1
|
Sadat Razavi Z, Sina Alizadeh S, Sadat Razavi F, Souri M, Soltani M. Advancing neurological disorders therapies: Organic nanoparticles as a key to blood-brain barrier penetration. Int J Pharm 2025; 670:125186. [PMID: 39788400 DOI: 10.1016/j.ijpharm.2025.125186] [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: 09/04/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier (BBB) plays a vital role in protecting the central nervous system (CNS) by preventing the entry of harmful pathogens from the bloodstream. However, this barrier also presents a significant obstacle when it comes to delivering drugs for the treatment of neurodegenerative diseases and brain cancer. Recent breakthroughs in nanotechnology have paved the way for the creation of a wide range of nanoparticles (NPs) that can serve as carriers for diagnosis and therapy. Regarding their promising properties, organic NPs have the potential to be used as effective carriers for drug delivery across the BBB based on recent advancements. These remarkable NPs have the ability to penetrate the BBB using various mechanisms. This review offers a comprehensive examination of the intricate structure and distinct properties of the BBB, emphasizing its crucial function in preserving brain balance and regulating the transport of ions and molecules. The disruption of the BBB in conditions such as stroke, Alzheimer's disease, and Parkinson's disease highlights the importance of developing creative approaches for delivering drugs. Through the encapsulation of therapeutic molecules and the precise targeting of transport processes in the brain vasculature, organic NP formulations present a hopeful strategy to improve drug transport across the BBB. We explore the changes in properties of the BBB in various pathological conditions and investigate the factors that affect the successful delivery of organic NPs into the brain. In addition, we explore the most promising delivery systems associated with NPs that have shown positive results in treating neurodegenerative and ischemic disorders. This review opens up new possibilities for nanotechnology-based therapies in cerebral diseases.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | | - Fateme Sadat Razavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada; Centre for Sustainable Business, International Business University, Toronto, Canada.
| |
Collapse
|
2
|
Razavi R, Khajouei G, Divsalar F, Dawi E, Amiri M. Recent advances on brain drug delivery via nanoparticles: alternative future materials for neuroscience applications; a review. Rev Neurosci 2025:revneuro-2024-0086. [PMID: 39829237 DOI: 10.1515/revneuro-2024-0086] [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: 06/19/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025]
Abstract
Essentially, the blood-brain barrier (BBB) serves as a line of demarcation between neural tissues and the bloodstream. A unique and protective characteristic of the blood-brain barrier is its ability to maintain cerebral homeostasis by regulating the flux of molecules and ions. The inability to uphold proper functioning in any of these constituents leads to the disruption of this specialized multicellular arrangement, consequently fostering neuroinflammation and neurodegeneration. Recent advancements in nanomedicine have been regarded as a promising avenue for improving the delivery of drugs to the central nervous system in the modern era. A major benefit of this innovation is that it allows drugs to accumulate selectively within the cerebral area by circumventing the blood-brain barrier. Although brain-targeted nanomedicines have demonstrated impressive achievements, certain limitations in targeting specificity still exist. In this examination, we scrutinize the distinctive physical and chemical attributes of nanoparticles (NPs) contributing to their facilitation in BBB traversal. We explore the various mechanisms governing NP passage over the BBB, encompassing paracellular conveyance, mediated transport, as well as adsorptive- and receptor-mediated transcytosis. The therapeutic success of NPs for the treatment of brain tumors has been extensively investigated through the use of various categories of NPs. Among these are polymeric nanoparticles, liposomes, solid lipid nanoparticles, dendrimers, metallic nanoparticles, quantum dots, and nanogels. The potential utility of nanoparticles goes beyond their ability to transport pharmaceuticals. They can serve as adept imaging contrast agents, capable of being linked with imaging probes. This will facilitate tumor visualization, delineate lesion boundaries and margins, and monitor drug delivery and treatment response. Versatile nanoparticles can be engineered to effectively target neoplastic lesions, serving dual roles in diagnostic imaging and therapeutic interventions. Subsequently, this discourse explores the constraints associated with nanoparticles in the context of treating brain tumors.
Collapse
Affiliation(s)
- Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran
| | - Ghazal Khajouei
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | - Fatemeh Divsalar
- Sina Hospital, Zarand Network and Health Center, 48463 Kerman University of Medical Sciences , Kerman, Iran
| | - Elmuez Dawi
- College of Humanities and Sciences, College of Humanities and Sciences, Department of Mathematics and Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
| | - Mahnaz Amiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| |
Collapse
|
3
|
Li A, Nicolas J, Mura S. Unlocking the Potential of Hybrid Nanocomposite Hydrogels: Design, Mechanical Properties and Biomedical Performances. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202409670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Indexed: 01/06/2025]
Abstract
AbstractHybrid nanocomposite hydrogels consist of the homogeneous incorporation of nano‐objects in a hydrogel matrix. The latter, whether made of natural or synthetic materials, possesses a microporous, soft structure that makes it an ideal host for a variety of polymer and lipid‐based nano‐objects as well as metal‐ and silica‐based ones. By carefully choosing the composition and the proportions of the different constituents, hybrid hydrogels can display a wide array of properties, from simple enhancement of mechanical characteristics to specific bioactivity. This review aims to provide an overview of the state of the art in hybrid hydrogels highlighting key aspects that make them a promising choice for a variety of biomedical applications. Strategies for the preparation of hybrid hydrogels are discussed by covering the selection of individual components. The review will also explore the physico‐chemical and rheological characterization of these materials, which is essential for understanding their structure and function, ultimately satisfying specifications for the intended use. Successful examples of biomedical applications will also be presented, and the main challenges to be met will be discussed, with the aim of stimulating the research community to exploit the full potential of these materials.
Collapse
Affiliation(s)
- Anqi Li
- Université Paris‐Saclay CNRS Institut Galien Paris‐Saclay Orsay 91400 France
| | - Julien Nicolas
- Université Paris‐Saclay CNRS Institut Galien Paris‐Saclay Orsay 91400 France
| | - Simona Mura
- Université Paris‐Saclay CNRS Institut Galien Paris‐Saclay Orsay 91400 France
| |
Collapse
|
4
|
Ghalwash MM, Fouad AG, Mohammed NH, Nagib MM, Khalil SFA, Belal A, Miski SF, Albezrah NKA, Elsayed A, Hassan AHE, Roh EJ, El-Housiny S. Fabrication and In Vivo Evaluation of In Situ pH-Sensitive Hydrogel of Sonidegib-Invasomes via Intratumoral Delivery for Basal Cell Skin Cancer Management. Pharmaceuticals (Basel) 2024; 18:31. [PMID: 39861094 PMCID: PMC11769384 DOI: 10.3390/ph18010031] [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: 12/13/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Basal cell skin cancer (BCSC) develops when skin cells proliferate uncontrollably. Sonidegib (SDB) is a therapeutic option for the treatment of BCSC by inhibiting hedgehog signaling. The problems with SDB's low solubility, poor bioavailability, resistance, poor targeting, and first-pass action make it less effective when taken orally. This investigation set out to design an intratumoral in situ pH-sensitive hydrogel of SDB-invasomes (IPHS-INV) that can effectively treat BCSC by improving SDB's bioavailability, sustainability, targeting, and efficacy while also reducing its resistance and undesirable side effects. METHODS Numerous S-INV formulations were developed using Box-Behnken Design Expert and tested before settling on the optimum S-INV formulation. An experimental 7, 12-dimethylbenzanthracene (DMBA) carcinoma rat model was used for in vivo studies of the IPHS-INV formulation after it was combined with chitosan. RESULTS Phospholipids (1.72% w/w), cholesterol (0.15% w/w), ethanol (1% v/v), and cineole (1.5% v/v) were shown to be the optimal components in the SDB-invasome formulation. The IPHS-INV formulation outperformed the permeation and bioavailability of free SDB by 7.14 and 6 times, respectively, and sustained its release by 57.41%. The IPHS-INV formulation showed a decrease in tumor volume of 99.05% and a reduction of hypercellular tumors, indicating its anti-cancer activity. The intratumoral IPHS-INV formulation maintained a higher concentration of SDB in tumors, indicating its targeting activity. CONCLUSIONS These findings support the use of the intratumoral IPHS-INV formulation as an effective strategy for the treatment of BCSC.
Collapse
Affiliation(s)
- Maha M. Ghalwash
- Department of Pharmaceutics and Drug Manufacturing, Faculty of Pharmacy, Modern University for Technology and Information, Cairo 11435, Egypt
| | - Amr Gamal Fouad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Nada H. Mohammed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Deraya University, Minya 61768, Egypt
| | - Marwa M. Nagib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Cairo 11435, Egypt
| | | | - Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Samar F. Miski
- Pharmacology and Toxicology Department, College of Pharmacy, Taibah University, Madina 42278, Saudi Arabia
| | | | - Amani Elsayed
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Ahmed H. E. Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Eun Joo Roh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Shaimaa El-Housiny
- Department of Pharmaceutics and Drug Manufacturing, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11435, Egypt
| |
Collapse
|
5
|
Zhang S, Zhong R, Younis MR, He H, Xu H, Li G, Yang R, Lui S, Wang Y, Wu M. Hydrogel Applications in the Diagnosis and Treatment of Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:65754-65778. [PMID: 39366948 DOI: 10.1021/acsami.4c11855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Glioblastoma multiforme (GBM), a common malignant neurological tumor, has boundaries indistinguishable from those of normal tissue, making complete surgical removal ineffective. The blood-brain barrier (BBB) further impedes the efficacy of radiotherapy and chemotherapy, leading to suboptimal treatment outcomes and a heightened probability of recurrence. Hydrogels offer multiple advantages for GBM diagnosis and treatment, including overcoming the BBB for improved drug delivery, controlled drug release for long-term efficacy, and enhanced relaxation properties of magnetic resonance imaging (MRI) contrast agents. Hydrogels, with their excellent biocompatibility and customizability, can mimic the in vivo microenvironment, support tumor cell culture, enable drug screening, and facilitate the study of tumor invasion and metastasis. This paper reviews the classification of hydrogels and recent research for the diagnosis and treatment of GBM, including their applications as cell culture platforms and drugs including imaging contrast agents carriers. The mechanisms of drug release from hydrogels and methods to monitor the activity of hydrogel-loaded drugs are also discussed. This review is intended to facilitate a more comprehensive understanding of the current state of GBM research. It offers insights into the design of integrated hydrogel-based GBM diagnosis and treatment with the objective of achieving the desired therapeutic effect and improving the prognosis of GBM.
Collapse
Affiliation(s)
- Shuaimei Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Renming Zhong
- Radiotherapy Physics & Technology Center, Cancer Center, West China Hospital, Chengdu, Sichuan 610041, P. R. China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Hualong He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ruiyan Yang
- Department of Biology, Macalester College, Saint Paul, Minnesota 55105, United States
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
6
|
Sharma P, Xu J, Puduvalli VK. Epichaperome Inhibition by PU-H71-Mediated Targeting of HSP90 Sensitizes Glioblastoma Cells to Alkylator-Induced DNA Damage. Cancers (Basel) 2024; 16:3934. [PMID: 39682123 DOI: 10.3390/cancers16233934] [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: 10/09/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Targeted therapies have been largely ineffective against glioblastoma (GBM) owing to the tumor's heterogeneity and intrinsic and adaptive treatment resistance. Targeting multiple pro-survival pathways simultaneously may overcome these limitations and yield effective treatments. Heat shock protein 90 (HSP90), an essential component of the epichaperome complex, is critical for the proper folding and activation of several pro-survival oncogenic proteins that drive GBM biology. METHODS Using a panel of biochemical and biological assays, we assessed the expression of HSP90 and its downstream targets and the effects of PU-H71, a highly specific and potent HSP90 inhibitor, on target modulation, downstream biochemical alterations, cell cycle progression, proliferation, migration, and apoptosis in patient-derived glioma stem-like cells (GSCs) with molecular profiles characteristic of GBM, as well as commercial glioma cell lines and normal human astrocytes (NHAs). RESULTS HSP90 inhibition by PU-H71 in GSCs significantly reduced cell proliferation, colony formation, wound healing, migration, and angiogenesis. In glioma cells, but not NHAs, potent PU-H71-mediated HSP90 inhibition resulted in the downregulation of pro-survival client proteins such as EGFR, MAPK, AKT, and S6. This reduction in pro-survival signals increased glioma cells' sensitivity to temozolomide, a monofunctional alkylator, and the combination of PU-H71 and temozolomide had greater anticancer efficacy than either agent alone. CONCLUSIONS These results confirm that HSP90 is a strong pro-survival factor in molecularly heterogeneous gliomas and suggest that epichaperome inhibition with HSP90 inhibitors warrants further investigation for the treatment of gliomas.
Collapse
Affiliation(s)
- Pratibha Sharma
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Jihong Xu
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Vinay K Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| |
Collapse
|
7
|
Rivero-Buceta E, Bernal-Gómez A, Vidaurre-Agut C, Lopez Moncholi E, María Benlloch J, Moreno Manzano V, David Vera Donoso C, Botella P. Prostate cancer chemotherapy by intratumoral administration of Docetaxel-Mesoporous silica nanomedicines. Int J Pharm 2024; 664:124623. [PMID: 39191333 DOI: 10.1016/j.ijpharm.2024.124623] [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: 05/31/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Docetaxel (DTX) is a recommended treatment in patients with metastasic prostate cancer (PCa), despite its therapeutic efficacy is limited by strong systemic toxicity. However, in localized PCa, intratumoral (IT) administration of DTX could be an alternative to consider that may help to overcome the disadvantages of conventional intravenous (IV) therapy. In this context, we here present the first in vivo preclinical study of PCa therapy with nanomedicines of mesoporous silica nanoparticles (MSN) and DTX by IT injection over a xenograft mouse model bearing human prostate adenocarcinoma tumors. The efficacy and tolerability, the biodistribution and the histopathology after therapy have been investigated for the DTX nanomedicine and the free drug, and compared with the IV administration of DTX. The obtained results demonstrate that IT injection of DTX and DTX nanomedicines allows precise and selective therapy of non-metastatic PCa and minimize systemic diffusion of the drug, showing superior activity than IV route. This allows reducing the therapeutic dose by one order and widens substantially the therapeutic window for this drug. Furthermore, the use of DTX nanomedicines as IT injection promotes strong antitumor efficacy and drug accumulation at the tumor site, improving the results obtained with the free drug by the same route.
Collapse
Affiliation(s)
- Eva Rivero-Buceta
- Instituto de Tecnología Química (UPV-CSIC), Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avda. de los Naranjos s/n, 46022 València, Spain
| | - Adrián Bernal-Gómez
- Escuela de Doctorado, Universidad Católica de Valencia, Plaza de San Agustín 3, 46001 Valencia, Spain; Hospital Universitario y Politécnico La Fe, Av. Fernando Abril Martorell, 106, 46026 Valencia, Spain
| | - Carla Vidaurre-Agut
- Instituto de Tecnología Química (UPV-CSIC), Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avda. de los Naranjos s/n, 46022 València, Spain
| | - Eric Lopez Moncholi
- Centro Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Jose María Benlloch
- Instituto de Instrumentación para Imagen Molecular (I3M), Centro Mixto CSIC-Universitat Politècnica de Valencia, Camino de Vera s/n, 46022 Valencia, Spain
| | | | - César David Vera Donoso
- Escuela de Doctorado, Universidad Católica de Valencia, Plaza de San Agustín 3, 46001 Valencia, Spain; Hospital Universitario y Politécnico La Fe, Av. Fernando Abril Martorell, 106, 46026 Valencia, Spain
| | - Pablo Botella
- Instituto de Tecnología Química (UPV-CSIC), Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avda. de los Naranjos s/n, 46022 València, Spain.
| |
Collapse
|
8
|
Butkiene G, Daugelaite AM, Poderys V, Marin R, Steponkiene S, Kazlauske E, Uzieliene I, Daunoravicius D, Jaque D, Rotomskis R, Skripka A, Vetrone F, Karabanovas V. Synergistic Enhancement of Photodynamic Cancer Therapy with Mesenchymal Stem Cells and Theranostic Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:49092-49103. [PMID: 39252643 PMCID: PMC11420871 DOI: 10.1021/acsami.4c10098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Nanoparticles engineered to combat cancer and other life-threatening diseases may significantly improve patient outcomes. However, inefficient nanoparticle delivery to tumors limits their use and necessitates the development of complex delivery approaches. Here, we examine this issue by harnessing the tumor-homing abilities of human mesenchymal stem cells (MSCs) to deliver a decoupled theranostic complex of rare earth-doped nanoparticles (dNPs) and photosensitizer chlorin e6 (Ce6) to tumors. We show that both bone-marrow- and skin-derived MSCs can transport the dNP-Ce6 complex inside tumor spheroids, which is challenging to accomplish by passive delivery alone. MSCs deliver the dNP-Ce6 complex across the tumor spheroid, facilitating more effective photodynamic damage and tumor destruction than passively accumulated dNP-Ce6. The dNP-Ce6 complex also provides the built-in ability to monitor the MSC migration without causing undesired phototoxicity, which is essential for maximal and side-effect-free delivery of nanoparticles. Our results demonstrate how MSCs can be used as delivery vehicles for the transportation of the dNP-Ce6 complex, addressing the limitations of passive nanoparticle delivery and providing light-based theranostics.
Collapse
Affiliation(s)
- Greta Butkiene
- Biomedical Physics Laboratory of the National Cancer Institute, P. Baublio St. 3b, Vilnius LT-08406, Lithuania
| | - Aleja Marija Daugelaite
- Biomedical Physics Laboratory of the National Cancer Institute, P. Baublio St. 3b, Vilnius LT-08406, Lithuania
- Faculty of Medicine, Vilnius University, M. K. Ciurlionio g. 21, Vilnius LT-03101, Lithuania
| | - Vilius Poderys
- Biomedical Physics Laboratory of the National Cancer Institute, P. Baublio St. 3b, Vilnius LT-08406, Lithuania
| | - Riccardo Marin
- Nano for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid 28049, Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Simona Steponkiene
- Biomedical Physics Laboratory of the National Cancer Institute, P. Baublio St. 3b, Vilnius LT-08406, Lithuania
| | - Evelina Kazlauske
- Biomedical Physics Laboratory of the National Cancer Institute, P. Baublio St. 3b, Vilnius LT-08406, Lithuania
- Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Sauletekio Ave. 11, Vilnius LT-10223, Lithuania
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu g. 5, Vilnius LT-08406, Lithuania
| | | | - Daniel Jaque
- Nano for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid 28049, Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, Madrid 28049, Spain
- Nano for Bioimaging Group (nanoBIG), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Ramón y Cajal, Madrid 28034, Spain
| | - Ricardas Rotomskis
- Biomedical Physics Laboratory of the National Cancer Institute, P. Baublio St. 3b, Vilnius LT-08406, Lithuania
- Biophotonics Group, Laser Research Center, Physics Faculty, Vilnius University, Sauletekio Ave. 9, Vilnius LT-10222, Lithuania
| | - Artiom Skripka
- Nano for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid 28049, Spain
- Centre Énergie, Matériaux et Télécommunications, Institut National de la Recherche Scientifique (INRS), Université du Québec, Varennes, Québec J3X 1P7, Canada
| | - Fiorenzo Vetrone
- Centre Énergie, Matériaux et Télécommunications, Institut National de la Recherche Scientifique (INRS), Université du Québec, Varennes, Québec J3X 1P7, Canada
- Centre Québécois sur les Matériaux Fonctionnels (CQMF)/Quebec Centre for Advanced Materials (QCAM), Montréal, Québec J3X 1P7, Canada
| | - Vitalijus Karabanovas
- Biomedical Physics Laboratory of the National Cancer Institute, P. Baublio St. 3b, Vilnius LT-08406, Lithuania
- Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Sauletekio Ave. 11, Vilnius LT-10223, Lithuania
| |
Collapse
|
9
|
Ascione F, Ferraro R, Dogra P, Cristini V, Guido S, Caserta S. Gradient-induced instability in tumour spheroids unveils the impact of microenvironmental nutrient changes. Sci Rep 2024; 14:20837. [PMID: 39242641 PMCID: PMC11379688 DOI: 10.1038/s41598-024-69570-6] [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: 04/03/2024] [Accepted: 08/06/2024] [Indexed: 09/09/2024] Open
Abstract
Tumours often display invasive behaviours that induce fingering, branching and fragmentation processes. The phenomenon, known as diffusional instability, is driven by differential cell proliferation, migration, and death due to the presence of metabolite and catabolite concentration gradients. An understanding of the intricate dynamics of this spatially heterogeneous process plays a key role in the investigation of tumour growth and invasion. In this study, we developed an in vitro tumour invasion assay to investigate cell invasiveness in tumour spheroids under a chemotactic stimulus. Our method, employing tumour spheroids seeded in a 3D collagen gel within a microfluidic chemotaxis chamber, focuses on the role of diffusive gradients. Using Time-Lapse Microscopy, the dynamic evolution of tumour spheroids was monitored in real-time, providing a comprehensive view of the morphological changes and cell migration patterns under different chemotactic conditions. Specifically, we explored the impact of fetal bovine serum (FBS) gradients on the behaviour of CT26 mouse colon carcinoma cells and compared the effects of varying FBS concentrations to two isotropic control conditions. Furthermore, a finite element in silico model was developed to quantify the diffusive flow of nutrients in the chemotaxis chamber and obtain a detailed understanding of tumour dynamics. Our findings reveal that the presence of a chemotactic gradient significantly influences tumour invasiveness, with higher concentrations of nutrients associated with increased cancer growth and cell migration.
Collapse
Affiliation(s)
- Flora Ascione
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, P. Le V. Tecchio 80, 80125, Naples, Italy
| | - Rosalia Ferraro
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, P. Le V. Tecchio 80, 80125, Naples, Italy
- CEINGE Advanced Biotechnologies Franco Salvatore, Via G. Salvatore 436, 80131, Naples, Italy
| | - Prashant Dogra
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Stefano Guido
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, P. Le V. Tecchio 80, 80125, Naples, Italy
- CEINGE Advanced Biotechnologies Franco Salvatore, Via G. Salvatore 436, 80131, Naples, Italy
| | - Sergio Caserta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, P. Le V. Tecchio 80, 80125, Naples, Italy.
- CEINGE Advanced Biotechnologies Franco Salvatore, Via G. Salvatore 436, 80131, Naples, Italy.
| |
Collapse
|
10
|
Delgado JF, Owen JW, Pritchard WF, Varble NA, Lopez-Silva TL, Mikhail AS, Arrichiello A, Ray T, Morhard R, Borde T, Saccenti L, Xu S, Rivera J, Schneider JP, Karanian JW, Wood BJ. Ultrasound and x-ray imageable poloxamer-based hydrogel for loco-regional therapy delivery in the liver. Sci Rep 2024; 14:20455. [PMID: 39227382 PMCID: PMC11372101 DOI: 10.1038/s41598-024-70992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
Intratumoral injections have the potential for enhanced cancer treatment efficacy while reducing costs and systemic exposure. However, intratumoral drug injections can result in substantial off-target leakage and are invisible under standard imaging modalities like ultrasound (US) and x-ray. A thermosensitive poloxamer-based gel for drug delivery was developed that is visible using x-ray imaging (computed tomography (CT), cone beam CT, fluoroscopy), as well as using US by means of integrating perfluorobutane-filled microbubbles (MBs). MBs content was optimized using tissue mimicking phantoms and ex vivo bovine livers. Gel formulations less than 1% MBs provided gel depositions that were clearly identifiable on US and distinguishable from tissue background and with minimal acoustic artifacts. The cross-sectional areas of gel depositions obtained with US and CT imaging were similar in studies using ex vivo bovine liver and postmortem in situ swine liver. The gel formulation enhanced multimodal image-guided navigation, enabling fusion of ultrasound and x-ray/CT imaging, which may enhance targeting, definition of spatial delivery, and overlap of tumor and gel. Although speculative, such a paradigm for intratumoral drug delivery might streamline clinical workflows, reduce radiation exposure by reliance on US, and boost the precision and accuracy of drug delivery targeting during procedures. Imageable gels may also provide enhanced temporal and spatial control of intratumoral conformal drug delivery.
Collapse
Affiliation(s)
- Jose F Delgado
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
- Fischell Department of Bioengineering, University of Maryland, College Park. Maryland, USA.
| | - Joshua W Owen
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - William F Pritchard
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| | - Nicole A Varble
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Philips Healthcare, Cambridge, MA, USA
| | - Tania L Lopez-Silva
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Andrew S Mikhail
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Antonio Arrichiello
- Department of Diagnostic and Interventional Radiology, UOS of Interventional Radiology, Ospedale Maggiore Di Lodi, Largo Donatori del Sangue, Lodi, Italy
| | - Trisha Ray
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Robert Morhard
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tabea Borde
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Laetitia Saccenti
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Sheng Xu
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jocelyne Rivera
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Institute of Biomedical Engineering, St. Catherine's College, University of Oxford, Oxford, UK
| | - Joel P Schneider
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - John W Karanian
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
- Fischell Department of Bioengineering, University of Maryland, College Park. Maryland, USA.
| |
Collapse
|
11
|
Longobardi G, Moore TL, Conte C, Ungaro F, Satchi‐Fainaro R, Quaglia F. Polyester nanoparticles delivering chemotherapeutics: Learning from the past and looking to the future to enhance their clinical impact in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1990. [PMID: 39217459 PMCID: PMC11670051 DOI: 10.1002/wnan.1990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Polymeric nanoparticles (NPs), specifically those comprised of biodegradable and biocompatible polyesters, have been heralded as a game-changing drug delivery platform. In fact, poly(α-hydroxy acids) such as polylactide (PLA), poly(lactide-co-glycolide) (PLGA), and poly(ε-caprolactone) (PCL) have been heavily researched in the past three decades as the material basis of polymeric NPs for drug delivery applications. As materials, these polymers have found success in resorbable sutures, biodegradable implants, and even monolithic, biodegradable platforms for sustained release of therapeutics (e.g., proteins and small molecules) and diagnostics. Few fields have gained more attention in drug delivery through polymeric NPs than cancer therapy. However, the clinical translational of polymeric nanomedicines for treating solid tumors has not been congruent with the fervor or funding in this particular field of research. Here, we attempt to provide a comprehensive snapshot of polyester NPs in the context of chemotherapeutic delivery. This includes a preliminary exploration of the polymeric nanomedicine in the cancer research space. We examine the various processes for producing polyester NPs, including methods for surface-functionalization, and related challenges. After a detailed overview of the multiple factors involved with the delivery of NPs to solid tumors, the crosstalk between particle design and interactions with biological systems is discussed. Finally, we report state-of-the-art approaches toward effective delivery of NPs to tumors, aiming at identifying new research areas and re-evaluating the reasons why some research avenues have underdelivered. We hope our effort will contribute to a better understanding of the gap to fill and delineate the future research work needed to bring polyester-based NPs closer to clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Thomas Lee Moore
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Claudia Conte
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Francesca Ungaro
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Sagol School of NeurosciencesTel Aviv UniversityTel AvivIsrael
| | - Fabiana Quaglia
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
12
|
Aizik G, Ostertag-Hill CA, Chakraborty P, Choi W, Pan M, Mankus DV, Lytton-Jean AKR, Kohane DS. Injectable hydrogel based on liposome self-assembly for controlled release of small hydrophilic molecules. Acta Biomater 2024; 183:101-110. [PMID: 38834149 PMCID: PMC11239275 DOI: 10.1016/j.actbio.2024.05.044] [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: 11/28/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Controlled release of low molecular weight hydrophilic drugs, administered locally, allows maintenance of high concentrations at the target site, reduces systemic side effects, and improves patient compliance. Injectable hydrogels are commonly used as a vehicle. However, slow release of low molecular weight hydrophilic drugs is very difficult to achieve, mainly due to a rapid diffusion of the drug out of the drug delivery system. Here we present an injectable and self-healing hydrogel based entirely on the self-assembly of liposomes. Gelation of liposomes, without damaging their structural integrity, was induced by modifying the cholesterol content and surface charge. The small hydrophilic molecule, sodium fluorescein, was loaded either within the extra-liposomal space or encapsulated into the aqueous cores of the liposomes. This encapsulation strategy enabled the achievement of controlled and adjustable release profiles, dependent on the mechanical strength of the gel. The hydrogel had a high mechanical strength, minimal swelling, and slow degradation. The liposome-based hydrogel had prolonged mechanical stability in vivo with benign tissue reaction. This work presents a new class of injectable hydrogel that holds promise as a versatile drug delivery system. STATEMENT OF SIGNIFICANCE: The porous nature of hydrogels poses a challenge for delivering small hydrophilic drug, often resulting in initial burst release and shorten duration of release. This issue is particularly pronounced with physically crosslinked hydrogels, since their matrix can swell and dissipate rapidly, but even in cases where the polymers in the hydrogel are covalently cross-linked, small molecules can be rapidly released through its porous mesh. Here we present an injectable self-healing hydrogel based entirely on the self-assembly of liposomes. Small hydrophilic molecules were entrapped inside the extra-liposomal space or loaded into the aqueous cores of the liposomes, allowing controlled and tunable release profiles.
Collapse
Affiliation(s)
- Gil Aizik
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Claire A Ostertag-Hill
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Priyadarshi Chakraborty
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wonmin Choi
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michelle Pan
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David V Mankus
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Abigail K R Lytton-Jean
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Jiang Z, Fu Y, Shen H. Development of Intratumoral Drug Delivery Based Strategies for Antitumor Therapy. Drug Des Devel Ther 2024; 18:2189-2202. [PMID: 38882051 PMCID: PMC11179649 DOI: 10.2147/dddt.s467835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
Research for tumor treatment with significant therapy effects and minimal side-effects has been widely carried over the past few decades. Different drug forms have received a lot of attention. However, systemic biodistribution induces efficacy and safety issues. Intratumoral delivery of agents might overcome these problems because of its abundant tumor accumulation and retention, thereby reducing side effects. Delivering hydrogels, nanoparticles, microneedles, and microspheres drug carriers directly to tumors can realize not only targeted tumor therapy but also low side-effects. Furthermore, intratumoral administration has been integrated with treatment strategies such as chemotherapy, enhancing radiotherapy, immunotherapy, phototherapy, magnetic fluid hyperthermia, and multimodal therapy. Some of these strategies are ongoing clinical trials or applied clinically. However, many barriers hinder it from being an ideal and widely used option, such as decreased drug penetration impeded by collagen fibers of a tumor, drug squeezed out by high density and high pressure, mature intratumoral injection technique. In this review, we systematically discuss intratumoral delivery of different drug carriers and current development of intratumoral therapy strategies.
Collapse
Affiliation(s)
- Zhimei Jiang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, People’s Republic of China
| | - Yuzhi Fu
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, People’s Republic of China
| | - Hongxin Shen
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, People’s Republic of China
| |
Collapse
|
14
|
Mao M, Wu Y, He Q. Recent advances in targeted drug delivery for the treatment of glioblastoma. NANOSCALE 2024; 16:8689-8707. [PMID: 38606460 DOI: 10.1039/d4nr01056f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is one of the highly malignant brain tumors characterized by significant morbidity and mortality. Despite the recent advancements in the treatment of GBM, major challenges persist in achieving controlled drug delivery to tumors. The management of GBM poses considerable difficulties primarily due to unresolved issues in the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB) and GBM microenvironment. These factors limit the uptake of anti-cancer drugs by the tumor, thus limiting the therapeutic options. Current breakthroughs in nanotechnology provide new prospects concerning unconventional drug delivery approaches for GBM treatment. Specifically, swimming nanorobots show great potential in active targeted delivery, owing to their autonomous propulsion and improved navigation capacities across biological barriers, which further facilitate the development of GBM-targeted strategies. This review presents an overview of technological progress in different drug administration methods for GBM. Additionally, the limitations in clinical translation and future research prospects in this field are also discussed. This review aims to provide a comprehensive guideline for researchers and offer perspectives on further development of new drug delivery therapies to combat GBM.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
15
|
Marques AC, Costa PC, Velho S, Amaral MH. Rheological and Injectability Evaluation of Sterilized Poloxamer-407-Based Hydrogels Containing Docetaxel-Loaded Lipid Nanoparticles. Gels 2024; 10:307. [PMID: 38786224 PMCID: PMC11121564 DOI: 10.3390/gels10050307] [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: 03/30/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Nanostructured lipid carriers (NLCs) have the potential to increase the bioavailability and reduce the side effects of docetaxel (DTX). However, only a small fraction of nanoparticles given intravenously can reach a solid tumor. In situ-forming gels combined with nanoparticles facilitate local administration and promote drug retention at the tumor site. Injectable hydrogels based on poloxamer 407 are excellent candidates for this hybrid nanoparticle-hydrogel system because of their thermoresponsive behavior and biocompatibility. Therefore, this work aimed to develop injectable poloxamer hydrogels containing NLCs for intratumoral delivery of DTX. To ensure sterility, the obtained hydrogels were autoclaved (121 °C for 15 min) after preparation. Then, the incorporation of NLCs into the poloxamer hydrogels and the impact of steam sterilization on the nanocomposite hydrogels were evaluated concerning sol-gel transition, injectability, and physicochemical stability. All formulations were extruded through the tested syringe-needle systems with acceptable force (2.2-13.4 N) and work (49.5-317.7 N·mm) of injection. Following steam sterilization, injection became easier in most cases, and the physicochemical properties of all hydrogels remained practically unchanged according to the spectroscopical and thermal analysis. The rheological evaluation revealed that the nanocomposite hydrogels were liquid at 25 °C and underwent rapid gelation at 37 °C. However, their sterilized counterparts gelled at 1-2 °C above body temperature, suggesting that the autoclaving conditions employed had rendered these nanocomposite hydrogels unsuitable for local drug delivery.
Collapse
Affiliation(s)
- Ana Camila Marques
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paulo C. Costa
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Sérgia Velho
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Maria Helena Amaral
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
16
|
Zhang Q, Zhang Y, Chen H, Sun LN, Zhang B, Yue DS, Wang CL, Zhang ZF. Injectable hydrogel with doxorubicin-loaded ZIF-8 nanoparticles for tumor postoperative treatments and wound repair. Sci Rep 2024; 14:9983. [PMID: 38693143 PMCID: PMC11063161 DOI: 10.1038/s41598-024-57664-0] [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: 11/09/2023] [Accepted: 03/20/2024] [Indexed: 05/03/2024] Open
Abstract
The need for tumor postoperative treatments aimed at recurrence prevention and tissue regeneration have raised wide considerations in the context of the design and functionalization of implants. Herein, an injectable hydrogel system encapsulated with anti-tumor, anti-oxidant dual functional nanoparticles has been developed in order to prevent tumor relapse after surgery and promote wound repair. The utilization of biocompatible gelatin methacryloyl (GelMA) was geared towards localized therapeutic intervention. Zeolitic imidazolate framework-8@ceric oxide (ZIF-8@CeO2, ZC) nanoparticles (NPs) were purposefully devised for their proficiency as reactive oxygen species (ROS) scavengers. Furthermore, injectable GelMA hydrogels loaded with ZC NPs carrying doxorubicin (ZC-DOX@GEL) were tailored as multifunctional postoperative implants, ensuring the efficacious eradication of residual tumor cells and alleviation of oxidative stress. In vitro and in vivo experiments were conducted to substantiate the efficacy in cancer cell elimination and the prevention of tumor recurrence through the synergistic chemotherapy approach employed with ZC-DOX@GEL. The acceleration of tissue regeneration and in vitro ROS scavenging attributes of ZC@GEL were corroborated using rat models of wound healing. The results underscore the potential of the multifaceted hydrogels presented herein for their promising application in tumor postoperative treatments.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin Lung Cancer Center, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yu Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin Lung Cancer Center, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hui Chen
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin Lung Cancer Center, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei-Na Sun
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin Lung Cancer Center, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Bin Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin Lung Cancer Center, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Dong-Sheng Yue
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin Lung Cancer Center, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chang-Li Wang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin Lung Cancer Center, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhen-Fa Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Tianjin Lung Cancer Center, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
17
|
Hsia T, Chen Y. RNA-encapsulating lipid nanoparticles in cancer immunotherapy: From pre-clinical studies to clinical trials. Eur J Pharm Biopharm 2024; 197:114234. [PMID: 38401743 DOI: 10.1016/j.ejpb.2024.114234] [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: 11/19/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2024]
Abstract
Nanoparticle-based delivery systems such as RNA-encapsulating lipid nanoparticles (RNA LNPs) have dramatically advanced in function and capacity over the last few decades. RNA LNPs boast of a diverse array of external and core configurations that enhance targeted delivery and prolong circulatory retention, advancing therapeutic outcomes. Particularly within the realm of cancer immunotherapies, RNA LNPs are increasingly gaining prominence. Pre-clinical in vitro and in vivo studies have laid a robust foundation for new and ongoing clinical trials that are actively enrolling patients for RNA LNP cancer immunotherapy. This review explores RNA LNPs, starting from their core composition to their external membrane formulation, set against a backdrop of recent clinical breakthroughs. We further elucidate the LNP delivery avenues, broach the prevailing challenges, and contemplate the future perspectives of RNA LNP-mediated immunotherapy.
Collapse
Affiliation(s)
- Tiffaney Hsia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
18
|
Li L, Xiang F, Wang F, Liu Y. Preparation and antitumor study of intelligent injectable hydrogel: Carboxymethyl chitosan-aldehyde gum Arabic composite graphene oxide hydrogel. Int J Biol Macromol 2024; 259:129429. [PMID: 38232874 DOI: 10.1016/j.ijbiomac.2024.129429] [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: 10/23/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
In this study, we used polyaldehyde gum Arabic (OGA) and carboxymethyl chitosan (CMCS) as a gel matrix to form an injectable self-healing hydrogel by Schiff-base bonding. Further, graphene oxide (GO) was loaded with doxorubicin (DOX) to the hydrogel, which resulted in a CMCS-OGA/GO@DOX hydrogel. We achieved a DOX drug loading capacity of 43.80 ± 1.13 %. Rheological studies showed that GO hydrogels have improved mechanical properties. The in vitro release profile showed pH responsiveness with 88.21 % DOX release at pH 5.5. Biocompatibility studies showed that the hydrogel composition had good cytocompatibility with L929 cells. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay showed a cell survival rate of 93.88 % within 48 h. The DOX-loaded hydrogel exhibited higher cell mortality in breast cancer cells (4 T1), with an inhibition rate of 79.4 % at 48 h. Acridine orange/ethidium bromide staining experiments on 4 T1 cells showed that when loaded with the same DOX concentration, the hydrogel significantly reduced the toxic effects on normal cells, whereas it had significant cytotoxic effects on cancer cells. This result indicates that the prepared GO hydrogel drug delivery system can serve as a novel approach for localized breast cancer treatment.
Collapse
Affiliation(s)
- Li Li
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Fengting Xiang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Fan Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Yu Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China; LiaoNing University Judicial Authentication Center, Shenyang 110036, China.
| |
Collapse
|
19
|
Baghdasaryan A, Liu H, Ren F, Hsu R, Jiang Y, Wang F, Zhang M, Grigoryan L, Dai H. Intratumor injected gold molecular clusters for NIR-II imaging and cancer therapy. Proc Natl Acad Sci U S A 2024; 121:e2318265121. [PMID: 38261618 PMCID: PMC10835035 DOI: 10.1073/pnas.2318265121] [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: 10/19/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Surgical resections of solid tumors guided by visual inspection of tumor margins have been performed for over a century to treat cancer. Near-infrared (NIR) fluorescence labeling/imaging of tumor in the NIR-I (800 to 900 nm) range with systemically administrated fluorophore/tumor-targeting antibody conjugates have been introduced to improve tumor margin delineation, tumor removal accuracy, and patient survival. Here, we show Au25 molecular clusters functionalized with phosphorylcholine ligands (AuPC, ~2 nm in size) as a preclinical intratumorally injectable agent for NIR-II/SWIR (1,000 to 3,000 nm) fluorescence imaging-guided tumor resection. The AuPC clusters were found to be uniformly distributed in the 4T1 murine breast cancer tumor upon intratumor (i.t.) injection. The phosphocholine coating afforded highly stealth clusters, allowing a high percentage of AuPC to fill the tumor interstitial fluid space homogeneously. Intra-operative surgical navigation guided by imaging of the NIR-II fluorescence of AuPC allowed for complete and non-excessive tumor resection. The AuPC in tumors were also employed as a photothermal therapy (PTT) agent to uniformly heat up and eradicate tumors. Further, we performed in vivo NIR-IIb (1,500 to 1,700 nm) molecular imaging of the treated tumor using a quantum dot-Annexin V (QD-P3-Anx V) conjugate, revealing cancer cell apoptosis following PTT. The therapeutic functionalities of AuPC clusters combined with rapid renal excretion, high biocompatibility, and safety make them promising for clinical translation.
Collapse
Affiliation(s)
- Ani Baghdasaryan
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| | - Haoran Liu
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| | - Fuqiang Ren
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| | - RuSiou Hsu
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| | - Yingying Jiang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| | - Feifei Wang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| | - Mengzhen Zhang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| | - Lilit Grigoryan
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA94305
| | - Hongjie Dai
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA94305
| |
Collapse
|
20
|
Bakhrushina EO, Mikhel IB, Buraya LM, Moiseev ED, Zubareva IM, Belyatskaya AV, Evzikov GY, Bondarenko AP, Krasnyuk II, Krasnyuk II. Implantation of In Situ Gelling Systems for the Delivery of Chemotherapeutic Agents. Gels 2024; 10:44. [PMID: 38247767 PMCID: PMC10815592 DOI: 10.3390/gels10010044] [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: 11/30/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Implantation is a modern method of administering chemotherapeutic agents, with a highly targeted effect and better patient tolerance due to the low frequency of administration. Implants are capable of controlled release, which makes them a viable alternative to infusional chemotherapy, allowing patients to enjoy a better quality of life without the need for prolonged hospitalization. Compared to subcutaneous implantation, intratumoral implantation has a number of significant advantages in terms of targeting and side effects, but this area of chemotherapy is still poorly understood in terms of clinical trials. At the same time, there are more known developments of drugs in the form of implants and injections for intratumoral administration. The disadvantages of classical intratumoral implants are the need for surgical intervention to install the system and the increased risk of tumor rupture noted by some specialists. The new generation of implants are in situ implants-systems formed in the tumor due to a phase transition (sol-gel transition) under the influence of various stimuli. Among this systems some are highly selective for a certain type of malignant neoplasm. Such systems are injected and have all the advantages of intratumoral injections, but due to the phase transition occurring in situ, they form depot forms that allow the long-term release of chemotherapeutic agents.
Collapse
Affiliation(s)
- Elena O. Bakhrushina
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Iosif B. Mikhel
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Liliya M. Buraya
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Egor D. Moiseev
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Irina M. Zubareva
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
- Department of Pharmacology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia
| | - Anastasia V. Belyatskaya
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Grigory Y. Evzikov
- Department of Nervous Diseases and Neurosurgery, N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia;
| | | | - Ivan I. Krasnyuk
- Department of Analytical, Physical and Colloidal Chemistry, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia;
| | - Ivan I. Krasnyuk
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| |
Collapse
|
21
|
Thenuwara G, Curtin J, Tian F. Advances in Diagnostic Tools and Therapeutic Approaches for Gliomas: A Comprehensive Review. SENSORS (BASEL, SWITZERLAND) 2023; 23:9842. [PMID: 38139688 PMCID: PMC10747598 DOI: 10.3390/s23249842] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
Gliomas, a prevalent category of primary malignant brain tumors, pose formidable clinical challenges due to their invasive nature and limited treatment options. The current therapeutic landscape for gliomas is constrained by a "one-size-fits-all" paradigm, significantly restricting treatment efficacy. Despite the implementation of multimodal therapeutic strategies, survival rates remain disheartening. The conventional treatment approach, involving surgical resection, radiation, and chemotherapy, grapples with substantial limitations, particularly in addressing the invasive nature of gliomas. Conventional diagnostic tools, including computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET), play pivotal roles in outlining tumor characteristics. However, they face limitations, such as poor biological specificity and challenges in distinguishing active tumor regions. The ongoing development of diagnostic tools and therapeutic approaches represents a multifaceted and promising frontier in the battle against this challenging brain tumor. The aim of this comprehensive review is to address recent advances in diagnostic tools and therapeutic approaches for gliomas. These innovations aim to minimize invasiveness while enabling the precise, multimodal targeting of localized gliomas. Researchers are actively developing new diagnostic tools, such as colorimetric techniques, electrochemical biosensors, optical coherence tomography, reflectometric interference spectroscopy, surface-enhanced Raman spectroscopy, and optical biosensors. These tools aim to regulate tumor progression and develop precise treatment methods for gliomas. Recent technological advancements, coupled with bioelectronic sensors, open avenues for new therapeutic modalities, minimizing invasiveness and enabling multimodal targeting with unprecedented precision. The next generation of multimodal therapeutic strategies holds potential for precision medicine, aiding the early detection and effective management of solid brain tumors. These innovations offer promise in adopting precision medicine methodologies, enabling early disease detection, and improving solid brain tumor management. This review comprehensively recognizes the critical role of pioneering therapeutic interventions, holding significant potential to revolutionize brain tumor therapeutics.
Collapse
Affiliation(s)
- Gayathree Thenuwara
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland;
- Institute of Biochemistry, Molecular Biology, and Biotechnology, University of Colombo, Colombo 00300, Sri Lanka
| | - James Curtin
- Faculty of Engineering and Built Environment, Technological University Dublin, Bolton Street, D01 K822 Dublin, Ireland;
| | - Furong Tian
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland;
| |
Collapse
|
22
|
Song X, Qian H, Yu Y. Nanoparticles Mediated the Diagnosis and Therapy of Glioblastoma: Bypass or Cross the Blood-Brain Barrier. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302613. [PMID: 37415556 DOI: 10.1002/smll.202302613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Glioblastoma is one of the most aggressive central nervous system malignancies with high morbidity and mortality. Current clinical approaches, including surgical resection, radiotherapy, and chemotherapy, are limited by the difficulty of targeting brain lesions accurately, leading to disease recurrence and fatal outcomes. The lack of effective treatments has prompted researchers to continuously explore novel therapeutic strategies. In recent years, nanomedicine has made remarkable progress and expanded its application in brain drug delivery, providing a new treatment for brain tumors. Against this background, this article reviews the application and progress of nanomedicine delivery systems in brain tumors. In this paper, the mechanism of nanomaterials crossing the blood-brain barrier is summarized. Furthermore, the specific application of nanotechnology in glioblastoma is discussed in depth.
Collapse
Affiliation(s)
- Xiaowei Song
- Department of Radiology, Anhui Provincial Institute of Translational Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, P. R. China
- Research Center of Clinical Medical Imaging, Hefei, 230022, China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230011, P. R. China
- Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, 230011, China
| | - Yongqiang Yu
- Department of Radiology, Anhui Provincial Institute of Translational Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, P. R. China
- Research Center of Clinical Medical Imaging, Hefei, 230022, China
| |
Collapse
|
23
|
Tsai LH, Young TH, Yen CH, Yao WC, Chang CH. Intratumoral thermo-chemotherapeutic alginate hydrogel containing doxorubicin loaded PLGA nanoparticle and heating agent. Int J Biol Macromol 2023; 251:126221. [PMID: 37572819 DOI: 10.1016/j.ijbiomac.2023.126221] [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: 05/09/2023] [Revised: 07/03/2023] [Accepted: 08/05/2023] [Indexed: 08/14/2023]
Abstract
Chemotherapy has been widely used to treat cancer; however, the non-specific systemic toxicity of chemotherapeutic agents has always been an issue. Local injection treatment is a strategy used to reduce the undesired adverse effects of chemotherapeutic drugs. In addition, chemotherapeutic agents combined with thermotherapy are effective in further enhancing therapeutic potency. In the present study, we prepared an injectable hydrogel, namely, doxorubicin (DOX)-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticle (DPN) and magnetite nanoparticle (MNP) embedded in alginate hydrogel (DPN/MNP-HG), where DPN and MNP were the chemotherapeutic and heating agents, respectively, for intratumoral thermo-chemotherapy. Injectable DPN/MNP-HG, which possesses solid-like elastic properties, was conveniently prepared via ionic cross-linking at room-temperature. When exposed to an alternating magnetic field (AMF), DPN/MNP-HG exhibited controllable heat generation with a reversible temperature-rise profile. Regarding the kinetics of DOX release, both with and without AMF, DPN/MNP-HG exhibited a slow initial burst and sustained release profile. In cytotoxicity studies and subcutaneous mouse cancer models, successful thermo-chemotherapy with DPN/MNP-HG resulted in significantly lower cell viability and increased tumor-growth suppression; mice also exhibited good tolerance to injected DPN/MNP-HG both with(+) and without AMF application. In conclusion, the proposed thermo-chemotherapeutic DPN/MNP-HG for local intratumoral injection is a promising formulation for cancer treatment.
Collapse
Affiliation(s)
- Li-Hui Tsai
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Tai-Horng Young
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan; Department of Biomedical Engineering, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chia-Hsiang Yen
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Wei-Cheng Yao
- Department of Anesthesiology and Pain Medicine, Min-Sheng General Hospital, Taoyuan 330, Taiwan
| | - Chih-Hao Chang
- Department of Orthopedics, National Taiwan University Hospital Jin-Shan Branch, New Taipei City 20844, Taiwan; Department of Orthopedics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| |
Collapse
|
24
|
Beola L, Iturrioz-Rodríguez N, Pucci C, Bertorelli R, Ciofani G. Drug-Loaded Lipid Magnetic Nanoparticles for Combined Local Hyperthermia and Chemotherapy against Glioblastoma Multiforme. ACS NANO 2023; 17:18441-18455. [PMID: 37698887 PMCID: PMC10540267 DOI: 10.1021/acsnano.3c06085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023]
Abstract
Glioblastoma multiforme (GBM) is a devastating tumor of the central nervous system, currently missing an effective treatment. The therapeutic gold standard consists of surgical resection followed by chemotherapy (usually with temozolomide, TMZ) and/or radiotherapy. TMZ does not, however, provide significant survival benefit after completion of treatment because of development of chemoresistance and of heavy side effects of systemic administration. Improvement of conventional treatments and complementary therapies are urgently needed to increase patient survival and quality of life. Stimuli-responsive lipid-based drug delivery systems offer promising prospects to overcome the limitations of the current treatments. In this work, multifunctional lipid-based magnetic nanovectors functionalized with the peptide angiopep-2 and loaded with TMZ (Ang-TMZ-LMNVs) were tested to enhance specific GBM therapy on an in vivo model. Exposure to alternating magnetic fields (AMFs) enabled magnetic hyperthermia to be performed, that works in synergy with the chemotherapeutic agent. Studies on orthotopic human U-87 MG-Luc2 tumors in nude mice have shown that Ang-TMZ-LMNVs can accumulate and remain in the tumor after local administration without crossing over into healthy tissue, effectively suppressing tumor invasion and proliferation and significantly prolonging the median survival time when combined with the AMF stimulation. This powerful synergistic approach has proven to be a robust and versatile nanoplatform for an effective GBM treatment.
Collapse
Affiliation(s)
- Lilianne Beola
- Smart
Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| | - Nerea Iturrioz-Rodríguez
- Smart
Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| | - Carlotta Pucci
- Smart
Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| | - Rosalia Bertorelli
- Translational
Pharmacology, Istituto Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Gianni Ciofani
- Smart
Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| |
Collapse
|
25
|
Sun B, Wu W, Narasipura EA, Ma Y, Yu C, Fenton OS, Song H. Engineering nanoparticle toolkits for mRNA delivery. Adv Drug Deliv Rev 2023; 200:115042. [PMID: 37536506 DOI: 10.1016/j.addr.2023.115042] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
The concept of using mRNA to produce its own medicine in situ in the body makes it an ideal drug candidate, holding great potential to revolutionize the way we approach medicine. The unique characteristics of mRNA, as well as its customizable biomedical functions, call for the rational design of delivery systems to protect and transport mRNA molecules. In this review, a nanoparticle toolkit is presented for the development of mRNA-based therapeutics from a drug delivery perspective. Nano-delivery systems derived from either natural systems or chemical synthesis, in the nature of organic or inorganic materials, are summarised. Delivery strategies in controlling the tissue targeting and mRNA release, as well as the role of nanoparticles in building and boosting the activity of mRNA drugs, have also been introduced. In the end, our insights into the clinical and translational development of mRNA nano-drugs are presented.
Collapse
Affiliation(s)
- Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Weixi Wu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
26
|
Yun WS, Kim J, Lim DK, Kim DH, Jeon SI, Kim K. Recent Studies and Progress in the Intratumoral Administration of Nano-Sized Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2225. [PMID: 37570543 PMCID: PMC10421122 DOI: 10.3390/nano13152225] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
Over the last 30 years, diverse types of nano-sized drug delivery systems (nanoDDSs) have been intensively explored for cancer therapy, exploiting their passive tumor targetability with an enhanced permeability and retention effect. However, their systemic administration has aroused some unavoidable complications, including insufficient tumor-targeting efficiency, side effects due to their undesirable biodistribution, and carrier-associated toxicity. In this review, the recent studies and advancements in intratumoral nanoDDS administration are generally summarized. After identifying the factors to be considered to enhance the therapeutic efficacy of intratumoral nanoDDS administration, the experimental results on the application of intratumoral nanoDDS administration to various types of cancer therapies are discussed. Subsequently, the reports on clinical studies of intratumoral nanoDDS administration are addressed in short. Intratumoral nanoDDS administration is proven with its versatility to enhance the tumor-specific accumulation and retention of therapeutic agents for various therapeutic modalities. Specifically, it can improve the efficacy of therapeutic agents with poor bioavailability by increasing their intratumoral concentration, while minimizing the side effect of highly toxic agents by restricting their delivery to normal tissues. Intratumoral administration of nanoDDS is considered to expand its application area due to its potent ability to improve therapeutic effects and relieve the systemic toxicities of nanoDDSs.
Collapse
Affiliation(s)
- Wan Su Yun
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jeongrae Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Kwon Lim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hwee Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
27
|
Marques AC, Costa PC, Velho S, Amaral MH. Injectable Poloxamer Hydrogels for Local Cancer Therapy. Gels 2023; 9:593. [PMID: 37504472 PMCID: PMC10379388 DOI: 10.3390/gels9070593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023] Open
Abstract
The widespread push to invest in local cancer therapies comes from the need to overcome the limitations of systemic treatment options. In contrast to intravenous administration, local treatments using intratumoral or peritumoral injections are independent of tumor vasculature and allow high concentrations of therapeutic agents to reach the tumor site with minimal systemic toxicity. Injectable biodegradable hydrogels offer a clear advantage over other delivery systems because the former requires no surgical procedures and promotes drug retention at the tumor site. More precisely, in situ gelling systems based on poloxamers have garnered considerable attention due to their thermoresponsive behavior, biocompatibility, ease of preparation, and possible incorporation of different anticancer agents. Therefore, this review focuses on the use of injectable thermoresponsive hydrogels based on poloxamers and their physicochemical and biological characterization. It also includes a summary of these hydrogel applications in local cancer therapies using chemotherapy, phototherapy, immunotherapy, and gene therapy.
Collapse
Affiliation(s)
- Ana Camila Marques
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paulo Cardoso Costa
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sérgia Velho
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - Maria Helena Amaral
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
28
|
Dam P, Celik M, Ustun M, Saha S, Saha C, Kacar EA, Kugu S, Karagulle EN, Tasoglu S, Buyukserin F, Mondal R, Roy P, Macedo MLR, Franco OL, Cardoso MH, Altuntas S, Mandal AK. Wound healing strategies based on nanoparticles incorporated in hydrogel wound patches. RSC Adv 2023; 13:21345-21364. [PMID: 37465579 PMCID: PMC10350660 DOI: 10.1039/d3ra03477a] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
The intricate, tightly controlled mechanism of wound healing that is a vital physiological mechanism is essential to maintaining the skin's natural barrier function. Numerous studies have focused on wound healing as it is a massive burden on the healthcare system. Wound repair is a complicated process with various cell types and microenvironment conditions. In wound healing studies, novel therapeutic approaches have been proposed to deliver an effective treatment. Nanoparticle-based materials are preferred due to their antibacterial activity, biocompatibility, and increased mechanical strength in wound healing. They can be divided into six main groups: metal NPs, ceramic NPs, polymer NPs, self-assembled NPs, composite NPs, and nanoparticle-loaded hydrogels. Each group shows several advantages and disadvantages, and which material will be used depends on the type, depth, and area of the wound. Better wound care/healing techniques are now possible, thanks to the development of wound healing strategies based on these materials, which mimic the extracellular matrix (ECM) microenvironment of the wound. Bearing this in mind, here we reviewed current studies on which NPs have been used in wound healing and how this strategy has become a key biotechnological procedure to treat skin infections and wounds.
Collapse
Affiliation(s)
- Paulami Dam
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Merve Celik
- Biomedical Engineering Graduate Program, TOBB University of Economics and Technology Ankara 06560 Turkey
| | - Merve Ustun
- Graduate School of Sciences and Engineering, Koç University Istanbul 34450 Turkey
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
| | - Sayantan Saha
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Chirantan Saha
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Elif Ayse Kacar
- Graduate Program of Tissue Engineering, Institution of Health Sciences, University of Health Sciences Turkey Istanbul Turkey
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
| | - Senanur Kugu
- Graduate Program of Tissue Engineering, Institution of Health Sciences, University of Health Sciences Turkey Istanbul Turkey
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
| | - Elif Naz Karagulle
- Biomedical Engineering Graduate Program, TOBB University of Economics and Technology Ankara 06560 Turkey
| | - Savaş Tasoglu
- Mechanical Engineering Department, School of Engineering, Koç University Istanbul Turkey
- Koç University Translational Medicine Research Center (KUTTAM), Koç University Istanbul Turkey
| | - Fatih Buyukserin
- Department of Biomedical Engineering, TOBB University of Economics and Technology Ankara 06560 Turkey
| | - Rittick Mondal
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Priya Roy
- Department of Law, Raiganj University North Dinajpur West Bengal India
| | - Maria L R Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária 79070900 Campo Grande Mato Grosso do Sul 70790160 Brazil
| | - Octávio L Franco
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco Campo Grande 79117900 Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília Brasília DF Brazil
| | - Marlon H Cardoso
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária 79070900 Campo Grande Mato Grosso do Sul 70790160 Brazil
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco Campo Grande 79117900 Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília Brasília DF Brazil
| | - Sevde Altuntas
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
- Department of Tissue Engineering, Institution of Health Sciences, University of Health Sciences Turkey Istanbul Turkey
| | - Amit Kumar Mandal
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
- Centre for Nanotechnology Sciences (CeNS), Raiganj University North Dinajpur West Bengal India
| |
Collapse
|
29
|
Padmakumar S, Amiji MM. Long-Acting Therapeutic Delivery Systems for the Treatment of Gliomas. Adv Drug Deliv Rev 2023; 197:114853. [PMID: 37149040 DOI: 10.1016/j.addr.2023.114853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115.
| |
Collapse
|
30
|
Li M, Tang H, Xiong Y, Yuan Z, He L, Han L. Pluronic F127 coating performance on PLGA nanoparticles: Enhanced flocculation and instability. Colloids Surf B Biointerfaces 2023; 226:113328. [PMID: 37156026 DOI: 10.1016/j.colsurfb.2023.113328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Nanoparticles (NPs) can be incorporated into hydrogels to obtain multifunctional hybrid systems to meet the delivery needs of different drugs. However, the stability of NPs in hydrogels is rarely revealed. In this article, we tried to explore the underlying mechanism of an interesting phenomenon that poly(lactic-co-glycolic acid) (PLGA) nanoparticles (PNPs) could flocculate and deposit in Pluronic F127 (F127) hydrogels at 4 °C. The results showed that this flocculation was relevant to the type of emulsifier formulated in PNPs, the particle materials and the F127 concentration, but independent of PLGA polymer end groups. Exactly, PNPs containing polyvinyl alcohol (PVA) as the emulsifier flocculated in F127 solution with a concentration above 15 %. The flocculated PNPs possessed increased particle size, decreased zeta potential, reduced hydrophobicity and an obvious coating layer, and these characteristics could be restored almost to the original state after two washes of flocculated PNPs with water. Moreover, the flocculation had no impact on the long-term size stability and drug-loading capacity of PNPs, and F127-treated PNPs showed improved cellular uptake than untreated PNPs. These results provide the evidence that adsorption of high concentrations of F127 on the surface of PNPs/PVA may lead to flocculation, and the flocculation is reversible by simply washing the flocs with water. To the best of our knowledge, this is the first study to scientifically explore the stability of PNPs in F127 hydrogels, providing theoretical and experimental support for the rational design and further development of nanoparticle-hydrogel composite.
Collapse
Affiliation(s)
- Meng Li
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Haiyu Tang
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Yu Xiong
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Zhixiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
31
|
Pal K, Sheth RA. Engineering the Tumor Immune Microenvironment through Minimally Invasive Interventions. Cancers (Basel) 2022; 15:196. [PMID: 36612192 PMCID: PMC9818918 DOI: 10.3390/cancers15010196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment (TME) is a unique landscape that poses several physical, biochemical, and immune barriers to anti-cancer therapies. The rapidly evolving field of immuno-engineering provides new opportunities to dismantle the tumor immune microenvironment by efficient tumor destruction. Systemic delivery of such treatments can often have limited local effects, leading to unwanted offsite effects such as systemic toxicity and tumor resistance. Interventional radiologists use contemporary image-guided techniques to locally deliver these therapies to modulate the immunosuppressive TME, further accelerating tumor death and invoking a better anti-tumor response. These involve local therapies such as intratumoral drug delivery, nanorobots, nanoparticles, and implantable microdevices. Physical therapies such as photodynamic therapy, electroporation, hyperthermia, hypothermia, ultrasound therapy, histotripsy, and radiotherapy are also available for local tumor destruction. While the interventional radiologist can only locally manipulate the TME, there are systemic offsite recruitments of the immune response. This is known as the abscopal effect, which leads to more significant anti-tumoral downstream effects. Local delivery of modern immunoengineering methods such as locoregional CAR-T therapy combined with immune checkpoint inhibitors efficaciously modulates the immunosuppressive TME. This review highlights the various advances and technologies available now to change the TME and revolutionize oncology from a minimally invasive viewpoint.
Collapse
Affiliation(s)
| | - Rahul A. Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
32
|
Meng L, Wang Z, Hou Z, Wang H, Zhang X, Zhang X, He X, Zhang X, Qin B, Li J, Zhang Z, Xue X, Wei Y. Study of epirubicin sustained-release chemoablation in tumor suppression and tumor microenvironment remodeling. Front Immunol 2022; 13:1064047. [PMID: 36605217 PMCID: PMC9807901 DOI: 10.3389/fimmu.2022.1064047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Although intratumoral chemoablation can obtain an impressive therapeutic effect, there is still incomplete ablation and tumor recurrence in some patients. This could be due to the short retention time of the drug in the tumor, the limited distribution of intratumoral drugs, and, beyond that, the immunotolerance caused by the tumor microenvironment (TME). There is still an urgent need to find an optimal drug sustained-release carrier and figure out the impact of regional injection to TME. Methods In this study, we supposed to use polyethylene glycol (PEG) hydrogel as a drug carrier to improve the retention time of the drug to extend the exposure of tumor cells and investigate the feasibility of combination local Epirubicin injection with anti-PD-L1. Results The results revealed obvious tumor suppression based on the tumor volume and the inhibition time of tumor growth in the A549 lung cancer mouse model after local injection. Furthermore, the enhanced antitumor effects of the combination of systematic anti- programmed death ligand 1 (PD-L1) therapy with local chemoablation (EPI-GEL/PD-L1) for abscopal tumor reduction in the 4T1 breast model were also observed. Flow cytometry analysis of the tumor and blood samples showed significant variations in the proportions of PD-L1+ and CD3+CD8+PD-1+ cells before and after anti-PD-L1 therapy. On day 4 after local injection of the EPI gel, the expression of PD-L1 in abscopal tumors was upregulated, while the expression of PD-L1 in bilateral tumors in mice was significantly reduced after anti-PD-L1 treatment. The proportion of CD3+CD8+PD-1+ cells in the tumor and circulating blood in the EPI-GEL/PD-L1 group was decreased compared with that in the EPI-GEL (single injection of epirubicin) group. Discussion The combination of local injection of the chemoablation agent with anti-PD-L1 monoclonal antibody (mAb) therapy may strengthen the antitumor activity, and the use of PEG hydrogel as the drug carrier can extend the retention time of the chemoablation agent around the tumor, maintaining a long-term tumor-killing activity.
Collapse
Affiliation(s)
- Liangliang Meng
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Zhenjun Wang
- Department of Radiology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhonghui Hou
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Hufei Wang
- National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Xiao Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xiaobo Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xiaofeng He
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xin Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Boyu Qin
- Department of Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jing Li
- Department of Radiology, Characteristic Medical Center, Chinese People’s Armed Police Force, Tianjin, China
| | - Zhongliang Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xiaodong Xue
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Yingtian Wei
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China,*Correspondence: Yingtian Wei,
| |
Collapse
|
33
|
Park S, Kim J, Lee C. Injectable rapidly dissolving needle-type gelatin implant capable of delivering high concentrations of H2O2 through intratumoral injection. Biomed Pharmacother 2022; 156:113910. [DOI: 10.1016/j.biopha.2022.113910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/24/2022] Open
|
34
|
Terracciano R, Carcamo-Bahena Y, Royal ALR, Messina L, Delk J, Butler EB, Demarchi D, Grattoni A, Wang Z, Cristini V, Dogra P, Filgueira CS. Zonal Intratumoral Delivery of Nanoparticles Guided by Surface Functionalization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:13983-13994. [PMID: 36318182 PMCID: PMC9671122 DOI: 10.1021/acs.langmuir.2c02319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/13/2022] [Indexed: 06/12/2023]
Abstract
Delivery of small molecules and anticancer agents to malignant cells or specific regions within a tumor is limited by penetration depth and poor spatial drug distribution, hindering anticancer efficacy. Herein, we demonstrate control over gold nanoparticle (GNP) penetration and spatial distribution across solid tumors by administering GNPs with different surface chemistries at a constant injection rate via syringe pump. A key finding in this study is the discovery of different zone-specific accumulation patterns of intratumorally injected nanoparticles dependent on surface functionalization. Computed tomography (CT) imaging performed in vivo of C57BL/6 mice harboring Lewis lung carcinoma (LLC) tumors on their flank and gross visualization of excised tumors consistently revealed that intratumorally administered citrate-GNPs accumulate in particle clusters in central areas of the tumor, while GNPs functionalized with thiolated phosphothioethanol (PTE-GNPs) and thiolated polyethylene glycol (PEG-GNPs) regularly accumulate in the tumor periphery. Further, PEG functionalization resulted in larger tumoral surface coverage than PTE, reaching beyond the outer zone of the tumor mass and into the surrounding stroma. To understand the dissimilarities in spatiotemporal evolution across the different GNP surface chemistries, we modeled their intratumoral transport with reaction-diffusion equations. Our results suggest that GNP surface passivation affects nanoparticle reactivity with the tumor microenvironment, leading to differential transport behavior across tumor zones. The present study provides a mechanistic understanding of the factors affecting spatiotemporal distribution of nanoparticles in the tumor. Our proof of concept of zonal delivery within the tumor may prove useful for directing anticancer therapies to regions of biomarker overexpression.
Collapse
Affiliation(s)
- Rossana Terracciano
- Department
of Nanomedicine, Houston Methodist Research
Institute, Houston, Texas77030, United States
- Department
of Electronics and Telecommunications, Politecnico
di Torino, Torino10129, Italy
| | - Yareli Carcamo-Bahena
- Department
of Nanomedicine, Houston Methodist Research
Institute, Houston, Texas77030, United States
| | - Amber Lee R. Royal
- Department
of Nanomedicine, Houston Methodist Research
Institute, Houston, Texas77030, United States
| | - Luca Messina
- Univestià
degli Studi di Napoli Federico II, Naples80138, Italy
| | - Jack Delk
- Texas
A&M University, College
Station, Texas77843, United States
| | - E. Brian Butler
- Department
of Radiation Oncology, Houston Methodist
Research Institute, Houston, Texas77030, United States
| | - Danilo Demarchi
- Department
of Electronics and Telecommunications, Politecnico
di Torino, Torino10129, Italy
| | - Alessandro Grattoni
- Department
of Nanomedicine, Houston Methodist Research
Institute, Houston, Texas77030, United States
- Department
of Radiation Oncology, Houston Methodist
Research Institute, Houston, Texas77030, United States
- Department
of Surgery, Houston Methodist Research Institute, Houston, Texas77030, United States
| | - Zhihui Wang
- Mathematics
in Medicine Program, Houston Methodist Research
Institute, Houston, Texas77030, United
States
- Department
of Imaging Physics, University of Texas
MD Anderson Cancer Center, Houston, Texas77030, United States
- Department
of Physiology and Biophysics, Weill Cornell
Medical College, New York, New York10022, United States
| | - Vittorio Cristini
- Mathematics
in Medicine Program, Houston Methodist Research
Institute, Houston, Texas77030, United
States
- Department
of Imaging Physics, University of Texas
MD Anderson Cancer Center, Houston, Texas77030, United States
- Physiology,
Biophysics, and Systems Biology Program, Graduate School of Medical
Sciences, Weill Cornell Medicine, New York, New York10022, United States
| | - Prashant Dogra
- Mathematics
in Medicine Program, Houston Methodist Research
Institute, Houston, Texas77030, United
States
- Department
of Physiology and Biophysics, Weill Cornell
Medical College, New York, New York10022, United States
| | - Carly S. Filgueira
- Department
of Nanomedicine, Houston Methodist Research
Institute, Houston, Texas77030, United States
- Department
of Cardiovascular Surgery, Houston Methodist
Research Institute, Houston, Texas77030, United States
| |
Collapse
|
35
|
Nunes D, Loureiro JA, Pereira MC. Drug Delivery Systems as a Strategy to Improve the Efficacy of FDA-Approved Alzheimer's Drugs. Pharmaceutics 2022; 14:2296. [PMID: 36365114 PMCID: PMC9694621 DOI: 10.3390/pharmaceutics14112296] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 08/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with a high impact worldwide, accounting for more than 46 million cases. The continuous increase of AD demands the fast development of preventive and curative therapeutic strategies that are truly effective. The drugs approved for AD treatment are classified into acetylcholinesterase inhibitors and N-methyl-D-aspartate receptor antagonists. The therapeutic effectiveness of those drugs is hindered by their restricted access to the brain due to the blood-brain barrier, low bioavailability, and poor pharmacokinetic properties. In addition, the drugs are reported to have undesirable side effects. Several drug delivery systems (DDSs) have been widely exploited to address these issues. DDSs serve as drug carriers, combining the ability to deliver drugs locally and in a targeted manner with the ability to release them in a controlled and sustained manner. As a result, the pharmacological therapeutic effectiveness is raised, while the unwanted side effects induced by the unspecific distribution decrease. This article reviews the recently developed DDSs to increase the efficacy of Food and Drug Administration-approved AD drugs.
Collapse
Affiliation(s)
- Débora Nunes
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana A. Loureiro
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria Carmo Pereira
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
36
|
Hydrogel on a Smart Nanomaterial Interface to Carry Therapeutics for Digitalized Glioma Treatment. Gels 2022; 8:gels8100664. [PMID: 36286164 PMCID: PMC9601840 DOI: 10.3390/gels8100664] [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: 09/05/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 12/02/2022] Open
Abstract
Glioma is considered the primary brain tumor to cause brain illnesses, and it is difficult to treat and shows resistance to various routine therapeutics. The most common treatments to cure glioma are the surgical removal of tumors followed by adjuvant chemotherapy and radiation therapy. The latest biocompatible interfaces have been incorporated into therapeutic modalities such as the targeted delivery of drugs using hydrogels to treat and manage brain glioma. This review illustrates the applications of the multimodal hydrogel as the carrier of therapeutics, gene therapy, therapeutic tactics, and glioma devices. The scientific articles were retrieved from 2019 to 2022 on Google Scholar and the Scopus database and screened to determine whether they were suitable for review. The 20 articles that fit the study are summarized in this review. These studies indicated that the sizes of the hydrogel range from 28 nm to 500 nm. There are 16 out of 20 articles that also explain the post-surgical application of hydrogels, and 13 out of 20 articles are employed in 3D culture and other structural manifestations of hydrogels. The pros of the hydrogel include the quick formulation for a sufficient filling of irregular damage sites, solubilizing hydrophobic drugs, continuously slowing drug release, provision of a 3D cell growth environment, improving efficacy, targetability of soluble biomolecules, increasing patient compliance, and decreased side effects. The cons of the hydrogel include difficult real-time monitoring, genetic manipulations, the cumbersome synchronized release of components, and lack of safety data. The prospects of the hydrogel may include the development of electronic hydrogel sensors that can be used to enhance guidance for the precise targeting patterns using patient-specific pathological idiosyncrasies. This technology has the potential to revolutionize the precision medicine approaches that would aid in the early detection and management of solid brain tumors.
Collapse
|
37
|
Ma Q, Li Q, Cai X, Zhou P, Wu Z, Wang B, Ma W, Fu S. Injectable hydrogels as drug delivery platform for in-situ treatment of malignant tumor. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
38
|
Zhao J, Wang L, Zhang H, Liao B, Li Y. Progress of Research in In Situ Smart Hydrogels for Local Antitumor Therapy: A Review. Pharmaceutics 2022; 14:pharmaceutics14102028. [PMID: 36297463 PMCID: PMC9611441 DOI: 10.3390/pharmaceutics14102028] [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: 07/26/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Cancer seriously threatens human health. Surgery, radiotherapy and chemotherapy are the three pillars of traditional cancer treatment, with targeted therapy and immunotherapy emerging over recent decades. Standard drug regimens are mostly executed via intravenous injection (IV), especially for chemotherapy agents. However, these treatments pose severe risks, including off-target toxic side effects, low drug accumulation and penetration at the tumor site, repeated administration, etc., leading to inadequate treatment and failure to meet patients’ needs. Arising from these challenges, a local regional anticancer strategy has been proposed to enhance therapeutic efficacy and concomitantly reduce systemic toxicity. With the advances in biomaterials and our understanding of the tumor microenvironment, in situ stimulus-responsive hydrogels, also called smart hydrogels, have been extensively investigated for local anticancer therapy due to their injectability, compatibility and responsiveness to various stimuli (pH, enzyme, heat, light, magnetic fields, electric fields etc.). Herein, we focus on the latest progress regarding various stimuli that cause phase transition and drug release from smart hydrogels in local regional anticancer therapy. Additionally, the challenges and future trends of the reviewed in situ smart hydrogels for local drug delivery are summarized and proposed.
Collapse
|
39
|
El Kheir W, Marcos B, Virgilio N, Paquette B, Faucheux N, Lauzon MA. Drug Delivery Systems in the Development of Novel Strategies for Glioblastoma Treatment. Pharmaceutics 2022; 14:1189. [PMID: 35745762 PMCID: PMC9227363 DOI: 10.3390/pharmaceutics14061189] [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: 05/01/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a grade IV glioma considered the most fatal cancer of the central nervous system (CNS), with less than a 5% survival rate after five years. The tumor heterogeneity, the high infiltrative behavior of its cells, and the blood-brain barrier (BBB) that limits the access of therapeutic drugs to the brain are the main reasons hampering the current standard treatment efficiency. Following the tumor resection, the infiltrative remaining GBM cells, which are resistant to chemotherapy and radiotherapy, can further invade the surrounding brain parenchyma. Consequently, the development of new strategies to treat parenchyma-infiltrating GBM cells, such as vaccines, nanotherapies, and tumor cells traps including drug delivery systems, is required. For example, the chemoattractant CXCL12, by binding to its CXCR4 receptor, activates signaling pathways that play a critical role in tumor progression and invasion, making it an interesting therapeutic target to properly control the direction of GBM cell migration for treatment proposes. Moreover, the interstitial fluid flow (IFF) is also implicated in increasing the GBM cell migration through the activation of the CXCL12-CXCR4 signaling pathway. However, due to its complex and variable nature, the influence of the IFF on the efficiency of drug delivery systems is not well understood yet. Therefore, this review discusses novel drug delivery strategies to overcome the GBM treatment limitations, focusing on chemokines such as CXCL12 as an innovative approach to reverse the migration of infiltrated GBM. Furthermore, recent developments regarding in vitro 3D culture systems aiming to mimic the dynamic peritumoral environment for the optimization of new drug delivery technologies are highlighted.
Collapse
Affiliation(s)
- Wiam El Kheir
- Advanced Dynamic Cell Culture Systems Laboratory, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
| | - Bernard Marcos
- Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
| | - Nick Virgilio
- Department of Chemical Engineering, Polytechnique Montréal, 2500 Chemin de Polytechnique, Montréal, QC H3T 1J4, Canada;
| | - Benoit Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada;
- Clinical Research Center of the Centre Hospitalier Universitaire de l’Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Nathalie Faucheux
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Clinical Research Center of the Centre Hospitalier Universitaire de l’Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Marc-Antoine Lauzon
- Advanced Dynamic Cell Culture Systems Laboratory, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Research Center on Aging, 1036 Rue Belvédère Sud, Sherbrooke, QC J1H 4C4, Canada
| |
Collapse
|
40
|
Karan S, Cho MY, Lee H, Park HS, Han EH, Song Y, Lee Y, Kim M, Cho JH, Sessler JL, Hong KS. Hypoxia-Responsive Luminescent CEST MRI Agent for In Vitro and In Vivo Tumor Detection and Imaging. J Med Chem 2022; 65:7106-7117. [PMID: 35580357 DOI: 10.1021/acs.jmedchem.1c01745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hypoxia is a feature of most solid tumors and a key determinant of cancer growth and propagation. Sensing hypoxia effectively could lead to more favorable clinical outcomes. Here, we report a molecular antenna-based bimodal probe designed to exploit the complementary advantages of magnetic resonance (MR)- and optical-based imaging. Specifically, we describe the synthesis and evaluation of a dual-action probe (NO2-Eu) that permits hypoxia-activated chemical exchange saturation transfer (CEST) MR and optical imaging. In CT26 cells, this NO2-Eu probe not only provides an enhanced CEST MRI signal but also turns "on" the optical signal under hypoxic conditions. Time-dependent in vivo CEST imaging in a hypoxic CT26 tumor xenograft mouse model revealed probe-dependent tumor detection by CEST MRI contrast in the tumor area. We thus suggest that dual-action hypoxia probes, like that reported here, could have a role to play in solid tumor diagnosis and monitoring.
Collapse
Affiliation(s)
- Sanu Karan
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Korea
| | - Mi Young Cho
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Hyunseung Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Hye Sun Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Eun Hee Han
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Youngkyu Song
- Research Equipment Operations Division, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Youlee Lee
- Research Equipment Operations Division, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Mina Kim
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Science, London WC1N 3BG, United Kingdom
| | - Jee-Hyun Cho
- Research Equipment Operations Division, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Kwan Soo Hong
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
41
|
Hersh AM, Alomari S, Tyler BM. Crossing the Blood-Brain Barrier: Advances in Nanoparticle Technology for Drug Delivery in Neuro-Oncology. Int J Mol Sci 2022; 23:4153. [PMID: 35456971 PMCID: PMC9032478 DOI: 10.3390/ijms23084153] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 12/10/2022] Open
Abstract
The blood-brain barrier (BBB) constitutes a microvascular network responsible for excluding most drugs from the brain. Treatment of brain tumors is limited by the impermeability of the BBB and, consequently, survival outcomes for malignant brain tumors remain poor. Nanoparticles (NPs) represent a potential solution to improve drug transport to brain tumors, given their small size and capacity to target tumor cells. Here, we review the unique physical and chemical properties of NPs that aid in BBB transport and discuss mechanisms of NP transport across the BBB, including paracellular transport, carrier-mediated transport, and adsorptive- and receptor-mediated transcytosis. The major types of NPs investigated for treatment of brain tumors are detailed, including polymeric NPs, liposomes, solid lipid NPs, dendrimers, metals, quantum dots, and nanogels. In addition to their role in drug delivery, NPs can be used as imaging contrast agents and can be conjugated with imaging probes to assist in visualizing tumors, demarcating lesion boundaries and margins, and monitoring drug delivery and treatment response. Multifunctional NPs can be designed that are capable of targeting tumors for both imaging and therapeutic purposes. Finally, limitations of NPs for brain tumor treatment are discussed.
Collapse
Affiliation(s)
| | | | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (S.A.)
| |
Collapse
|
42
|
Dogra P, Ramírez JR, Butner JD, Peláez MJ, Chung C, Hooda-Nehra A, Pasqualini R, Arap W, Cristini V, Calin GA, Ozpolat B, Wang Z. Translational Modeling Identifies Synergy between Nanoparticle-Delivered miRNA-22 and Standard-of-Care Drugs in Triple-Negative Breast Cancer. Pharm Res 2022; 39:511-528. [PMID: 35294699 PMCID: PMC8986735 DOI: 10.1007/s11095-022-03176-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/21/2022] [Indexed: 12/29/2022]
Abstract
Purpose Downregulation of miRNA-22 in triple-negative breast cancer (TNBC) is associated with upregulation of eukaryotic elongation 2 factor kinase (eEF2K) protein, which regulates tumor growth, chemoresistance, and tumor immunosurveillance. Moreover, exogenous administration of miRNA-22, loaded in nanoparticles to prevent degradation and improve tumor delivery (termed miRNA-22 nanotherapy), to suppress eEF2K production has shown potential as an investigational therapeutic agent in vivo. Methods To evaluate the translational potential of miRNA-22 nanotherapy, we developed a multiscale mechanistic model, calibrated to published in vivo data and extrapolated to the human scale, to describe and quantify the pharmacokinetics and pharmacodynamics of miRNA-22 in virtual patient populations. Results Our analysis revealed the dose-response relationship, suggested optimal treatment frequency for miRNA-22 nanotherapy, and highlighted key determinants of therapy response, from which combination with immune checkpoint inhibitors was identified as a candidate strategy for improving treatment outcomes. More importantly, drug synergy was identified between miRNA-22 and standard-of-care drugs against TNBC, providing a basis for rational therapeutic combinations for improved response Conclusions The present study highlights the translational potential of miRNA-22 nanotherapy for TNBC in combination with standard-of-care drugs. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-022-03176-3.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
| | - Maria J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Anupama Hooda-Nehra
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, 07101, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, 07101, USA
- Department of Radiation Oncology, Division of Cancer Biology, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, 07101, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
- Department of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77230, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York, 10065, USA
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA.
| |
Collapse
|
43
|
Nunes D, Andrade S, Ramalho MJ, Loureiro JA, Pereira MC. Polymeric Nanoparticles-Loaded Hydrogels for Biomedical Applications: A Systematic Review on In Vivo Findings. Polymers (Basel) 2022; 14:polym14051010. [PMID: 35267833 PMCID: PMC8912535 DOI: 10.3390/polym14051010] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/18/2022] [Accepted: 03/01/2022] [Indexed: 02/07/2023] Open
Abstract
Clinically available medications face several hurdles that limit their therapeutic activity, including restricted access to the target tissues due to biological barriers, low bioavailability, and poor pharmacokinetic properties. Drug delivery systems (DDS), such as nanoparticles (NPs) and hydrogels, have been widely employed to address these issues. Furthermore, the DDS improves drugs’ therapeutic efficacy while reducing undesired side effects caused by the unspecific distribution over the different tissues. The integration of NPs into hydrogels has emerged to improve their performance when compared with each DDS individually. The combination of both DDS enhances the ability to deliver drugs in a localized and targeted manner, paired with a controlled and sustained drug release, resulting in increased drug therapeutic effectiveness. With the incorporation of the NPs into hydrogels, it is possible to apply the DDS locally and then provide a sustained release of the NPs in the site of action, allowing the drug uptake in the required location. Additionally, most of the materials used to produce the hydrogels and NPs present low toxicity. This article provides a systematic review of the polymeric NPs-loaded hydrogels developed for various biomedical applications, focusing on studies that present in vivo data.
Collapse
Affiliation(s)
- Débora Nunes
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (D.N.); (S.A.); (M.J.R.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Stéphanie Andrade
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (D.N.); (S.A.); (M.J.R.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria João Ramalho
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (D.N.); (S.A.); (M.J.R.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana A. Loureiro
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (D.N.); (S.A.); (M.J.R.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- Correspondence: (J.A.L.); (M.C.P.)
| | - Maria Carmo Pereira
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (D.N.); (S.A.); (M.J.R.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- Correspondence: (J.A.L.); (M.C.P.)
| |
Collapse
|
44
|
Quader S, Kataoka K, Cabral H. Nanomedicine for brain cancer. Adv Drug Deliv Rev 2022; 182:114115. [PMID: 35077821 DOI: 10.1016/j.addr.2022.114115] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/18/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
CNS tumors remain among the deadliest forms of cancer, resisting conventional and new treatment approaches, with mortality rates staying practically unchanged over the past 30 years. One of the primary hurdles for treating these cancers is delivering drugs to the brain tumor site in therapeutic concentration, evading the blood-brain (tumor) barrier (BBB/BBTB). Supramolecular nanomedicines (NMs) are increasingly demonstrating noteworthy prospects for addressing these challenges utilizing their unique characteristics, such as improving the bioavailability of the payloadsviacontrolled pharmacokinetics and pharmacodynamics, BBB/BBTB crossing functions, superior distribution in the brain tumor site, and tumor-specific drug activation profiles. Here, we review NM-based brain tumor targeting approaches to demonstrate their applicability and translation potential from different perspectives. To this end, we provide a general overview of brain tumor and their treatments, the incidence of the BBB and BBTB, and their role on NM targeting, as well as the potential of NMs for promoting superior therapeutic effects. Additionally, we discuss critical issues of NMs and their clinical trials, aiming to bolster the potential clinical applications of NMs in treating these life-threatening diseases.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
45
|
Ruiz-Garcia H, Ramirez-Loera C, Malouff TD, Seneviratne DS, Palmer JD, Trifiletti DM. Novel Strategies for Nanoparticle-Based Radiosensitization in Glioblastoma. Int J Mol Sci 2021; 22:9673. [PMID: 34575840 PMCID: PMC8465220 DOI: 10.3390/ijms22189673] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/09/2023] Open
Abstract
Radiotherapy (RT) is one of the cornerstones in the current treatment paradigm for glioblastoma (GBM). However, little has changed in the management of GBM since the establishment of the current protocol in 2005, and the prognosis remains grim. Radioresistance is one of the hallmarks for treatment failure, and different therapeutic strategies are aimed at overcoming it. Among these strategies, nanomedicine has advantages over conventional tumor therapeutics, including improvements in drug delivery and enhanced antitumor properties. Radiosensitizing strategies using nanoparticles (NP) are actively under study and hold promise to improve the treatment response. We aim to describe the basis of nanomedicine for GBM treatment, current evidence in radiosensitization efforts using nanoparticles, and novel strategies, such as preoperative radiation, that could be synergized with nanoradiosensitizers.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| | | | - Timothy D. Malouff
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Danushka S. Seneviratne
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Joshua D. Palmer
- Department of Radiation Oncology, Ohio State University, Columbus, OH 43210, USA;
| | - Daniel M. Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
46
|
Janjua TI, Rewatkar P, Ahmed-Cox A, Saeed I, Mansfeld FM, Kulshreshtha R, Kumeria T, Ziegler DS, Kavallaris M, Mazzieri R, Popat A. Frontiers in the treatment of glioblastoma: Past, present and emerging. Adv Drug Deliv Rev 2021; 171:108-138. [PMID: 33486006 DOI: 10.1016/j.addr.2021.01.012] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/13/2020] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is one of the most aggressive cancers of the brain. Despite extensive research over the last several decades, the survival rates for GBM have not improved and prognosis remains poor. To date, only a few therapies are approved for the treatment of GBM with the main reasons being: 1) significant tumour heterogeneity which promotes the selection of resistant subpopulations 2) GBM induced immunosuppression and 3) fortified location of the tumour in the brain which hinders the delivery of therapeutics. Existing therapies for GBM such as radiotherapy, surgery and chemotherapy have been unable to reach the clinical efficacy necessary to prolong patient survival more than a few months. This comprehensive review evaluates the current and emerging therapies including those in clinical trials that may potentially improve both targeted delivery of therapeutics directly to the tumour site and the development of agents that may specifically target GBM. Particular focus has also been given to emerging delivery technologies such as focused ultrasound, cellular delivery systems nanomedicines and immunotherapy. Finally, we discuss the importance of developing novel materials for improved delivery efficacy of nanoparticles and therapeutics to reduce the suffering of GBM patients.
Collapse
|
47
|
Colucci F, Mancini V, Mattu C, Boffito M. Designing Multifunctional Devices for Regenerative Pharmacology Based on 3D Scaffolds, Drug-Loaded Nanoparticles, and Thermosensitive Hydrogels: A Proof-of-Concept Study. Pharmaceutics 2021; 13:pharmaceutics13040464. [PMID: 33808138 PMCID: PMC8066789 DOI: 10.3390/pharmaceutics13040464] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/25/2022] Open
Abstract
Regenerative pharmacology combines tissue engineering/regenerative medicine (TERM) with drug delivery with the aim to improve the outcomes of traditional TERM approaches. In this work, we aimed to design a multicomponent TERM platform comprising a three-dimensional scaffold, a thermosensitive hydrogel, and drug-loaded nanoparticles. We used a thermally induced phase separation method to obtain scaffolds with anisotropic mechanical properties, suitable for soft tissue engineering. A thermosensitive hydrogel was developed using a Poloxamer® 407-based poly(urethane) to embed curcumin-loaded nanoparticles, obtained by the single emulsion nanoprecipitation method. We found that encapsulated curcumin could retain its antioxidant activity and that embedding nanoparticles within the hydrogel did not affect the hydrogel gelation kinetics nor the possibility to progressively release the drug. The porous scaffold was easily loaded with the hydrogel, resulting in significantly enhanced (4-fold higher) absorption of a model molecule of nutrients (fluorescein isothiocyanate dextran 4kDa) from the surrounding environment compared to pristine scaffold. The developed platform could thus represent a valuable alternative in the treatment of many pathologies affecting soft tissues, by concurrently exploiting the therapeutic effects of drugs, with the 3D framework acting as a physical support for tissue regeneration and the cell-friendly environment represented by the hydrogel.
Collapse
Affiliation(s)
- Francesco Colucci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy; (F.C.); (V.M.)
| | - Vanessa Mancini
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy; (F.C.); (V.M.)
- Department of Anatomy & Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy; (F.C.); (V.M.)
- PolitoBIOMed Laboratory, Politecnico di Torino, 10129 Turin, Italy
- Correspondence: (C.M.); (M.B.)
| | - Monica Boffito
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy; (F.C.); (V.M.)
- PolitoBIOMed Laboratory, Politecnico di Torino, 10129 Turin, Italy
- Institute for Chemical-Physical Processes, National Research Council (CNR-IPCF), 56124 Pisa, Italy
- Correspondence: (C.M.); (M.B.)
| |
Collapse
|
48
|
Anaya DA, Dogra P, Wang Z, Haider M, Ehab J, Jeong DK, Ghayouri M, Lauwers GY, Thomas K, Kim R, Butner JD, Nizzero S, Ramírez JR, Plodinec M, Sidman RL, Cavenee WK, Pasqualini R, Arap W, Fleming JB, Cristini V. A Mathematical Model to Estimate Chemotherapy Concentration at the Tumor-Site and Predict Therapy Response in Colorectal Cancer Patients with Liver Metastases. Cancers (Basel) 2021; 13:cancers13030444. [PMID: 33503971 PMCID: PMC7866038 DOI: 10.3390/cancers13030444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary It is known that drug transport barriers in the tumor determine drug concentration at the tumor site, causing disparity from the systemic (plasma) drug concentration. However, current clinical standard of care still bases dosage and treatment optimization on the systemic concentration of drugs. Here, we present a proof of concept observational cohort study to accurately estimate drug concentration at the tumor site from mathematical modeling using biologic, clinical, and imaging/perfusion data, and correlate it with outcome in colorectal cancer liver metastases. We demonstrate that drug concentration at the tumor site, not in systemic circulation, can be used as a credible biomarker for predicting chemotherapy outcome, and thus our mathematical modeling approach can be applied prospectively in the clinic to personalize treatment design to optimize outcome. Abstract Chemotherapy remains a primary treatment for metastatic cancer, with tumor response being the benchmark outcome marker. However, therapeutic response in cancer is unpredictable due to heterogeneity in drug delivery from systemic circulation to solid tumors. In this proof-of-concept study, we evaluated chemotherapy concentration at the tumor-site and its association with therapy response by applying a mathematical model. By using pre-treatment imaging, clinical and biologic variables, and chemotherapy regimen to inform the model, we estimated tumor-site chemotherapy concentration in patients with colorectal cancer liver metastases, who received treatment prior to surgical hepatic resection with curative-intent. The differential response to therapy in resected specimens, measured with the gold-standard Tumor Regression Grade (TRG; from 1, complete response to 5, no response) was examined, relative to the model predicted systemic and tumor-site chemotherapy concentrations. We found that the average calculated plasma concentration of the cytotoxic drug was essentially equivalent across patients exhibiting different TRGs, while the estimated tumor-site chemotherapeutic concentration (eTSCC) showed a quadratic decline from TRG = 1 to TRG = 5 (p < 0.001). The eTSCC was significantly lower than the observed plasma concentration and dropped by a factor of ~5 between patients with complete response (TRG = 1) and those with no response (TRG = 5), while the plasma concentration remained stable across TRG groups. TRG variations were driven and predicted by differences in tumor perfusion and eTSCC. If confirmed in carefully planned prospective studies, these findings will form the basis of a paradigm shift in the care of patients with potentially curable colorectal cancer and liver metastases.
Collapse
Affiliation(s)
- Daniel A. Anaya
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
- Correspondence: (D.A.A.); (V.C.); Tel.: +1-813-745-1432 (D.A.A.); +1-505-934-1813 (V.C.); Fax: +1-813-745-7229 (D.A.A.)
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Mintallah Haider
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Jasmina Ehab
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Daniel K. Jeong
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Masoumeh Ghayouri
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Gregory Y. Lauwers
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Kerry Thomas
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Richard Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute & ARTIDIS AG, University of Basel, 4056 Basel, Switzerland;
| | - Richard L. Sidman
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA;
| | - Webster K. Cavenee
- Ludwig Institute for Cancer Research, University of California-San Diego, La Jolla, CA 92093, USA;
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey & Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey & Division of Hematology/Oncology, Department of Medicine Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
| | - Jason B. Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
- Correspondence: (D.A.A.); (V.C.); Tel.: +1-813-745-1432 (D.A.A.); +1-505-934-1813 (V.C.); Fax: +1-813-745-7229 (D.A.A.)
| |
Collapse
|