms/drug therapy , Peripheral Nervous System Neoplasms/metabolism , Peripheral Nervous System Neoplasms/pathology , Polyethylene Glycols/pharmacology , Primary Cell Culture , Recombinant Proteins/pharmacology , Schwann Cells/pathology , Signal Transduction
13.
Acta Neuropathol ; 125(1): 159-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23099891

ABSTRACT

Neurofibromatosis type 1 (NF1) is a common genetic disease that predisposes 30-50 % of affected individuals to develop plexiform neurofibromas. We found that macrophage infiltration of both mouse and human neurofibromas correlates with disease progression. Macrophages accounted for almost half of neurofibroma cells, leading us to hypothesize that nerve macrophages are inflammatory effectors in neurofibroma development and/or growth. We tested the effects of PLX3397, a dual kit/fms kinase inhibitor that blocks macrophage infiltration, in the Dhh-Cre; Nf1(flox/flox) mouse model of GEM grade I neurofibroma. In mice aged 1-4 months, prior to development of nerve pathology and neurofibroma formation, PLX3397 did not impair tumor initiation and increased tumor volume compared to controls. However, in mice aged 7-9 months, after tumor establishment, a subset of mice demonstrating the largest reductions in macrophages after PLX3397 exhibited cell death and tumor volume regression. Macrophages are likely to provide an initial line of defense against developing tumors. Once tumors are established, they become tumor permissive. Macrophage depletion may result in impaired tumor maintenance and represent a therapeutic strategy for neurofibroma therapy.


Subject(s)
Enzyme Inhibitors/therapeutic use , Macrophages/cytology , Neurofibroma/drug therapy , Neurofibromin 1/metabolism , Age Factors , Animals , Disease Models, Animal , Humans , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neurofibroma/genetics , Neurofibroma/metabolism , Neurofibroma/pathology , Neurofibromin 1/genetics , Neurons/ultrastructure , Schwann Cells/metabolism , Schwann Cells/pathology , Tumor Burden
14.
J. bras. med ; 101(3): 45-49, 2013.
Article in Portuguese | LILACS | ID: lil-698227

ABSTRACT

Nos pacientes com neurofibromatose tipo 1 (NF1), o número de neurofibromas varia grandemente de pessoa para pessoa, mesmo em casos intrafamiliares, com a mesma mutação na linhagem germinativa no gene NF1. Além das mutações nesse gene, provavelmente fatores adicionais são importantes no desenvolvimento ou no crescimento dos neurofibromas. O objetivo deste artigo é revisar a literatura sobre o papel dos hormônios no desenvolvimento e crescimento dos neurofibromas e a segurança da terapia hormonal nos pacientes com NF1


In patients with neurofibromatosis type 1 (NF1), the number of neurofibromas varies greatly from person to person, even in intrafamilial cases, with the same germline mutation in the NF1 gene. In addition to the mutations in this gene, it is likely that additional factors are important in neurofibroma development or growth. The purpose of this article is to review the literature on the role of hormones in the development and growth of neurofibromas and the safety of hormonal therapy in patients with NF1


Subject(s)
Humans , Neurofibroma/drug therapy , Neurofibromatosis 1/therapy , Genes, Neurofibromatosis 1 , Gonadal Steroid Hormones , Growth Hormone , Neoplasms/chemically induced , Hormone Replacement Therapy/adverse effects , Hormone Replacement Therapy
15.
Head Neck Oncol ; 4: 30, 2012.
Article in English | MEDLINE | ID: mdl-22673101

ABSTRACT

Photodynamic therapy (PDT) - the fourth modality - has been successfully used in the management of early and advanced pathologies of the head and neck. We studied the effect of this modality on a giant solitary neurofibroma of the neck. A 70-year-old Caucasian female presented with left neck pain and disfigurement associated with slight shortness of breath and dysphagia. Examination revealed a large mass in the neck with no neurovascular compromise. Magnetic resonance imaging (MRI) reported a heterogeneously enhancing mass extending from the left angle of the mandible to the base of the neck. A core biopsy was performed and histopathological examination revealed a disorganised array of peripheral nerve fascicles. The patient elected to receive photodynamic therapy as the primary intervention. The multi-disciplinary meeting approved the treatment plan. The photosensitizing agent was mTHPC (0.15 mg/kg), which was systemically administered 96-hours prior to ultrasound (US)-guided light delivery to the mass, which was undertaken under general anaesthesia. Recovery was uneventful. Post-PDT follow-up showed that the patient's pain, dysphagia and shortness of breath issues had improved. The disfigurement of the neck caused by the mass was no longer a problem. Three months post-PDT, MRI revealed a significant reduction in the neurofibroma size. PDT was proven as a successful primary intervention for this pathology. However, higher evidence-based studies are required before this therapy can be proposed as a replacement to any of the other conventional therapies.


Subject(s)
Head and Neck Neoplasms/drug therapy , Neurofibroma/drug therapy , Photochemotherapy/methods , Aged , Female , Humans , Mesoporphyrins/administration & dosage , Photosensitizing Agents/administration & dosage , Treatment Outcome
17.
Blood ; 116(2): 157-64, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20233971

ABSTRACT

Neurofibromatosis type 1 (NF1) is the most common genetic disorder with a predisposition to malignancy and affects 1 in 3500 persons worldwide. NF1 is caused by a mutation in the NF1 tumor suppressor gene that encodes the protein neurofibromin. Patients with NF1 have cutaneous, diffuse, and plexiform neurofibromas, tumors comprised primarily of Schwann cells, blood vessels, fibroblasts, and mast cells. Studies from human and murine models that closely recapitulate human plexiform neurofibroma formation indicate that tumorigenesis necessitates NF1 loss of heterozygosity in the Schwann cell. In addition, our most recent studies with bone marrow transplantation and pharmacologic experiments implicate haploinsufficiency of Nf1 (Nf1(+/-)) and c-kit signaling in the hematopoietic system as required and sufficient for tumor progression. Here, we review recent studies implicating the hematopoietic system in plexiform neurofibroma genesis, delineate the physiology of stem cell factor-dependent hematopoietic cells and their contribution to the neurofibroma microenvironment, and highlight the application of this research toward the first successful, targeted medical treatment of a patient with a nonresectable and debilitating neurofibroma. Finally, we emphasize the importance of the tumor microenvironment hypothesis, asserting that tumorigenic cells in the neurofibroma do not arise and grow in isolation.


Subject(s)
Mast Cells/immunology , Neurofibroma/immunology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/immunology , Animals , Antineoplastic Agents/therapeutic use , Benzamides , Child, Preschool , Female , Humans , Imatinib Mesylate , Neurofibroma/drug therapy , Neurofibroma/genetics , Neurofibromatosis 1/drug therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use
19.
Lasers Med Sci ; 24(5): 769-75, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19377913

ABSTRACT

Photodynamic therapy is an emerging technology and results from the interactions between a photosensitiser, oxygen and light. The delivery of light may either be by surface illumination or by interstitial application. We describe the first clinical application of ultrasound-guided interstitial photodynamic therapy (US-iPDT). A total of 23 treatments with meta-tetra-hydroxyphenyl chlorine (mTHPC) and ultrasound-guided interstitial photodynamic therapy were performed on 21 patients with various conditions at the Head & Neck Centre, University College London Hospital. The needles could be clearly identified during insertion in all 23 treatments, and it was possible to guide parallel needle insertions using ultrasound. Although the resolution of ultrasound is not as high as that of other imaging modalities [i.e. computed tomography (CT) and magnetic resonance imaging (MRI)] it was satisfactory in identifying the centre and the peripheries of the pathological lesions. Ultrasound is very easy to perform, non-invasive, relatively inexpensive, quick, convenient, non-ionising, suited to the imaging of soft tissues and does not cause any discomfort. Ultrasound can be used to guide 'real-time' photodynamic therapy in deep-seated tumours and other malformations and can augment the information from other imaging modalities without affecting the patient's treatment outcome.


Subject(s)
Head and Neck Neoplasms/drug therapy , Photochemotherapy/methods , Vascular Malformations/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Arteriovenous Malformations/diagnostic imaging , Arteriovenous Malformations/drug therapy , Child , Female , Head and Neck Neoplasms/diagnostic imaging , Hemangioma/diagnostic imaging , Hemangioma/drug therapy , Humans , Lymphangioma/diagnostic imaging , Lymphangioma/drug therapy , Male , Middle Aged , Neurofibroma/diagnostic imaging , Neurofibroma/drug therapy , Photochemotherapy/instrumentation , Ultrasonography , Vascular Malformations/diagnostic imaging , Young Adult
20.
Cell ; 135(3): 408-10, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18984150

ABSTRACT

The tumor microenvironment is known to play an important role in tumorigenesis. In this issue, Yang et al. (2008) demonstrate that mast cells heterozygous for the Nf1 gene promote the growth of neurofibromas in a mouse model of neurofibromatosis and that genetic and pharmacological inhibition of these cells is sufficient to block tumor growth.


Subject(s)
Mast Cells/metabolism , Neurofibroma/metabolism , Animals , Humans , Mice , Neurofibroma/drug therapy , Neurofibroma/genetics , Neurofibroma/pathology , Neurofibromin 1/genetics , Neurofibromin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...