The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×

Abstract

In the past, psychotherapy and neuropharmacological approaches have been the most common treatments for disordered thoughts, moods, and behaviors. One new path of brain therapeutics is in the deployment of noninvasive approaches designed to reprogram brain function at the cellular level. Treatment at the cellular level may be considered for a wide array of disorders, ranging from mood disorders to neurodegenerative disorders. Brain-targeted biological therapy may provide minimally invasive and accurate delivery of treatment. The present article discusses the hurdles and advances that characterize the pathway to this goal.

Over the years, therapy to change aberrant thoughts, moods, and behaviors has largely deployed techniques that use verbal communication or neuropharmacological manipulations. Rarely, invasive neurosurgical interventions have been proposed. There is a hope that noninvasive approaches may be created that will aim to reprogram brain function at the cellular level, so that dysfunction caused by inflammation, intoxication, and genetic aberration may be rectified in the treatment of depression, anxiety, dementia, and other neuropsychiatric disorders. To date, there have been few attempts to reprogram brain function, most of which have used invasive approaches. Stem cells have been implanted to restore mobility in Parkinsonism (1). Stem cells have been injected to revive the brain after stroke in humans (2). Intraventricular stem cells have improved memory in subjects with Alzheimer’s (3, 4). In aging mice, exosome injection into the hypothalamus appears to restore youth, as if the biological clock of aging may be reset (5). Although all these demonstrations promise renewal, widespread application of brain-targeted biological therapy will require minimally invasive, inexpensive, and accurate delivery systems. The present article discusses the challenges and triumphs that characterize the pathway to this goal.

Emerging Techniques for Facilitating Delivery of Therapeutic Substances Across the Blood-Brain Barrier

The interfaces of the blood vessels with parenchyma, and of blood vessels with cerebrospinal fluid, present relative barriers to the ingress of small molecules, oligonucleotides, particulates, and cellular elements into the brain (6). Small molecules may have specific carrier systems that allow for transendothelial transport (7). Phosphorothioate oligonucleotides, chemically modified to ensure nuclease resistance, are reportedly transported across the blood-brain barrier (BBB) through yet unidentified transporters, albeit with limited efficiency (8, 9). Transcytotic mechanisms also exist for larger materials, such as large proteins, exosomes, microbes, and immune cells (10). In the latter circumstances, there are specific ligands that allow luminal attachment and a complex sequence of events that eventuate in endo-vesicular delivery to the parenchymal side of the barrier.

The integrity of the tight intercellular junctions plays a role in essentially excluding bloodborne elements that are toxic to the brain, including glutamate, prothrombin, and plasminogen (6). The ionic channels active in endothelial membranes are also tasked with providing a relatively stable concentration of calcium and potassium in the brain interstitium, which is required for the normal electrophysiological function of neurons (11). By contrast, blood concentrations of these elements may fluctuate under differing physiological conditions, including fasting or aerobic exercise (12), and engagement during motor, affective, and cognitive tasks. Cerebral ischemic neuronal dysfunction can follow the incorrect balance of neural ions (13), and profound or prolonged disruption of these systems can lead to neurodegeneration (14). Therefore, successful delivery of therapeutics to the brain through the BBB must be accomplished without causing permanent damage or disruption to these systems.

One such mechanism for increasing permeability of the BBB for the delivery of therapeutics to the brain is transcranial focused ultrasound. Various forms of ultrasound equipment are available for human treatment and investigation, ranging from multiprobe devices used in a magnetic resonance imaging (MRI) environment, to single probe devices applied in the temple region. Transtemporal probes are targetable with integrated Doppler technology (15) or with optical tracking devices. Before the development of clot retrieval devices (16, 17), ultrasound delivered by a transtemporal window was used extensively for mechanical clot fragmentation and for improved blood flow, along with intravenous clot-dissolving agents (18). Safety has been established at frequencies of 2 MHz, but an increased risk of brain hemorrhage has characterized more penetrating, lower frequency treatment (19). Earlier work (20) on the opening of tight junctions after acute stroke or trauma has afforded delivery of stem cells to the brain through intravenous infusion. Mechanical effects of sonification have been used to pry open tight junctions after microbubble infusion (21). The latter technique presents significant safety issues, because the tight junction opening may lead to intracerebral hemorrhage (22, 23). Thus, increasing permeability of the BBB without using microbubbles has been shown to be effective. This experience with ultrasound treatment has suggested that such technology may have several effects on brain tissue, including temporary alteration of vascular properties (24, 25). Indeed, ultrasound has been shown to increase the adhesiveness of the luminal surface of capillaries (26, 27), fostering potential noninvasive delivery to targeted brain tissue (21, 2832). Furthermore, in our experience of focused ultrasound in humans, blood flow can be increased up to eightfold, while also monitoring effects with quantitative arterial spin labeling (33). In the latter circumstance, poorly penetrating agents may be more efficiently delivered by increasing local capillary delivery. In other words, ultrasound may be used to facilitate focal delivery without risking injury to blood vessel integrity (34).

Wish List of Biologicals and Delivery Systems

Stem cells may be given intravenously and then channeled to specific brain sites by using focused ultrasound. Much of the early research with stem cells in neurological conditions used fetal progenitors of unspecialized mesenchymal stem cells from autologous or allogeneic sources (3537). Although transplanted stem cells in small numbers may survive to a certain extent, most will die while releasing packets of nucleic acids (e.g., microRNA, messenger RNA, genomic and mitochondrial DNA) and growth factors in the form of exosomes (3840). There are several advantages to using stem cells, including their quality to migrate and home in on areas of inflammation and hypoxia in tumors (4143). Furthermore, when they persist in tissue, they may provide a more extended period of exosome secretion, allowing for more effective delivery of exosome-encapsulated molecules. These qualities have been used to advantage in treating chronic stroke (44, 45), traumatic brain injury, and in earlier work in treating Parkinson’s disease (46, 47). The ability to migrate, home in on, and contain strategic cargo, such as lethal virions or prodrug-converting enzymes, has encouraged stem cell therapy to be utilized as a Trojan horse in the search and destruction of gliomas (42, 4850).

A disadvantage to the use of stem cells is the relative frailty of these therapeutic agents, which presents challenges to storage, transportation, reconstitution, and deployment (50). As an alternative, there has been increased interest in the use of exosomes (5154). Exosomes are not motile, but they can diffuse through the interstitium and, with specific surface ligands, they stick to receptive cell surfaces (55, 56). The pioneering study by Alvarez-Erviti et al. (57) provided a blueprint for such targeted and exosome-dependent delivery of therapeutic short interfering RNA (siRNA) into neurons, microglia, and oligodendrocytes through intravenous administration. The exosomes were derived from dendritic cells engineered to secrete extracellular vesicles with neuron-specific rabies viral glycoprotein peptide on the surface and electroporated with therapeutic siRNAs before administration (57). Manufactured liposomes may also act in similar ways when coated with relevant ligands (58, 59). Lipid-modified ligands, such as DNA or RNA aptamers, provide a simpler strategy to modify exosome surface and thus target specificity by incorporating the ligand into the exosomal membrane (6062). Because exosomes get into the brain by transcellular mechanisms from the bloodstream (63), no forceful opening of tight junctions is required (34). The packaging of therapeutic elements inside a lipid shell, such as an exosome, is likely to protect these elements from the enzymatic degradation that would likely occur with an unprotected bloodborne state.

Early efforts at stem cell and exosome therapy have often used regenerative elements from fetal or youthful donors. The initial hope was that these harvested elements would already complement progrowth and anti-inflammatory factors that would be useful in deployment. There is increasing interest in identifying and deploying natural or synthetic nucleic acid cargo that has specific actions on the recipient. The required effect will depend on the necessities of the condition being treated; desired outcomes may range from stimulating proliferation to suppressing inflammation (64, 65) to the promotion of intracellular processes, such as autophagy (66).

Targets that are based on clinical research experience with deep brain stimulation (DBS), such as the nucleus accumbens or the subgenual cingulate, may be considered for ultrasound-facilitated delivery of exosomes to treat refractory depression (67, 68). Degenerative conditions, such as Alzheimer’s disease or Parkinson’s disease, may be treated with exosome delivery to affected structures, such as the nucleus basalis of Meynert (69) and substantia nigra (70). Exosomes from youthful donors could be delivered to the hypothalamus to reverse aging-related frailty (5).

Other small molecules can be delivered to the brain by using focused ultrasound. Bosutinib, which has relatively poor intrinsic brain penetration, has been used along with ultrasound to treat Alzheimer’s disease and Parkinson’s dementia (71). Lipids generally fare better than hydrophilic substances in terms of brain penetration (72). Nevertheless, facilitated delivery to areas at risk for inflammation and degeneration may be targeted effectively, as in the delivery of plasmalogen precursors (73, 74) to treat Alzheimer’s disease and Parkinson’s disease. The anti-inflammatory effects of the docosahexaenoic acid (DHA) component of the plasmalogen precursor or the coadministration of curcumin extracts may be considered for targeting the nucleus accumbens of the subgenual cingulate in treating depression. Peptides, such as TREK-1 inhibitors, have been considered as treatments for mood disorders (75, 76). The short half-lives of these agents, due in part to renal clearance, limit their clinical applications. Facilitated delivery of peptides with focused ultrasound may be one strategy that may circumvent the limitations of short plasma half-lives (7780).

Notably, the effects of ultrasound go beyond circumvention of the BBB. Ultrasound appears to produce mechanical effects on glial cells and neurons; mechanical transduction changes the biochemistry and physiology of nuclear, cytosolic, and membrane components, with potentially long-lasting impacts on targeted cells (19, 20, 81, 82). The potential use of ultrasound as a direct stand-alone therapy is the subject of other work.

Other Techniques to Deliver Biologicals to the Brain

For many years, mannitol infusions have been used to shrink endothelial cells (83, 84), thereby opening spaces between them to allow for entry of small (85) and large (86, 87) therapeutic agents (88) into the brain. There is little control over targeting with this technique, unless by combining with real-time monitoring of injection via advanced MRI (89). Nasal insufflation has also been proposed (9092). Nasal application presents an opportunity for rapid systemic uptake, with mucosal vascularity making this an ideal solution for drug delivery among children and those with poor intravenous access. Depending on the insufflation delivery device, some product may be delivered to the underside of the cribriform plate (93, 94) at the apex of the nasal cavity; numerous devices have been designed to allow targeted delivery to the olfactory mucosa (95). At this location, potential cerebrospinal fluid entry is available; however, with age and even more so in those with anosmia, the nose-to-brain pathways are potentially compromised. Uptake may be through olfactory nerves and trigeminal afferents; assuming that the latter pathways may be questionably patent among elderly individuals, product will likely be delivered off target to traverse the orbitofrontal cortex and brain stem, respectively. Pharmacokinetic studies have generally demonstrated very low penetration of substances through these paths. Producing carrier systems with improved penetrance is an area of active research (96).

In the meantime, there are other targetable energy sources that may play a role in the delivery of biologicals. Transcranial magnetic stimulation (TMS) is used extensively as a stand-alone therapy for depression (9799) and obsessive-compulsive disorder (100, 101). After stimulation, blood flow to the targeted area is temporarily increased (102, 103), allowing for facilitated delivery of biologicals. A recent study (104) found successful delivery of tracers across the BBB with high amplitude repetitive transcranial magnetic stimulation (rTMS), in which large areas are potentially treated with rTMS; however, precision deployment and depth penetration beyond 3 cm are severely limited, making this energy source problematic for access to deep nuclear groups. Laser therapy at 1,024 nm has better potential penetration (down to more than 7 cm), and it is free of scalp stimulation that further limits rTMS deployment (105, 106). The use of multiple probes may enhance physiological changes and focal effects in deeper sites. Research relating to probe design and optimization of laser dosage is needed to understand the extent laser therapy may have in the future of delivery of biologicals to the brain (107). Microwave sources may also be considered for widespread and superficial delivery, but relevant research on this application is in its infancy (108, 109). There is potential in the combination of technologies, such as microwave and near infrared, for controlled drug delivery, which may be useful in getting medicine across the BBB (108). More research is needed to investigate the use of TMS, laser therapy, and microwave therapy in delivery of biologicals.

Successful Symptom-Based Neuronavigation

As previously stated, the success of therapeutics relies heavily on the accuracy of targeted delivery to the incipient region. By improving permeability of the BBB, ultrasound and other energy mechanisms offer the ability to improve the delivery of drugs and molecules. However, the accuracy of this delivery is directly proportional to the spatial resolution and precision of the method used to target the energy device. Until recently, TMS was conducted in the United Stated without any neuroimaging (97, 99, 100). Only recently has structural and functional neuronavigation been applied to TMS and other neuromodulation treatments (110112), and the clinical outcomes have improved in direct accordance with the improved targeting of the brain structures (98, 105, 113). As this field continues to evolve, the use of multimodal neuroimaging to ensure accurate and successful targeting of specific brain regions will be of paramount importance to ensuring both safety and clinical outcomes. With the ability to target structural and functional regions with high specificity comes the ability to choose clinically relevant targets. Much of DBS requires a specific nucleus as the target; neuronavigated drug and molecule delivery will allow for small brain structures and nuclei to be identified as targets for therapeutic delivery. The targeting of these structures is the subject of another article within this issue.

Conclusion

The application of biological therapy to neuropsychiatric illness will likely result in an expansion of the therapeutic armamentarium. Several challenges to the use of biologicals include high cost, variable brain penetration, and uncertain target delivery. Focused ultrasound and other targetable energy sources promise to meet these challenges. More efficient delivery would reduce cost by enabling the potential use of lower systemic doses. Brain penetration may be improved by local enhancement of blood flow, facilitation of capillary adhesion, and stimulation of transcellular transport mechanisms. Specific target delivery is enhanced by aiming the energy source at network nodes that appear to be part of the relevant pathophysiology.

Department of Neurology, University of California, Los Angeles and Department of Neurology, University of Southern California, Los Angeles (Jordan);Neurological Associates—The Interventional Group, Los Angeles (Jordan, Zielinski);Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Los Angeles (Kortylewski);Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Kuhn, Bystritsky).
Send correspondence to Dr. Jordan ().

Dr. Jordan reports owning a focused ultrasound device and a transcranial magnetic stimulation device that may be used in some of the research discussed. Dr. Jordan also reports being part owner and member of the board of Synaptic Research, which performs research on focused ultrasound delivery of substances to the brain.

Dr. Kortylewski reports serving on the Scientific Advisory Board of Scopus BioPharma and its subsidiary Duet Therapeutics. Dr. Bystritsky reports being chief executive officer of BrainSonix Corp., president of Westside Neurotherapeutics, president of the Institute for Advanced Consciousness Studies, a partner at CalNeuro Research Group, president of the Pacific Institute for Medical Research, and a partner and board member of Synaptic Research. The other authors report no financial relationships with commercial interests.

References

1 Yasuhara T, Shingo T, Date I: Glial cell line-derived neurotrophic factor (GDNF) therapy for Parkinson’s disease. Acta Med Okayama 2007; 61:51–56Google Scholar

2 Borlongan CV: Concise review: stem cell therapy for stroke patients: are we there yet? Stem Cells Transl Med 2019; 8:983–988CrossrefGoogle Scholar

3 McGinley LM, Kashlan ON, Bruno ES, et al.: Human neural stem cell transplantation improves cognition in a murine model of Alzheimer’s disease. Sci Rep 2018; 8:14776CrossrefGoogle Scholar

4 Wang SM, Lee CU, Lim HK: Stem cell therapies for Alzheimer’s disease: is it time? Curr Opin Psychiatry 2019; 32:105–116CrossrefGoogle Scholar

5 Zhang Y, Kim MS, Jia B, et al.: Hypothalamic stem cells control ageing speed partly through exosomal miRNAs. Nature 2017; 548:52–57CrossrefGoogle Scholar

6 Sharif Y, Jumah F, Coplan L, et al.: Blood brain barrier: a review of its anatomy and physiology in health and disease. Clin Anat 2018; 31:812–823CrossrefGoogle Scholar

7 Pulido RS, Munji RN, Chan TC, et al.: Neuronal activity regulates blood-brain barrier efflux transport through endothelial circadian genes. Neuron 2020; 108:937–952.e7CrossrefGoogle Scholar

8 Banks WA, Farr SA, Butt W, et al.: Delivery across the blood-brain barrier of antisense directed against amyloid beta: reversal of learning and memory deficits in mice overexpressing amyloid precursor protein. J Pharmacol Exp Ther 2001; 297:1113–1121Google Scholar

9 Bennett CF, Krainer AR, Cleveland DW: Antisense oligonucleotide therapies for neurodegenerative diseases. Annu Rev Neurosci 2019; 42:385–406CrossrefGoogle Scholar

10 Mäger I, Meyer AH, Li J, et al.: Targeting blood-brain-barrier transcytosis—perspectives for drug delivery. Neuropharmacology 2017; 120:4–7CrossrefGoogle Scholar

11 Santin JM, Schulz DJ: Membrane voltage is a direct feedback signal that influences correlated ion channel expression in neurons. Curr Biol 2019; 29:1683–1688.e2CrossrefGoogle Scholar

12 Jiménez-Maldonado A, Rentería I, García-Suárez PC, et al.: The impact of high-intensity interval training on brain derived neurotrophic factor in brain: a mini-review. Front Neurosci 2018; 12:839CrossrefGoogle Scholar

13 Yamazaki Y, Harada S, Wada T, et al.: Sodium influx through cerebral sodium-glucose transporter type 1 exacerbates the development of cerebral ischemic neuronal damage. Eur J Pharmacol 2017; 799:103–110CrossrefGoogle Scholar

14 Cunnane SC, Trushina E, Morland C, et al.: Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov 2020; 19:609–633CrossrefGoogle Scholar

15 Rabut C, Yoo S, Hurt RC, et al.: Ultrasound technologies for imaging and modulating neural activity. Neuron 2020; 108:93–110CrossrefGoogle Scholar

16 Chaplin V, Phipps MA, Jonathan SV, et al.: On the accuracy of optically tracked transducers for image-guided transcranial ultrasound. Int J CARS 2019; 14:1317–1327CrossrefGoogle Scholar

17 Hameed A, Zafar H, Mylotte D, et al.: Recent trends in clot retrieval devices: a review. Cardiol Ther 2017; 6:193–202CrossrefGoogle Scholar

18 Prada F, Kalani MYS, Yagmurlu K, et al.: Applications of focused ultrasound in cerebrovascular diseases and brain tumors. Neurotherapeutics 2019; 16:67–87CrossrefGoogle Scholar

19 Blackmore J, Shrivastava S, Sallet J, et al.: Ultrasound neuromodulation: a review of results, mechanisms and safety. Ultrasound Med Biol 2019; 45:1509–1536CrossrefGoogle Scholar

20 Baek H, Pahk KJ, Kim H: A review of low-intensity focused ultrasound for neuromodulation. Biomed Eng Lett 2017; 7:135–142CrossrefGoogle Scholar

21 Wu SK, Chu PC, Chai WY, et al.: Characterization of different microbubbles in assisting focused ultrasound-induced blood-brain barrier opening. Sci Rep 2017; 7:46689CrossrefGoogle Scholar

22 McMahon D, Poon C, Hynynen K: Evaluating the safety profile of focused ultrasound and microbubble-mediated treatments to increase blood-brain barrier permeability. Expert Opin Drug Deliv 2019; 16:129–142CrossrefGoogle Scholar

23 Wong KH, Riaz MK, Xie Y, et al.: Review of current strategies for delivering Alzheimer’s disease drugs across the blood-brain barrier. Int J Mol Sci 2019; 20:381CrossrefGoogle Scholar

24 Fomenko A, Neudorfer C, Dallapiazza RF, et al.: Low-intensity ultrasound neuromodulation: an overview of mechanisms and emerging human applications. Brain Stimul 2018; 11:1209–1217CrossrefGoogle Scholar

25 Fini M, Tyler WJ: Transcranial focused ultrasound: a new tool for non-invasive neuromodulation. Int Rev Psychiatry 2017; 29:168–177CrossrefGoogle Scholar

26 Poon C, Pellow C, Hynynen K: Neutrophil recruitment and leukocyte response following focused ultrasound and microbubble mediated blood-brain barrier treatments. Theranostics 2021; 11:1655–1671CrossrefGoogle Scholar

27 Man VH, Li MS, Derreumaux P, et al.: Molecular mechanism of ultrasound interaction with a blood brain barrier model. J Chem Phys 2020; 153:045104CrossrefGoogle Scholar

28 Janowicz PW, Leinenga G, Götz J, et al.: Ultrasound-mediated blood-brain barrier opening enhances delivery of therapeutically relevant formats of a tau-specific antibody. Sci Rep 2019; 9:9255CrossrefGoogle Scholar

29 Morse SV, Pouliopoulos AN, Chan TG, et al.: Rapid short-pulse ultrasound delivers drugs uniformly across the murine blood-brain barrier with negligible disruption. Radiology 2019; 291:459–466CrossrefGoogle Scholar

30 Valdez MA, Fernandez E, Matsunaga T, et al.: Distribution and diffusion of macromolecule delivery to the brain via focused ultrasound using magnetic resonance and multispectral fluorescence imaging. Ultrasound Med Biol 2020; 46:122–136CrossrefGoogle Scholar

31 Ozdas MS, Shah AS, Johnson PM, et al.: Non-invasive molecularly-specific millimeter-resolution manipulation of brain circuits by ultrasound-mediated aggregation and uncaging of drug carriers. Nat Commun 2020; 11:4929CrossrefGoogle Scholar

32 Conaty P, Sherman LS, Naaldijk Y, et al.: Methods of mesenchymal stem cell homing to the blood-brain barrier; in Somatic Stem Cells. Edited by Singh SR, Rameshwar P. New York, Humana Press, 2018CrossrefGoogle Scholar

33 Becerra S, Duncan J, Jordan S, et al.: Case study: comparison of MRI techniques for demonstrating successful ultrasound targeting: BOLD compared with ASL functional imaging. Brain Stimul 2019; 12:547CrossrefGoogle Scholar

34 Alptekin A, Khan MB, Ara R, et al.: Pulsed focal ultrasound as a non-invasive method to deliver exosomes in the brain/stroke. J Biomed Nanotechnol 2021; 17:1170–1183CrossrefGoogle Scholar

35 Prinz M, Masuda T, Wheeler MA, et al.: Microglia and central nervous system-associated macrophages—from origin to disease modulation. Annu Rev Immunol 2021; 39:251–277CrossrefGoogle Scholar

36 Abe Y, Ochiai D, Sato Y, et al.: Amniotic fluid stem cells as a novel strategy for the treatment of fetal and neonatal neurological diseases. Placenta 2021; 104:247–252CrossrefGoogle Scholar

37 Willing AE, Das M, Howell M, et al.: Potential of mesenchymal stem cells alone, or in combination, to treat traumatic brain injury. CNS Neurosci Ther 2020; 26:616–627CrossrefGoogle Scholar

38 Vogel A, Upadhya R, Shetty AK: Neural stem cell derived extracellular vesicles: attributes and prospects for treating neurodegenerative disorders. EBioMedicine 2018; 38:273–282CrossrefGoogle Scholar

39 Yasuhara T, Kawauchi S, Kin K, et al.: Cell therapy for central nervous system disorders: current obstacles to progress. CNS Neurosci Ther 2020; 26:595–602CrossrefGoogle Scholar

40 Al-Kharboosh R, ReFaey K, Lara-Velazquez M, et al.: Inflammatory mediators in glioma microenvironment play a dual role in gliomagenesis and mesenchymal stem cell homing: implication for cellular therapy. Mayo Clin Proc Innov Qual Outcomes 2020; 4:443–459CrossrefGoogle Scholar

41 Gupta A, Cady C, Fauser AM, et al.: Cell-free stem cell-derived extract formulation for regenerative medicine applications. Int J Mol Sci 2020; 21:9364CrossrefGoogle Scholar

42 Mooney R, Hammad M, Batalla-Covello J, et al.: Concise review: neural stem cell-mediated targeted cancer therapies. Stem Cells Transl Med 2018; 7:740–747. doi: 10.1002/sctm.18-0003CrossrefGoogle Scholar

43 Kim HY, Kim TJ, Kang L, et al.: Mesenchymal stem cell-derived magnetic extracellular nanovesicles for targeting and treatment of ischemic stroke. Biomaterials 2020; 243:119942CrossrefGoogle Scholar

44 Lalu MM, Montroy J, Dowlatshahi D, et al.: From the lab to patients: a systematic review and meta-analysis of mesenchymal stem cell therapy for stroke. Transl Stroke Res 2020; 11:345–364CrossrefGoogle Scholar

45 Fan Y, Winanto, Ng SY: Replacing what’s lost: a new era of stem cell therapy for Parkinson’s disease. Transl Neurodegener 2020; 9:2CrossrefGoogle Scholar

46 Doi D, Magotani H, Kikuchi T, et al.: Pre-clinical study of induced pluripotent stem cell-derived dopaminergic progenitor cells for Parkinson’s disease. Nat Commun 2020; 11:3369CrossrefGoogle Scholar

47 Ibarra LE: Cellular Trojan horses for delivery of nanomedicines to brain tumors: where do we stand and what is next? Nanomedicine (Lond) 2021; 16:517–522CrossrefGoogle Scholar

48 Luo M, Zhou Y, Gao N, et al.: Mesenchymal stem cells transporting black phosphorus-based biocompatible nanospheres: active Trojan horse for enhanced photothermal cancer therapy. Chem Eng J 2020; 385:123942CrossrefGoogle Scholar

49 Hadryś A, Sochanik A, McFadden G, et al.: Mesenchymal stem cells as carriers for systemic delivery of oncolytic viruses. Eur J Pharmacol 2020; 874:172991CrossrefGoogle Scholar

50 Krueger TEG, Thorek DLJ, Denmeade SR, et al.: Concise review: mesenchymal stem cell‐based drug delivery: the good, the bad, the ugly, and the promise. Stem Cells Transl Med 2018; 7:651–663CrossrefGoogle Scholar

51 Kalluri R, LeBleu VS: The biology, function, and biomedical applications of exosomes. Science 2020b; 367:eaau6977. doi: 10.1126/science.aau6977CrossrefGoogle Scholar

52 Donoso-Quezada J, Ayala-Mar S, González-Valdez J: State-of-the-art exosome loading and functionalization techniques for enhanced therapeutics: a review. Crit Rev Biotechnol 2020; 40:804–820CrossrefGoogle Scholar

53 Qi Y, Guo L, Jiang Y, et al.: Brain delivery of quercetin-loaded exosomes improved cognitive function in AD mice by inhibiting phosphorylated tau-mediated neurofibrillary tangles. Drug Deliv 2020; 27:745–755CrossrefGoogle Scholar

54 Lai N, Wu D, Liang T, et al.: Systemic exosomal miR-193b-3p delivery attenuates neuroinflammation in early brain injury after subarachnoid hemorrhage in mice. J Neuroinflammation 2020; 17:74CrossrefGoogle Scholar

55 Gurung S, Perocheau D, Touramanidou L, et al.: The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal 2021; 19:47CrossrefGoogle Scholar

56 Hu Q, Su H, Li J, et al.: Clinical applications of exosome membrane proteins. Precis Clin Med 2020; 3:54–66CrossrefGoogle Scholar

57 Alvarez-Erviti L, Seow Y, Yin H, et al.: Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol 2011; 29:341–345CrossrefGoogle Scholar

58 Eroğlu İ, İbrahim M: Liposome-ligand conjugates: a review on the current state of art. J Drug Target 2020; 28:225–244CrossrefGoogle Scholar

59 Filipczak N, Pan J, Yalamarty SSK, et al.: Recent advancements in liposome technology. Adv Drug Deliv Rev 2020; 156:4–22CrossrefGoogle Scholar

60 Zou J, Shi M, Liu X, et al.: Aptamer-functionalized exosomes: elucidating the cellular uptake mechanism and the potential for cancer-targeted chemotherapy. Anal Chem 2019; 91:2425–2430CrossrefGoogle Scholar

61 Terstappen GC, Meyer AH, Bell RD, et al.: Strategies for delivering therapeutics across the blood-brain barrier. Nat Rev Drug Discov 2021; 20:362–383CrossrefGoogle Scholar

62 Gudbergsson JM, Jønsson K, Simonsen JB, et al.: Systematic review of targeted extracellular vesicles for drug delivery—considerations on methodological and biological heterogeneity. J Control Release 2019; 306:108–120CrossrefGoogle Scholar

63 Tashima T: Smart strategies for therapeutic agent delivery into brain across the blood-brain barrier using receptor-mediated transcytosis. Chem Pharm Bull (Tokyo) 2020; 68:316–325CrossrefGoogle Scholar

64 Kahroba H, Davatgaran-Taghipour Y: Exosomal Nrf2: from anti-oxidant and anti-inflammation response to wound healing and tissue regeneration in aged-related diseases. Biochimie 2020; 171–172:103–109CrossrefGoogle Scholar

65 Schwarzenbach H, Gahan PB: Exosomes in immune regulation. Noncoding RNA 2021; 7:4Google Scholar

66 Gu X, Li Y, Chen K, et al.: Exosomes derived from umbilical cord mesenchymal stem cells alleviate viral myocarditis through activating AMPK/mTOR-mediated autophagy flux pathway. J Cell Mol Med 2020; 24:7515–7530CrossrefGoogle Scholar

67 Silva-Dos-Santos A, Sales M, Sebastião A, et al.: A new viewpoint on the etiopathogenesis of depression: insights from the neurophysiology of deep brain stimulation in Parkinson’s disease and treatment-resistant depression. Front Psychiatry 2021; 12:607339CrossrefGoogle Scholar

68 Khairuddin S, Ngo FY, Lim WL, et al.: A decade of progress in deep brain stimulation of the subcallosal cingulate for the treatment of depression. J Clin Med 2020; 9:3260CrossrefGoogle Scholar

69 Hanna Al-Shaikh FS, Duara R, Crook JE, et al.: Selective vulnerability of the nucleus basalis of Meynert among neuropathologic subtypes of Alzheimer disease. JAMA Neurol 2020; 77:225–233CrossrefGoogle Scholar

70 Delva A, Van Weehaeghe D, Koole M, et al.: Loss of presynaptic terminal integrity in the substantia nigra in early Parkinson’s disease. Mov Disord 2020; 35:1977–1986CrossrefGoogle Scholar

71 Mahdavi KD, Jordan SE, Barrows HR, et al.: Treatment of dementia with bosutinib: an open-label study of a tyrosine kinase inhibitor. Neurol Clin Pract 2021; 11:e294–e302CrossrefGoogle Scholar

72 Salem LH, El-Feky GS, Fahmy RH, et al.: Coated lipidic nanoparticles as a new strategy for enhancing nose-to-brain delivery of a hydrophilic drug molecule. J Pharm Sci 2020; 109:2237–2251CrossrefGoogle Scholar

73 Fujino T, Yamada T, Asada T, et al.: Efficacy and blood plasmalogen changes by oral administration of plasmalogen in patients with mild Alzheimer’s disease and mild cognitive impairment: a multicenter, randomized, double-blind, placebo-controlled trial. EBioMedicine 2017; 17:199–205CrossrefGoogle Scholar

74 Su XQ, Wang J, Sinclair AJ: Plasmalogens and Alzheimer’s disease: a review. Lipids Health Dis 2019; 18:100CrossrefGoogle Scholar

75 Wu F, Sun H, Gong W, et al.: Genetic and pharmacological inhibition of two-pore domain potassium channel TREK-1 alters depression-related behaviors and neuronal plasticity in the hippocampus in mice. CNS Neurosci Ther 2021; 27:220–232CrossrefGoogle Scholar

76 Giannoni-Guzmán MA, Kamitakahara A, Magalong V, et al.: Circadian photoperiod alters TREK-1 channel function and expression in dorsal raphe serotonergic neurons via melatonin receptor 1 signaling. J Pineal Res 2021; 70:e12705CrossrefGoogle Scholar

77 Bak M, Park J, Min K, et al.: Recombinant peptide production platform coupled with site-specific albumin conjugation enables a convenient production of long-acting therapeutic peptide. Pharmaceutics 2020; 12:364CrossrefGoogle Scholar

78 Ho E, Chen D, Zhao Y, et al.: Modified exosomes for extended intravitreal drug delivery. Invest Ophthalmol Vis Sci 2021; 62:1208Google Scholar

79 Dong X, Lei Y, Yu Z, et al.: Exosome-mediated delivery of an anti-angiogenic peptide inhibits pathological retinal angiogenesis. Theranostics 2021; 11:5107–5126CrossrefGoogle Scholar

80 Xu H, Liao C, Liang S, et al.: A novel peptide-equipped exosomes platform for delivery of antisense oligonucleotides. ACS Appl Mater Interfaces 2021; 13:10760–10767CrossrefGoogle Scholar

81 Kim T, Park C, Chhatbar PY, et al.: Effect of low intensity transcranial ultrasound stimulation on neuromodulation in animals and humans: an updated systematic review. Front Neurosci 2021; 15:620863CrossrefGoogle Scholar

82 Bowary P, Greenberg BD: Noninvasive focused ultrasound for neuromodulation: a review. Psychiatr Clin North Am 2018; 41:505–514CrossrefGoogle Scholar

83 Burks SR, Kersch CN, Witko JA, et al.: Blood-brain barrier opening by intracarotid artery hyperosmolar mannitol induces sterile inflammatory and innate immune responses. Proc Natl Acad Sci USA 2021; 118:e2021915118CrossrefGoogle Scholar

84 Fu H, McCarty DM: Crossing the blood-brain-barrier with viral vectors. Curr Opin Virol 2016; 21:87–92CrossrefGoogle Scholar

85 Greene C, Campbell M: Tight junction modulation of the blood brain barrier: CNS delivery of small molecules. Tissue Barriers 2016; 4:e1138017CrossrefGoogle Scholar

86 Rodriguez A, Tatter SB, Debinski W: Neurosurgical techniques for disruption of the blood-brain barrier for glioblastoma treatment. Pharmaceutics 2015; 7:175–187CrossrefGoogle Scholar

87 Doolittle ND, Muldoon LL, Culp AY, et al.: Delivery of chemotherapeutics across the blood-brain barrier: challenges and advances. Adv Pharmacol 2014; 71:203–243CrossrefGoogle Scholar

88 Parrish KE, Sarkaria JN, Elmquist WF: Improving drug delivery to primary and metastatic brain tumors: strategies to overcome the blood-brain barrier. Clin Pharmacol Ther 2015; 97:336–346CrossrefGoogle Scholar

89 Guzman R, Janowski M, Walczak P: Intra-arterial delivery of cell therapies for stroke. Stroke 2018; 49:1075–1082CrossrefGoogle Scholar

90 Giuliani A, Balducci AG, Zironi E, et al.: In vivo nose-to-brain delivery of the hydrophilic antiviral ribavirin by microparticle agglomerates. Drug Deliv 2018; 25:376–387CrossrefGoogle Scholar

91 Tiozzo Fasiolo L, Manniello MD, Tratta E, et al.: Opportunity and challenges of nasal powders: drug formulation and delivery. Eur J Pharm Sci 2018; 113:2–17CrossrefGoogle Scholar

92 Bourganis V, Kammona O, Alexopoulos A, et al.: Recent advances in carrier mediated nose-to-brain delivery of pharmaceutics. Eur J Pharm Biopharm 2018; 128:337–362CrossrefGoogle Scholar

93 Sonawane RO, Bachhav Y, Tekade AR, et al.: Nanoparticles for direct nose-to-brain drug delivery: implications of targeting approaches; in Direct Nose-to-Brain Drug Delivery. Edited by Pardeshi, CV, Souto EB, Cambridge, MA, Academic Press, 2021CrossrefGoogle Scholar

94 Lansley A, Martin G: Nasal drug delivery; in Aulton’s Pharmaceutics: The Design and Manufacture of Medicines, International ed. Edited by Aulton M, Taylor K. Edinburgh, UK, Elsevier Ltd., 2018Google Scholar

95 Pandey A, Nikam A, Basavraj S, et al.: Nose-to-brain drug delivery: regulatory aspects, clinical trials, patents, and future perspectives; in Direct Nose-to-Brain Drug Delivery. Edited by Pardeshi, CV, Souto EB, Cambridge, MA, Academic Press, 2021CrossrefGoogle Scholar

96 Forbes B, Bommer R, Goole J, et al.: A consensus research agenda for optimising nasal drug delivery. Expert Opin Drug Deliv 2020; 17:127–132CrossrefGoogle Scholar

97 Sonmez AI, Camsari DD, Nandakumar AL, et al.: Accelerated TMS for depression: a systematic review and meta-analysis. Psychiatry Res 2019; 273:770–781CrossrefGoogle Scholar

98 Packham HR, Nicodemus NE, Jordan SE, et al.: Effects of fMRI navigated TMS on treatment outcomes in major depressive disorder. Brain Stimul 2018; 11:e18CrossrefGoogle Scholar

99 Levkovitz Y, Isserles M, Padberg F, et al.: Efficacy and safety of deep transcranial magnetic stimulation for major depression: a prospective multicenter randomized controlled trial. World Psychiatry 2015; 14:64–73CrossrefGoogle Scholar

100 Carmi L, Tendler A, Bystritsky A, et al.: Efficacy and safety of deep transcranial magnetic stimulation for obsessive-compulsive disorder: a prospective multicenter randomized double-blind placebo-controlled trial. Am J Psychiatry 2019; 176:931–938CrossrefGoogle Scholar

101 Roth Y, Tendler A, Arikan MK, et al.: Real-world efficacy of deep TMS for obsessive-compulsive disorder: post-marketing data collected from twenty-two clinical sites. J Psychiatr Res 2021; 137:667–672CrossrefGoogle Scholar

102 Orosz A, Jann K, Wirth M, et al.: Theta burst TMS increases cerebral blood flow in the primary motor cortex during motor performance as assessed by arterial spin labeling (ASL). Neuroimage 2012; 61:599–605CrossrefGoogle Scholar

103 Hamano T, Kaji R, Fukuyama H, et al.: Lack of prolonged cerebral blood flow change after transcranial magnetic stimulation. Electroencephalogr Clin Neurophysiol 1993; 89: 207–210CrossrefGoogle Scholar

104 Vazana U, Schori L, Monsonego U, et al.: TMS-induced controlled BBB opening: preclinical characterization and implications for treatment of brain cancer. Pharmaceutics 2020; 12:946CrossrefGoogle Scholar

105 Vargas E, Barrett DW, Saucedo CL, et al.: Beneficial neurocognitive effects of transcranial laser in older adults. Lasers Med Sci 2017; 32:1153–1162CrossrefGoogle Scholar

106 Tian F, Hase SN, Gonzalez-Lima F, et al.: Transcranial laser stimulation improves human cerebral oxygenation. Lasers Surg Med 2016; 48:343–349CrossrefGoogle Scholar

107 Huang L-D, Kao T-C, Sung K-B, et al.: Simulation study on the optimization of photon energy delivered to the prefrontal cortex in low-level-light therapy using red to near-infrared light. IEEE J Sel Top Quantum Electron 2021; 27:1–10Google Scholar

108 Peng H, Wang M, Hu C, et al.: A new type of MgFe2O4@CuS-APTES nanocarrier for magnetic targeting and light-microwave dual controlled drug release. Int J Nanomedicine 2020; 15:8783–8802CrossrefGoogle Scholar

109 Eden BD, Rice AJ, Lovett TD, et al.: Microwave-assisted synthesis and in vitro stability of N-benzylamide non-steroidal anti-inflammatory drug conjugates for CNS delivery. Bioorg Med Chem Lett 2019; 29:1487–1491CrossrefGoogle Scholar

110 Sack AT, Cohen Kadosh R, Schuhmann T, et al.: Optimizing functional accuracy of TMS in cognitive studies: a comparison of methods. J Cogn Neurosci 2009; 21:207–221CrossrefGoogle Scholar

111 Julkunen P, Säisänen L, Danner N, et al.: Comparison of navigated and non-navigated transcranial magnetic stimulation for motor cortex mapping, motor threshold and motor evoked potentials. Neuroimage 2009; 44:790–795CrossrefGoogle Scholar

112 Kaushik A, Rodriguez J, Rothen D: MRI-guided, noninvasive delivery of magneto-electric drug nanocarriers to the brain in a nonhuman primate. ACS Appl Bio Mater 2019; 2:4826–4836CrossrefGoogle Scholar

113 Sollmann N, Krieg SM, Säisänen L, et al.: Mapping of motor function with neuronavigated transcranial magnetic stimulation: a review on clinical application in brain tumors and methods for ensuring feasible accuracy. Brain Sci 2021; 11:897CrossrefGoogle Scholar