Figure 2 - uploaded by Ji Song
Content may be subject to copyright.
Prediction of normal brain extracellular spacings based on nanoparticle diffusion (A) Distribution of the logarithmic D eff of individual nanoparticles (■ PS-PEG5k and □ PSCOOH) at τ = 1 s. Data represents 4 independent experiments, with an average of n > 100 nanoparticles per experiment. (B) Normalized ensemble-averaged diffusivities (<D eff >/D W ) for different sized PEG-coated PS particles (60 nm (■), 110 nm (•), and 240 nm (▲)) at τ = 1 s, where D W is the theoretical diffusivities of neutral particles in PBS. The solid and dashed lines represent the range of theoretical <D eff >/D W ratios for various sized 60-, 110-, and 240-nm particles predicted by the obstruction scaling model. This leads to an estimated mesh spacing range in normal rat brain tissue of 20-230 nm obtained by maximum likelihood estimation fitting to experimental diffusivity ratios. <D eff >/D W from Thorne et al [34] are provided (3 nm (■), 14 nm (▲), and 35 nm(•)) for reference to previously estimated values. (C) Percent pores larger than 100-and 200-nm. Data represents the ensemble average of four independent experiments with n > 100 particles tracked for each experiment. 

Prediction of normal brain extracellular spacings based on nanoparticle diffusion (A) Distribution of the logarithmic D eff of individual nanoparticles (■ PS-PEG5k and □ PSCOOH) at τ = 1 s. Data represents 4 independent experiments, with an average of n > 100 nanoparticles per experiment. (B) Normalized ensemble-averaged diffusivities (<D eff >/D W ) for different sized PEG-coated PS particles (60 nm (■), 110 nm (•), and 240 nm (▲)) at τ = 1 s, where D W is the theoretical diffusivities of neutral particles in PBS. The solid and dashed lines represent the range of theoretical <D eff >/D W ratios for various sized 60-, 110-, and 240-nm particles predicted by the obstruction scaling model. This leads to an estimated mesh spacing range in normal rat brain tissue of 20-230 nm obtained by maximum likelihood estimation fitting to experimental diffusivity ratios. <D eff >/D W from Thorne et al [34] are provided (3 nm (■), 14 nm (▲), and 35 nm(•)) for reference to previously estimated values. (C) Percent pores larger than 100-and 200-nm. Data represents the ensemble average of four independent experiments with n > 100 particles tracked for each experiment. 

Source publication
Article
Full-text available
The blood-brain barrier (BBB) presents a significant obstacle for the treatment of many central nervous system (CNS) disorders, including invasive brain tumors, Alzheimer's, Parkinson's and stroke. Therapeutics must be capable of bypassing the BBB and also penetrate the brain parenchyma to achieve a desired effect within the brain. In this study, w...

Contexts in source publication

Context 1
... ensure that the observed rapid transport of PEGylated nanoparticles was not biased by a small fraction of fast moving outlier particles, we evaluated the heterogeneity in particle transport rates by examining the distribution of individual particle diffusivities at 1 s ( Figure 2A). The fastest 75 percent of 60-nm and fastest 65 percent of 110-nm PS-PEG particles exhibited uniformly rapid transport, compared to the fastest 15 percent of 240-nm PS-PEG particles. ...
Context 2
... on the determination that transport was limited due to steric obstruction for larger particles and not due to particle interaction with brain ECS components, diffusion rates for 240-nm PEG-PS particles were incorporated into this model. Using maximum likelihood estimation, the average pore size of normal rat brain ECS was estimated to be between 60-120 nm ( Figure 2B). More specifically, we calculated the pore size distribution to range from 20 nm to 230 nm. ...
Context 3
... specifically, we calculated the pore size distribution to range from 20 nm to 230 nm. The largest 20% of pore sizes experienced by particles were > 100 nm and 14% of pore sizes experienced by particles were > 200 nm ( Figure 2C). Overall approximately 60% of the pores sampled by probe particles were larger than 50 nm. ...
Context 4
... weighted MR images, which detect red blood cell leakage across the BBB, were taken 2-5 minutes postsonication at pressures ranging from 0.4 to 2.0 MPa. Hypointense regions indicating the presence of blood in the tissue appeared at 92% of locations (n=39) sonicated with pressures greater than or equal to 0.8 MPa, while only 16% of locations (n=43) sonicated with 0.6 MPa developed hypointensity ( Figure S2A). No erythrocyte leakage was detected at 0.4 MPa (n=3). ...
Context 5
... note, hypointense regions produced at 0.6 MPa were much smaller and fainter than those produced at higher pressures, and only occurred when sonications were performed close to the olfactory bulb. Additionally, MR thermometry indicated that no thermal rise occurred at the focal spot during the sonications ( Figure S2B). ...

Similar publications

Article
Full-text available
Background and Purpose The blood-brain barrier (BBB), a critical interface of specialized endothelial cells, plays a pivotal role in regulating molecular and ion transport between the central nervous system (CNS) and systemic circulation. Experimental Approach This review aims to delve into the intricate architecture and functions of the BBB while...
Preprint
Full-text available
Phagocytic immunotherapies such as CD47 blockade have emerged as promising strategies for glioblastoma (GB) therapy, but the blood brain/tumor barriers (BBB/BTB) pose a persistent challenge for mCD47 delivery that can be overcome by focused ultrasound (FUS)-mediated BBB/BTB disruption. We here leverage immuno-PET imaging to determine how timing of...
Article
Full-text available
Focused ultrasound (FUS) coupled with microbubbles (MB) has been found as a promising approach to disrupt the blood-brain barrier (BBB). However, how this disruption affects drug transport remains unclear. In this study, drug transport in the combination therapy of liposomes and FUS-MB induced BBB disruption (BBBD) is investigated based on a multip...
Article
Full-text available
Background Focused ultrasound (FUS) combined with microbubbles (MBs) has emerged as a potential approach for opening the blood–brain barrier (BBB) for delivering drugs into the brain. However, MBs range in size of microns and thus can hardly extravasate into the brain parenchyma. Recently, growing attention has been paid to gas vesicles (GVs), whic...
Article
Full-text available
Glioblastoma multiforme (GBM) is a deadly and aggressive malignant brain cancer that is highly resistant to treatments. A particular challenge of treatment is caused by the blood–brain barrier (BBB), the relatively impermeable vasculature of the brain. The BBB prevents large molecules from entering the brain parenchyma. This protective characterist...

Citations

... These techniques aim to open the BBB temporarily and safely or utilize specialized mechanisms to transport drugs. These technologies, including focused ultrasound, electromagnetic fields, and intranasal delivery, promise to revolutionize CNS drug delivery [50]. ...
Article
Full-text available
Non-invasive drug delivery across the blood–brain barrier (BBB) represents a significant advancement in treating neurological diseases. The BBB is a tightly packed layer of endothelial cells that shields the brain from harmful substances in the blood, allowing necessary nutrients to pass through. It is a highly selective barrier, which poses a challenge to delivering therapeutic agents into the brain. Several non-invasive procedures and devices have been developed or are currently being investigated to enhance drug delivery across the BBB. This paper presents a review and a prospective analysis of the art and science that address pharmacology, technology, delivery systems, regulatory approval, ethical concerns, and future possibilities.
... The safety and efficacy of FUS BBBO has been assessed in numerous preclinical trials involving animals and human subjects [14]. The increase in temperature in the FUS target region and surrounding tissue is minimal (less than two degrees in several rodent and porcine models), regardless of the operating parameters [15,16]. Note however that the mechanical effects of FUS BBBO can only be induced under optimal operating parameters, including ultrasound intensity, transducer frequency, microbubble size, and bubble concentration [17][18][19]. ...
... Extensive research on the use of FUS to induce BBBO has made it clear that parameter optimization is crucial to the effectiveness of BBBO for drug delivery applications, particularly when used for immunotherapy and targeted therapy in the CNS [29]. Here, we discuss the effects of various therapeutic agents entering the CNS following FUS-induced BBBO, including drug coated microbubbles [30][31][32], and nanoparticles [15]. Numerous animal studies have investigated the efficacy of FUS-MB enhanced drug delivery based on LIFU. ...
... Researchers have determined that it is safe to deliver 60 nm pressure-dependent brain-penetrating nanoparticles (BPNs) in regions where the BBB has been disrupted by FUS-MBs. Note that the clearance of BPNs from the body is delayed and they pass only into those areas where BBB has been disrupted [15], thereby making it possible to target specific areas of interest without off-target toxicity. ...
Article
Full-text available
Background Despite advances in immunotherapy and targeted treatments for malignancies of the central nervous system (CNS), the treatment of brain metastases (BMs) remains a formidable challenge, due largely to difficulties in crossing the blood-brain barrier (BBB), drug resistance, and molecular discrepancies. Focused ultrasound (FUS) is a non-invasive tool for BBB breaching, tumor ablation, enhancing drug delivery, promoting the release of tumor biomarkers for liquid biopsy, or the tumor microenvironment disruption. This paper presents a comprehensive review of the current literature related to FUS and its application in the treatment of brain metastasis. Methods This review of the current literature via PubMed, Google Scholar, and Clincaltrials.gov focused on clinical trials in which FUS is used in the intracranial treatment of metastatic tumor, glioma, or GBM. Results FUS is safe and effective for treatment of primary or metastatic brain tumors. FUS-augmented drug delivery can open BBB to facilitate the transport of chemotherapeutic agents, immunotherapies, and targeted treatments. The integration of FUS with liquid biopsy has considerable potential for early tumor detection, precise gene profiling, and personalized therapy. Sonodynamic therapy can induce tumor cell apoptosis and could potentially be used to enhance the outcomes of other tumor treatments, such as surgery and chemotherapy. Conclusion Further work is required to establish FUS as a standard therapy for BMs. FUS has the potential to transform brain tumor treatment, particularly when combined with immunotherapy and targeted therapy as a non-invasive alternative to surgery and radiation therapy.
... These technologies, including focused ultrasound, electromagnetic fields, and intranasal delivery, promise to revolutionize CNS drug delivery. [109] The development of effective, non-invasive, device-mediated techniques for drug delivery across the BBB has been fraught with challenges but illuminated by moments of innovation and breakthrough. As the field progresses, it is imperative to prioritize safety, ensuring that the integrity and function of the BBB are not compromised in the long term. ...
Preprint
Full-text available
Device-mediated, non-invasive drug delivery across the blood-brain barrier (BBB) represents a significant advancement in treating neurological diseases. The BBB is a tightly packed layer of endothelial cells that shields the brain from harmful substances in the blood, allowing necessary nutrients to pass through. It is a highly selective barrier, which poses a challenge to delivering therapeutic agents into the brain. Several non-invasive techniques and devices have been proposed or investigated to enhance drug delivery across the BBB. This paper presents the current state of the art and case studies that address the pharmacology, technology, delivery systems, regulatory approval, ethical concerns, and future possibilities.
... However, disruption of the BBB does not uniformly result in increased drug penetration into the brain, as it does not only increase influx but also facilitates rapid clearance out of the brain [245,246]. Notwithstanding, Nance et al. showed improved delivery of long-circulating PEGylated PS PNPs to the brain using MRI-guided FUS, suggesting that local and temporary BBB disruption in combination with longer circulating NP might improve the in vivo efficacy of administered therapeutics [247]. ...
Article
Full-text available
Primary brain and central nervous system (CNS) tumors are a diverse group of neoplasms that occur within the brain and spinal cord. Although significant advances in our understanding of the intricate biological underpinnings of CNS neoplasm tumorigenesis and progression have been made, the translation of these discoveries into effective therapies has been stymied by the unique challenges presented by these tumors’ exquisitely sensitive location and the body’s own defense mechanisms (e.g., the brain–CSF barrier and blood–brain barrier), which normally protect the CNS from toxic insult. These barriers effectively prevent the delivery of therapeutics to the site of disease. To overcome these obstacles, new methods for therapeutic delivery are being developed, with one such approach being the utilization of nanoparticles. Here, we will cover the current state of the field with a particular focus on the challenges posed by the BBB, the different nanoparticle classes which are under development for targeted CNS tumor therapeutics delivery, and strategies which have been developed to bypass the BBB and enable effective therapeutics delivery to the site of disease.
... For instance, PEGylated PLGA nanoparticles of nanometric size exhibited a prominent brain permeation and prolonged circulation time than bigger nanoparticles, especially 200 and 800 nm nanoparticles [80]. Likewise, it was observed that PEGylated nanoparticles integrated with an ultrasound approach were able to disrupt BBB, which augmented the drug concentration in the normal rat brain by more than 110 nm nanoparticles [81]. Another investigation utilized different sizes of biocompatible NIR nanomaterial (10, 30, and 60 nm) that exhibited insignificant selectivity. ...
Article
The combination of nanotechnology and controlled release techniques is vital to effective drug delivery into the brain. An in-depth and comprehensive overview of nanotechnology applications for bioactive shuttle across the blood brain barrier (BBB) is presented in this review article. Essentially, the article is concerned with tailoring the surface functionalities of nanoparticles such as peptides, proteins, and antibodies for delivery to the brain. Design and structure of nanoparticulate platforms, along with their physicochemical properties, including drug encapsulation, impact permeating the BBB. The use of nanocargoes for specific targeting at the subcellular level, including mitochondria, as well as using the transporter for active targeting (receptor-mediated transcytosis) has been discussed to provide the best knowledge of approaches to improve the effectiveness of delivery platforms, including transferrin, folate, and low-density lipoproteins (LDL) receptors. Various nanocargoes, including lipidic nanocarriers, polymeric nanocarriers, camouflaged nanocarriers (exosomes), and inorganic nanocarriers, are explored to improve therapeutic administration into the brain or modulate therapeutic applications in the diseased brain. The key protein corona effect on nanotherapeutics delivery into the CNS is also thoroughly described in the review. The key protein corona effect on nanotherapeutics delivery into the CNS is also thoroughly described in the review. In conclusion, the review provides a comprehensive overview of recent research investigating nanotechnology as a means of treating brain-related disorders.
... In the laboratory setting, blood-brain barrier permeability is consistently increased through mechanical effects of FUS on injected microbubbles. [28][29][30][31] Using FUS, we were able to achieve enhanced delivery of macromolecules to the distal ischemic penumbra (Figure 2, Figure S1). Achieving enhanced delivery through the use of FUS seems appealing and we show that effective, yet safe, and hemorrhage-free conditions can be established in an animal stroke model ( Figure 3). ...
Article
Full-text available
BACKGROUND Transplantation of autologous mitochondria into ischemic tissue may mitigate injury caused by ischemia and reperfusion. METHODS Using murine stroke models of middle cerebral artery occlusion, we sought to evaluate feasibility of delivery of viable mitochondria to ischemic brain parenchyma. We evaluated the effects of concurrent focused ultrasound activation of microbubbles, which serves to open the blood–brain barrier, on efficacy of delivery of mitochondria. RESULTS Following intraarterial delivery, mitochondria distribute through the stroked hemisphere and integrate into neural and glial cells in the brain parenchyma. Consistent with functional integration in the ischemic tissue, the transplanted mitochondria elevate concentration of adenosine triphosphate in the stroked hemisphere, reduce infarct volume and increase cell viability. The addition of focused ultrasound leads to improved blood–brain barrier opening without hemorrhagic complications. CONCLUSION Our results have implications for the development of interventional strategies after ischemic stroke and suggest a novel potential modality of therapy after mechanical thrombectomy.
... 46 In addition, MR-guided focused ultrasound can reversibly disrupt the blood-brain barrier and facilitate the delivery of polystyrene NPs. 47 Notably, the application of external fields is commonly accompanied with changes in the temperature of treated tissues, which may affect NP transport and NP-cell interactions. Generally, the experimental research on the transport and adhesion behaviors of NPs is commonly carried out in an artificial blood vessel. ...
Article
Full-text available
Background A good understanding of the adhesion behaviors of the nanocarriers in microvessels in chemo-hyperthermia synergistic therapy is conducive to nanocarrier design for targeted drug delivery. Methods In this study, we constructed an artificial blood vessel system using gelatins with a complete endothelial monolayer formed on the inner vessel wall. The numbers of adhered NPs under different conditions were measured, as well as the interaction forces between the arginine–glycine–aspartic acid (RGD) ligands and endothelial cells. Results The experimental results on the adhesion of ligand–coated nanoparticles (NPs) with different sizes and morphologies in the blood vessel verified that the gelatin-based artificial vessel possessed good cytocompatibility and mechanical properties, which are suitable for the investigation on NP adhesion characteristics in microvessels. When the temperature deviated from 37 °C, an increase or decrease in temperature resulted in a decrease in the number of adhered NPs, but the margination probability of NP adhesion increased at high temperatures due to the enhanced Brownian movement and flow disturbance. It is found that the effect of cooling was less than that of heating according to the observed changes in cell morphology and a decrease in cell activity under the static and perfusion culture conditions within the temperature range of 25 °C–43 °C. Furthermore, the measurement results of change in the RGD ligand-cell interaction with temperature showed good agreement with those in the number of adhered NPs. Conclusion The Findings suggest that designing ligands that can bind to the receptor and are least susceptible to temperature variation can be an effective means to enhance drug retention.
... BBB-crossing drug delivery in a relatively mild way. 91 Apart from the general advantages about the flexibility in surface modification and controllability to load and release drugs, 114,115 NPs are able to target microglia due to the intrinsic capability of microglia to internalize foreign bodies. 116,117 The microglial targeting extent of NPs can be modulated by their dimension and surface charge as well. ...
Article
Traumatic central nervous system (CNS) injuries, including spinal cord injury and traumatic brain injury, are challenging enemies of human health. Microglia, the main component of the innate immune system in CNS, can be activated postinjury and are key participants in the pathological procedure and development of CNS trauma. Activated microglia can be typically classified into pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. Reducing M1 polarization while promoting M2 polarization is thought to be promising for CNS injury treatment. However, obstacles such as the low permeability of the blood-brain barrier and short retention time in circulation limit the therapeutic outcomes of administrated drugs, and rational delivery strategies are necessary for efficient microglial regulation. To this end, proper administration methods and delivery systems like nano/microcarriers and scaffolds are investigated to augment the therapeutic effects of drugs, while some of these delivery systems have self-efficacies in microglial manipulation. Besides, systems based on cell and cell-derived exosomes also show impressive effects, and some underlying targeting mechanisms of these delivery systems have been discovered. In this review, we introduce the roles of microglia play in traumatic CNS injuries, discuss the potential targets for the polarization regulation of microglial phenotype, and summarize recent studies and clinical trials about delivery strategies on enhancing the effect of microglial regulation and therapeutic outcome, as well as targeting mechanisms post CNS trauma.
... Recombinant tissue plasminogen activator (rt-PA) is currently the only FDAapproved therapy [36]. In addition, the systemic delivery of candidate neural repair drugs is limited by the BBB and off-target effects; thus, promising candidate therapies for neural repair with potential clinical application value are limited [37]. For example, delivering promising therapeutic growth factors to the brain is very difficult [38]. ...
Article
Full-text available
Ischemic stroke is a major cause of death and disability worldwide. There is almost no effective treatment for this disease. Therefore, developing effective treatment for ischemic stroke is urgently needed. Efficient delivery of therapeutic drugs to ischemic sites remained a great challenge for improved treatment of strokes. In recent years, hydrogel-based strategies have been widely investigated for new and improved therapies. They have the advantage of delivering therapeutics in a controlled manner to the poststroke sites, aiming to enhance the intrinsic repair and regeneration. In this review, we discuss the pathophysiology of stroke and the development of injectable hydrogels in the application of both stroke treatment and neural tissue engineering. We also discuss the prospect and the challenges of hydrogels in the treatment of ischemic strokes.
... Thus, to achieve favorable delivery outcomes without tissue damaging, we did not choose higher or lower peak negative pressures than 0.53 MPa. This value was quite close to the one (0.55 MPa) used in another study investigating the effect of FUS/MB on the transport of nanoparticles in the glioma tissue because they also found that 16% of locations sonicated with 0.6 MPa developed red blood cell leakage [36,37]. ...
... However, there is limited literature regarding the impact of acoustic pressures on the penetration distance of nanoparticles. Nance et al. found that the extravasation areas of 60-nm nanoparticles were dependent on the acoustic pressure in normal rat brains [36]. Higher FUS pressure (0.6 MPa) increased the coverage within the interstitium outside the vasculature by nearly two-fold when compared to the lower pressure (0.4 MPa). ...
Article
DNA nanostructures, with good biosafety, highly programmable assembly, flexible modification, and precise control, are tailored as drug carriers to deliver therapeutic agents for cancer therapy. However, they face considerable challenges regarding their delivery into the brain, mainly due to the blood-brain barrier (BBB). By controlling the acoustic parameters, focused ultrasound combined with microbubbles (FUS/MB) can temporarily, noninvasively, and reproducibly open the BBB in a localized region. We investigated the delivery outcome of pH-responsive DNA octahedra loading Epirubicin ([email protected]) via FUS/MB and its therapeutic efficiency in a mouse model bearing intracranial glioma xenograft. Using FUS/MB to locally disrupt the BBB or the blood-tumor barrier (BTB) and systemic administration of [email protected] ([email protected] + FUS/MB) (2 mg/kg of loaded Epr), we achieved an Epr concentration of 292.3 ± 10.1 ng/g tissue in glioma, a 4.4-fold increase compared to unsonicated animals (p < 0.001). The in vitro findings indicated that Epr released from DNA strands accumulated in lysosomes and induced enhanced cytotoxicity compared to free Epr. Further two-photon intravital imaging of spatiotemporal patterns of the DNA-Octa leakage revealed that the FUS/MB treatment enhanced DNA-Octa delivery across several physiological barriers at microscopic level, including the first extravasation across the BBB/BTB and then deep penetration into the glioma center and engulfment of DNA-Octa into the tumor cell body. Longitudinal in vivo bioluminescence imaging and histological analysis indicated that the intracranial glioma progression in nude mice treated with [email protected] + FUS/MB was effectively retarded compared to other groups. The beneficial effect on survival was most significant in the [email protected] + FUS/MB group, with a 50% increase in median survival and a 73% increase in the maximum survival compared to control animals. Our work demonstrates the potential viability of FUS/MB as an alternative strategy for glioma delivery of anticancer drugs using DNA nanostructures as the drug delivery platform for brain cancer therapy.