Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Eur J Cancer ; 140: 130-139, 2020 11.
Article in English | MEDLINE | ID: mdl-33091717

ABSTRACT

BACKGROUND: The protease inhibitor ritonavir (RTV) is a clinical-stage inhibitor of the human immunodeficiency virus. In a drug repositioning approach, we here exhibit the additional potential of RTV to augment current treatment of glioblastoma, the most aggressive primary brain tumour of adulthood. METHODS: We explored the antitumour activity of RTV and mechanisms of action in a broad spectrum of short-term expanded clinical cell samples from primary and recurrent glioblastoma and in a cohort of conventional cell lines and non-tumour human neural controls in vitro. To validate RTV efficacy in monotherapeutic and in combinatorial settings, we used patient-derived xenograft models in a series of in vivo studies. RESULTS: RTV monotherapy induced a selective antineoplastic response and demonstrated cytostatic and anti-migratory activity at clinical plasma peak levels. Additional exposure to temozolomide or irradiation further enhanced the effects synergistically, fostered by mechanisms of autophagy and increased endoplasmic reticulum stress. In xenograft models, we consequently observed increasing overall survival under the combinatorial effect of RTV and temozolomide. CONCLUSIONS: Our data establish RTV as a valuable repositioning candidate for further exploration as an adjunct therapeutic in the clinical care of glioblastoma.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Glioblastoma/drug therapy , Ritonavir/therapeutic use , Adult , Autophagy/drug effects , Cell Line , Drug Repositioning/methods , Drug Therapy, Combination/methods , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Male , Neoplasm Recurrence, Local/drug therapy , Temozolomide/therapeutic use
2.
J Transl Med ; 17(1): 96, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894200

ABSTRACT

BACKGROUND: Molecularly targeted therapies using receptor inhibitors, small molecules or monoclonal antibodies are routinely applied in oncology. Verification of target expression should be mandatory prior to initiation of therapy, yet, determining the expression status is most challenging in recurrent glioblastoma (GBM) where most patients are not eligible for second-line surgery. Because very little is known on the consistency of expression along the clinical course we here explored common drug targets in paired primary vs. recurrent GBM tissue samples. METHODS: Paired surgical tissue samples were derived from a homogeneously treated cohort of 34 GBM patients. All patients received radiotherapy and temozolomide chemotherapy. Verification of common drug targets included immunohistological analysis of PDGFR-ß, FGFR-2, FGFR-3, and mTOR-pathway component (phospho-mTORSer2448) as well as molecular, MLPA-based analysis of specific copy number aberrations at the gene loci of ALK, PDGFRA, VEGFR2/KDR, EGFR, MET, and FGFR1. RESULTS: Paired tumor tissue exhibited significant changes of expression in 9 of the 10 investigated druggable targets (90%). Only one target (FGFR1) was found "unchanged", since dissimilar expression was observed in only one of the 34 paired tumor tissue samples. All other targets were variably expressed with an 18-56% discordance rate between primary and recurrent tissue. CONCLUSIONS: The high incidence of dissimilar target expression status in clinical samples from primary vs. recurrent GBM suggests clinically relevant heterogeneity along the course of disease. Molecular target expression, as determined at primary diagnosis, may not necessarily present rational treatment clues for the clinical care of recurrent GBM. Further studies need to analyze the therapeutic impact of longitudinal heterogeneity in GBM.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/therapy , Genetic Heterogeneity , Glioblastoma/therapy , Molecular Targeted Therapy/methods , Neoplasm Recurrence, Local/therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Disease Progression , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/physiology , Female , Glioblastoma/diagnosis , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Longitudinal Studies , Male , Middle Aged , Molecular Targeted Therapy/trends , Neoplasm Recurrence, Local/genetics , Patient Care Planning , Retrospective Studies , Young Adult
3.
Clin Cancer Res ; 23(2): 562-574, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27521447

ABSTRACT

PURPOSE: Investigation of clonal heterogeneity may be key to understanding mechanisms of therapeutic failure in human cancer. However, little is known on the consequences of therapeutic intervention on the clonal composition of solid tumors. EXPERIMENTAL DESIGN: Here, we used 33 single cell-derived subclones generated from five clinical glioblastoma specimens for exploring intra- and interindividual spectra of drug resistance profiles in vitro In a personalized setting, we explored whether differences in pharmacologic sensitivity among subclones could be employed to predict drug-dependent changes to the clonal composition of tumors. RESULTS: Subclones from individual tumors exhibited a remarkable heterogeneity of drug resistance to a library of potential antiglioblastoma compounds. A more comprehensive intratumoral analysis revealed that stable genetic and phenotypic characteristics of coexisting subclones could be correlated with distinct drug sensitivity profiles. The data obtained from differential drug response analysis could be employed to predict clonal population shifts within the naïve parental tumor in vitro and in orthotopic xenografts. Furthermore, the value of pharmacologic profiles could be shown for establishing rational strategies for individualized secondary lines of treatment. CONCLUSIONS: Our data provide a previously unrecognized strategy for revealing functional consequences of intratumor heterogeneity by enabling predictive modeling of treatment-related subclone dynamics in human glioblastoma. Clin Cancer Res; 23(2); 562-74. ©2016 AACR.


Subject(s)
Drug Combinations , Drug Resistance, Neoplasm/genetics , Genetic Heterogeneity , Glioblastoma/drug therapy , Animals , Clonal Evolution/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Xenograft Model Antitumor Assays
4.
J Cancer Res Clin Oncol ; 142(7): 1581-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27100354

ABSTRACT

PURPOSE: Dovitinib (TKI258) is an oral multi-tyrosine kinase inhibitor of FGFR, VEGFR, PDGFR ß, and c-Kit. Since dovitinib is able to cross the blood-brain barrier and targets brain tumor-relevant pathways, we conducted a phase I trial to demonstrate its safety in recurrent glioblastoma (GBM). PATIENTS AND METHODS: Patients with first or second GBM recurrence started treatment with the maximal tolerated dose (MTD) previously established in systemic cancer patients (500 mg/d, 5 days on/2 days off). A modified 3 + 3 design in three cohorts (500, 400, 300 mg) was used. RESULTS: Twelve patients were enrolled. Seventy-two adverse events (AEs) occurred and 16.7 % of AEs were classified as ≥CTC grade 3 toxicity, mainly including hepatotoxicity and hematotoxicity. Only one out of six patients of the 300-mg cohort showed grade 3 toxicity. The PFS-6 rate was 16.7 %, and it was not associated with detection of the FGFR-TACC gene fusion in the tumor. CONCLUSION: Dovitinib is safe in patients with recurrent GBM and showed efficacy in only some patients unselected for target expression. The recommended phase II dose of 300 mg would be substantially lower than the recently established MTD in systemic cancer patients. Further personalized trials are recommended.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Quinolones/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Biomarkers, Tumor/metabolism , Cohort Studies , Female , Humans , Male , Microtubule-Associated Proteins/metabolism , Middle Aged , Quinolones/administration & dosage , Quinolones/adverse effects , Receptor, Fibroblast Growth Factor, Type 3/metabolism
5.
Clin Cancer Res ; 19(15): 4124-36, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23908450

ABSTRACT

PURPOSE: Glioblastoma is a highly malignant, invariably fatal brain tumor for which effective pharmacotherapy remains an unmet medical need. EXPERIMENTAL DESIGN: Screening of a compound library of 160 synthetic and natural toxic substances identified the antihelmintic niclosamide as a previously unrecognized candidate for clinical development. Considering the cellular and interindividual heterogeneity of glioblastoma, a portfolio of short-term expanded primary human glioblastoma cells (pGBM; n = 21), common glioma lines (n = 5), and noncancer human control cells (n = 3) was applied as a discovery platform and for preclinical validation. Pharmacodynamic analysis, study of cell-cycle progression, apoptosis, cell migration, proliferation, and on the frequency of multipotent/self-renewing pGBM cells were conducted in vitro, and orthotopic xenotransplantation was used to confirm anticancer effects in vivo. RESULTS: Niclosamide led to cytostatic, cytotoxic, and antimigratory effects, strongly reduced the frequencies of multipotent/self-renewing cells in vitro, and after exposure significantly diminished the pGBMs' malignant potential in vivo. Mechanism of action analysis revealed that niclosamide simultaneously inhibited intracellular WNT/CTNNB1-, NOTCH-, mTOR-, and NF-κB signaling cascades. Furthermore, combinatorial drug testing established that a heterozygous deletion of the NFKBIA locus in glioblastoma samples could serve as a genomic biomarker for predicting a synergistic activity of niclosamide with temozolomide, the current standard in glioblastoma therapy. CONCLUSIONS: Together, our data advocate the use of pGBMs for exploration of compound libraries to reveal unexpected leads, for example, niclosamide that might be suited for further development toward personalized clinical application.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioblastoma/genetics , Niclosamide/administration & dosage , Animals , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioblastoma/pathology , Humans , Mice , NF-kappa B/metabolism , Receptors, Notch/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
6.
Stem Cells ; 31(6): 1064-74, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23390110

ABSTRACT

Cellular heterogeneity, for example, the intratumoral coexistence of cancer cells with and without stem cell characteristics, represents a potential root of therapeutic resistance and a significant challenge for modern drug development in glioblastoma (GBM). We propose here that activation of the innate immune system by stimulation of innate immune receptors involved in antiviral and antitumor responses can similarly target different malignant populations of glioma cells. We used short-term expanded patient-specific primary human GBM cells to study the stimulation of the cytosolic nucleic acid receptors melanoma differentiation-associated gene 5 (MDA5) and retinoic acid-inducible gene I (RIG-I). Specifically, we analyzed cells from the tumor core versus "residual GBM cells" derived from the tumor resection margin as well as stem cell-enriched primary cultures versus specimens without stem cell properties. A portfolio of human, nontumor neural cells was used as a control for these studies. The expression of RIG-I and MDA5 could be induced in all of these cells. Receptor stimulation with their respective ligands, p(I:C) and 3pRNA, led to in vitro evidence for an effective activation of the innate immune system. Most intriguingly, all investigated cancer cell populations additionally responded with a pronounced induction of apoptotic signaling cascades revealing a second, direct mechanism of antitumor activity. By contrast, p(I:C) and 3pRNA induced only little toxicity in human nonmalignant neural cells. Granted that the challenge of effective central nervous system (CNS) delivery can be overcome, targeting of RIG-I and MDA5 could thus become a quintessential strategy to encounter heterogeneous cancers in the sophisticated environments of the brain.


Subject(s)
Antineoplastic Agents/pharmacology , Cytosol/immunology , DEAD-box RNA Helicases/immunology , Glioblastoma/drug therapy , Glioblastoma/immunology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon-Induced Helicase, IFIH1 , Ligands , Receptors, Immunologic , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/immunology , Stem Cells/metabolism
7.
Ann Neurol ; 68(2): 264-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20695020

ABSTRACT

Residual tumor cells remain beyond the margins of every glioblastoma (GBM) resection. Their resistance to postsurgical therapy is considered a major driving force of mortality, but their biology remains largely uncharacterized. In this study, residual tumor cells were derived via experimental biopsy of the resection margin after standard neurosurgery for direct comparison with samples from the routinely resected tumor tissue. In vitro analysis of proliferation, invasion, stem cell qualities, GBM-typical antigens, genotypes, and in vitro drug and irradiation challenge studies revealed these cells as unique entities. Our findings suggest a need for characterization of residual tumor cells to optimize diagnosis and treatment of GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Adult , Aged , Antigens, Neoplasm/metabolism , Brain Neoplasms/drug therapy , Cell Culture Techniques , Cell Proliferation , Cell Separation , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured
8.
Nat Genet ; 40(11): 1279-81, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18849994

ABSTRACT

We carried out a genome-wide association study in 296 individuals with male-pattern baldness (androgenetic alopecia) and 347 controls. We then investigated the 30 best SNPs in an independent replication sample and found highly significant association for five SNPs on chromosome 20p11 (rs2180439 combined P = 2.7 x 10(-15)). No interaction was detected with the X-chromosomal androgen receptor locus, suggesting that the 20p11 locus has a role in a yet-to-be-identified androgen-independent pathway.


Subject(s)
Alopecia/genetics , Chromosomes, Human, Pair 20/genetics , Genetic Predisposition to Disease , Adult , Female , Genome, Human/genetics , Germany , Humans , Male , Polymorphism, Single Nucleotide/genetics
9.
Am J Hum Genet ; 82(3): 737-43, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18304493

ABSTRACT

Androgenetic alopecia (AGA, male pattern baldness) is the most common form of hair loss. The origin of AGA is genetic, with the X chromosome located androgen receptor gene (AR) being the only risk gene identified to date. We present the results of a genome-wide linkage study of 95 families and linkage fine mapping of the 3q21-q29, 11q14-q25, 18p11-q23, and 19p13-q13 regions in an extended sample of 125 families of German descent. The locus with strongest evidence for linkage was mapped to 3q26 with a nonparametric linkage (NPL) score of 3.97 (empirical p value = 0.00055). This is the first step toward the identification of new susceptibility genes in AGA, a process which will provide important insights into the molecular and cellular basis of scalp hair loss.


Subject(s)
Alopecia/genetics , Chromosomes, Human, Pair 3/genetics , Genetic Linkage , Genetic Predisposition to Disease , Adult , Age of Onset , Chromosome Mapping , Family , Genetic Testing , Genome, Human/genetics , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...