Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
bioRxiv ; 2024 May 18.
Article in English | MEDLINE | ID: mdl-38798583

ABSTRACT

The rapid and sustained proliferation in cancer cells requires accelerated protein synthesis. Accelerated protein synthesis and disordered cell metabolism in cancer cells greatly increase the risk of translation errors. ribosome-associated quality control (RQC) is a recently discovered mechanism for resolving ribosome collisions caused by frequent translation stalls. The role of the RQC pathway in cancer initiation and progression remains controversial and confusing. In this study, we investigated the pathogenic role of mitochondrial stress-induced protein carboxyl-terminal terminal alanine and threonine tailing (msiCAT-tailing) in glioblastoma (GBM), which is a specific RQC response to translational arrest on the outer mitochondrial membrane. We found that msiCAT-tailed mitochondrial proteins frequently exist in glioblastoma stem cells (GSCs). Ectopically expressed msiCAT-tailed mitochondrial ATP synthase F1 subunit alpha (ATP5α) protein increases the mitochondrial membrane potential and blocks mitochondrial permeability transition pore (MPTP) formation/opening. These changes in mitochondrial properties confer resistance to staurosporine (STS)-induced apoptosis in GBM cells. Therefore, msiCAT-tailing can promote cell survival and migration, while genetic and pharmacological inhibition of msiCAT-tailing can prevent the overgrowth of GBM cells. Highlights: The RQC pathway is disturbed in glioblastoma (GBM) cellsmsiCAT-tailing on ATP5α elevates mitochondrial membrane potential and inhibits MPTP openingmsiCAT-tailing on ATP5α inhibits drug-induced apoptosis in GBM cellsInhibition of msiCAT-tailing impedes overall growth of GBM cells.

2.
Cancers (Basel) ; 16(2)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38254796

ABSTRACT

Glioma progression is a complex process controlled by molecular factors that coordinate the crosstalk between tumor cells and components of the tumor microenvironment (TME). Among these, immune cells play a critical role in cancer survival and progression. The complex interplay between cancer cells and the immune TME influences the outcome of immunotherapy and other anti-cancer therapies. Here, we present an updated view of the pro- and anti-tumor activities of the main myeloid and lymphocyte cell populations in the glioma TME. We review the underlying mechanisms involved in crosstalk between cancer cells and immune cells that enable gliomas to evade the immune system and co-opt these cells for tumor growth. Lastly, we discuss the current and experimental therapeutic options being developed to revert the immunosuppressive activity of the glioma TME. Knowledge of the complex interplay that elapses between tumor and immune cells may help develop new combination treatments able to overcome tumor immune evasion mechanisms and enhance response to immunotherapies.

3.
Brain Sci ; 13(11)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-38002476

ABSTRACT

Primitive neuroectodermal tumors of the central nervous system, or CNS neuroblastoma, are rare neoplasms in children. Recently, methylation profiling enabled the discovery of four distinct entities of these tumors. The current treatment paradigm involves surgical resection followed by chemotherapy and radiation. However, upfront surgical resection carries high surgical morbidity in this patient population due to their young age, tumor vascularity, and often deep location in the brain. We report a case of CNS neuroblastoma that can be successfully treated with neoadjuvant chemotherapy followed by minimally invasive laser interstitial thermal therapy and radiation. The patient has complete treatment with no evidence of recurrence at one year follow-up. This case illustrates a potential paradigm shift in the treatment of these rare tumors can be treated using minimally invasive surgical approach to achieve a favorable outcome.

4.
Cancer Res ; 83(15): 2527-2542, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37219874

ABSTRACT

Glioblastoma (GBM) is an immunologically "cold" tumor that does not respond to current immunotherapy. Here, we demonstrate a fundamental role for the α-isoform of the catalytic subunit of protein phosphatase-2A (PP2Ac) in regulating glioma immunogenicity. Genetic ablation of PP2Ac in glioma cells enhanced double-stranded DNA (dsDNA) production and cGAS-type I IFN signaling, MHC-I expression, and tumor mutational burden. In coculture experiments, PP2Ac deficiency in glioma cells promoted dendritic cell (DC) cross-presentation and clonal expansion of CD8+ T cells. In vivo, PP2Ac depletion sensitized tumors to immune-checkpoint blockade and radiotherapy treatment. Single-cell analysis demonstrated that PP2Ac deficiency increased CD8+ T-cell, natural killer cell, and DC accumulation and reduced immunosuppressive tumor-associated macrophages. Furthermore, loss of PP2Ac increased IFN signaling in myeloid and tumor cells and reduced expression of a tumor gene signature associated with worse patient survival in The Cancer Genome Atlas. Collectively, this study establishes a novel role for PP2Ac in inhibiting dsDNA-cGAS-STING signaling to suppress antitumor immunity in glioma. SIGNIFICANCE: PP2Ac deficiency promotes cGAS-STING signaling in glioma to induce a tumor-suppressive immune microenvironment, highlighting PP2Ac as a potential therapeutic target to enhance tumor immunogenicity and improve response to immunotherapy.


Subject(s)
Glioblastoma , Glioma , Interferon Type I , Humans , Immunity, Innate , Interferon Type I/metabolism , Nucleotidyltransferases/genetics , Tumor Microenvironment
5.
J Clin Invest ; 133(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36757811

ABSTRACT

Stimulator of IFN genes type I (STING-Type I) IFN signaling in myeloid cells plays a critical role in effective antitumor immune responses, but STING agonists as monotherapy have shown limited efficacy in clinical trials. The mechanisms that downregulate STING signaling are not fully understood. Here, we report that protein phosphatase 2A (PP2A), with its specific B regulatory subunit Striatin 4 (STRN4), negatively regulated STING-Type I IFN in macrophages. Mice with macrophage PP2A deficiency exhibited reduced tumor progression. The tumor microenvironment showed decreased immunosuppressive and increased IFN-activated macrophages and CD8+ T cells. Mechanistically, we demonstrated that Hippo kinase MST1/2 was required for STING activation. STING agonists induced dissociation of PP2A from MST1/2 in normal macrophages, but not in tumor conditioned macrophages. Furthermore, our data showed that STRN4 mediated PP2A binding to and dephosphorylation of Hippo kinase MST1/2, resulting in stabilization of YAP/TAZ to antagonize STING activation. In human patients with glioblastoma (GBM), YAP/TAZ was highly expressed in tumor-associated macrophages but not in nontumor macrophages. We also demonstrated that PP2A/STRN4 deficiency in macrophages reduced YAP/TAZ expression and sensitized tumor-conditioned macrophages to STING stimulation. In summary, we demonstrated that PP2A/STRN4-YAP/TAZ has, in our opinion, been an unappreciated mechanism that mediates immunosuppression in tumor-associated macrophages, and targeting the PP2A/STRN4-YAP/TAZ axis can sensitize tumors to immunotherapy.


Subject(s)
Glioblastoma , Tumor-Associated Macrophages , Animals , Humans , Mice , Calmodulin-Binding Proteins , Macrophages , Protein Processing, Post-Translational , Signal Transduction , Tumor Microenvironment , Interferon Type I/metabolism
6.
Cancers (Basel) ; 14(9)2022 May 03.
Article in English | MEDLINE | ID: mdl-35565414

ABSTRACT

Pediatric medulloblastoma (MB) is the most common pediatric brain tumor with varying prognoses depending on the distinct molecular subtype. The four consensus subgroups are WNT, Sonic hedgehog (SHH), Group 3, and Group 4, which underpin the current 2021 WHO classification of MB. While the field of knowledge for treating this disease has significantly advanced over the past decade, a deeper understanding is still required to improve the clinical outcomes for pediatric patients, who are often vulnerable in ways that adult patients are not. Here, we discuss how recent insights into the pathogenesis of pediatric medulloblastoma have directed current and future research. This review highlights new developments in understanding the four molecular subtypes' pathophysiology, epigenetics, and therapeutic targeting. In addition, we provide a focused discussion of recent developments in imaging, and in the surgery, chemotherapy, and radiotherapy of pediatric medulloblastoma. The article includes a brief explanation of healthcare costs associated with medulloblastoma treatment.

7.
Neuromolecular Med ; 24(1): 18-22, 2022 03.
Article in English | MEDLINE | ID: mdl-34487301

ABSTRACT

As a multi-functional cellular organelle, mitochondrial metabolic reprogramming is well recognized as a hallmark of cancer. The center of mitochondrial metabolism is oxidative phosphorylation (OXPHOS), in which cells use enzymes to oxidize nutrients, thereby converting the chemical energy to the biological energy currency ATPs. OXPHOS also creates the mitochondrial membrane potential and serve as the driving force of other mitochondrial metabolic pathways and experiences significant reshape in the different stages of tumor progression. In this minireview, we reviewed the major mitochondrial pathways that are connected to OXPHOS and are affected in cancer cells. In addition, we summarized the function of novel bio-active molecules targeting mitochondrial metabolic processes such as OXPHOS, mitochondrial membrane potential and mitochondrial dynamics. These molecules exhibit intriguing preclinical and clinical results and have been proven to be promising antitumor candidates in recent studies.


Subject(s)
Glioblastoma , Oxidative Phosphorylation , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Dynamics
8.
J Neurosurg Pediatr ; 28(1): 50-53, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34020419

ABSTRACT

Laser interstitial thermal therapy (LITT) is increasingly used as a surgical option for the treatment of epilepsy. Placement of the laser fibers relies on stereotactic navigation with cranial fixation pins. In addition, the laser fiber is stabilized in the cranium during the ablation using a cranial bolt that assumes maturity of the skull. Therefore, younger infants (< 2 years of age) have traditionally not been considered as candidates for LITT. However, LITT is an appealing option for patients with familial epilepsy syndromes, such as tuberous sclerosis complex (TSC), due to the multiplicity of lesions and the likely need for multiple procedures. A 4-month-old infant with TSC presented with refractory focal seizures despite receiving escalating doses of 5 antiepileptic medications. Electrographic and clinical seizures occurred numerous times daily. Noninvasive evaluations, including MRI, magnetoencephalography, scalp EEG, and SPECT, localized the ictal onset to a left frontal cortical tuber in the premotor area. In this paper, the authors report a novel technique to overcome the challenges of performing LITT in an infant with an immature skull by repurposing the Navigus biopsy skull mount for stereotactic placement of a laser fiber using electromagnetic-based navigation. The patient underwent successful ablation of the tuber and remained seizure free 4 months postoperatively. To the authors' knowledge, this is the youngest reported patient to undergo LITT. A safe method is described to perform LITT in an infant using commonly available tools without dedicated instrumentation beyond standard stereotactic navigation, a biopsy platform, and the Visualase system.

9.
J Neurooncol ; 148(2): 231-244, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32342332

ABSTRACT

PURPOSE: Glioblastoma (GBM) carries a dismal prognosis despite standard multimodal treatment with surgery, chemotherapy and radiation. Immune checkpoint inhibitors, such as PD1 blockade, for treatment of GBM failed to show clinical benefit. Rational combination strategies to overcome resistance of GBM to checkpoint monotherapy are needed to extend the promise of immunotherapy to GBM management. Emerging evidence suggests that protein phosphatase 2A (PP2A) plays a critical role in the signal transduction pathways of both adaptive and innate immune cells and that inhibition of PP2A could enhance cancer immunity. We investigated the use of a PP2A inhibitor, LB-100, to enhance antitumor efficacy of PD1 blockade in a syngeneic glioma model. METHODS: C57BL/6 mice were implanted with murine glioma cell line GL261-luc or GL261-WT and randomized into 4 treatment arms: (i) control, (ii) LB-100, (iii) PD1 blockade and (iv) combination. Survival was assessed and detailed profiling of tumor infiltrating leukocytes was performed. RESULTS: Dual PP2A and PD1 blockade significantly improved survival compared with monotherapy alone. Combination therapy resulted in complete regression of tumors in about 25% of mice. This effect was dependent on CD4 and CD8 T cells and cured mice established antigen-specific secondary protective immunity. Analysis of tumor lymphocytes demonstrated enhanced CD8 infiltration and effector function. CONCLUSION: This is the first preclinical investigation of the effect of combining PP2A inhibition with PD1 blockade for GBM. This novel combination provided effective tumor immunotherapy and long-term survival in our animal GBM model.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Brain Neoplasms/immunology , Glioblastoma/immunology , Piperazines/administration & dosage , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Phosphatase 2/antagonists & inhibitors , Animals , Brain Neoplasms/prevention & control , Cell Line, Tumor , Drug Therapy, Combination/methods , Female , Glioblastoma/prevention & control , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Protein Phosphatase 2/immunology
10.
Front Cell Dev Biol ; 8: 620788, 2020.
Article in English | MEDLINE | ID: mdl-33520994

ABSTRACT

Glioblastoma (GBM) is the most common malignant brain cancer. Increasing evidence suggests that mitochondrial dysfunction plays a key role in GBM progression as mitochondria is essential in regulating cell metabolism, oxidative stress, and cell death. Meanwhile, the immune microenvironment in GBM is predominated by tumor-associated macrophages and microglia (TAM), which is a heterogenous population of myeloid cells that, in general, create an immunosuppressive milieu to support tumor growth. However, subsets of TAMs can be pro-inflammatory and thereby antitumor. Therapeutic strategies targeting TAMs are increasingly explored as novel treatment strategies for GBM. The connection between mitochondrial dysfunction and TAMs phenotype in the tumor microenvironment is unclear. This review aims to provide perspectives and discuss possible molecular mechanisms mediating the interplay between glioma mitochondrial dysfunction and TAMs phenotype in shaping tumor immune microenvironment.

11.
Neurosurg Focus ; 47(4): E4, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31574478

ABSTRACT

OBJECTIVE: Prenatal imaging has several critical roles in the diagnosis and management of myelomeningocele, including specific family counseling and the selection of fetal surgery or postnatal repair. In this study, the authors compared the accuracy of fetal MRI and prenatal ultrasonography (US) in predicting the spinal lesion level and assessed the correlation between imaging findings and motor function as independently evaluated by a physical therapist (PT) after birth. METHODS: A retrospective review of demographic and clinical data was performed to identify children who had been treated with postnatal myelomeningocele closure at a single institution between March 2013 and December 2018. Patients were eligible for inclusion if they had all of the following: prenatal US identifying the neural tube defect level, fetal MRI identifying the neural tube defect level, and postoperative PT evaluation identifying the motor deficit level. Statistical analysis was performed using Cohen's kappa coefficient to compare the US- and MRI-demonstrated lesion level and correlate these findings with the motor level assigned postnatally by a PT via manual muscle testing. RESULTS: Thirty-four patients met the inclusion criteria. The mean gestational age at US was 23.0 ± 4.7 weeks, whereas the mean gestational age at MRI was 24.0 ± 4.1 weeks. The mean time from surgery to the PT evaluation was 2.9 ± 1.9 days. Prenatal US and MRI were in agreement within one spinal level in 74% of cases (25/34, k = 0.43). When comparing the US-demonstrated spinal level with the PT-assigned motor level, the two were in agreement within one level in 65% of cases (22/34, k = 0.40). When comparing MRI-demonstrated spinal level with the PT motor level, the two were in agreement within one level in 59% of cases (20/34, k = 0.37). MRI and US were within two spinal levels of the PT evaluation in 79.4% and 85.3% of cases, respectively. MRI and US agreed within two levels in 97.1% of cases. Prenatal US and MRI were equivalent when comparing the difference between the imaged level and the postnatal motor deficit level (mean level difference: 1.12 ± 1.16 vs 1.17 ± 1.11, p = 0.86). CONCLUSIONS: Prenatal US and MRI equivalently predicted the postnatal motor deficit level in children with myelomeningocele. These data may be valuable in prenatal prognostication.


Subject(s)
Gestational Age , Meningomyelocele/surgery , Neural Tube Defects/surgery , Spine/surgery , Child , Female , Humans , Male , Pregnancy , Retrospective Studies , Ultrasonography, Prenatal/methods , Ventriculostomy/methods
13.
Eur J Hum Genet ; 27(10): 1599-1610, 2019 10.
Article in English | MEDLINE | ID: mdl-31227808

ABSTRACT

The posterior fossa of the cranium contains the cerebellum and brainstem. Processes that reduce the volume of the posterior fossa squeeze the cerebellum and brainstem caudally, resulting in Chiari I malformation (CM1). CM1 causes neck pain, balance issues, decreased motor skills and headaches in those affected. We have posterior fossa measurements and whole exome sequence data on individuals from 7 extended families from Russia that have a family history of CM1. We performed parametric linkage analyses using an autosomal dominant inheritance model with a disease allele frequency of 0.01 and a penetrance of 0.8 for carriers and 0.0 for non-carriers. Variant-based two-point linkage analysis and gene-based linkage analysis was performed. Our results found a genome-wide significant signal on chromosome 1q43-44 (max HLOD = 3.3) in the variant-based analysis and 12q23 (max HLOD = 4.2) in the gene-based analysis. In both cases, the signal was driven by a single (different) family that contained a long, linked haplotype across the region in question. Using functional annotation, we were able to identify several rare nonsynonymous variants that were enriched in each family. The best candidate genes were rs765865412:G>A in MYBPC1 for the 12q haplotype and rs61749963:A>G in COX20 for the 1q haplotype. Good candidate variants in the 1q haplotype were also identified in CEP170 and AKT. Further laboratory work is planned to verify the causality of these genes.


Subject(s)
Arnold-Chiari Malformation/diagnosis , Arnold-Chiari Malformation/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 1 , Genome-Wide Association Study , Phenotype , Computational Biology/methods , Cranial Fossa, Posterior/abnormalities , Female , Genetic Linkage , Genome-Wide Association Study/methods , Genotype , Humans , Lod Score , Magnetic Resonance Imaging , Male , Exome Sequencing
14.
Childs Nerv Syst ; 35(10): 1847-1851, 2019 10.
Article in English | MEDLINE | ID: mdl-31139903

ABSTRACT

The complex Chiari, characterized by abnormal craniocervical bony anatomy in addition to Chiari tonsillar herniation, is a relatively recent addition to the concepts surrounding the Chiari literature. The primary findings of complex Chiari include craniocervical kyphosis and retroflexed odontoid, both of which can be described with radiographic measurements. This manuscript will outline the background literature regarding Chiari craniocervical morphometrics and supply an algorithm for the general management of complex Chiari patients.


Subject(s)
Arnold-Chiari Malformation/diagnostic imaging , Atlanto-Axial Joint/diagnostic imaging , Atlanto-Occipital Joint/diagnostic imaging , Cervical Atlas/diagnostic imaging , Clinical Decision-Making , Practice Guidelines as Topic/standards , Arnold-Chiari Malformation/surgery , Atlanto-Axial Joint/surgery , Atlanto-Occipital Joint/surgery , Cervical Atlas/surgery , Clinical Decision-Making/methods , Decompression, Surgical/methods , Decompression, Surgical/standards , Humans
15.
J Immunother Cancer ; 7(1): 131, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31113486

ABSTRACT

Tumor immunology has changed the landscape of cancer treatment. Yet, not all patients benefit as cancer immune responsiveness (CIR) remains a limitation in a considerable proportion of cases. The multifactorial determinants of CIR include the genetic makeup of the patient, the genomic instability central to cancer development, the evolutionary emergence of cancer phenotypes under the influence of immune editing, and external modifiers such as demographics, environment, treatment potency, co-morbidities and cancer-independent alterations including immune homeostasis and polymorphisms in the major and minor histocompatibility molecules, cytokines, and chemokines. Based on the premise that cancer is fundamentally a disorder of the genes arising within a cell biologic process, whose deviations from normality determine the rules of engagement with the host's response, the Society for Immunotherapy of Cancer (SITC) convened a task force of experts from various disciplines including, immunology, oncology, biophysics, structural biology, molecular and cellular biology, genetics, and bioinformatics to address the complexity of CIR from a holistic view. The task force was launched by a workshop held in San Francisco on May 14-15, 2018 aimed at two preeminent goals: 1) to identify the fundamental questions related to CIR and 2) to create an interactive community of experts that could guide scientific and research priorities by forming a logical progression supported by multiple perspectives to uncover mechanisms of CIR. This workshop was a first step toward a second meeting where the focus would be to address the actionability of some of the questions identified by working groups. In this event, five working groups aimed at defining a path to test hypotheses according to their relevance to human cancer and identifying experimental models closest to human biology, which include: 1) Germline-Genetic, 2) Somatic-Genetic and 3) Genomic-Transcriptional contributions to CIR, 4) Determinant(s) of Immunogenic Cell Death that modulate CIR, and 5) Experimental Models that best represent CIR and its conversion to an immune responsive state. This manuscript summarizes the contributions from each group and should be considered as a first milestone in the path toward a more contemporary understanding of CIR. We appreciate that this effort is far from comprehensive and that other relevant aspects related to CIR such as the microbiome, the individual's recombined T cell and B cell receptors, and the metabolic status of cancer and immune cells were not fully included. These and other important factors will be included in future activities of the taskforce. The taskforce will focus on prioritization and specific actionable approach to answer the identified questions and implementing the collaborations in the follow-up workshop, which will be held in Houston on September 4-5, 2019.


Subject(s)
Immunotherapy , Neoplasms/therapy , Tumor Microenvironment/immunology , Advisory Committees , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Congresses as Topic , Disease Models, Animal , Humans , Medical Oncology/organization & administration , Neoplasms/genetics , Neoplasms/immunology , Societies, Medical/organization & administration , Treatment Outcome , Tumor Microenvironment/genetics
16.
J Neurosurg Pediatr ; 22(4): 348-351, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29979129

ABSTRACT

Osteogenesis imperfecta (OI) is an inherited connective tissue disorder that causes bone fragility and deformity. Neurological manifestations, including macrocephaly and hydrocephalus, have been reported. Increased vascular fragility or bleeding diathesis also predisposes OI patients to intracranial hemorrhage. The development of chronic subdural fluid collections or hydrocephalus may require CSF diversion. The authors report a previously unrecognized complication of CSF diversion in a patient with OI, that is, a delayed severe cranial deformity, presumably due to over-shunting. In addition to the cosmetic concern, the deformity caused severe headaches and tenderness. The patient underwent craniectomy and titanium mesh cranioplasty, which resulted in the complete resolution of symptoms. This report raises the possibility that over-shunting in patients with OI could predispose to the formation of cranial deformity requiring surgical intervention.


Subject(s)
Osteogenesis Imperfecta/surgery , Plastic Surgery Procedures/methods , Skull/pathology , Ventriculoperitoneal Shunt/adverse effects , Adolescent , Craniotomy/methods , Female , Humans , Skull/surgery
17.
Nat Commun ; 9(1): 2126, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844427

ABSTRACT

Mounting evidence suggests that inhibition of protein phosphatase-2A (PP2A), a serine/threonine phosphatase, could enhance anticancer immunity. However, drugs targeting PP2A are not currently available. Here, we report that a PP2A inhibitor, LB-100, when combined with anti-PD-1 (aPD-1) blockade can synergistically elicit a durable immune-mediated antitumor response in a murine CT26 colon cancer model. This effect is T-cell dependent, leading to regression of a significant proportion of tumors. Analysis of tumor lymphocytes demonstrates enhanced effector T-cell and reduced suppressive regulatory T-cell infiltration. Clearance of tumor establishes antigen-specific secondary protective immunity. A synergistic effect of LB-100 and aPD-1 blockade is also observed in B16 melanoma model. In addition, LB-100 activates the mTORC1 signaling pathway resulting in decreased differentiation of naive CD4 cells into regulatory T cells. There is also increased expression of Th1 and decreased expression of Th2 cytokines. These data highlight the translational potential of PP2A inhibition in combination with checkpoint inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Colonic Neoplasms/therapy , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , Piperazines/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Phosphatase 2/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Combined Chemotherapy Protocols , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Signal Transduction/immunology , Th1 Cells/immunology , Th2 Cells/immunology
18.
Cancer Lett ; 415: 217-226, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29199006

ABSTRACT

Atypical and anaplastic meningiomas (AAM) represent 20% of all meningiomas. They are associated with poor outcomes due to their tendency to recur. While surgery and radiation (RT) are first line therapy, no effective systemic medical treatment has been identified. Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase involved in cell cycle regulation and DNA repair. Here, we examined radiosensitizing effects of LB-100, a novel inhibitor of PP2A against AAM as a novel treatment strategy. Three human-derived immortalized meningioma cell lines, IOMM-LEE, GAR, and CH-157, were used to investigate the radio-sensitizing potential of LB-100 in AAM. Survival fraction by clonogenic assay, immunofluorescence, cell cycle analysis and protein expression were evaluated in vitro. The antitumor effects of combining LB-100 with RT were verified in vivo by using intracranial orthotopic xenograft mouse model. Pharmacologic PP2A inhibition with LB-100 prior to RT enhanced the radiosensitivity of meningioma cells and reduced survival fraction in clonogenic assays. LB-100 increased DNA double-strand breakage (measured by γ-H2AX), mitotic catastrophe cell death, and G2/M cell cycle arrest in irradiated meningioma cells. Also, LB-100 decreased activation of STAT3 and expression of its downstream proteins. In vivo, LB-100 and RT combined treatment prolonged the survival of mice with xenografts compared to RT alone. Taken together, these results provide convincing preclinical data to support the use of LB-100 as a radiosensitizing agent for treatment of malignant meningioma. Its potential for clinical application deserves further investigation.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Meningeal Neoplasms/therapy , Meningioma/therapy , Piperazines/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chemoradiotherapy/methods , Disease-Free Survival , Female , Humans , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningioma/metabolism , Meningioma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Phosphatase 2/metabolism
19.
Sci Rep ; 6: 36670, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830712

ABSTRACT

The blood-brain barrier (BBB) restricts the uptake of many neuro-therapeutic molecules, presenting a formidable hurdle to drug development in brain diseases. We proposed a new and dynamic in vivo-like three-dimensional microfluidic system that replicates the key structural, functional and mechanical properties of the blood-brain barrier in vivo. Multiple factors in this system work synergistically to accentuate BBB-specific attributes-permitting the analysis of complex organ-level responses in both normal and pathological microenvironments in brain tumors. The complex BBB microenvironment is reproduced in this system via physical cell-cell interaction, vascular mechanical cues and cell migration. This model possesses the unique capability to examine brain metastasis of human lung, breast and melanoma cells and their therapeutic responses to chemotherapy. The results suggest that the interactions between cancer cells and astrocytes in BBB microenvironment might affect the ability of malignant brain tumors to traverse between brain and vascular compartments. Furthermore, quantification of spatially resolved barrier functions exists within a single assay, providing a versatile and valuable platform for pharmaceutical development, drug testing and neuroscientific research.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Cell Communication , Models, Biological , Neoplasms, Experimental/metabolism , Tumor Microenvironment , A549 Cells , Animals , Astrocytes/pathology , Blood-Brain Barrier/pathology , Brain Neoplasms/pathology , Humans , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Rats
20.
Oncotarget ; 7(11): 12447-63, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26799670

ABSTRACT

The protein phosphatase 2A (PP2A) inhibitor, LB100, has been shown in pre-clinical studies to be an effective chemo- and radio-sensitizer for treatment of various cancers. We investigated effects associated with LB100 treatment alone and in combination with cisplatin for medulloblastoma (MB) in vitro and in vivo in an intracranial xenograft model. We demonstrated that LB100 had a potent effect on MB cells. By itself, LB100 inhibited proliferation and induced significant apoptosis in a range of pediatric MB cell lines. It also attenuated MB cell migration, a pre-requirement for invasion. When used in combination, LB100 enhanced cisplatin-mediated cytotoxic effects. Cell viability in the presence of 1 uM cisplatin alone was 61% (DAOY), 100% (D341), and 58% (D283), but decreased with the addition of 2 µM of LB100 to 26% (DAOY), 67% (D341), and 27% (D283), (p < 0.005). LB100 suppressed phosphorylation of the STAT3 protein and several STAT3 downstream targets. Also, LB100 directly increased cisplatin uptake and overcame cisplatin-resistance in vitro. Finally, LB100 exhibited potent in vivo anti-neoplastic activity in combination with cisplatin in an intracranial xenograft model.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cerebellar Neoplasms/drug therapy , Cisplatin/pharmacology , Medulloblastoma/drug therapy , Piperazines/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Line, Tumor , Cerebellar Neoplasms/enzymology , Cerebellar Neoplasms/pathology , Cisplatin/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Humans , Medulloblastoma/enzymology , Medulloblastoma/pathology , Mice , Mice, SCID , Piperazines/administration & dosage , Protein Phosphatase 2/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...