Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38674001

RESUMO

Medulloblastoma (MB) encompasses diverse subgroups, and leptomeningeal disease/metastasis (LMD) plays a substantial role in associated fatalities. Despite extensive exploration of canonical genes in MB, the molecular mechanisms underlying LMD and the involvement of the orthodenticle homeobox 2 (OTX2) gene, a key driver in aggressive MB Group 3, remain insufficiently understood. Recognizing OTX2's pivotal role, we investigated its potential as a catalyst for aggressive cellular behaviors, including migration, invasion, and metastasis. OTX2 overexpression heightened cell growth, motility, and polarization in Group 3 MB cells. Orthotopic implantation of OTX2-overexpressing cells in mice led to reduced median survival, accompanied by the development of spinal cord and brain metastases. Mechanistically, OTX2 acted as a transcriptional activator of the Mechanistic Target of Rapamycin (mTOR) gene's promoter and the mTORC2 signaling pathway, correlating with upregulated downstream genes that orchestrate cell motility and migration. Knockdown of mTOR mRNA mitigated OTX2-mediated enhancements in cell motility and polarization. Analysis of human MB tumor samples (N = 952) revealed a positive correlation between OTX2 and mTOR mRNA expression, emphasizing the clinical significance of OTX2's role in the mTORC2 pathway. Our results reveal that OTX2 governs the mTORC2 signaling pathway, instigating LMD in Group 3 MBs and offering insights into potential therapeutic avenues through mTORC2 inhibition.


Assuntos
Regulação Neoplásica da Expressão Gênica , Alvo Mecanístico do Complexo 2 de Rapamicina , Meduloblastoma , Neoplasias Meníngeas , Fatores de Transcrição Otx , Animais , Feminino , Humanos , Masculino , Camundongos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Neoplasias Cerebelares/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Meduloblastoma/genética , Meduloblastoma/patologia , Meduloblastoma/metabolismo , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/secundário , Fatores de Transcrição Otx/metabolismo , Fatores de Transcrição Otx/genética , Transdução de Sinais
2.
Cancer ; 128(8): 1605-1615, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35132615

RESUMO

BACKGROUND: Brain tumors are the leading cause of death from disease in children. Racial/ethnic minority children have poorer outcomes than White children; however, it is not clear whether this association is mediated by treatment received. METHODS: Children (aged 0-19 years) diagnosed with brain tumors in the National Cancer Database (2004-2016) were identified. Cox proportional hazards models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) between race/ethnicity (Black, Hispanic, Asian/Pacific Islander, American Indian/Alaska Native, or White [reference]) and death. An inverse odds weighted mediation analysis was performed with treatment received as the mediator. RESULTS: Among 22,469 cases, White children (69% of the sample) had significantly better overall 12.5-year survival (P < .01). Black children (13% of the sample) and Hispanic children (14% of the sample) had an increased risk of death overall and for glioblastoma and oligodendroglioma. Compared with Whites, Asian/Pacific Islander children had a higher risk of death from choroid plexus tumors and a lower risk of death from medulloblastoma. There were no statistically significant meditating effects by treatment received, although the estimate was borderline in Hispanic children (indirect HR, 1.08; 95% CI, 0.99-1.18). A treatment-independent association between race/ethnicity and death remained for Hispanic children (direct HR, 1.18; 95% CI, 1.04-1.33) and Black children (direct HR, 1.28; 95% CI, 1.13-1.45). If deaths in minorities had equaled those in White children, 5% fewer total deaths and 15% fewer minority deaths would have occurred. CONCLUSIONS: Survival disparities exist in pediatric brain tumors and are largely independent of treatment received, but other mechanisms linked to race/ethnicity remain important.


Assuntos
Neoplasias do Sistema Nervoso Central , Etnicidade , Adolescente , Adulto , Criança , Pré-Escolar , Hispânico ou Latino , Humanos , Lactente , Recém-Nascido , Grupos Minoritários , Programa de SEER , Estados Unidos/epidemiologia , Adulto Jovem
3.
Cancer ; 128(8): 1616-1625, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35132626

RESUMO

BACKGROUND: Central nervous system (CNS) tumors rank among the top 5 cancers diagnosed in young adults aged 20 to 39 years at diagnosis and show a clear male excess in incidence. It is unknown whether sex differences in survival persist across histologic types and depend on the treatment received. METHODS: From the National Cancer Database (2004-2016), young adults (aged 20-39 years) who had been diagnosed with CNS tumors were identified. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated as measures of association between sex and death via Cox regression. An inverse odds weighting mediation analysis was performed with treatment received as a mediator. RESULTS: There were 47,560 cases (47% male). Males had worse overall survival than females for 9 of 16 histologic types, including diffuse astrocytoma, glioblastoma, and meningioma (all P < .05). Males had an increased risk of death after a brain tumor diagnosis overall (HR, 1.47; 95% CI, 1.41-1.53) and for 8 histologies. There was a significant association between male sex and death overall that was mediated by treatment received (indirect-effect HR, 1.17; 95% CI, 1.15-1.18), but no single histology had a significant indirect effect. All histologies examined in mediation analyses had significant direct effects for sex. The excess mortality due to sex was 20% for all CNS tumors combined and 34% among males with CNS tumors. CONCLUSIONS: Overall, treatment received may mediate a portion of the association between sex and death after a CNS tumor, but sex itself appears to be a stronger risk factor for death in this study.


Assuntos
Neoplasias Encefálicas , Neoplasias do Sistema Nervoso Central , Glioblastoma , Adulto , Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/terapia , Neoplasias do Sistema Nervoso Central/epidemiologia , Neoplasias do Sistema Nervoso Central/terapia , Bases de Dados Factuais , Feminino , Humanos , Incidência , Masculino , Adulto Jovem
4.
J Am Acad Dermatol ; 84(6): 1554-1561, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32682884

RESUMO

BACKGROUND: Treatment with BRAF inhibitors (BRAFI) and MEK inhibitors (MEKI) causes cutaneous reactions in children, limiting dosing or resulting in treatment cessation. The spectrum and severity of these reactions is not defined. OBJECTIVE: To determine the frequency and spectrum of cutaneous reactions in children receiving BRAFI and MEKI and their effects on continued therapy. METHODS: A multicenter, retrospective study was conducted at 11 clinical sites in the United States and Canada enrolling 99 children treated with BRAFI and/or MEKI for any indication from January 1, 2012, to January 1, 2018. RESULTS: All children in this study had a cutaneous reaction; most had multiple, with a mean per patient of 3.5 reactions on BRAFI, 3.7 on MEKI, and 3.4 on combination BRAFI/MEKI. Three patients discontinued treatment because of a cutaneous reaction. Treatment was altered in 27% of patients on BRAFI, 39.5% on MEKI, and 33% on combination therapy. The cutaneous reactions most likely to alter treatment were dermatitis, panniculitis, and keratosis pilaris-like reactions for BRAFI and dermatitis, acneiform eruptions, and paronychia for MEKI. CONCLUSIONS: Cutaneous reactions are common in children receiving BRAFI and MEKI, and many result in alterations or interruptions in oncologic therapy. Implementing preventative strategies at the start of therapy may minimize cutaneous reactions.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Toxidermias/epidemiologia , Neoplasias/tratamento farmacológico , Paroniquia/epidemiologia , Inibidores de Proteínas Quinases/efeitos adversos , Adolescente , Canadá/epidemiologia , Criança , Pré-Escolar , Toxidermias/diagnóstico , Toxidermias/etiologia , Feminino , Humanos , Lactente , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Paroniquia/induzido quimicamente , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Estudos Retrospectivos , Estados Unidos/epidemiologia
5.
Curr Oncol Rep ; 23(4): 45, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33721151

RESUMO

PURPOSE OF REVIEW: Patients with neurofibromatosis type 1 (NF1) are at increased risk for benign and malignant neoplasms. Recently, targeted therapy with the MEK inhibitor class has helped address these needs. We highlight recent successes with selumetinib while acknowledging ongoing challenges for NF1 patients and future directions. RECENT FINDINGS: MEK inhibitors have demonstrated efficacy for NF1-related conditions, including plexiform neurofibromas and low-grade gliomas, two common causes of NF1-related morbidity. Active investigations for NF1-related neoplasms have benefited from advanced understanding of the genomic and cell signaling alterations in these conditions and development of sound preclinical animal models. Selumetinib has become the first FDA-approved targeted therapy for NF1 following its demonstrated efficacy for inoperable plexiform neurofibroma. Investigations of combination therapy and the development of a representative NF1 swine model hold promise for translating therapies for other NF1-associated pathology.


Assuntos
Benzimidazóis/uso terapêutico , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Neurofibromatose 1/tratamento farmacológico , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos , Neurofibroma Plexiforme/tratamento farmacológico , Neurofibromatose 1/genética , Medicina de Precisão , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais , Suínos
6.
J Neurooncol ; 147(3): 701-709, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32239432

RESUMO

BACKGROUND: The incidence of malignant peripheral nerve sheath tumors (MPNSTs) is low in the general population, although individuals with Neurofibromatosis Type I (NF1) are particularly susceptible. These tumors generally have a high probability of metastasis. The rate and types of second malignancies (SMNs) after a primary diagnosis of MPNST are not well characterized. We aimed to quantify the rate of SMNs among individuals with a first primary MPNST using population-based data. METHODS: We estimated age-standardized incidence rates (SIRs) for SMNs among 1,579 primary MPNST cases between ages 0-85+ using SEER 18 (2000-2015). We estimated incidence rate ratios (IRRs) and 95% confidence intervals (95% CI) for SMNs in MPNST cases compared with general population rates. We conducted sex-stratified and age-restricted analyses (< 30 years at diagnosis). RESULTS: Seven percent (108/1579) of MPNST cases developed a SMN (SIR of 4635 cases/million). Compared to the general population, MPNST cases were more likely to develop SMNs (IRR: 29.3; 95% CI 23.8-34.8) and had a much higher rate of second MPNSTs (IRR: 15,992.9; 95% CI 9594.5-22,391.3). Aside from a second MPNST, second cancers were frequently diagnosed in the breast, lung, skin, and soft tissue in females and were myeloid and skin malignancies in males. When restricted to < 30 years of age, second MPNSTs were the most common cancers diagnosed. CONCLUSIONS: The rate of SMNs among MPNST cases is tremendously higher than that observed among individuals with other cancers, particularly for second MPNSTs. These findings suggest rates of SMNs may also be higher in NF1 individuals.


Assuntos
Segunda Neoplasia Primária/epidemiologia , Neurofibrossarcoma/epidemiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Feminino , Humanos , Incidência , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Adulto Jovem
7.
Am J Med Genet A ; 179(12): 2433-2446, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31566897

RESUMO

While neurofibromatosis type 1 (NF1) and Noonan syndrome (NS) are clinically distinct genetic syndromes, they have overlapping features because they are caused by pathogenic variants in genes encoding molecules within the Ras-mitogen-activated protein kinase signaling pathway. Increased risk for emotional and behavioral challenges has been reported in both children and adults with these syndromes. The current study examined parent-report and self-report measures of emotional functioning among children with NF1 and NS as compared to their unaffected siblings. Parents and children with NS (n = 39), NF1 (n = 39), and their siblings without a genetic condition (n = 32) completed well-validated clinical symptom rating scales. Results from parent questionnaires indicated greater symptomatology on scales measuring internalizing behaviors and symptoms of attention deficit hyperactivity disorder (ADHD) in both syndrome groups as compared with unaffected children. Frequency and severity of emotional and behavioral symptoms were remarkably similar across the two clinical groups. Symptoms of depression and anxiety were higher in children who were also rated as meeting symptom criteria for ADHD. While self-report ratings by children generally correlated with parent ratings, symptom severity was less pronounced. Among unaffected siblings, parent ratings indicated higher than expected levels of anxiety. Study findings may assist with guiding family-based interventions to address emotional challenges.


Assuntos
Emoções , Neurofibromatose 1/diagnóstico , Síndrome de Noonan/diagnóstico , Adolescente , Fatores Etários , Criança , Comportamento Infantil , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Masculino , Neurofibromatose 1/genética , Síndrome de Noonan/genética , Pais , Fenótipo , Autorrelato , Irmãos , Inquéritos e Questionários
8.
J Pediatr Hematol Oncol ; 39(4): 306-308, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28085744

RESUMO

INTRODUCTION: Neurofibromatosis type 1 (NF1) is the most commonly inherited autosomal dominant disorder in humans. NF1 patients have increased risk for gastrointestinal stromal tumors (GISTs). A Meckel's diverticulum (MD) represents a persistent embryonic omphalomesenteric duct characterized as a true diverticulum located near the ileocecal valve. We report a unique clinical case whereby a patient with NF1 developed a GIST within a MD. CASE: An adolescent male with NF1 presented with persistent lower abdominal pain. Clinical evaluation demonstrated a large pelvic mass. In the operating room, the mass was noted to emerge from a MD. Final pathology demonstrated a GIST with negative margins and CD117 positivity. DISCUSSION: Patients with NF1 are at increased risk for mesenchymal tumors including malignant peripheral nerve sheath tumors. GISTs are the most important and frequent non-neurological malignancy in NF1 and develop in ∼7% of NF1 patients. GISTs tend to be multifocal in NF1; however, they rarely occur within a Meckel's diverticula. CONCLUSIONS: Our case represents a rare case of a patient with NF1 who developed a symptomatic GIST within a MD. We recommend utilizing laparoscopy to determine resectability and clarify the diagnosis in this unique patient population who are at risk for multiple neoplasms.


Assuntos
Tumores do Estroma Gastrointestinal/patologia , Divertículo Ileal/diagnóstico , Neurofibromatose 1/complicações , Adolescente , Diagnóstico Diferencial , Humanos , Laparoscopia , Masculino , Neurofibromatose 1/patologia
10.
J Pediatr Hematol Oncol ; 38(6): e196-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27203570

RESUMO

We report a patient with stage 3 ganglioneuroblastoma who initially presented with clinical and laboratory features consistent with mucopolysaccharidosis including coarse facial features, developmental delay, and an elevated quantitative urine glycosaminoglycan (GAG) level. All mucopolysaccharidosis features resolved following successful treatment of neuroblastoma. High GAG levels have been documented in the pediatric oncology literature, yet not as a potential marker of malignancy or other target for clinical utility. This patient prompts further investigation into the relationship between neuroblastoma and elevated GAG levels.


Assuntos
Ganglioneuroblastoma/complicações , Glicosaminoglicanos/urina , Mucopolissacaridoses/etiologia , Ganglioneuroblastoma/terapia , Humanos , Lactente
11.
Am J Med Genet A ; 167A(12): 3186-91, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26364901

RESUMO

We report a patient with a constitutional missense mutation in SMARCB1, Coffin-Siris Syndrome (CSS), and schwannomatosis. CSS is a rare congenital syndrome with characteristic clinical findings. This thirty-three-year-old man was diagnosed early in life with the constellation of moderate intellectual disability, hypotonia, mild microcephaly, coarse facies, wide mouth with full lips, hypoplasia of the digits, and general hirsutism. At age 26, he was found to have schwannomatosis after presenting with acute spinal cord compression. Blood and tissue analysis of multiple subsequent schwannoma resections revealed a germline missense mutation of SMARCB1, acquired loss of 22q including SMARCB1 and NF2 and mutation of the remaining NF2 wild-type allele-thus completing the four-hit, three-event mechanism associated with schwannomatosis. Variations in five genes have been associated with the Coffin-Siris phenotype: ARID1A, ARID1B, SMARCA4, SMARCB1, and SMARCE1. Of these genes, SMARCB1 has a well-established association with schwannomatosis and malignancy. This is the first report of a patient with a constitutional missense mutation of SMARCB1 resulting in CSS and subsequent development of schwannomatosis. This finding demonstrates that a SMARCB1 mutation may be the initial "hit" (constitutional) for a genetic disorder with subsequent risk of developing schwannomas and other malignancies, and raises the possibility that other patients with switch/sucrose non-fermenting (SWI/SNF) mutations may be at increased risk for tumors.


Assuntos
Anormalidades Múltiplas/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Face/anormalidades , Deformidades Congênitas da Mão/genética , Deficiência Intelectual/genética , Micrognatismo/genética , Mutação de Sentido Incorreto/genética , Pescoço/anormalidades , Neurilemoma/genética , Neurofibromatoses/genética , Neoplasias Cutâneas/genética , Fatores de Transcrição/genética , Anormalidades Múltiplas/patologia , Adulto , Exoma/genética , Face/patologia , Deformidades Congênitas da Mão/patologia , Humanos , Deficiência Intelectual/patologia , Masculino , Micrognatismo/patologia , Pescoço/patologia , Neurilemoma/patologia , Neurofibromatoses/patologia , Fenótipo , Prognóstico , Proteína SMARCB1 , Neoplasias Cutâneas/patologia
12.
Crit Rev Immunol ; 34(5): 399-432, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25404047

RESUMO

Anti-tumor immunotherapy using tumor lysate-based vaccines has made great advances over recent decades. Cancer vaccines aim to elicit adaptive immune responses through various pathways by providing tumor and tumor-associated antigens with an immune stimulant or adjuvant. These anti-tumor vaccines are therefore developed as personalized treatments. Utilizing tumors as a source of vaccine antigens in immunotherapy has demonstrated promising results with minimal toxicity. However, to date, researchers have failed to overcome the overpowering immune suppressive effects within the tumor microenvironment. Immune suppression occurs naturally via multiple mechanisms. These mechanisms serve an important homeostatic role restoring a normal tissue microenvironment following an inflammatory response. Due to these suppressive mechanisms and the inherent heterogeneity of tumors, it is imperative to then elicit and maintain a specific tumoricidal response if vaccine therapy or some other combination of reagents is chosen. In this review, we focus on the historical use of tumors as a source of antigens to elicit a tumoricidal response and the limitations encountered that prevent greater success in immunotherapy. We describe the advantages and disadvantages of various vaccines and their ineffectiveness due to tumor-induced immune suppression.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/uso terapêutico , Doenças do Cão/terapia , Adjuvantes Imunológicos/uso terapêutico , Animais , Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/veterinária , Doenças do Cão/imunologia , Cães , Humanos , Terapia de Imunossupressão , Medicina de Precisão , Indução de Remissão , Falha de Tratamento , Evasão Tumoral , Microambiente Tumoral
13.
Pediatr Blood Cancer ; 61(4): 737-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24019247

RESUMO

Castleman disease is a rare lymphoproliferative disorder, which presents in a unicentric or multicentric fashion. Multicentric Castleman disease (MCD) is associated with significant systemic symptoms, in part related to the underlying role of interleukin-6 in disease pathogenesis. Treatment for MCD has not been well established and prognosis has historically been poor. We present a case of severe MCD in a pediatric patient who has shown sustained remission following multi-agent chemotherapy and targeted maintenance therapy with the interleukin-6 receptor inhibitor, tocilizumab. This represents the first case report of sustained remission of MCD in a pediatric patient following discontinuation of tocilizumab therapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Hiperplasia do Linfonodo Gigante/tratamento farmacológico , Adolescente , Quimioterapia Combinada , Humanos , Masculino , Prognóstico , Receptores de Interleucina-6/antagonistas & inibidores , Indução de Remissão
14.
J Pediatr Hematol Oncol ; 36(3): e177-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24309598

RESUMO

Turcot syndrome is a rare inherited condition of colonic polyposis associated with central nervous system tumors. We report a patient with a novel adenomatous polyposis coli gene mutation leading to a severe phenotype including medulloblastoma, low-grade fibromyxoid sarcoma following cranial radiation, pilomatrixomas, colonic adenomas, and abdominal desmoid tumor following colectomy, all of which were successfully treated. Multiple tumors may be seen in patients with Turcot syndrome but the occurrence of sarcomas is rare. This case highlights the importance of close follow-up for patients with Turcot syndrome and the importance of a broad differential diagnosis in evaluating a condition in which multiple tumors are frequently seen.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias Encefálicas/genética , Neoplasias Colorretais/genética , Mutação/genética , Síndromes Neoplásicas Hereditárias/genética , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/patologia , Criança , Neoplasias Colorretais/complicações , Neoplasias Colorretais/patologia , Feminino , Fibromatose Agressiva/genética , Fibromatose Agressiva/patologia , Fibromatose Agressiva/cirurgia , Fibrossarcoma/genética , Fibrossarcoma/patologia , Fibrossarcoma/radioterapia , Humanos , Meduloblastoma/genética , Meduloblastoma/patologia , Meduloblastoma/radioterapia , Síndromes Neoplásicas Hereditárias/complicações , Síndromes Neoplásicas Hereditárias/patologia , Fenótipo , Prognóstico
15.
bioRxiv ; 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38585724

RESUMO

Neurofibromatosis Type 1 (NF1) is a common cancer predisposition syndrome, caused by heterozygous loss of function mutations in the tumor suppressor gene NF1. Individuals with NF1 develop benign tumors of the peripheral nervous system (neurofibromas), originating from the Schwann cell linage after somatic loss of the wild type NF1 allele, some of which progress further to malignant peripheral nerve sheath tumors (MPNST). There is only one FDA approved targeted therapy for symptomatic plexiform neurofibromas and none approved for MPNST. The genetic basis of NF1 syndrome makes associated tumors ideal for using synthetic drug sensitivity approaches to uncover therapeutic vulnerabilities. We developed a drug discovery pipeline to identify therapeutics for NF1-related tumors using isogeneic pairs of NF1-proficient and deficient immortalized human Schwann cells. We utilized these in a large-scale high throughput screen (HTS) for drugs that preferentially kill NF1-deficient cells, through which we identified 23 compounds capable of killing NF1-deficient Schwann cells with selectivity. Multiple hits from this screen clustered into classes defined by method of action. Four clinically interesting drugs from these classes were tested in vivo using both a genetically engineered mouse model of high-grade peripheral nerve sheath tumors and human MPNST xenografts. All drugs tested showed single agent efficacy in these models as well as significant synergy when used in combination with the MEK inhibitor selumetinib. This HTS platform yielded novel therapeutically relevant compounds for the treatment of NF1-associated tumors and can serve as a tool to rapidly evaluate new compounds and combinations in the future.

16.
Front Oncol ; 13: 1113121, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37035203

RESUMO

Background: Medulloblastoma, the most common malignant pediatric brain tumor, displays marked sex differences in prevalence of the four main molecular subgroups: SHH, WNT, Group 3 and Group 4. Males are more frequently diagnosed with SHH, Group 3 and 4 tumors, which have worse prognoses than WNT tumors. Little is known about sex differences in methylation profiles within subgroups. Methods: Using publicly available methylation data (Illumina HumanMethylation450K array), we compared beta values for males versus females. Differentially methylated positions (DMP) by sex within medulloblastoma subgroups were identified on the autosomes. DMPs were mapped to genes and Reactome pathway analysis was run by subgroup. Kaplan-Meier survival curves (Log-Rank p-values) were assessed for each sex within subgroup. MethylCIBERSORT was used to investigate the tumor microenvironment using deconvolution to estimate the abundances of immune cell types using DNA methylation data. Results: There were statistically significant differences in sex by medulloblastoma subgroups (chi-squared p-value=0.00004): Group 3 (n=144; 65% male), Group 4 (n=326; 67% male), SHH (n=223; 57% male) and WNT (n=70; 41% male). Females had worse survival than males for SHH (p-value=0.02). DMPs by sex were identified within subgroups: SHH (n=131), Group 4 (n=29), Group 3 (n=19), and WNT (n=16) and validated in an independent dataset. Unsupervised hierarchical clustering showed that sex-DMPs in SHH did not correlate with other tumor attributes. Ten genes with sex DMPs (RFTN1, C1orf103, FKBP1B, COL25A1, NPDC1, B3GNT1, FOXN3, RNASEH2C, TLE1, and PHF17) were shared across subgroups. Significant pathways (p<0.05) associated with DMPs were identified for SHH (n=22) and Group 4 (n=4) and included signaling pathways for RET proto-oncogene, advanced glycosylation end product receptor, regulation of KIT, neurotrophic receptors, NOTCH, and TGF-ß. In SHH, we identified DMPs in four genes (CDK6, COL25A1, MMP16, PRIM2) that encode proteins which are the target of therapies in clinical trials for other cancers. There were few sex differences in immune cell composition within tumor subgroups. Conclusion: There are sexually dimorphic methylation profiles for SHH medulloblastoma where survival differences were observed. Sex-specific therapies in medulloblastoma may impact outcomes.

17.
BMC Res Notes ; 16(1): 275, 2023 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-37848948

RESUMO

OBJECTIVE: In epidemiological and experimental research, high folic acid intake has been demonstrated to accelerate tumor development among populations with genetic and/or molecular susceptibility to cancer. Neurofibromatosis type 1 (NF1) is a common autosomal dominant disorder predisposing affected individuals to tumorigenesis, including benign plexiform neurofibromas; however, understanding of factors associated with tumor risk in NF1 patients is limited. Therefore, we investigated whether pregestational folic acid intake modified plexiform-like peripheral nerve sheath tumor risk in a transgenic NF1 murine model. RESULTS: We observed no significant differences in overall survival according to folate group. Relative to controls (180 days), median survival did not statistically differ in deficient (174 days, P = 0.56) or supplemented (177 days, P = 0.13) folate groups. Dietary folate intake was positively associated with RBC folate levels at weaning, (P = 0.023, 0.0096, and 0.0006 for deficient vs. control, control vs. supplemented, and deficient vs. supplemented groups, respectively). Dorsal root ganglia (DRG), brachial plexi, and sciatic nerves were assessed according to folate group. Mice in the folate deficient group had significantly more enlarged DRG relative to controls (P = 0.044), but no other groups statistically differed. No significant differences for brachial plexi or sciatic nerve enlargement were observed according to folate status.


Assuntos
Neoplasias de Bainha Neural , Neurofibroma Plexiforme , Neurofibroma , Neurofibromatose 1 , Humanos , Gravidez , Feminino , Animais , Camundongos , Neurofibromatose 1/genética , Neurofibromatose 1/complicações , Neurofibromatose 1/patologia , Ácido Fólico , Neurofibroma/complicações , Neurofibroma/patologia , Neurofibroma Plexiforme/complicações , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/patologia
18.
Cancer Epidemiol Biomarkers Prev ; 31(9): 1830-1838, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-35437584

RESUMO

BACKGROUND: Recent investigations of malignant peripheral nerve sheath tumor (MPNST) survival have reported higher mortality among non-White individuals. However, previous analyses have not examined the impact of socioeconomic status (SES) on these observations. This study aims to characterize factors associated with cause-specific MPNST survival, including information related to census-tract-level SES (CT-SES). METHODS: We identified 2,432 primary MPNSTs using the Surveillance, Epidemiology, and End Results (SEER) 18 (2000-2016) database. We used Cox proportional hazards modeling to estimate the effects of sex, race/ethnicity, CT-SES quintile, metastasis at diagnosis, tumor site, age at diagnosis, and treatment by surgery on survival. Models were fit in both the full population and, separately, stratified by race/ethnicity and age at diagnosis (<40 vs. ≥40). RESULTS: In adjusted models, age at diagnosis, CT-SES, and metastasis at diagnosis were associated with mortality. In race/ethnicity-stratified analysis, higher CT-SES was found to improve survival only in the White population. Among those diagnosed before age 40, metastasis at diagnosis and American Indian/Alaska Native race/ethnicity were associated with mortality, and both Hispanic ethnicity and Asian/Pacific Islander race were suggestive for increased mortality. Among cases, diagnoses at age 40 and above, age at diagnosis, male sex, and CT-SES were associated with mortality. CONCLUSIONS: This analysis provides evidence that among pediatric and young adult patients, non-White populations experience inferior survival compared with Whites, independent of CT-SES. Our findings also suggest that the effect of CT-SES on MPNST survival may differ by racial/ethnic group. IMPACT: These findings suggest that barriers to healthcare for certain racial/ethnic groups extend beyond SES.


Assuntos
Neoplasias , Neurofibrossarcoma , Adulto , Criança , Etnicidade , Humanos , Masculino , Grupos Raciais , Programa de SEER , Classe Social , Adulto Jovem
19.
J Clin Invest ; 132(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34882581

RESUMO

BACKGROUNDLong-term prognosis of WHO grade II low-grade gliomas (LGGs) is poor, with a high risk of recurrence and malignant transformation into high-grade gliomas. Given the relatively intact immune system of patients with LGGs and the slow tumor growth rate, vaccines are an attractive treatment strategy.METHODSWe conducted a pilot study to evaluate the safety and immunological effects of vaccination with GBM6-AD, lysate of an allogeneic glioblastoma stem cell line, with poly-ICLC in patients with LGGs. Patients were randomized to receive the vaccines before surgery (arm 1) or not (arm 2) and all patients received adjuvant vaccines. Coprimary outcomes were to evaluate safety and immune response in the tumor.RESULTSA total of 17 eligible patients were enrolled - 9 in arm 1 and 8 in arm 2. This regimen was well tolerated with no regimen-limiting toxicity. Neoadjuvant vaccination induced upregulation of type-1 cytokines and chemokines and increased activated CD8+ T cells in peripheral blood. Single-cell RNA/T cell receptor sequencing detected CD8+ T cell clones that expanded with effector phenotype and migrated into the tumor microenvironment (TME) in response to neoadjuvant vaccination. Mass cytometric analyses detected increased tissue resident-like CD8+ T cells with effector memory phenotype in the TME after the neoadjuvant vaccination.CONCLUSIONThe regimen induced effector CD8+ T cell response in peripheral blood and enabled vaccine-reactive CD8+ T cells to migrate into the TME. Further refinements of the regimen may have to be integrated into future strategies.TRIAL REGISTRATIONClinicalTrials.gov NCT02549833.FUNDINGNIH (1R35NS105068, 1R21CA233856), Dabbiere Foundation, Parker Institute for Cancer Immunotherapy, and Daiichi Sankyo Foundation of Life Science.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Carboximetilcelulose Sódica/análogos & derivados , Glioma , Terapia Neoadjuvante , Poli I-C/administração & dosagem , Polilisina/análogos & derivados , Microambiente Tumoral/imunologia , Vacinação , Adulto , Idoso , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Carboximetilcelulose Sódica/administração & dosagem , Feminino , Glioma/imunologia , Glioma/terapia , Humanos , Masculino , Pessoa de Meia-Idade , Polilisina/administração & dosagem
20.
Neurotherapeutics ; 18(3): 1980-1994, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33829411

RESUMO

Numerous therapies aimed at driving an effective anti-glioma response have been employed over the last decade; nevertheless, survival outcomes for patients remain dismal. This may be due to the expression of immune-checkpoint ligands such as PD-L1 by glioblastoma (GBM) cells which interact with their respective receptors on tumor-infiltrating effector T cells curtailing the activation of anti-GBM CD8+ T cell-mediated responses. Therefore, a combinatorial regimen to abolish immunosuppression would provide a powerful therapeutic approach against GBM. We developed a peptide ligand (CD200AR-L) that binds an uncharacterized CD200 immune-checkpoint activation receptor (CD200AR). We sought to test the hypothesis that CD200AR-L/CD200AR binding signals via he DAP10&12 pathways through in vitro studies by analyzing transcription, protein, and phosphorylation, and in vivo loss of function studies using inhibitors to select signaling molecules. We report that CD200AR-L/CD200AR binding induces an initial activation of the DAP10&12 pathways followed by a decrease in activity within 30 min, followed by reactivation via a positive feedback loop. Further in vivo studies using DAP10&12KO mice revealed that DAP10, but not DAP12, is required for tumor control. When we combined CD200AR-L with an immune-stimulatory gene therapy, in an intracranial GBM model in vivo, we observed increased median survival, and long-term survivors. These studies are the first to characterize the signaling pathway used by the CD200AR, demonstrating a novel strategy for modulating immune checkpoints for immunotherapy currently being analyzed in a phase I adult trial.


Assuntos
Antígenos CD/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Inibidores de Checkpoint Imunológico/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/deficiência , Sequência de Aminoácidos , Animais , Antígenos CD/administração & dosagem , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Feminino , Terapia Genética/métodos , Glioma/tratamento farmacológico , Glioma/genética , Inibidores de Checkpoint Imunológico/administração & dosagem , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Terciária de Proteína , Receptores Imunológicos/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa