1
|
Liu N, Haziyihan A, Zhao W, Chen Y, Chao H. Trajectory of brain-derived amyloid beta in Alzheimer's disease: where is it coming from and where is it going? Transl Neurodegener 2024; 13:42. [PMID: 39160618 PMCID: PMC11331646 DOI: 10.1186/s40035-024-00434-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder that primarily impacts cognitive function. Currently there are no disease-modifying treatments to stop or slow its progression. Recent studies have found that several peripheral and systemic abnormalities are associated with AD, and our understanding of how these alterations contribute to AD is becoming more apparent. In this review, we focuse on amyloid‑beta (Aβ), a major hallmark of AD, summarizing recent findings on the source of brain-derived Aβ and discussing where and how the brain-derived Aβ is cleared in vivo. Based on these findings, we propose future strategies for AD prevention and treatment, from a novel perspective on Aβ metabolism.
Collapse
Affiliation(s)
- Ni Liu
- Zhengzhou University, Zhengzhou, 450001, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | | | - Wei Zhao
- Zhengzhou University, Zhengzhou, 450001, China
| | - Yu Chen
- Zhengzhou University, Zhengzhou, 450001, China
| | - Hongbo Chao
- Zhengzhou University, Zhengzhou, 450001, China.
- Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
2
|
Madadi AK, Sohn MJ. Advances in Intrathecal Nanoparticle Delivery: Targeting the Blood-Cerebrospinal Fluid Barrier for Enhanced CNS Drug Delivery. Pharmaceuticals (Basel) 2024; 17:1070. [PMID: 39204177 PMCID: PMC11357388 DOI: 10.3390/ph17081070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
The blood-cerebrospinal fluid barrier (BCSFB) tightly regulates molecular exchanges between the bloodstream and cerebrospinal fluid (CSF), creating challenges for effective central nervous system (CNS) drug delivery. This review assesses intrathecal (IT) nanoparticle (NP) delivery systems that aim to enhance drug delivery by circumventing the BCSFB, complementing approaches that target the blood-brain barrier (BBB). Active pharmaceutical ingredients (APIs) face hurdles like restricted CNS distribution and rapid clearance, which diminish the efficacy of IT therapies. NPs can be engineered to extend drug circulation times, improve CNS penetration, and facilitate sustained release. This review discusses key pharmacokinetic (PK) parameters essential for the effectiveness of these systems. NPs can quickly traverse the subarachnoid space and remain within the leptomeninges for extended periods, often exceeding three weeks. Some designs enable deeper brain parenchyma penetration. Approximately 80% of NPs in the CSF are cleared through the perivascular glymphatic pathway, with microglia-mediated transport significantly contributing to their paravascular clearance. This review synthesizes recent progress in IT-NP delivery across the BCSFB, highlighting critical findings, ongoing challenges, and the therapeutic potential of surface modifications and targeted delivery strategies.
Collapse
Affiliation(s)
- Ahmad Khalid Madadi
- Department of Biomedical Science, Graduate School of Medicine, Inje University, 75, Bokji-ro, Busanjingu, Busan 47392, Republic of Korea;
| | - Moon-Jun Sohn
- Department of Biomedical Science, Graduate School of Medicine, Inje University, 75, Bokji-ro, Busanjingu, Busan 47392, Republic of Korea;
- Department of Neurosurgery, Neuroscience & Radiosurgery Hybrid Research Center, Inje University Ilsan Paik Hospital, College of Medicine, Juhwa-ro 170, Ilsanseo-gu, Goyang City 10380, Republic of Korea
| |
Collapse
|
3
|
Kim S, Ullah I, Beloor J, Chung K, Kim J, Yi Y, Kang E, Yun G, Heo S, Pyun SH, Kim SH, Kumar P, Lee SK. Systemic Treatment with siRNA Targeting Gamma-Secretase Activating Protein Inhibits Amyloid-β Accumulation in Alzheimer's Disease. Biomater Res 2024; 28:0027. [PMID: 38868092 PMCID: PMC11168191 DOI: 10.34133/bmr.0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/31/2024] [Indexed: 06/14/2024] Open
Abstract
Amyloid-β (Aβ) peptide aggregation in the brain is a key factor in Alzheimer's disease. However, direct inhibition of β-secretase or γ-secretase proves ineffective in reducing Aβ accumulation and improving cognition in Alzheimer's. Recent findings suggest that inhibiting gamma-secretase activating protein (GSAP) can decrease Aβ generation without affecting crucial γ-secretase substrates. Dimerization of Lep9R3LC (diLep9R3LC) was confirmed by Ellman's test. The peptide-small interfering RNA (siRNA) complex ratio, particle size, and surface charge were analyzed using electrophoretic mobility shift assay, and dynamic light scattering, respectively. In a 3xTg mice model of Alzheimer's disease, diLep9R3LC:siRNA complexes were intravenously administered twice a week for 8 weeks. Assessments included gene silencing, protein expression, and behavioral improvement using reverse transcription polymerase chain reaction, quantitative polymerase chain reaction, western blotting, Y-maze, and object recognition tests. The efficacy of Lep9R3LC dimerization was ~80% after a 3-d reaction by Ellman's test. In N2a cells, diLep9R3LC:siGSAP complexes achieved ~70% silencing at 48 h posttransfection. In 7-month-old male 3xTg mice, GSAP knockdown was ~30% in the cortex and ~50% in the hippocampus. The behavior improved in mice treated with diLep9R3LC:siGSAP complexes, showing a 60% increase in entries and an 80% increase object recognition. A novel dipeptide, diLep9R3LC, complexed with siRNA targeting GSAP (siGSAP), efficiently delivers siRNA to the mouse brain, targeting the hippocampus. The treatment inhibits Aβ accumulation, reduces GSK-3β-associated with tau hyperphosphorylation, and improves Alzheimer's behavior. Our findings highlight diLep9R3LC:siGSAP's potential for Alzheimer's and as a siRNA carrier for central nervous system-related diseases.
Collapse
Affiliation(s)
- Sunghwa Kim
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Department of Internal Medicine,
Yale University, New Haven, CT, USA
| | - Jagadish Beloor
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Department of Internal Medicine,
Yale University, New Haven, CT, USA
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Lerner Research Institute,
Cleveland Clinic, Cleveland, OH, USA
| | - Jongkil Kim
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
- Harvard Medical School, Boston, MA, USA
| | - Yujong Yi
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Eunhwa Kang
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Gyeongju Yun
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Seoyoun Heo
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Seon-Hong Pyun
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine,
Hanyang University, Seoul, Korea
| | - Priti Kumar
- Department of Internal Medicine,
Yale University, New Haven, CT, USA
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology,
Hanyang University, Seoul, Korea
| |
Collapse
|
4
|
Cooper CG, Kafetzis KN, Patabendige A, Tagalakis AD. Blood-brain barrier disruption in dementia: Nano-solutions as new treatment options. Eur J Neurosci 2024; 59:1359-1385. [PMID: 38154805 DOI: 10.1111/ejn.16229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/30/2023]
Abstract
Candidate drugs targeting the central nervous system (CNS) demonstrate extremely low clinical success rates, with more than 98% of potential treatments being discontinued due to poor blood-brain barrier (BBB) permeability. Neurological conditions were shown to be the second leading cause of death globally in 2016, with the number of people currently affected by neurological disorders increasing rapidly. This increasing trend, along with an inability to develop BBB permeating drugs, is presenting a major hurdle in the treatment of CNS-related disorders, like dementia. To overcome this, it is necessary to understand the structure and function of the BBB, including the transport of molecules across its interface in both healthy and pathological conditions. The use of CNS drug carriers is rapidly gaining popularity in CNS research due to their ability to target BBB transport systems. Further research and development of drug delivery vehicles could provide essential information that can be used to develop novel treatments for neurological conditions. This review discusses the BBB and its transport systems and evaluates the potential of using nanoparticle-based delivery systems as drug carriers for CNS disease with a focus on dementia.
Collapse
Affiliation(s)
| | | | - Adjanie Patabendige
- Department of Biology, Edge Hill University, Ormskirk, UK
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| | - Aristides D Tagalakis
- Department of Biology, Edge Hill University, Ormskirk, UK
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
5
|
Chung S, Yi Y, Ullah I, Chung K, Park S, Lim J, Kim C, Pyun SH, Kim M, Kim D, Lee M, Rhim T, Lee SK. Systemic Treatment with Fas-Blocking Peptide Attenuates Apoptosis in Brain Ischemia. Int J Mol Sci 2024; 25:661. [PMID: 38203830 PMCID: PMC10780202 DOI: 10.3390/ijms25010661] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Apoptosis plays a crucial role in neuronal injury, with substantial evidence implicating Fas-mediated cell death as a key factor in ischemic strokes. To address this, inhibition of Fas-signaling has emerged as a promising strategy in preventing neuronal cell death and alleviating brain ischemia. However, the challenge of overcoming the blood-brain barrier (BBB) hampers the effective delivery of therapeutic drugs to the central nervous system (CNS). In this study, we employed a 30 amino acid-long leptin peptide to facilitate BBB penetration. By conjugating the leptin peptide with a Fas-blocking peptide (FBP) using polyethylene glycol (PEG), we achieved specific accumulation in the Fas-expressing infarction region of the brain following systemic administration. Notably, administration in leptin receptor-deficient db/db mice demonstrated that leptin facilitated the delivery of FBP peptide. We found that the systemic administration of leptin-PEG-FBP effectively inhibited Fas-mediated apoptosis in the ischemic region, resulting in a significant reduction of neuronal cell death, decreased infarct volumes, and accelerated recovery. Importantly, neither leptin nor PEG-FBP influenced apoptotic signaling in brain ischemia. Here, we demonstrate that the systemic delivery of leptin-PEG-FBP presents a promising and viable strategy for treating cerebral ischemic stroke. Our approach not only highlights the therapeutic potential but also emphasizes the importance of overcoming BBB challenges to advance treatments for neurological disorders.
Collapse
Affiliation(s)
- Sungeun Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Yujong Yi
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
- Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Seongjun Park
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Jaeyeoung Lim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Chaeyeon Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Seon-Hong Pyun
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Minkyung Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Minhyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Taiyoun Rhim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| |
Collapse
|
6
|
Arora S, Bajaj T, Kumar J, Goyal M, Singh A, Singh C. Recent Advances in Delivery of Peptide and Protein Therapeutics to the Brain. J Pharmacol Exp Ther 2024; 388:54-66. [PMID: 37977811 DOI: 10.1124/jpet.123.001690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023] Open
Abstract
The classes of neuropharmaceuticals known as proteins and peptides serve as diagnostic tools and are involved in specific communication in the peripheral and central nervous systems. However, due to tight junctions resembling epithelial cells found in the blood-brain barrier (BBB) in vivo, they are typically excluded from transport from the blood to the brain. The drugs having molecular weight of less than 400 Dalton are able to cross the BBB via lipid-mediated free diffusion. However, large molecule therapeutics are devoid of these characteristics. As an alternative, these substances may be carried via chimeric peptide drug delivery systems, and assist in transcytosis through BBB with the aid of linker strategies. With their recent developments, several forms of nanoparticles, including poly (ethylene glycol)-poly(ε-caprolactone) copolymers, nanogels, liposomes, nanostructured lipid carriers, poly (D, L-lactide-co-glycolide) nanoparticles, chitosan, and solid lipid nanoparticles, have also been considered for their therapeutic applications. Moreover, the necessity for physiologic optimization of current drug delivery methods and their carriers to deliver therapeutic doses of medication into the brain for the treatment of various neurologic illnesses has also been emphasized. Therapeutic use of proteins and peptides has no neuroprotective impact in the absence of all these methods. Each tactic, however, has unique drawbacks and considerations. In this review, we discuss different drug delivery methods for therapeutic distribution of pharmaceuticals, primarily neuroproteins and neuropeptides, through endothelial capillaries via blood-brain barrier. Finally, we have also discussed the challenges and future perspective of protein and peptide therapeutics delivery to the brain. SIGNIFICANCE STATEMENT: Very few reports on the delivery of therapeutic protein and peptide nanoformulations are available in the literature. Herein, we attempted to discuss these nanoformulations of protein and peptide therapeutics used to treat brain diseases.
Collapse
Affiliation(s)
- Sanchit Arora
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Tania Bajaj
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Jayant Kumar
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Manoj Goyal
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Arti Singh
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Charan Singh
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| |
Collapse
|
7
|
He Y, Guan J, Lai L, Zhang X, Chen B, Wang X, Wu R. Imaging of brain clearance pathways via MRI assessment of the glymphatic system. Aging (Albany NY) 2023; 15:14945-14956. [PMID: 38149988 PMCID: PMC10781494 DOI: 10.18632/aging.205322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/03/2023] [Indexed: 12/28/2023]
Abstract
Glymphatic clearance dysfunction may play an important role in a variety of neurodegenerative diseases and the progression of ageing. However, in vivo imaging of the glymphatic system is challenging. In this study, we describe an MRI method based on chemical exchange saturation transfer (CEST) of the Angiopep-2 probe to visualize the clearance function of the glymphatic system. We injected rats with Angiopep-2 via the tail vein and performed in vivo MRI at 7 T to track differences in Angiopep-2 signal changes; we then applied the same principles in a bilateral deep cervical lymph node ligation rat model and in ageing rats. We demonstrated the feasibility of Angiopep-2 CEST for visualizing the clearance function of the glymphatic system. Finally, a pathological assessment was performed. Within the model group, the deep cervical lymph node ligation group and the ageing group showed higher CEST signal than the control group. We conclude that this new MRI method can visualize clearance in the glymphatic system.
Collapse
Affiliation(s)
- Yi He
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- Department of Ultrasound, Shantou Central Hospital, Shantou, Guangdong, China
| | - Jitian Guan
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Lingfeng Lai
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xiaolei Zhang
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Beibei Chen
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xueqing Wang
- Department of Ultrasound, Shantou Central Hospital, Shantou, Guangdong, China
| | - Renhua Wu
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| |
Collapse
|
8
|
Wu W, Huang J, Han P, Zhang J, Wang Y, Jin F, Zhou Y. Research Progress on Natural Plant Molecules in Regulating the Blood-Brain Barrier in Alzheimer's Disease. Molecules 2023; 28:7631. [PMID: 38005352 PMCID: PMC10674591 DOI: 10.3390/molecules28227631] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder. With the aging population and the continuous development of risk factors associated with AD, it will impose a significant burden on individuals, families, and society. Currently, commonly used therapeutic drugs such as Cholinesterase inhibitors, N-methyl-D-aspartate antagonists, and multiple AD pathology removal drugs have been shown to have beneficial effects on certain pathological conditions of AD. However, their clinical efficacy is minimal and they are associated with certain adverse reactions. Furthermore, the underlying pathological mechanism of AD remains unclear, posing a challenge for drug development. In contrast, natural plant molecules, widely available, offer multiple targeting pathways and demonstrate inherent advantages in modifying the typical pathologic features of AD by influencing the blood-brain barrier (BBB). We provide a comprehensive review of recent in vivo and in vitro studies on natural plant molecules that impact the BBB in the treatment of AD. Additionally, we analyze their specific mechanisms to offer novel insights for the development of safe and effective targeted drugs as well as guidance for experimental research and the clinical application of drugs for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Weidong Wu
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Jiahao Huang
- Department of Chinese Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Pengfei Han
- Science and Education Section, Zhangjiakou First Hospital, Zhangjiakou 075041, China;
| | - Jian Zhang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Yuxin Wang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Fangfang Jin
- Department of Internal Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yanyan Zhou
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| |
Collapse
|
9
|
Leal AF, Inci OK, Seyrantepe V, Rintz E, Celik B, Ago Y, León D, Suarez DA, Alméciga-Díaz CJ, Tomatsu S. Molecular Trojan Horses for treating lysosomal storage diseases. Mol Genet Metab 2023; 140:107648. [PMID: 37598508 DOI: 10.1016/j.ymgme.2023.107648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 08/22/2023]
Abstract
Lysosomal storage diseases (LSDs) are caused by monogenic mutations in genes encoding for proteins related to the lysosomal function. Lysosome plays critical roles in molecule degradation and cell signaling through interplay with many other cell organelles, such as mitochondria, endoplasmic reticulum, and peroxisomes. Even though several strategies (i.e., protein replacement and gene therapy) have been attempted for LSDs with promising results, there are still some challenges when hard-to-treat tissues such as bone (i.e., cartilages, ligaments, meniscus, etc.), the central nervous system (mostly neurons), and the eye (i.e., cornea, retina) are affected. Consistently, searching for novel strategies to reach those tissues remains a priority. Molecular Trojan Horses have been well-recognized as a potential alternative in several pathological scenarios for drug delivery, including LSDs. Even though molecular Trojan Horses refer to genetically engineered proteins to overcome the blood-brain barrier, such strategy can be extended to strategies able to transport and deliver drugs to specific tissues or cells using cell-penetrating peptides, monoclonal antibodies, vesicles, extracellular vesicles, and patient-derived cells. Only some of those platforms have been attempted in LSDs. In this paper, we review the most recent efforts to develop molecular Trojan Horses and discuss how this strategy could be implemented to enhance the current efficacy of strategies such as protein replacement and gene therapy in the context of LSDs.
Collapse
Affiliation(s)
- Andrés Felipe Leal
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, Colombia; Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Orhan Kerim Inci
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Betul Celik
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Yasuhiko Ago
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Daniel León
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Diego A Suarez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland; Faculty of Arts and Sciences, University of Delaware, Newark, DE, USA; Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Xue S, Zhou X, Yang ZH, Si XK, Sun X. Stroke-induced damage on the blood-brain barrier. Front Neurol 2023; 14:1248970. [PMID: 37840921 PMCID: PMC10569696 DOI: 10.3389/fneur.2023.1248970] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/08/2023] [Indexed: 10/17/2023] Open
Abstract
The blood-brain barrier (BBB) is a functional phenotype exhibited by the neurovascular unit (NVU). It is maintained and regulated by the interaction between cellular and non-cellular matrix components of the NVU. The BBB plays a vital role in maintaining the dynamic stability of the intracerebral microenvironment as a barrier layer at the critical interface between the blood and neural tissues. The large contact area (approximately 20 m2/1.3 kg brain) and short diffusion distance between neurons and capillaries allow endothelial cells to dominate the regulatory role. The NVU is a structural component of the BBB. Individual cells and components of the NVU work together to maintain BBB stability. One of the hallmarks of acute ischemic stroke is the disruption of the BBB, including impaired function of the tight junction and other molecules, as well as increased BBB permeability, leading to brain edema and a range of clinical symptoms. This review summarizes the cellular composition of the BBB and describes the protein composition of the barrier functional junction complex and the mechanisms regulating acute ischemic stroke-induced BBB disruption.
Collapse
Affiliation(s)
| | | | | | | | - Xin Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Chaparro CIP, Simões BT, Borges JP, Castanho MARB, Soares PIP, Neves V. A Promising Approach: Magnetic Nanosystems for Alzheimer's Disease Theranostics. Pharmaceutics 2023; 15:2316. [PMID: 37765284 PMCID: PMC10536416 DOI: 10.3390/pharmaceutics15092316] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Among central nervous system (CNS) disorders, Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and a major cause of dementia worldwide. The yet unclear etiology of AD and the high impenetrability of the blood-brain barrier (BBB) limit most therapeutic compounds from reaching the brain. Although many efforts have been made to effectively deliver drugs to the CNS, both invasive and noninvasive strategies employed often come with associated side effects. Nanotechnology-based approaches such as nanoparticles (NPs), which can act as multifunctional platforms in a single system, emerged as a potential solution for current AD theranostics. Among these, magnetic nanoparticles (MNPs) are an appealing strategy since they can act as contrast agents for magnetic resonance imaging (MRI) and as drug delivery systems. The nanocarrier functionalization with specific moieties, such as peptides, proteins, and antibodies, influences the particles' interaction with brain endothelial cell constituents, facilitating transport across the BBB and possibly increasing brain penetration. In this review, we introduce MNP-based systems, combining surface modifications with the particles' physical properties for molecular imaging, as a novel neuro-targeted strategy for AD theranostics. The main goal is to highlight the potential of multifunctional MNPs and their advances as a dual nanotechnological diagnosis and treatment platform for neurodegenerative disorders.
Collapse
Affiliation(s)
- Catarina I. P. Chaparro
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University of Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Beatriz T. Simões
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
| | - João P. Borges
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University of Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Miguel A. R. B. Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
| | - Paula I. P. Soares
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University of Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
| |
Collapse
|
12
|
Shetti AU, Ramakrishnan A, Romanova L, Li W, Vo K, Volety I, Ratnayake I, Stephen T, Minshall RD, Cologna SM, Lazarov O. Reduced endothelial caveolin-1 underlies deficits in brain insulin signalling in type 2 diabetes. Brain 2023; 146:3014-3028. [PMID: 36731883 PMCID: PMC10316766 DOI: 10.1093/brain/awad028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/07/2023] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Patients with type 2 diabetes exhibit severe impairments in insulin signalling in the brain and are five times more likely to develop Alzheimer's disease. However, what leads to these impairments is not fully understood. Here, we show reduced expression of endothelial cell caveolin-1 (Cav-1) in the db/db (Leprdb) mouse model of type 2 diabetes. This reduction correlated with alterations in insulin receptor expression and signalling in brain microvessels as well as brain parenchyma. These findings were recapitulated in the brains of endothelial cell-specific Cav-1 knock-out (Tie2Cre; Cav-1fl/fl) mice. Lack of Cav-1 in endothelial cells led to reduced response to insulin as well as reduced insulin uptake. Furthermore, we observed that Cav-1 was necessary for the stabilization of insulin receptors in lipid rafts. Interactome analysis revealed that insulin receptor interacts with Cav-1 and caveolae-associated proteins, insulin-degrading enzyme and the tight junction protein Zonula Occludence-1 in brain endothelial cells. Restoration of Cav-1 in Cav-1 knock-out brain endothelial cells rescued insulin receptor expression and localization. Overall, these results suggest that Cav-1 regulates insulin signalling and uptake by brain endothelial cells by modulating IR-α and IR-β localization and function in lipid rafts. Furthermore, depletion of endothelial cell-specific Cav-1 and the resulting impairment in insulin transport leads to alteration in insulin signalling in the brain parenchyma of type 2 diabetics.
Collapse
Affiliation(s)
- Aashutosh U Shetti
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Abhirami Ramakrishnan
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Liudmila Romanova
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Wenping Li
- Department of Chemistry, College of Liberal Arts and Sciences, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Khanh Vo
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ipsita Volety
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ishara Ratnayake
- Electron Microscopy Core, Research Resource Center, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Terilyn Stephen
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Richard D Minshall
- Department of Pharmacology and Regenerative Medicine, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Anesthesiology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Stephanie M Cologna
- Department of Chemistry, College of Liberal Arts and Sciences, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Placci M, Giannotti MI, Muro S. Polymer-based drug delivery systems under investigation for enzyme replacement and other therapies of lysosomal storage disorders. Adv Drug Deliv Rev 2023; 197:114683. [PMID: 36657645 PMCID: PMC10629597 DOI: 10.1016/j.addr.2022.114683] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 12/25/2022] [Indexed: 01/18/2023]
Abstract
Lysosomes play a central role in cellular homeostasis and alterations in this compartment associate with many diseases. The most studied example is that of lysosomal storage disorders (LSDs), a group of 60 + maladies due to genetic mutations affecting lysosomal components, mostly enzymes. This leads to aberrant intracellular storage of macromolecules, altering normal cell function and causing multiorgan syndromes, often fatal within the first years of life. Several treatment modalities are available for a dozen LSDs, mostly consisting of enzyme replacement therapy (ERT) strategies. Yet, poor biodistribution to main targets such as the central nervous system, musculoskeletal tissue, and others, as well as generation of blocking antibodies and adverse effects hinder effective LSD treatment. Drug delivery systems are being studied to surmount these obstacles, including polymeric constructs and nanoparticles that constitute the focus of this article. We provide an overview of the formulations being tested, the diseases they aim to treat, and the results observed from respective in vitro and in vivo studies. We also discuss the advantages and disadvantages of these strategies, the remaining gaps of knowledge regarding their performance, and important items to consider for their clinical translation. Overall, polymeric nanoconstructs hold considerable promise to advance treatment for LSDs.
Collapse
Affiliation(s)
- Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Marina I Giannotti
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; CIBER-BBN, ISCIII, Barcelona, Spain; Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona 08028, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; Institute of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08010, Spain; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
14
|
Nguyen NHT, Nguyen NT, Kim YH, Min J. Yeast-derived vacuoles as a novel carrier with enhanced hCMEC/D3 cell monolayer penetration. Biotechnol J 2023; 18:e2200393. [PMID: 36321515 DOI: 10.1002/biot.202200393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier (BBB) is a brain protection structure that restricts drug delivery from the blood to the central nervous system. Thus, we developed a novel drug carrier using yeast vacuoles to overcome this problem. The purpose of this study was to assess the drug transportability of yeast vacuoles using a human cerebral microvascular endothelial cell line (hCMEC/D3) cell monolayer. Here, we used daunorubicin (DNR) as a microtubule-targeting agent with the ability to disaggregate pre-formed fibrils and prevent Tau fibrillization. An in vitro model was developed by culturing hCMEC/D3 cells on Transwell inserts in EBM-2 endothelial basal medium until the cells formed a monolayer. Next, nano-sized yeast vacuoles were loaded with DNR, and the signals inside and outside the hMEC/D3 cell monolayer were detected using the GloMax® Explorer fluorometer. DNR penetrated the cell monolayer and was regulated by endocytosis via receptor-mediated macropinocytosis on the surface of the cell. Confocal imaging showed a significant increase in intracellular DNR fluorescence when the cells were treated with the vacuole-encapsulated drug. These results indicate that the drug penetrated the hCMEC/D3 cell monolayer via encapsulation into the vacuoles. Overall, yeast-derived vacuoles are promising candidates as drug carriers to the brain.
Collapse
Affiliation(s)
- Ngoc-Han Thi Nguyen
- Department of Bioprocess Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Ngoc-Tu Nguyen
- School of Chemical Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Yang Hoon Kim
- School of Biological Sciences, Chungbuk National University, Cheongju, South Korea
| | - Jiho Min
- Department of Bioprocess Engineering, Jeonbuk National University, Jeonju, South Korea.,School of Chemical Engineering, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
15
|
Padilla-Godínez FJ, Ruiz-Ortega LI, Guerra-Crespo M. Nanomedicine in the Face of Parkinson's Disease: From Drug Delivery Systems to Nanozymes. Cells 2022; 11:3445. [PMID: 36359841 PMCID: PMC9657131 DOI: 10.3390/cells11213445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 01/02/2024] Open
Abstract
The complexity and overall burden of Parkinson's disease (PD) require new pharmacological approaches to counteract the symptomatology while reducing the progressive neurodegeneration of affected dopaminergic neurons. Since the pathophysiological signature of PD is characterized by the loss of physiological levels of dopamine (DA) and the misfolding and aggregation of the alpha-synuclein (α-syn) protein, new proposals seek to restore the lost DA and inhibit the progressive damage derived from pathological α-syn and its impact in terms of oxidative stress. In this line, nanomedicine (the medical application of nanotechnology) has achieved significant advances in the development of nanocarriers capable of transporting and delivering basal state DA in a controlled manner in the tissues of interest, as well as highly selective catalytic nanostructures with enzyme-like properties for the elimination of reactive oxygen species (responsible for oxidative stress) and the proteolysis of misfolded proteins. Although some of these proposals remain in their early stages, the deepening of our knowledge concerning the pathological processes of PD and the advances in nanomedicine could endow for the development of potential treatments for this still incurable condition. Therefore, in this paper, we offer: (i) a brief summary of the most recent findings concerning the physiology of motor regulation and (ii) the molecular neuropathological processes associated with PD, together with (iii) a recapitulation of the current progress in controlled DA release by nanocarriers and (iv) the design of nanozymes, catalytic nanostructures with oxidoreductase-, chaperon, and protease-like properties. Finally, we conclude by describing the prospects and knowledge gaps to overcome and consider as research into nanotherapies for PD continues, especially when clinical translations take place.
Collapse
Affiliation(s)
- Francisco J. Padilla-Godínez
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
- Regenerative Medicine Laboratory, Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
| | - Leonardo I. Ruiz-Ortega
- Institute for Physical Sciences, National Autonomous University of Mexico, Cuernavaca 62210, Mexico
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Magdalena Guerra-Crespo
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
- Regenerative Medicine Laboratory, Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
| |
Collapse
|
16
|
Zhao P, Zhang N, An Z. Engineering antibody and protein therapeutics to cross the blood-brain barrier. Antib Ther 2022; 5:311-331. [PMID: 36540309 PMCID: PMC9759110 DOI: 10.1093/abt/tbac028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/10/2022] [Accepted: 11/01/2022] [Indexed: 08/17/2023] Open
Abstract
Diseases in the central nervous system (CNS) are often difficult to treat. Antibody- and protein-based therapeutics hold huge promises in CNS disease treatment. However, proteins are restricted from entering the CNS by the blood-brain barrier (BBB). To achieve enhanced BBB crossing, antibody-based carriers have been developed by utilizing the endogenous macromolecule transportation pathway, known as receptor-mediated transcytosis. In this report, we first provided an overall review on key CNS diseases and the most promising antibody- or protein-based therapeutics approved or in clinical trials. We then reviewed the platforms that are being explored to increase the macromolecule brain entry to combat CNS diseases. Finally, we have analyzed the lessons learned from past experiences and have provided a perspective on the future engineering of novel delivery vehicles for antibody- and protein-based therapies for CNS diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| |
Collapse
|
17
|
Mitusova K, Peltek OO, Karpov TE, Muslimov AR, Zyuzin MV, Timin AS. Overcoming the blood-brain barrier for the therapy of malignant brain tumor: current status and prospects of drug delivery approaches. J Nanobiotechnology 2022; 20:412. [PMID: 36109754 PMCID: PMC9479308 DOI: 10.1186/s12951-022-01610-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/18/2022] [Indexed: 01/06/2023] Open
Abstract
Besides the broad development of nanotechnological approaches for cancer diagnosis and therapy, currently, there is no significant progress in the treatment of different types of brain tumors. Therapeutic molecules crossing the blood-brain barrier (BBB) and reaching an appropriate targeting ability remain the key challenges. Many invasive and non-invasive methods, and various types of nanocarriers and their hybrids have been widely explored for brain tumor treatment. However, unfortunately, no crucial clinical translations were observed to date. In particular, chemotherapy and surgery remain the main methods for the therapy of brain tumors. Exploring the mechanisms of the BBB penetration in detail and investigating advanced drug delivery platforms are the key factors that could bring us closer to understanding the development of effective therapy against brain tumors. In this review, we discuss the most relevant aspects of the BBB penetration mechanisms, observing both invasive and non-invasive methods of drug delivery. We also review the recent progress in the development of functional drug delivery platforms, from viruses to cell-based vehicles, for brain tumor therapy. The destructive potential of chemotherapeutic drugs delivered to the brain tumor is also considered. This review then summarizes the existing challenges and future prospects in the use of drug delivery platforms for the treatment of brain tumors.
Collapse
Affiliation(s)
- Ksenia Mitusova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Oleksii O Peltek
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Timofey E Karpov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Albert R Muslimov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
- Sirius University of Science and Technology, Olympic Ave 1, Sirius, 354340, Russian Federation
| | - Mikhail V Zyuzin
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Alexander S Timin
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation.
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation.
| |
Collapse
|
18
|
Ahmadi Kamalabadi M, Neshastehriz A, Ghaznavi H, Amini SM. Folate functionalized gold-coated magnetic nanoparticles effect in combined electroporation and radiation treatment of HPV-positive oropharyngeal cancer. Med Oncol 2022; 39:196. [PMID: 36071293 DOI: 10.1007/s12032-022-01780-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
The rate of HPV-positive oropharyngeal cancer incidence is increasing, especially in the young population. While these patients show good responses to radiotherapy. The major complication of radiotherapy is normal tissue involvement. Thus, finding an effective treatment method is essential. Multimodal therapy with the lowest side effect could be an effective treatment method. Theranostic gold magnetic core-shell nanostructure was developed as a platform for multimodal therapy of HPV-positive oropharyngeal cancer. The folate functionalized gold-magnetic core-shell nanostructure has been synthesized in a stepwise approach and characterized with various techniques including TEM, UV-Vis, and FTIR spectroscopy. KB was selected as a host for HPV and folate receptor-positive cancer. HGF as normal cell lines was selected. Both cell lines have been treated with nanoparticles, electric field and radiotherapy, either separately or in combination. Cell viability and apoptosis rate were determined by MTT and flow cytometry assay. In addition, cellular uptake of the nanoparticles was measured by ICP-OES analysis. The average size of folate functionalized gold-magnetic core-shell nanostructure was 13.8 ± 6.4 nm. A characteristic plasmonic peak of gold nanoshells was observed in the UV-Vis spectrum. The functionalization of synthesized nanostructure was confirmed with FTIR spectroscopy. None of the treatments alone can cause a significant death in cancerous cells. Combination treatments can increase cancer cell mortality and increase the proportion of apoptotic cells in them. Furthermore, it has been observed that the electric field enhanced the cellular uptake of nanoparticles just in cancerous cells. Based on our findings, we conclude that the combination of folate functionalized nanoparticles and electroporation opens a new way to improve radiation therapy efficacy of HPV-positive cancers.
Collapse
Affiliation(s)
- Mahdieh Ahmadi Kamalabadi
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Radiation Sciences, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Neshastehriz
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Radiation Sciences, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Seyed Mohammad Amini
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Yawalkar AN, Pawar MA, Vavia PR. Microspheres for targeted drug delivery- A review on recent applications. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Arjun P, Freeman JL, Kannan RR. Neurospecific fabrication and toxicity assessment of a PNIPAM nanogel encapsulated with trans-tephrostachin for blood-brain-barrier permeability in zebrafish model. Heliyon 2022; 8:e10237. [PMID: 36042734 PMCID: PMC9420489 DOI: 10.1016/j.heliyon.2022.e10237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/18/2022] [Accepted: 08/05/2022] [Indexed: 12/02/2022] Open
Abstract
Biocompatible Poly(N-isopropylacrylamide) (PNIPAM) nanogels (NGs) were developed at 40–65 nm to deliver Trans-Tephrostachin (TT) in zebrafish brain. Neurospecific PNIPAM NGs are functionalized with polysorbate 80 (PS80) to overcome the Blood Brain Barrier (BBB). The TT loaded with NG (NG + TT) was confirmed in UV-spectroscopy and transmission electron microscopy (TEM) with 90% efficiency of controlled release at 37 °C. The neurospecificity of NG was confirmed in 144 hours post fertilization (hpf) larvae with PS80 surface-treated rhodamine-B (Rh–B) conjugated NG and visualized in the zebrafish CNS. Oral gavaging of TT loaded NG with PS80 surface treatment (NG + TT + PS80) was confirmed to cross the BBB in adult zebrafish at 37 °C. TT release was detected by RP-HPLC. LC50 was determined as 250 μg/ml for NG, 172 μg/ml for NG + TT, and 0.9 μg/ml for TT at 96 hpf and confirmed the lesser toxicity in TT bound NG. Delays in growth and malformations were observed at concentrations above the 96 hpf-LC50. The behavior outcomes were varied with phase - and concentration-dependent hypo- or hyperactivity. The altered expression of genes associated with Alzheimer’s disease (AD) was found at 96 hpf of its LC50 concentration. The expression of appa was significantly increased for TT and supporting the TT to bind NG without altering the AD genes. Thus the study suggests the biocompatible potential of PNIPAM and its neurospecific delivery to the brain.
Collapse
Affiliation(s)
- Pitchai Arjun
- Neuroscience Lab, Centre for Molecular and Nanomedical Sciences (CMNS), Centre for Nanoscience and Nanotechnology (CNSNT), School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, (Deemed to Be University) Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai, 600119, Tamil Nadu, India.,School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Rajaretinam Rajesh Kannan
- Neuroscience Lab, Centre for Molecular and Nanomedical Sciences (CMNS), Centre for Nanoscience and Nanotechnology (CNSNT), School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, (Deemed to Be University) Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai, 600119, Tamil Nadu, India
| |
Collapse
|
21
|
Neganova ME, Aleksandrova YR, Sukocheva OA, Klochkov SG. Benefits and limitations of nanomedicine treatment of brain cancers and age-dependent neurodegenerative disorders. Semin Cancer Biol 2022; 86:805-833. [PMID: 35779712 DOI: 10.1016/j.semcancer.2022.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 02/07/2023]
Abstract
The treatment of central nervous system (CNS) malignancies, including brain cancers, is limited by a number of obstructions, including the blood-brain barrier (BBB), the heterogeneity and high invasiveness of tumors, the inaccessibility of tissues for early diagnosis and effective surgery, and anti-cancer drug resistance. Therapies employing nanomedicine have been shown to facilitate drug penetration across the BBB and maintain biodistribution and accumulation of therapeutic agents at the desired target site. The application of lipid-, polymer-, or metal-based nanocarriers represents an advanced drug delivery system for a growing group of anti-cancer chemicals. The nanocarrier surface is designed to contain an active ligand (cancer cell marker or antibody)-binding structure which can be modified to target specific cancer cells. Glioblastoma, ependymoma, neuroblastoma, medulloblastoma, and primary CNS lymphomas were recently targeted by easily absorbed nanocarriers. The metal- (such as transferrin drug-loaded systems), polymer- (nanocapsules and nanospheres), or lipid- (such as sulfatide-containing nanoliposomes)-based nano-vehicles were loaded with apoptosis- and/or ferroptosis-stimulating agents and demonstrated promising anti-cancer effects. This review aims to discuss effective nanomedicine approaches designed to overcome the current limitations in the therapy of brain cancers and age-dependent neurodegenerative disorders. To accent current obstacles for successful CNS-based cancer therapy, we discuss nanomedicine perspectives and limitations of nanodrug use associated with the specificity of nervous tissue characteristics and the effects nanocarriers have on cognition.
Collapse
Affiliation(s)
- Margarita E Neganova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Yulia R Aleksandrova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Olga A Sukocheva
- School of Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| | - Sergey G Klochkov
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| |
Collapse
|
22
|
Jadhav S, Yenorkar N, Bondre R, Karemore M, Bali N. Nanomedicines encountering HIV dementia: A guiding star for neurotherapeutics. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
23
|
Ball K, Bruin G, Escandon E, Funk C, Pereira JN, Yang TY, Yu H. Characterizing the pharmacokinetics and biodistribution of therapeutic proteins: an industry white paper. Drug Metab Dispos 2022; 50:858-866. [PMID: 35149542 DOI: 10.1124/dmd.121.000463] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022] Open
Abstract
Characterization of the pharmacokinetics (PK) and biodistribution of therapeutic proteins (TPs) is a hot topic within the pharmaceutical industry, particularly with an ever-increasing catalog of novel modality TPs. Here, we review the current practices, and provide a summary of extensive cross-company discussions as well as a survey completed by International Consortium for Innovation and Quality (IQ consortium) members on this theme. A wide variety of in vitro, in vivo and in silico techniques are currently used to assess PK and biodistribution of TPs, and we discuss the relevance of these from an industry perspective, focusing on PK/PD understanding at the preclinical stage of development, and translation to human. We consider that the 'traditional in vivo biodistribution study' is becoming insufficient as a standalone tool, and thorough characterization of the interaction of the TP with its target(s), target biology, and off-target interactions at a microscopic scale are key to understand the overall biodistribution at a full-body scale. Our summary of the current challenges and our recommendations to address these issues could provide insight into the implementation of best practices in this area of drug development, and continued cross-company collaboration will be of tremendous value. Significance Statement The Innovation & Quality Consortium (IQ) Translational and ADME Sciences Leadership Group (TALG) working group for the ADME of therapeutic proteins evaluates the current practices, recent advances, and challenges in characterizing the PK and biodistribution of therapeutic proteins during drug development, and proposes recommendations to address these issues. Incorporating the in vitro, in vivo and in silico approaches discussed herein may provide a pragmatic framework to increase early understanding of PK/PD relationships, and aid translational modelling for first-in-human dose predictions.
Collapse
Affiliation(s)
| | - Gerard Bruin
- Novartis Institutes for Biomedical Research, Switzerland
| | | | - Christoph Funk
- Dept. of Drug Metabolism and Pharmacokinetics, F. Hoffmann-La Roche Ltd., Switzerland
| | | | | | - Hongbin Yu
- Boehringer Ingelheim Pharmaceuticals, Inc, United States
| |
Collapse
|
24
|
Sadekar SS, Bowen M, Cai H, Jamalian S, Rafidi H, Shatz‐Binder W, Lafrance‐Vanasse J, Chan P, Meilandt WJ, Oldendorp A, Sreedhara A, Daugherty A, Crowell S, Wildsmith KR, Atwal J, Fuji RN, Horvath J. Translational approaches for brain delivery of biologics via cerebrospinal fluid. Clin Pharmacol Ther 2022; 111:826-834. [PMID: 35064573 PMCID: PMC9305158 DOI: 10.1002/cpt.2531] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/04/2022] [Indexed: 11/14/2022]
Abstract
Delivery of biologics via cerebrospinal fluid (CSF) has demonstrated potential to access the tissues of the central nervous system (CNS) by circumventing the blood‐brain barrier and blood‐CSF barrier. Developing an effective CSF drug delivery strategy requires optimization of multiple parameters, including choice of CSF access point, delivery device technology, and delivery kinetics to achieve effective therapeutic concentrations in the target brain region, whereas also considering the biologic modality, mechanism of action, disease indication, and patient population. This review discusses key preclinical and clinical examples of CSF delivery for different biologic modalities (antibodies, nucleic acid‐based therapeutics, and gene therapy) to the brain via CSF or CNS access routes (intracerebroventricular, intrathecal‐cisterna magna, intrathecal‐lumbar, intraparenchymal, and intranasal), including the use of novel device technologies. This review also discusses quantitative models of CSF flow that provide insight into the effect of fluid dynamics in CSF on drug delivery and CNS distribution. Such models can facilitate delivery device design and pharmacokinetic/pharmacodynamic translation from preclinical species to humans in order to optimize CSF drug delivery to brain regions of interest.
Collapse
Affiliation(s)
- Shraddha S Sadekar
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Mayumi Bowen
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Hao Cai
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Samira Jamalian
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Hanine Rafidi
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Whitney Shatz‐Binder
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Julien Lafrance‐Vanasse
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Pamela Chan
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - William J. Meilandt
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Amy Oldendorp
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Alavattam Sreedhara
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Ann Daugherty
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Susan Crowell
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Kristin R. Wildsmith
- Clinical pharmacology and translational medicine Neurology business Eisai, Nutley NJ 07110 USA
| | - Jasvinder Atwal
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Reina N. Fuji
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Josh Horvath
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| |
Collapse
|
25
|
Qamar Z, Ashhar MU, Annu, Qizilibash FF, Sahoo PK, Ali A, Ali J, Baboota S. Lipid nanocarrier of selegiline augmented anti-Parkinson's effect via P-gp modulation using quercetin. Int J Pharm 2021; 609:121131. [PMID: 34563617 DOI: 10.1016/j.ijpharm.2021.121131] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 01/08/2023]
Abstract
In the present study, SEL was loaded in a lipid nanocarrier (LNC) formulation with a P-gp pump inhibitor i.e., Quercetin (QUR) for improving the bioavailability of the SEL in the brain via the oral route. SEL-QUR LNC was formulated using modified emulsiosonication method and optimized using central composite rotatable design (CCRD) design. The results showed that optimized SEL-QUR LNC formulation was spherical with globule size, polydispersity index, entrapment efficiency and zeta potential within the range of 92.46-95.34 nm, 0.239-0.248, 88.94-91.26%, and -6.21 to -7.75 mV respectively. A 4-fold and 6-fold increase was observed in the permeation of SEL from SEL-QUR LNC across the gut sac in comparison with SEL-QUR and SEL suspensions respectively. CLSM images showed 2-fold deeper permeation of SEL across intestinal membrane demonstrating excellent in vivo prospect of the formulation. The behavioural studies including forced swimming, muscle coordination, locomotor activity, akinesia, and catalepsy were performed in the haloperidol-induced PD rats that demonstrated increased efficacy of the formulation in contrast to the SEL-QUR and SEL suspensions. These studies concluded that developed LNC formulation loaded SEL with P-gp inhibitor had the potential in improving bioavailability of SEL in the brain via oral route.
Collapse
Affiliation(s)
- Zufika Qamar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Muhammad Usama Ashhar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Annu
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Farheen Fatima Qizilibash
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Pravat Kumar Sahoo
- Delhi Institute of Pharmaceutical Science and Research (DIPSAR), Pushp Vihar, Sector III, MB Road, New Delhi 110017, India
| | - Asgar Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
26
|
Varricchio A, Ramesh SA, Yool AJ. Novel Ion Channel Targets and Drug Delivery Tools for Controlling Glioblastoma Cell Invasiveness. Int J Mol Sci 2021; 22:ijms222111909. [PMID: 34769339 PMCID: PMC8584308 DOI: 10.3390/ijms222111909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 12/13/2022] Open
Abstract
Comprising more than half of all brain tumors, glioblastoma multiforme (GBM) is a leading cause of brain cancer-related deaths worldwide. A major clinical challenge is presented by the capacity of glioma cells to rapidly infiltrate healthy brain parenchyma, allowing the cancer to escape control by localized surgical resections and radiotherapies, and promoting recurrence in other brain regions. We propose that therapies which target cellular motility pathways could be used to slow tumor dispersal, providing a longer time window for administration of frontline treatments needed to directly eradicate the primary tumors. An array of signal transduction pathways are known to be involved in controlling cellular motility. Aquaporins (AQPs) and voltage-gated ion channels are prime candidates as pharmacological targets to restrain cell migration in glioblastoma. Published work has demonstrated AQPs 1, 4 and 9, as well as voltage-gated potassium, sodium and calcium channels, chloride channels, and acid-sensing ion channels are expressed in GBM and can influence processes of cell volume change, extracellular matrix degradation, cytoskeletal reorganization, lamellipodial and filopodial extension, and turnover of cell-cell adhesions and focal assembly sites. The current gap in knowledge is the identification of optimal combinations of targets, inhibitory agents, and drug delivery systems that will allow effective intervention with minimal side effects in the complex environment of the brain, without disrupting finely tuned activities of neuro-glial networks. Based on published literature, we propose that co-treatments using AQP inhibitors in addition to other therapies could increase effectiveness, overcoming some limitations inherent in current strategies that are focused on single mechanisms. An emerging interest in nanobodies as drug delivery systems could be instrumental for achieving the selective delivery of combinations of agents aimed at multiple key targets, which could enhance success in vivo.
Collapse
Affiliation(s)
- Alanah Varricchio
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| | - Sunita A. Ramesh
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia;
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
- Correspondence:
| |
Collapse
|
27
|
Calabrese G, De Luca G, Nocito G, Rizzo MG, Lombardo SP, Chisari G, Forte S, Sciuto EL, Conoci S. Carbon Dots: An Innovative Tool for Drug Delivery in Brain Tumors. Int J Mol Sci 2021; 22:11783. [PMID: 34769212 PMCID: PMC8583729 DOI: 10.3390/ijms222111783] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 01/16/2023] Open
Abstract
Brain tumors are particularly aggressive and represent a significant cause of morbidity and mortality in adults and children, affecting the global population and being responsible for 2.6% of all cancer deaths (as well as 30% of those in children and 20% in young adults). The blood-brain barrier (BBB) excludes almost 100% of the drugs targeting brain neoplasms, representing one of the most significant challenges to current brain cancer therapy. In the last decades, carbon dots have increasingly played the role of drug delivery systems with theranostic applications against cancer, thanks to their bright photoluminescence, solubility in bodily fluids, chemical stability, and biocompatibility. After a summary outlining brain tumors and the current drug delivery strategies devised in their therapeutic management, this review explores the most recent literature about the advances and open challenges in the employment of carbon dots as both diagnostic and therapeutic agents in the treatment of brain cancers, together with the strategies devised to allow them to cross the BBB effectively.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Giovanna De Luca
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Giuseppe Nocito
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Maria Giovanna Rizzo
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Sofia Paola Lombardo
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (S.P.L.); (G.C.)
| | - Giulia Chisari
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (S.P.L.); (G.C.)
| | - Stefano Forte
- IOM Ricerca, Via Penninazzo 11, 95029 Viagrande, Italy;
| | - Emanuele Luigi Sciuto
- A.O.-Universitaria Policlinico “G. Rodolico–San Marco”, Via Santa Sofia 78, 95123 Catania, Italy;
| | - Sabrina Conoci
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| |
Collapse
|
28
|
Rajendran R, Menon KN, Nair SC. Nanotechnology Approaches for Enhanced CNS Drug Delivery in the Management of Schizophrenia. Adv Pharm Bull 2021; 12:490-508. [PMID: 35935056 PMCID: PMC9348538 DOI: 10.34172/apb.2022.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 06/02/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022] Open
Abstract
Schizophrenia is a neuropsychiatric disorder mainly affecting the central nervous system, presented with auditory and visual hallucinations, delusion and withdrawal from society. Abnormal dopamine levels mainly characterise the disease; various theories of neurotransmitters explain the pathophysiology of the disease. The current therapeutic approach deals with the systemic administration of drugs other than the enteral route, altering the neurotransmitter levels within the brain and providing symptomatic relief. Fluid biomarkers help in the early detection of the disease, which would improve the therapeutic efficacy. However, the major challenge faced in CNS drug delivery is the blood-brain barrier. Nanotherapeutic approaches may overcome these limitations, which will improve safety, efficacy, and targeted drug delivery. This review article addresses the main challenges faced in CNS drug delivery and the significance of current therapeutic strategies and nanotherapeutic approaches for a better understanding and enhanced drug delivery to the brain, which improve the quality of life of schizophrenia patients.
Collapse
Affiliation(s)
| | - Krishnakumar Neelakandha Menon
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi-682041, Kerala, India
| | | |
Collapse
|
29
|
Lucas AT, Moody A, Schorzman AN, Zamboni WC. Importance and Considerations of Antibody Engineering in Antibody-Drug Conjugates Development from a Clinical Pharmacologist's Perspective. Antibodies (Basel) 2021; 10:30. [PMID: 34449544 PMCID: PMC8395454 DOI: 10.3390/antib10030030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/04/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Antibody-drug conjugates (ADCs) appear to be in a developmental boom, with five FDA approvals in the last two years and a projected market value of over $4 billion by 2024. Major advancements in the engineering of these novel cytotoxic drug carriers have provided a few early success stories. Although the use of these immunoconjugate agents are still in their infancy, valuable lessons in the engineering of these agents have been learned from both preclinical and clinical failures. It is essential to appreciate how the various mechanisms used to engineer changes in ADCs can alter the complex pharmacology of these agents and allow the ADCs to navigate the modern-day therapeutic challenges within oncology. This review provides a global overview of ADC characteristics which can be engineered to alter the interaction with the immune system, pharmacokinetic and pharmacodynamic profiles, and therapeutic index of ADCs. In addition, this review will highlight some of the engineering approaches being explored in the creation of the next generation of ADCs.
Collapse
Affiliation(s)
- Andrew T. Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Amber Moody
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Allison N. Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
| | - William C. Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Glolytics, LLC, Chapel Hill, NC 27517, USA
| |
Collapse
|
30
|
Kouhi A, Pachipulusu V, Kapenstein T, Hu P, Epstein AL, Khawli LA. Brain Disposition of Antibody-Based Therapeutics: Dogma, Approaches and Perspectives. Int J Mol Sci 2021; 22:ijms22126442. [PMID: 34208575 PMCID: PMC8235515 DOI: 10.3390/ijms22126442] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Due to their high specificity, monoclonal antibodies have been widely investigated for their application in drug delivery to the central nervous system (CNS) for the treatment of neurological diseases such as stroke, Alzheimer’s, and Parkinson’s disease. Research in the past few decades has revealed that one of the biggest challenges in the development of antibodies for drug delivery to the CNS is the presence of blood–brain barrier (BBB), which acts to restrict drug delivery and contributes to the limited uptake (0.1–0.2% of injected dose) of circulating antibodies into the brain. This article reviews the various methods currently used for antibody delivery to the CNS at the preclinical stage of development and the underlying mechanisms of BBB penetration. It also describes efforts to improve or modulate the physicochemical and biochemical properties of antibodies (e.g., charge, Fc receptor binding affinity, and target affinity), to adapt their pharmacokinetics (PK), and to influence their distribution and disposition into the brain. Finally, a distinction is made between approaches that seek to modify BBB permeability and those that use a physiological approach or antibody engineering to increase uptake in the CNS. Although there are currently inherent difficulties in developing safe and efficacious antibodies that will cross the BBB, the future prospects of brain-targeted delivery of antibody-based agents are believed to be excellent.
Collapse
|
31
|
Huang CW, Chuang CP, Chen YJ, Wang HY, Lin JJ, Huang CY, Wei KC, Huang FT. Integrin α 2β 1-targeting ferritin nanocarrier traverses the blood-brain barrier for effective glioma chemotherapy. J Nanobiotechnology 2021; 19:180. [PMID: 34120610 PMCID: PMC8201891 DOI: 10.1186/s12951-021-00925-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ferritin, the natural iron storage protein complex, self-assembles into a uniform cage-like structure. Human H-ferritin (HFn) has been shown to transverse the blood-brain barrier (BBB) by binding to transferrin receptor 1 (TfR1), which is abundant in endothelial cells and overexpressed in tumors, and enters cells via endocytosis. Ferritin is easily genetically modified with various functional molecules, justifying that it possesses great potential for development into a nanocarrier drug delivery system. RESULTS In this study, a unique integrin α2β1-targeting H-ferritin (2D-HFn)-based drug delivery system was developed that highlights the feasibility of receptor-mediated transcytosis (RMT) for glioma tumor treatment. The integrin targeting α2β1 specificity was validated by biolayer interferometry in real time monitoring and followed by cell binding, chemo-drug encapsulation stability studies. Compared with naïve HFn, 2D-HFn dramatically elevated not only doxorubicin (DOX) drug loading capacity (up to 458 drug molecules/protein cage) but also tumor targeting capability after crossing BBB in an in vitro transcytosis assay (twofold) and an in vivo orthotopic glioma model. Most importantly, DOX-loaded 2D-HFn significantly suppressed subcutaneous and orthotopic U-87MG tumor progression; in particular, orthotopic glioma mice survived for more than 80 days. CONCLUSIONS We believe that this versatile nanoparticle has established a proof-of-concept platform to enable more accurate brain tumor targeting and precision treatment arrangements. Additionally, this unique RMT based ferritin drug delivery technique would accelerate the clinical development of an innovative drug delivery strategy for central nervous system diseases with limited side effects in translational medicine.
Collapse
Affiliation(s)
- Chiun-Wei Huang
- Center for Advanced Molecular Imaging and Translation (CAMIT), Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chia-Pao Chuang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, AC2-414, No.1, Sec. 4, Roosevelt Rd., Taipei, 106319, Taiwan
| | - Yan-Jun Chen
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, AC2-414, No.1, Sec. 4, Roosevelt Rd., Taipei, 106319, Taiwan
| | - Hsu-Yuan Wang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, AC2-414, No.1, Sec. 4, Roosevelt Rd., Taipei, 106319, Taiwan
| | - Jia-Jia Lin
- Center for Advanced Molecular Imaging and Translation (CAMIT), Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Feng-Ting Huang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, AC2-414, No.1, Sec. 4, Roosevelt Rd., Taipei, 106319, Taiwan.
| |
Collapse
|
32
|
Naziris N, Pippa N, Sereti E, Chrysostomou V, Kędzierska M, Kajdanek J, Ionov M, Miłowska K, Balcerzak Ł, Garofalo S, Limatola C, Pispas S, Dimas K, Bryszewska M, Demetzos C. Chimeric Stimuli-Responsive Liposomes as Nanocarriers for the Delivery of the Anti-Glioma Agent TRAM-34. Int J Mol Sci 2021; 22:ijms22126271. [PMID: 34200955 PMCID: PMC8230631 DOI: 10.3390/ijms22126271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Nanocarriers are delivery platforms of drugs, peptides, nucleic acids and other therapeutic molecules that are indicated for severe human diseases. Gliomas are the most frequent type of brain tumor, with glioblastoma being the most common and malignant type. The current state of glioma treatment requires innovative approaches that will lead to efficient and safe therapies. Advanced nanosystems and stimuli-responsive materials are available and well-studied technologies that may contribute to this effort. The present study deals with the development of functional chimeric nanocarriers composed of a phospholipid and a diblock copolymer, for the incorporation, delivery and pH-responsive release of the antiglioma agent TRAM-34 inside glioblastoma cells. Nanocarrier analysis included light scattering, protein incubation and electron microscopy, and fluorescence anisotropy and thermal analysis techniques were also applied. Biological assays were carried out in order to evaluate the nanocarrier nanotoxicity in vitro and in vivo, as well as to evaluate antiglioma activity. The nanosystems were able to successfully manifest functional properties under pH conditions, and their biocompatibility and cellular internalization were also evident. The chimeric nanoplatforms presented herein have shown promise for biomedical applications so far and should be further studied in terms of their ability to deliver TRAM-34 and other therapeutic molecules to glioblastoma cells.
Collapse
Affiliation(s)
- Nikolaos Naziris
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece; (N.N.); (N.P.)
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.K.); (J.K.); (M.I.); (K.M.)
| | - Natassa Pippa
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece; (N.N.); (N.P.)
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (V.C.); (S.P.)
| | - Evangelia Sereti
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece; (E.S.); (K.D.)
| | - Varvara Chrysostomou
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (V.C.); (S.P.)
| | - Marta Kędzierska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.K.); (J.K.); (M.I.); (K.M.)
| | - Jakub Kajdanek
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.K.); (J.K.); (M.I.); (K.M.)
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.K.); (J.K.); (M.I.); (K.M.)
| | - Katarzyna Miłowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.K.); (J.K.); (M.I.); (K.M.)
| | - Łucja Balcerzak
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland;
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (S.G.); (C.L.)
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (S.G.); (C.L.)
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (V.C.); (S.P.)
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece; (E.S.); (K.D.)
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.K.); (J.K.); (M.I.); (K.M.)
- Correspondence: (M.B.); (C.D.); Tel.: +48-426354474 (M.B.); +30-2107274596 (C.D.)
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece; (N.N.); (N.P.)
- Correspondence: (M.B.); (C.D.); Tel.: +48-426354474 (M.B.); +30-2107274596 (C.D.)
| |
Collapse
|
33
|
Delivery of Therapeutic Agents to the Central Nervous System and the Promise of Extracellular Vesicles. Pharmaceutics 2021; 13:pharmaceutics13040492. [PMID: 33916841 PMCID: PMC8067091 DOI: 10.3390/pharmaceutics13040492] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
The central nervous system (CNS) is surrounded by the blood–brain barrier (BBB), a semipermeable border of endothelial cells that prevents pathogens, solutes and most molecules from non-selectively crossing into the CNS. Thus, the BBB acts to protect the CNS from potentially deleterious insults. Unfortunately, the BBB also frequently presents a significant barrier to therapies, impeding passage of drugs and biologicals to target cells within the CNS. This review provides an overview of different approaches to deliver therapeutics across the BBB, with an emphasis in extracellular vesicles as delivery vehicles to the CNS.
Collapse
|
34
|
Persano F, Batasheva S, Fakhrullina G, Gigli G, Leporatti S, Fakhrullin R. Recent advances in the design of inorganic and nano-clay particles for the treatment of brain disorders. J Mater Chem B 2021; 9:2756-2784. [PMID: 33596293 DOI: 10.1039/d0tb02957b] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inorganic materials, in particular nanoclays and silica nanoparticles, have attracted enormous attention due to their versatile and tuneable properties, making them ideal candidates for a wide range of biomedical applications, such as drug delivery. This review aims at overviewing recent developments of inorganic nanoparticles (like porous or mesoporous silica particles) and different nano-clay materials (like montmorillonite, laponites or halloysite nanotubes) employed for overcoming the blood brain barrier (BBB) in the treatment and therapy of major brain diseases such as Alzheimer's, Parkinson's, glioma or amyotrophic lateral sclerosis. Recent strategies of crossing the BBB through invasive and not invasive administration routes by using different types of nanoparticles compared to nano-clays and inorganic particles are overviewed.
Collapse
Affiliation(s)
- Francesca Persano
- University of Salento, Department of Mathematics and Physics, Via Per Arnesano 73100, Lecce, Italy
| | | | | | | | | | | |
Collapse
|
35
|
Development of a New Polymeric Nanocarrier Dedicated to Controlled Clozapine Delivery at the Dopamine D 2-Serotonin 5-HT 1A Heteromers. Polymers (Basel) 2021; 13:polym13071000. [PMID: 33805130 PMCID: PMC8036403 DOI: 10.3390/polym13071000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Clozapine, the second generation antipsychotic drug, is one of the prominent compounds used for treatment of schizophrenia. Unfortunately, use of this drug is still limited due to serious side effects connected to its unspecific and non-selective action. Nevertheless, clozapine still remains the first-choice drug for the situation of drug-resistance schizophrenia. Development of the new strategy of clozapine delivery into well-defined parts of the brain has been a great challenge for modern science. In the present paper we focus on the presentation of a new nanocarrier for clozapine and its use for targeted transport, enabling its interaction with the dopamine D2 and serotonin 5-HT1A heteromers (D2-5-HT1A) in the brain tissue. Clozapine polymeric nanocapsules (CLO-NCs) were prepared using anionic surfactant AOT (sodium docusate) as an emulsifier, and bio-compatible polyelectrolytes such as: poly-l-glutamic acid (PGA) and poly-l-lysine (PLL). Outer layer of the carrier was grafted by polyethylene glycol (PEG). Several variants of nanocarriers containing the antipsychotic varying in physicochemical parameters were tested. This kind of approach may enable the availability and safety of the drug, improve the selectivity of its action, and finally increase effectiveness of schizophrenia therapy. Moreover, the purpose of the manuscript is to cover a wide scope of the issues, which should be considered while designing a novel means for drug delivery. It is important to determine the interactions of a new nanocarrier with many cell components on various cellular levels in order to be sure that the new nanocarrier will be safe and won’t cause undesired effects for a patient.
Collapse
|
36
|
Sheth V, Wang L, Bhattacharya R, Mukherjee P, Wilhelm S. Strategies for Delivering Nanoparticles across Tumor Blood Vessels. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007363. [PMID: 37197212 PMCID: PMC10187772 DOI: 10.1002/adfm.202007363] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Indexed: 05/19/2023]
Abstract
Nanoparticle transport across tumor blood vessels is a key step in nanoparticle delivery to solid tumors. However, the specific pathways and mechanisms of this nanoparticle delivery process are not fully understood. Here, the biological and physical characteristics of the tumor vasculature and the tumor microenvironment are explored and how these features affect nanoparticle transport across tumor blood vessels is discussed. The biological and physical methods to deliver nanoparticles into tumors are reviewed and paracellular and transcellular nanoparticle transport pathways are explored. Understanding the underlying pathways and mechanisms of nanoparticle tumor delivery will inform the engineering of safer and more effective nanomedicines for clinical translation.
Collapse
Affiliation(s)
- Vinit Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, 800 NE 10th St, Oklahoma City, OK 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, 800 NE 10th St, Oklahoma City, OK 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| |
Collapse
|
37
|
Choudhari M, Hejmady S, Narayan Saha R, Damle S, Singhvi G, Alexander A, Kesharwani P, Kumar Dubey S. Evolving new-age strategies to transport therapeutics across the blood-brain-barrier. Int J Pharm 2021; 599:120351. [PMID: 33545286 DOI: 10.1016/j.ijpharm.2021.120351] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/24/2021] [Accepted: 01/30/2021] [Indexed: 12/25/2022]
Abstract
A basic understanding of the blood-brain barrier (BBB) is essential for the novel advancements in targeting drugs specific to the brain. Neoplasm compromising the internal structure of BBB that results in impaired vasculature is called as blood tumor barrier (BTB). Besides, the BBB serves as a chief hindrance to the passage of a drug into the brain parenchyma. The small and hydrophilic drugs majorly display an absence of desired molecular characteristics required to cross the BBB. Furthermore, all classes of biologics have failed in the clinical trials of brain diseases over the past years since these biologics are large molecules that do not cross the BBB. Also, new strategies have been discovered that use the Trojan horse technology with the re-engineered biologics for BBB transport. Thus, this review delivers information about the different grades of tumors (I-IV) i.e. examples of BBB/BTB heterogenicity along with the different mechanisms for transporting the therapeutics into the brain tumors by crossing BBB. This review also provides insights into the emerging approaches of peptide delivery and the non-invasive and brain-specific molecular Trojan horse targeting technologies. Also, the several challenges in the clinical development of BBB penetrating IgG fusion protein have been discussed.
Collapse
Affiliation(s)
- Manisha Choudhari
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Siddhanth Hejmady
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Ranendra Narayan Saha
- Birla Institute of Technology and Science, Pilani, Dubai Campus, United Arab Emirates
| | - Shantanu Damle
- Colorcon Asia Pvt. Ltd., Verna Industrial Estate, Verna 403722, Goa, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Amit Alexander
- National Institute of Pharmaceutical Education and Research (NIPER GUWAHATI), Department of Pharmaceutical Technology (Formulations), Department of Pharmaceuticals, Ministry of Chemical and Fertilizers, Government of India, Sila Village, Nizsundarighopa, Changsari, Kamrup (R), Guwahati, Assam 781101, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India; R&D Healthcare Division Emami Ltd., 13, BT Road, Belgharia, Kolkata 700056, India.
| |
Collapse
|
38
|
WANG KAI, ZHANG FENGTIAN, WEN CHANGLONG, HUANG ZHIHUA, HU ZHIHAO, ZHANG YUWEN, HU FUQIANG, WEN LIJUAN. Regulation of pathological blood-brain barrier for intracranial enhanced drug delivery and anti-glioblastoma therapeutics. Oncol Res 2021. [DOI: 10.32604/or.2022.025696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
|
39
|
Pandian SRK, Pavadai P, Vellaisamy S, Ravishankar V, Palanisamy P, Sundar LM, Chandramohan V, Sankaranarayanan M, Panneerselvam T, Kunjiappan S. Formulation and evaluation of rutin-loaded solid lipid nanoparticles for the treatment of brain tumor. Naunyn Schmiedebergs Arch Pharmacol 2020; 394:735-749. [PMID: 33156389 DOI: 10.1007/s00210-020-02015-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
The primary requirement for curing cancer is the delivery of essential drug load at the cancer microenvironment with therapeutic efficacy. Considering this, the present study aims to formulate "Rutin"-encapsulated solid lipid nanoparticles (SLNs) for effective brain delivery across the blood-brain barrier (BBB). Rutin-loaded SLNs were fabricated by oil-in-water microemulsion technique and were characterized for their physicochemical properties. The in vivo biodistribution study of rutin-loaded SLNs was studied using Rattus norvegicus rats. Subsequently, in silico molecular docking and dynamic calculations were performed to examine the binding affinity as well as stability of rutin at the active site of target protein "epidermal growth factor receptor (EGFR)." Formulated rutin-loaded SLNs were predominantly spherical in shape with an average particle diameter of 100 nm. Additionally, the biocompatibility and stability have been proved in vitro. The presence and biodistribution of rutin in vivo after 54 h of injection were observed as 15.23 ± 0.32% in the brain, 8.68 ± 0.63% in the heart, 4.78 ± 0.28% in the kidney, 5.04 ± 0.37% in the liver, 0.92 ± 0.04% in the lung, and 11.52 ± 0.65% in the spleen, respectively. Molecular docking results revealed the higher binding energy of - 150.973 kJ/mol of rutin with EGFR. Molecular dynamic simulation studies demonstrated that rutin with EGFR receptor complex was highly stable at 30 ns. The observed results exemplified that the formulated rutin-loaded SLNs were stable in circulation for a period up to 5 days. Thus, rutin-encapsulated SLN formulations can be used as a promising vector to target tumors across BBB. Graphical abstract.
Collapse
Affiliation(s)
- Sureshbabu Ram Kumar Pandian
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, 626126, India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M S R Nagar, Bengaluru, Karnataka, 560054, India
| | - Sivakumar Vellaisamy
- Department of Pharmaceutics, Arulmigu Kalasalingam College of Pharmacy, Krishnankoil, Tamilnadu, 626126, India
| | - Vigneshwaran Ravishankar
- Department of Biotechnology, Mepco Schlenk Engineering College, Sivakasi, Tamilnadu, 626005, India
| | - Ponnusamy Palanisamy
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, Tamilnadu, 632014, India
| | - Lakshmi M Sundar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M S R Nagar, Bengaluru, Karnataka, 560054, India
| | - Vivek Chandramohan
- Department of Biotechnology, Siddaganga Institute of Technology, Tumakuru, Karnataka, 572103, India
| | | | - Theivendren Panneerselvam
- Department of Pharmaceutical Chemistry, Swamy Vivekananda College of Pharmacy, Elayampalayam, Namakkal, Tamilnadu, 637205, India.
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, 626126, India.
| |
Collapse
|
40
|
Ladel S, Maigler F, Flamm J, Schlossbauer P, Handl A, Hermann R, Herzog H, Hummel T, Mizaikoff B, Schindowski K. Impact of Glycosylation and Species Origin on the Uptake and Permeation of IgGs through the Nasal Airway Mucosa. Pharmaceutics 2020; 12:E1014. [PMID: 33114132 PMCID: PMC7690786 DOI: 10.3390/pharmaceutics12111014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/18/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022] Open
Abstract
Although we have recently reported the involvement of neonatal Fc receptor (FcRn) in intranasal transport, the transport mechanisms are far from being elucidated. Ex vivo porcine olfactory tissue, primary cells from porcine olfactory epithelium (OEPC) and the human cell line RPMI 2650 were used to evaluate the permeation of porcine and human IgG antibodies through the nasal mucosa. IgGs were used in their wild type and deglycosylated form to investigate the impact of glycosylation. Further, the expression of FcRn and Fc-gamma receptor (FCGR) and their interaction with IgG were analyzed. Comparable permeation rates for human and porcine IgG were observed in OEPC, which display the highest expression of FcRn. Only traces of porcine IgGs could be recovered at the basolateral compartment in ex vivo olfactory tissue, while human IgGs reached far higher levels. Deglycosylated human IgG showed significantly higher permeation in comparison to the wild type in RPMI 2650 and OEPC, but insignificantly elevated in the ex vivo model. An immunoprecipitation with porcine primary cells and tissue identified FCGR2 as a potential interaction partner in the nasal mucosa. Glycosylation sensitive receptors appear to be involved in the uptake, transport, but also degradation of therapeutic IgGs in the airway epithelial layer.
Collapse
Affiliation(s)
- Simone Ladel
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Frank Maigler
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Johannes Flamm
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Patrick Schlossbauer
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| | - Alina Handl
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Rebecca Hermann
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| | - Helena Herzog
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Thomas Hummel
- Smell & Taste Clinic, Department of Otorhinolaryngology, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany;
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany;
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| |
Collapse
|
41
|
Designing peptide nanoparticles for efficient brain delivery. Adv Drug Deliv Rev 2020; 160:52-77. [PMID: 33031897 DOI: 10.1016/j.addr.2020.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
The targeted delivery of therapeutic compounds to the brain is arguably the most significant open problem in drug delivery today. Nanoparticles (NPs) based on peptides and designed using the emerging principles of molecular engineering show enormous promise in overcoming many of the barriers to brain delivery faced by NPs made of more traditional materials. However, shortcomings in our understanding of peptide self-assembly and blood-brain barrier (BBB) transport mechanisms pose significant obstacles to progress in this area. In this review, we discuss recent work in engineering peptide nanocarriers for the delivery of therapeutic compounds to the brain: from synthesis, to self-assembly, to in vivo studies, as well as discussing in detail the biological hurdles that a nanoparticle must overcome to reach the brain.
Collapse
|
42
|
Al-azzawi S, Masheta D, Guildford A, Phillips G, Santin M. A Peptide-Based Nanocarrier for an Enhanced Delivery and Targeting of Flurbiprofen into the Brain for the Treatment of Alzheimer's Disease: An In Vitro Study. NANOMATERIALS 2020; 10:nano10081590. [PMID: 32823499 PMCID: PMC7466704 DOI: 10.3390/nano10081590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is an age-related disease caused by abnormal accumulation of amyloid-β in the brain leading to progressive tissue degeneration. Flurbiprofen (FP), a drug used to mitigate the disease progression, has low efficacy due to its limited permeability across the blood-brain barrier (BBB). In a previous work, FP was coupled at the uppermost branching of an ε-lysine-based branched carrier, its root presenting a phenylalanine moiety able to increase the hydrophobicity of the complex and enhance the transport across the BBB by adsorptive-mediated transcytosis (AMT). The present study explores a different molecular design of the FP-peptide delivery system, whereby its root presents an ApoE-mimicking peptide, a targeting ligand that could enhance transport across the BBB by receptor-mediated transcytosis (RMT). The functionalised complex was synthesised using a solid-phase peptide synthesis and characterised by mass spectrometry and FTIR. Cytotoxicity and permeability of this complex across an in vitro BBB model were analysed. Moreover, its activity and degradation to release the drug were investigated. The results revealed successful synthesis and grafting of FP molecules at the uppermost molecular branches of the lysine terminal without observed cytotoxicity. When covalently linked to the nanocarrier, FP was still active on target cells, albeit with a reduced activity, and was released as a free drug upon hydrolysis in a lysosome-mimicking medium. Noticeably, this work shows the high efficiency of RMT-driven FP delivery over delivery systems relying on AMT.
Collapse
Affiliation(s)
- Shafq Al-azzawi
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Bimolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK; (S.A.-a.); (D.M.); (A.G.); (G.P.)
- College of Pharmacy, University of Babylon, Ministry of Higher Education and Scientific Research, Hilla 51002, Iraq
| | - Dhafir Masheta
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Bimolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK; (S.A.-a.); (D.M.); (A.G.); (G.P.)
- College of Pharmacy, University of Babylon, Ministry of Higher Education and Scientific Research, Hilla 51002, Iraq
| | - Anna Guildford
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Bimolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK; (S.A.-a.); (D.M.); (A.G.); (G.P.)
- Tissue Click Ltd., Brighton BN2 6SJ, UK
| | - Gary Phillips
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Bimolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK; (S.A.-a.); (D.M.); (A.G.); (G.P.)
- Tissue Click Ltd., Brighton BN2 6SJ, UK
| | - Matteo Santin
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Bimolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK; (S.A.-a.); (D.M.); (A.G.); (G.P.)
- Correspondence:
| |
Collapse
|
43
|
Salehi A, Paturu MR, Patel B, Cain MD, Mahlokozera T, Yang AB, Lin TH, Leuthardt EC, Yano H, Song SK, Klein RS, Schmidt R, Kim AH. Therapeutic enhancement of blood-brain and blood-tumor barriers permeability by laser interstitial thermal therapy. Neurooncol Adv 2020; 2:vdaa071. [PMID: 32666049 PMCID: PMC7344247 DOI: 10.1093/noajnl/vdaa071] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background The blood–brain and blood–tumor barriers (BBB and BTB), which restrict the entry of most drugs into the brain and tumor, respectively, are a significant challenge in the treatment of glioblastoma. Laser interstitial thermal therapy (LITT) is a minimally invasive surgical technique increasingly used clinically for tumor cell ablation. Recent evidence suggests that LITT might locally disrupt BBB integrity, creating a potential therapeutic window of opportunity to deliver otherwise brain-impermeant agents. Methods We established a LITT mouse model to test if laser therapy can increase BBB/BTB permeability in vivo. Mice underwent orthotopic glioblastoma tumor implantation followed by LITT in combination with BBB tracers or the anticancer drug doxorubicin. BBB/BTB permeability was measured using fluorimetry, microscopy, and immunofluorescence. An in vitro endothelial cell model was also used to corroborate findings. Results LITT substantially disrupted the BBB and BTB locally, with increased permeability up to 30 days after the intervention. Remarkably, molecules as large as human immunoglobulin extravasated through blood vessels and permeated laser-treated brain tissue and tumors. Mechanistically, LITT decreased tight junction integrity and increased brain endothelial cell transcytosis. Treatment of mice bearing glioblastoma tumors with LITT and adjuvant doxorubicin, which is typically brain-impermeant, significantly increased animal survival. Conclusions Together, these results suggest that LITT can locally disrupt the BBB and BTB, enabling the targeted delivery of systemic therapies, including, potentially, antibody-based agents.
Collapse
Affiliation(s)
- Afshin Salehi
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Mounica R Paturu
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Bhuvic Patel
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Matthew D Cain
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Tatenda Mahlokozera
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Alicia B Yang
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Tsen-Hsuan Lin
- Department of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Eric C Leuthardt
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Hiroko Yano
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Robyn S Klein
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.,Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Robert Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Albert H Kim
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.,Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
44
|
Asha Spandana K, Bhaskaran M, Karri V, Natarajan J. A comprehensive review of nano drug delivery system in the treatment of CNS disorders. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101628] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
45
|
Dogra P, Butner JD, Nizzero S, Ruiz Ramírez J, Noureddine A, Peláez MJ, Elganainy D, Yang Z, Le AD, Goel S, Leong HS, Koay EJ, Brinker CJ, Cristini V, Wang Z. Image-guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1628. [PMID: 32314552 PMCID: PMC7507140 DOI: 10.1002/wnan.1628] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/06/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug-loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size-dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non-invasive in vivo imaging modalities. This allows for visualization and quantification of the whole-body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non-invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state-of-the-art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - María J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Applied Physics Graduate Program, Rice University, Houston, Texas, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhen Yang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anh-Dung Le
- Nanoscience and Microsystems Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - Shreya Goel
- Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
46
|
Extracellular Vesicles as Signaling Mediators and Disease Biomarkers across Biological Barriers. Int J Mol Sci 2020; 21:ijms21072514. [PMID: 32260425 PMCID: PMC7178048 DOI: 10.3390/ijms21072514] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/20/2020] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles act as shuttle vectors or signal transducers that can deliver specific biological information and have progressively emerged as key regulators of organized communities of cells within multicellular organisms in health and disease. Here, we survey the evolutionary origin, general characteristics, and biological significance of extracellular vesicles as mediators of intercellular signaling, discuss the various subtypes of extracellular vesicles thus far described and the principal methodological approaches to their study, and review the role of extracellular vesicles in tumorigenesis, immunity, non-synaptic neural communication, vascular-neural communication through the blood-brain barrier, renal pathophysiology, and embryo-fetal/maternal communication through the placenta.
Collapse
|
47
|
On-Chip Synthesis of Hyaluronic Acid-Based Nanoparticles for Selective Inhibition of CD44+ Human Mesenchymal Stem Cell Proliferation. Pharmaceutics 2020; 12:pharmaceutics12030260. [PMID: 32183027 PMCID: PMC7151101 DOI: 10.3390/pharmaceutics12030260] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
In this study, an innovative microfluidics-based method was developed for one-step synthesis of hyaluronic acid (HA)-based nanoparticles (NPs), by exploiting polyelectrolytic interactions between HA and chitosan (CS), in order to improve reliability, reproducibility and possible scale-up of the NPs preparation. The on-chip synthesis, using a staggered herringbone micromixer, allowed to produce HA/CS NPs with tailored-made size and suitable for both parenteral (117.50 ± 4.51 nm) and loco-regional (349.15 ± 38.09 nm) administration, mainly composed by HA (more than 85% wt) with high negative surface charge (< −20 mV). HA/CS NPs were successfully loaded with a challenging water-insoluble molecule, Everolimus (EVE), an FDA- and EMA-approved anticancer drug able to lead to cell cycle arrest, reduced angiogenesis and promotion of apoptosis. HA/CS NPs resulted to be massively internalized in CD44+ human mesenchymal stem cells via CD44 receptor-mediated endocytosis. HA/CS NPs selectiveness towards CD44 was highlighted by blocking CD44 receptor by anti-CD44 primary antibody and by comparison to CS-based NPs cellular uptake. Eventually, high effectiveness in inhibiting cell proliferation was demonstrated on-chip synthetized EVE loaded HA/CS NPs by tracking in vitro DNA synthesis.
Collapse
|
48
|
Xie J, Shen Z, Anraku Y, Kataoka K, Chen X. Nanomaterial-based blood-brain-barrier (BBB) crossing strategies. Biomaterials 2019; 224:119491. [PMID: 31546096 DOI: 10.1016/j.biomaterials.2019.119491] [Citation(s) in RCA: 287] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/31/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
Increasing attention has been paid to the diseases of central nervous system (CNS). The penetration efficiency of most CNS drugs into the brain parenchyma is rather limited due to the existence of blood-brain barrier (BBB). Thus, BBB crossing for drug delivery to CNS remains a significant challenge in the development of neurological therapeutics. Because of the advantageous properties (e.g., relatively high drug loading content, controllable drug release, excellent passive and active targeting, good stability, biodegradability, biocompatibility, and low toxicity), nanomaterials with BBB-crossability have been widely developed for the treatment of CNS diseases. This review summarizes the current understanding of the physiological structure of BBB, and provides various nanomaterial-based BBB-crossing strategies for brain delivery of theranostic agents, including intranasal delivery, temporary disruption of BBB, local delivery, cell penetrating peptide (CPP) mediated BBB-crossing, receptor mediated BBB-crossing, shuttle peptide mediated BBB-crossing, and cells mediated BBB-crossing. Clinicians, biologists, material scientists and chemists are expected to be interested in this review.
Collapse
Affiliation(s)
- Jinbing Xie
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China; Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Zheyu Shen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Yasutaka Anraku
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
49
|
Mahalingaiah PK, Ciurlionis R, Durbin KR, Yeager RL, Philip BK, Bawa B, Mantena SR, Enright BP, Liguori MJ, Van Vleet TR. Potential mechanisms of target-independent uptake and toxicity of antibody-drug conjugates. Pharmacol Ther 2019; 200:110-125. [DOI: 10.1016/j.pharmthera.2019.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/19/2019] [Indexed: 12/13/2022]
|
50
|
Kevadiya BD, Ottemann BM, Thomas MB, Mukadam I, Nigam S, McMillan J, Gorantla S, Bronich TK, Edagwa B, Gendelman HE. Neurotheranostics as personalized medicines. Adv Drug Deliv Rev 2019; 148:252-289. [PMID: 30421721 PMCID: PMC6486471 DOI: 10.1016/j.addr.2018.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022]
Abstract
The discipline of neurotheranostics was forged to improve diagnostic and therapeutic clinical outcomes for neurological disorders. Research was facilitated, in largest measure, by the creation of pharmacologically effective multimodal pharmaceutical formulations. Deployment of neurotheranostic agents could revolutionize staging and improve nervous system disease therapeutic outcomes. However, obstacles in formulation design, drug loading and payload delivery still remain. These will certainly be aided by multidisciplinary basic research and clinical teams with pharmacology, nanotechnology, neuroscience and pharmaceutic expertise. When successful the end results will provide "optimal" therapeutic delivery platforms. The current report reviews an extensive body of knowledge of the natural history, epidemiology, pathogenesis and therapeutics of neurologic disease with an eye on how, when and under what circumstances neurotheranostics will soon be used as personalized medicines for a broad range of neurodegenerative, neuroinflammatory and neuroinfectious diseases.
Collapse
Affiliation(s)
- Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Midhun Ben Thomas
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saumya Nigam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|