Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
Neurooncol Adv ; 4(1): vdab184, 2022.
Article in English | MEDLINE | ID: mdl-35118381

ABSTRACT

BACKGROUND: Hypoxia is a prominent feature of solid tumors and can function as fertile environment for oncolytic anaerobic bacteria such as Clostridium novyi-NT (C. novyi-NT) where it can induce tumor destruction in mice and patients. However, two major obstacles have limited its use, namely the host inflammatory response and the incomplete clearance of normoxic tumor areas. METHODS: In this study, we first used a subcutaneous tumor model of a glioblastoma (GBM) cell line in immunocompetent mice to investigate the local distribution of tumor hypoxia, kinetics of C. novyi-NT germination and spread, and the local host immune response. We subsequently applied the acquired knowledge to develop a C. novyi-NT therapy in an orthotopic rabbit brain tumor model. RESULTS: We found that local accumulation of granular leukocytes, mainly neutrophils, could impede the spread of bacteria through the tumor and prevent complete oncolysis. Depletion of neutrophils via anti-Ly6G antibody or bone marrow suppression using hydroxyurea significantly improved tumor clearance. We then applied this approach to rabbits implanted with an aggressive intracranial brain tumor and achieved long-term survival in majority of the animals without apparent toxicity. CONCLUSION: These results indicated that depleting neutrophils can greatly enhance the safety and efficacy of C. novyi-NT cancer therapy for brain tumors.

3.
Sci Rep ; 11(1): 6517, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33753770

ABSTRACT

Signal Transducer and Activator of Transcription 3 (STAT3) is a transcription factor and an oncogene product, which plays a pivotal role in tumor progression. Therefore, targeting persistent STAT3 signaling directly is an attractive anticancer strategy. The aim of this study is to test the efficacy of a novel STAT3 small molecule inhibitor, LLL12B, in suppressing medulloblastoma cells in vitro and tumor growth in vivo. LLL12B selectively inhibited the induction of STAT3 phosphorylation by interleukin-6 but not induction of STAT1 phosphorylation by INF-γ. LLL12B also induced apoptosis in human medulloblastoma cells. In addition, LLL12B exhibited good oral bioavailability in vivo and potent suppressive activity in tumor growth of medulloblastoma cells in vivo. Besides, combining LLL12B with cisplatin showed greater inhibition of cell viability and tumorsphere formation as well as induction of apoptosis comparing to single agent treatment in medulloblastoma cells. Furthermore, LLL12B and cisplatin combination exhibited greater suppression of medulloblastoma tumor growth than monotherapy in vivo. The present study supported that LLL12B is a novel therapeutic agent for medulloblastoma and the combination of LLL12B with a chemotherapeutic agent cisplatin may be an effective approach for medulloblastoma therapy.


Subject(s)
Anthraquinones/pharmacology , Interferon-gamma/genetics , Medulloblastoma/drug therapy , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Sulfonamides/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Heterografts , Humans , Interleukin-6/genetics , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Phosphorylation/drug effects
4.
Br J Cancer ; 124(9): 1566-1580, 2021 04.
Article in English | MEDLINE | ID: mdl-33658640

ABSTRACT

BACKGROUND: MPNST is a rare soft-tissue sarcoma that can arise from patients with NF1. Existing chemotherapeutic and targeted agents have been unsuccessful in MPNST treatment, and recent findings implicate STAT3 and HIF1-α in driving MPNST. The DNA-binding and transcriptional activity of both STAT3 and HIF1-α is regulated by Redox factor-1 (Ref-1) redox function. A first-generation Ref-1 inhibitor, APX3330, is being tested in cancer clinical trials and could be applied to MPNST. METHODS: We characterised Ref-1 and p-STAT3 expression in various MPNST models. Tumour growth, as well as biomarkers of apoptosis and signalling pathways, were measured by qPCR and western blot following treatment with inhibitors of Ref-1 or STAT3. RESULTS: MPNSTs from Nf1-Arfflox/floxPostnCre mice exhibit significantly increased positivity of p-STAT3 and Ref-1 expression when malignant transformation occurs. Inhibition of Ref-1 or STAT3 impairs MPNST growth in vitro and in vivo and induces apoptosis. Genes highly expressed in MPNST patients are downregulated following inhibition of Ref-1 or STAT3. Several biomarkers downstream of Ref-1 or STAT3 were also downregulated following Ref-1 or STAT3 inhibition. CONCLUSIONS: Our findings implicate a unique therapeutic approach to target important MPNST signalling nodes in sarcomas using new first-in-class small molecules for potential translation to the clinic.


Subject(s)
Biomarkers, Tumor/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gene Expression Regulation, Neoplastic , Neurofibrosarcoma/pathology , STAT3 Transcription Factor/metabolism , Adolescent , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neurofibrosarcoma/genetics , Neurofibrosarcoma/metabolism , Prognosis , STAT3 Transcription Factor/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Neurooncol Adv ; 3(1): vdaa154, 2021.
Article in English | MEDLINE | ID: mdl-33506200

ABSTRACT

BACKGROUND: Mebendazole is an anthelmintic drug introduced for human use in 1971 that extends survival in preclinical models of glioblastoma and other brain cancers. METHODS: A single-center dose-escalation and safety study of mebendazole in 24 patients with newly diagnosed high-grade gliomas in combination with temozolomide was conducted. Patients received mebendazole in combination with adjuvant temozolomide after completing concurrent radiation plus temozolomide. Dose-escalation levels were 25, 50, 100, and 200 mg/kg/day of oral mebendazole. A total of 15 patients were enrolled at the highest dose studied of 200 mg/kg/day. Trough plasma levels of mebendazole were measured at 4, 8, and 16 weeks. RESULTS: Twenty-four patients (18 glioblastoma and 6 anaplastic glioma) were enrolled with a median age of 49.8 years. Four patients (at 200 mg/kg) developed elevated grade 3 alanine aminotransferase (ALT) and/or aspartate transaminase (AST) after 1 month, which reversed with lower dosing or discontinuation. Plasma levels of mebendazole were variable but generally increased with dose. Kaplan-Meier analysis showed a 21-month median overall survival with 41.7% of patients alive at 2 years and 25% at 3 and 4 years. Median progression-free survival (PFS) from the date of diagnosis for 17 patients taking more than 1 month of mebendazole was 13.1 months (95% confidence interval [CI]: 8.8-14.6 months) but for 7 patients who received less than 1 month of mebendazole PFS was 9.2 months (95% CI: 5.8-13.0 months). CONCLUSION: Mebendazole at doses up to 200 mg/kg demonstrated long-term safety and acceptable toxicity. Further studies are needed to determine mebendazole's efficacy in patients with malignant glioma.

6.
Genes (Basel) ; 11(7)2020 07 08.
Article in English | MEDLINE | ID: mdl-32650362

ABSTRACT

Patients with RASopathy Neurofibromatosis 1 (NF1) are at a markedly increased risk of the development of benign and malignant tumors. Malignant tumors are often recalcitrant to treatments and associated with poor survival; however, no chemopreventative strategies currently exist. We thus evaluated the effect of mebendazole, alone or in combination with cyclooxygenase-2 (COX-2) inhibitors, on the prevention of NF1-related malignancies in a cisNf1+/-;Tp53+/- (NPcis) mouse model of NF1. Our in vitro findings showed that mebendazole (MBZ) inhibits the growth of NF1-related malignant peripheral nerve sheath tumors (MPNSTs) through a reduction in activated guanosine triphosphate (GTP)-bound Ras. The daily MBZ treatment of NPcis mice dosed at 195 mg/kg daily, initiated 60 days after birth, substantially delayed the formation of solid malignancies and increased median survival (p < 0.0001). Compared to placebo-treated mice, phosphorylated extracellular signal-regulated kinase (pERK) levels were decreased in the malignancies of MBZ-treated mice. The combination of MBZ with COX-2 inhibitor celecoxib (CXB) further enhanced the chemopreventative effect in female mice beyond each drug alone. These findings demonstrate the feasibility of a prevention strategy for malignancy development in high-risk NF1 individuals.


Subject(s)
Antineoplastic Agents/therapeutic use , Mebendazole/therapeutic use , Nerve Sheath Neoplasms/prevention & control , Neurofibromatosis 1/genetics , Animals , Antineoplastic Agents/administration & dosage , Celecoxib/administration & dosage , Celecoxib/therapeutic use , Cell Line, Tumor , Chemoprevention , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/therapeutic use , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Male , Mebendazole/administration & dosage , Mice , Mice, Inbred C57BL , Nerve Sheath Neoplasms/genetics , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , ras Proteins/metabolism
8.
Gene Ther ; 26(6): 277-286, 2019 06.
Article in English | MEDLINE | ID: mdl-31127187

ABSTRACT

Neurofibromatosis type 1, including the highly aggressive malignant peripheral nerve sheath tumors (MPNSTs), is featured by the loss of functional neurofibromin 1 (NF1) protein resulting from genetic alterations. A major function of NF1 is suppressing Ras activities, which is conveyed by an intrinsic GTPase-activating protein-related domain (GRD). In this study, we explored the feasibility of restoring Ras GTPase via exogenous expression of various GRD constructs, via gene delivery using a panel of adeno-associated virus (AAV) vectors in MPNST and human Schwann cells (HSCs). We demonstrated that several AAV serotypes achieved favorable transduction efficacies in those cells and a membrane-targeting GRD fused with an H-Ras C-terminal motif (C10) dramatically inhibited the Ras pathway and MPNST cells in a NF1-specific manner. Our results opened up a venue of gene replacement therapy in NF1-related tumors.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Neurofibromatosis 1/therapy , Neurofibromin 1/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Feasibility Studies , Genetic Vectors/genetics , Humans , Neurofibromin 1/chemistry , Neurofibromin 1/metabolism , Protein Domains , Schwann Cells/metabolism , ras Proteins/genetics , ras Proteins/metabolism
9.
Nature ; 564(7735): 273-277, 2018 12.
Article in English | MEDLINE | ID: mdl-30542164

ABSTRACT

Cytokine release syndrome (CRS) is a life-threatening complication of several new immunotherapies used to treat cancers and autoimmune diseases1-5. Here we report that atrial natriuretic peptide can protect mice from CRS induced by such agents by reducing the levels of circulating catecholamines. Catecholamines were found to orchestrate an immunodysregulation resulting from oncolytic bacteria and lipopolysaccharide through a self-amplifying loop in macrophages. Myeloid-specific deletion of tyrosine hydroxylase inhibited this circuit. Cytokine release induced by T-cell-activating therapeutic agents was also accompanied by a catecholamine surge and inhibition of catecholamine synthesis reduced cytokine release in vitro and in mice. Pharmacologic catecholamine blockade with metyrosine protected mice from lethal complications of CRS resulting from infections and various biotherapeutic agents including oncolytic bacteria, T-cell-targeting antibodies and CAR-T cells. Our study identifies catecholamines as an essential component of the cytokine release that can be modulated by specific blockers without impairing the therapeutic response.


Subject(s)
Catecholamines/antagonists & inhibitors , Catecholamines/metabolism , Cytokines/adverse effects , Syndrome , Animals , Atrial Natriuretic Factor/pharmacology , CD3 Complex/antagonists & inhibitors , Catecholamines/biosynthesis , Cytokines/immunology , Epinephrine/metabolism , Female , Humans , Immunotherapy, Adoptive , In Vitro Techniques , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Norepinephrine/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , alpha-Methyltyrosine/pharmacology
10.
Oncotarget ; 8(16): 25955-25962, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28412733

ABSTRACT

Needle biopsy is an indispensable diagnostic tool in obtaining tumor tissue for diagnostic examination. Tumor cell seeding in the needle track during percutaneous needle biopsies has been reported for various types of cancers. The mechanical force of the biopsy both directly displaces the malignant cells and causes bleeding and fluid movement that can further disseminate cells. To prevent the risk of tumor cell seeding during biopsy, we developed a gelatin stick loaded with chemotherapeutics such as doxorubicin (DXR) that was inserted into the biopsy canal. The gelatin-doxorubicin sticks (GDSs) were created by passively loading precut gelatin foam strips (Gelfoam) with doxorubicin solution. The dried GDSs were inserted into the needle track through the sheath during the needle biopsy and eventually self-absorbed. We showed that this procedure prevented iatrogenic tumor seeding during needle biopsies in two subcutaneous tumor models. In an alternative application, using GDSs in intracranial brain tumor implantation avoided the outgrowth of tumor from the rodent brain, which could otherwise potentially fuse the tumor with the meninges and distort the results in therapeutic studies in rodent brain tumor models.


Subject(s)
Antineoplastic Agents/administration & dosage , Biopsy, Needle/instrumentation , Biopsy, Needle/methods , Gelatin , Neoplasm Seeding , Animals , Biopsy, Needle/adverse effects , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Disease Models, Animal , Doxorubicin/administration & dosage , Female , Gelatin/chemistry , Humans , Mice
11.
Neurotherapeutics ; 14(2): 298-306, 2017 04.
Article in English | MEDLINE | ID: mdl-28349408

ABSTRACT

The RASopathy neurofibromatosis 1 is an autosomal dominant hereditary cancer syndrome that represents a major risk for the development of malignancies, particularly malignant peripheral nerve sheath tumors (MPNSTs). MPNSTs are unique sarcomas that originate from the peripheral nerve and represent the only primary cancer of the peripheral nervous system. To date, surgery is the only treatment modality proven to have survival benefit for MPNSTs and even when maximal surgery is feasible, these tumors are rarely curable, despite the use of chemotherapy and radiation. In this review, we discuss the current state-of-the-art treatments for MPNSTs, latest therapeutic developments, and critical aspects of the underlying molecular and pathophysiology that appear promising for therapeutic developments in the future. In particular, we discuss the specific elements of cancer in the peripheral nerve and how that may impel development of unique therapies for this form of sarcoma.


Subject(s)
Neurilemmoma/genetics , Neurilemmoma/therapy , Neurofibromatosis 1/complications , Peripheral Nervous System Neoplasms/genetics , Peripheral Nervous System Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Humans , Neurilemmoma/complications , Peripheral Nervous System Neoplasms/complications , Treatment Outcome
12.
Oncotarget ; 7(42): 68571-68584, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27612418

ABSTRACT

Inheritance of a gene mutation leads to the initiation of 5 to 10% of most cancers, including colon cancer cases. We developed a chemoprevention strategy using a novel combination of the non-steroidal anti-inflammatory (NSAID) sulindac plus the anthelminthic benzimidazole, mebendazole. This oral drug combination was effective in the ApcMin/+ mouse model of Familial Adenomatous Polyposis (FAP). Treatment with 35 mg/kg daily mebendazole reduced the number of intestinal adenomas by 56% (P = 0.0002), 160 ppm sulindac by 74% (P < 0.0001), and the combination by 90% (P < 0.0001). The combination significantly reduced microadenomas, polyp number and size in both the small intestines and colon when compared to untreated controls or sulindac alone. Mebendazole as a single agent decreased COX2 expression, blood vessel formation, VEGFR2 phosphorylation, and worked synergistically with sulindac to reduce overexpression of MYC, BCL2, and various pro-inflammatory cytokines. Given the low toxicity of mebendazole, these preclinical findings support the consideration of clinical trials for high risk cancer patients using mebendazole either alone or in combination. The findings have implications for populations with moderate and above risk for developing cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Transformation, Neoplastic/drug effects , Colon/drug effects , Colonic Neoplasms/prevention & control , Xenograft Model Antitumor Assays , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cell Line, Tumor , Colon/pathology , Colonic Neoplasms/pathology , HCT116 Cells , HT29 Cells , Humans , Intestine, Small/drug effects , Intestine, Small/pathology , Male , Mebendazole/administration & dosage , Mice, Inbred C57BL , Mice, Nude , Sulindac/administration & dosage
13.
Expert Opin Investig Drugs ; 25(8): 937-56, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27170161

ABSTRACT

INTRODUCTION: Despite substantial improvements in standards of care, the most common aggressive pediatric and adult high-grade gliomas (HGG) carry uniformly fatal diagnoses due to unique treatment limitations, high recurrence rates and the absence of effective treatments following recurrence. Recent advancements in our understanding of the pathophysiology, genetics and epigenetics as well as mechanisms of immune surveillance during gliomagenesis have created new knowledge to design more effective and target-directed therapies to improve patient outcomes. AREAS COVERED: In this review, the authors discuss the critical genetic, epigenetic and immunologic aberrations found in gliomas that appear rational and promising for therapeutic developments in the presence and future. The current state of the latest therapeutic developments including tumor-specific targeted drug therapies, metabolic targeting, epigenetic modulation and immunotherapy are summarized and suggestions for future directions are offered. Furthermore, they highlight contemporary issues related to the clinical development, such as challenges in clinical trials and toxicities. EXPERT OPINION: The commitment to understanding the process of gliomagenesis has created a catalogue of aberrations that depict multiple mechanisms underlying this disease, many of which are suitable to therapeutic inhibition and are currently tested in clinical trials. Thus, future treatment endeavors will employ multiple treatment modalities that target disparate tumor characteristics personalized to the patient's individual tumor.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Drug Design , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Epigenesis, Genetic , Glioma/genetics , Glioma/pathology , Humans , Immunotherapy/methods , Molecular Targeted Therapy , Neoplasm Recurrence, Local
14.
Genes Dis ; 3(2): 144-152, 2016 Jun.
Article in English | MEDLINE | ID: mdl-30258882

ABSTRACT

The attenuated anaerobic bacterium Clostridium novyi-NT (C. novyi-NT) is known for its ability to precisely germinate in and eradicate treatment-resistant hypoxic tumors in various experimental animal models and spontaneously occurring canine sarcomas. In this article, we review the therapeutic and toxicologic aspects of C. novyi-NT therapy, key challenges and limitations, and promising strategies to optimize its performance via recombinant DNA technology and immunotherapeutic approaches, to establish C. novyi-NT as an essential tool in cancer therapy.

15.
Clin Cancer Res ; 21(15): 3462-3470, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25862759

ABSTRACT

PURPOSE: Mebendazole (MBZ), first used as an antiparasitic drug, shows preclinical efficacy in models of glioblastoma and medulloblastoma. Three different mebendazole polymorphs (A, B, and C) exist, and a detailed assessment of the brain penetration, pharmacokinetics, and antitumor properties of each individual mebendazole polymorph is necessary to improve mebendazole-based brain cancer therapy. EXPERIMENTAL DESIGN AND RESULTS: In this study, various marketed and custom-formulated mebendazole tablets were analyzed for their polymorph content by IR spectroscopy and subsequently tested in an orthotopic GL261 mouse glioma model for efficacy and tolerability. The pharmacokinetics and brain concentration of mebendazole polymorphs and two main metabolites were analyzed by LC/MS. We found that polymorph B and C both increased survival in a GL261 glioma model, as B exhibited greater toxicity. Polymorph A showed no benefit. Polymorph B and C both reached concentrations in the brain that exceeded the IC50 in GL261 cells 29-fold. In addition, polymorph C demonstrated an AUC0₋24h brain-to-plasma (B/P) ratio of 0.82, whereas B showed higher plasma AUC and lower B/P ratio. In contrast, polymorph A presented markedly lower levels in the plasma and brain. Furthermore, the combination with elacridar was able to significantly improve the efficacy of polymorph C in GL261 glioma and D425 medulloblastoma models in mice. CONCLUSIONS: Among mebendazole polymorphs, C reaches therapeutically effective concentrations in the brain tissue and tumor with fewer side effects, and is the better choice for brain cancer therapy. Its efficacy can be further enhanced by combination with elacridar.


Subject(s)
Brain Neoplasms/drug therapy , Mebendazole/administration & dosage , Medulloblastoma/drug therapy , Neoplasms, Experimental/drug therapy , Animals , Brain Neoplasms/pathology , Chemistry, Pharmaceutical , Disease Models, Animal , Humans , Mebendazole/chemistry , Mebendazole/pharmacokinetics , Medulloblastoma/pathology , Mice , Neoplasms, Experimental/pathology , Neutrophils/drug effects
16.
Oncotarget ; 6(8): 5536-46, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25849940

ABSTRACT

Glioblastoma (GBM) is a highly aggressive primary brain tumor that is especially difficult to treat. The tumor's ability to withstand hypoxia leads to enhanced cancer cell survival and therapy resistance, but also yields a microenvironment that is in many aspects unique within the human body, thus offering potential therapeutic opportunities. The spore-forming anaerobic bacterium Clostridium novyi-NT(C. novyi-NT) has the ability to propagate in tumor-generated hypoxia, leading to oncolysis. Here, we show that intravenously injected spores of C. novyi-NT led to dramatic tumor destructions and significant survival increases in implanted, intracranial syngeneic F98 and human xenograft 060919 rat GBM models. C. novyi-NT germination was specific and confined to the neoplasm, with sparing of the normal brain parenchyma. All animals tolerated the bacteriolytic treatment, but edema and increased intracranial pressure could quickly be lethal if not monitored and medically managed with hydration and antibiotics. These results provide pre-clinical data supporting the development of this therapeutic approach for the treatment of patients with GBM.


Subject(s)
Brain Neoplasms/microbiology , Brain Neoplasms/therapy , Clostridium/physiology , Glioblastoma/microbiology , Glioblastoma/therapy , Injections, Intravenous/veterinary , Animals , Antineoplastic Agents/administration & dosage , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Hypoxia/physiology , Clostridium/growth & development , Clostridium/metabolism , Clostridium Infections/metabolism , Clostridium Infections/microbiology , Clostridium Infections/pathology , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Rats , Rats, Inbred F344 , Rats, Nude , Spores, Bacterial , Xenograft Model Antitumor Assays
17.
Neuro Oncol ; 17(4): 545-54, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25253417

ABSTRACT

BACKGROUND: Medulloblastoma is the most common malignant brain tumor in children. Current standard treatments cure 40%-60% of patients, while the majority of survivors suffer long-term neurological sequelae. The identification of 4 molecular groups of medulloblastoma improved the clinical management with the development of targeted therapies; however, the tumor acquires resistance quickly. Mebendazole (MBZ) has a long safety record as antiparasitic in children and has been recently implicated in inhibition of various tyrosine kinases in vitro. Here, we investigated the efficacy of MBZ in various medulloblastoma subtypes and MBZ's impact on vascular endothelial growth factor receptor 2 (VEGFR2) and tumor angiogenesis. METHODS: The inhibition of MBZ on VEGFR2 kinase was investigated in an autophosphorylation assay and a cell-free kinase assay. Mice bearing orthotopic PTCH1-mutant medulloblastoma allografts, a group 3 medulloblastoma xenograft, and a PTCH1-mutant medulloblastoma with acquired resistance to the smoothened inhibitor vismodegib were treated with MBZ. The survival benefit and the impact on tumor angiogenesis and VEGFR2 kinase function were analyzed. RESULTS: We determined that MBZ interferes with VEGFR2 kinase by competing with ATP. MBZ selectively inhibited tumor angiogenesis but not the normal brain vasculatures in orthotopic medulloblastoma models and suppressed VEGFR2 kinase in vivo. MBZ significantly extended the survival of medulloblastoma models derived from different molecular backgrounds. CONCLUSION: Our findings support testing of MBZ as a possible low-toxicity therapy for medulloblastomas of various molecular subtypes, including tumors with acquired vismodegib resistance. Its antitumor mechanism may be partially explained by inhibition of tumor angiogenesis.


Subject(s)
Antineoplastic Agents/therapeutic use , Cerebellar Neoplasms/drug therapy , Mebendazole/therapeutic use , Medulloblastoma/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Cell Line, Tumor , Cerebellar Neoplasms/enzymology , Female , Humans , Medulloblastoma/enzymology , Mice , Mice, Nude , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Mol Cancer Ther ; 14(1): 3-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25376612

ABSTRACT

The hedgehog (Hh) signaling pathway is activated in many types of cancer and therefore presents an attractive target for new anticancer agents. Here, we show that mebendazole, a benzamidazole with a long history of safe use against nematode infestations and hydatid disease, potently inhibited Hh signaling and slowed the growth of Hh-driven human medulloblastoma cells at clinically attainable concentrations. As an antiparasitic, mebendazole avidly binds nematode tubulin and causes inhibition of intestinal microtubule synthesis. In human cells, mebendazole suppressed the formation of the primary cilium, a microtubule-based organelle that functions as a signaling hub for Hh pathway activation. The inhibition of Hh signaling by mebendazole was unaffected by mutants in the gene that encodes human Smoothened (SMO), which are selectively propagated in cell clones that survive treatment with the Hh inhibitor vismodegib. Combination of vismodegib and mebendazole resulted in additive Hh signaling inhibition. Because mebendazole can be safely administered to adults and children at high doses over extended time periods, we propose that mebendazole could be rapidly repurposed and clinically tested as a prospective therapeutic agent for many tumors that are dependent on Hh signaling.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Repositioning/methods , Hedgehog Proteins/antagonists & inhibitors , Mebendazole/administration & dosage , Medulloblastoma/drug therapy , Anilides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , HEK293 Cells , Humans , Mebendazole/pharmacology , Medulloblastoma/genetics , Mice , Mice, Nude , Mutation , NIH 3T3 Cells , Pyridines/pharmacology , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Smoothened Receptor , Xenograft Model Antitumor Assays
19.
Sci Transl Med ; 6(249): 249ra111, 2014 Aug 13.
Article in English | MEDLINE | ID: mdl-25122639

ABSTRACT

Species of Clostridium bacteria are notable for their ability to lyse tumor cells growing in hypoxic environments. We show that an attenuated strain of Clostridium novyi (C. novyi-NT) induces a microscopically precise, tumor-localized response in a rat orthotopic brain tumor model after intratumoral injection. It is well known, however, that experimental models often do not reliably predict the responses of human patients to therapeutic agents. We therefore used naturally occurring canine tumors as a translational bridge to human trials. Canine tumors are more like those of humans because they occur in animals with heterogeneous genetic backgrounds, are of host origin, and are due to spontaneous rather than engineered mutations. We found that intratumoral injection of C. novyi-NT spores was well tolerated in companion dogs bearing spontaneous solid tumors, with the most common toxicities being the expected symptoms associated with bacterial infections. Objective responses were observed in 6 of 16 dogs (37.5%), with three complete and three partial responses. On the basis of these encouraging results, we treated a human patient who had an advanced leiomyosarcoma with an intratumoral injection of C. novyi-NT spores. This treatment reduced the tumor within and surrounding the bone. Together, these results show that C. novyi-NT can precisely eradicate neoplastic tissues and suggest that further clinical trials of this agent in selected patients are warranted.


Subject(s)
Clostridium/physiology , Injections, Intralesional , Neoplasms/microbiology , Neoplasms/therapy , Animals , Dogs , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Necrosis , Neoplasms/diagnostic imaging , Neoplasms/pathology , Rats , Reproducibility of Results , Sarcoma/diagnostic imaging , Sarcoma/pathology , Sarcoma/therapy , Spores, Bacterial , Tomography, X-Ray Computed , Treatment Outcome
20.
PLoS One ; 8(10): e75945, 2013.
Article in English | MEDLINE | ID: mdl-24146797

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary malignant adult brain tumor and is associated with poor survival. Recently, stem-like cell populations have been identified in numerous malignancies including GBM. To identify genes whose expression is changed with differentiation, we compared transcript profiles from a GBM oncosphere line before and after differentiation. Bioinformatic analysis of the gene expression profiles identified podocalyxin-like protein (PODXL), a protein highly expressed in human embryonic stem cells, as a potential marker of undifferentiated GBM stem-like cells. The loss of PODXL expression upon differentiation of GBM stem-like cell lines was confirmed by quantitative real-time PCR and flow cytometry. Analytical flow cytometry of numerous GBM oncosphere lines demonstrated PODXL expression in all lines examined. Knockdown studies and flow cytometric cell sorting experiments demonstrated that PODXL is involved in GBM stem-like cell proliferation and oncosphere formation. Compared to PODXL-negative cells, PODXL-positive cells had increased expression of the progenitor/stem cell markers Musashi1, SOX2, and BMI1. Finally, PODXL expression directly correlated with increasing glioma grade and was a marker for poor outcome in patients with GBM. In summary, we have demonstrated that PODXL is expressed in GBM stem-like cells and is involved in cell proliferation and oncosphere formation. Moreover, high PODXL expression correlates with increasing glioma grade and decreased overall survival in patients with GBM.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , Sialoglycoproteins/genetics , Adult , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Neoplasm Grading , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Prognosis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Sialoglycoproteins/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...