Skip to main content
Neuro-Oncology Advances logoLink to Neuro-Oncology Advances
. 2020 Oct 24;3(1):vdaa145. doi: 10.1093/noajnl/vdaa145

Adult precision medicine: learning from the past to enhance the future

Ashley Ghiaseddin 1,, Lan B Hoang Minh 1, Michalina Janiszewska 2, David Shin 1, Wolfgang Wick 3, Duane A Mitchell 1, Patrick Y Wen 4,5,6, Stuart A Grossman 7
PMCID: PMC7846182  PMID: 33543142

Abstract

Despite therapeutic advances for other malignancies, gliomas remain challenging solid tumors to treat. Complete surgical resection is nearly impossible due to gliomas’ diffuse infiltrative nature, and treatment is hampered by restricted access to the tumors due to limited transport across the blood–brain barrier. Recent advances in genomic studies and next-generation sequencing techniques have led to a better understanding of gliomas and identification of potential aberrant signaling pathways. Targeting the specific genomic abnormalities via novel molecular therapies has opened a new avenue in the management of gliomas, with encouraging results in preclinical studies and early clinical trials. However, molecular characterization of gliomas revealed significant heterogeneity, which poses a challenge for targeted therapeutic approaches. In this context, leading neuro-oncology researchers and clinicians, industry innovators, and patient advocates convened at the inaugural annual Remission Summit held in Orlando, FL in February 2019 to discuss the latest advances in immunotherapy and precision medicine approaches for the treatment of adult and pediatric brain tumors and outline the unanswered questions, challenges, and opportunities that lay ahead for advancing the duration and quality of life for patients with brain tumors. Here, we provide historical context for precision medicine in other cancers, present emerging approaches for gliomas, discuss their limitations, and outline the steps necessary for future success. We focus on the advances in small molecule targeted therapy, as the use of immunotherapy as an emerging precision medicine modality for glioma treatment has recently been reviewed by our colleagues.

Keywords: blood–brain barrier (BBB), glioma, next-generation sequencing (NGS), precision medicine, tumor heterogeneity


Key Points.

  • For precision medicine to be successful in gliomas, targets should be ‘drivers’ and drugs must reach targets in therapeutic concentrations.

  • Future clinical trial design should include molecular classification, highlight certain biomarkers and promote Bayesian adaptive randomization designs.

Definition

Precision medicine for adult brain tumors is an innovative treatment approach tailored to the genetic profile of both the patient and cancer.1 Precision medicine approaches for adult brain tumors have yielded some success, specifically in the targeting of BRAF, H3K27 demethylation, and NTRK fusions.2,3 However, spatial and temporal heterogeneity is an important challenge that the brain cancer field must address before precision medicine can be considered a viable option for brain tumor patients. Spatial heterogeneity has elegantly been described in a published transcriptional atlas,4 where genomic alterations and gene expression patterns are shown to differ between from the leading edge, infiltrating tumor, cellular tumor, pseudopalisading cells around necrosis, and microvascular proliferation regions of glioblastoma (GBM) resected tumors.4 Temporal heterogeneity, which describes the changes in gene expression patterns occurring between the time of diagnosis, treatment, and ultimately tumor recurrence, was highlighted when a review of phase III clinical trial (ACT-IV) investigating rindopepimut with temozolomide for patients with newly diagnosed, epidermal growth factor receptor variant III (EGFRvIII)-expressing GBM revealed a loss of EGFRvIII expression in approximately 60% of recurrent tumor tissues over time, independently of the treatment arm.5 Other commonly used molecular targets displaying temporal heterogeneity between primary and recurrent GBM include FGFR-2, FGFR-3, ALK, PDGFRA, PDGFRB, VEGFR2/KDR, EGFR, and MET.6 In GBM isocitrate dehydrogenase (IDH) wild-type tumors, molecular events appear stable in nearly 80% of occurrences, whereas changes to mutational status do frequently occur (nearly 90% of TERT and 60% of EGFR mutations).7 It is important that the research and data generated from using precision medicine in the treatment of other cancers be leveraged in the application of precision medicine for adult brain tumors.

Successes in Other Cancers

The success of precision medicine is evident in many cancer types, with therapies targeting ERBB2 (HER2) amplification in breast cancer,8,9 breakpoint cluster region (BCR)-ABL fusion gene in chronic myelogenous leukemia,10,11 epidermal growth factor receptor (EGFR) mutations in non-small-cell lung cancer (NSCLC),12,13 anaplastic lymphoma kinase (ALK) fusions in non-small-cell lung carcinoma,14,15 and proto-oncogene B-Raf (BRAF) mutations in melanoma.16–18 Identification of these specific genetic alterations have led to the development of standardized treatment plans focused on targeting the protein products of these alterations.19

The BRAF kinase inhibitor vemurafenib was shown to improve survival in patients with untreated BRAFV600E-mutated metastatic melanoma, with over a 50% response rate.20 After implementing the BRAFV600E inhibitors dabrafenib and vemurafenib into standard practice for the treatment of BRAF-mutated melanomas, further studies demonstrated improved efficacy when combining these drugs with inhibition of the downstream target mitogen-activated protein kinase (MAPK). The combinations of dabrafenib plus trametinib, vemurafenib plus cobimetinib, and encorafenib plus binimetanib have shown improved response and delay in acquired resistance in BRAF-mutated melanoma patients.16,21

Isocitrate dehydrogenase is a particularly attractive target for therapeutic intervention since IDH mutations appear to be an early event in carcinogenesis.22 IDH inhibitors have received positive attention following research in leukemia, in which approximately 10% of patients with acute myeloid leukemia (AML) harbor IDH1 mutations. Ivosidenib, an IDH1 oral inhibitor, has been shown to be well tolerated and induce durable remissions in AML patients with poor prognosis.23 Recently, both ivosidenib and the IDH2 inhibitor enasidenib have received approval from the Food and Drug Administration (FDA) for treatment of refractory IDH-mutated AML patients.24 Ivosidenib and vorasidenib, a brain-penetrant IDH1/2 inhibitor, have both been evaluated in recurrent IDH-mutated gliomas with a significant subset of grade II non-enhancing tumors showing prolonged stable disease.25 A phase III trial evaluating vorasidenib in newly diagnosed IDH-mutated grade II gliomas is currently underway (INDIGO trial).

The approval of EGFR tyrosine kinase inhibitors (TKIs) has allowed a dramatic improvement in clinical outcomes for patients with EGFR-mutated NSCLC. EGFR is a cell surface membrane receptor that controls intracellular signal transduction pathways regulating cell proliferation, apoptosis, angiogenesis, adhesion, and motility. Although first-generation (erlotinib, gefitinib) and second-generation (afatinib) TKIs have been standard of care for initial management of EGFR-mutant NSCLC, newer data suggest improved outcomes with front-line treatment with the third-generation agent, osimertinib.26 Osimertinib was approved by the FDA in 2018 as a first-line treatment for people with advanced NSCLC with specific EGFR mutations, based on earlier findings from the FLAURA trial showing a median overall survival of 38.6 months among participants in the osimertinib treatment group, compared with 31.8 months in the erlotinib/gefitinib group.27 In addition, osimertinib has been shown to cross the blood–brain barrier (BBB) with activity demonstrated against brain and leptomeningeal metastases.28–30

BRAF, IDH1, and EGFR mutations are also frequently found in gliomas. Thus, the benefits of targeting products of these genes in other cancers may provide useful insights for furthering the care of brain tumor patients. However, several limitations, specific to the biology and location of gliomas, have hampered the progress of targeted therapies in these tumors.

Inherent Limitations for Precision Medicine for Gliomas

The success of precision oncology is largely dependent on the identification of targetable biological features in the tumor. This poses a great challenge, especially for high-grade gliomas, as these tumors are inherently heterogeneous. Cells within gliomas differ in their morphology, the underlying gene expression, and genetic mutational landscape.31–34 Thus, any selected therapeutic target might be expressed by most, but unlikely by all cells within a tumor, leading to incomplete elimination of cancer cells. Genetic, epigenetic, and microenvironmental factors can all contribute to the intratumoral heterogeneity.35 In glioma, 4 major cellular subtypes (pro-neural, neural, mesenchymal, and classical) have been recently identified.32 These subpopulations are present at various proportions in different patients’ tumors; however, the relevance of this complex biology in the development of therapy resistance is not yet understood. Furthermore, no therapeutic benefit has been connected to a specific subtype. Glioma cells’ ability to shift between these different phenotypes is of particular concern, as it decreases the likelihood of therapeutic success with a single-agent targeted treatment, unless it very efficiently targets an overarching mechanism. In addition, recent studies have highlighted the temporal molecular heterogeneity leading to differential mutational profiles of the tumors at recurrence following treatment, which might be driving universal therapeutic resistance in adult patients with diffuse glioma.8,36 Therefore, understanding the fundamental biology of glioma at the molecular level is paramount and the relevance of any molecular data has to be discussed with any subsequent treatment inflicting (resistance) pressure.

As mentioned in the previous section, several potential therapeutic targets for gliomas have been successfully pursued in other tumor types. Apart from the cellular heterogeneity issue, some of these drugs have failed trials in gliomas due to insufficient drug penetration into the tumor tissue.37,38 Thus, overcoming the BBB and ensuring sufficient distribution of the drug will be required for advances in precision neuro-oncology.

Another challenge in effective targeted therapies for glioma is the difficulty of biomarker assessment. Since repeat on-treatment biopsy is not possible, it is hard to evaluate treatment effects at early timepoints, which in other tumor types is a valuable guide for tailoring the treatment to a particular tumor and patient. Thus, for gliomas, longitudinal follow-up after treatment is solely based on radiological imaging, which is not suitable for measuring the effects of treatment every few days. Many of the above-mentioned limitations could be overcome, and we discuss in detail the most significant approaches that could drive the field forward below.

Requirements for Success

Identify Targets That Are “Drivers”

CNS tumors were among the first tumor types undergoing molecular characterization by the Cancer Genome Atlas Network.33,34 Advances in next-generation sequencing (NGS) provided an extensive view of genomic alterations frequently found in these tumors.39,40 Most brain tumor-related genes play crucial roles in mitogen-activated signal transduction, cellular immortalization, or DNA repair. Highly expressed receptor tyrosine kinases (RTKs), including EGFR and platelet-derived growth factor receptor A (PDGFRA), are products of frequent amplifications in GBM.40 Despite extensive efforts to use drugs that target these molecules, most have failed to produce durable responses in clinical trials. Even if the drugs can cross the BBB in adequate concentrations, they may not be sufficiently potent to inhibit the targeted molecular pathway. Many of these genes can activate redundant signaling pathways, making drugs directed at single targets insufficient.41 Moreover, the RTKs are often amplified only in a subset of cells within a tumor, thus generating a heterogeneous mosaic of gene expression.42

The identification of mutations that are uniformly present in all cancer cells could have a higher clinical impact when considering targeted therapies for gliomas. One such case is the mutation of IDH1, which is present in up to 70% of lower-grade gliomas and most secondary GBMs.43,44 Upcoming classifications will increasingly consider molecular characteristics in the nomenclature of glioma. Round 2 of the cIMPACT classification updates has recommended re-classifying IDH-mutated GBM as astrocytoma, IDH mutant, WHO grade IV. The WHO grade IV designation for IDH-mutant astrocytoma would involve the presence of necrosis or microvascular proliferation on histology or genetic alteration with CDKN2A/B homozygous deletion.45 Mutations in the IDH1 gene generate a new catalytic activity of the resulting IDH enzyme, production of 2-hydroxyglutarate, and alterations in the metabolism and epigenetic regulation of these tumor cells.46 Moreover, these mutations are clonal, suggesting an early event in tumor evolution47 and providing an attractive opportunity for drug design. The value of targeting mutant IDH1 is controversial as the early occurrence of IDH1 mutations may be a “hit-and-run” event48 that facilitates malignant transformation but may not be essential for the survival of fully malignant cells later in tumor evolution. Studies of IDH inhibitors are ongoing in the glioma population, with the oral IDH1 inhibitor AG120 (ivosidenib) and the IDH1/2 inhibitor AG-881 (vorasidenib) showing favorable safety profiles in phase I clinical trials and a subset of patients with prolonged stable disease.25,49,50 Early clinical trials of IDH1 inhibitors in glioma have shown some effects,51 with a high percentage of patients with prolonged stable disease. These promising initial results have led to an ongoing phase III placebo-controlled trial evaluating vorasidenib in IDH-mutated grade II gliomas following initial surgery (INDIGO trial, NCT04164901). Another example of a ubiquitous mutation in cancers is a mutation of the hTERT promoter,52 which results in the upregulation of telomere lengthening protein hTERT and epigenetic reprogramming of cancer cells. As it is overexpressed in 90% of all human tumors, and increases cellular aging defense mechanisms, hTERT protein has been extensively studied as a therapeutic target in glioma.53 However, as in the case of RTKs, the presence of alternative mechanisms for telomere lengthening54 has rendered the hTERT-targeting agents tested in these trials ineffective. Nonetheless, hTERT remains a potentially promising target and the focus of significant research.

An attractive strategy for more effective treatment of brain tumors has been to test targets for which tailored therapies have been developed against other tumor types. In particular, melanoma and hematologic malignancies have been early adopters of precision medicine, with targets that are relevant to brain tumors. Case reports have suggested a treatment response after targeting BRAF-mutated gliomas with BRAF kinase inhibitors that have demonstrated efficacy in the treatment of melanoma. However, the evidence is limited due to a lack of large randomized studies as well as a limited sample size, and it is estimated that less than 3% of high-grade gliomas harbor BRAFV600E mutations.55 A recent phase II study of dabrafenib and trametinib in patients with recurrent gliomas harboring BRAFV600E mutations has shown durable responses in 29% of patients and 62% in patients with lower-grade gliomas.56

How could we identify the potential drug targets that are crucial for the survival of glioma or other CNS tumor cells? While many of the tumor-initiating genetic events and pathways leading to tumor cell proliferation/death have been identified, more research is needed to understand the networks of pathways and the interplay between genetic and epigenetic alterations that drive CNS tumor growth. Inhibiting common downstream signaling pathways, such as proteasomes using the proteasome inhibitor marizomib57 (NCT03345095) or nuclear export with Selinexor,58 could be crucial in order to overcome redundant signaling pathways and heterogeneity. In gliomas, communicating networks that render tumor cells resistant against cytotoxic therapies were identified.59 Synapses on the connections originating from the glioma cells allow tumor cells to retrieve neuronal input that fosters growth.60 Single-cell sequencing further revealed that primary brain tumors recapitulate many steps of neurodevelopment.61 Thus, exploring the intersection of cancer biology and neuroscience will open new opportunities for repurposing drugs used in neurological conditions.62 These include glutamate inhibitors, such as perampanel, which can cross the BBB, and could inhibit glioma cell proliferation. A list of selected published case reports and clinical trial results highlighting targets that are currently being tested in precision medicine for brain tumors is given in Table 1.

Table 1.

Summary of Targeted Therapies for Gliomas in Completed or Currently Enrolling Clinical Trials

Target Drug Clinical Trial
BRAF Vemurafenib NCT01524978
RAS/RAF/MEK/ERK TAK-580 NCT03429803
Dabrafenib/Trametinib NCT02034110, NCT02684058
Encorafenib/Binimetinib NCT03973918
EGFR ABT-414 NCT01800695
H3 K27M Panobinostat NCT02717455
IDH1/2 Vorasidenib (AG-881) NCT04164901
VEGF(R) Bevacizumab NCT00345163
Regorafenib NCT03970447

In addition, our current insight into new targeted therapies will be significantly broadened by the use of preclinical tumor models that better recapitulate the complexity of brain tumors. Currently employed preclinical glioma models include carcinogen (ethyl-nitrosourea)-induced gliomas in animals, in vitro glioma cell cultures derived from human or animal gliomas, patient-derived and murine glioma xenograft models (subcutaneous, orthotopic), and transgenic mouse models (eg, with conditional expression of oncogenes or loss of tumor-suppressor genes).63 However, the intratumoral heterogeneity of diffuse gliomas cannot be recapitulated to the fullest in those in vitro or in vivo models. Genetically engineered animal models lack the diversity of the tumor-driving mutations. While patient-derived xenografts retain high levels of heterogeneity, they need to be established in immunocompromised animals, which lack key subsets of immune cells that shape the tumor landscape in human gliomas. In addition, for most therapies, the BBB, which is relatively intact in large parts of diffuse glioma, is a true barrier, and modeling this barrier is a challenging task. Increased efforts in improving patient tumor-derived xenografts models64 and in vitro cerebral organoid systems,65,66 as well as progress in microfluidic67,68 and single-cell technologies33,34,69,70 that can assist in reproducing crucial aspects of brain tumors such as the BBB and molecular heterogeneity, will allow us to learn more about the interactions between distinct subtypes of glioma cells and their microenvironment. Systems biology and mathematical modeling71,72 will also help us in finding new, more effective ways to target the dependencies of brain tumors. Although preclinical models do not perfectly recapitulate human gliomas, these models may be very useful if carefully selected based on the targeted therapy to be tested.

Find Currently Available Drugs to Hit the Targets

Novel treatments for gliomas are urgently needed, but the process of developing new agents that reach clinical application is highly time-consuming and expensive. Therefore, drug repurposing has been employed to facilitate and accelerate the discovery of new cancer therapies.73,74 Several compounds that have been successfully used for non-oncology indications have been repositioned for the treatment of gliomas, based on findings of tumor cell characteristics that might render them sensitive to those therapeutic agents. Those repurposed drugs have key advantages, such as known drug CNS activity, BBB penetration, pharmacokinetics/pharmacodynamics, and clinical safety information.75 Furthermore, they are less costly than newly marketed drugs and have already been approved for clinical use by regulatory authorities. Antiepileptic drugs, such as valproic acid, levetiracetam, and talampanel, have been investigated, but no favorable survival outcome has been associated with their use in clinical studies for GBM.76–79 The antidiabetic drug metformin has also been investigated as an adjunct to GBM treatment due to its modulatory effects on metabolism; however, pooled retrospective analyses of clinical trial data have not produced conclusive evidence supporting its use in the treatment of GBM.80 Disulfiram, a drug used to manage chronic alcoholism, has recently been found to have preclinical activity in glioma81–84 and have been evaluated in clinical trials ( NCT01777919). Mebendazole, an FDA-approved anthelmintic drug, has been shown to exhibit antitumor effects by inhibiting protein kinases and potentially induce microtubule destabilization in preclinical glioma models.85 Due to mebendazole’s efficacy in tumor suppression, and evidence from laboratory studies suggesting that it can help improve the effectiveness of radiotherapy and temozolomide,86,87 numerous clinical trials have been underway to investigate this possibility (NCT01729260, NCT01837862, and NCT02644291). In addition, as gliomas are a molecularly complex disease with high dynamic heterogeneity, multi-targeted approaches have been explored, such as in the Coordinated Undermining of Survival Paths by 9 repurposed drugs (CUSP9) trial88 by the International Initiative for Accelerated Improvement of Glioblastoma Care.89 The initially proposed regimen consisted of 9 already-marketed drugs that had not been previously used for oncological indications but showed some inhibitory effects on signaling pathways that promote GBM growth. Preliminary results confirming the safety of CUSP9 (version 3) combined with metronomic temozolomide were presented at the Society for Neuro-Oncology meeting in 2018. Whether these regimens have any activity remains to be determined.

Furthermore, the integration of modern OMICS technology, systems biology, and high-throughput drug screens would allow for a more systematic approach and individually tailored, patient-specific therapeutic strategy. This type of approach was explored by Byron et al.90 in a single-arm clinical trial (NCT02060890). While this study did not show therapeutic efficacy, it demonstrated that a molecularly driven, integrative approach is technically feasible. A more collaborative and transparent setting with respect to industry-generated drugs, granting access to deprioritized drugs and related information, would greatly facilitate and accelerate successful drug repurposing for the treatment of malignant gliomas. An example of such an initiative is the Discovering New Therapeutic Uses for Existing Molecules program, which represents a collaboration between the NIH National Center for Advancing Translational Sciences and pharmaceutical companies such as Astra Zeneca, Janssen Research and Development, LLC, and Pfizer Inc.91 Moreover, widely accessible comprehensive data libraries integrating results from OMICS analyses as well as molecular and clinical data on available drugs, such as the drug repurposing hub or the ReFRAME library, need to be developed.92,93 Additional integrative platforms and funding opportunities would further promote drug repurposing and the discovery of novel treatments for malignant gliomas.

Demonstrate That These Drugs Reach the Target in Therapeutic Concentrations

Achieving effective concentrations of anticancer drugs within solid tumors is difficult as these are challenging to measure. In brain tumors, exclusion of therapeutic agents due to the BBB coupled with the limited and likely non-uniform distribution of compounds within the brain and tumor region makes adequate drug delivery a critical challenge.75 Published reports of in vivo animal and human clinical studies evaluating drug levels and target inhibition in the brain provide an initial resource for evaluating the potential CNS activity of a compound. In addition, in silico predictive models have been developed to aid in predicting BBB penetration.94 Clinical trials that attempt to assess the extent of drug delivery to targeted GBM tissue typically involve treatment of patients prior to surgery; drug concentration is then measured in a small sample of the contrast-enhancing resected tumor. However, these analyses need to take into account that most of the tumor remaining after surgery is non-enhancing and infiltrative, likely with different genomic changes and a more intact BBB when compared with the resected tumor.95 Therefore, understanding the differences in drug distribution between the enhancing and non-enhancing tumor tissue, as well as the potential differences in the biological response to the drug in these distinct regions, is critically important. Future therapeutic planning will thus need to consider BBB penetration of the selected therapy and/or potential methods to circumvent this barrier to effective drug delivery. Another challenge is the fact that the drug concentrations measured are typically total drug concentration and what is likely more relevant is the free-drug concentration. This is hard to measure and requires labor-intensive approaches such as microdialysis or modeling approaches with uncertain reliability.96

Modify or Circumvent the BBB to Increase Drug Delivery to the Brain

One method for optimizing the delivery of drugs to gliomas would be to modify or circumvent the BBB. Various techniques have been researched in an effort to increase BBB permeability and optimize treatment success. Magnetic resonance-guided focused ultrasound (MRgFUS), coupled with injected microbubbles, has been used to disrupt the BBB in a way that is both temporary and customizable. Animal studies have shown enhanced drug delivery following MRgFUS, with no axonal or neuronal injury, and a recent phase I study has shown early safety and feasibility, though further investigation is warranted.97 Another phase I study conducted in recurrent GBM patients recently showed that pulsed ultrasound was well tolerated and may increase the effectiveness of systemic drug therapies, such as carboplatin, in the brain without inducing neurotoxicity.98 Laser Interstitial Thermal Therapy (LITT) has also been shown to temporarily disrupt the BBB, and research is ongoing to analyze the effects on drug delivery. In 2016, one of the pivotal clinical trials testing these effects reported that BBB permeability increased within 1–2 weeks after LITT, with resolution over 4–6 weeks, suggesting a window of opportunity for drug delivery.99 Recently, preclinical data evaluating Tumor Treating Fields’ effects on the BBB have indicated their potential for increasing BBB permeability, and further studies are being conducted.100 In addition to physical methods of BBB disruption, several pharmacological approaches have also been tested. Cereport (RMP-7), a bradykinin B2 receptor agonist, transiently increases the permeability of the BBB.101,102 It has been shown to effectively enhance chemotherapeutic effect when combined with carboplatin,103 as well as improve the delivery of other drugs into the CNS.104 More recently, the A2A adenosine receptor signaling pathway has also been targeted to achieve BBB disruption.105–107 Co-administration of regadenoson, an FDA-approved adenosine receptor agonist used for cardiac stress testing, has been shown to increase temozolomide levels in rat brain by 60%, without affecting plasma concentrations,108 but had no effect in a pilot study on BBB in normal human CNS.109 An initial study in GBM patients did not show improvement in temozolomide concentrations in the brain.110 Whether these approaches disrupt the BBB sufficiently to significantly enhance the delivery of therapeutic agents remains to be seen. Alternative dosing strategies may be required to maximize the benefit of transient BBB disruption.

Pharmacologic approaches are an important method to circumvent the BBB, especially when intervention with a device is contraindicated due to tumor location and/or size. Transmembrane diffusion and transporters are 2 important pathways for substances to cross the BBB. Transmembrane diffusion is a non-saturable process that is enhanced when a drug is hydrophobic and small in terms of molecular weight. However, one important limitation of a drug’s hydrophobicity/high lipid solubility is that its uptake by peripheral tissues is increased, thus decreasing the drug’s blood concentration and amount reaching the BBB, in the case of glioma-targeting drugs. Transporter-targeted drug delivery systems have emerged as promising platforms for efficient drug delivery. Transporters use a saturable transport system that allows the uptake of large, hydrophilic substances. Furthermore, transporters for peptides and regulatory proteins can be selective of the areas they target within the central nervous system.111

Osmotic BBB opening is a method used to increase drug concentration within the CNS. Hypertonic mannitol is infused into the vessel, with barrier disruption within the tumor evaluated by either contrast-enhanced computed tomography or radionucleotide scanning. Early clinical studies using this method were able to identify increased methotrexate levels in the brain that corresponded with contrast enhancement on imaging.112 However, several issues with this approach have limited its widespread use. First, the procedure remains labor intensive and invasive, with complications related to catheter use including thrombosis as well as groin hematoma post catheterization. Other issues that arise are related to BBB disruption and include seizures and other neurological symptoms.113 The costs associated with osmotic BBB opening procedures also remain an issue. In addition, infusion in one vessel is a major limitation, since tumors are not contained within the distribution of only one vessel, and thus multiple vessels must be infused. Lastly, this approach has been tested for several decades, with the initial phase I study performed in 1979, but there have not been significant clinical improvements.114

Convection-enhanced delivery (CED) is another mechanism that has been used in GBM treatment that utilizes a pressure gradient to drive fluid flow throughout the tumor. In comparison to diffusion-based mechanisms, CED delivers drugs evenly, at increased quantities, and throughout larger tissue volumes. Limitations of CED include the challenging selection of agents that specifically target the intra- and peritumoral regions while minimizing toxicity to normal cells within the peritumoral region. Challenges also include trajectory planning in the use of more than one cannula to target the intra- and peritumoral regions. Furthermore, cannula design for CED must take into consideration several key principles, which include minimizing injury to normal cells, combining flexibility with increased speed of drug delivery, and optimizing the durability of the device. Improvements in cannula design have allowed for increased flexibility (once the rigid stylet has been removed following insertion) and longer duration for infusion of therapy, as demonstrated by cannulas used in recent clinical trials,115,116 they are still a limiting factor when considering CED.117

Biodegradable polymers have been used in GBM and implanted after surgical tumor debulking to allow local drug delivery, while minimizing systemic side effects.118 Furthermore, polymeric delivery allows prolonged and controlled exposure to the drug. Further studies are being explored to expand polymeric delivery to various chemotherapies and biologics focusing on novel targets.119 There remain questions regarding whether this approach allows sufficient diffusion of therapeutic agents into the tumor against bulk flow.

Design Clinical Trials With Goals That Progress Logically

Design and implementation of a clinical trial typically require the full support of a major institution, given the resources required to successfully achieve its goals, which are mainly to determine whether new treatments are both safe and effective. These goals should be reasonably achievable based on existing literature and preclinical data. Progression through the phases of any trial will determine the safety profile of any interventional arm and also allow for the monitoring of toxicity. These data will allow for the quantification of the intervention and increase its broad applicability. Patients should typically be monitored during the trial for biomarkers that are specific for the intervention in question. Finally, in addition to overall survival, logical endpoints should include 6-month progression-free survival as well as durable response rates. As life expectancy increases, greater emphasis on quality of life endpoints will be paramount to ensure meaningful response is achieved.

For clinical trials for gliomas, radiologic endpoints to assess tumor response have been continuously updated by the Response Assessment in Neuro-Oncology (RANO) working groups,120 which discuss gliomas, brain metastases, and leptomeningeal disease. The earliest RANO working group, RANO-high-grade glioma (RANO-HGG), aimed to address issues surrounding pseudo-progression following completion of fractionated radiation treatment and concomitant chemotherapy, as well as pseudo-response following the use of antiangiogenic treatments affecting vascular permeability.121 The immunotherapy RANO (iRANO) working group has provided guidance on radiographic assessment for patients receiving immunotherapy to allow for close monitoring with imaging over the first 3 months of starting immunotherapy, when patients typically have radiographic worsening in the setting of clinical stability. This recommendation was developed from observations in other solid tumors that show transient radiographic worsening prior to response, which is similar to the phenomenon of pseudo-progression post-radiation and is often interpreted as immune-mediated inflammatory changes.121,122 Future directions for RANO include assessment of PET imaging in the management of glioma patients, involving both prognostic information and response to treatment. As a supplement to MRI-based assessment, PET imaging through amino acid PET tracers O-(2-[18F]-fluoroethyl)-l-tyrosine and 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine has been shown to accurately determine changes related to progressive tumor, versus treatment effects related to pseudo-progression or delayed radiation-related changes.121,123 Advances in MR imaging techniques and utilization of PET imaging will play a significant role in identifying radiologic endpoints in future glioma studies.

Next-generation sequencing and molecular testing of gliomas have become more widespread due to a greater emphasis on molecular characterization to be integrated with histological diagnosis, as described in the updated 2016 World Health Organization Classification of Tumors of the Central Nervous System124 and further elaborated in the next update as already discussed by the cIMPACT approach. Molecular classification may reduce concerns of histological ambiguity in biopsied tissues, while leveraging our understanding of certain biomarkers to be predictive, prognostic, and/or diagnostic.125

In addition, a number of recent platform trials with Bayesian adaptive randomization designs, such as the INSIGhT126 and GBM AGILE127 trials, are attempting to accelerate the development of novel targeted agents and match biomarkers to specific agents. For the INSIGhT trial, experimental arms are compared with a common control arm of fractionated radiation therapy with concomitant temozolomide, followed by adjuvant temozolomide. The initial stage uses experimental arms with defined genomic biomarkers that are tested simultaneously. The trial evolves as the acquired results alter the randomization probabilities of treatment effects on progression-free survival. The model incorporates biomarker-specific probability and methods incorporate Bayesian estimation. The results that accumulate during the second stage may lead to several treatment arms dropping off the study due to low probability affecting overall survival with potential for new arms being added. GBM AGILE similarly uses 2 stages and employs Bayesian estimation in their statistical method. The initial stage employs adaptive randomization among various treatment arms (considering clinical and biomarker endpoints) to evaluate multiple treatment options simultaneously and move treatments that hold promise into the second stage which utilizes fixed randomization versus control to accelerate the clinical trial process. The same principles are used in platform basket trials, such as the N2M2 trial,128 that is assigning patients into multiple arms base on tumor genotype.

Summary

Precision medicine offers an individualized treatment plan based on the patient’s tumor molecular markers and genetic signature. Tumor heterogeneity must be addressed by identifying targets that are “drivers.” Modeling systems using advanced mathematics and computational biology can aid in target identification while inhibition of downstream signaling pathways may address their redundancy. Once drugs that reach the desired target are identified, we must demonstrate effective drug delivery that maintains therapeutic concentration in non-contrast-enhancing tumors. Clinical trials with novel adaptive designs will allow targeted drug testing but maintain flexibility to allow pivoting to different treatment pathways at the earliest time a drug is suspected to be ineffective. Clinical trials remain the preferred recommendation for both newly diagnosed and recurrent GBM patients, as standard-of-care treatment is simply not good enough for the majority of patients due to poor prognosis. Given the lack of competing therapies, NGS panels that can provide information on potentially actionable targets remain a very attractive tool when tailoring treatment for individual patients. However, if the drug cannot reach the desired target in therapeutic concentrations, then the patient may sustain systemic toxicities without a meaningful therapeutic response. Tailored treatments must consider ease of drug delivery, effective therapeutic concentrations, and the adverse effects when assessing efficacy.

Funding

This research has been funded by Adam Michael Rosen Foundation, ReMission Alliance Against Brain Tumors, and Wells Brain Tumor Research Fund at UF Foundation.

Conflict of interest statement. The authors do not have conflicts of interest related to the information in this manuscript. Details of paid consulting and funding not related to this content are below. A.G. has received personal fees from Monteris Medical and Novocure. A.G. has also received research funding support from Orbus Therapeutics. L.H.M. declares that she has no conflict of interest. M.J. declares that she has no conflict of interest. D.S. declares that he has no conflict of interest. W.W. is a paid consultant for Bayer, Merck Sharp and Dohme, and Roche (with the compensation received by the University of Heidelberg), and reports receiving other commercial research support from Roche, Apogenix, and Pfizer. D.A.M. has intellectual property licensed or optioned to Celldex Therapeutics, Annias Immunotherapeutics, and iOncologi. D.A.M. has served as an advisor/consultant to Bristol-Myers Squibb, Tocagen, and Oncorus, and is cofounder of iOncologi, Inc., an immuno-oncology biotechnology company. Neither any reagent nor any funding from these organizations was used in this study. P.Y.W. is a member of the Speaker’s Bureau for Merck and Prime Oncology. P.Y.W. serves as a consultant/advisory board member for Agios, Astra Zeneca, Bayer, Blue Earth Diagnostics, Immunomic Therapeutics, Karyopharm, Kiyatec, Puma, Taiho, Vascular Biogenics, Deciphera, and VBI Vaccines. P.Y.W. also has received research support from Agios, Astra Zeneca, Bayer, Blue Earth Diagnostics, Immunomic Therapeutics, Karyopharm, Kiyatec, Puma, Taiho, Vascular Biogenics, Deciphera, and VBI Vaccines. S.A.G. declares that he has no conflict of interest.

Authorship Statement. Conception and design: all authors; manuscript writing: all authors; and final approval of the manuscript: all authors.

References

  • 1. Shin SH, Bode AM, Dong Z. Precision medicine: the foundation of future cancer therapeutics. NPJ Precis Oncol.  2017;1(1):12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Kaley T, Touat M, Subbiah V, et al.  BRAF inhibition in BRAFV600-mutant gliomas: results from the VE-BASKET study. J Clin Oncol.  2018;36(35):3477–3484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Gambella A, Senetta R, Collemi G, et al.  NTRK fusions in central nervous system tumors: a rare, but worthy target. Int J Mol Sci. 2020; 21(3):753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Puchalski RB, Shah N, Miller J, et al.  An anatomic transcriptional atlas of human glioblastoma. Science.  2018;360(6389):660–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Platten M. EGFRvIII vaccine in glioblastoma-InACT-IVe or not ReACTive enough?  Neuro Oncol.  2017;19(11):1425–1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Schäfer N, Gielen GH, Rauschenbach L, et al.  Longitudinal heterogeneity in glioblastoma: moving targets in recurrent versus primary tumors. J Transl Med.  2019;17(1):96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Draaisma K, Chatzipli A, Taphoorn M, et al.  Molecular evolution of IDH wild-type glioblastomas treated with standard of care affects survival and design of precision medicine trials: a report from the EORTC 1542 study. J Clin Oncol.  2020;38(1):81–99. [DOI] [PubMed] [Google Scholar]
  • 8. Waks AG, Winer EP. Breast cancer treatment: a review. JAMA.  2019;321(3):288–300. [DOI] [PubMed] [Google Scholar]
  • 9. Brown-Glaberman U, Dayao Z, Royce M. HER2-targeted therapy for early-stage breast cancer: a comprehensive review. Oncology (Williston Park).  2014;28(4):281–289. [PubMed] [Google Scholar]
  • 10. Stagno F, Stella S, Spitaleri A, Pennisi MS, Di Raimondo F, Vigneri P. Imatinib mesylate in chronic myeloid leukemia: frontline treatment and long-term outcomes. Expert Rev Anticancer Ther.  2016;16(3): 273–278. [DOI] [PubMed] [Google Scholar]
  • 11. Iqbal N, Iqbal N. Imatinib: a breakthrough of targeted therapy in cancer. Chemother Res Pract.  2014;2014:357027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Bonomi PD, Gandara D, Hirsch FR, et al.  Predictive biomarkers for response to EGFR-directed monoclonal antibodies for advanced squamous cell lung cancer. Ann Oncol.  2018;29(8):1701–1709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Agustoni F, Suda K, Yu H, et al.  EGFR-directed monoclonal antibodies in combination with chemotherapy for treatment of non-small-cell lung cancer: an updated review of clinical trials and new perspectives in biomarkers analysis. Cancer Treat Rev.  2019;72:15–27. [DOI] [PubMed] [Google Scholar]
  • 14. Lin JJ, Riely GJ, Shaw AT. Targeting ALK: precision medicine takes on drug resistance. Cancer Discov.  2017;7(2):137–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Kerr KM, López-Ríos F. Precision medicine in NSCLC and pathology: how does ALK fit in the pathway?  Ann Oncol. 2016;27(Suppl. 3): iii16–iii24. [DOI] [PubMed] [Google Scholar]
  • 16. Dummer R, Ascierto PA, Gogas HJ, et al.  Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol.  2018;19(5):603–615. [DOI] [PubMed] [Google Scholar]
  • 17. Kim A, Cohen MS. The discovery of vemurafenib for the treatment of BRAF-mutated metastatic melanoma. Expert Opin Drug Discov.  2016;11(9):907–916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Larkin J, Ascierto PA, Dréno B, et al.  Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med.  2014;371(20):1867–1876. [DOI] [PubMed] [Google Scholar]
  • 19. Gil J, Laczmanska I, Pesz KA, Sasiadek MM. Personalized medicine in oncology. New perspectives in management of gliomas. Contemp Oncol (Pozn).  2018;22(1A):1–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Chapman PB, Hauschild A, Robert C, et al. ; BRIM-3 Study Group Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med.  2011;364(26):2507–2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Cheng L, Lopez-Beltran A, Massari F, MacLennan GT, Montironi R. Molecular testing for BRAF mutations to inform melanoma treatment decisions: a move toward precision medicine. Mod Pathol.  2018;31(1):24–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Madala HR, Punganuru SR, Arutla V, Misra S, Thomas TJ, Srivenugopal KS. Beyond brooding on oncometabolic havoc in IDH-mutant gliomas and AML: current and future therapeutic strategies. Cancers (Basel). 2018;10(2):49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. DiNardo CD, Watts JM, Stein EM, et al.  Ivosidenib (AG-120) induced durable remissions and transfusion independence in patients with IDH1-mutant relapsed or refractory myelodysplastic syndrome: results from a phase 1 dose escalation and expansion study. Blood. 2018;132(Suppl. 1):1812–1812. [Google Scholar]
  • 24. Stein EM, DiNardo CD, Fathi AT, et al.  Ivosidenib or enasidenib combined with induction and consolidation chemotherapy in patients with newly diagnosed AML with an IDH1 or IDH2 mutation is safe, effective, and leads to MRD-negative complete remissions. Blood. 2018;132(Suppl. 1):560–560. [Google Scholar]
  • 25. Mellinghoff IK, Ellingson BM, Touat M, et al.  Ivosidenib in isocitrate dehydrogenase 1-mutated advanced glioma. J Clin Oncol. 2020;38(29):3398–3406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Yuan M, Huang LL, Chen JH, Wu J, Xu Q. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer. Signal Transduct Target Ther.  2019;4:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Ramalingam SS, Vansteenkiste J, Planchard D, et al. ; FLAURA Investigators Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med.  2020;382(1):41–50. [DOI] [PubMed] [Google Scholar]
  • 28. Lee J, Choi Y, Han J, et al.  Osimertinib improves overall survival in patients with EGFR-mutated NSCLC with leptomeningeal metastases regardless of T790M mutational status. J Thorac Oncol. 2020;15(11):1758–1766. [DOI] [PubMed] [Google Scholar]
  • 29. Park S, Lee MH, Seong M, et al.  A phase II, multicenter, two cohort study of 160 mg osimertinib in EGFR T790M-positive non-small-cell lung cancer patients with brain metastases or leptomeningeal disease who progressed on prior EGFR TKI therapy. Ann Oncol.  2020;31(10):1397–1404. [DOI] [PubMed] [Google Scholar]
  • 30. Reungwetwattana T, Nakagawa K, Cho BC, et al.  CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-mutated advanced non-small-cell lung cancer. J Clin Oncol. 2018:JCO2018783118. [DOI] [PubMed] [Google Scholar]
  • 31. Hovestadt V, Smith KS, Bihannic L, et al.  Resolving medulloblastoma cellular architecture by single-cell genomics. Nature.  2019;572(7767):74–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Neftel C, Laffy J, Filbin MG, et al.  An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell. 2019;178(4):835–849.e821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Filbin MG, Tirosh I, Hovestadt V, et al.  Developmental and oncogenic programs in H3K27M gliomas dissected by single-cell RNA-seq. Science.  2018;360(6386):331–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Patel AP, Tirosh I, Trombetta JJ, et al.  Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science.  2014;344(6190):1396–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell.  2015;27(1):15–26. [DOI] [PubMed] [Google Scholar]
  • 36. Barthel FP, Johnson KC, Varn FS, et al. ; GLASS Consortium Longitudinal molecular trajectories of diffuse glioma in adults. Nature.  2019;576(7785):112–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Harder BG, Blomquist MR, Wang J, et al.  Developments in blood-brain barrier penetrance and drug repurposing for improved treatment of glioblastoma. Front Oncol.  2018;8:462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Sarkaria JN, Hu LS, Parney IF, et al.  Is the blood-brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro Oncol.  2018;20(2):184–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Brennan CW, Verhaak RG, McKenna A, et al. ; TCGA Research Network The somatic genomic landscape of glioblastoma. Cell.  2013;155(2):462–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Wen PY, Touat M, Alexander BM, et al.  Buparlisib in patients with recurrent glioblastoma harboring phosphatidylinositol 3-kinase pathway activation: an open-label, multicenter, multi-arm, phase II trial. J Clin Oncol.  2019;37(9):741–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Snuderl M, Fazlollahi L, Le LP, et al.  Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. Cancer Cell.  2011;20(6):810–817. [DOI] [PubMed] [Google Scholar]
  • 43. Yan H, Parsons DW, Jin G, et al.  IDH1 and IDH2 mutations in gliomas. N Engl J Med.  2009;360(8):765–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Parsons DW, Jones S, Zhang X, et al.  An integrated genomic analysis of human glioblastoma multiforme. Science.  2008;321(5897):1807–1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Louis DN, Wesseling P, Aldape K, et al.  cIMPACT-NOW update 6: new entity and diagnostic principle recommendations of the cIMPACT-Utrecht meeting on future CNS tumor classification and grading. Brain Pathol.  2020;30(4):844–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Dang L, White DW, Gross S, et al.  Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature.  2009;462(7274):739–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Mazor T, Chesnelong C, Pankov A, et al.  Clonal expansion and epigenetic reprogramming following deletion or amplification of mutant IDH1. Proc Natl Acad Sci U S A.  2017;114(40):10743–10748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Sasaki M, Knobbe CB, Munger JC, et al.  IDH1(R132H) mutation increases murine haematopoietic progenitors and alters epigenetics. Nature.  2012;488(7413):656–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Mellinghoff I, Penas-Prado M, Peters K, et al.  ACTR-31. phase 1 study of AG-881, an inhibitor of mutant IDH1 and IDH2: results from the recurrent/progressive glioma population. Neuro Oncol. 2018;20:vi18. [Google Scholar]
  • 50. Mellinghoff I, Touat M, Maher E, et al.  ACTR-46. AG-120, A first-in-class mutant IDH1 inhibitor in patients with recurrent or progressive IDH1 mutant glioma: updated results from the phase 1 non-enhancing glioma population. Neuro Oncol. 2017;19:vi10–vi11. [Google Scholar]
  • 51. IDH inhibitors target common glioma mutation. Cancer Discov. 2019;9(8):992. [DOI] [PubMed] [Google Scholar]
  • 52. Killela PJ, Reitman ZJ, Jiao Y, et al.  TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc Natl Acad Sci U S A.  2013;110(15):6021–6026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Zanetti M. A second chance for telomerase reverse transcriptase in anticancer immunotherapy. Nat Rev Clin Oncol. 2017;14(2):115–128. [DOI] [PubMed] [Google Scholar]
  • 54. Cesare AJ, Reddel RR. Alternative lengthening of telomeres: models, mechanisms and implications. Nat Rev Genet.  2010;11(5):319–330. [DOI] [PubMed] [Google Scholar]
  • 55. Kaley T, Touat M, Subbiah V, et al.  BRAF inhibition in BRAF(V600)-mutant gliomas: results from the VE-BASKET study. J Clin Oncol. 2018:3477–3484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wen P, Stein A, van den Bent M, et al.  ACTR-30. Updated efficacy and safety of dabrafenib plus trametinib in patients with recurrent/refractory BRAF V600E–mutated high-grade glioma (HGG) and low-grade glioma (LGG). Neuro Oncol. 2019;21:vi19–vi20. [Google Scholar]
  • 57. Roth P, Reijneveld JC, Gorlia T, et al.  EORTC 1709/CCTG CE.8: a phase III trial of marizomib in combination with standard temozolomide-based radiochemotherapy versus standard temozolomide-based radiochemotherapy alone in patients with newly diagnosed glioblastoma. J Clin Oncol. 2019;37(15 Suppl):TPS2072–TPS2072. [Google Scholar]
  • 58. Lassman AB, Bent MJVD, Wen PY, et al.  OS07.5 Interim analysis data from Phase 2 study on efficacy, safety & intratumoral pharmacokinetics of oral Selinexor (KPT-330) in patients with recurrent glioblastoma (GBM). Neuro Oncol. 2017;19(Suppl. 3):iii14–iii14. [Google Scholar]
  • 59. Osswald M, Jung E, Sahm F, et al.  Brain tumour cells interconnect to a functional and resistant network. Nature.  2015;528(7580):93–98. [DOI] [PubMed] [Google Scholar]
  • 60. Venkataramani V, Tanev DI, Strahle C, et al.  Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature.  2019;573(7775):532–538. [DOI] [PubMed] [Google Scholar]
  • 61. Jung E, Alfonso J, Osswald M, Monyer H, Wick W, Winkler F. Emerging intersections between neuroscience and glioma biology. Nat Neurosci.  2019;22(12):1951–1960. [DOI] [PubMed] [Google Scholar]
  • 62. Monje M, Borniger JC, D’Silva NJ, et al.  Roadmap for the emerging field of cancer neuroscience. Cell.  2020;181(2):219–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W. Glioma: experimental models and reality. Acta Neuropathol.  2017;133(2):263–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Oh BH, Westler WM, Darba P, Markley JL. Protein carbon-13 spin systems by a single two-dimensional nuclear magnetic resonance experiment. Science.  1988;240(4854):908–911. [DOI] [PubMed] [Google Scholar]
  • 65. Linkous A, Balamatsias D, Snuderl M, et al.  Modeling patient-derived glioblastoma with cerebral organoids. Cell Rep. 2019;26(12):3203–3211.e3205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Lancaster MA, Renner M, Martin CA, et al.  Cerebral organoids model human brain development and microcephaly. Nature.  2013;501(7467):373–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Park TE, Mustafaoglu N, Herland A, et al.  Hypoxia-enhanced blood-brain barrier chip recapitulates human barrier function and shuttling of drugs and antibodies. Nat Commun.  2019;10(1):2621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Ayuso JM, Monge R, Martínez-González A, et al.  Glioblastoma on a microfluidic chip: generating pseudopalisades and enhancing aggressiveness through blood vessel obstruction events. Neuro Oncol.  2017;19(4):503–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Venteicher AS, Tirosh I, Hebert C, et al.  Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science. 2017;355(6332). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Tirosh I, Venteicher AS, Hebert C, et al.  Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature.  2016;539(7628):309–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Miranda A, Cova T, Sousa J, Vitorino C, Pais A. Computational modeling in glioblastoma: from the prediction of blood-brain barrier permeability to the simulation of tumor behavior. Future Med Chem.  2018;10(1):121–131. [DOI] [PubMed] [Google Scholar]
  • 72. Altrock PM, Liu LL, Michor F. The mathematics of cancer: integrating quantitative models. Nat Rev Cancer.  2015;15(12):730–745. [DOI] [PubMed] [Google Scholar]
  • 73. Siegelin MD, Schneider E, Westhoff MA, Wirtz CR, Karpel-Massler G. Current state and future perspective of drug repurposing in malignant glioma. Semin Cancer Biol. 2019. [DOI] [PubMed] [Google Scholar]
  • 74. Abbruzzese C, Matteoni S, Signore M, et al.  Drug repurposing for the treatment of glioblastoma multiforme. J Exp Clin Cancer Res.  2017;36(1):169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Prados MD, Byron SA, Tran NL, et al.  Toward precision medicine in glioblastoma: the promise and the challenges. Neuro Oncol.  2015;17(8):1051–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Happold C, Gorlia T, Chinot O, et al.  Does valproic acid or levetiracetam improve survival in glioblastoma? A pooled analysis of prospective clinical trials in newly diagnosed glioblastoma. J Clin Oncol.  2016;34(7):731–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Krauze AV, Myrehaug SD, Chang MG, et al.  A phase 2 study of concurrent radiation therapy, temozolomide, and the histone deacetylase inhibitor valproic acid for patients with glioblastoma. Int J Radiat Oncol Biol Phys.  2015;92(5):986–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Bobustuc GC, Baker CH, Limaye A, et al.  Levetiracetam enhances p53-mediated MGMT inhibition and sensitizes glioblastoma cells to temozolomide. Neuro Oncol.  2010;12(9):917–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Grossman SA, Ye X, Chamberlain M, et al.  Talampanel with standard radiation and temozolomide in patients with newly diagnosed glioblastoma: a multicenter phase II trial. J Clin Oncol.  2009;27(25):4155–4161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Seliger C, Genbrugge E, Gorlia T, et al. ; EORTC Brain Tumor Group Use of metformin and outcome of patients with newly diagnosed glioblastoma: pooled analysis. Int J Cancer.  2020;146(3):803–809. [DOI] [PubMed] [Google Scholar]
  • 81. Paranjpe A, Zhang R, Ali-Osman F, Bobustuc GC, Srivenugopal KS. Disulfiram is a direct and potent inhibitor of human O6-methylguanine-DNA methyltransferase (MGMT) in brain tumor cells and mouse brain and markedly increases the alkylating DNA damage. Carcinogenesis.  2014;35(3):692–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Wang W, Darling JL. How could a drug used to treat alcoholism also be effective against glioblastoma?  Expert Rev Anticancer Ther.  2013;13(3):239–241. [DOI] [PubMed] [Google Scholar]
  • 83. Hothi P, Martins TJ, Chen L, et al.  High-throughput chemical screens identify disulfiram as an inhibitor of human glioblastoma stem cells. Oncotarget.  2012;3(10):1124–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Triscott J, Lee C, Hu K, et al.  Disulfiram, a drug widely used to control alcoholism, suppresses the self-renewal of glioblastoma and over-rides resistance to temozolomide. Oncotarget.  2012;3(10):1112–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. De Witt M, Gamble A, Hanson D, et al.  Repurposing mebendazole as a replacement for vincristine for the treatment of brain tumors. Mol Med.  2017;23:50–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Kipper FC, Silva AO, Marc AL, et al.  Vinblastine and antihelmintic mebendazole potentiate temozolomide in resistant gliomas. Invest New Drugs.  2018;36(2):323–331. [DOI] [PubMed] [Google Scholar]
  • 87. Markowitz D, Ha G, Ruggieri R, Symons M. Microtubule-targeting agents can sensitize cancer cells to ionizing radiation by an interphase-based mechanism. Onco Targets Ther.  2017;10:5633–5642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Kast RE, Boockvar JA, Brüning A, et al.  A conceptually new treatment approach for relapsed glioblastoma: coordinated undermining of survival paths with nine repurposed drugs (CUSP9) by the International Initiative for Accelerated Improvement of Glioblastoma Care. Oncotarget.  2013;4(4):502–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Kast RE, Karpel-Massler G, Halatsch ME. CUSP9* treatment protocol for recurrent glioblastoma: aprepitant, artesunate, auranofin, captopril, celecoxib, disulfiram, itraconazole, ritonavir, sertraline augmenting continuous low dose temozolomide. Oncotarget.  2014;5(18):8052–8082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Byron SA, Tran NL, Halperin RF, et al.  Prospective feasibility trial for genomics-informed treatment in recurrent and progressive glioblastoma. Clin Cancer Res.  2018;24(2):295–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Allison M. NCATS launches drug repurposing program. Nat Biotechnol.  2012;30(7):571–572. [DOI] [PubMed] [Google Scholar]
  • 92. Corsello SM, Bittker JA, Liu Z, et al.  The Drug Repurposing Hub: a next-generation drug library and information resource. Nat Med.  2017;23(4):405–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Frail DE, Brady M, Escott KJ, et al.  Pioneering government-sponsored drug repositioning collaborations: progress and learning. Nat Rev Drug Discov.  2015;14(12):833–841. [DOI] [PubMed] [Google Scholar]
  • 94. Lanevskij K, Japertas P, Didziapetris R. Improving the prediction of drug disposition in the brain. Expert Opin Drug Metab Toxicol.  2013;9(4):473–486. [DOI] [PubMed] [Google Scholar]
  • 95. Grossman SA, Romo CG, Rudek MA, et al.  Baseline requirements for novel agents being considered for phase II/III brain cancer efficacy trials: conclusions from the Adult Brain Tumor Consortium’s first workshop on CNS drug delivery. Neuro Oncol.  2020;22(10):1422–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Bao X, Wu J, Sanai N, Li J. Determination of total and unbound ribociclib in human plasma and brain tumor tissues using liquid chromatography coupled with tandem mass spectrometry. J Pharm Biomed Anal.  2019;166:197–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Mainprize T, Lipsman N, Huang Y, et al.  Blood-brain barrier opening in primary brain tumors with non-invasive MR-guided focused ultrasound: a clinical safety and feasibility study. Sci Rep.  2019;9(1):321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Idbaih A, Canney M, Belin L, et al.  Safety and feasibility of repeated and transient blood-brain barrier disruption by pulsed ultrasound in patients with recurrent glioblastoma. Clin Cancer Res.  2019;25(13): 3793–3801. [DOI] [PubMed] [Google Scholar]
  • 99. Leuthardt EC, Duan C, Kim MJ, et al.  Hyperthermic laser ablation of recurrent glioblastoma leads to temporary disruption of the peritumoral blood brain barrier. PLoS One.  2016;11(2):e0148613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Kessler A, Schaeffer C, Burek M, et al.  EXTH-41. Effects of tumor treating fields (TTFields) on blood brain barrier (BBB) permeability. Neuro Oncol. 2018;20:vi93–vi94. [Google Scholar]
  • 101. Dean RL, Emerich DF, Hasler BP, Bartus RT. Cereport (RMP-7) increases carboplatin levels in brain tumors after pretreatment with dexamethasone. Neuro Oncol.  1999;1(4):268–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Mackic JB, Stins M, Jovanovic S, Kim KS, Bartus RT, Zlokovic BV. Cereport (RMP-7) increases the permeability of human brain microvascular endothelial cell monolayers. Pharm Res.  1999;16(9): 1360–1365. [DOI] [PubMed] [Google Scholar]
  • 103. Prados MD, Schold SC Jr, Fine HA, et al.  A randomized, double-blind, placebo-controlled, phase 2 study of RMP-7 in combination with carboplatin administered intravenously for the treatment of recurrent malignant glioma. Neuro Oncol.  2003;5(2):96–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Borlongan CV, Emerich DF. Facilitation of drug entry into the CNS via transient permeation of blood brain barrier: laboratory and preliminary clinical evidence from bradykinin receptor agonist, Cereport. Brain Res Bull.  2003;60(3):297–306. [DOI] [PubMed] [Google Scholar]
  • 105. Bynoe MS, Viret C, Yan A, Kim DG. Adenosine receptor signaling: a key to opening the blood-brain door. Fluids Barriers CNS.  2015;12:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Kim DG, Bynoe MS. A2A adenosine receptor regulates the human blood-brain barrier permeability. Mol Neurobiol.  2015;52(1):664–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Carman AJ, Mills JH, Krenz A, Kim DG, Bynoe MS. Adenosine receptor signaling modulates permeability of the blood-brain barrier. J Neurosci.  2011;31(37):13272–13280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Jackson S, Anders NM, Mangraviti A, et al.  The effect of regadenoson-induced transient disruption of the blood-brain barrier on temozolomide delivery to normal rat brain. J Neurooncol.  2016;126(3):433–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Jackson S, George RT, Lodge MA, et al.  The effect of regadenoson on the integrity of the human blood-brain barrier, a pilot study. J Neurooncol.  2017;132(3):513–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Jackson S, Weingart J, Nduom EK, et al.  The effect of an adenosine A2A agonist on intra-tumoral concentrations of temozolomide in patients with recurrent glioblastoma. Fluids Barriers CNS.  2018; 15(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Banks WA. Characteristics of compounds that cross the blood-brain barrier. BMC Neurol.  2009;9(Suppl. 1):S3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Neuwelt EA, Specht HD, Howieson J, et al.  Osmotic blood-brain barrier modification: clinical documentation by enhanced CT scanning and/or radionuclide brain scanning. AJR Am J Roentgenol.  1983;141(4):829–835. [DOI] [PubMed] [Google Scholar]
  • 113. McAllister LD, Doolittle ND, Guastadisegni PE, et al.  Cognitive outcomes and long-term follow-up results after enhanced chemotherapy delivery for primary central nervous system lymphoma. Neurosurgery. 2000;46(1):51–60;discussion 60-1. [PubMed] [Google Scholar]
  • 114. Bellavance MA, Blanchette M, Fortin D. Recent advances in blood-brain barrier disruption as a CNS delivery strategy. AAPS J.  2008;10(1):166–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Sampson JH, Akabani G, Archer GE, et al.  Intracerebral infusion of an EGFR-targeted toxin in recurrent malignant brain tumors. Neuro Oncol.  2008;10(3):320–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Kunwar S, Chang S, Westphal M, et al. ; PRECISE Study Group Phase III randomized trial of CED of IL13-PE38QQR vs Gliadel wafers for recurrent glioblastoma. Neuro Oncol.  2010;12(8):871–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Jahangiri A, Chin AT, Flanigan PM, Chen R, Bankiewicz K, Aghi MK. Convection-enhanced delivery in glioblastoma: a review of preclinical and clinical studies. J Neurosurg.  2017;126(1):191–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Brem H, Piantadosi S, Burger PC, et al.  Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group. Lancet.  1995;345(8956):1008–1012. [DOI] [PubMed] [Google Scholar]
  • 119. Guerin C, Olivi A, Weingart JD, Lawson HC, Brem H. Recent advances in brain tumor therapy: local intracerebral drug delivery by polymers. Invest New Drugs.  2004;22(1):27–37. [DOI] [PubMed] [Google Scholar]
  • 120. Wen PY, Chang SM, Van den Bent MJ, Vogelbaum MA, Macdonald DR, Lee EQ. Response assessment in neuro-oncology clinical trials. J Clin Oncol.  2017;35(21):2439–2449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Chukwueke UN, Wen PY. Use of the Response Assessment in Neuro-Oncology (RANO) criteria in clinical trials and clinical practice. CNS Oncol. 2019;8(1):Cns28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Hodi FS, O’Day SJ, McDermott DF, et al.  Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med.  2010;363(8):711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Galldiks N, Lohmann P, Albert NL, Tonn JC, Langen K-J. Current status of PET imaging in neuro-oncology. Neuro Oncol Adv. 2019;1(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Louis DN, Perry A, Reifenberger G, et al.  The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol.  2016;131(6):803–820. [DOI] [PubMed] [Google Scholar]
  • 125. Zacher A, Kaulich K, Stepanow S, et al.  Molecular diagnostics of gliomas using next generation sequencing of a glioma-tailored gene panel. Brain Pathol.  2017;27(2):146–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Alexander BM, Trippa L, Gaffey S, et al.  Individualized screening trial of innovative glioblastoma therapy (INSIGhT): a Bayesian adaptive platform trial to develop precision medicines for patients with glioblastoma. JCO Precis Oncol. 2019(3):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Alexander BM, Ba S, Berger MS, et al. ; GBM AGILE Network Adaptive global innovative learning environment for glioblastoma: GBM AGILE. Clin Cancer Res.  2018;24(4):737–743. [DOI] [PubMed] [Google Scholar]
  • 128. Wick W, Dettmer S, Berberich A, et al.  N2M2 (NOA-20) phase I/II trial of molecularly matched targeted therapies plus radiotherapy in patients with newly diagnosed non-MGMT hypermethylated glioblastoma. Neuro Oncol.  2019;21(1):95–105. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neuro-oncology Advances are provided here courtesy of Oxford University Press

RESOURCES