Abstract INTRODUCTION Little progress has been made in the development of effective new therapies for glioblastoma (GBM) the past decades. Fresh patient-derived GBM cell culture models have become the gold standard for GBM drug discovery and development. One of the major obstacles in identifying novel candidate drugs against GBM remains the blood-brain barrier (BBB). Therefore, it is crucial to select drugs with favourable physicochemical properties to cross BBB and reach the tumour tissue in therapeutically effective concentrations. In current drug repurposing approach, we evaluated available anti-cancer agents in our patient-derived drug screening platform against GBM. METHODS The FDA-approved Oncology Drug Set II library was tested on 45 primary GBM cell cultures. We developed a drug shortlisting pipeline combining efficacy data with pharmacodynamic and pharmacokinetic characteristics of each compound. The therapeutic efficacy of the selected agent was assessed in an orthotopic mouse PDX model, while penetration into the CNS by LC/MS/MS. RESULTS Omacetaxine mepesuccinate (OMA) was ranked as one of the most promising candidates applying our drug selection approach. In vitro, OMA revealed anti-tumour activity at IC50 values well-below reported Cmax plasma values in approximately 80% of GBM cultures. NanoString nCounter analysis, revealed DNA damage repair as the main pathway involved in OMA’s anti-tumour effect. Activation of caspase 3/7 activity and decrease of glioma cell invasiveness were also linked to its anti-tumour effect. In vivo, 1mg/kg dose of OMA was found to reach the brain tumour tissue in concentrations similar to the reported IC50 values in vitro. No adverse reactions were noted and a survival benefit was observed in a proportion of the treated mice. CONCLUSIONS At 1 mg/kg, OMA reaches the tumour brain tissue in therapeutically effective concentrations in mice while a moderate therapeutic benefit was observed. Additional in vivo experiments are ongoing investigating higher dosages of OMA and longer exposure.
Abstract Background Patient-derived glioma stem-like cells (GSCs) have become the gold-standard in neuro-oncological research; however, it remains to be established whether loss of in situ microenvironment affects the clinically-predictive value of this model. We implemented a GSC monolayer system to investigate in situ - in vitro molecular correspondence and the relationship between in vitro and patient response to temozolomide (TMZ). Methods DNA/RNA-sequencing was performed on 56 glioblastoma tissues and 19 derived GSC cultures. Sensitivity to TMZ was screened across 66 GSC cultures. Viability readouts were related to clinical parameters of corresponding patients and whole-transcriptome data. Results Tumour DNA and RNA sequences revealed strong similarity to corresponding GSCs despite loss of neuronal and immune interactions. In vitro TMZ screening yielded three response categories which significantly correlated with patient survival, therewith providing more specific prediction than the binary MGMT marker. Transcriptome analysis identified 121 genes related to TMZ sensitivity of which 21were validated in external datasets. Conclusion GSCs retain patient-unique hallmark gene expressions despite loss of their natural environment. Drug screening using GSCs predicted patient response to TMZ more specifically than MGMT status, while transcriptome analysis identified potential biomarkers for this response. GSC drug screening therefore provides a tool to improve drug development and precision medicine for glioblastoma.
Abstract BACKGROUND Little progress has been made in the development of effective new therapies for glioblastoma (GBM) the past decades. One of the major obstacles in identifying novel candidate drugs against GBM remains the blood-brain barrier (BBB) and the efflux transporters. Therefore, it is crucial to select drugs able to cross BBB and reach the tumor tissue in therapeutically effective concentrations. In current study, we evaluated the anti-glioma effect of the protein synthesis inhibitor omacetaxine mepesuccinate (OMA) in vitro and its CNS penetrating properties in vivo. MATERIAL AND METHODS The FDA-approved Oncology Drug Set II library was tested on 55 patient-derived GBM cell cultures. We designed a drug shortlisting pipeline combining efficacy data with pharmacodynamic and pharmacokinetic characteristics of each compound. We developed and characterized 3 orthotopic mouse PDX models in terms of brain vascular integrity, BBB permeability and expression of ABC transporters. The PK profile of omacetaxine was assessed in these orthotopic mouse PDX models. RESULTS In vitro, OMA revealed anti-tumor activity at IC50 values well-below reported Cmax plasma value (Cmax=43nM) in approximately 80% of GBM cultures. Unexpectedly, OMA does not downregulate the expression of its acknowledged target, ribosomal protein 3 (RPL3). In glioma cells while activation of caspase 3/7 activity is linked to its anti-tumor effect. OMA is a substrate for ABCB1 (P-glycoprotein) but not for ABCG2 (BCRP). In vivo, dose escalation showed safety of the drug and OMA was found to reach the brain tumor tissue in concentrations similar or higher to the reported IC50 values in vitro, depending on the PDX model used. No adverse reactions were noted. CONCLUSION OMA is a CNS penetrant drug that reaches the brain tumor tissue in all tested concentrations. The inter-model variation in accumulation of OMA is associated to the extent of BBB disruption of each PDX model. Additional in vivo experiments are ongoing to investigate the therapeutic effect of OMA and longer exposure using in vivo models.
Chemotherapy using temozolomide is the standard treatment for patients with glioblastoma. Despite treatment, prognosis is still poor largely due to the emergence of temozolomide resistance. This resistance is closely linked to the widely recognized inter- and intra-tumoral heterogeneity in glioblastoma, although the underlying mechanisms are not yet fully understood. To induce temozolomide resistance, we subjected 21 patient-derived glioblastoma cell cultures to Temozolomide treatment for a period of up to 90 days. Prior to treatment, the cells' molecular characteristics were analyzed using bulk RNA sequencing. Additionally, we performed single-cell RNA sequencing on four of the cell cultures to track the evolution of temozolomide resistance. The induced temozolomide resistance was associated with two distinct phenotypic behaviors, classified as "adaptive" (ADA) or "non-adaptive" (N-ADA) to temozolomide. The ADA phenotype displayed neurodevelopmental and metabolic gene signatures, whereas the N-ADA phenotype expressed genes related to cell cycle regulation, DNA repair, and protein synthesis. Single-cell RNA sequencing revealed that in ADA cell cultures, one or more subpopulations emerged as dominant in the resistant samples, whereas N-ADA cell cultures remained relatively stable. The adaptability and heterogeneity of glioblastoma cells play pivotal roles in temozolomide treatment and contribute to the tumor's ability to survive. Depending on the tumor's adaptability potential, subpopulations with acquired resistance mechanisms may arise.
Abstract BACKGROUND The development of new therapeutic agents generally takes many years to translate to clinically-effective new treatments. Among the strategies to reduce this time frame, efforts are now being undertaken to investigate drug repurposing. With this approach, compounds available for a specific disease are evaluated for their therapeutic efficacy in other diseases. We have set up a patient-derived cell culture model to apply this strategy for glioblastoma (GBM). MATERIAL AND METHODS Fresh patient-derived tumour tissue was dissociated and cultured in serum-free medium supplemented with EGF and bFGF. MGMT status was determined by methylation-specific PCR.Drug screening was performed using the NIH anti-cancer collection containing 114 approved oncology drugs. Compounds include chemotherapeutic agents as well as small molecule targeted agents. Readout for drug effects is based on ATP-based viability assay. Using systems modelling approaches, integrated analysis of both mutational and expression data of each tumour is applied to identify key pathways involved in response to specific compounds. RESULTS Molecular analysis demonstrated that copy number variations are preserved under serum-free culture conditions and that the MGMT methylation status is retained in over 75% of cases. Screening of the NIH anti-cancer collection on 55 GBM cell cultures revealed high intertumoral variation in response to most drugs. This included subsets of GBM revealing exceptionally high sensitivity to specific agents at clinically-feasible concentrations. Further ranking of the compounds was made based on the therapeutic index (IC50 tumour versus normal human astrocytes) and predicted blood-brain-barrier crossing capability. Currently, integrated analysis of molecular profiles of the tumour in relation to the drug response data is ongoing with the aim of identifying response predictors to these clinically-approved anti-cancer agents. CONCLUSION Our patient-derived in vitro drug screening assay may offer a tool to identify available anti-cancer agents that are effective in a subpopulation of GBM patients and that may be implemented in future stratified clinical trials for this patient group.
Abstract Background Radiation therapy and chemotherapy using Temozolomide are the standard adjuvant treatments for patients with glioblastoma. Despite maximal treatment prognosis is still poor largely due to the emergence of Temozolomide resistance. This resistance is closely linked to the widely recognized inter- and intra-tumoral heterogeneity in glioblastoma, although the underlying mechanisms are not yet fully understood. This study aims to investigate the diverse molecular mechanisms involved in temozolomide resistance. Methods To induce temozolomide resistance, we subjected 21 patient-derived glioblastoma cell cultures to Temozolomide treatment for a period of up to 90 days. Prior to treatment, the cells’ molecular characteristics were analyzed using bulk RNA sequencing. Additionally, we performed single-cell sequencing on four of the cell cultures to track the evolution of temozolomide resistance. Results The induced temozolomide resistance was associated with two distinct phenotypic behaviors, classified as “adaptive” (ADA) or “non-adaptive” (N-ADA) to temozolomide. The ADA phenotype displayed neurodevelopmental and metabolic gene signatures, whereas the N-ADA phenotype expressed genes related to cell cycle regulation, DNA repair, and protein synthesis. Single-cell RNA sequencing revealed that in ADA cell cultures, one or more subpopulations emerged as dominant in the resistant samples, whereas N-ADA cell cultures remained relatively stable. Conclusions The adaptability and heterogeneity of glioblastoma cells play pivotal roles in temozolomide treatment and contribute to the tumor’s ability to survive. Depending on the tumor’s adaptability potential, subpopulations with acquired resistance mechanisms may arise. Further research is necessary to deepen our understanding of these mechanisms and develop strategies to overcome them.
Abstract BACKGROUND Patient-derived glioma stem-like cells (GSCs) have become the gold-standard in neuro-oncological research; however, it remains to be established whether loss of in situ microenvironment affects the clinically-predictive value of this model. We implemented a GSC monolayer system to investigate in situ-in vitro molecular correspondence and the relationship between in vitro and patient response to temozolomide (TMZ). METHODS DNA and RNA-sequencing was performed on 56 glioblastoma tissues and 19 derived GSC cultures. Sensitivity to TMZ was screened across 66 GSC cultures. Drug response readouts were related to clinical parameters of corresponding patients and whole-transcriptome data. RESULTS Tumor DNA and RNA sequences revealed strong similarity to corresponding GSCs despite loss of neuronal and immune interactions. Patient-specific hallmark gene expression levels were retained in vitro. Functional screening for TMZ sensitivity yielded three response categories (responders, intermediates and non-responders) which significantly correlated with patient survival, therewith providing a more specific response prediction than the binary MGMT marker. Transcriptome analysis of GSCs and parental tissues identified 121 genes related to TMZ sensitivity and overall survival, of which 21, including MGMT, could be validated in external datasets. CONCLUSION GSCs retain patient-unique hallmark gene expressions despite loss of their natural environment. Drug screening using GSCs predicted patient response to TMZ more specifically than MGMT status, while transcriptome analysis identified potential biomarkers for this response. GSC drug screening therefore provides a tool to improve drug development and precision medicine for glioblastoma patients.
Abstract BACKGROUND Major obstacles that have impeded the development of effective new therapies for GBM include inter- and intratumoral heterogeneity, the blood-brain-barrier and use of sub-optimal cell line-based preclinical models. Taking these hurdles into account, we have set up a patient-derived GBM drug-screening platform. Molecular similarities and dissimilarities between tumors and derived cultures were investigated and the predictive power of our assay for patient response to TMZ was assessed. A large panel of GBM cells was screened for sensitivity to available oncological agents with the aim of deriving a set of drugs with favorable physicochemical properties for BBB crossing and potent activity in (a subset of) GBM cultures. Finally, we determined the success rate of performing a small-scale screen with 20 selected agents within 4 weeks post-surgery. MATERIAL AND METHODS Tissue samples as well as ultrasonic surgical aspirates (USA) were received from GBM surgeries and utilized to establish glioma stem cell (GSC) cultures. Correspondence of GSCs to parental tumors was assessed by DNA/RNA-sequencing. In vitro sensitivity to temozolomide was screened across 55 GSC cultures. Cell viability readouts were related to clinical parameters of corresponding patients. FDA-approved anticancer agents (n=107) were screened on 45 GBM cultures by dose-response testing and viability was assessed using Cell Titer-Glo. Twenty selected agents were screened on 24 fresh tumor samples within 4 weeks of receiving the tissue. RESULTS By combining both USA and tissue piece-derived dissociation protocols, culture success increased to 95%, ensuring representation of the near-complete spectrum of GBM subtypes. Tumor DNA and RNA sequences revealed strong similarities to corresponding GSC cultures although differences related to neuronal signaling, immune interactions, and cell cycle were observed. Tumor expression profiles of hallmark genes of GBM were faithfully retained in derived cell cultures. In vitro screening of TMZ on a large cohort identified 3 response categories (responders/intermediates/non-responders) for which Cox regression analysis revealed significantly different overall survival curves of corresponding patients. Screening of approved anticancer agents on 45 GBM cultures underscored the tremendous intertumoral heterogeneity in drug sensitivities. We identified 20 potent agents each effective at clinically-achievable concentrations in (a subset of) GBM cultures with favorable BBB penetration properties (CNS-MPO score). Screening of these agents on a per patient basis within 4 weeks of receiving tissue was successful in 18 out of 24 (75%) tested tumors. CONCLUSION Patient-derived functional drug screening offers a tool to predict TMZ response in GBM patients and assess tumor sensitivity to candidate treatments, either for GBM subsets or on a per patient basis.
RNA-seq data of glioma samples and their matched primary cultures. Cultures were exposed to the IDH mutant-specific inhibitor AGI-5198 or DMSO control.
RNA-seq data of glioma samples and their matched primary cultures. Cultures were exposed to the IDH mutant-specific inhibitor AGI-5198 or DMSO control.