Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
1.
Oncogene ; 43(19): 1411-1430, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38480916

RÉSUMÉ

Malignant peripheral nerve sheath tumors (MPNSTs) are chemotherapy resistant sarcomas that are a leading cause of death in neurofibromatosis type 1 (NF1). Although NF1-related MPNSTs derive from neural crest cell origin, they also exhibit intratumoral heterogeneity. TP53 mutations are associated with significantly decreased survival in MPNSTs, however the mechanisms underlying TP53-mediated therapy responses are unclear in the context of NF1-deficiency. We evaluated the role of two commonly altered genes, MET and TP53, in kinome reprograming and cellular differentiation in preclinical MPNST mouse models. We previously showed that MET amplification occurs early in human MPNST progression and that Trp53 loss abrogated MET-addiction resulting in MET inhibitor resistance. Here we demonstrate a novel mechanism of therapy resistance whereby p53 alters MET stability, localization, and downstream signaling leading to kinome reprogramming and lineage plasticity. Trp53 loss also resulted in a shift from RAS/ERK to AKT signaling and enhanced sensitivity to MEK and mTOR inhibition. In response to MET, MEK and mTOR inhibition, we observed broad and heterogeneous activation of key differentiation genes in Trp53-deficient lines suggesting Trp53 loss also impacts lineage plasticity in MPNSTs. These results demonstrate the mechanisms by which p53 loss alters MET dependency and therapy resistance in MPNSTS through kinome reprogramming and phenotypic flexibility.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Neurofibromatose de type 1 , Inhibiteurs de protéines kinases , Protéine p53 suppresseur de tumeur , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Souris , Humains , Résistance aux médicaments antinéoplasiques/génétique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Neurofibromatose de type 1/génétique , Neurofibromatose de type 1/anatomopathologie , Neurofibromine-1/génétique , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme , Tumeurs des gaines nerveuses/génétique , Tumeurs des gaines nerveuses/anatomopathologie , Tumeurs des gaines nerveuses/traitement médicamenteux , Lignée cellulaire tumorale , Transduction du signal , Lignage cellulaire/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Neurofibrosarcome/génétique , Neurofibrosarcome/anatomopathologie , Neurofibrosarcome/traitement médicamenteux , Plasticité cellulaire/effets des médicaments et des substances chimiques , Plasticité cellulaire/génétique
2.
Clin Cancer Res ; 29(17): 3484-3497, 2023 09 01.
Article de Anglais | MEDLINE | ID: mdl-37410426

RÉSUMÉ

PURPOSE: Malignant peripheral nerve sheath tumors (MPNST) are lethal, Ras-driven sarcomas that lack effective therapies. We investigated effects of targeting cyclin-dependent kinases 4 and 6 (CDK4/6), MEK, and/or programmed death-ligand 1 (PD-L1) in preclinical MPNST models. EXPERIMENTAL DESIGN: Patient-matched MPNSTs and precursor lesions were examined by FISH, RNA sequencing, IHC, and Connectivity-Map analyses. Antitumor activity of CDK4/6 and MEK inhibitors was measured in MPNST cell lines, patient-derived xenografts (PDX), and de novo mouse MPNSTs, with the latter used to determine anti-PD-L1 response. RESULTS: Patient tumor analyses identified CDK4/6 and MEK as actionable targets for MPNST therapy. Low-dose combinations of CDK4/6 and MEK inhibitors synergistically reactivated the retinoblastoma (RB1) tumor suppressor, induced cell death, and decreased clonogenic survival of MPNST cells. In immune-deficient mice, dual CDK4/6-MEK inhibition slowed tumor growth in 4 of 5 MPNST PDXs. In immunocompetent mice, combination therapy of de novo MPNSTs caused tumor regression, delayed resistant tumor outgrowth, and improved survival relative to monotherapies. Drug-sensitive tumors that regressed contained plasma cells and increased cytotoxic T cells, whereas drug-resistant tumors adopted an immunosuppressive microenvironment with elevated MHC II-low macrophages and increased tumor cell PD-L1 expression. Excitingly, CDK4/6-MEK inhibition sensitized MPNSTs to anti-PD-L1 immune checkpoint blockade (ICB) with some mice showing complete tumor regression. CONCLUSIONS: CDK4/6-MEK inhibition induces a novel plasma cell-associated immune response and extended antitumor activity in MPNSTs, which dramatically enhances anti-PD-L1 therapy. These preclinical findings provide strong rationale for clinical translation of CDK4/6-MEK-ICB targeted therapies in MPNST as they may yield sustained antitumor responses and improved patient outcomes.


Sujet(s)
Neurofibrosarcome , Souris , Humains , Animaux , Neurofibrosarcome/traitement médicamenteux , Plasmocytes/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Mitogen-Activated Protein Kinase Kinases , Lignée cellulaire tumorale , Microenvironnement tumoral , Kinase-4 cycline-dépendante
3.
Oncologist ; 28(5): 453-459, 2023 05 08.
Article de Anglais | MEDLINE | ID: mdl-36724001

RÉSUMÉ

BACKGROUND: Evaluation of prior phase II trials for malignant peripheral nerve sheath tumors (MPNST) may help develop more suitable trial endpoints in future studies. METHODS: We analyzed outcomes of patients with recurrent or unresectable/metastatic MPNST enrolled on prior Sarcoma Alliance for Research through Collaboration (SARC) phase II trials and estimated the progression-free survival (PFS). PFS from SARC006 (NCT00304083), the phase II trial of upfront chemotherapy in chemotherapy naïve patients, was analyzed separately. Impact of baseline enrollment characteristics on PFS was evaluated. RESULTS: Sixty-four patients (29 male, 35 female, median age 39 years (range 15-81)) with MPNST were enrolled on 1 of 5 trials of single agent or combination therapy that were determined to be inactive. Patients had received a median of 1 (range 0-5) prior systemic therapy, and most had undergone prior surgery (77%) and radiation (61%). Seventy-three percent had metastatic disease at enrollment. Median PFS was 1.77 months (95% CI, 1.61-3.45), and the PFS rate at 4 months was 15%. Greater number of prior systemic therapies and worse performance status were associated with inferior PFS. There was no significant difference in PFS based on age at enrollment, treatment trial, response criteria, presence of metastatic disease, disease site at enrollment, and prior surgery or radiation. In comparison, on the SARC006 trial the PFS rate at 4 months was 94% in 40 patients. CONCLUSION: These data provide a historical baseline PFS that may be used as a comparator in future clinical trials for patients with MPNST.


Sujet(s)
Neurofibrosarcome , Sarcomes , Tumeurs des tissus mous , Humains , Mâle , Femelle , Adolescent , Jeune adulte , Adulte , Adulte d'âge moyen , Sujet âgé , Sujet âgé de 80 ans ou plus , Neurofibrosarcome/traitement médicamenteux , Sarcomes/traitement médicamenteux , Survie sans progression , Tumeurs des tissus mous/traitement médicamenteux , Association thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
4.
Genes Chromosomes Cancer ; 62(1): 47-51, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-35906852

RÉSUMÉ

Neurotropic tropomyosin receptor kinase (NTRK) gene rearrangements have been reported in limited cases of sarcomas; however, to date, there has been only one report of such rearrangements in malignant peripheral nerve sheath tumors (MPNSTs). Herein, we describe a 51-year-old male patient with a buttock tumor arising from the sciatic nerve, which was diagnosed as MPNST with positive S-100 staining, negative SOX10 staining, and loss of trimethylation at lysine 27 of histone H3 (H3K27me3) confirmed by immunohistochemistry. Soon after the resection of the primary tumor, the patient was found to have pulmonary and lymph node metastases. Chemotherapy with eribulin and trabectedin showed limited effects. However, the patient responded rapidly to pazopanib, but severe side effects caused discontinuation of the treatment. RNA panel testing revealed a novel fusion gene between Small Nuclear Ribonucleoprotein U1 Subunit 70 (SNRNP70) gene and NTRK3 gene. Furthermore, loss of NF1, SUZ12, and CDKN2A genes was confirmed by DNA panel testing, which is compatible with a histological diagnosis of MPNST. SNRNP70 possesses a coiled-coiled domain and seems to induce constitutive activation of NTRK3 through dimerization. In fact, immunohistochemistry revealed diffuse staining of pan-TRK within tumor cells. Treatment with entrectinib, which is an NTRK inhibitor, showed a quick and durable response for 10 months. Although NTRK rearrangements are very rare in MPNST, this case highlights the importance of genetic testing in MPNST, especially using an RNA panel for the detection of rare fusion genes.


Sujet(s)
Neurofibrosarcome , Mâle , Humains , Adulte d'âge moyen , Neurofibrosarcome/traitement médicamenteux , Neurofibrosarcome/génétique , Marqueurs biologiques tumoraux/génétique , Immunohistochimie , ARN , Petites ribonucléoprotéines nucléaires U1
5.
Clin Cancer Res ; 28(15): 3185-3195, 2022 08 02.
Article de Anglais | MEDLINE | ID: mdl-35446392

RÉSUMÉ

Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft-tissue sarcomas that represent an important clinical challenge, particularly given their strong tendency to relapse and metastasize and their relatively poor response to conventional therapies. To date, targeted, noncytotoxic treatments have demonstrated limited clinical success with MPNSTs, highlighting the need to explore other key pathways to find novel, improved therapeutic approaches. Here, we review evidence supporting the crucial role of the RAS/MEK/ERK pathway and angiogenesis in MPNST pathogenesis, and we focus on the potential of therapies targeting these pathways to treat this disease. We also present works suggesting that the combination of MEK inhibitors and antiangiogenic agents could represent a promising therapeutic strategy to manage MPNSTs. In support of this notion, we discuss the preclinical rational and clinical benefits of this combination therapy in other solid tumor types. Finally, we describe other emerging therapeutic approaches that could improve patient outcomes in MPNSTs, such as immune-based therapies.


Sujet(s)
Tumeurs des gaines nerveuses , Neurofibrosarcome , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Humains , Mitogen-Activated Protein Kinase Kinases , Récidive tumorale locale/traitement médicamenteux , Tumeurs des gaines nerveuses/traitement médicamenteux , Neurofibrosarcome/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique
6.
Ann Afr Med ; 20(3): 228-231, 2021.
Article de Anglais | MEDLINE | ID: mdl-34558453

RÉSUMÉ

Malignant peripheral nerve sheath tumor (MPNST) contains properties and histologic markers of both neural crest cells and mesenchymal cells. It is a rare diagnosis, with an incidence of 1:100,000/year or 4%-10% of soft-tissue sarcomas. There are very few cases reported and studied. Therefore, establishing a proper diagnosis and treatment of MPNST provides a challenge. We present this unique and rare case of metastatic MPNST of the small and large bowel with bone, pulmonary, liver, and splenic metastases. The patient subsequently developed hemorrhagic brain metastases and died 6 months after THE initial diagnosis.


Résumé La tumeur maligne de la gaine nerveuse périphérique (MPNST) contient des propriétés et des marqueurs histologiques des cellules de la crête neurale et du mésenchyme cellules. Il s'agit d'un diagnostic rare, avec une incidence de 1: 100 000 / an ou 4 à 10% des sarcomes des tissus mous. Il y a très peu de cas signalés et étudié. Par conséquent, l'établissement d'un diagnostic et d'un traitement appropriés de la MPNST constitue un défi. Nous présentons ce cas unique et rare de MPNST métastatique du petit et du gros intestin avec métastases osseuses, pulmonaires, hépatiques et spléniques. Le patient a ensuite développé métastases cérébrales hémorragiques et est décédé 6 mois après LE diagnostic initial.


Sujet(s)
Tumeurs de l'intestin/secondaire , Gros intestin/anatomopathologie , Intestin grêle/anatomopathologie , Tumeurs des gaines nerveuses/secondaire , Neurofibrosarcome/anatomopathologie , Tumeurs des tissus mous/anatomopathologie , Sujet âgé , Antibiotiques antinéoplasiques/usage thérapeutique , Tumeurs osseuses/secondaire , Tumeurs osseuses/chirurgie , Traitement médicamenteux adjuvant , Doxorubicine/usage thérapeutique , Humains , Tumeurs de l'intestin/traitement médicamenteux , Tumeurs de l'intestin/anatomopathologie , Mâle , Métastase tumorale , Seconde tumeur primitive , Tumeurs des gaines nerveuses/chirurgie , Neurofibrosarcome/traitement médicamenteux , Neurofibrosarcome/chirurgie , Tumeurs des tissus mous/traitement médicamenteux , Tumeurs des tissus mous/chirurgie , Résultat thérapeutique
7.
Int J Cancer ; 148(1): 140-149, 2021 01 01.
Article de Anglais | MEDLINE | ID: mdl-32638374

RÉSUMÉ

Malignant peripheral nerve sheath tumor (MPNST) often does not respond well to chemotherapy and develops against a background of NF1. The purpose of our study was to examine the efficacy of pazopanib against MPNST. Our study was designed as a physician-initiated phase II clinical trial in patients with advanced MPNST. Patients were registered from 11 large hospitals. The primary endpoint was set to clarify the clinical benefit rate (CBR) at 12 weeks according to response evaluation criteria in solid tumors (RECIST). Progression-free survival (PFS), overall survival (OS) and the CBR based on modified Choi evaluation at week 12 were set as secondary endpoints along with treatment-related safety. The study enrolled 12 patients. Median age was 49 years. Seven had Grade 2 and five Grade 3 according to the FNCLCC evaluation. Median follow-up period was 10.6 months. CBR at 12 weeks was both 50.0% (RECIST and Choi). The median PFS was 5.4 months for both RECIST and Choi, and the median OS was 10.6 months. Of special interest, the median PFS was 2.9 months for patients with FNCLCC Grade 2 and 10.2 months for Grade 3 (both RECIST and Choi). Grade 4 adverse events of neutropenia and lipase elevation were noted in one patient each. The results of this pazopanib therapy were generally better than those of any of the other single molecular targeted therapies reported previously. Although accumulation of more cases remains necessary, we conclude pazopanib treatment for MPNST to be a safe and promising treatment after doxorubicin-based chemotherapy.


Sujet(s)
Indazoles/administration et posologie , Neurofibrosarcome/traitement médicamenteux , Neutropénie/diagnostic , Inhibiteurs de protéines kinases/administration et posologie , Pyrimidines/administration et posologie , Sulfonamides/administration et posologie , Adulte , Sujet âgé , Femelle , Études de suivi , Humains , Indazoles/effets indésirables , Mâle , Adulte d'âge moyen , Grading des tumeurs , Neurofibrosarcome/diagnostic , Neurofibrosarcome/mortalité , Neutropénie/induit chimiquement , Survie sans progression , Inhibiteurs de protéines kinases/effets indésirables , Pyrimidines/effets indésirables , Évaluation de la réponse des tumeurs solides aux traitements , Indice de gravité de la maladie , Sulfonamides/effets indésirables , Jeune adulte
8.
BMC Cancer ; 20(1): 896, 2020 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-32948135

RÉSUMÉ

BACKGROUND: Anticancer compound 3-bromopyruvate (3-BrPA) suppresses cancer cell growth via targeting glycolytic and mitochondrial metabolism. The malignant peripheral nerve sheath tumor (MPNST), a very aggressive, therapy resistant, and Neurofibromatosis type 1 associated neoplasia, shows a high metabolic activity and affected patients may therefore benefit from 3-BrPA treatment. To elucidate the specific mode of action, we used a controlled cell model overexpressing proteasome activator (PA) 28, subsequently leading to p53 inactivation and oncogenic transformation and therefore reproducing an important pathway in MPNST and overall tumor pathogenesis. METHODS: Viability of MPNST cell lines S462, NSF1, and T265 in response to increasing doses (0-120 µM) of 3-BrPA was analyzed by CellTiter-Blue® assay. Additionally, we investigated viability, reactive oxygen species (ROS) production (dihydroethidium assay), nicotinamide adenine dinucleotide dehydrogenase activity (NADH-TR assay) and lactate production (lactate assay) in mouse B8 fibroblasts overexpressing PA28 in response to 3-BrPA application. For all experiments normal and nutrient deficient conditions were tested. MPNST cell lines were furthermore characterized immunohistochemically for Ki67, p53, bcl2, bcl6, cyclin D1, and p21. RESULTS: MPNST significantly responded dose dependent to 3-BrPA application, whereby S462 cells were most responsive. Human control cells showed a reduced sensitivity. In PA28 overexpressing cancer cell model 3-BrPA application harmed mitochondrial NADH dehydrogenase activity mildly and significantly failed to inhibit lactate production. PA28 overexpression was associated with a functional glycolysis as well as a partial resistance to stress provoked by nutrient deprivation. 3-BrPA treatment was not associated with an increase of ROS. Starvation sensitized MPNST to treatment. CONCLUSIONS: Aggressive MPNST cells are sensitive to 3-BrPA therapy in-vitro with and without starvation. In a PA28 overexpression cancer cell model leading to p53 inactivation, thereby reflecting a key molecular feature in human NF1 associated MPNST, known functions of 3-BrPA to block mitochondrial activity and glycolysis were reproduced, however oncogenic cells displayed a partial resistance. To conclude, 3-BrPA was sufficient to reduce NF1 associated MPNST viability potentially due inhibition of glycolysis which should lead to the initiation of further studies and promises a potential benefit for NF1 patients.


Sujet(s)
Antienzymes/usage thérapeutique , Neurofibrosarcome/traitement médicamenteux , Pyruvates/usage thérapeutique , Prolifération cellulaire , Antienzymes/pharmacologie , Humains , Voies et réseaux métaboliques , Pyruvates/pharmacologie
9.
Cancer ; 126(24): 5303-5310, 2020 12 15.
Article de Anglais | MEDLINE | ID: mdl-32914879

RÉSUMÉ

BACKGROUND: Lorvotuzumab mertansine (IMGN901) is an antibody-drug conjugate linking an antimitotic agent (DM1) to an anti-CD56 antibody (lorvotuzumab). Preclinical efficacy has been noted in Wilms tumor, rhabdomyosarcoma, and neuroblastoma. Synovial sarcoma, malignant peripheral nerve sheath tumor (MPNST), and pleuropulmonary blastoma also express CD56. A phase 2 trial of lorvotuzumab mertansine was conducted to assess its efficacy, recommended phase 2 dose, and toxicities. METHODS: Eligible patients had relapsed after or progressed on standard therapy for their tumor type. Lorvotuzumab mertansine (110 mg/m2 per dose) was administered at the adult recommended phase 2 dose intravenously on days 1 and 8 of 21-day cycles. Dexamethasone premedication was used. Pharmacokinetic samples, peripheral blood CD56-positive cell counts, and tumor CD56 expression were assessed. RESULTS: Sixty-two patients enrolled. The median age was 14.3 years (range, 2.8-29.9 years); 35 were male. Diagnoses included Wilms tumor (n = 17), rhabdomyosarcoma (n = 17), neuroblastoma (n = 12), synovial sarcoma (n = 10), MPNST (n = 5), and pleuropulmonary blastoma (n = 1). Five patients experienced 9 dose-limiting toxicities: hyperglycemia (n = 1), colonic fistula (n = 1) with perforation (n = 1), nausea (n = 1) with vomiting (n = 1), increased alanine aminotransferase in cycle 1 (n = 2), and increased alanine aminotransferase in cycle 2 (n = 1) with increased aspartate aminotransferase (n = 1). Non-dose-limiting toxicities (grade 3 or higher) attributed to lorvotuzumab mertansine were rare. The median values of the maximum concentration, half-life, and area under the curve from zero to infinity for DM1 were 0.87 µg/mL, 35 hours, and 27.9 µg/mL h, respectively. Peripheral blood CD56+ leukocytes decreased by 71.9% on day 8. One patient with rhabdomyosarcoma had a partial response, and 1 patient with synovial sarcoma achieved a delayed complete response. CONCLUSIONS: Lorvotuzumab mertansine (110 mg/m2 ) is tolerated in children at the adult recommended phase 2 dose; clinical activity is limited.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Maitansine/analogues et dérivés , Neuroblastome/traitement médicamenteux , Neurofibrosarcome/traitement médicamenteux , Blastome pulmonaire/traitement médicamenteux , Rhabdomyosarcome/traitement médicamenteux , Sarcome synovial/traitement médicamenteux , Tumeur de Wilms/traitement médicamenteux , Adolescent , Adulte , Anticorps monoclonaux/effets indésirables , Aire sous la courbe , Antigènes CD56/métabolisme , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Mâle , Dose maximale tolérée , Maitansine/administration et posologie , Maitansine/effets indésirables , Neuroblastome/métabolisme , Neurofibrosarcome/métabolisme , Blastome pulmonaire/métabolisme , Rhabdomyosarcome/métabolisme , Sarcome synovial/métabolisme , Analyse de survie , Résultat thérapeutique , Tumeur de Wilms/métabolisme , Jeune adulte
10.
Pediatr Blood Cancer ; 67(12): e28639, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32975370

RÉSUMÉ

BACKGROUND: Malignant peripheral nerve sheath tumor (MPNST) is an aggressive form of soft-tissue sarcoma (STS) in children. Despite intensive therapy, relatively few children with metastatic and unresectable disease survive beyond three years. RAS pathway activation is common in MPNST, suggesting MEK pathway inhibition as a targeted therapy, but the impact on clinical outcome has been small to date. PROCEDURE: We conducted preclinical pharmacokinetic (PK) and pharmacodynamic studies of two MEK inhibitors, trametinib and selumetinib, in two MPNST models and analyzed tumors for intratumor drug levels. We then investigated 3'-deoxy-3'-[18 F]fluorothymidine (18 F-FLT) PET imaging followed by 18 F-FDG PET/CT imaging of MPNST xenografts coupled to short-term or longer-term treatment with selumetinib focusing on PET-based imaging as a biomarker of MEK inhibition. RESULTS: Trametinib decreased pERK expression in MPNST xenografts but did not prolong survival or decrease Ki67 expression. In contrast, selumetinib prolonged survival of animals bearing MPNST xenografts, and this correlated with decreased pERK and Ki67 staining. PK studies revealed a significantly higher fraction of unbound selumetinib within a responsive MPNST xenograft model. Thymidine uptake, assessed by 18 F-FLT PET/CT, positively correlated with Ki67 expression in different xenograft models and in response to selumetinib. CONCLUSION: The ability of MEK inhibitors to control MPNST growth cannot simply be predicted by serum drug levels or drug-induced changes in pERK expression. Tumor cell proliferation assessed by 18 F-FLT PET imaging might be useful as an early response marker to targeted therapies, including MEK inhibition, where a primary effect is cell-cycle arrest.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Neurofibrosarcome/anatomopathologie , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Protéines G ras/antagonistes et inhibiteurs , Animaux , Apoptose , Benzimidazoles/administration et posologie , Prolifération cellulaire , Fluorodésoxyglucose F18/pharmacocinétique , Humains , Souris , Souris de lignée NOD , Souris SCID , Neurofibrosarcome/imagerie diagnostique , Neurofibrosarcome/traitement médicamenteux , Neurofibrosarcome/métabolisme , Pyridones/administration et posologie , Pyrimidinones/administration et posologie , Radiopharmaceutiques/pharmacocinétique , Distribution tissulaire , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Dis Model Mech ; 13(8)2020 08 27.
Article de Anglais | MEDLINE | ID: mdl-32651197

RÉSUMÉ

Polycomb repressive complex 2 (PRC2) is an epigenetic regulator of gene expression that possesses histone methyltransferase activity. PRC2 trimethylates lysine 27 of histone H3 proteins (H3K27me3) as a chromatin modification associated with repressed transcription of genes frequently involved in cell proliferation or self-renewal. Loss-of-function mutations in the PRC2 core subunit SUZ12 have been identified in a variety of tumors, including malignant peripheral nerve sheath tumors (MPNSTs). To determine the consequences of SUZ12 loss in the pathogenesis of MPNST and other cancers, we used CRISPR-Cas9 to disrupt the open reading frame of each of two orthologous suz12 genes in zebrafish: suz12a and suz12b We generated these knockout alleles in the germline of our previously described p53 (also known as tp53)- and nf1-deficient zebrafish model of MPNSTs. Loss of suz12 significantly accelerated the onset and increased the penetrance of MPNSTs compared to that in control zebrafish. Moreover, in suz12-deficient zebrafish, we detected additional types of tumors besides MPNSTs, including leukemia with histological characteristics of lymphoid malignancies, soft tissue sarcoma and pancreatic adenocarcinoma, which were not detected in p53/nf1-deficient control fish, and are also contained in the human spectrum of SUZ12-deficient malignancies identified in the AACR Genie database. The suz12-knockout tumors displayed reduced or abolished H3K27me3 epigenetic marks and upregulation of gene sets reported to be targeted by PRC2. Thus, these zebrafish lines with inactivation of suz12 in combination with loss of p53/nf1 provide a model of human MPNSTs and multiple other tumor types, which will be useful for mechanistic studies of molecular pathogenesis and targeted therapy with small molecule inhibitors.


Sujet(s)
Transformation cellulaire néoplasique/génétique , Extinction de l'expression des gènes , Neurofibromine-1/génétique , Neurofibrosarcome/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéines de poisson-zèbre/génétique , Danio zébré/génétique , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Animaux , Animal génétiquement modifié , Antinéoplasiques/pharmacologie , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie , Méthylation de l'ADN , Modèles animaux de maladie humaine , Épigenèse génétique , Régulation de l'expression des gènes tumoraux , Leucémies/génétique , Leucémies/métabolisme , Leucémies/anatomopathologie , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , MAP Kinase Kinase Kinases/métabolisme , Neurofibromine-1/déficit , Neurofibrosarcome/traitement médicamenteux , Neurofibrosarcome/métabolisme , Neurofibrosarcome/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Sarcomes/génétique , Sarcomes/métabolisme , Sarcomes/anatomopathologie , Transduction du signal , Tumeurs des tissus mous/génétique , Tumeurs des tissus mous/métabolisme , Tumeurs des tissus mous/anatomopathologie , Protéine p53 suppresseur de tumeur/déficit , Danio zébré/métabolisme , Protéines de poisson-zèbre/déficit
12.
Medicine (Baltimore) ; 99(26): e20725, 2020 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-32590748

RÉSUMÉ

RATIONALE: Malignant peripheral nerve sheath tumor (MPNST) is a rare sarcoma. Owing to the lack of specific histological criteria, immunohistochemical, and molecular diagnostic markers, several differential diagnoses must be considered. Advances in molecular testing can provide significant insights for management of rare tumor. PATIENT CONCERNS: The patient was a 50-year-old man with a history of lumpectomy on the right back 30 years ago. He felt a stabbing pain at the right iliac fossa and went to the local hospital. DIAGNOSIS: By immunohistochemistry, the tumor cells stained positively for S-100 (focal +), CD34 (strong +++) and Ki-67 (20%), and negatively for smooth muscle actin, pan-cytokeratin, neurofilament, pan-cytokeratin-L, GFAP, CD31, STAT6, ERG, myogenin, and MyoD1. Combined with the histopathology and immunohistochemistry results, our initial diagnosis was solitary fibrous tumor (SFT) or MPNST. The tissue biopsy was sent for next-generation sequencing. neurofibromatosis type 1 Q1395Hfs*22 somatic mutation, neurofibromatosis type 1 D483Tfs*15 germline mutation, and amplifications of BTK, MDM2, ATF1, BMPR1A, EBHA2, GNA13, PTPN11, RAD52, RPTOR, and SOX9, as well as TJP1-ROS1 fusion, CDKN2A-IL1RAPL2 fusion and CDKN2A/UBAP1 rearrangement were identified. Given that NAB2-STAT6 fusion, a specific biomarker of SFT, was not identified in our patient's tumor, the SFT was excluded by through genetic testing results. Therefore, our finally diagnosis was a MPNST by 2 or more pathologists. INTERVENTIONS AND OUTCOMES: Subsequently, the patient received crizotinib therapy for 2 months and showed stable disease. However, after crizotinib continued treatment for 4 months, the patient's disease progressed. Soon after, the patient stopped crizotinib treatment and died in home. LESSONS: To our knowledge, this is the first report of the TJP1-ROS1 fusion, which expands the list of gene fusions and highlights new targets for targeted therapy. Also, our case underlines the value of multi-gene panel next-generation sequencing for diagnosis of MPNST.


Sujet(s)
Crizotinib/administration et posologie , Fusion de gènes/génétique , Séquençage nucléotidique à haut débit/méthodes , Neurofibrosarcome , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes/génétique , Protéine-1 de la zonula occludens/génétique , Antinéoplasiques/administration et posologie , Diagnostic différentiel , Évolution de la maladie , Surveillance des médicaments , Humains , Immunohistochimie , Mâle , Adulte d'âge moyen , Tumeurs du tissu fibreux/diagnostic , Neurofibrosarcome/traitement médicamenteux , Neurofibrosarcome/génétique , Neurofibrosarcome/anatomopathologie , Résultat thérapeutique
13.
Int J Mol Sci ; 21(4)2020 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-32102484

RÉSUMÉ

Persistent signalling via the PI3K/AKT/mTOR pathway is a major driver of malignancy in NF1-associated malignant peripheral nerve sheath tumours (MPNST). Nevertheless, single targeting of this pathway is not sufficient to inhibit MPNST growth. In this report, we demonstrate that combined treatment with the allosteric pan-AKT inhibitor MK-2206 and the mTORC1/mTORC2 inhibitor AZD8055 has synergistic effects on the viability of MPNST cell lines in comparison to the treatment with each compound alone. However, when treating animals bearing experimental MPNST with the combined AKT/mTOR regime, no influence on tumour growth was observed. Further analysis of the MPNST xenograft tumours resistant to AKT/mTOR treatment revealed a reactivation of both AKT and mTOR in several tumour samples. Additional targeting of the RAS/RAF/MEK/MAPK pathway with the allosteric MEK1/2 inhibitor AZD6244 showed synergistic effects on the viability of MPNST cell lines in vitro in comparison to the dual AKT/mTOR inhibition. In summary, these data indicate that combined treatment with AKT and mTOR inhibitors is effective on MPNST cells in vitro but tumour resistance can occur rapidly in vivo by restoration of AKT/mTOR signalling. Our data further suggest that a triple treatment with inhibitors against AKT, mTORC1/2 and MEK1/2 may be a promising treatment option that should be further analysed in an experimental MPNST mouse model in vivo.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Neurofibrosarcome/traitement médicamenteux , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Animaux , Benzimidazoles/administration et posologie , Lignée cellulaire tumorale , Synergie des médicaments , Composés hétérocycliques 3 noyaux/administration et posologie , Humains , Souris SCID , Morpholines/administration et posologie , Neurofibromatose de type 1/complications , Neurofibromatose de type 1/métabolisme , Neurofibrosarcome/complications , Neurofibrosarcome/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme
14.
Clin Cancer Res ; 26(12): 2997-3011, 2020 06 15.
Article de Anglais | MEDLINE | ID: mdl-32086342

RÉSUMÉ

PURPOSE: Malignant peripheral nerve sheath tumors (MPNST) are deadly sarcomas that lack effective therapies. In most MPNSTs, the retinoblastoma (RB1) tumor suppressor is disabled by hyperactivation of cyclin-dependent kinases (CDK), commonly through loss of CDK-inhibitory proteins such as p27(Kip1). RABL6A is an inhibitor of RB1 whose role in MPNSTs is unknown. To gain insight into MPNST development and establish new treatment options, we investigated RABL6A-RB1 signaling and CDK inhibitor-based therapy in MPNSTs. EXPERIMENTAL DESIGN: We examined patient-matched MPNSTs and precursor lesions by RNA sequencing (RNA-Seq) and IHC. Molecular and biological effects of silencing RABL6A and/or p27 in MPNST lines and normal human Schwann cells were determined. Tumor-suppressive effects of CDK inhibitors were measured in MPNST cells and orthotopic tumors. RESULTS: RABL6A was dramatically upregulated in human MPNSTs compared with precursor lesions, which correlated inversely with p27 levels. Silencing RABL6A caused MPNST cell death and G1 arrest that coincided with p27 upregulation, CDK downregulation, and RB1 activation. The growth-suppressive effects of RABL6A loss, and its regulation of RB1, were largely rescued by p27 depletion. Importantly, reactivation of RB1 using a CDK4/6 inhibitor (palbociclib) killed MPNST cells in vitro in an RABL6A-dependent manner and suppressed MPNST growth in vivo. Low-dose combination of drugs targeting multiple RB1 kinases (CDK4/6, CDK2) had enhanced antitumorigenic activity associated with potential MPNST cell redifferentiation. CONCLUSIONS: RABL6A is a new driver of MPNST pathogenesis that acts in part through p27-RB1 inactivation. Our results suggest RB1 targeted therapy with multiple pathway drugs may effectively treat MPNSTs.


Sujet(s)
Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques , Neurofibrosarcome/traitement médicamenteux , Protéines oncogènes/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Protéines de liaison à la protéine du rétinoblastome/métabolisme , Ubiquitin-protein ligases/métabolisme , Protéines G rab/métabolisme , Animaux , Apoptose , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Prolifération cellulaire , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Souris , Souris de lignée NOD , Souris SCID , Neurofibrosarcome/génétique , Neurofibrosarcome/métabolisme , Neurofibrosarcome/anatomopathologie , Protéines oncogènes/génétique , Protéines de liaison à la protéine du rétinoblastome/génétique , Transduction du signal , Cellules cancéreuses en culture , Ubiquitin-protein ligases/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines G rab/génétique
15.
Mol Cancer Ther ; 19(3): 731-741, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31848295

RÉSUMÉ

Malignant peripheral nerve sheath tumors (MPNST) frequently overexpress eukaryotic initiation factor 4F components, and the eIF4A inhibitor silvestrol potently suppresses MPNST growth. However, silvestrol has suboptimal drug-like properties, including a bulky structure, poor oral bioavailability (<2%), sensitivity to MDR1 efflux, and pulmonary toxicity in dogs. We compared ten silvestrol-related rocaglates lacking the dioxanyl ring and found that didesmethylrocaglamide (DDR) and rocaglamide (Roc) had growth-inhibitory activity comparable with silvestrol. Structure-activity relationship analysis revealed that the dioxanyl ring present in silvestrol was dispensable for, but may enhance, cytotoxicity. Both DDR and Roc arrested MPNST cells at G2-M, increased the sub-G1 population, induced cleavage of caspases and PARP, and elevated the levels of the DNA-damage response marker γH2A.X, while decreasing the expression of AKT and ERK1/2, consistent with translation inhibition. Unlike silvestrol, DDR and Roc were not sensitive to MDR1 inhibition. Pharmacokinetic analysis confirmed that Roc had 50% oral bioavailability. Importantly, Roc, when administered intraperitoneally or orally, showed potent antitumor effects in an orthotopic MPNST mouse model and did not induce pulmonary toxicity in dogs as found with silvestrol. Treated tumors displayed degenerative changes and had more cleaved caspase-3-positive cells, indicative of increased apoptosis. Furthermore, Roc effectively suppressed the growth of osteosarcoma, Ewing sarcoma, and rhabdomyosarcoma cells and patient-derived xenografts. Both Roc- and DDR-treated sarcoma cells showed decreased levels of multiple oncogenic kinases, including insulin-like growth factor-1 receptor. The more favorable drug-like properties of DDR and Roc and the potent antitumor activity of Roc suggest that these rocaglamides could become viable treatments for MPNST and other sarcomas.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Benzofuranes/composition chimique , Benzofuranes/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Neurofibrosarcome/traitement médicamenteux , Maturation post-traductionnelle des protéines/effets des médicaments et des substances chimiques , Aglaia/composition chimique , Animaux , Apoptose , Caspase-3/métabolisme , Cycle cellulaire , Prolifération cellulaire , Humains , Souris , Neurofibrosarcome/métabolisme , Neurofibrosarcome/anatomopathologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Neuro Oncol ; 21(11): 1389-1400, 2019 11 04.
Article de Anglais | MEDLINE | ID: mdl-31127849

RÉSUMÉ

BACKGROUND: The Ras signaling pathway is commonly dysregulated in human malignant peripheral nerve sheath tumors (MPNSTs). It is well known that galectin-1 (Gal-1) is essential to stabilize membrane Ras and thereby induce the activation of Ras. However, the role of Gal-1 in MPNST progression remains unknown. The aim of this study was to examine whether Gal-1 knockdown could have an effect on the Ras signaling pathway. METHODS: Cell viability, apoptosis assay, and colony formation were performed to examine the effects of inhibition of Gal-1 in MPNST cells. We used a human MPNST xenograft model to assess growth and metastasis inhibitory effects of Gal-1 inhibitor LLS2. RESULTS: Gal-1 was upregulated in MPNST patients and was highly expressed in MPNST cells. Knockdown of Gal-1 by small interfering (si)RNA in Gal-1 expressing MPNST cells significantly reduces cell proliferation through the suppression of C-X-C chemokine receptor type 4 (CXCR4) and the rat sarcoma viral oncogene homolog (RAS)/extracellular signal-regulated kinase (ERK) pathway, which are important oncogenic signaling in MPNST development. Moreover, Gal-1 knockdown induces apoptosis and inhibits colony formation. LLS2, a novel Gal-1 allosteric small molecule inhibitor, is cytotoxic against MPNST cells and was able to induce apoptosis and suppress colony formation in MPNST cells. LLS2 treatment and Gal-1 knockdown exhibited similar effects on the suppression of CXCR4 and RAS/ERK pathways. More importantly, inhibition of Gal-1 expression or function by treatment with either siRNA or LLS2 resulted in significant tumor responses in an MPNST xenograft model. CONCLUSION: Our results identified an oncogenic role of Gal-1 in MPNST and that its inhibitor, LLS2, is a potential therapeutic agent, applied topically or systemically, against MPNST.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Galectine 1/antagonistes et inhibiteurs , Neurofibrosarcome/anatomopathologie , Récepteurs CXCR4/antagonistes et inhibiteurs , Bibliothèques de petites molécules/pharmacologie , Protéines G ras/antagonistes et inhibiteurs , Animaux , Marqueurs biologiques tumoraux , Mouvement cellulaire , Prolifération cellulaire , Femelle , Galectine 1/génétique , Galectine 1/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , Souris , Souris nude , Neurofibrosarcome/traitement médicamenteux , Neurofibrosarcome/métabolisme , Pronostic , Petit ARN interférent/génétique , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , Transduction du signal , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines G ras/génétique , Protéines G ras/métabolisme
17.
Clin Nucl Med ; 44(6): 494-495, 2019 Jun.
Article de Anglais | MEDLINE | ID: mdl-30889004

RÉSUMÉ

MPNSTs (malignant peripheral nerve sheath tumors) are a highly malignant group of soft tissue sarcomas and carry a very poor prognosis. Metastasis to bilateral adrenal glands is very rare in such group of neoplasms. We discuss a case of 85-year-old man who was diagnosed with MPNST from prevertebral mass with metastases to bilateral adrenal glands and bone marrow from the beginning and role of serial F-FDG PET/CT scans in response assessment first to sunitinib (tyrosine kinase inhibitor) and then to liposomal doxorubicin.


Sujet(s)
Tumeurs de la surrénale/imagerie diagnostique , Tumeurs des gaines nerveuses/imagerie diagnostique , Neurofibrosarcome/imagerie diagnostique , Tomographie par émission de positons couplée à la tomodensitométrie , Tumeurs de la surrénale/secondaire , Sujet âgé de 80 ans ou plus , Antinéoplasiques/usage thérapeutique , Doxorubicine/usage thérapeutique , Fluorodésoxyglucose F18 , Humains , Mâle , Tumeurs des gaines nerveuses/traitement médicamenteux , Tumeurs des gaines nerveuses/anatomopathologie , Neurofibrosarcome/traitement médicamenteux , Neurofibrosarcome/anatomopathologie , Radiopharmaceutiques
18.
Oncologist ; 24(6): 857-863, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30126857

RÉSUMÉ

BACKGROUND: Sorafenib and dacarbazine have low single-agent response rates in metastatic sarcomas. As angiogenesis inhibitors can enhance the efficacy of chemotherapy, we investigated the combination of sorafenib and dacarbazine in select sarcoma subtypes. MATERIALS AND METHODS: Patients with leiomyosarcoma (LMS), synovial sarcoma (SS), or malignant peripheral nerve sheath tumors (MPNST) with up to two previous lines of therapy and adequate hepatic, renal, and marrow function received 3-week cycles of sorafenib at 400 mg oral twice daily and dacarbazine 1,000 mg/m2 intravenously (later reduced to 850 mg/m2). Patients were evaluated for response every 6 weeks. The primary objective was to determine the disease control rate (DCR) of sorafenib plus dacarbazine in the selected sarcoma subtypes. RESULTS: The study included 37 patients (19 female); median age was 55 years (range 26-87); and histologies included LMS (22), SS (11), and MPNST (4). The DCR was 46% (17/37). Median progression-free survival was 13.4 weeks. The RECIST response rate was 14% (5/37). The Choi response rate was 51% (19/37). Median overall survival was 13.2 months. Of the first 25 patients, 15 (60%) required dacarbazine dose reductions for hematologic toxicity, with one episode of grade 5 neutropenic fever. After reducing the starting dose of dacarbazine to 850 mg/m2, only 3 of the final 12 (25%) patients required dose reduction. CONCLUSION: This phase II study met its primary endpoint with an 18-week DCR of 46%. The clinical activity of dacarbazine plus sorafenib in patients with these diagnoses is modest. IMPLICATIONS FOR PRACTICE: Metastatic soft tissue sarcomas are a heterogeneous group of relatively rare malignancies. Most patients are treated with cytotoxic chemotherapy or targeted therapy in the form of tyrosine kinase inhibitors. Response rates are relatively low, and there is a need for better therapies. This clinical trial demonstrates that combining a cytotoxic therapy (dacarbazine) with an antiangiogenic small molecule (sorafenib) is feasible and associated with favorable disease-control rates; however, it also increases the potential for significant toxicity.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Neutropénie fébrile/épidémiologie , Léiomyosarcome/traitement médicamenteux , Neurofibrosarcome/traitement médicamenteux , Sarcome synovial/traitement médicamenteux , Administration par voie orale , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Dacarbazine/administration et posologie , Dacarbazine/effets indésirables , Relation dose-effet des médicaments , Calendrier d'administration des médicaments , Études de faisabilité , Neutropénie fébrile/diagnostic , Neutropénie fébrile/étiologie , Femelle , Humains , Léiomyosarcome/mortalité , Léiomyosarcome/anatomopathologie , Mâle , Adulte d'âge moyen , Neurofibrosarcome/mortalité , Neurofibrosarcome/anatomopathologie , Survie sans progression , Évaluation de la réponse des tumeurs solides aux traitements , Sarcome synovial/mortalité , Sarcome synovial/anatomopathologie , Indice de gravité de la maladie , Sorafénib/administration et posologie , Sorafénib/effets indésirables
19.
Acta Clin Belg ; 68(4): 309-10, 2013.
Article de Anglais | MEDLINE | ID: mdl-24455804

RÉSUMÉ

Drug-induced pancreatitis is rare (1.4-2%). This report describes a 20-year-old female patient who developed acute pancreatitis while being treated for neurosarcoma of abdominal wall with the ifosfamide and doxorubicin regimen. Although it is unusual, it is important to consider chemotherapeutic agents as a possible etiology for acute pancreatitis in patients presenting with gastrointestinal symptoms.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Neurofibrosarcome/traitement médicamenteux , Pancréatite/induit chimiquement , Paroi abdominale , Doxorubicine/administration et posologie , Femelle , Humains , Ifosfamide/administration et posologie , Neurofibrosarcome/imagerie diagnostique , Radiographie , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...