Next Article in Journal
Non-Invasive Delivery of Nano-Emulsified Sesame Oil-Extract of Turmeric Attenuates Lung Inflammation
Next Article in Special Issue
Focused Ultrasound and Microbubbles-Mediated Drug Delivery to Brain Tumor
Previous Article in Journal
Development and Characterization of Biointeractive Gelatin Wound Dressing Based on Extract of Punica granatum Linn
Previous Article in Special Issue
Pharmacokinetic Principles and Their Application to Central Nervous System Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Addressing BBB Heterogeneity: A New Paradigm for Drug Delivery to Brain Tumors

1
Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
2
Department of Biomedical Sciences, University of Minnesota Medical School—Duluth, Duluth, MN 55812, USA
3
Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902, USA
*
Authors to whom correspondence should be addressed.
Submission received: 20 November 2020 / Revised: 8 December 2020 / Accepted: 10 December 2020 / Published: 11 December 2020
(This article belongs to the Special Issue Drug Delivery to Brain Tumors)

Abstract

:
Effective treatments for brain tumors remain one of the most urgent and unmet needs in modern oncology. This is due not only to the presence of the neurovascular unit/blood–brain barrier (NVU/BBB) but also to the heterogeneity of barrier alteration in the case of brain tumors, which results in what is referred to as the blood–tumor barrier (BTB). Herein, we discuss this heterogeneity, how it contributes to the failure of novel pharmaceutical treatment strategies, and why a “whole brain” approach to the treatment of brain tumors might be beneficial. We discuss various methods by which these obstacles might be overcome and assess how these strategies are progressing in the clinic. We believe that by approaching brain tumor treatment from this perspective, a new paradigm for drug delivery to brain tumors might be established.

Graphical Abstract

1. Introduction

The blood–brain barrier (BBB) remains one of the greatest obstacles to effective pharmaceutical interventions in the treatment of central nervous system (CNS) disease, including brain tumors. While it is true that some loss of neurovascular and barrier integrity may occur in and around brain tumors, the magnitude of this change is not consistent, and new pharmaceutical strategies for the treatment of brain tumors have yet to show significant efficacy in the clinic [1,2,3]. This lack of efficacy is largely attributed to insufficient drug delivery due to the presence of the BBB. The dense vascular network of the brain works to strictly regulate the transport of substances into and out of the brain parenchyma in order to maintain ionic homeostasis, nutrient supply, and removal of waste for optimal neuronal function. In recent decades, research has revealed that the BBB is composed of specialized endothelial cells (ECs), which are surrounded and supported by pericytes and astrocytes and are regulated by neuronal signaling, forming what is referred to as the neurovascular unit (NVU) [4]. A lack of vesicular transport across these specialized ECs and the presence of active efflux proteins help to further restrict the access of drugs to the CNS [5]. Currently, treatment for the majority of brain tumors involves maximal surgical resection, if possible, followed by radiation, and in the case of glioblastoma multiforme (GBM), concomitant temozolomide (TMZ) [3]. However, these treatments often prove to be palliative, and malignant brain tumors are nearly always fatal within five years of initial diagnosis [6,7].
While treatments for peripheral malignancies have improved dramatically in recent decades with the advent of earlier diagnosis, improved imaging, targeted small molecule inhibitors, and large molecule biologics, the treatment of brain tumors has lagged far behind, and their incidence is on the rise [7]. Therefore, it is imperative to understand how the NVU/BBB may be altered in the case of brain tumors and how to design pharmaceutical interventions specifically to overcome this challenge while maintaining neurovascular integrity as much as possible. To this end, a number of strategies have been proposed to improve drug delivery to the brain and brain tumors. Invasive strategies to bypass the NVU/BBB include convection-enhanced delivery (CED) and direct injection, in addition to polymer-based, biodegradable implants for drug delivery. Noninvasive strategies might include focused ultrasound (FUS) and hyperosmotic disruption of the NVU/BBB, as well as inhibition of efflux transporters, nanoparticle-based strategies, and the use of endogenous transport mechanisms across the brain EC by receptor-mediated transcytosis. In this review, we will introduce brain barrier anatomy and physiology, discuss the heterogeneous impacts of tumor growth and signaling on NVU/BBB integrity, and provide brief overviews of the strategies investigated to deliver drugs to CNS tumors.

2. Barriers and Boundaries in the Brain

Rational drug delivery to any organ requires a thorough understanding of the structures and properties of the target tissue. This section includes detailed features of the dynamic NVU model that is rapidly supplanting the former static BBB concept. Furthermore, the role that these features serve in guiding the molecular basis for current therapies for CNS tumors is illustrated.

2.1. CNS Blood–Tissue Barriers

Any strategy for blood-borne drug delivery into the CNS must consider several structural obstacles related to blood–tissue interfaces [8,9]. First, the brain is covered by layers of cells collectively described as the dura–arachnoid–pia membranes. The dura separates peripheral vessels within the cranium from the cerebrospinal fluid (CSF) in which the brain resides. Several compact layers of epithelial cells, with tight junctional contacts and relatively low surface areas, prevent materials from traversing this boundary. Within the CSF compartment, the pial layer contains vessels that penetrate the brain parenchyma. These vessels also exhibit very low permeability and surface area. Other major barriers include relatively small regions of CNS circulation and include the choroid plexus, circumventricular organs (CVOs), and ependymal cells. Each of these has specialized epithelial cells with properties that highly restrict water-soluble chemicals from penetrating the mass of parenchymal tissue. The choroid plexus that produces CSF, for example, contains specialized epithelial cells with tight junctions (TJs) encasing a fenestrated vascular endothelium that allows the exchange of blood constituents with the extracellular space. The CVOs are also composed of fenestrated endothelial cells, but their location is confined by surrounding tanycytes, a specialized epithelial cell that also has tight junctional contacts that localize and restrict the interstitial fluid. Direct exchange between the CSF and interstitial fluid is restricted by ependymal cells that line the ventricular surface and form a selectively permeable cellular barrier.

2.2. Neurovascular Unit

By far, the major blood–tissue interface in the brain is the microvasculature network extending from arterioles to capillaries and to venules. By illustration, in one gram of human brain, the length of vessels, if joined end to end, approximates the length of 4.5 football fields and almost a quarter (~23%) of the surface area of a sheet of photocopy paper. The current model of the brain microvasculature consists of several different cell types working collectively to form a functional NVU [4,10]. The endothelial cells and their tight junctional contacts that block paracellular diffusion are surrounded by pericytes that form gap junctions with multiple adjacent endothelial cells and by astrocytic endfeet that cover >99% of the endothelial–pericyte cell surface (Figure 1). The astrocytes, in turn, extend processes that monitor synaptic activity and react by signaling endothelial cells and pericytes to respond to increased metabolic demands by increasing nutrient delivery. Microglia, the resident immune cells, are extravascular when dormant but react swiftly to remove cellular debris (phagocytosis) or respond to inflammatory signals associated with disease or injury. Loss of pericytes and their signaling in the NVU by injury or genetic means leads to reduced expression of endothelial tight junction proteins and dysfunction of their permeability barrier [11,12]. These examples illustrate the remarkable dynamics, plasticity, and interdependence of signaling among the NVU cells to maintain functional stability in the contemporary model of the neurovasculature.

2.3. Blood to Brain Permeability and Transport

As the brain depends on external nutrients for growth and development and yet is protected from the influence of circulating toxins or xenobiotics, the endothelial cell exhibits critical membrane-imbedded proteins that function as transporters. One group of transporters includes facilitated carriers and secondary active transporters for the delivery of energy substrates and essential nutrients (Table 1). The glucose transporter (GLUT1), monocarboxylic acid transporter (MCT1), and amino acid transporters are examples of the >40 transporters detected by functional and transcriptomic analyses [13].
A second type of transporter that is critical to brain drug delivery is the ABC (ATP-binding cassette) superfamily that uses the energy of ATP hydrolysis to expel endogenous and exogenous xenobiotics from the cell (and from the brain) and return them to the blood for transformation and excretion (Table 2). The most relevant ABC transporters expressed by brain endothelial cells are P-glycoprotein (P-gp, ABCB1), breast-cancer-related protein (BCRP, ABCG2), and the multiple drug-related proteins (MRP1, -4, -5; ABCC1, -4, -5). Many anticancer drugs are substrates of the ABC transporters and, therefore, may influence their effectiveness as brain cancer drugs (Figure 1). The relevance of ABC transporters in anticancer drug delivery in brain tumors with apparently high permeability is illustrated by the fact that the most permeable tumor vasculatures have influx rate constants several-fold less than the rate constants for other organs such as muscle, heart, lung, kidney, and liver [14]. Therefore, efflux mechanisms are likely important even in CNS tumors, in which the permeability of the brain vasculature is compromised and elevated compared to the surrounding tissue.

3. Heterogeneous Blood–Tumor Barrier Permeability

The understanding of the BBB’s physical and biochemical barrier functions, including the expression of tight junction proteins, restricted paracellular transport, and active efflux mechanisms, has been well established. However, determining the integrity of the NVU/BBB in and around tumors and how this affects tumor treatment has been less straightforward. In the case of both primary and metastatic tumors, the NVU/BBB is subject to changes due to tumor growth and signaling, and these alterations in NVU/BBB integrity and physiology result in what will hereafter be referred to as the blood–tumor barrier (BTB). The BTB may be characterized by an inflammatory environment with increased numbers of activated astrocytes, vascular endothelial growth factor (VEGF)-induced reduction in the expression of tight junction proteins like claudin-5, breakdown of the basal lamina, and tumor cell interference in associations between endothelial cells and astrocytic endfeet (Figure 2) [15,16,17]. There is also evidence for a change in the phenotype of BTB-associated pericytes, which may show decreased platelet-derived growth factor receptor-β (PDGFR-β) expression in addition to increased desmin expression [18]. As a result of these changes, the BTB can be, on average, somewhat “leakier” (more permeable) than the normal NVU/BBB in the absence of disease [17,19,20]. The predominant question with regards to BBB breakdown and the treatment of brain tumors has therefore been, is the breakdown of the NVU/BBB in the case of brain tumors significant and uniform enough to allow for the accumulation of efficacious drug concentrations?
As this question has been repeatedly investigated, various preclinical tumor models have routinely led to conflicting results. In some cases, tumor vascular permeability, assessed by the accumulation of fluorescent tracers, has been previously correlated with growth patterns, tumor size, or peripheral tumor of origin [21]. In other cases, including a variety of brain-trophic metastatic breast cancer models developed at the National Institutes of Health (NIH), no correlation between tumor size and permeability has been found [14,19]. These studies also found that the variability of BTB permeability among tumors in the same animal and even among regions of the same tumors, as assessed by the accumulation of fluorescent tracers and small molecules like paclitaxel, doxorubicin, and lapatinib, could be as much as 100-fold [14,22,23]. More recent studies in HER2+ brain-trophic breast cancer metastasis models have shown a poor correlation between drug accumulation and tracer accumulation, as well as inconsistent drug uptake and variable efficacy of biologics like trastuzumab and other antibody-based therapies [22,24,25]. Another model of lung cancer brain metastases found two-fold increases in permeability to small molecules like 3H-mannitol but concluded that this small relative increase in addition to functional P-gp was still a significant limitation to systemic drug therapy [26]. In addition, a number of studies utilizing transporter-knockout mice and patient-derived xenograft (PDX) models of GBMs and brain metastases have shown that the efficacy of systemic administration of various small molecules is consistently limited by the presence of the NVU/BBB and BTB, active efflux, and the fact that vascular permeability is widely variable within and around the tumor region [20,27,28,29,30,31,32,33]. This heterogeneity in permeability at the BBB leads to wide variability in drug/tracer accumulation and has also been confirmed by elegant correlated ultramicroscopy and MRI techniques in preclinical tumor models [34]. These studies point to the conclusion that relying on the potential for increased BTB permeability is unlikely to result in efficacious treatment through the systemic administration of novel therapies and their subsequent regulatory approval for such applications.
Although the aforementioned evidence has been largely preclinical, it agrees with clinical observations when considered in the appropriate context. Increased permeability of the BTB, relative to normal brain, is observed clinically, as increased uptake of tracers in magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging allows for definitive diagnosis of brain tumors and informs many aspects of their treatment [35]. However, especially in the case of diffuse and invasive tumors like GBM, it has also been shown that nonenhancing, infiltrating regions of brain tumors often exist outside of the region of T1-weighted contrast enhancement [36,37]. This indicates that some portions of the malignant tumor are protected by a relatively uncompromised NVU/BBB. The patterns of treatment failure are strongly correlated with and attributed to these nonenhancing regions, and maximal resection that includes these regions improves survival [38,39,40]. Increasingly, early-phase studies, in which patients receive drugs prior to tumor resection and biopsy, are being utilized to determine the real extent of antineoplastic drug permeability to the BTB [1,2]. Although fold-increases in drug concentrations relative to normal brain may be observed at the core of the tumor, this may still not be adequate to cause cell death. As has been evidenced in many of the aforementioned preclinical models, it is unlikely that these drug concentrations are representative of concentrations in the entirety of the tumor. In the case of GBM, the infiltrative boundaries of the tumor are likely to have a more competent and intact BTB, closer to that of “unaffected brain” [20,31,41,42]. This heterogeneous drug distribution among different regions of the tumor is also clinically evidenced in drug concentrations from biopsies of non-contrast-enhancing tumor regions [43].
As there has been a great success with novel treatments of peripheral disease, the culmination of decades of brain tumor research has led to the conclusion that it is imperative that molecules and delivery strategies be designed foremost with an intact NVU/BBB in mind. As an example, GNE317, a small molecule that was designed specifically to avoid active efflux, showed significantly higher activity in a model of brain metastases of lung cancer than another counterpart PI3K inhibitor not designed to penetrate the BBB/NVU [44,45]. Other brain-penetrant inhibitors like osimertinib, an EGFR inhibitor, have also shown better preclinical and potential clinical efficacy [46,47]. While designing small lipophilic molecules in an attempt to optimize tumor penetration and minimize active efflux is certainly one potential method towards effective treatments for brain tumors, there are a vast number of other drug delivery strategies and novel molecules in development for this application. These strategies will be discussed in the following sections.

4. Invasive Technologies

The NVU/BBB poses numerous challenges for efficient drug delivery to the brain and brain tumors, as discussed in the previous section. To address these challenges, various invasive and noninvasive strategies have been developed to improve the delivery of therapeutic agents to the brain. Invasive technologies are based on local delivery of therapeutics to the brain, bypassing the NVU/BBB entirely. They include drug delivery to the cerebrospinal fluid (CSF) via intrathecal or intraventricular injections and interstitial delivery via biodegradable wafers or catheters (Figure 3).

4.1. Intrathecal and Intraventricular Injections

Intrathecal (IT) administration involves direct injection of therapeutics into the CSF that fills the thecal space and encompasses intrathecal–lumbar injection but can also be used to describe intracerebroventricular or intracisternal magna injections [48]. Chemotherapy may be administered directly into the lumbar thecal sac via lumbar puncture or infused into the lateral ventricle through a subcutaneous reservoir and a ventricular catheter, allowing the drug to distribute into the target sites via diffusion [49]. Drug delivery via lumbar puncture may require multiple administrations, is highly invasive, causes discomfort to the patient, and is not likely to allow effective drug delivery to brain tumors. Alternatively, intraventricular infusions are often administered via the Ommaya reservoir, invented in 1963 by Ayub Ommaya, which is inserted into one of the lateral ventricles (Figure 3) [50,51]. Clinically, it is essential to ensure correct placement of the catheter in the ventricle, and, to this end, new state of the art technology using smartphones is being developed as a guide for accurate neuronavigation and catheter placement, which may make these procedures more accessible to a variety of clinics [52].
In the landscape of brain tumor treatment utilizing this technique, there are several clinical reports of intrathecal (IT) administration of large molecules, including trastuzumab, in patients with leptomeningeal metastatic brain tumors [53,54,55,56,57,58,59]. However, these studies also utilized concomitant systemic administration of trastuzumab, and, therefore, the positive symptomatic benefit cannot be solely attributed to IT administration. There are other case reports that have combined trastuzumab with other cytotoxic agents like methotrexate and/or cytarabine, resulting in prolongation of disease control in HER2+ brain tumor patients [60,61,62,63,64,65,66]. In the preclinical setting, prophylactic IT administration of adeno-associated virus serotype 9 (AAV9) gene therapy vectors and tumor tropic neural stem cells (NSCs) loaded with chemotherapeutic agents shows that IT administration has the potential for efficient drug delivery of large molecules and biological vectors to the subarachnoid space [48,67,68,69].
From amongst small molecules, methotrexate and cytarabine are frequently prescribed for IT administration. However, there are numerous reports of neurotoxicity and other complications such as transverse myelopathy associated with the IT administration of these drugs [70,71,72,73,74,75]. Therefore, although high concentrations can be attained in the CSF using IT injections, reducing the total dose and risk of systemic toxicity, this method of administration has its drawbacks. The rate of drug distribution is slow and inversely proportional to the molecular weight, meaning large molecules often have very low or undetectable concentrations when distant from the site of injection [76]. Additionally, rapid CSF turnover, as compared to the rate of diffusion, results in faster clearance of the therapeutics from the site of administration [77,78]. For years, there has been a common misconception that the distribution of drugs into the CSF is indicative of NVU/BBB permeability and that delivery of drugs to the CSF would ensure delivery to the deeper brain tissues. However, it is now more widely accepted that this is not the case, and the reader is directed to a review of this topic [79].

4.2. Convection-Enhanced Delivery

Convection-enhanced delivery (CED) is one of the most explored techniques to bypass the NVU/BBB; it was developed in the early 1990s by Edward Oldfield’s group at the NIH [80]. CED involves the infusion of fluids locally under pressure into the interstitial space in the brain or tumor using stereotactically placed catheters. CED primarily utilizes bulk flow, and diffusion is a minor component. While diffusion relies on the concentration gradient, and macromolecules penetrate only up to a few millimeters under diffusive forces, the distribution pattern attained with CED can be described by Darcy’s law, in which the velocity of the molecule is dependent on the pressure gradient and hydraulic conductivity of the medium [81,82].
CED is being widely studied in preclinical and clinical studies for GBM and diffuse intrinsic pontine glioma (DIPG). Souweidane et al. studied CED in combination with PET imaging to deliver a PET-visible histone deacetylase (HDAC) inhibitor, PETobinostat, for theranostic applications in DIPG [83]. Various nanotechnology-based drug delivery systems, like liposomes, nanoparticles, and polymeric micelles, are being administered via CED to increase the volume of the brain tissue accessible to these systems, which are otherwise limited by poor diffusion [84,85,86,87,88,89,90,91]. Models of CED could help inform treatment design and optimization of other parameters like volume of infusate, duration of infusion, catheter design and placement and can guide treatment design [92]. A model was recently developed to understand the flow and distribution of carmustine and paclitaxel solutions and doxorubicin-loaded liposomes post-CED [93,94]. Gill et al. studied CED of panobinostat to understand its pharmacokinetics in brain tissue [95]. A vast number of clinical trials have been initiated to investigate CED for delivery of both large and small molecules, and some of these have been compiled in Table 3.
CED is a promising technique that has the potential to overcome the limitations posed by systemic delivery. Successful translation of this technique to the clinic would have varied applications to treat a multitude of CNS disorders. For CED to reach its full therapeutic potential, characteristic challenges like catheter design and placement, prevention of reflux, tracking infusate delivery, reduction in mechanical tissue damage and edema, and the potential requirements of multiple infusions need to be addressed. Other challenges associated with CED include cost of the procedure, specific clinical expertise, and postprocedural imaging [86].
Table 3. Summary of clinical trials with convection-enhanced delivery (CED) for brain tumors from the clinicaltrials.gov database (accessed on 20 September 2020).
Table 3. Summary of clinical trials with convection-enhanced delivery (CED) for brain tumors from the clinicaltrials.gov database (accessed on 20 September 2020).
TitlePurposeNCT NumberPhaseStatus/OutcomeRef
MTX110 by Convection-Enhanced Delivery in Treating Participants With Newly-Diagnosed Diffuse Intrinsic Pontine Glioma (PNOC015)To study the side effects of panobinostat nanoparticles formulation MTX110 in participants with newly diagnosed DIPGNCT03566199I/IIActive, not recruiting[96]
Chronic Convection Enhanced Delivery of TopotecanPrimarily to establish the safety of prolonged intracerebral CED of chemotherapy in patients with recurrent HGG. Secondly to determine topotecan distribution and radiographic tumor response under the given CED conditionsi. NCT03154996
ii. NCT03927274
iii. NCT02278510
iv. NCT00308165
i. I
ii., iii. Early phase I
iv. I/II
i.Active, not recruiting
ii. Recruiting
iii. Completed: Safety of CMC catheters has been reported
iv. Recruiting
[97,98,99,100,101]
CED With Irinotecan Liposome Injection Using Real-Time Imaging in Children With Diffuse Intrinsic Pontine Glioma (DIPG; PNOC 009)Phase I and Early Efficacy Study of CED of irinotecan liposome injection (nal-IRI) using real-time imaging with gadolinium in children with DIPG who have completed focal radiotherapyNCT03086616IRecruiting[102]
CED of 124I-Omburtamab for Patients With Non-Progressive Diffuse Pontine Gliomas Previously Treated With External Beam Radiation TherapyTo studythe safety of 124I-omburtamab given by CED at different dose levels for DIPGNCT01502917IRecruiting[103]
CED of MTX110 Newly Diagnosed Diffuse Midline GliomasTo find the maximum tolerated dose of MTX110 (a water-soluble Panobinostat nanoparticle formulation) and Gadolinium that can be given safely in children with newly DIPGNCT04264143IRecruiting[104]
Carboplatin in Treating Patients With Recurrent High-Grade GliomasTo evaluate the safety and toxicity of carboplatin administered by CED in HGG. It is a dose-escalating study.NCT01644955ICompleted[105]
Convection-Enhanced Delivery (CED) of MDNA55 in Adults With Recurrent or Progressive GlioblastomaSingle-arm study with the primary endpoint of median overall survival (mOS) and a secondary endpoint of objective response rate (ORR) following a single intra-tumoral infusion of MDNA55 in adult recurrent GBM subjectsNCT02858895IICompleted: mOS 12.4 months for all patients vs. 7.2 months in synthetic control arm (SCA); 13.2 months in patients with high IL4R expression vs. 6.1 months in SCA[106,107,108]
An Open-Label Dose Escalation Safety Study of Convection-Enhanced Delivery of IL13-PE38QQR in Patients With Progressive Pediatric Diffuse Infiltrating Brainstem Glioma and Supratentorial High-Grade GliomaTest the safety and feasibility of giving IL13-PE38QQR directly into regions of the brain in pediatric patients with DIPG or HGG, using CEDNCT00880061ITerminated: did not reach the entire MRI-defined tumor volume in any patient, short-term radiographic effects were observed in 2 of the 5 patients treated.[109,110]
Study of Convection-Enhanced, Image-Assisted Delivery of Liposomal-Irinotecan In Recurrent High-Grade GliomaDose toleration study to determine MTD of nanoliposomal irinotecan in adults with recurrent HGG by CEDNCT02022644IRecruiting[111]
IL13-PE38QQR Infusion After Tumor Resection, Followed by Radiation Therapy With or Without Temozolomide in Patients With Newly Diagnosed Malignant GliomaDetermine the highest dose of IL13-PE38QQR that can be safely administered by CED to the area around the tumor site after surgical resection and concurrent radiation or TMZ NCT00089427ICompleted: Positive results, overall survival linked to catheter placement[112,113]
Safety and Efficacy Study to Treat Recurrent Grade 4 Malignant Brain TumorsTo study the safety and efficacy of TP-38 at 100 ng/mLNCT00104091IICompleted: Results pending[114]
Maximum Tolerated Dose, Safety, and Efficacy of Rhenium Nanoliposomes in Recurrent Glioma (ReSPECT)A multicenter, sequential cohort, open-label, volume, and dose-escalation study of the safety, tolerability, and distribution of 186RNL given by CED to patients with recurrent or progressive malignant glioma after standard surgical, radiation, and/or chemotherapy treatmentNCT01906385I/IIRecruiting[115]
Safety Study of Replication-competent Adenovirus (Delta-24-RGD) in Patients With Recurrent GlioblastomaTo determine the safety and tolerability of Delta-24-RGD administered by CED to the tumor and the surrounding infiltrated brain in patients with recurrent GBMNCT01582516I/IICompleted: Safe and robust replication of the AAV, killing of rHGG cells. ≥95% reduction in tumor size in some patients, 5 patients survived >3 years[116,117]
A Dose-Escalation Phase I Study Of Human-Recombinant Bone Morphogenetic Protein 4 Administered Via CED In GBM PatientsTo evaluate the feasibility and safety of intratumor and interstitial therapy with hBMP4 in increasing doses in patients with progressive and/or multiple recurrent GBMNCT02869243IRecruiting[118]
The PRECISE Trial: Study of IL13-PE38QQR Compared to GLIADEL Wafer in Patients With Recurrent Glioblastoma MultiformeTo determine whether overall survival duration, safety, and quality of life are improved for patients treated with IL13-PE38QQR compared to patients treated with GLIADEL® Wafer following surgical tumor removal in treatment of first recurrence GBMNCT00076986IIICompleted: There was no survival difference between CB administered via CED and Gliadel® Wafer[119,120]
Phase 1 Trial of D2C7-IT in Combination With i. 2141-V11 for Recurrent Malignant Glioma
ii. Atezolimab for recurrent gliomas
Phase 1 study of D2C7-IT in combination with monoclonal antibodiesi. NCT04547777
ii. NCT04160494
iii. NCT02303678
Ii. Not yet recruiting
ii, iii. Recruiting
[121,122,123]
Phase 1b Study PVSRIPO for Recurrent Malignant Glioma in ChildrenConfirm the safety of the selected dose and potential toxicity of oncolytic poliovirus (PV) immunotherapy with PVSRIPO for pediatric patients with recurrent WHO grade III or IV malignant glioma, to determine MTD for phase 2i. NCT03043391, ii. NCT01491893, iii. NCT04479241Ii. Recruiting
ii. Active, not recruiting
[124,125,126]
Phase IIb Clinical Trial With TGF-β2 Antisense Compound AP 12009 for Recurrent or Refractory High-Grade GliomaMultinational dose-finding Phase IIb study of the efficacy and safety of two doses of AP 12009 (OT-101/trabedersen) compared to standard chemotherapy (TMZ or PCV) in adult patients with confirmed recurrent high-grade gliomaNCT00431561IIbCompleted: OT-101 is an effective agent against recurrent gliomas without the myelosuppression effects of chemotherapy, which is unavailable[127,128,129]

4.3. Biodegradable Wafers

The development of biodegradable polymers for drug delivery has surged ahead in recent decades, and numerous controlled release implants have been developed. Such implants allow precise control over the rate of drug release, prolong local exposure, and reduce systemic toxicities. A major landmark of this technology in brain tumor treatment is the Gliadel® polymeric wafer technology that was approved by the Food and Drug Administration (FDA) in September 1996 as an adjunct to surgery to prolong survival in patients with recurrent GBM, for whom surgical resection is indicated [130,131]. The wafers are surgically implanted at the time of tumor resection and gradually release the loaded DNA and RNA alkylating agent, carmustine (BCNU), with the intention of drug diffusion into surrounding tissue. The polymer is polyanhydride poly[1,3-bis (carboxyphenoxy) propane-co-sebacic-acid] (PCPP:SA), of which more than 70% is biodegraded within three weeks of implantation [131]. Stea et al. conducted a systematic literature review of clinical trials and reports of Gliadel® wafers in combination with radiation therapy and TMZ, and their findings suggest a positive additive effect without an increase in toxicity [132]. This warrants the need for larger prospective trials that combine Gliadel® with TMZ and radiation therapy with scientifically backed study design and patient selection for confirming and establishing the anticipated synergy between these treatments [133,134,135,136,137,138,139,140].
Recent research efforts are examining the efficacy of wafers after coloading BCNU and TMZ to determine if there is prolonged survival compared to wafers of the individually loaded drug or orally administered TMZ in rodent glioma models. These PLGA wafers used a pre-encapsulation process and reported 25% long-term survivors (survived >120 days compared to median survival of 28 days) in the F344 rat model [141]. Other chemotherapeutic agents that have been explored for sustained local delivery via polymeric implants include taxol, camptothecin, minocycline, doxorubicin, and others [142,143,144,145,146,147,148,149,150,151,152,153,154,155,156]. Novel polymeric implants and microchips can be used to deliver several drugs locally at varying time points in a controlled manner, and various formulations such as nanoparticles, liposomes, and microparticles can also be delivered via these implants.
A major challenge for this technology is to ensure biocompatibility and biodegradation, as there are reports of incompletely biodegraded materials found up to 68 weeks after implantation; hence, patients must be monitored carefully [157]. The success of these therapies is limited due to their inability to reach distant, invasive, and dense tumor cells due to poor diffusion characteristics. Focused treatments, in combination with localized delivery, are required to target these cells, and local delivery via implants will play a critical role in this mode of drug delivery [158,159].

5. Blood–Brain Barrier Disrupting Strategies

Apart from these invasive methods, other noninvasive techniques have been investigated to transiently disrupt the neurovasculature to enhance drug delivery to the CNS. These methods may have better patient compatibility compared to invasive approaches such as CED and IT injection and will perhaps allow lower dosage, thus reducing toxicity compared to traditional systemic administration routes like intravenous injection.

5.1. Osmotic Blood-Brain Barrier Disruption

The tight junctions of the cerebrovascular endothelium can be transiently and reversibly disrupted by the infusion of a hyperosmolar solution into a cerebral artery putatively because of the shrinkage of endothelial cells following the splitting of tight junctions. The resulting intracellular spaces increase paracellular diffusion and facilitate the delivery of therapeutic, diagnostic, and functional agents relevant to CNS disease. This method was first proposed by Rapoport et al. in 1972, who exposed the pia arachnoid surface of the cerebral cortex of healthy rabbits to different osmotic concentrations, resulting in osmotically induced and reversible cell shrinkage (Figure 4) [160]. In practice, this method involves the infusion of 1.4 M mannitol, which has been FDA-approved for administration to patients [161]. Besides mannitol, other hypertonic solutions used for transient barrier disruption include arabinose, lactamide, saline, urea, and several radiographic contrast agents [162]. The first Phase I clinical trial on osmotic BBB disruption (BBBD) for enhanced drug delivery to the brain was initiated in 1979 [163]. Using this technique in experimental and clinical treatment of brain tumors, permeability enhancements of greater magnitude were observed for tumors with low, rather than high, initial permeability relative to that of normal brain [164].
In the clinical setting, chemotherapeutics used in combination with osmotic BBBD include methotrexate (MTX), carboplatin, melphalan, cyclophosphamide, etoposide, and etoposide phosphate [165]. A clinical study by Neuwelt et al. during the 1980s demonstrated that osmotic BBBD plus MTX produced long-term remission and improved survival in patients with primary CNS lymphoma (PCNSL) [166]. Another clinical study from 1982 to 2005, involving more PCNSL patients treated with osmotic BBBD and methotrexate at four institutions, showed durable tumor control over a 23-year period [167]. Additional clinical studies have demonstrated relatively low toxicity [168,169].
Although the disruption is transient and is fully reversed within several hours [170,171], one risk of osmotic BBB disruption is the additional mass effect in the brain that results from a 1.5% increase in brain fluid content [172,173]. Assessment of the extent of the tumor and the associated mass effect prior to osmotic BBB disruption is important for optimizing protocols and minimizing the risks of this procedure [174].

5.2. Microbubble-Mediated Focused Ultrasound

A localized disruption of the neurovasculature using focused ultrasound (FUS) has been suggested as an anatomically or functionally targeted method for drug delivery from the vasculature into the brain parenchyma. FUS-induced BBB opening in the presence of microbubbles is local, transient, and reversible, usually within several hours [175]. Its feasibility and efficacy to promote the delivery of therapeutic agents into the brain have been examined extensively since 1997 when Kullervo Hynynen and Ferenc Jolesz first demonstrated the potential feasibility of BBBD through the intact human skull utilizing short, high-intensity ultrasound [176]. More recently, with the application of magnetic resonance-compatible transducers, image-guided FUS has allowed targeted localization to brain tumors and reduced the risk of off-target effects [177,178,179].
Therapeutic FUS is generally applied in conjunction with intravenously administered microbubbles (Figure 4). These microbubbles are lipid, protein, or polymer-shelled, inert gas-filled bubbles that are usually between 0.5 to 10 µm in diameter [180]. They are currently FDA-approved for use as contrast agents in ultrasound imaging and are utilized in the context of drug delivery to help reduce the energy threshold required for BBBD [181]. The energy threshold is, to some extent, determined by the size of microbubbles, and, typically, the smaller the diameter of these microbubbles, the higher the pressure required for effectiveness [182]. It is important to carefully control the energy level of FUS, as high pressure and frequency may cause an inflammatory response and/or tissue damage, such as hemorrhage and apoptotic neuronal damage [183]. Extensive research into the safety and feasibility of FUS has been initiated in a variety of CNS diseases, and recently, clinical studies have been conducted to determine the safety and efficacy of the application of FUS with intravenously injected microbubbles in human brain tumors [184,185,186].
FUS with microbubbles has been applied to glioma treatment to assist in the delivery of free therapeutic agents, including small molecules such as doxorubicin [187,188], as well as large molecules like bevacizumab and trastuzumab [189,190,191]. It has also been used for gene- [192,193] and drug-loaded nanoparticle delivery [192,193,194,195]. An alternative way to increase the penetration of therapeutic drugs into the brain is to load the drug of interest into the microbubbles themselves [196,197]. These studies demonstrated that the drug release process could be controlled by the acoustic emission provided by ultrasound imaging. FUS has also been combined with magnetic targeting of magnetic nanoparticles (MNPs) in conjunction with MRI monitoring for CNS drug delivery [198,199]. In this synergistic system, FUS facilitates delivery through the vascular wall via passive enhanced permeability and retention (EPR) effects, while magnetic forces actively enhance the deposition of MNPs into the brain.
This approach for BBBD has been shown to be relatively safe, but it also has limitations [200]. Despite the early increase in drug delivery to the CNS, recent studies with large molecules showed that enhanced permeability was diminished after 5 days [201]. This could be one explanation for why many animal studies on FUS conducted with trastuzumab showed nonsignificant differences regarding survival when comparing FUS- and non-FUS-treated groups [189,190]. Repeated FUS treatment before drug administration may, therefore, be required, which increases risk. Other obstacles in a wide clinical FUS application include issues with repeatability of the FUS procedure and dependence on MRI and specially-trained operators [202].
Overall, FUS-mediated BBBD has provided a promising approach to therapeutic delivery to brain tumors and other CNS diseases such as Alzheimer’s and Parkinson’s diseases [203]. Meanwhile, successful and wider clinical translation requires a more extensive and thorough examination of possible safety issues due to repeated BBBD, the repeatability of FUS treatment, and optimization of ultrasound parameter settings.

6. Nanoparticles

Nanoparticles are a large category of nanoscale particles (1–1000 nm) with the capacity to adsorb, entrap, or be modified with various therapeutic agents. These particles are promising strategies to improve brain drug delivery [204]. This section is focused on nanoparticle strategies to overcome low neurovascular permeability and increase drug delivery into brain tumors. A summary of the major categories of the current nanoparticles is shown in Table 4 and can be broadly categorized into biological vectors and synthetic vectors of various types.

6.1. Biological Vectors

6.1.1. Viral Vectors

Viral vectors have been repeatedly used in GBM gene therapy clinical trials [205]. Viral vectors have the ability to naturally infect cells with nucleic acids with high transfection efficiency [206]. Currently, several viruses have been developed into vectors for brain delivery, including retroviruses, adenoviruses, and adeno-associated viruses (AAVs) [13,205,207,208,209]. Although viral vectors have been studied for over two decades, they have only resulted in marginal increases in overall survival. Long et al. undertook a Phase I clinical trial of p53 gene therapy using an adenovirus vector (Ad-p53). However, transfected cells were found residing only around the injection site [209]. The limitations of using viral vectors for drug delivery include poor brain tumor penetration, highly invasive administration methods, and a prevailing risk of oncogenesis and lethality of viral vectors [13,205,207,208,209].

6.1.2. Exosomes

Exosomes are small endogenous extracellular vesicles (40–100 nm in diameter) that are secreted by various types of cells and have drug-loading and signal-carrying capacity [210]. Exosomes can be loaded with various kinds of cargos, such as nucleic acids, proteins, and small molecules, due to their bubble-like structure [211,212]. Exosomes are generally stable in circulation and lack significant immunogenicity [13,212]. They transport cargos among cells and may even cross BBB via endogenous pathways of intercellular communication [13,210,211,212]. In addition, exosomes have also played important roles in cancer immunotherapy by virtue of the biological signals enclosed in exosomes [210,213]. However, the technologies and strategies to isolate and purify exosomes must be further developed to ensure quality control, and other side effects such as the potential tumor induction risk of tumor-cell-derived exosomes have to be taken into account [206,210,212].

6.1.3. Cell Delivery

Cell-based drug delivery is another exciting strategy for the delivery of therapeutics across the BBB via the innate mobility of cells. There are two cell types that have been evaluated as therapeutic carriers: immune cells and stem cells. In particular, neural stem cells (NSCs), mesenchymal stem cells (MSCs), and neutrophils have been studied for cell-based therapy [214,215,216]. These cell carriers can deliver a variety of therapeutics, including genes, cytokines, enzymes, and nanoparticles across the BBB and are naturally recruited to the sites of brain tumors by an inflammation-mediated pathway [217]. Xue et al. demonstrated that neutrophil-mediated paclitaxel cationic liposomes could penetrate the brain efficiently and slowed the recurrent growth of tumors in mice, with significantly improved survival rates [216]. Balyasnikova et al. developed engineered neural stem cells to express membrane-bound TNF-α-related apoptosis-inducing ligands to induce apoptosis in glioma cells [218]. Detailed mechanisms of the cell carrier’s delivery can be found in the recommended reviews [217,219]. The major difficulties associated with this strategy are the limited therapeutics loading and potential toxicity of the cargo to the cell carriers themselves. Moreover, the spatial and temporal release of the therapeutic agents from the cell carriers must be well-controlled during drug delivery in order to achieve the expected efficacy [217,220].

6.2. Synthetic Vehicles

Synthetic nanoparticles have been broadly investigated to deliver drugs to the brain. The physicochemical properties of the nanoparticles, including size, surface charge, and lipophilicity, are important in the brain passive-diffusion process. A growing interest in the application of inorganic nanoparticles, especially metallic nanoparticles and metallic oxide nanoparticles, in CNS delivery has emerged among the BBB research community [221,222]. Iron oxide nanoparticles, such as maghemite (γ-Fe2O3) and magnetite (Fe3O4), are extensively explored due to their inherent magnetic properties, coupled with tunable size and surface functionality [223,224,225]. Mesoporous silica nanoparticles (MSNPs) are nanoscale silica particles with good loading capacity due to their porous structure and easily modified surface; they are the most commonly applied silica-based delivery vehicles [226]. These inorganic nanoparticles are produced on the scale of nanometers in order to increase their ability to cross the BBB, providing photodynamic or contrast imaging functions due to material properties [223]. However, the potential for neurotoxicity and unspecific distribution are serious barriers to the broad application of metallic nanoparticles [224].
Actively targeted nanoparticles account for the majority of brain drug delivery systems currently under investigation. Surface-modified nanoparticles are transported into the brain, bypassing the BBB by three main routes: adsorptive-mediated transcytosis (AMT), receptor-mediated transcytosis (RMT), and transporter-mediated transcytosis (TMT) [219,227]. Adsorptive-mediated brain targeting largely depends on the electrostatic interaction between the positively-charged drug delivery systems and the negatively-charged BBB [228]. In 2007, Lu et al. evaluated the transcytosis of cationic bovine serum albumin conjugated poly(ethyleneglycol)-poly(lactide) (PEG-PLA) nanoparticles (CBSA-NP) on the BBB. They found that the transcytosis ability of CBSA-NP increased with the increase of CBSA surface density per nanoparticle [229]. However, this nonspecific targeting is the inherent limitation of AMT since negatively charged membranes are present throughout all the vascular system [219]. Moreover, positive nanoparticles have more tendency to adsorb surrounding proteins and form protein coronas [230]. RMT and TMT target the brain more specifically than AMT through ligand-receptor recognition. These receptor-mediated strategies will be further introduced in the following section. The transporters for TMT are usually transporters of nutrient materials like sugars, vitamins, hormones, and amino acids [219,231,232]. These actively targeted nanoparticles target brain tumors more specifically and, therefore, have higher accumulation and lower systemic side effects. However, some concerns such as protein adsorption and corona formation around the nanoparticles, potential neurotoxicity, and difficulty of manufacturing due to the complex structures need to be further addressed [219,233].
Table 4. Nanoparticles for brain drug delivery.
Table 4. Nanoparticles for brain drug delivery.
Ref[207,208,209][210,211,213][214,215,216,218][221,222][227,228][230,233,234]
[231,235]
ExamplesAAV9-hIFNβ, retroviral herpes simplex virus-thymidine kinase (HSV-tk), Toca 511 delivers suicide gene, cytosine deaminase (CD), and in combination with oral prodrug, adenoviral vector carrying the wild-type p53 gene (Ad-p53)Paclitaxel with bEND.3 cell-derived exosome, doxorubicin with U-87 MG cell-derived exosome, miRNA-486-5p transferred exosomes, siKrasG12D iExosomes, tumor-cell-derived exosomes and α-GalCer on a DC-based vaccineNeutrophil-mediated paclitaxel cationic liposomes, carboxylesterase-expressing allogeneic neural stem cells, bone morphogenetic protein 4 (BMP4) expressing adipose-derived mesenchymal stem cells, neural stem cells engineered to express membrane-bound TRAIL (NSCs-mTRAIL)Iron oxide nanoparticles, gold nanoclusters, mesoporous silica nanoparticles, lanthanide upconversion particlesCationized bovine serum albumin modified NPs, polysorbate 80 or poloxamer 188 overcoated NPs, apolipoprotein bound nanoparticles, Transferrin receptor-targeted (OX26) immunoliposomes, LDLR-DHA nanoparticles, insulin-mAb-modified HSA NPs; Glutathione-modified liposomes, choline-derivate-modified NPs
Disadvantagesi. Limited brain tumor penetration
ii. Highly invasive administration method
iii. Prevailing risk of oncogenesis and lethality
i. Lacking standardized isolation and purification procedure,
ii. Donor cells choice
iii. Potential tumor induction risk of tumor cell-derived exosomes
i. Potentially toxic effects of the cargo on the cell carrier itself
ii. Spatial and temporal release of the therapeutic agent
iii. Limited loading efficiency
i. Neurotoxicity
ii. Unspecific distribution
i. Poor selectivity
ii. Protein adsorption and corona formation
i. Protein adsorption and corona formation
ii. Potential neurotoxicity
iii. Difficulty of manufacturing
Advantagesi. High efficiency for gene delivery,
ii. Innate ability to infect cells
i. Nonimmunogenic
ii. Stable and long circulation
iii. Cross BBB
iv. Target the tissue via their natural surface proteins
i. Cross BBB
ii. Naturally recruited to sites of brain tumors
i. Ultrasmall size
ii. Easily modified
iii. Contrast imaging
iv. Phototherapeutics
i. Electrostatic adsorption
ii. Improve cellular uptake
iii. Improve penetrating efficiency
i. High selectivity
ii. Enhanced brain accumulation
iii. Cross BBB
iv. Decrease systemic toxicity
Strategy AMTRMT and TMT
Viral vectorsExosomesCell carriersPassive diffusionActively targeted delivery
Biological vectorsSynthetic vehicles

7. Receptor-Mediated Transcytosis

The final strategy to be discussed in this section is the use of endogenous active transport mechanisms to improve drug delivery to brain tumors, in particular, RMT. This transport is accomplished by three basic steps: binding of the cargo to the target receptor on the luminal side of the brain EC; endocytosis, sorting, and transport across the EC cytoplasm; release of the cargo from the basolateral membrane of the EC into the brain interstitium (Figure 5). As discussed in reference to the heterogeneity of BTB permeability, it may be beneficial to address the treatment of brain tumors by utilizing a “whole brain” delivery strategy to address intact NVU/BBB [236]. RMT-based strategies can be viewed as such an approach because they are generally designed to target transport mechanisms that are functional throughout the extensive vasculature of the brain. Successful brain delivery using RMT requires that target receptors have a high relative expression on the luminal side of the brain endothelium, must mediate transcytosis, and should have high turnover [237,238]. Importantly, the receptor-binding moiety should also have a relatively low affinity for the target in order to optimize drug delivery and release in the brain parenchyma and to limit trafficking to the lysosome [237,239].
Common receptor targets at the BBB include transferrin receptors (TfR1), insulin receptors (IRs), insulin-like growth factor receptors (IGFRs), the low-density lipoprotein-related protein receptor 1 (LRP1), and nicotinic acetylcholine receptors (nAchRs) [240,241]. This strategy has been widely explored in the brain barriers research community for use in a number of CNS diseases, from brain tumors to lysosomal storage disorders, Alzheimer’s disease, Parkinson’s disease, and others. As noted in the previous sections, these various RMT-based delivery mechanisms are often combined with other technologies like BBBD, CED, nanoparticle formulations, gene delivery, and novel biologics. Herein, we will classify delivery constructs into two categories: shuttle peptides and antibody-based constructs.

7.1. BBB Shuttle Peptides

Shuttle peptides are relatively short sequences of amino acids (<50 AAs) that bind to a receptor on the luminal side of the EC to induce endocytosis of the cargo. They are often based on the known sequences from receptor-binding domains of endogenously transported substances like insulin, ApoE, and transferrin, but they may also be discovered by phage display biopanning. These peptides can be directly bound to cargo or associated via noncovalent interactions [238]. Covalently bound shuttle peptides are more likely to have known and relatively consistent stoichiometry, kinetics, and affinity. On the other hand, some investigation into noncovalent associations that may be more prone to cargo release might be more rapidly translated across a number of different drugs for various applications [242,243]. However, their binding affinity for cargo and optimal stoichiometry must be determined. The benefits of shuttle peptides, in general, include their relatively small size, simplicity of synthesis and purification, versatility, and discovery through biopanning. Their limitations include their liability to proteolytic degradation and relatively short half-life in circulation, which may be ameliorated somewhat by cyclization [244].
All of the aforementioned receptors have been targeted for drug delivery. Though there is debate as to the location of LRP1 expression on brain ECs [245,246], a number of shuttle peptides have been developed to target this receptor, including agiopeps and K16ApoE. The K16ApoE peptide consists of 16 lysine residues and the LRP1-binding domain of ApoE [243,247]. While this has shown some evidence of improved drug delivery to the brain, the therapeutic window is narrow and may not be suitable for clinical translation due to acute toxicities observed in mice [242]. Angiopep-2 is one of the most well-characterized shuttle peptides for brain delivery, and it is derived from the Kunitz domain of aprotinin [248]. Angiopep-2 has been widely utilized as a targeting moiety for nanoparticle formulations of antineoplastic agents like TMZ and docetaxel, as well as siRNA, monoclonal antibodies (mAbs), and various radiosensitizing agents for the treatment of CNS tumors [249,250,251,252,253,254,255]. The most developed shuttle peptide construct is likely ANG1005, an angiopep–paclitaxel conjugate recently investigated in clinical trials for the treatment of brain metastases from breast cancer, as well as meningiomas [256,257,258]. Other shuttle peptides include peptide-22, which binds to LDLR, and glutathione, which binds to the GSH transporter. GSH-coated pegylated nanoparticles show increased CNS penetration and have been investigated in clinical trials for the delivery of doxorubicin to brain tumors [259,260]. TfR1 has also been widely investigated as a delivery mechanism due to its expression in tumor cells and brain ECs [261]. T7 targets TfR1 and is a shuttle peptide that has been investigated to deliver antisense oligonucleotides to gliomas [262,263]. Delivery of radiosensitizing gold nanoparticles to brain tumors has been shown to be enhanced by Tfpep [264], and another TfR1-directed peptide, THR, was recently compared with other previously mentioned peptides for the delivery of AAVs and gold nanoparticles to the brain but without specific applications towards the treatment of brain tumors [224,265]. A vast variety of BBB shuttle peptides have been explored, and we direct the reader to an excellent review of the topic for further reading [238].

7.2. Antibody-Based Delivery Systems

Antibody-based therapies are one of the most rapidly evolving fields in pharmaceutics due to their plasma stability, long half-life, and specificity. Antibodies, specifically immunoglobulin G (IgG), are large, bivalent molecules (~150kDa) composed of two identical heavy chains and two identical light chains bound by disulfide bonds. These proteins have proved to be incredibly effective in the treatment of peripheral tumors, but they do not generally cross the BBB from blood into brain [265,266,267,268]. In fact, without enhanced delivery mechanisms, drug accumulation in the brain is likely to be much less than 1% [266,269]. However, there is still significant interest in delivering these drugs to the brain [24,245,270,271,272], and antibodies are also well-suited to serve the same purpose as shuttle peptides to promote RMT. With the recent blossoming of innovative protein engineering and the use of antibody fragments, the somewhat “modular” structure of IgGs has been exploited to modify and utilize different domains. This allows them to be optimized for use as brain-targeted therapies and brain delivery vehicles.
A variety of therapeutic antibodies and antibody-decorated NPs have been targeted at the brain via the TfRs and IRs for treatments of CNS diseases, most notably Alzheimer’s disease and brain tumors [273,274]. In recent years, a nanocarrier of p53 gene therapy, decorated with anti-TfR1 single-chain variable fragments (scFvs), SGT-53, has been successful in preclinical studies and has moved into clinical trials [275,276,277]. Although a study in adult refractory CNS tumors was terminated, actively recruiting studies for children with refractory solid tumors and planned clinical trials in refractory CNS tumors in pediatric patients (NCT02354547, NCT03554707) are still ongoing. Other imaginative antibody constructs explore bispecific or multivalent targeting [278,279]. Recent work from AbbVie demonstrates the targeting of multivalent, dual-variable-domain IgGs (DVD-Igs) with a dual affinity for precision targeting. These molecules can bind two targets, TfR1 for RMT and HER2, for prospective targeting to HER2+ brain tumors while maintaining the Fc domain unchanged, allowing for beneficial FcRn recycling [280]. Furthermore, recently published work from Denali Therapeutics demonstrates a novel protein transport vehicle (TV) with an affinity for TfR1 incorporated into the Fc region of the IgG, allowing for retention of bivalent binding to the therapeutic target [281,282]. Although these are not explicitly intended to treat brain tumors, they are an exciting contribution.

8. Conclusions and Future Perspectives

In the past decade, there has been a tremendous increase in the understanding of the physiology of the BBB. However, this has not translated to efficacious treatment of CNS disorders, which range from epilepsy to brain tumors. In the case of both primary and metastatic brain tumors, the BBB is disrupted heterogeneously, leading to the formation of the blood–tumor barrier (BTB). The BTB harbors considerable structural and functional heterogeneity within the tumor microenvironment and varies across different cancer subtypes [17]. It compels us to question if the leakiness can be used to our advantage to deliver drugs in desired concentrations to the target site.
While some reports have shown a positive correlation between increased permeability in the tumor to tumor size and growth patterns, there are reports, including those from NIH, that demonstrate no correlation between the two. These inconsistencies highlight the problem of heterogeneity of the BBB breakdown, and this challenge is encountered in the clinical setting as well. Diagnosis of brain tumors using fluorescent tracers is facilitated by the increased permeability of the BTB, but why does it not extrapolate to the treatment modalities like chemotherapy? Instead of relying on the altered BBB permeability to deliver cytotoxic cargo to the tumor cells, it would be better to prepare the delivery systems to face the most challenging barrier—the intact BBB—and strategize the delivery to efficiently target the tumor cells and reduce any off-target toxicity.
Over years of research, various strategies have been developed to invasively or noninvasively overcome the BBB. The invasive strategies bypass the BBB altogether and deliver the therapeutic agents directly into the brain parenchyma or into the CSF. These strategies prevent systemic exposure of the drug, thereby limiting its toxicity and side-effects. They have been widely explored in the clinical setting and there are numerous on-going clinical trials, demonstrating the huge potential of this strategy. However, invasive procedures need highly specialized instruments and personnel. From a patient’s perspective, noninvasive strategies are preferred. Various noninvasive BBB-disrupting strategies and nanoparticle drug delivery systems that bypass the BBB by a number of transport routes are discussed in the review. Transient disruption of the BBB using focused ultrasound enables the delivery of a wide range of therapeutics to the brain, ranging from small molecules to large molecules. It is imperative to understand the kinetics and time duration of the temporary disruption to effectively plan the delivery of therapeutics. The first decade of the 21st century saw the “nanoparticle boom” and nanoparticles proved to be able to deliver conventional drugs, recombinant proteins, vaccines, and nucleotides. This versatile carrier system can be modified to target various transcytosis pathways to ensure improved drug delivery using the enhanced permeation retention (EPR) effect [283].
Understanding the physiology of the BBB at the cellular and molecular levels helps researchers design delivery systems that selectively target the receptors and transporters on the cell surface of the BBB. A challenge while developing these constructs is to avoid off-target effects. Ensuring delivery at the site of action is critical to achieving the desired concentrations at that site and minimal off-target effects. Thus, understanding the pharmacokinetics of the delivery systems would be imperative for their progress from the preclinical research settings to the clinical scenario.
In conclusion, a better understanding of the BBB/BTB physiology has led to the development of a multitude of strategies to target the tumor cells present beyond these barriers. As has been demonstrated on numerous occasions in the past, a “one size fits all” approach is not effective. A rational combination of drugs and their delivery is now being designed to attain optimal concentrations in brain tumors. In this way, a comprehensive treatment regime will be established.

Author Contributions

Writing—original draft preparation, J.I.G., S.R., W.Z. (Wenqiu Zhang), W.Z. (Wenjuan Zhang), L.R.D., and W.F.E.; writing—review and editing, J.I.G., S.R., L.R.D., and W.F.E.; visualization, J.I.G. and S.R.; supervision and conceptualization, L.R.D., W.F.E., and J.N.S. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was fundsed by National Cancer Institute: CA220378, National Cancer Institute: CA210180, National Institute of Neurological Disorders and Stroke: NS077921, National Institute of Neurological Disorders and Stroke: NS073610.

Acknowledgments

All figures were created in Biorender.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAVAdeno-associated virus
ABCATP-binding cassette protein
AMTAdsorptive-mediated transcytosis
BBBBlood–brain barrier
BBBDBlood–brain barrier disruption
BCRPBreast cancer resistance protein
BTBBlood–tumor barrier
CEDConvection-enhanced delivery
CNSCentral nervous system
CSFCerebrospinal fluid
CVOCircumventricular organs
DIPGDiffuse intrinsic pontine glioma
ECEndothelial cells
EGFREpidermal growth factor receptor
FDAFood and Drug Administration
FUSFocused ultrasound
GBMGlioblastoma miltiforme
GLUT1Glucose transporter 1
HDACHistone deacetylase
IGFRInsulin-like growth factor receptor
IRInsulin receptor
ITIntrathecal
LDLRLow-density lipoprotein receptor
LRP1Low-density lipoprotein-related protein 1
MCT1Monocarboxylate transporter 1
MNPMagnetic nanoparticles
MSCMesenchymal stem cells
MRIMagnetic resonance imaging
MRPMultidrug resistance protein
NSCNeural stem cell
NVUNeurovascular unit
NVU/BBBNeurovascular unit/blood–brain barrier
PDGFR-βPlatelet-derived growth factor-β
P-gpP-glycoprotein
PLGApoly(lactic-co-glycolic acid)
RMTReceptor-mediated transcytosis
TfR1Transferrin receptor 1
TMTTransporter-mediated transcytosis
VEGFVascular endothelial growth factor

References

  1. Sarkaria, J.N.; Hu, L.S.; Parney, I.F.; Pafundi, D.H.; Brinkmann, D.H.; Laack, N.N.; Giannini, C.; Burns, T.C.; Kizilbash, S.H.; Laramy, J.K.; et al. Is the blood-brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro-Oncology 2018, 20, 184–191. [Google Scholar] [CrossRef] [PubMed]
  2. Pitz, M.W.; Desai, A.; Grossman, S.A.; Blakeley, J.O. Tissue concentration of systemically administered antineoplastic agents in human brain tumors. J. Neuro-Oncol. 2011, 104, 629–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Arvold, N.D.; Lee, E.Q.; Mehta, M.P.; Margolin, K.; Alexander, B.M.; Lin, N.U.; Anders, C.K.; Soffietti, R.; Camidge, D.R.; Vogelbaum, M.A.; et al. Updates in the management of brain metastases. Neuro-Oncology 2016, 18, 1043–1065. [Google Scholar] [CrossRef] [PubMed]
  4. Abbott, N.J.; Patabendige, A.A.K.; Dolman, D.E.M.; Yusof, S.R.; Begley, D.J. Structure and function of the blood–brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
  5. Terasaki, T.; Ohtsuki, S. Brain-to-blood transporters for endogenous substrates and xenobiotics at the blood-brain barrier: An overview of biology and methodology. Neurotherapeutics 2005, 2, 63–72. [Google Scholar] [CrossRef] [PubMed]
  6. Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol. Biomarkers Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Nayak, L.; Lee, E.Q.; Wen, P.Y. Epidemiology of brain metastases. Curr. Oncol. Rep. 2012. [Google Scholar] [CrossRef]
  8. Neuwelt, E.A.; Bauer, B.; Fahlke, C.; Fricker, G.; Iadecola, C.; Janigro, D.; Leybaert, L.; Molnár, Z.; O’Donnell, M.E.; Povlishock, J.T.; et al. Engaging neuroscience to advance translational research in brain barrier biology. Nat. Rev. Neurosci. 2011, 12, 169–182. [Google Scholar] [CrossRef] [Green Version]
  9. Mastorakos, P.; McGAVERN, D.B. The anatomy and immunology of vasculature in the central nervous system. Sci. Immunol. 2019, 4, eaav0492. [Google Scholar] [CrossRef]
  10. Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef]
  11. Daneman, R.; Zhou, L.; Kebede, A.A.; Barres, B.A. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 2010, 468, 562–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Liu, Q.; Lin, W.J.; Tang, Y. New Insights into the Dysfunctions of Pericytes and Neurovascular Units in Neurodegenerative Diseases. Neurosci. Bull. 2020. [Google Scholar] [CrossRef] [PubMed]
  13. Pandit, R.; Chen, L.; Götz, J. The blood-brain barrier: Physiology and strategies for drug delivery. Adv. Drug Deliv. Rev. 2019. [Google Scholar] [CrossRef] [PubMed]
  14. Lockman, P.R.; Mittapalli, R.K.; Taskar, K.S.; Rudraraju, V.; Gril, B.; Bohn, K.A.; Adkins, C.E.; Roberts, A.; Thorsheim, H.R.; Gaasch, J.A.; et al. Heterogeneous Blood-Tumor Barrier Permeability Determines Drug Efficacy in Experimental Brain Metastases of Breast Cancer. Clin. Cancer Res. 2010, 16, 5664–5678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Argaw, A.T.; Gurfein, B.T.; Zhang, Y.; Zameer, A.; John, G.R. VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown. Proc. Natl. Acad. Sci. USA 2009, 106, 1977–1982. [Google Scholar] [CrossRef] [Green Version]
  16. Watkins, S.; Robel, S.; Kimbrough, I.F.; Robert, S.M.; Ellis-Davies, G.; Sontheimer, H. Disruption of astrocyte-vascular coupling and the blood-brain barrier by invading glioma cells. Nat. Commun. 2014, 5, 1–15. [Google Scholar] [CrossRef] [Green Version]
  17. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef]
  18. Lyle, L.T.; Lockman, P.R.; Adkins, C.E.; Mohammad, A.S.; Sechrest, E.; Hua, E.; Palmieri, D.; Liewehr, D.J.; Steinberg, S.M.; Kloc, W.; et al. Alterations in Pericyte Subpopulations Are Associated with Elevated Blood-Tumor Barrier Permeability in Experimental Brain Metastasis of Breast Cancer. Clin. Cancer Res. 2016, 22, 5287–5299. [Google Scholar] [CrossRef] [Green Version]
  19. Adkins, C.E.; Mohammad, A.S.; Terrell-Hall, T.B.; Dolan, E.L.; Shah, N.; Sechrest, E.; Griffith, J.; Lockman, P.R. Characterization of passive permeability at the blood-tumor barrier in five preclinical models of brain metastases of breast cancer. Clin. Exp. Metastasis 2016, 33, 373–383. [Google Scholar] [CrossRef] [Green Version]
  20. Gampa, G.; Kenchappa, R.S.; Mohammad, A.S.; Parrish, K.E.; Kim, M.; Crish, J.F.; Luu, A.; West, R.; Hinojosa, A.Q.; Sarkaria, J.N.; et al. Enhancing Brain Retention of a KIF11 Inhibitor Significantly Improves its Efficacy in a Mouse Model of Glioblastoma. Sci. Rep. 2020, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
  21. Zhang, R.D.; Price, J.E.; Fujimaki, T.; Bucana, C.D.; Fidler, I.J. Differential permeability of the blood-brain barrier in experimental brain metastases produced by human neoplasms implanted into nude mice. Am. J. Pathol. 1992, 141, 1115–1124. [Google Scholar]
  22. Terrell-Hall, T.B.; Nounou, M.I.; El-Amrawy, F.; Griffith, J.I.G.; Lockman, P.R. Trastuzumab distribution in an in-vivo and in-vitro model of brain metastases of breast cancer. Oncotarget 2017, 8, 83734–83744. [Google Scholar] [CrossRef]
  23. Taskar, K.S.; Rudraraju, V.; Mittapalli, R.K.; Samala, R.; Thorsheim, H.R.; Lockman, J.; Gril, B.; Hua, E.; Palmieri, D.; Polli, J.W.; et al. Lapatinib Distribution in HER2 Overexpressing Experimental Brain Metastases of Breast Cancer. Pharm. Res. 2012, 29, 770–781. [Google Scholar] [CrossRef] [Green Version]
  24. Gril, B.; Wei, D.; Zimmer, A.S.; Robinson, C.; Khan, I.; Difilippantonio, S.; Overstreet, M.G.; Steeg, P.S. HER2 antibody-drug conjugate controls growth of breast cancer brain metastases in hematogenous xenograft models, with heterogeneous blood–tumor barrier penetration unlinked to a passive marker. Neuro-Oncology 2020, 22, 1625–1636. [Google Scholar] [CrossRef]
  25. Askoxylakis, V.; Ferraro, G.B.; Kodack, D.P.; Badeaux, M.; Shankaraiah, R.C.; Seano, G.; Kloepper, J.; Vardam, T.; Martin, J.D.; Naxerova, K.; et al. Preclinical Efficacy of Ado-trastuzumab Emtansine in the Brain Microenvironment. J. Natl. Cancer Inst. 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. On, N.H.; Mitchell, R.; Savant, S.D.; Bachmeier, C.J.; Hatch, G.M.; Miller, D.W. Examination of blood-brain barrier (BBB) integrity in a mouse brain tumor model. J. Neuro-Oncol. 2013, 111, 133–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Agarwal, S.; Elmquist, W.F. Insight into the Cooperation of P-glycoprotein (ABCB1) and Breast Cancer Resistance Protein (ABCG2) at the Blood–Brain Barrier: A Case Study Examining Sorafenib Efflux Clearance. Mol. Pharm. 2012, 9, 678–684. [Google Scholar] [CrossRef] [PubMed]
  28. Parrish, K.E.; Pokorny, J.; Mittapalli, R.K.; Bakken, K.; Sarkaria, J.N.; Elmquist, W.F. Efflux Transporters at the Blood-Brain Barrier Limit Delivery and Efficacy of Cyclin-Dependent Kinase 4/6 Inhibitor Palbociclib (PD-0332991) in an Orthotopic Brain Tumor Model. J. Pharmacol. Exp. Ther. 2015, 355, 264–271. [Google Scholar] [CrossRef]
  29. Gampa, G.; Kim, M.; Mohammad, A.S.; Parrish, K.E.; Mladek, A.C.; Sarkaria, J.N.; Elmquist, W.F. Brain Distribution and Active Efflux of Three panRAF Inhibitors: Considerations in the Treatment of Melanoma Brain Metastases. J. Pharmacol. Exp. Ther. 2019, 368, 446–461. [Google Scholar] [CrossRef] [Green Version]
  30. Mittapalli, R.K.; Chung, A.H.; Parrish, K.E.; Crabtree, D.; Halvorson, K.G.; Hu, G.; Elmquist, W.F.; Becher, O.J. ABCG2 and ABCB1 Limit the Efficacy of Dasatinib in a PDGF-B-Driven Brainstem Glioma Model. Mol. Cancer Ther. 2016, 15, 819–829. [Google Scholar] [CrossRef] [Green Version]
  31. Pokorny, J.L.; Calligaris, D.; Gupta, S.K.; Iyekegbe, D.O.; Mueller, D.; Bakken, K.K.; Carlson, B.L.; Schroeder, M.A.; Evans, D.L.; Lou, Z.; et al. The efficacy of the wee1 inhibitor MK-1775 combined with temozolomide is limited by heterogeneous distribution across the blood-brain barrier in glioblastoma. Clin. Cancer Res. 2015, 21, 1916–1924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Lakoma, A.; Barbieri, E.; Agarwal, S.; Jackson, J.; Chen, Z.; Kim, Y.; McVay, M.; Shohet, J.M.; Kim, E.S. The MDM2 small-molecule inhibitor RG7388 leads to potent tumor inhibition in p53 wild-type neuroblastoma. Cell Death Discov. 2015, 1, 15026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kim, M.; Ma, D.J.; Calligaris, D.; Zhang, S.; Feathers, R.W.; Vaubel, R.A.; Meaux, I.; Mladek, A.C.; Parrish, K.E.; Jin, F.; et al. Efficacy of the MDM2 Inhibitor SAR405838 in Glioblastoma Is Limited by Poor Distribution Across the Blood–Brain Barrier. Mol. Cancer Ther. 2018, 17, 1893–1901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Breckwoldt, M.O.; Bode, J.; Sahm, F.; Krüwel, T.; Solecki, G.; Hahn, A.; Wirthschaft, P.; Berghoff, A.S.; Haas, M.; Venkataramani, V.; et al. Correlated MRI and ultramicroscopy (MR-UM) of brain tumors reveals vast heterogeneity of tumor infiltration and neoangiogenesis in preclinical models and human disease. Front. Neurosci. 2019, 12, 1004. [Google Scholar] [CrossRef] [Green Version]
  35. Fink, J.R.; Muzi, M.; Peck, M.; Krohn, K.A. Multimodality brain tumor imaging: MR imaging, PET, and PET/MR imaging. J. Nucl. Med. 2015, 56, 1554–1561. [Google Scholar] [CrossRef] [Green Version]
  36. Watanabe, M.; Tanaka, R.; Takeda, N. Magnetic resonance imaging and histopathology of cerebral gliomas. Neuroradiology 1992, 34, 463–469. [Google Scholar] [CrossRef]
  37. Kelly, P.J.; Daumas-Duport, C.; Kispert, D.B.; Kall, B.A.; Scheithauer, B.W.; Illig, J.J. Imaging-based stereotaxic serial biopsies in untreated intracranial glial neoplasms. J. Neurosurg. 1987, 66, 865–874. [Google Scholar] [CrossRef]
  38. Pessina, F.; Navarria, P.; Cozzi, L.; Ascolese, A.M.; Simonelli, M.; Santoro, A.; Clerici, E.; Rossi, M.; Scorsetti, M.; Bello, L. Maximize surgical resection beyond contrast-enhancing boundaries in newly diagnosed glioblastoma multiforme: Is it useful and safe? A single institution retrospective experience. J. Neuro-Oncol. 2017, 135, 129–139. [Google Scholar] [CrossRef]
  39. Sanai, N.; Berger, M.S. Glioma extent of resection and its impact on patient outcome. Neurosurgery 2008, 62, 753–766. [Google Scholar] [CrossRef] [Green Version]
  40. Brown, T.J.; Brennan, M.C.; Li, M.; Church, E.W.; Brandmeir, N.J.; Rakszawski, K.L.; Patel, A.S.; Rizk, E.B.; Suki, D.; Sawaya, R.; et al. Association of the Extent of Resection With Survival in Glioblastoma. JAMA Oncol. 2016, 2, 1460–1469. [Google Scholar] [CrossRef] [Green Version]
  41. Agarwal, S.; Mittapalli, R.K.; Zellmer, D.M.; Gallardo, J.L.; Donelson, R.; Seiler, C.; Decker, S.A.; SantaCruz, K.S.; Pokorny, J.L.; Sarkaria, J.N.; et al. Active efflux of dasatinib from the brain limits efficacy against murine glioblastoma: Broad implications for the clinical use of molecularly targeted agents. Mol. Cancer Ther. 2012, 11, 2183–2192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Sanai, N.; Li, J.; Boerner, J.; Stark, K.; Wu, J.; Kim, S.; Derogatis, A.; Mehta, S.; Dhruv, H.D.; Heilbrun, L.K.; et al. Phase 0 trial of azd1775 in first-recurrence glioblastoma patients. Clin. Cancer Res. 2018, 24, 3820–3828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Milano, M.T.; Okunieff, P.; Donatello, R.S.; Mohile, N.A.; Sul, J.; Walter, K.A.; Korones, D.N. Patterns and timing of recurrence after temozolomide-based chemoradiation for glioblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2010, 78, 1147–1155. [Google Scholar] [CrossRef]
  44. Osswald, M.; Blaes, J.; Liao, Y.; Solecki, G.; Gömmel, M.; Berghoff, A.S.; Salphati, L.; Wallin, J.J.; Phillips, H.S.; Wick, W.; et al. Impact of blood-brain barrier integrity on tumor growth and therapy response in brain metastases. Clin. Cancer Res. 2016, 22, 6078–6087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Salphati, L.; Heffron, T.P.; Alicke, B.; Nishimura, M.; Barck, K.; Carano, R.A.; Cheong, J.; Edgar, K.A.; Greve, J.; Kharbanda, S.; et al. Targeting the PI3K pathway in the brain—Efficacy of a PI3K inhibitor optimized to cross the blood-brain barrier. Clin. Cancer Res. 2012, 18, 6239–6248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ballard, P.; Yates, J.W.T.; Yang, Z.; Kim, D.W.; Yang, J.C.H.; Cantarini, M.; Pickup, K.; Jordan, A.; Hickey, M.; Grist, M.; et al. Preclinical comparison of osimertinib with other EGFR-TKIs in EGFR-mutant NSCLC brain metastases models, and early evidence of clinical brain metastases activity. Clin. Cancer Res. 2016, 22, 5130–5140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Reungwetwattana, T.; Nakagawa, K.; Cho, B.C.; Cobo, M.; Cho, E.K.; Bertolini, A.; Bohnet, S.; Zhou, C.; Lee, K.H.; Nogami, N.; 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, 36, 3290–3297. [Google Scholar] [CrossRef]
  48. Fowler, M.J.; Cotter, J.D.; Knight, B.E.; Sevick-Muraca, E.M.; Sandberg, D.I.; Sirianni, R.W. Intrathecal drug delivery in the era of nanomedicine. Adv. Drug Deliv. Rev. 2020. [Google Scholar] [CrossRef]
  49. Costa, R.; Kumthekar, P. Management of central nervous system metastases in breast cancer. In The Breast: Comprehensive Management of Benign and Malignant Diseases; Elsevier Inc.: Amsterdam, The Netherlands, 2018; pp. 942–960.e7. ISBN 9780323359559. [Google Scholar]
  50. Ammaya, A.K. Subcutaneous Reservoir and Pump For Sterile Access to Ventricular Cerebrospinal Fluid. Lancet 1963, 186, 983–984. [Google Scholar] [CrossRef]
  51. Witorsch, P.; Williams, T.W.; Ommaya, A.K.; Utz, J.P. Intraventricular Administration of Amphotericin B: Use of Subcutaneous Reservoir in Four Patients With Mycotic Meningitis. JAMA J. Am. Med. Assoc. 1965, 194, 699–702. [Google Scholar] [CrossRef]
  52. Ozerov, S.; Thomale, U.W.; Schulz, M.; Schaumann, A.; Samarin, A.; Kumirova, E. The use of a smartphone-assisted ventricle catheter guide for Ommaya reservoir placement—Experience of a retrospective bi-center study. Child’s Nerv. Syst. 2018, 34, 853–859. [Google Scholar] [CrossRef] [PubMed]
  53. Malani, R.; Fleisher, M.; Kumthekar, P.; Lin, X.; Omuro, A.; Groves, M.D.; Lin, N.U.; Melisko, M.; Lassman, A.B.; Jeyapalan, S.; et al. Cerebrospinal fluid circulating tumor cells as a quantifiable measurement of leptomeningeal metastases in patients with HER2 positive cancer. J. Neuro-Oncol. 2020, 148, 599–606. [Google Scholar] [CrossRef] [PubMed]
  54. Pluchart, H.; Jacquet, E.; Charlety, D.; Allenet, B.; Bedouch, P.; Mousseau, M. Long-Term Survivor with Intrathecal and Intravenous Trastuzumab Treatment in Metastatic Breast Cancer. Target. Oncol. 2016, 11, 687–691. [Google Scholar] [CrossRef] [PubMed]
  55. Lu, N.T.; Raizer, J.; Gabor, E.P.; Liu, N.M.; Vu, J.Q.; Slamon, D.J.; Barstis, J.L. Intrathecal trastuzumab: Immunotherapy improves the prognosis of leptomeningeal metastases in HER-2+ breast cancer patient. J. Immunother. Cancer 2015, 3, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Gulia, S.; Gupta, S.; Singh, A. Intrathecal trastuzumab for leptomeningeal carcinomatosis in patients with human epidermal growth factor receptor 2 positive breast cancer. Indian J. Med. Paediatr. Oncol. 2016, 37, 196–198. [Google Scholar] [CrossRef] [Green Version]
  57. Figura, N.B.; Long, W.; Yu, M.; Robinson, T.J.; Mokhtari, S.; Etame, A.B.; Tran, N.D.; Diaz, R.; Soliman, H.; Han, H.S.; et al. Intrathecal trastuzumab in the management of HER2+ breast leptomeningeal disease: A single institution experience. Breast Cancer Res. Treat. 2018, 169, 391–396. [Google Scholar] [CrossRef]
  58. García, F.J.V.; Carrión, N.P.; de la Cruz-Merino, L. Long-term complete response to intrathecal trastuzumab in a patient with leptomeningeal carcinomatosis due to her2- overexpressing breast cancer. Medicine 2020, 99, e18298. [Google Scholar] [CrossRef]
  59. Zagouri, F.; Zoumpourlis, P.; Le Rhun, E.; Bartsch, R.; Zografos, E.; Apostolidou, K.; Dimopoulos, M.-A.; Preusser, M. Intrathecal administration of anti-HER2 treatment for the treatment of meningeal carcinomatosis in breast cancer: A metanalysis with meta-regression. Cancer Treat. Rev. 2020, 88, 102046. [Google Scholar] [CrossRef]
  60. Mrugala, M.M.; Kim, B.; Sharma, A.; Johnson, N.; Graham, C.; Kurland, B.F.; Gralow, J. Phase II Study of Systemic High-dose Methotrexate and Intrathecal Liposomal Cytarabine for Treatment of Leptomeningeal Carcinomatosis From Breast Cancer. Clin. Breast Cancer 2019, 19, 311–316. [Google Scholar] [CrossRef]
  61. Scott, B.J.; Oberheim-Bush, N.A.; Kesari, S. Leptomeningeal metastasis in breast cancer-a systematic review. Oncotarget 2016, 7, 3740–3747. [Google Scholar] [CrossRef] [Green Version]
  62. Taillibert, S.; Chamberlain, M.C. Leptomeningeal Metastasis. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2018; Volume 149, pp. 169–204. [Google Scholar]
  63. Niwińska, A.; Rudnicka, H.; Murawska, M. Breast cancer leptomeningeal metastasis: The results of combined treatment and the comparison of methotrexate and liposomal cytarabine as intra-cerebrospinal fluid chemotherapy. Clin. Breast Cancer 2015, 15, 66–72. [Google Scholar] [CrossRef] [PubMed]
  64. Pan, Z.; Yang, G.; He, H.; Zhao, G.; Yuan, T.; Li, Y.; Shi, W.; Gao, P.; Dong, L.; Li, Y. Concurrent radiotherapy and intrathecal methotrexate for treating leptomeningeal metastasis from solid tumors with adverse prognostic factors: A prospective and single-arm study. Int. J. Cancer 2016, 139, 1864–1872. [Google Scholar] [CrossRef] [PubMed]
  65. Assi, H.I.; Mahmoud, T.; Saadeh, F.S.; El Darsa, H. Management of leptomeningeal metastasis in breast cancer. Clin. Neurol. Neurosurg. 2018, 172, 151–159. [Google Scholar] [CrossRef] [PubMed]
  66. Scott, B.J.; van Vugt, V.A.; Rush, T.; Brown, T.; Chen, C.C.; Carter, B.S.; Schwab, R.; Fanta, P.; Helsten, T.; Bazhenova, L.; et al. Concurrent intrathecal methotrexate and liposomal cytarabine for leptomeningeal metastasis from solid tumors: A retrospective cohort study. J. Neuro-Oncol. 2014, 119, 361–368. [Google Scholar] [CrossRef]
  67. Rothwell, W.T.; Bell, P.; Richman, L.K.; Limberis, M.P.; Tretiakova, A.P.; Li, M.; Wilson, J.M. Intrathecal viral vector delivery of trastuzumab prevents or inhibits tumor growth of human HER2-positive xenografts in mice. Cancer Res. 2018, 78, 6171–6182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Gutova, M.; Flores, L.; Adhikarla, V.; Tsaturyan, L.; Tirughana, R.; Aramburo, S.; Metz, M.; Gonzaga, J.; Annala, A.; Synold, T.W.; et al. Quantitative Evaluation of Intraventricular Delivery of Therapeutic Neural Stem Cells to Orthotopic Glioma. Front. Oncol. 2019, 9, 68. [Google Scholar] [CrossRef] [Green Version]
  69. Householder, K.T.; Dharmaraj, S.; Sandberg, D.I.; Wechsler-Reya, R.J.; Sirianni, R.W. Fate of nanoparticles in the central nervous system after intrathecal injection in healthy mice. Sci. Rep. 2019, 9, 1–11. [Google Scholar] [CrossRef] [Green Version]
  70. Chen, Y.M.; Chen, M.C.; Tsai, C.M.; Perng, R.P. Intrathecal gemcitabine chemotherapy for non-small cell lung cancer patients with meningeal carcinomatosis—A case report. Lung Cancer 2003, 40, 99–101. [Google Scholar] [CrossRef]
  71. Jabbour, E.; O’Brien, S.; Kantarjian, H.; Garcia-Manero, G.; Ferrajoli, A.; Ravandi, F.; Cabanillas, M.; Thomas, D.A. Neurologic complications associated with intrathecal liposomal cytarabine given prophylactically in combination with high-dose methotrexate and cytarabine to patients with acute lymphocytic leukemia. Blood 2007, 109, 3214–3218. [Google Scholar] [CrossRef]
  72. Pan, Y.; Wang, C.; Wang, H.; Tao, Q.; Xiong, S.; Zhai, Z. Transverse myelopathy occurring with intrathecal administration of methotrexate and cytarabine chemotherapy: A case report. Oncol. Lett. 2016, 11, 4066–4068. [Google Scholar] [CrossRef] [Green Version]
  73. Chotsampancharoen, T.; Sripornsawan, P.; Wongchanchailert, M. Two Fatal Cases of Accidental Intrathecal Vincristine Administration: Learning from Death Events. Chemotherapy 2016, 61, 108–110. [Google Scholar] [CrossRef] [PubMed]
  74. Nair, A. Implications of Intrathecal Chemotherapy for Anaesthesiologists: A Brief Review. Scientifica 2016, 2016, 3759845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Partap, S.; Murphy, P.A.; Vogel, H.; Barnes, P.D.; Edwards, M.S.B.; Fisher, P.G. Liposomal cytarabine for central nervous system embryonal tumors in children and young adults. J. Neuro-Oncol. 2011, 103, 561–566. [Google Scholar] [CrossRef] [PubMed]
  76. Ferguson, R.K.; Woodbury, D.M. Penetration of 14C-inulin and 14C-sucrose into brain, cerebrospinal fluid, and skeletal muscle of developing rats. Exp. Brain Res. 1969, 7, 181–194. [Google Scholar] [CrossRef]
  77. Bergman, I.; Burckart, G.; Pohi, C.R.; Venkataramanan, R.; Barmada, M.A.; Griffin, J.A.; Cheung, N.K. Pharmacokinetics of IgG and IgM anti-ganglioside antibodies in rats and monkeys after intrathecal administration. ASPET 1998, 284, 111–115. [Google Scholar]
  78. Collins, J.M. Pharmacokinetics of intraventricular administration. J. Neuro-Oncol. 1983, 1, 283–291. [Google Scholar] [CrossRef]
  79. Pardridge, W.M. CSF, blood-brain barrier, and brain drug delivery. Expert Opin. Drug Deliv. 2016, 13, 963–975. [Google Scholar] [CrossRef]
  80. Hunt Bobo, R.; Laske, D.W.; Akbasak, A.; Morrison, P.F.; Dedrick, R.L.; Oldfield, E.H. Convection-enhanced delivery of macromolecules in the brain. Proc. Natl. Acad. Sci. USA 1994, 91, 2076–2080. [Google Scholar] [CrossRef] [Green Version]
  81. Mehta, A.M.; Sonabend, A.M.; Bruce, J.N. Convection-Enhanced Delivery. Neurotherapeutics 2017, 14, 358–371. [Google Scholar] [CrossRef] [Green Version]
  82. Saka, R.; Sathe, P.; Khan, W. Brain local delivery strategy. In Brain Targeted Drug Delivery System; Elsevier: Amsterdam, The Netherlands, 2019; pp. 241–286. [Google Scholar]
  83. Tosi, U.; Kommidi, H.; Adeuyan, O.; Guo, H.; Maachani, U.B.; Chen, N.; Su, T.; Zhang, G.; Pisapia, D.J.; Dahmane, N.; et al. PET, image-guided HDAC inhibition of pediatric diffuse midline glioma improves survival in murine models. Sci. Adv. 2020, 6, eabb4105. [Google Scholar] [CrossRef]
  84. Pang, H.H.; Chen, P.Y.; Wei, K.C.; Huang, C.W.; Shiue, Y.L.; Huang, C.Y.; Yang, H.W. Convection-enhanced delivery of a virus-like nanotherapeutic agent with dual-modal imaging for besiegement and eradication of brain tumors. Theranostics 2019, 9, 1752–1763. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, C.; Nance, E.A.; Mastorakos, P.; Chisholm, J.; Berry, S.; Eberhart, C.; Tyler, B.; Brem, H.; Suk, J.S.; Hanes, J. Convection enhanced delivery of cisplatin-loaded brain penetrating nanoparticles cures malignant glioma in rats. J. Control. Release 2017, 263, 112–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Jahangiri, A.; Chin, A.T.; Flanigan, P.M.; Chen, R.; Bankiewicz, K.; Aghi, M.K. Convection-enhanced delivery in glioblastoma: A review of preclinical and clinical studies. J. Neurosurg. 2017, 126, 191–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Yokosawa, M.; Sonoda, Y.; Sugiyama, S.; Saito, R.; Yamashita, Y.; Nishihara, M.; Satoh, T.; Kumabe, T.; Yokoyama, M.; Tominaga, T. Convection-enhanced delivery of a synthetic retinoid Am80, loaded into polymeric micelles, prolongs the survival of rats bearing intracranial glioblastoma xenografts. Tohoku J. Exp. Med. 2010, 221, 257–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Grahn, A.Y.; Bankiewicz, K.S.; Dugich-Djordjevic, M.; Bringas, J.R.; Hadaczek, P.; Johnson, G.A.; Eastman, S.; Luz, M. Non-PEGylated liposomes for convection-enhanced delivery of topotecan and gadodiamide in malignant glioma: Initial experience. J. Neuro-Oncol. 2009, 95, 185–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Lin, C.Y.; Li, R.J.; Huang, C.Y.; Wei, K.C.; Chen, P.Y. Controlled release of liposome-encapsulated temozolomide for brain tumour treatment by convection-enhanced delivery. J. Drug Target. 2018, 26, 325–332. [Google Scholar] [CrossRef]
  90. Nordling-David, M.M.; Yaffe, R.; Guez, D.; Meirow, H.; Last, D.; Grad, E.; Salomon, S.; Sharabi, S.; Levi-Kalisman, Y.; Golomb, G.; et al. Liposomal temozolomide drug delivery using convection enhanced delivery. J. Control. Release 2017, 261, 138–146. [Google Scholar] [CrossRef]
  91. Zhang, R.; Saito, R.; Mano, Y.; Sumiyoshi, A.; Kanamori, M.; Sonoda, Y.; Kawashima, R.; Tominaga, T. Convection-enhanced delivery of SN-38-loaded polymeric micelles (NK012) enables consistent distribution of SN-38 and is effective against rodent intracranial brain tumor models. Drug Deliv. 2016, 23, 2780–2786. [Google Scholar] [CrossRef] [Green Version]
  92. Rechberger, J.S.; Power, E.A.; Lu, V.M.; Zhang, L.; Sarkaria, J.N.; Daniels, D.J. Evaluating infusate parameters for direct drug delivery to the brainstem: A comparative study of convection-enhanced delivery versus osmotic pump delivery. Neurosurg. Focus 2020, 48. [Google Scholar] [CrossRef] [Green Version]
  93. Zhan, W.; Wang, C.H. Convection enhanced delivery of liposome encapsulated doxorubicin for brain tumour therapy. J. Control. Release 2018, 285, 212–229. [Google Scholar] [CrossRef]
  94. Zhan, W.; Arifin, D.Y.; Lee, T.K.; Wang, C.H. Mathematical Modelling of Convection Enhanced Delivery of Carmustine and Paclitaxel for Brain Tumour Therapy. Pharm. Res. 2017, 34, 860–873. [Google Scholar] [CrossRef] [PubMed]
  95. Singleton, W.G.B.; Bieneman, A.S.; Woolley, M.; Johnson, D.; Lewis, O.; Wyatt, M.J.; Damment, S.J.P.; Boulter, L.J.; Killick-Cole, C.L.; Asby, D.J.; et al. The distribution, clearance, and brainstem toxicity of panobinostat administered by convection-enhanced delivery. Proc. J. Neurosurg. Pediatrics Am. Assoc. Neurol. Surg. 2018, 22, 288–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. MTX110 by Convection-Enhanced Delivery in Treating Participants with Newly-Diagnosed Diffuse Intrinsic Pontine Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03566199?term=convection+enhanced+delivery&draw=2&rank=1 (accessed on 21 September 2020).
  97. Chronic Convection Enhanced Delivery of Topotecan-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03154996?term=convection+enhanced+delivery&draw=2&rank=2 (accessed on 21 September 2020).
  98. Intratumorally-Administered Topotecan Using Convection-Enhanced Delivery in Patients With Grade III/IV Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03927274?term=convection+enhanced+delivery&draw=2&rank=4 (accessed on 21 September 2020).
  99. Topotecan Using Convection-Enhanced Delivery (CED) in High Grade Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02278510 (accessed on 3 October 2020).
  100. Vogelbaum, M.A.; Brewer, C.; Barnett, G.H.; Mohammadi, A.M.; Peereboom, D.M.; Ahluwalia, M.S.; Gao, S. First-in-human evaluation of the Cleveland Multiport Catheter for convection-enhanced delivery of topotecan in recurrent high-grade glioma: Results of pilot trial 1. J. Neurosurg. 2018, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Safety Study of Intracerebral Topotecan for Recurrent Brain Tumors-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00308165?term=convection+enhanced+delivery&draw=3&rank=17 (accessed on 21 September 2020).
  102. CED With Irinotecan Liposome Injection Using Real Time Imaging in Children with Diffuse Intrinsic Pontine Glioma (DIPG) (PNOC 009)-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03086616?term=convection+enhanced+delivery&draw=2&rank=3 (accessed on 21 September 2020).
  103. Convection-Enhanced Delivery of 124I-Omburtamab for Patients With Non-Progressive Diffuse Pontine Gliomas Previously Treated With External Beam Radiation Therapy-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT01502917?term=convection+enhanced+delivery&draw=2&rank=5 (accessed on 21 September 2020).
  104. CED of MTX110 Newly Diagnosed Diffuse Midline Gliomas-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04264143?term=convection+enhanced+delivery&draw=2&rank=6 (accessed on 21 September 2020).
  105. Carboplatin in Treating Patients With Recurrent High-Grade Gliomas-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT01644955?term=convection+enhanced+delivery&draw=2&rank=7 (accessed on 21 September 2020).
  106. Zacks Small Cap Research-MDNA.TO: New Data for MDNA55 and MDNA11 Presented at ASCO 2020. Available online: https://scr.zacks.com/News/Press-Releases/Press-Release-Details/2020/MDNATO-New-Data-for-MDNA55-and-MDNA11-Presented-at-ASCO-2020/default.aspx (accessed on 21 September 2020).
  107. Randazzo, D.; Achrol, A.; Aghi, M.K.; Bexon, M.; Brem, S.; Brenner, A.J.; Butowski, N.A.; Chandhasin, C.; Chowdhary, S.A.; Coello, M.; et al. MDNA55: A locally administered IL4 guided toxin as a targeted treatment for recurrent glioblastoma. J. Clin. Oncol. 2019, 37, 2039. [Google Scholar] [CrossRef]
  108. Convection-Enhanced Delivery (CED) of MDNA55 in Adults with Recurrent or Progressive Glioblastoma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02858895?term=convection+enhanced+delivery&draw=2&rank=10 (accessed on 21 September 2020).
  109. Chittiboina, P.; Heiss, J.D.; Warren, K.E.; Lonser, R.R. Magnetic resonance imaging properties of convective delivery in diffuse intrinsic pontine gliomas: Clinical article. J. Neurosurg. Pediatr. 2014, 13, 273–275. [Google Scholar] [CrossRef]
  110. An Open Label Dose Escalation Safety Study of Convection-Enhanced Delivery of IL13-PE38QQR in Patients with Progressive Pediatric Diffuse Infiltrating Brainstem Glioma and Supratentorial High-Grade Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00880061?term=convection+enhanced+delivery&draw=3&rank=11 (accessed on 21 September 2020).
  111. Study of Convection-Enhanced, Image-Assisted Delivery of Liposomal-Irinotecan In Recurrent High Grade Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02022644?term=convection+enhanced+delivery&draw=3&rank=18 (accessed on 21 September 2020).
  112. Kunwar, S.; Prados, M.D.; Chang, S.M.; Berger, M.S.; Lang, F.F.; Piepmeier, J.M.; Sampson, J.H.; Ram, Z.; Gutin, P.H.; Gibbons, R.D.; et al. Direct intracerebral delivery of cintredekin besudotox (IL13-PE38QQR) in recurrent malignant glioma: A report by the cintredekin besudotox intraparenchymal study group. J. Clin. Oncol. 2007, 25, 837–844. [Google Scholar] [CrossRef]
  113. IL13-PE38QQR Infusion After Tumor Resection, Followed by Radiation Therapy With or Without Temozolomide in Patients With Newly Diagnosed Malignant Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00089427 (accessed on 3 October 2020).
  114. Safety and Efficacy Study to Treat Recurrent Grade 4 Malignant Brain Tumors-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00104091?term=convection+enhanced+delivery&draw=5&rank=34 (accessed on 21 September 2020).
  115. Maximum Tolerated Dose, Safety, and Efficacy of Rhenium Nanoliposomes in Recurrent Glioma (ReSPECT)-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT01906385?term=convection+enhanced+delivery&draw=5&rank=31 (accessed on 21 September 2020).
  116. Lang, F.F.; Conrad, C.; Gomez-Manzano, C.; Alfred Yung, W.K.; Sawaya, R.; Weinberg, J.S.; Prabhu, S.S.; Rao, G.; Fuller, G.N.; Aldape, K.D.; et al. Phase I study of DNX-2401 (delta-24-RGD) oncolytic adenovirus: Replication and immunotherapeutic effects in recurrent malignant glioma. J. Clin. Oncol. 2018, 36, 1419–1427. [Google Scholar] [CrossRef]
  117. Safety Study of Replication-Competent Adenovirus (Delta-24-Rgd) in Patients with Recurrent Glioblastoma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT01582516?term=convection+enhanced+delivery&draw=4&rank=22 (accessed on 21 September 2020).
  118. A Dose Escalation Phase I Study Of Human-Recombinant Bone Morphogenetic Protein 4 Administrated Via CED In GBM Patients-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02869243?term=convection+enhanced+delivery&draw=4&rank=23 (accessed on 21 September 2020).
  119. Kunwar, S.; Chang, S.; Westphal, M.; Vogelbaum, M.; Sampson, J.; Barnett, G.; Shaffrey, M.; Ram, Z.; Piepmeier, J.; Prados, M.; et al. Phase III randomized trial of CED of IL13-PE38QQR vs Gliadel wafers for recurrent glioblastoma. Neuro-Oncology 2010, 12, 871–881. [Google Scholar] [CrossRef] [Green Version]
  120. The PRECISE Trial: Study of IL13-PE38QQR Compared to GLIADEL Wafer in Patients With Recurrent Glioblastoma Multiforme-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00076986?term=convection+enhanced+delivery&draw=4&rank=25 (accessed on 21 September 2020).
  121. Phase 1 Trial of D2C7-IT in Combination With 2141-V11 for Recurrent Malignant Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04547777?term=convection+enhanced+delivery&draw=4&rank=26 (accessed on 21 September 2020).
  122. D2C7-IT With Atezolizumab for Recurrent Gliomas-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04160494?term=convection+enhanced+delivery&draw=4&rank=29 (accessed on 21 September 2020).
  123. D2C7 for Adult Patients With Recurrent Malignant Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02303678?term=convection+enhanced+delivery&draw=4&rank=21 (accessed on 21 September 2020).
  124. Phase 1b Study PVSRIPO for Recurrent Malignant Glioma in Children-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03043391?term=convection+enhanced+delivery&draw=4&rank=28 (accessed on 21 September 2020).
  125. PVSRIPO for Recurrent Glioblastoma (GBM)-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT01491893?term=convection+enhanced+delivery&draw=4&rank=30 (accessed on 21 September 2020).
  126. PVSRIPO and Pembrolizumab in Patients With Recurrent Glioblastoma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04479241?term=convection+enhanced+delivery&draw=5&rank=36 (accessed on 21 September 2020).
  127. Available online: http://oncotelic.com/wp-content/uploads/AACR-2019-GBM-Final-For-Web.pdf (accessed on 21 September 2020).
  128. Phase IIb Clinical Trial With TGF-β2 Antisense Compound AP 12009 for Recurrent or Refractory High-Grade Glioma-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00431561?term=convection+enhanced+delivery&draw=5&rank=38 (accessed on 21 September 2020).
  129. Uckun, F.M.; Qazi, S.; Hwang, L.; Trieu, V.N. Recurrent or refractory high-grade gliomas treated by convection-enhanced delivery of a TGFΒ2-targeting RNA therapeutic: A post-hoc analysis with long-term follow-up. Cancers 2019, 11, 1892. [Google Scholar] [CrossRef] [Green Version]
  130. FDA. GLIADEL® WAFER (Carmustine Implant), for Intracranial Use. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/020637s029lbl.pdf (accessed on 21 September 2020).
  131. Brem, H.; Mahaley, M.S.; Vick, N.A.; Black, K.L.; Schold, S.C.; Burger, P.C.; Friedman, A.H.; Ciric, I.S.; Eller, T.W.; Cozzens, J.W.; et al. Interstitial chemotherapy with drug polymer implants for the treatment of recurrent gliomas. J. Neurosurg. 1991, 74, 441–446. [Google Scholar] [CrossRef]
  132. Ashby, L.S.; Smith, K.A.; Stea, B. Gliadel wafer implantation combined with standard radiotherapy and concurrent followed by adjuvant temozolomide for treatment of newly diagnosed high-grade glioma: A systematic literature review. World J. Surg. Oncol. 2016, 14, 225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Sasaki, T.; Fukai, J.; Kodama, Y.; Hirose, T.; Okita, Y.; Moriuchi, S.; Nonaka, M.; Tsuyuguchi, N.; Terakawa, Y.; Uda, T.; et al. Characteristics and outcomes of elderly patients with diffuse gliomas: A multi-institutional cohort study by Kansai Molecular Diagnosis Network for CNS Tumors. J. Neuro-Oncol. 2018, 140, 329–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Wait, S.D.; Prabhu, R.S.; Burri, S.H.; Atkins, T.G.; Asher, A.L. Polymeric drug delivery for the treatment of glioblastoma. Neuro. Oncol. 2015, 17, ii9–ii23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Lechapt-Zalcman, E.; Levallet, G.; Dugué, A.E.; Vital, A.; Diebold, M.D.; Menei, P.; Colin, P.; Peruzzy, P.; Emery, E.; Bernaudin, M.; et al. O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation and low MGMT-encoded protein expression as prognostic markers in glioblastoma patients treated with biodegradable carmustine wafer implants after initial surgery followed by radiotherapy with concomitant and adjuvant temozolomide. Cancer 2012, 118, 4545–4554. [Google Scholar] [CrossRef] [PubMed]
  136. Metellus, P.; Coulibaly, B.; Nanni, I.; Fina, F.; Eudes, N.; Giorgi, R.; Barrie, M.; Chinot, O.; Fuentes, S.; Dufour, H.; et al. Prognostic impact of O6-methylguanine-DNA methyltransferase silencing in patients with recurrent glioblastoma multiforme who undergo surgery and carmustine wafer implantation: A prospective patient cohort. Cancer 2009, 115, 4783–4794. [Google Scholar] [CrossRef]
  137. Gutenberg, A.; Bock, H.C.; Brück, W.; Doerner, L.; Mehdorn, H.M.; Roggendorf, W.; Westphal, M.; Felsberg, J.; Reifenberger, G.; Giese, A. MGMT promoter methylation status and prognosis of patients with primary or recurrent glioblastoma treated with carmustine wafers. Br. J. Neurosurg. 2013, 27, 772–778. [Google Scholar] [CrossRef]
  138. Ducray, F.; Honnorat, J. New adjuvant chemotherapy for glioblastoma. Press. Medicale 2007, 36, 1249–1254. [Google Scholar] [CrossRef]
  139. Fukai, J.; Nishibayashi, H.; Uematsu, Y.; Kanemura, Y.; Fujita, K.; Nakao, N. Rapid regression of glioblastoma following carmustine wafer implantation: A case report. Mol. Clin. Oncol. 2016, 5, 153–157. [Google Scholar] [CrossRef] [Green Version]
  140. Sippl, C.; Ketter, R.; Braun, L.; Teping, F.; Schoeneberger, L.; Kim, Y.J.; List, M.; Nakhoda, A.; Wemmert, S.; Oertel, J.; et al. miRNA-26a expression influences the therapy response to carmustine wafer implantation in patients with glioblastoma multiforme. Acta Neurochir. 2019, 161, 2299–2309. [Google Scholar] [CrossRef]
  141. Shapira-Furman, T.; Serra, R.; Gorelick, N.; Doglioli, M.; Tagliaferri, V.; Cecia, A.; Peters, M.; Kumar, A.; Rottenberg, Y.; Langer, R.; et al. Biodegradable wafers releasing Temozolomide and Carmustine for the treatment of brain cancer. J. Control. Release 2019, 295, 93–101. [Google Scholar] [CrossRef]
  142. Lee, J.; Cho, H.R.; Cha, G.D.; Seo, H.; Lee, S.; Park, C.K.; Kim, J.W.; Qiao, S.; Wang, L.; Kang, D.; et al. Flexible, sticky, and biodegradable wireless device for drug delivery to brain tumors. Nat. Commun. 2019, 10, 1–9. [Google Scholar] [CrossRef] [PubMed]
  143. Santini, J.T.; Cima, M.J.; Langer, R. A controlled-release microchip. Nature 1999, 397, 335–338. [Google Scholar] [CrossRef] [PubMed]
  144. Frazier, J.L.; Wang, P.P.; Case, D.; Tyler, B.M.; Pradilla, G.; Weingart, J.D.; Brem, H. Local delivery of minocycline and systemic BCNU have synergistic activity in the treatment of intracranial glioma. J. Neuro-Oncol. 2003, 64, 203–209. [Google Scholar] [CrossRef] [PubMed]
  145. Bow, H.; Hwang, L.S.; Schildhaus, N.; Xing, J.; Murray, L.; Salditch, Q.; Ye, X.; Zhang, Y.; Weingart, J.; Brem, H.; et al. Local delivery of angiogenesis-inhibitor minocycline combined with radiotherapy and oral temozolomide chemotherapy in 9L glioma: Laboratory investigation. J. Neurosurg. 2014, 120, 662–669. [Google Scholar] [CrossRef] [PubMed]
  146. Weingart, J.D.; Sipos, E.P.; Brem, H. The role of minocycline in the treatment of intracranial 9L glioma. J. Neurosurg. 1995, 82, 635–640. [Google Scholar] [CrossRef] [PubMed]
  147. Li, K.W.; Dang, W.; Tyler, B.M.; Troiano, G.; Tihan, T.; Brem, H.; Walter, K.A. Polilactofate microspheres for paclitaxel delivery to central nervous system malignancies. Clin. Cancer Res. 2003, 9, 3441–3447. [Google Scholar]
  148. Graham-Gurysh, E.; Moore, K.M.; Satterlee, A.B.; Sheets, K.T.; Lin, F.C.; Bachelder, E.M.; Miller, C.R.; Hingtgen, S.D.; Ainslie, K.M. Sustained Delivery of Doxorubicin via Acetalated Dextran Scaffold Prevents Glioblastoma Recurrence after Surgical Resection. Mol. Pharm. 2018, 15, 1309–1318. [Google Scholar] [CrossRef]
  149. Yuan, X.; Dillehay, L.E.; Williams, J.R.; Williams, J.A. Synthetic, implantable polymers for IUdR radiosensitization of experimental human malignant glioma. Cancer Biother. Radiopharm. 1999, 14, 187–202. [Google Scholar] [CrossRef]
  150. Yuan, X.; Tabassi, K.; Williams, J.A. Implantable polymers for tirapazamine treatments of experimental intracranial malignant glioma. Radiat. Oncol. Investig. 1999, 7, 218–230. [Google Scholar] [CrossRef]
  151. Ranganath, S.H.; Wang, C.H. Biodegradable microfiber implants delivering paclitaxel for post-surgical chemotherapy against malignant glioma. Biomaterials 2008, 29, 2996–3003. [Google Scholar] [CrossRef]
  152. Fleming, A.B.; Saltzman, W.M. Pharmacokinetics of the carmustine implant. Clin. Pharmacokinet. 2002, 41, 403–419. [Google Scholar] [CrossRef] [PubMed]
  153. Graham-Gurysh, E.G.; Murthy, A.B.; Moore, K.M.; Hingtgen, S.D.; Bachelder, E.M.; Ainslie, K.M. Synergistic drug combinations for a precision medicine approach to interstitial glioblastoma therapy. J. Control. Release 2020, 323, 282–292. [Google Scholar] [CrossRef] [PubMed]
  154. Storm, P.B.; Moriarity, J.L.; Tyler, B.; Burger, P.C.; Brem, H.; Weingart, J. Polymer delivery of camptothecin against 9L gliosarcoma: Release, distribution, and efficacy. J. Neuro-Oncol. 2002, 56, 209–217. [Google Scholar] [CrossRef] [PubMed]
  155. Sampath, P.; Amundson, E.; Wall, M.E.; Tyler, B.M.; Wani, M.C.; Alderson, L.M.; Colvin, M.; Brem, H.; Weingart, J.D. Camptothecin analogs in malignant gliomas: Comparative analysis and characterization. J. Neurosurg. 2003, 98, 570–577. [Google Scholar] [CrossRef]
  156. Sampath, P.; Rhines, L.D.; DiMeco, F.; Tyler, B.M.; Park, M.C.; Brem, H. Interstitial docetaxel (Taxotere), ccarmustine and ccombined interstitial ttherapy: A novel treatment for experimental malignant glioma. J. Neuro-Oncol. 2006, 80, 9–17. [Google Scholar] [CrossRef] [Green Version]
  157. Shibahara, I.; Hanihara, M.; Watanabe, T.; Dan, M.; Sato, S.; Kuroda, H.; Inamura, A.; Inukai, M.; Hara, A.; Yasui, Y.; et al. Tumor microenvironment after biodegradable BCNU wafer implantation: Special consideration of immune system. J. Neuro-Oncol. 2018, 137, 417–427. [Google Scholar] [CrossRef]
  158. Wolinsky, J.B.; Colson, Y.L.; Grinstaff, M.W. Local drug delivery strategies for cancer treatment: Gels, nanoparticles, polymeric films, rods, and wafers. J. Control. Release 2012, 159, 14–26. [Google Scholar] [CrossRef] [Green Version]
  159. Thiagarajah, J.R.; Kim, J.K.; Magzoub, M.; Verkman, A.S. Slowed diffusion in tumors revealed by microfiberoptic epifluorescence photobleaching. Nat. Methods 2006, 3, 275–280. [Google Scholar] [CrossRef]
  160. Rapoport, S.I.; Hori, M.; Klatzo, I. Testing of a hypothesis for osmotic opening of the blood-brain barrier. Am. J. Physiol. 1972, 223, 323–331. [Google Scholar] [CrossRef] [Green Version]
  161. Neuwelt, E.A. Reversible osmotic blood-brain barrier disruption in humans: Implications for the chemotherapy of malignant brain tumors. Neurosurgery 1980, 7, 204. [Google Scholar] [CrossRef]
  162. Kroll, R.A.; Neuwelt, E.A. Outwitting the blood-brain barrier for therapeutic purposes: Osmotic opening and other means. Neurosurgery 1998, 42, 1083–1100. [Google Scholar] [CrossRef] [PubMed]
  163. Levin, A.B.; Duff, T.A.; Javid, M.J. Treatment of increased intracranial pressure: A comparison of different hyperosmotic agents and the use of thiopental. Neurosurgery 1979, 5, 570–575. [Google Scholar] [CrossRef] [PubMed]
  164. Rapoport, S.I. Osmotic opening of the blood-brain barrier: Principles, mechanism, and therapeutic applications. Cell. Mol. Neurobiol. 2000, 20, 217–230. [Google Scholar] [CrossRef] [PubMed]
  165. Kenta, A. Sex differences in Drosophila behavior: Qualitative and quantitative dimorphism. Curr. Opin. Physiol. 2018, 6, 35–45. [Google Scholar] [CrossRef]
  166. Neuwelt, E.A.; Goldman, D.L.; Dahlborg, S.A.; Crossen, J.; Ramsey, F.; Roman-Goldstein, S.; Braziel, R.; Dana, B. Primary CNS lymphoma treated with osmotic blood-brain barrier disruption: Prolonged survival and preservation of cognitive function. J. Clin. Oncol. 1991, 9, 1580–1590. [Google Scholar] [CrossRef]
  167. Angelov, L.; Doolittle, N.D.; Kraemer, D.F.; Siegal, T.; Barnett, G.H.; Peereboom, D.M.; Stevens, G.; McGregor, J.; Jahnke, K.; Lacy, C.A.; et al. Blood-brain barrier disruption and intra-arterial methotrexate-based therapy for newly diagnosed primary CNS lymphoma: A multi-institutional experience. J. Clin. Oncol. 2009, 27, 3503–3509. [Google Scholar] [CrossRef] [Green Version]
  168. Doolittle, N.D.; Korfel, A.; Lubow, M.A.; Schorb, E.; Schlegel, U.; Rogowski, S.; Fu, R.; Dósa, E.; Illerhaus, G.; Kraemer, D.F.; et al. Long-term cognitive function, neuroimaging, and quality of life in primary CNS lymphoma. Neurology 2013, 81, 84–92. [Google Scholar] [CrossRef] [Green Version]
  169. Tyson, R.M.; Siegal, T.; Doolittle, N.D.; Lacy, C.; Kraemer, D.F.; Neuwelt, E.A. Current status and future of relapsed primary central nervous system lymphoma (PCNSL). Leuk. Lymphoma 2003, 44, 627–633. [Google Scholar] [CrossRef]
  170. Siegal, T.; Rubinstein, R.; Bokstein, F.; Schwartz, A.; Lossos, A.; Shalom, E.; Chisin, R.; Gomori, J.M. In vivo assessment of the window of barrier opening after osmotic blood- brain barrier disruption in humans. J. Neurosurg. 2000, 92, 599–605. [Google Scholar] [CrossRef] [Green Version]
  171. Rapoport, S.I.; Fredericks, W.R.; Ohno, K.; Pettigrew, K.D. Quantitative aspects of reversible osmotic opening of the blood-brain barrier. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 1980, 7, 421–431. [Google Scholar] [CrossRef]
  172. Bellavance, M.A.; Blanchette, M.; Fortin, D. Recent advances in blood-brain barrier disruption as a CNS delivery strategy. AAPS J. 2008, 10, 166–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Kemper, E.M.; Boogerd, W.; Thuis, I.; Beijnen, J.H.; van Tellingen, O. Modulation of the blood-brain barrier in oncology: Therapeutic opportunities for the treatment of brain tumours? Cancer Treat. Rev. 2004, 30, 415–423. [Google Scholar] [CrossRef] [PubMed]
  174. Doolittle, N.D.; Miner, M.E.; Hall, W.A.; Siegal, T.; Hanson, E.J.; Osztie, E.; McAllister, L.D.; Bubalo, J.S.; Kraemer, D.F.; Fortin, D.; et al. Safety and efficacy of a multicenter study using intraarterial chemotherapy in conjunction with osmotic opening of the blood-brain barrier for the treatment of patients with malignant brain tumors. Cancer 2000, 88, 637–647. [Google Scholar] [CrossRef]
  175. Sheikov, N.; McDannold, N.; Sharma, S.; Hynynen, K. Effect of Focused Ultrasound Applied With an Ultrasound Contrast Agent on the Tight Junctional Integrity of the Brain Microvascular Endothelium. Ultrasound Med. Biol. 2008, 34, 1093–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Hynynen, K.; Jolesz, F.A. Demonstration of potential noninvasive ultrasound brain therapy through an intact skull. Ultrasound Med. Biol. 1998, 24, 275–283. [Google Scholar] [CrossRef]
  177. Lipsman, N.; Meng, Y.; Bethune, A.J.; Huang, Y.; Lam, B.; Masellis, M.; Herrmann, N.; Heyn, C.; Aubert, I.; Boutet, A.; et al. Blood–brain barrier opening in Alzheimer’s disease using MR-guided focused ultrasound. Nat. Commun. 2018, 9, 1–8. [Google Scholar] [CrossRef] [Green Version]
  178. Treat, L.H.; McDannold, N.; Vykhodtseva, N.; Zhang, Y.; Tam, K.; Hynynen, K. Targeted delivery of doxorubicin to the rat brain at therapeutic levels using MRI-guided focused ultrasound. Int. J. Cancer 2007, 121, 901–907. [Google Scholar] [CrossRef]
  179. Choi, J.J.; Pernot, M.; Small, S.A.; Konofagou, E.E. Noninvasive, transcranial and localized opening of the blood-brain barrier using focused ultrasound in mice. Ultrasound Med. Biol. 2007, 33, 95–104. [Google Scholar] [CrossRef]
  180. Himuro, S. Physicochemical characteristics of microbubbles. Kagaku Koguku 2007, 71, 165–169. [Google Scholar] [CrossRef]
  181. Timbie, K.F.; Mead, B.P.; Price, R.J. Drug and gene delivery across the blood-brain barrier with focused ultrasound. J. Control. Release 2015, 219, 61–75. [Google Scholar] [CrossRef] [Green Version]
  182. Tung, Y.-S.; Vlachos, F.; Feshitan, J.A.; Borden, M.A.; Konofagou, E.E. The mechanism of interaction between focused ultrasound and microbubbles in blood-brain barrier opening in mice. J. Acoust. Soc. Am. 2011, 130, 3059–3067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Tsai, H.C.; Tsai, C.H.; Chen, W.S.; Inserra, C.; Wei, K.C.; Liu, H.L. Safety evaluation of frequent application of microbubble-enhanced focused ultrasound blood-brain-barrier opening. Sci. Rep. 2018, 8, 1–13. [Google Scholar] [CrossRef] [PubMed]
  184. Idbaih, A.; Canney, M.; Belin, L.; Desseaux, C.; Vignot, A.; Bouchoux, G.; Asquier, N.; Law-Ye, B.; Leclercq, D.; Bissery, A.; 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, 3793–3801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Carpentier, A.; Canney, M.; Vignot, A.; Reina, V.; Beccaria, K.; Horodyckid, C.; Karachi, C.; Leclercq, D.; Lafon, C.; Chapelon, J.Y.; et al. Clinical trial of blood-brain barrier disruption by pulsed ultrasound. Sci. Transl. Med. 2016, 8. [Google Scholar] [CrossRef]
  186. Mainprize, T.; Lipsman, N.; Huang, Y.; Meng, Y.; Bethune, A.; Ironside, S.; Heyn, C.; Alkins, R.; Trudeau, M.; Sahgal, A.; 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, 1–7. [Google Scholar] [CrossRef] [Green Version]
  187. Park, J.; Aryal, M.; Vykhodtseva, N.; Zhang, Y.Z.; McDannold, N. Evaluation of permeability, doxorubicin delivery, and drug retention in a rat brain tumor model after ultrasound-induced blood-tumor barrier disruption. J. Control. Release 2017, 250, 77–85. [Google Scholar] [CrossRef] [Green Version]
  188. Kovacs, Z.; Werner, B.; Rassi, A.; Sass, J.O.; Martin-Fiori, E.; Bernasconi, M. Prolonged survival upon ultrasound-enhanced doxorubicin delivery in two syngenic glioblastoma mouse models. J. Control. Release 2014, 187, 74–82. [Google Scholar] [CrossRef]
  189. Park, E.J.; Zhang, Y.Z.; Vykhodtseva, N.; McDannold, N. Ultrasound-mediated blood-brain/blood-tumor barrier disruption improves outcomes with trastuzumab in a breast cancer brain metastasis model. J. Control. Release 2012, 163, 277–284. [Google Scholar] [CrossRef] [Green Version]
  190. Kobus, T.; Zervantonakis, I.K.; Zhang, Y.; McDannold, N.J. Growth inhibition in a brain metastasis model by antibody delivery using focused ultrasound-mediated blood-brain barrier disruption. J. Control. Release 2016, 238, 281–288. [Google Scholar] [CrossRef] [Green Version]
  191. Liu, H.L.; Hsu, P.H.; Lin, C.Y.; Huang, C.W.; Chai, W.Y.; Chu, P.C.; Huang, C.Y.; Chen, P.Y.; Yang, L.Y.; Kuo, J.S.; et al. Focused ultrasound enhances central nervous system delivery of Bevacizumab for malignant glioma treatment. Radiology 2016, 281, 99–108. [Google Scholar] [CrossRef]
  192. Yang, F.Y.; Chang, W.Y.; Lin, W.T.; Hwang, J.J.; Chien, Y.C.; Wang, H.E.; Tsai, M.L. Focused ultrasound enhanced molecular imaging and gene therapy for multifusion reporter gene in glioma-bearing rat model. Oncotarget 2015, 6, 36260–36268. [Google Scholar] [CrossRef] [Green Version]
  193. Chang, E.L.; Ting, C.Y.; Hsu, P.H.; Lin, Y.C.; Liao, E.C.; Huang, C.Y.; Chang, Y.C.; Chan, H.L.; Chiang, C.S.; Liu, H.L.; et al. Angiogenesis-targeting microbubbles combined with ultrasound-mediated gene therapy in brain tumors. J. Control. Release 2017, 255, 164–175. [Google Scholar] [CrossRef] [PubMed]
  194. Treat, L.H.; McDannold, N.; Zhang, Y.; Vykhodtseva, N.; Hynynen, K. Improved Anti-Tumor Effect of Liposomal Doxorubicin After Targeted Blood-Brain Barrier Disruption by MRI-Guided Focused Ultrasound in Rat Glioma. Ultrasound Med. Biol. 2012, 38, 1716–1725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Yang, F.Y.; Wang, H.E.; Liu, R.S.; Teng, M.C.; Li, J.J.; Lu, M.; Wei, M.C.; Wong, T.T. Pharmacokinetic Analysis of 111In-Labeled Liposomal Doxorubicin in Murine Glioblastoma after Blood-Brain Barrier Disruption by Focused Ultrasound. PLoS ONE 2012, 7, 1–7. [Google Scholar] [CrossRef] [PubMed]
  196. Ting, C.Y.; Fan, C.H.; Liu, H.L.; Huang, C.Y.; Hsieh, H.Y.; Yen, T.C.; Wei, K.C.; Yeh, C.K. Concurrent blood-brain barrier opening and local drug delivery using drug-carrying microbubbles and focused ultrasound for brain glioma treatment. Biomaterials 2012, 33, 704–712. [Google Scholar] [CrossRef] [PubMed]
  197. Fan, C.H.; Ting, C.Y.; Chang, Y.C.; Wei, K.C.; Liu, H.L.; Yeh, C.K. Drug-loaded bubbles with matched focused ultrasound excitation for concurrent blood-brain barrier opening and brain-tumor drug delivery. Acta Biomater. 2015, 15, 89–101. [Google Scholar] [CrossRef]
  198. Chen, P.Y.; Liu, H.L.; Hua, M.Y.; Yang, H.W.; Huang, C.Y.; Chu, P.C.; Lyu, L.A.; Tseng, I.C.; Feng, L.Y.; Tsai, H.C.; et al. Novel magnetic/ultrasound focusing system enhances nanoparticle drug delivery for glioma treatment. Neuro. Oncol. 2010, 12, 1050–1060. [Google Scholar] [CrossRef] [Green Version]
  199. Liu, H.L.; Hua, M.Y.; Yang, H.W.; Huang, C.Y.; Chu, P.N.; Wu, J.S.; Tseng, I.C.; Wang, J.J.; Yen, T.C.; Chen, P.Y.; et al. Magnetic resonance monitoring of focused ultrasound/magnetic nanoparticle targeting delivery of therapeutic agents to the brain. Proc. Natl. Acad. Sci. USA 2010, 107, 15205–15210. [Google Scholar] [CrossRef] [Green Version]
  200. Burgess, A.; Hynynen, K. Noninvasive and targeted drug delivery to the brain using focused ultrasound. ACS Chem. Neurosci. 2013, 4, 519–526. [Google Scholar] [CrossRef] [Green Version]
  201. Arvanitis, C.D.; Askoxylakis, V.; Guo, Y.; Datta, M.; Kloepper, J.; Ferraro, G.B.; Bernabeu, M.O.; Fukumura, D.; McDannold, N.; Jain, R.K. Mechanisms of enhanced drug delivery in brain metastases with focused ultrasound-induced blood–tumor barrier disruption. Proc. Natl. Acad. Sci. USA 2018, 115, E8717–E8726. [Google Scholar] [CrossRef] [Green Version]
  202. Burgess, A.; Shah, K.; Hough, O.; Hynynen, K. Endothelial cell metabolism in normal and diseased vasculature. Circ. Res. 2016, 116, 1231–1244. [Google Scholar] [CrossRef]
  203. Miller, D.B.; O’Callaghan, J.P. New horizons for focused ultrasound (FUS)—Therapeutic applications in neurodegenerative diseases. Metabolism 2017, 69, S3–S7. [Google Scholar] [CrossRef] [PubMed]
  204. Lockman, P.R.; Mumper, R.J.; Khan, M.A.; Allen, D.D. Nanoparticle Technology for Drug Delivery Across the Blood-Brain Barrier. Drug Dev. Ind. Pharm. 2002, 28, 1–13. [Google Scholar] [CrossRef] [PubMed]
  205. Caffery, B.; Lee, J.S.; Alexander-Bryant, A.A. Vectors for Glioblastoma Gene Therapy: Viral & Non-Viral Delivery Strategies. Nanomaterials 2019, 9, 105. [Google Scholar] [CrossRef] [Green Version]
  206. Dong, X. Current Strategies for Brain Drug Delivery. Theranostics 2018, 8, 1481–1493. [Google Scholar] [CrossRef] [PubMed]
  207. Huang, T.T.; Hlavaty, J.; Ostertag, D.; Espinoza, F.L.; Martin, B.; Petznek, H.; Rodriguez-Aguirre, M.; Ibañez, C.E.; Kasahara, N.; Gunzburg, W.; et al. Toca 511 gene transfer and 5-fluorocytosine in combination with temozolomide demonstrates synergistic therapeutic efficacy in a temozolomide-sensitive glioblastoma model. Cancer Gene Ther. 2013, 20, 544–551. [Google Scholar] [CrossRef] [PubMed]
  208. Guhasarkar, D.; Su, Q.; Gao, G.; Sena-Esteves, M. Systemic AAV9-IFNβ gene delivery treats highly invasive glioblastoma. Neuro-Oncology 2016, 18, 1508–1518. [Google Scholar] [CrossRef] [Green Version]
  209. Lang, F.F.; Bruner, J.M.; Fuller, G.N.; Aldape, K.; Prados, M.D.; Chang, S.; Berger, M.S.; McDermoff, M.W.; Kunwar, S.M.; Junck, L.R.; et al. Phase I trial of adenovirus-mediated p53 gene therapy for recurrent glioma: Biological and clinical results. J. Clin. Oncol. 2003, 21, 2508–2518. [Google Scholar] [CrossRef]
  210. Liao, W.; Du, Y.; Zhang, C.; Pan, F.; Yao, Y.; Zhang, T.; Peng, Q. Exosomes: The next generation of endogenous nanomaterials for advanced drug delivery and therapy. Acta Biomater. 2019, 86, 1–14. [Google Scholar] [CrossRef]
  211. Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio Rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef]
  212. Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Liu, H.; Chen, L.; Liu, J.; Meng, H.; Zhang, R.; Ma, L.; Wu, L.; Yu, S.; Shi, F.; Li, Y.; et al. Co-delivery of tumor-derived exosomes with alpha-galactosylceramide on dendritic cell-based immunotherapy for glioblastoma. Cancer Lett. 2017, 411, 182–190. [Google Scholar] [CrossRef] [PubMed]
  214. Aboody, K.S.; Najbauer, J.; Danks, M.K. Stem and progenitor cell-mediated tumor selective gene therapy. Gene Ther. 2008, 15, 739–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Sheets, K.T.; Bagó, J.R.; Hingtgen, S.D. Delivery of cytotoxic mesenchymal stem cells with biodegradable scaffolds for treatment of postoperative brain cancer. In Targeted Drug Delivery; Sirianni, R.W., Behkam, B., Eds.; Springer International Publishing: Cham, Switzerland, 2018; Volume 1831, pp. 49–58. [Google Scholar]
  216. Xue, J.; Zhao, Z.; Zhang, L.; Xue, L.; Shen, S.; Wen, Y.; Wei, Z.; Wang, L.; Kong, L.; Sun, H.; et al. Neutrophil-mediated anticancer drug delivery for suppression of postoperative malignant glioma recurrence. Nat. Nanotechnol. 2017, 12, 692–700. [Google Scholar] [CrossRef]
  217. Hersh, D.S.; Wadajkar, A.S.; Roberts, N.B.; Perez, J.G.; Connolly, N.P.; Frenkel, V.; Winkles, J.A.; Woodworth, G.F.; Kim, A.J. Evolving Drug Delivery Strategies to Overcome the Blood Brain Barrier. Curr. Pharm. Des. 2016, 22, 1177–1193. [Google Scholar] [CrossRef] [Green Version]
  218. Balyasnikova, I.V.; Ferguson, S.D.; Han, Y.; Liu, F.; Lesniak, M.S. Therapeutic effect of neural stem cells expressing TRAIL and bortezomib in mice with glioma xenografts. Cancer Lett. 2011, 310, 148–159. [Google Scholar] [CrossRef] [Green Version]
  219. Parodi, A.; Rudzinska, M.; Deviatkin, A.A.; Soond, S.M.; Baldin, A.V.; Zamyatnin, J.A.A. Established and Emerging Strategies for Drug Delivery Across the Blood-Brain Barrier in Brain Cancer. Pharmaceutics 2019, 11, 245. [Google Scholar] [CrossRef] [Green Version]
  220. Batrakova, E.V.; Gendelman, H.E.; Kabanov, A.V. Cell-mediated drug delivery. Expert Opin. Drug Deliv. 2011, 8, 415–433. [Google Scholar] [CrossRef] [Green Version]
  221. Luther, D.C.; Huang, R.; Jeon, T.; Zhang, X.; Lee, Y.-W.; Nagaraj, H.; Rotello, V.M. Delivery of drugs, proteins, and nucleic acids using inorganic nanoparticles. Adv. Drug Deliv. Rev. 2020. [Google Scholar] [CrossRef]
  222. Sawicki, K.; Czajka, M.; Matysiak-Kucharek, M.; Fal, B.; Drop, B.; Mȩczyńska-Wielgosz, S.; Sikorska, K.; Kruszewski, M.; Kapka-Skrzypczak, L. Toxicity of metallic nanoparticles in the central nervous system. Nanotechnol. Rev. 2019, 8, 175–200. [Google Scholar] [CrossRef] [Green Version]
  223. Azcona, P.; Zysler, R.; Lassalle, V. Simple and novel strategies to achieve shape and size control of magnetite nanoparticles intended for biomedical applications. Colloids Surfaces A Physicochem. Eng. Asp. 2016, 504, 320–330. [Google Scholar] [CrossRef] [Green Version]
  224. Prades, R.; Guerrero, S.; Araya, E.; Molina, C.; Salas, E.; Zurita, E.; Selva, J.; Egea, G.; López-Iglesias, C.; Teixidó, M.; et al. Delivery of gold nanoparticles to the brain by conjugation with a peptide that recognizes the transferrin receptor. Biomaterials 2012, 33, 7194–7205. [Google Scholar] [CrossRef] [PubMed]
  225. Múzquiz-Ramos, E.M.; Guerrero-Chávez, V.; Macías-Martínez, B.I.; López-Badillo, C.M.; García-Cerda, L.A. Synthesis and characterization of maghemite nanoparticles for hyperthermia applications. Ceram. Int. 2015, 41, 397–402. [Google Scholar] [CrossRef]
  226. Tang, F.; Li, L.; Chen, D. Mesoporous silica nanoparticles: Synthesis, biocompatibility and drug delivery. Adv. Mater. 2012, 24, 1504–1534. [Google Scholar] [CrossRef] [PubMed]
  227. Kreuter, J. Mechanism of polymeric nanoparticle-based drug transport across the blood-brain barrier (BBB). J. Microencapsul. 2013. [Google Scholar] [CrossRef] [PubMed]
  228. Jallouli, Y.; Paillard, A.; Chang, J.; Sevin, E.; Betbeder, D. Influence of surface charge and inner composition of porous nanoparticles to cross blood-brain barrier in vitro. Int. J. Pharm. 2007, 344, 103–109. [Google Scholar] [CrossRef]
  229. Lu, W.; Wan, J.; She, Z.; Jiang, X. Brain delivery property and accelerated blood clearance of cationic albumin conjugated pegylated nanoparticle. J. Control. Release 2007, 118, 38–53. [Google Scholar] [CrossRef]
  230. Mendes, M.; Sousa, J.; Pais, A.A.; Vitorino, C. Targeted Theranostic Nanoparticles for Brain Tumor Treatment. Pharmaceutics 2018, 10, 181. [Google Scholar] [CrossRef] [Green Version]
  231. Li, J.; Zhou, L.; Ye, D.; Huang, S.; Shao, K.; Huang, R.; Han, L.; Liu, Y.; Liu, S.; Ye, L.; et al. Choline-derivate-modified nanoparticles for brain-targeting gene delivery. Adv. Mater. 2011, 23, 4516–4520. [Google Scholar] [CrossRef]
  232. Rip, J.; Chen, L.; Hartman, R.; Van Den Heuvel, A.; Reijerkerk, A.; Van Kregten, J.; Van Der Boom, B.; Appeldoorn, C.; De Boer, M.; Maussang, D.; et al. Glutathione PEGylated liposomes: Pharmacokinetics and delivery of cargo across the blood-brain barrier in rats. J. Drug Target. 2014, 22, 460–467. [Google Scholar] [CrossRef] [Green Version]
  233. Johnsen, K.B.; Burkhart, A.; Melander, F.; Kempen, P.J.; Vejlebo, J.B.; Siupka, P.; Nielsen, M.S.; Andresen, T.L.; Moos, T. Targeting transferrin receptors at the blood-brain barrier improves the uptake of immunoliposomes and subsequent cargo transport into the brain parenchyma. Sci. Rep. 2017, 7, 1–13. [Google Scholar] [CrossRef] [PubMed]
  234. Ulbrich, K.; Knobloch, T.; Kreuter, J. Targeting the insulin receptor: Nanoparticles for drug delivery across the blood-brain barrier (BBB). J. Drug Target. 2011. [Google Scholar] [CrossRef] [PubMed]
  235. Mulik, R.S.; Bing, C.; Ladouceur-Wodzak, M.; Munaweera, I.; Chopra, R.; Corbin, I.R. Localized delivery of low-density lipoprotein docosahexaenoic acid nanoparticles to the rat brain using focused ultrasound. Biomaterials 2016, 83, 257–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Agarwal, S.; Sane, R.; Oberoi, R.; Ohlfest, J.R.; Elmquist, W.F. Delivery of molecularly targeted therapy to malignant glioma, a disease of the whole brain. Expert Rev. Mol. Med. 2011, 13, e17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Yu, Y.J.; Zhang, Y.; Kenrick, M.; Hoyte, K.; Luk, W.; Lu, Y.; Atwal, J.; Elliott, J.M.; Prabhu, S.; Watts, R.J.; et al. Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci. Transl. Med. 2011, 3, 84ra44. [Google Scholar] [CrossRef] [PubMed]
  238. Oller-Salvia, B.; Sánchez-Navarro, M.; Giralt, E.; Teixidó, M. Blood-brain barrier shuttle peptides: An emerging paradigm for brain delivery. Chem. Soc. Rev. 2016, 45, 4690–4707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Haqqani, A.S.; Thom, G.; Burrell, M.; Delaney, C.E.; Brunette, E.; Baumann, E.; Sodja, C.; Jezierski, A.; Webster, C.; Stanimirovic, D.B. Intracellular sorting and transcytosis of the rat transferrin receptor antibody OX26 across the blood–brain barrier in vitro is dependent on its binding affinity. J. Neurochem. 2018, 146, 735–752. [Google Scholar] [CrossRef] [Green Version]
  240. Uchida, Y.; Yagi, Y.; Takao, M.; Takao, M.; Tano, M.; Umetsu, M.; Hirano, S.; Usui, T.; Tachikawa, M.; Terasaki, T. Comparison of Absolute Protein Abundances of Transporters and Receptors among Blood-Brain Barriers at Different Cerebral Regions and the Blood-Spinal Cord Barrier in Humans and Rats. Mol. Pharm. 2020, 17, 2006–2020. [Google Scholar] [CrossRef]
  241. Uchida, Y.; Ohtsuki, S.; Katsukura, Y.; Ikeda, C.; Suzuki, T.; Kamiie, J.; Terasaki, T. Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. J. Neurochem. 2011, 117, 333–345. [Google Scholar] [CrossRef]
  242. Aasen, S.N.; Espedal, H.; Holte, C.F.; Keunen, O.; Karlsen, T.V.; Tenstad, O.; Maherally, Z.; Miletic, H.; Hoang, T.; Eikeland, A.V.; et al. Improved drug delivery to brain metastases by peptide-mediated permeabilization of the blood–brain barrier. Mol. Cancer Ther. 2019, 18, 2171–2181. [Google Scholar] [CrossRef] [Green Version]
  243. Sarkar, G.; Curran, G.L.; Sarkaria, J.N.; Lowe, V.J.; Jenkins, R.B. Peptide carrier-mediated non-covalent delivery of unmodified cisplatin, methotrexate and other agents via intravenous route to the brain. PLoS ONE 2014, 9, e97655. [Google Scholar] [CrossRef] [PubMed]
  244. Bird, G.H.; Madani, N.; Perry, A.F.; Princiotto, A.M.; Supko, J.G.; He, X.; Gavathiotis, E.; Sodroski, J.G.; Walensky, L.D. Hydrocarbon double-stapling remedies the proteolytic instability of a lengthy peptide therapeutic. Proc. Natl. Acad. Sci. USA 2010, 107, 14093–14098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Pardridge, W.M. Blood-Brain Barrier and Delivery of Protein and Gene Therapeutics to Brain. Front. Aging Neurosci. 2020. [Google Scholar] [CrossRef] [PubMed]
  246. Richardson, D.R.; Morgan, E.H. The transferrin homologue, melanotransferrin (p97), is rapidly catabolized by the liver of the rat and does not effectively donate iron to the brain. Biochim. Biophys. Acta-Mol. Basis Dis. 2004, 1690, 124–133. [Google Scholar] [CrossRef] [Green Version]
  247. Gobinda, S.; Geoffry, C.; Robert, J. Crossing the blood-brain barrier: Brain delivery of unmodified cancer therapeutics via intravenous route mediated by a peptide transporter. Neuro. Oncol. 2013, 15, iii37–iii61. [Google Scholar] [CrossRef] [Green Version]
  248. Demeule, M.; Regina, A.; Ché, C.; Poirier, J.; Nguyen, T.; Gabathuler, R.; Castaigne, J.P.; Béliveau, R. Identification and design of peptides as a new drug delivery system for the brain. J. Pharmacol. Exp. Ther. 2008, 324, 1064–1072. [Google Scholar] [CrossRef] [Green Version]
  249. Luo, Z.; Jin, K.; Pang, Q.; Shen, S.; Yan, Z.; Jiang, T.; Zhu, X.; Yu, L.; Pang, Z.; Jiang, X. On-Demand Drug Release from Dual-Targeting Small Nanoparticles Triggered by High-Intensity Focused Ultrasound Enhanced Glioblastoma-Targeting Therapy. ACS Appl. Mater. Interfaces 2017, 9, 31612–31625. [Google Scholar] [CrossRef]
  250. Zhu, Y.; Jiang, Y.; Meng, F.; Deng, C.; Cheng, R.; Zhang, J.; Feijen, J.; Zhong, Z. Highly efficacious and specific anti-glioma chemotherapy by tandem nanomicelles co-functionalized with brain tumor-targeting and cell-penetrating peptides. J. Control. Release 2018. [Google Scholar] [CrossRef]
  251. Shi, Y.; Jiang, Y.; Cao, J.; Yang, W.; Zhang, J.; Meng, F.; Zhong, Z. Boosting RNAi therapy for orthotopic glioblastoma with nontoxic brain-targeting chimaeric polymersomes. J. Control. Release 2018, 292, 163–171. [Google Scholar] [CrossRef]
  252. Regina, A.; Demeule, M.; Tripathy, S.; Lord-Dufour, S.; Currie, J.C.; Iddir, M.; Annabi, B.; Castaigne, J.P.; Lachowicz, J.E. ANG4043, a novel brain-penetrant peptide-mAb conjugate, is efficacious against HER2-positive intracranial tumors in mice. Mol. Cancer Ther. 2015. [Google Scholar] [CrossRef] [Green Version]
  253. Hoyos-Ceballos, G.P.; Ruozi, B.; Ottonelli, I.; Da Ros, F.; Vandelli, M.A.; Forni, F.; Daini, E.; Vilella, A.; Zoli, M.; Tosi, G.; et al. PLGA-PEG-Ang–2 nanoparticles for blood–brain barrier crossing: Proof-of-concept study. Pharmaceutics 2020, 12, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Wang, X.; Xiong, Z.; Liu, Z.; Huang, X.; Jiang, X. Angiopep-2/IP10-EGFRvIIIscFv modified nanoparticles and CTL synergistically inhibit malignant glioblastoma. Sci. Rep. 2018, 8, 12827. [Google Scholar] [CrossRef] [PubMed]
  255. Tian, X.; Nyberg, S.; Sharp, P.S.; Madsen, J.; Daneshpour, N.; Armes, S.P.; Berwick, J.; Azzouz, M.; Shaw, P.; Abbott, N.J.; et al. LRP-1-mediated intracellular antibody delivery to the Central Nervous System. Sci. Rep. 2015, 5, srep11990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Kumthekar, P.; Tang, S.; Brenner, A.J.; Kesari, S.; Anders, C.K.; Carrillo, J.A.; Chalasani, P.; Kabos, P.; Ahluwalia, M.S.; Ibrahim, N.K. OS7.2 A Phase II Study of ANG1005, a novel BBB/BCB Penetratant Taxane in Patients with Recurrent Brain Metastases and Leptomeningeal Carcinomatosis from Breast Cancer. Neuro-Oncology 2016, 18, iv16. [Google Scholar] [CrossRef] [Green Version]
  257. Kumthekar, P.; Tang, S.C.; Brenner, A.J.; Kesari, S.; Piccioni, D.E.; Anders, C.; Carrillo, J.; Chalasani, P.; Kabos, P.; Puhalla, S.; et al. ANG1005, a Brain-Penetrating Peptide–Drug Conjugate, Shows Activity in Patients with Breast Cancer with Leptomeningeal Carcinomatosis and Recurrent Brain Metastases. Clin. Cancer Res. 2020, 26, 2789–2799. [Google Scholar] [CrossRef] [Green Version]
  258. Thomas, F.C.; Taskar, K.; Rudraraju, V.; Goda, S.; Thorsheim, H.R.; Gaasch, J.A.; Mittapalli, R.K.; Palmieri, D.; Steeg, P.S.; Lockman, P.R.; et al. Uptake of ANG1005, a novel paclitaxel derivative, through the blood-brain barrier into brain and experimental brain metastases of breast cancer. Pharm. Res. 2009, 26, 2486–2494. [Google Scholar] [CrossRef] [Green Version]
  259. Gaillard, P.J.; Appeldoorn, C.C.M.; Dorland, R.; Van Kregten, J.; Manca, F.; Vugts, D.J.; Windhorst, B.; Van Dongen, G.A.M.S.; De Vries, H.E.; Maussang, D.; et al. Pharmacokinetics, brain delivery, and efficacy in brain tumor-bearing mice of glutathione pegylated liposomal doxorubicin (2B3-101). PLoS ONE 2014, 9, e82331. [Google Scholar] [CrossRef] [Green Version]
  260. Brandsma, D.; Dieras, V.; Altintas, S.; Anders, C.; Arnedos, M.; Gelderblom, H.; Soetekouw, P.; Jager, A.; van Linde, M.; Aftimos, P. p08.03 * 2b3-101, glutathione pegylated liposomal doxorubicin, in patients with recurrent high grade gliomas and breast cancer brain metastases. Neuro. Oncol. 2014, 16, ii50–ii51. [Google Scholar] [CrossRef] [Green Version]
  261. Recht, L.; Torres, C.O.; Smith, T.W.; Raso, V.; Griffin, T.W. Transferrin receptor in normal and neoplastic brain tissue: Implications for brain-tumor immunotherapy. J. Neurosurg. 1990, 72, 941–945. [Google Scholar] [CrossRef]
  262. Kim, G.; Kim, M.; Lee, Y.; Byun, J.W.; Hwang, D.W.; Lee, M. Systemic delivery of microRNA-21 antisense oligonucleotides to the brain using T7-peptide decorated exosomes. J. Control. Release 2020, 317, 273–281. [Google Scholar] [CrossRef]
  263. Kuang, Y.; An, S.; Guo, Y.; Huang, S.; Shao, K.; Liu, Y.; Li, J.; Ma, H.; Jiang, C. T7 peptide-functionalized nanoparticles utilizing RNA interference for glioma dual targeting. Int. J. Pharm. 2013, 454, 11–20. [Google Scholar] [CrossRef] [PubMed]
  264. Dixit, S.; Novak, T.; Miller, K.; Zhu, Y.; Kenney, M.E.; Broome, A.M. Transferrin receptor-targeted theranostic gold nanoparticles for photosensitizer delivery in brain tumors. Nanoscale 2015, 7, 1782–1790. [Google Scholar] [CrossRef] [PubMed]
  265. Zhang, X.; He, T.; Chai, Z.; Samulski, R.J.; Li, C. Blood-brain barrier shuttle peptides enhance AAV transduction in the brain after systemic administration. Biomaterials 2018, 176, 71–83. [Google Scholar] [CrossRef]
  266. Abuqayyas, L.; Balthasar, J.P. Investigation of the role of FcγR and FcRn in mAb distribution to the brain. Mol. Pharm. 2013, 10, 1505–1513. [Google Scholar] [CrossRef] [PubMed]
  267. Zhang, Y.; Pardridge, W.M. Mediated efflux of IgG molecules from brain to blood across the blood-brain barrier. J. Neuroimmunol. 2001, 114, 168–172. [Google Scholar] [CrossRef]
  268. Schlachetzki, F.; Zhu, C.; Pardridge, W.M. Expression of the neonatal Fc receptor (FcRn) at the blood-brain barrier. J. Neurochem. 2002, 81, 203–206. [Google Scholar] [CrossRef]
  269. St-Amour, I.; Paré, I.; Alata, W.; Coulombe, K.; Ringuette-Goulet, C.; Drouin-Ouellet, J.; Vandal, M.; Soulet, D.; Bazin, R.; Calon, F. Brain bioavailability of human intravenous immunoglobulin and its transport through the murine blood-brain barrier. J. Cereb. Blood Flow Metab. 2013, 33, 1983–1992. [Google Scholar] [CrossRef] [Green Version]
  270. Kumar, N.N.; Pizzo, M.E.; Nehra, G.; Wilken-Resman, B.; Boroumand, S.; Thorne, R.G. Passive Immunotherapies for Central Nervous System Disorders: Current Delivery Challenges and New Approaches. Bioconjug. Chem. 2018, 29, 3937–3966. [Google Scholar] [CrossRef]
  271. Gan, H.K.; van den Bent, M.; Lassman, A.B.; Reardon, D.A.; Scott, A.M. Antibody–drug conjugates in glioblastoma therapy: The right drugs to the right cells. Nat. Rev. Clin. Oncol. 2017, 14, 695–707. [Google Scholar] [CrossRef]
  272. Cavaco, M.; Gaspar, D.; Castanho, M.; Neves, V. Antibodies for the Treatment of Brain Metastases, a Dream or a Reality? Pharmaceutics 2020, 12, 62. [Google Scholar] [CrossRef] [Green Version]
  273. Zhang, Y.; Zhu, C.; Pardridge, W.M. Antisense gene therapy of brain cancer with an artificial virus gene delivery system. Mol. Ther. 2002, 6, 67–72. [Google Scholar] [CrossRef] [PubMed]
  274. Boado, R.J.; Hui, E.K.W.; Lu, J.Z.; Pardridge, W.M. Very high plasma concentrations of a monoclonal antibody against the human insulin receptor are produced by subcutaneous injection in the rhesus monkey. Mol. Pharm. 2016, 13, 3241–3246. [Google Scholar] [CrossRef] [PubMed]
  275. Xu, L.; Huang, C.C.; Huang, W.; Tang, W.H.; Rait, A.; Yin, Y.Z.; Cruz, I.; Xiang, L.M.; Pirollo, K.F.; Chang, E.H. Systemic tumor-targeted gene delivery by anti-transferrin receptor scFv-immunoliposomes. Mol. Cancer Ther. 2002, 1, 5. [Google Scholar]
  276. Kim, S.S.; Harford, J.B.; Moghe, M.; Slaughter, T.; Doherty, C.; Chang, E.H. A tumor-targeting nanomedicine carrying the p53 gene crosses the blood–brain barrier and enhances anti-PD-1 immunotherapy in mouse models of glioblastoma. Int. J. Cancer 2019, 145, 2535–2546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Senzer, N.; Nemunaitis, J.; Nemunaitis, D.; Bedell, C.; Edelman, G.; Barve, M.; Nunan, R.; Pirollo, K.F.; Rait, A.; Chang, E.H. Phase I Study of a Systemically Delivered p53 Nanoparticle in Advanced Solid Tumors. Mol. Ther. 2013, 21, 1096–1103. [Google Scholar] [CrossRef] [Green Version]
  278. Stanimirovic, D.B.; Kemmerich, K.; Haqqani, A.S.; Farrington, G.K. Engineering and Pharmacology of Blood–Brain Barrier-Permeable Bispecific Antibodies. In Advances in Pharmacology; Elsevier: Amsterdam, The Netherlands, 2014; Volume 71, pp. 301–335. [Google Scholar]
  279. VVerdino, P.; Atwell, S.; Demarest, S.J. Emerging trends in bispecific antibody and scaffold protein therapeutics. Curr. Opin. Chem. Eng. 2018, 19, 107–123. [Google Scholar] [CrossRef]
  280. Karaoglu Hanzatian, D.; Schwartz, A.; Gizatullin, F.; Erickson, J.; Deng, K.; Villanueva, R.; Stedman, C.; Harris, C.; Ghayur, T.; Goodearl, A. Brain uptake of multivalent and multi-specific DVD-Ig proteins after systemic administration. mAbs 2018, 10, 765–777. [Google Scholar] [CrossRef] [Green Version]
  281. Kariolis, M.S.; Wells, R.C.; Getz, J.A.; Kwan, W.; Mahon, C.S.; Tong, R.; Kim, D.J.; Srivastava, A.; Bedard, C.; Henne, K.R.; et al. Brain delivery of therapeutic proteins using an Fc fragment blood-brain barrier transport vehicle in mice and monkeys. Sci. Transl. Med. 2020, 12, eaay1359. [Google Scholar] [CrossRef]
  282. Ullman, J.C.; Arguello, A.; Getz, J.A.; Bhalla, A.; Mahon, C.S.; Wang, J.; Giese, T.; Bedard, C.; Kim, D.J.; Blumenfeld, J.R.; et al. Brain delivery and activity of a lysosomal enzyme using a blood-brain barrier transport vehicle in mice. Sci. Transl. Med. 2020, 12, eaay1163. [Google Scholar] [CrossRef]
  283. Tzeng, S.Y.; Green, J.J. Therapeutic nanomedicine for brain cancer. Ther. Deliv. 2013, 4, 687–704. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The neurovascular unit/blood–brain barrier (NVU/BBB) is composed of specialized endothelial cells and support cells, including pericytes and astrocytes. The cross-sectional view illustrates that the majority of the abluminal surface of the endothelial cell is covered by pericytes and astrocytic foot processes. Paracellular transport across the BBB/NVU is restricted by tight junction proteins, and even small, lipophilic molecules that might diffuse across the BBB may be subject to active efflux by a variety of proteins. Facilitated active transport, receptor-mediated transport, and ion transporters allow the brain to be supplied with nutrients while maintaining strict homeostasis.
Figure 1. The neurovascular unit/blood–brain barrier (NVU/BBB) is composed of specialized endothelial cells and support cells, including pericytes and astrocytes. The cross-sectional view illustrates that the majority of the abluminal surface of the endothelial cell is covered by pericytes and astrocytic foot processes. Paracellular transport across the BBB/NVU is restricted by tight junction proteins, and even small, lipophilic molecules that might diffuse across the BBB may be subject to active efflux by a variety of proteins. Facilitated active transport, receptor-mediated transport, and ion transporters allow the brain to be supplied with nutrients while maintaining strict homeostasis.
Pharmaceutics 12 01205 g001
Figure 2. The blood–tumor barrier (BTB) is characterized by increased cytokine and VEGF signaling from the tumor, which may lead to decreased expression of tight junction (TJ) proteins like claudin-5. Alterations in pericyte phenotype and disruption of astrocytic associations with endothelial cells may contribute to decreased barrier integrity. However, this is not a uniform phenomenon within or among tumors, and the expression of efflux transporters limits drug permeation into the tumor. Evidence exists showing decreased permeability of the BTB in regions distant to the core of the tumor, which more closely resemble “unaffected” brain.
Figure 2. The blood–tumor barrier (BTB) is characterized by increased cytokine and VEGF signaling from the tumor, which may lead to decreased expression of tight junction (TJ) proteins like claudin-5. Alterations in pericyte phenotype and disruption of astrocytic associations with endothelial cells may contribute to decreased barrier integrity. However, this is not a uniform phenomenon within or among tumors, and the expression of efflux transporters limits drug permeation into the tumor. Evidence exists showing decreased permeability of the BTB in regions distant to the core of the tumor, which more closely resemble “unaffected” brain.
Pharmaceutics 12 01205 g002
Figure 3. Various invasive technologies to increase drug delivery to the brain by bypassing the BBB/NVU include intrathecal injection via a lumbar puncture and various intracranial techniques. These include (A) intracerebroventricular injection using the Ommaya reservoir, (B) convection-enhanced delivery (CED) by way of intracerebral catheter placement, and (C) placement of drug-loaded polymeric wafers.
Figure 3. Various invasive technologies to increase drug delivery to the brain by bypassing the BBB/NVU include intrathecal injection via a lumbar puncture and various intracranial techniques. These include (A) intracerebroventricular injection using the Ommaya reservoir, (B) convection-enhanced delivery (CED) by way of intracerebral catheter placement, and (C) placement of drug-loaded polymeric wafers.
Pharmaceutics 12 01205 g003
Figure 4. Drug delivery to the brain may be increased by noninvasive BBB disruption (BBBD) techniques, including (A) osmotic disruption and (B) focused ultrasound. In osmotic disruption, infusion of a hyperosmolar solution via a cerebral artery results in endothelial cell shrinkage, temporarily disrupting tight junctions. Focused ultrasound uses an infusion of inert gas-filled microbubbles, which, upon application of focused ultrasound, may burst and temporarily disrupt tight junction proteins. Advantages and disadvantages of both are listed.
Figure 4. Drug delivery to the brain may be increased by noninvasive BBB disruption (BBBD) techniques, including (A) osmotic disruption and (B) focused ultrasound. In osmotic disruption, infusion of a hyperosmolar solution via a cerebral artery results in endothelial cell shrinkage, temporarily disrupting tight junctions. Focused ultrasound uses an infusion of inert gas-filled microbubbles, which, upon application of focused ultrasound, may burst and temporarily disrupt tight junction proteins. Advantages and disadvantages of both are listed.
Pharmaceutics 12 01205 g004
Figure 5. Receptor-mediated transcytosis is one of the most common techniques to increase the delivery of large molecules, nanoparticles, and brain-impermeant drugs to brain tumors. Cargo bound to endothelial-membrane-bound receptors is pulled into endothelial cells (ECs) and sorted in the early endosome. Bivalent-binding and high-affinity cargo-receptor complexes are often trafficked to the lysosome for degradation, whereas cargoes bound with lower affinity are more likely to be trafficked for transport across the cell. The cargo is then released on the abluminal side of the endothelium, and the receptor may be recycled back to the luminal membrane.
Figure 5. Receptor-mediated transcytosis is one of the most common techniques to increase the delivery of large molecules, nanoparticles, and brain-impermeant drugs to brain tumors. Cargo bound to endothelial-membrane-bound receptors is pulled into endothelial cells (ECs) and sorted in the early endosome. Bivalent-binding and high-affinity cargo-receptor complexes are often trafficked to the lysosome for degradation, whereas cargoes bound with lower affinity are more likely to be trafficked for transport across the cell. The cargo is then released on the abluminal side of the endothelium, and the receptor may be recycled back to the luminal membrane.
Pharmaceutics 12 01205 g005
Table 1. Endothelial cell membrane transporters: partial list of common carriers.
Table 1. Endothelial cell membrane transporters: partial list of common carriers.
Transport SystemTypical SubstrateSLC FamilyCommon Name
Carbohydrates
HexoseGlucoseSLC2A1Glut1
Sodium Myo-inositolMyo-inositolSLC5A3SMIT
Monocarboxylates
Monocarboxylic acidLactic acid
ketones
SLC16A1MCT1
Amino Acids
Large neutral amino acidPhenylalanineSLC7A5LAT1
Small neutral amino acidAlanineSLC38A2SNAT2, -3, -5
Cationic amino acidLysineSLC7A1Cat1, CAT3
Beta amino acidTaurineSLC6A6TauT
Ala-Ser-CysAla, ser, cysSLC1A4ASCT1, -2
Excitatory amino acidGlutamic acidSLC1A2EAAT-1, -2, -3
GlycineGlycineSLC6A9, A5GT-1
Others
Fatty acidsEssential FA LPC-PC (DHA)SLC44A1/2 Mfsd2AFATP-1, -4 Mfsd2A
NucleosideAdenosineSLC29A1 SLC28A1ENT-1, -2; CNT1–3
HormonesThyroid T3
Thyroid T4
SLC16A2
OATP1C1
MCT8
OATP1C1
Biotin, pantothenic acidbiotinSLC5A6SMVT
Folic acidFolinic acidSLC46A1PCFT
CopperCu+SLC31A1CTR1
Table 2. Brain endothelial cell transporters of xenobiotics/drugs. Members of the ABC (ATP-binding cassette) superfamily of transporters demonstrated in brain endothelial cells and non-ABC transporters of organic chemicals potentially present are listed.
Table 2. Brain endothelial cell transporters of xenobiotics/drugs. Members of the ABC (ATP-binding cassette) superfamily of transporters demonstrated in brain endothelial cells and non-ABC transporters of organic chemicals potentially present are listed.
Transport SystemCommon NameTypical Substrate
ATP Binding Cassette Transporter (ABC)
ABCB1P-gpBroad-spectrum, xenobiotics
ABCG2BCRPmitoxantrone anthracycline
xenobiotics
ABCC1MRP1GSSG, leukotrienes
ABCC5MRP5Thiopurines, cyclic nucleotides
ABCC4MRP4Organic anions
Non-ABC Transporters
SLC22A7OAT2-3Organic ions
SLC22A8OATP1A4
SLC20A2OATP2B1
SLCO1A4OCTN2
SLCO2B1OCT1-3
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Griffith, J.I.; Rathi, S.; Zhang, W.; Zhang, W.; Drewes, L.R.; Sarkaria, J.N.; Elmquist, W.F. Addressing BBB Heterogeneity: A New Paradigm for Drug Delivery to Brain Tumors. Pharmaceutics 2020, 12, 1205. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics12121205

AMA Style

Griffith JI, Rathi S, Zhang W, Zhang W, Drewes LR, Sarkaria JN, Elmquist WF. Addressing BBB Heterogeneity: A New Paradigm for Drug Delivery to Brain Tumors. Pharmaceutics. 2020; 12(12):1205. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics12121205

Chicago/Turabian Style

Griffith, Jessica I., Sneha Rathi, Wenqiu Zhang, Wenjuan Zhang, Lester R. Drewes, Jann N. Sarkaria, and William F. Elmquist. 2020. "Addressing BBB Heterogeneity: A New Paradigm for Drug Delivery to Brain Tumors" Pharmaceutics 12, no. 12: 1205. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics12121205

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop