1
|
Proshkina GM, Shramova EI, Mirkasyimov AB, Griaznova OY, Konovalova EV, Schulga AA, Deyev SM. The Barnase-Barstar-based pre-targeting strategy for enhanced antitumor therapy in vivo. Biochimie 2025; 228:158-166. [PMID: 39307408 DOI: 10.1016/j.biochi.2024.09.011] [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: 08/08/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
There is a great need for novel approaches to the treatment of epithelial ovarian carcinoma, which is the leading cause of mortality from gynecological malignancies. In this study, the pre-targeting technology was used to enhance the in vivo targeting of cytotoxic module composed of nanoliposomes loaded with a truncated form of Pseudomonas aeruginosa exotoxin A (PE40) to cancer cells. Pre-targeting system used in this study is composed of bacterial ribonuclease Barnase and its natural antitoxin Barstar. Barstar, genetically fused to various engineered scaffold proteins specific to tumor-associated antigens (HER2, EpCAM) serves as a primary module for precise cancer cell recognition. Barnase conjugated to a therapeutic agent serves as a cytotoxic or secondary module for malignant cell elimination. Due to strong non-covalent interaction (KD10-14 M) of Barstar and Barnase, the primary and secondary modules efficiently interact with each other on the cell surface, which has been proven by confocal microscopy and flow cytometry. Using mice with SKOV-3 ovarian cancer xenografts, we have shown that regardless of the targeting module, the pre-targeting approach is much more effective than a single-step active targeting.
Collapse
Affiliation(s)
- G M Proshkina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow, 117997, Russia.
| | - E I Shramova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow, 117997, Russia
| | - A B Mirkasyimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow, 117997, Russia; Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - O Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow, 117997, Russia
| | - E V Konovalova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow, 117997, Russia
| | - A A Schulga
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow, 117997, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow, 117997, Russia; National Research Center "Kurchatov Institute", Moscow, 123182, Russia; Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| |
Collapse
|
2
|
Rana D, Rangel VR, Padmanaban P, Trikalitis VD, Kandar A, Kim HW, Rouwkema J. Bioprinting of Aptamer-Based Programmable Bioinks to Modulate Multiscale Microvascular Morphogenesis in 4D. Adv Healthc Mater 2025; 14:e2402302. [PMID: 39487611 DOI: 10.1002/adhm.202402302] [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: 06/24/2024] [Revised: 10/17/2024] [Indexed: 11/04/2024]
Abstract
Dynamic growth factor presentation influences how individual endothelial cells assemble into complex vascular networks. Here, programmable bioinks are developed that facilitate dynamic vascular endothelial growth factor (VEGF) presentation to guide vascular morphogenesis within 3D-bioprinted constructs. Aptamer's high affinity is leveraged for rapid VEGF sequestration in spatially confined regions and utilized aptamer-complementary sequence (CS) hybridization to tune VEGF release kinetics temporally, days after bioprinting. It is shown that spatial resolution of programmable bioink, combined with CS-triggered VEGF release, significantly influences the alignment, organization, and morphogenesis of microvascular networks in bioprinted constructs. The presence of aptamer-tethered VEGF and the generation of instantaneous VEGF gradients upon CS-triggering restricted hierarchical network formation to the printed aptamer regions at all spatial resolutions. Network properties improved as the spatial resolution decreased, with low-resolution designs yielding the highest network properties. Specifically, CS-treated low-resolution designs exhibited significant vascular network remodeling, with an increase in vessel density(1.35-fold), branching density(1.54-fold), and average vessel length(2.19-fold) compared to non-treated samples. The results suggest that CS acts as an external trigger capable of inducing time-controlled changes in network organization and alignment on-demand within spatially localized regions of a bioprinted construct. It is envisioned that these programmable bioinks will open new opportunities for bioengineering functional, hierarchically self-organized vascular networks within engineered tissues.
Collapse
Affiliation(s)
- Deepti Rana
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, 7522NB, The Netherlands
| | - Vincent R Rangel
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, 7522NB, The Netherlands
| | - Prasanna Padmanaban
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, 7522NB, The Netherlands
| | - Vasileios D Trikalitis
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, 7522NB, The Netherlands
| | - Ajoy Kandar
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, 7522NB, The Netherlands
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, 7522NB, The Netherlands
| |
Collapse
|
3
|
Kandell RM, Wu JR, Kwon EJ. Reprograming Clots for In Vivo Chemical Targeting in Traumatic Brain Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301738. [PMID: 38780012 PMCID: PMC11293973 DOI: 10.1002/adma.202301738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2024] [Indexed: 05/25/2024]
Abstract
Traumatic brain injury (TBI) is a critical public health concern, yet there are no therapeutics available to improve long-term outcomes. Drug delivery to TBI remains a challenge due to the blood-brain barrier and increased intracranial pressure. In this work, a chemical targeting approach to improve delivery of materials to the injured brain, is developed. It is hypothesized that the provisional fibrin matrix can be harnessed as an injury-specific scaffold that can be targeted by materials via click chemistry. To accomplish this, the brain clot is engineered in situ by delivering fibrinogen modified with strained cyclooctyne (SCO) moieties, which incorporated into the injury lesion and is retained there for days. Improved intra-injury capture and retention of diverse, clickable azide-materials including a small molecule azide-dye, 40 kDa azide-PEG nanomaterial, and a therapeutic azide-protein in multiple dosing regimens is subsequently observed. To demonstrate therapeutic translation of this approach, a reduction in reactive oxygen species levels in the injured brain after delivery of the antioxidant catalase, is achieved. Further, colocalization between azide and SCO-fibrinogen is specific to the brain over off-target organs. Taken together, a chemical targeting strategy leveraging endogenous clot formation is established which can be applied to improve therapeutic delivery after TBI.
Collapse
Affiliation(s)
- Rebecca M. Kandell
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Jason R. Wu
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Ester J. Kwon
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
4
|
Singhal R, Sarangi MK, Rath G. Injectable Hydrogels: A Paradigm Tailored with Design, Characterization, and Multifaceted Approaches. Macromol Biosci 2024; 24:e2400049. [PMID: 38577905 DOI: 10.1002/mabi.202400049] [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: 02/05/2024] [Revised: 03/22/2024] [Indexed: 04/06/2024]
Abstract
Biomaterials denoting self-healing and versatile structural integrity are highly curious in the biomedicine segment. The injectable and/or printable 3D printing technology is explored in a few decades back, which can alter their dimensions temporarily under shear stress, showing potential healing/recovery tendency with patient-specific intervention toward the development of personalized medicine. Thus, self-healing injectable hydrogels (IHs) are stunning toward developing a paradigm for tissue regeneration. This review comprises the designing of IHs, rheological characterization and stability, several benchmark consequences for self-healing IHs, their translation into tissue regeneration of specific types, applications of IHs in biomedical such as anticancer and immunomodulation, wound healing and tissue/bone regeneration, antimicrobial potentials, drugs, gene and vaccine delivery, ocular delivery, 3D printing, cosmeceuticals, and photothermal therapy as well as in other allied avenues like agriculture, aerospace, electronic/electrical industries, coating approaches, patents associated with therapeutic/nontherapeutic avenues, and numerous futuristic challenges and solutions.
Collapse
Affiliation(s)
- Rishika Singhal
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Manoj Kumar Sarangi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, 751030, India
| |
Collapse
|
5
|
Gharios R, Francis RM, DeForest CA. Chemical and Biological Engineering Strategies to Make and Modify Next-Generation Hydrogel Biomaterials. MATTER 2023; 6:4195-4244. [PMID: 38313360 PMCID: PMC10836217 DOI: 10.1016/j.matt.2023.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
There is a growing interest in the development of technologies to probe and direct in vitro cellular function for fundamental organoid and stem cell biology, functional tissue and metabolic engineering, and biotherapeutic formulation. Recapitulating many critical aspects of the native cellular niche, hydrogel biomaterials have proven to be a defining platform technology in this space, catapulting biological investigation from traditional two-dimensional (2D) culture into the 3D world. Seeking to better emulate the dynamic heterogeneity characteristic of all living tissues, global efforts over the last several years have centered around upgrading hydrogel design from relatively simple and static architectures into stimuli-responsive and spatiotemporally evolvable niches. Towards this end, advances from traditionally disparate fields including bioorthogonal click chemistry, chemoenzymatic synthesis, and DNA nanotechnology have been co-opted and integrated to construct 4D-tunable systems that undergo preprogrammed functional changes in response to user-defined inputs. In this Review, we highlight how advances in synthetic, semisynthetic, and bio-based chemistries have played a critical role in the triggered creation and customization of next-generation hydrogel biomaterials. We also chart how these advances stand to energize the translational pipeline of hydrogels from bench to market and close with an outlook on outstanding opportunities and challenges that lay ahead.
Collapse
Affiliation(s)
- Ryan Gharios
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
| | - Ryan M. Francis
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
| | - Cole A. DeForest
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
- Department of Bioengineering, University of Washington, Seattle WA 98105, USA
- Department of Chemistry, University of Washington, Seattle WA 98105, USA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle WA 98109, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle WA 98105, USA
| |
Collapse
|
6
|
Rochet LNC, Bahou C, Wojciechowski JP, Koutsopetras I, Britton P, Spears RJ, Thanasi IA, Shao B, Zhong L, Bučar DK, Aliev AE, Porter MJ, Stevens MM, Baker JR, Chudasama V. Use of pyridazinediones for tuneable and reversible covalent cysteine modification applied to peptides, proteins and hydrogels. Chem Sci 2023; 14:13743-13754. [PMID: 38075666 PMCID: PMC10699563 DOI: 10.1039/d3sc04976k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/27/2023] [Indexed: 05/02/2024] Open
Abstract
Reversible cysteine modification has been found to be a useful tool for a plethora of applications such as selective enzymatic inhibition, activity-based protein profiling and/or cargo release from a protein or a material. However, only a limited number of reagents display reliable dynamic/reversible thiol modification and, in most cases, many of these reagents suffer from issues of stability, a lack of modularity and/or poor rate tunability. In this work, we demonstrate the potential of pyridazinediones as novel reversible and tuneable covalent cysteine modifiers. We show that the electrophilicity of pyridazinediones correlates to the rates of the Michael addition and retro-Michael deconjugation reactions, demonstrating that pyridazinediones provide an enticing platform for readily tuneable and reversible thiol addition/release. We explore the regioselectivity of the novel reaction and unveil the reason for the fundamental increased reactivity of aryl bearing pyridazinediones by using DFT calculations and corroborating findings with SCXRD. We also applied this fundamental discovery to making more rapid disulfide rebridging agents in related work. We finally provide the groundwork for potential applications in various areas with exemplification using readily functionalised "clickable" pyridazinediones on clinically relevant cysteine and disulfide conjugated proteins, as well as on a hydrogel material.
Collapse
Affiliation(s)
- Léa N C Rochet
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Calise Bahou
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Jonathan P Wojciechowski
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Ilias Koutsopetras
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Phyllida Britton
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Richard J Spears
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Baihao Shao
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Lisha Zhong
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Dejan-Krešimir Bučar
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Abil E Aliev
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Michael J Porter
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
7
|
Mor N, Raghav N. In-vitro simulation of modified-alginate ester as sustained release delivery system for curcumin. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
8
|
Ciciriello AJ, Surnar B, Medy GD, Su X, Dhar S, Dumont CM. Biomaterial-targeted precision nanoparticle delivery to the injured spinal cord. Acta Biomater 2022; 152:532-545. [PMID: 36087868 PMCID: PMC10551882 DOI: 10.1016/j.actbio.2022.08.077] [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: 05/04/2022] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 11/01/2022]
Abstract
Drug delivery requires precision in timing, location, and dosage to achieve therapeutic benefits. Challenges in addressing all three of these critical criteria result in poor temporal dexterity, widespread accumulation and off-target effects, and high doses with the potential for toxicity. To address these challenges, we have developed the BiomatErial Accumulating Carriers for On-demand Nanotherapy (BEACON) platform that utilizes an implantable biomaterial to serve as a target for systemically delivered nanoparticles (NPs). With the BEACON system, administered NPs are conjugated with a ligand that has high affinity for a receptor in the implanted biomaterial. To test BEACON, an in vivo spinal cord injury (SCI) model was used as it provides an injury model where the three identified criteria can be tested as it is a dynamic and complicated injury model with no currently approved therapies. Through our work, we have demonstrated temporal dexterity in NP administration by injecting 6 days post-SCI, decreased off-target accumulation with a significant drop in liver accumulation, and retention of our NPs in the target biomaterial. The BEACON system can be applied broadly, beyond the nervous system, to improve systemically delivered NP accumulation at an implanted biomaterial target. STATEMENT OF SIGNIFICANCE: Targeted drug delivery approaches have the potential to improve therapeutic regimens for patients on a case-by-case basis. Improved localization of a therapeutic to site of interest can result in increased efficacy and limit the need for repeat dosing. Unfortunately, targeted strategies can fall short when receptors on cells or tissues are too widespread or change over the course of disease or injury progression. The BEACON system developed herein eliminates the need to target a cell or tissue receptor by targeting an implantable biomaterial with location-controllable accumulation and sustained presentation over time. The targeting paradigm presented by BEACON is widely applicable throughout tissue engineering and regenerative medicine without the need to retool for each new application.
Collapse
Affiliation(s)
- Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States
| | - Bapurao Surnar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Giovanni D Medy
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Xiaoyu Su
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Shanta Dhar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami, 1475 NW 12th Avenue, Miami, Florida 33136, United States
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States.
| |
Collapse
|
9
|
Abstract
Biomaterials with the ability to self-heal and recover their structural integrity offer many advantages for applications in biomedicine. The past decade has witnessed the rapid emergence of a new class of self-healing biomaterials commonly termed injectable, or printable in the context of 3D printing. These self-healing injectable biomaterials, mostly hydrogels and other soft condensed matter based on reversible chemistry, are able to temporarily fluidize under shear stress and subsequently recover their original mechanical properties. Self-healing injectable hydrogels offer distinct advantages compared to traditional biomaterials. Most notably, they can be administered in a locally targeted and minimally invasive manner through a narrow syringe without the need for invasive surgery. Their moldability allows for a patient-specific intervention and shows great prospects for personalized medicine. Injected hydrogels can facilitate tissue regeneration in multiple ways owing to their viscoelastic and diffusive nature, ranging from simple mechanical support, spatiotemporally controlled delivery of cells or therapeutics, to local recruitment and modulation of host cells to promote tissue regeneration. Consequently, self-healing injectable hydrogels have been at the forefront of many cutting-edge tissue regeneration strategies. This study provides a critical review of the current state of self-healing injectable hydrogels for tissue regeneration. As key challenges toward further maturation of this exciting research field, we identify (i) the trade-off between the self-healing and injectability of hydrogels vs their physical stability, (ii) the lack of consensus on rheological characterization and quantitative benchmarks for self-healing injectable hydrogels, particularly regarding the capillary flow in syringes, and (iii) practical limitations regarding translation toward therapeutically effective formulations for regeneration of specific tissues. Hence, here we (i) review chemical and physical design strategies for self-healing injectable hydrogels, (ii) provide a practical guide for their rheological analysis, and (iii) showcase their applicability for regeneration of various tissues and 3D printing of complex tissues and organoids.
Collapse
Affiliation(s)
- Pascal Bertsch
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands
| | - Mani Diba
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - David J. Mooney
- John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - Sander C. G. Leeuwenburgh
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,
| |
Collapse
|
10
|
Rana D, Kandar A, Salehi-Nik N, Inci I, Koopman B, Rouwkema J. Spatiotemporally controlled, aptamers-mediated growth factor release locally manipulates microvasculature formation within engineered tissues. Bioact Mater 2022; 12:71-84. [PMID: 35087964 PMCID: PMC8777207 DOI: 10.1016/j.bioactmat.2021.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- Deepti Rana
- Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500, AE, Enschede, the Netherlands
| | - Ajoy Kandar
- Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500, AE, Enschede, the Netherlands
| | - Nasim Salehi-Nik
- Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500, AE, Enschede, the Netherlands
| | - Ilyas Inci
- Izmir Democracy University, Vocational School of Health Services, Department of Dentistry Services, Dental Prosthetics Technology, Izmir, 35140, Turkey
| | - Bart Koopman
- Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500, AE, Enschede, the Netherlands
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500, AE, Enschede, the Netherlands
- Corresponding author.
| |
Collapse
|
11
|
Lin ZP, Nguyen LNM, Ouyang B, MacMillan P, Ngai J, Kingston BR, Mladjenovic SM, Chan WCW. Macrophages Actively Transport Nanoparticles in Tumors After Extravasation. ACS NANO 2022; 16:6080-6092. [PMID: 35412309 DOI: 10.1021/acsnano.1c11578] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanoparticles need to navigate a complex microenvironment to target cells in solid tumors after extravasation. Diffusion is currently the accepted primary mechanism for nanoparticle distribution in tumors. However, the extracellular matrix can limit nanoparticle diffusion. Here, we identified tumor-associated macrophages as another key player in transporting and redistributing nanoparticles in the tumor microenvironment. We found tumor-associated macrophages actively migrate toward nanoparticles extravasated from the vessels, engulfing and redistributing them in the tumor stroma. The macrophages can carry the nanoparticles 2-5 times deeper in the tumor than passive diffusion. The amount of nanoparticles transported by the tumor-associated macrophages is size-dependent. Understanding the nanoparticle behavior after extravasation will provide strategies to engineer them to navigate the microenvironment for improved intratumoral targeting and therapeutic effectiveness.
Collapse
Affiliation(s)
- Zachary Pengju Lin
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Luan N M Nguyen
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Ben Ouyang
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- M.D./PhD Program, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Presley MacMillan
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Jessica Ngai
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Benjamin R Kingston
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97201, United States
| | - Stefan M Mladjenovic
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Department of Material Science and Engineering, University of Toronto, 184 College Street, Toronto, Ontario M5S 3E4, Canada
| |
Collapse
|
12
|
Abstract
Stimuli-responsive, on-demand release of drugs from drug-eluting depots could transform the treatment of many local diseases, providing intricate control over local dosing. However, conventional on-demand drug release approaches rely on locally implanted drug depots, which become spent over time and cannot be refilled or reused without invasive procedures. New strategies to noninvasively refill drug-eluting depots followed by on-demand release could transform clinical therapy. Here we report an on-demand drug delivery paradigm that combines bioorthogonal click chemistry to locally enrich protodrugs at a prelabeled site and light-triggered drug release at the target tissue. This approach begins with introduction of the targetable depot through local injection of chemically reactive azide groups that anchor to the extracellular matrix. The anchored azide groups then capture blood-circulating protodrugs through bioorthogonal click chemistry. After local capture and retention, active drugs can be released through external light irradiation. In this report, a photoresponsive protodrug was constructed consisting of the chemotherapeutic doxorubicin (Dox), conjugated to dibenzocyclooctyne (DBCO) through a photocleavable ortho-nitrobenzyl linker. The protodrug exhibited excellent on-demand light-triggered Dox release properties and light-mediated in vitro cytotoxicity in U87 glioblastoma cell lines. Furthermore, in a live animal setting, azide depots formed in mice through intradermal injection of activated azide-NHS esters. After i.v. administration, the protodrug was captured by the azide depots with intricate local specificity, which could be increased with multiple refills. Finally, doxorubicin could be released from the depot upon light irradiation. Multiple rounds of depot refilling and light-mediated release of active drug were accomplished, indicating that this system has the potential for multiple rounds of treatment. Taken together, these in vitro and in vivo proof of concept studies establish a novel method for in vivo targeting and on-demand delivery of cytotoxic drugs at target tissues.
Collapse
Affiliation(s)
- Sandeep Palvai
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Christopher T Moody
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Sharda Pandit
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
13
|
Abstract
The fields of precision imaging and drug delivery have revealed a number of tools to improve target specificity and increase efficacy in diagnosing and treating disease. Biological molecules, such as antibodies, continue to be the primary means of assuring active targeting of various payloads. However, molecular-scale recognition motifs have emerged in recent decades to achieve specificity through the design of interacting chemical motifs. In this regard, an assortment of bioorthogonal covalent conjugations offer possibilities for in situ complexation under physiological conditions. Herein, a related concept is discussed that leverages interactions from noncovalent or supramolecular motifs to facilitate in situ recognition and complex formation in the body. Classic supramolecular motifs based on host-guest complexation offer one such means of facilitating recognition. In addition, synthetic bioinspired motifs based on oligonucleotide hybridization and coiled-coil peptide bundles afford other routes to form complexes in situ. The architectures to include recognition of these various motifs for targeting enable both monovalent and multivalent presentation, seeking high affinity or engineered avidity to facilitate conjugation even under dilute conditions of the body. Accordingly, supramolecular "click chemistry" offers a complementary tool in the growing arsenal targeting improved healthcare efficacy.
Collapse
Affiliation(s)
| | | | - Matthew J. Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| |
Collapse
|
14
|
Zhu D, Hou J, Qian M, Jin D, Hao T, Pan Y, Wang H, Wu S, Liu S, Wang F, Wu L, Zhong Y, Yang Z, Che Y, Shen J, Kong D, Yin M, Zhao Q. Nitrate-functionalized patch confers cardioprotection and improves heart repair after myocardial infarction via local nitric oxide delivery. Nat Commun 2021; 12:4501. [PMID: 34301958 PMCID: PMC8302626 DOI: 10.1038/s41467-021-24804-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a short-lived signaling molecule that plays a pivotal role in cardiovascular system. Organic nitrates represent a class of NO-donating drugs for treating coronary artery diseases, acting through the vasodilation of systemic vasculature that often leads to adverse effects. Herein, we design a nitrate-functionalized patch, wherein the nitrate pharmacological functional groups are covalently bound to biodegradable polymers, thus transforming small-molecule drugs into therapeutic biomaterials. When implanted onto the myocardium, the patch releases NO locally through a stepwise biotransformation, and NO generation is remarkably enhanced in infarcted myocardium because of the ischemic microenvironment, which gives rise to mitochondrial-targeted cardioprotection as well as enhanced cardiac repair. The therapeutic efficacy is further confirmed in a clinically relevant porcine model of myocardial infarction. All these results support the translational potential of this functional patch for treating ischemic heart disease by therapeutic mechanisms different from conventional organic nitrate drugs.
Collapse
Affiliation(s)
- Dashuai Zhu
- State key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Jingli Hou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Meng Qian
- State key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Dawei Jin
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tian Hao
- State key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Yanjun Pan
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - He Wang
- State key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Shuting Wu
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuo Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Fei Wang
- State key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Lanping Wu
- Department of Cardiac Ultrasound, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yumin Zhong
- Diagnostic Imaging Center, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhilu Yang
- Key Laboratory of Advanced Technology for Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Yongzhe Che
- School of Medicine, Nankai University, Tianjin, China
| | - Jie Shen
- College of Pharmacy, Nankai University, Tianjin, China
| | - Deling Kong
- State key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Meng Yin
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qiang Zhao
- State key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China.
- Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, Henan Province, China.
| |
Collapse
|
15
|
Deng Y, Shavandi A, Okoro OV, Nie L. Alginate modification via click chemistry for biomedical applications. Carbohydr Polym 2021; 270:118360. [PMID: 34364605 DOI: 10.1016/j.carbpol.2021.118360] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/28/2022]
Abstract
Alginate biopolymers are characterized by favorable properties, of biocompatibility, degradability, and non-toxicity. However, the poor stability properties of alginate have limited its suitability for diverse applications. Recently, click chemistry has generated significant research interest due to its high reaction efficiency, high selectivity for a single product, harmless byproducts, and processing simplicity. Alginate modified using click chemistry enables the production of alginate derivatives with enhanced physical and chemical properties. Herein, we review the employment of click chemistry in the development of alginate-based materials or systems. Various click chemistries were highlighted, including azide and alkyne cycloaddition (e.g. Copper-(I)-catalyzed azide-alkyne cycloaddition (CuAAC), Strain-promoted alkyne-azide cycloaddition (SPAAC)), Diels-Alder reaction (Inverse electron demand Diels-Alder (IEDDA) cycloaddition, Tetrazine-norbornene Diels-Alder reactions), Thiol-ene/yne addition (Free-radical thiol-ene addition click reactions, Thiol-Michael addition click reactions, Thiol-yne addition click reaction), Oxime based click reactions, and other click reactions. Alginate functionalized with click chemistry and its properties were also discussed. The present study shows that click chemistry may be employed in modifying the mechanical strength, biochemical/biological properties of alginate-based materials. Finally, the applications of alginate-based materials in wound dressing, drug delivery, protein delivery, tissue regeneration, and 3D bioprinting were described and the future perspectives of alginates modified with click chemistry, are subsequently presented. This review provides new insights for readers to design structures and expand applications of alginate using click chemistry reactions in a detailed and more rational manner.
Collapse
Affiliation(s)
- Yaling Deng
- College of Intelligent Science and Control Engineering, Jinling Institute of Technology, Nanjing 211169, China
| | - Amin Shavandi
- BioMatter unit - 3BIO - École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium.
| | - Oseweuba Valentine Okoro
- BioMatter unit - 3BIO - École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Lei Nie
- College of Life Sciences, Xinyang Normal University, Xinyang 464000, China.
| |
Collapse
|
16
|
Scinto SL, Bilodeau DA, Hincapie R, Lee W, Nguyen SS, Xu M, am Ende CW, Finn MG, Lang K, Lin Q, Pezacki JP, Prescher JA, Robillard MS, Fox JM. Bioorthogonal chemistry. NATURE REVIEWS. METHODS PRIMERS 2021; 1:30. [PMID: 34585143 PMCID: PMC8469592 DOI: 10.1038/s43586-021-00028-z] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/05/2021] [Indexed: 12/11/2022]
Abstract
Bioorthogonal chemistry represents a class of high-yielding chemical reactions that proceed rapidly and selectively in biological environments without side reactions towards endogenous functional groups. Rooted in the principles of physical organic chemistry, bioorthogonal reactions are intrinsically selective transformations not commonly found in biology. Key reactions include native chemical ligation and the Staudinger ligation, copper-catalysed azide-alkyne cycloaddition, strain-promoted [3 + 2] reactions, tetrazine ligation, metal-catalysed coupling reactions, oxime and hydrazone ligations as well as photoinducible bioorthogonal reactions. Bioorthogonal chemistry has significant overlap with the broader field of 'click chemistry' - high-yielding reactions that are wide in scope and simple to perform, as recently exemplified by sulfuryl fluoride exchange chemistry. The underlying mechanisms of these transformations and their optimal conditions are described in this Primer, followed by discussion of how bioorthogonal chemistry has become essential to the fields of biomedical imaging, medicinal chemistry, protein synthesis, polymer science, materials science and surface science. The applications of bioorthogonal chemistry are diverse and include genetic code expansion and metabolic engineering, drug target identification, antibody-drug conjugation and drug delivery. This Primer describes standards for reproducibility and data deposition, outlines how current limitations are driving new research directions and discusses new opportunities for applying bioorthogonal chemistry to emerging problems in biology and biomedicine.
Collapse
Affiliation(s)
- Samuel L. Scinto
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Didier A. Bilodeau
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Robert Hincapie
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Wankyu Lee
- Pfizer Worldwide Research and Development, Cambridge, MA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Sean S. Nguyen
- Department of Chemistry, University of California, Irvine, CA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Minghao Xu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | | | - M. G. Finn
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kathrin Lang
- Department of Chemistry, Technical University of Munich, Garching, Germany
- Laboratory of Organic Chemistry, ETH Zurich, Zurich, Switzerland
| | - Qing Lin
- Department of Chemistry, State University of New York at Buffalo, Buffalo, NY, USA
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Jennifer A. Prescher
- Department of Chemistry, University of California, Irvine, CA, USA
- Molecular Biology & Biochemistry, University of California, Irvine, CA, USA
| | | | - Joseph M. Fox
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| |
Collapse
|
17
|
Maslarska V, Bozhanov S, Ivanova S, Angelova VT. Development and Validation of a Liquid Chromatographic Method for Aroylhydrazones at Hydrolytic Conditions. CURR PHARM ANAL 2021. [DOI: 10.2174/1573412916666191231094046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
The indole-containing aroylhydrazone derivatives 3a-c with potent antimycobacterial
activity against a referent strain M. tuberculosis H37Rv and low cytotoxicity were evaluated
for their stability via the precise and accurate HPLC analytical method in aqueous media of different
pH (2.0, 7.0, 9.0 and 12.0).
Objective:
The study describes the development and validation of a simple and reliable HPLC-UV procedure
for the determination of aroylhydrazone derivatives and their hydrolytic stability. Additionally,
to recognize if hydrolysis leads to generating undesired products, the degradation processes were identified.
Method:
The separation was achieved with a LiChrosorb®RP-18 (250 x 4.6 mm) column, at ambient
temperature with isocratic mode with mobile phase containing mixture of component A (acetonitrile)
and component B (0.001M NaH2PO4, with 5 mM 1-heptane sulfonic acid sodium salt, adjusted to pH
3.0) in a ratio 60:40 (v/v). The flow rate was 1.0 ml/min and the eluent was monitored at 297 nm. The
proposed method was validated as per ICH guidelines.
Result:
The obtained results showed that the compounds were sensitive to hydrolytic decomposition in
aqueous media, resulting in the splitting of the hydrazone bond. Rapid hydrolysis of substances was
observed in the acid medium. The elevated temperature significantly accelerated the hydrolytic reaction.
Relatively slow hydrolysis of 3a-c was observed in a neutral solution and aqueous solutions buffered
to pH 9. The hydrolysis of 3a-c in neutral, alkaline and strong alkaline medium followed the pseudo-
first-order reaction rate and showed a linear dependence of lnC versus time.
Conclusion:
A validated high-performance liquid chromatographic assay for the determination of the
hydrolytic stability of a series of aroylhydrazones was developed and optimized for the first time. The
methods devised are successfully applicable to the development of pharmaceutical formulations.
Collapse
Affiliation(s)
- Vania Maslarska
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2, Dunav St., 1000 Sofia,Bulgaria
| | - Stanislav Bozhanov
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2, Dunav St., 1000 Sofia,Bulgaria
| | - Stefka Ivanova
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2, Dunav St., 1000 Sofia,Bulgaria
| | - Violina T. Angelova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2, Dunav St., 1000 Sofia,Bulgaria
| |
Collapse
|
18
|
Liu R, Zuo R, Hudalla GA. Harnessing molecular recognition for localized drug delivery. Adv Drug Deliv Rev 2021; 170:238-260. [PMID: 33484737 PMCID: PMC8274479 DOI: 10.1016/j.addr.2021.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
A grand challenge in drug delivery is providing the right dose, at the right anatomic location, for the right duration of time to maximize therapeutic efficacy while minimizing off-target toxicity and other deleterious side-effects. Two general modalities are receiving broad attention for localized drug delivery. In the first, referred to as "targeted accumulation", drugs or drug carriers are engineered to have targeting moieties that promote their accumulation at a specific tissue site from circulation. In the second, referred to as "local anchoring", drugs or drug carriers are inserted directly into the tissue site of interest where they persist for a specified duration of time. This review surveys recent advances in harnessing molecular recognition between proteins, peptides, nucleic acids, lipids, and carbohydrates to mediate targeted accumulation and local anchoring of drugs and drug carriers.
Collapse
Affiliation(s)
- Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
19
|
Chemically-Gated and Sustained Molecular Transport through Nanoporous Gold Thin Films in Biofouling Conditions. NANOMATERIALS 2021; 11:nano11020498. [PMID: 33669404 PMCID: PMC7920421 DOI: 10.3390/nano11020498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/09/2021] [Accepted: 02/13/2021] [Indexed: 11/22/2022]
Abstract
Sustained release and replenishment of the drug depot are essential for the long-term functionality of implantable drug-delivery devices. This study demonstrates the use nanoporous gold (np-Au) thin films for in-plane transport of fluorescein (a small-molecule drug surrogate) over large (mm-scale) distances from a distal reservoir to the site of delivery, thereby establishing a constant flux of molecular release. In the absence of halides, the fluorescein transport is negligible due to a strong non-specific interaction of fluorescein with the pore walls. However, in the presence of physiologically relevant concentration of ions, halides preferentially adsorb onto the gold surface, minimizing the fluorescein–gold interactions and thus enabling in-plane fluorescein transport. In addition, the nanoporous film serves as an intrinsic size-exclusion matrix and allows for sustained release in biofouling conditions (dilute serum). The molecular release is reproducibly controlled by gating it in response to the presence of halides at the reservoir (source) and the release site (sink) without external triggers (e.g., electrical and mechanical).
Collapse
|
20
|
Srinivasan S, Yee NA, Wu K, Zakharian M, Mahmoodi A, Royzen M, Oneto JMM. SQ3370 Activates Cytotoxic Drug via Click Chemistry at Tumor and Elicits Sustained Responses in Injected & Non-injected Lesions. ADVANCED THERAPEUTICS 2021; 4. [PMID: 33869738 DOI: 10.1002/adtp.202000243] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
While systemic immuno-oncology therapies have shown remarkable success, only a limited subset of patients benefit from them. Our Click Activated Protodrugs Against Cancer (CAPAC™) Platform is a click chemistry-based approach that activates cancer drugs at a specific tumor with minimal systemic toxicity. CAPAC Platform is agnostic to tumor characteristics that can vary across patients and hence applicable to several types of tumors. We describe the benefits of SQ3370 (lead candidate of CAPAC) to achieve systemic anti-tumor responses in mice bearing two tumors. SQ3370 consists of a biopolymer, injected in a single lesion, followed by systemic doses of an attenuated protodrug™ of doxorubicin (Dox). SQ3370 was well-tolerated at 5.9-times the maximum dose of conventional Dox, increased survival by 63% and induced a systemic anti-tumor response against injected and non-injected lesions. The sustained anti-tumor response also correlated with immune activation measured at both lesions. SQ3370 could potentially benefit patients with micro-metastatic lesions.
Collapse
Affiliation(s)
- S Srinivasan
- Shasqi, Inc., 665 3 St., Suite 501, San Francisco, CA 94107
| | - N A Yee
- Shasqi, Inc., 665 3 St., Suite 501, San Francisco, CA 94107
| | - K Wu
- University of Albany, 1400 Washington Ave., LS-1136, Albany, NY 12222
| | - M Zakharian
- Shasqi, Inc., 665 3 St., Suite 501, San Francisco, CA 94107
| | - A Mahmoodi
- Shasqi, Inc., 665 3 St., Suite 501, San Francisco, CA 94107
| | - M Royzen
- University of Albany, 1400 Washington Ave., LS-1136, Albany, NY 12222
| | | |
Collapse
|
21
|
Cho HJ, Park SJ, Jung WH, Cho Y, Ahn DJ, Lee YS, Kim S. Injectable Single-Component Peptide Depot: Autonomously Rechargeable Tumor Photosensitization for Repeated Photodynamic Therapy. ACS NANO 2020; 14:15793-15805. [PMID: 33175520 DOI: 10.1021/acsnano.0c06881] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The general practice of photodynamic therapy (PDT) comprises repeated multiple sessions, where photosensitizers are repeatedly administered prior to each operation of light irradiation. To address potential problems arising from the total overdose of photosensitizer by such repeated injections, we here introduce an internalizing RGD peptide (iRGD) derivative (Ppa-iRGDC-BK01) that self-aggregates into an injectable single-component supramolecular depot. Ppa-iRGDC-BK01 is designed as an in situ self-implantable photosensitizer so that it forms a depot by itself upon injection, and its molecular functions (cancer cell internalization and photosensitization) are activated by sustained release, tumor targeting, and tumor-selective proteolytic/reductive cleavage of the iRGD segment. The experimental and theoretical studies revealed that when exposed to body temperature, Ppa-iRGDC-BK01 undergoes thermally accelerated self-assembly to form a supramolecular depot through the hydrophobic interaction of the Ppa pendants and the reorganization of the interpeptide hydrogen bonding. It turned out that the self-aggregation of Ppa-iRGDC-BK01 into a depot exerts a multiple-quenching effect on the photosensitivity to effectively prevent nonspecific phototoxicity and protect it from photobleaching outside the tumor, while enabling autonomous tumor rephotosensitization by long sustained release, tumor accumulation, and intratumoral activation over time. We demonstrate that depot formation through a single peritumoral injection and subsequent quintuple laser irradiations at intervals resulted in complete eradication of the tumor. During the repeated PDT, depot-implanted normal tissues around the tumor exhibited no phototoxic damage under laser exposure. Our approach of single-component photosensitizing supramolecular depot, combined with a strategy of tumor-targeted therapeutic activation, would be a safer and more precise operation of PDT through a nonconventional protocol composed of one-time photosensitizer injection and multiple laser irradiations.
Collapse
Affiliation(s)
- Hong-Jun Cho
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sung-Jun Park
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Woo Hyuk Jung
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Yuri Cho
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong June Ahn
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Yoon-Sik Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehoon Kim
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
22
|
Oliva N, Almquist BD. Spatiotemporal delivery of bioactive molecules for wound healing using stimuli-responsive biomaterials. Adv Drug Deliv Rev 2020; 161-162:22-41. [PMID: 32745497 DOI: 10.1016/j.addr.2020.07.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/23/2020] [Indexed: 12/28/2022]
Abstract
Wound repair is a fascinatingly complex process, with overlapping events in both space and time needed to pave a pathway to successful healing. This additional complexity presents challenges when developing methods for the controlled delivery of therapeutics for wound repair and tissue engineering. Unlike more traditional applications, where biomaterial-based depots increase drug solubility and stability in vivo, enhance circulation times, and improve retention in the target tissue, when aiming to modulate wound healing, there is a desire to enable localised, spatiotemporal control of multiple therapeutics. Furthermore, many therapeutics of interest in the context of wound repair are sensitive biologics (e.g. growth factors), which present unique challenges when designing biomaterial-based delivery systems. Here, we review the diverse approaches taken by the biomaterials community for creating stimuli-responsive materials that are beginning to enable spatiotemporal control over the delivery of therapeutics for applications in tissue engineering and regenerative medicine.
Collapse
|
23
|
Kang SM, Lee JH, Huh YS, Takayama S. Alginate Microencapsulation for Three-Dimensional In Vitro Cell Culture. ACS Biomater Sci Eng 2020; 7:2864-2879. [PMID: 34275299 DOI: 10.1021/acsbiomaterials.0c00457] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advances in microscale 3D cell culture systems have helped to elucidate cellular physiology, understand mechanisms of stem cell differentiation, produce pathophysiological models, and reveal important cell-cell and cell-matrix interactions. An important consideration for such studies is the choice of material for encapsulating cells and associated extracellular matrix (ECM). This Review focuses on the use of alginate hydrogels, which are versatile owing to their simple gelation process following an ionic cross-linking mechanism in situ, with no need for procedures that can be potentially toxic to cells, such as heating, the use of solvents, and UV exposure. This Review aims to give some perspectives, particularly to researchers who typically work more with poly(dimethylsiloxane) (PDMS), on the use of alginate as an alternative material to construct microphysiological cell culture systems. More specifically, this Review describes how physicochemical characteristics of alginate hydrogels can be tuned with regards to their biocompatibility, porosity, mechanical strength, ligand presentation, and biodegradability. A number of cell culture applications are also described, and these are subcategorized according to whether the alginate material is used to homogeneously embed cells, to micropattern multiple cellular microenvironments, or to provide an outer shell that creates a space in the core for cells and other ECM components. The Review ends with perspectives on future challenges and opportunities for 3D cell culture applications.
Collapse
Affiliation(s)
- Sung-Min Kang
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, 30332, United States of America.,The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, 30332, United States of America.,NanoBio High-Tech Materials Research Center, Department of Biological Engineering, Inha University, 100 Inha-ro, Incheon, 22212, Republic of Korea
| | - Ji-Hoon Lee
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, 30332, United States of America.,The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, 30332, United States of America
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Engineering, Inha University, 100 Inha-ro, Incheon, 22212, Republic of Korea
| | - Shuichi Takayama
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, 30332, United States of America.,The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, 30332, United States of America
| |
Collapse
|
24
|
Hsu RS, Fang JH, Shen WT, Sheu YC, Su CK, Chiang WH, Hu SH. Injectable DNA-architected nanoraspberry depot-mediated on-demand programmable refilling and release drug delivery. NANOSCALE 2020; 12:11153-11164. [PMID: 32400827 DOI: 10.1039/d0nr01185a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Drug delivery depots boosting a local concentration of therapeutic agents have received great attention in clinical applications due to their low occurrence of side effects and high therapeutic efficacy. However, once the payload is exhausted, the local drug concentration will be lower than the therapeutic window. To address this issue, an injectable double-strand deoxyribonucleic acid (DNA)-architected nanoraspberry depot (DNR-depot) was developed that can refill doxorubicin (Dox, an anticancer drug) from the blood and remotely control drug release on demand. The large porous surface on a uniform nanoraspberry (NR) filled covalently with DNA serves as a Dox sponge-like refilling reservoir, and the NR serves as a magnetic electrical absorber. Via the strong affinity between Dox and DNA molecules, the refilling process of Dox can be achieved on DNR-depot both in vitro and in vivo. Upon high-frequency magnetic field (HFMF) treatment, the remotely triggered release of Dox is actuated by the dissociation of Dox and DNA molecules, facilitating an approximately 800% improvement in drug concentration at the tumor site compared to free Dox injection alone. Furthermore, the cycles of refilling and release can be carried out more than 3 times in vivo within 21 days. The combination of refilling and HFMF-programmable Dox release in tumors via DNR-depot can effectively inhibit tumor growth for 30 days.
Collapse
Affiliation(s)
- Ru-Siou Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Jen-Hung Fang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Wei-Ting Shen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Yu-Chen Sheu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Cheng-Kuan Su
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan
| | - Wen-Hsuan Chiang
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| |
Collapse
|
25
|
McNamara SL, Brudno Y, Miller AB, Ham HO, Aizenberg M, Chaikof EL, Mooney DJ. Regenerating Antithrombotic Surfaces through Nucleic Acid Displacement. ACS Biomater Sci Eng 2020; 6:2159-2166. [PMID: 33455325 DOI: 10.1021/acsbiomaterials.0c00038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Blood-contacting devices are commonly coated with antithrombotic agents to prevent clot formation and to extend the lifespan of the device. However, in vivo degradation of these bioactive surface agents ultimately limits device efficacy and longevity. Here, a regenerative antithrombotic catheter surface treatment is developed using oligodeoxynucleotide (ODN) toehold exchange. ODN strands modified to carry antithrombotic payloads can inhibit the thrombin enzyme when bound to a surface and exchange with rapid kinetics over multiple cycles, even while carrying large payloads. The surface-bound ODNs inhibit thrombin activity to significantly reduce fibrinogen cleavage and fibrin formation, and this effect is sustained after ODN exchange of the surface-bound strands with a fresh antithrombotic payload. This study presents a unique strategy for achieving a continuous antithrombotic state for blood-contacting devices using an ODN-based regeneration method.
Collapse
Affiliation(s)
- Stephanie L McNamara
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02138, United States.,Harvard-MIT Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02138, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, 911 Oval Drive, Raleigh, North Carolina 27695, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| | - Alex B Miller
- Harvard-MIT Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02138, United States
| | - Hyun Oki Ham
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States.,Department of Surgery, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Michael Aizenberg
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| | - Elliot L Chaikof
- Harvard-MIT Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02138, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States.,Department of Surgery, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02138, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| |
Collapse
|
26
|
Adams MR, Moody CT, Sollinger JL, Brudno Y. Extracellular-Matrix-Anchored Click Motifs for Specific Tissue Targeting. Mol Pharm 2020; 17:392-403. [PMID: 31829613 DOI: 10.1021/acs.molpharmaceut.9b00589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Local presentation of cancer drugs by injectable drug-eluting depots reduces systemic side effects and improves efficacy. However, local depots deplete their drug stores and are difficult to introduce into stiff tissues, or organs, such as the brain, that cannot accommodate increased pressure. We present a method for introducing targetable depots through injection of activated ester molecules into target tissues that react with and anchor themselves to the local extracellular matrix (ECM) and subsequently capture systemically administered small molecules through bioorthogonal click chemistry. A computational model of tissue-anchoring depot formation and distribution was verified by histological analysis and confocal imaging of cleared tissues. ECM-anchored click groups do not elicit any noticeable local or systemic toxicity or immune response and specifically capture systemically circulating molecules at intradermal, intratumoral, and intracranial sites for multiple months. Taken together, ECM anchoring of click chemistry motifs is a promising approach to specific targeting of both small and large therapeutics, enabling repeated local presentation for cancer therapy and other diseases.
Collapse
Affiliation(s)
- Mary R Adams
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Christopher T Moody
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Jennifer L Sollinger
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States.,Lineberger Comprehensive Cancer Center , University of North Carolina, Chapel Hill , 450 West Dr. , Chapel Hill , North Carolina 27599 , United States
| |
Collapse
|
27
|
Abstract
This chapter is a brief overview of use of nanobiotechnology in drug delivery. Several types of nanoparticles are available. Nanoparticulate formulations of normally used drugs have increased efficacy due to improved absorption and require lower dosage with less side effects than standard formulations. Nanobiotechnology also facilitates targeted drug delivery of anticancer drugs, which is important for the management of cancer. Nanoparticles also facilitate crossing of biological barriers in the human body for drug delivery to targeted organs, for example, crossing the blood-brain barrier to reach the brain. Nanobiotechnology applications in delivery of biological therapies are expanding in areas such as cell and gene therapies, siRNAs, and monoclonal antibodies. Some nanoparticles can carry more than one therapeutic molecule enabling multimodal therapy and combination with physical modalities such as radiotherapy in cancer. Nanorobotics is developing with applications in drug delivery, particularly for cancer. Other anticipated developments in this area include use of nanotechnology for creating intelligent drug release devices.
Collapse
|
28
|
Wang H, Sobral MC, Snyder T, Brudno Y, Gorantla VS, Mooney DJ. Clickable, acid labile immunosuppressive prodrugs forin vivotargeting. Biomater Sci 2020; 8:266-277. [DOI: 10.1039/c9bm01487j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clickable immunosuppressive prodrugs enablein vivoreplenishment of drugs in biomaterial depots to maintain long-term immunosuppression in tissue/organ transplantation.
Collapse
Affiliation(s)
- Hua Wang
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| | - Miguel C. Sobral
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| | - Tracy Snyder
- Wyss Institute for Biologically Inspired Engineering
- Cambridge
- USA
| | - Yevgeny Brudno
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| | - Vijay S. Gorantla
- Surgery
- Ophthalmology and Bioengineering
- Wake Forest School of Medicine
- Winston-Salem
- USA
| | - David J. Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| |
Collapse
|
29
|
Czuban M, Srinivasan S, Yee NA, Agustin E, Koliszak A, Miller E, Khan I, Quinones I, Noory H, Motola C, Volkmer R, Di Luca M, Trampuz A, Royzen M, Mejia Oneto JM. Bio-Orthogonal Chemistry and Reloadable Biomaterial Enable Local Activation of Antibiotic Prodrugs and Enhance Treatments against Staphylococcus aureus Infections. ACS CENTRAL SCIENCE 2018; 4:1624-1632. [PMID: 30648146 PMCID: PMC6311693 DOI: 10.1021/acscentsci.8b00344] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Indexed: 05/08/2023]
Abstract
Systemic administration of antibiotics can cause severe side-effects such as liver and kidney toxicity, destruction of healthy gut bacteria, as well as multidrug resistance. Here, we present a bio-orthogonal chemistry-based strategy toward local prodrug concentration and activation. The strategy is based on the inverse electron-demand Diels-Alder chemistry between trans-cyclooctene and tetrazine and involves a biomaterial that can concentrate and activate multiple doses of systemic antibiotic therapy prodrugs at a local site. We demonstrate that a biomaterial, consisting of alginate hydrogel modified with tetrazine, is efficient at activating multiple doses of prodrugs of vancomycin and daptomycin in vitro as well as in vivo. These results support a drug delivery process that is independent of endogenous environmental markers. This approach is expected to improve therapeutic efficacy with decreased side-effects of antibiotics against bacterial infections. The platform has a wide scope of possible applications such as wound healing, and cancer and immunotherapy.
Collapse
Affiliation(s)
- Magdalena Czuban
- Berlin-Brandenburg
Center for Regenerative Therapies and Berlin-Brandenburg School for
Regenerative Therapies, Charité Universitätsmedizin
Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Institute
of Chemistry and Biochemistry, Freie Universität, Takustr. 3, 14195 Berlin, Germany
| | | | - Nathan A. Yee
- Shasqi
Inc., 665 Third Street, San Francisco, California 94107, United States
| | - Edgar Agustin
- Department
of Chemistry, University at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Anna Koliszak
- Berlin-Brandenburg
Center for Regenerative Therapies, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Ethan Miller
- Shasqi
Inc., 665 Third Street, San Francisco, California 94107, United States
| | - Irfan Khan
- Department
of Chemistry, University at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Ilenis Quinones
- Department
of Chemistry, University at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Hasina Noory
- Department
of Chemistry, University at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Christopher Motola
- Department
of Chemistry, University at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Rudolf Volkmer
- Institute
for Medical Immunology and Leibniz-Institut für Molekulare
Pharmakologie, Charité Universitätsmedizin
Berlin, 10117 Berlin, Germany
| | - Mariagrazia Di Luca
- Berlin-Brandenburg
Center for Regenerative Therapies, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andrej Trampuz
- Charité −
Universitätsmedizin Berlin, corporate member
of Freie Universität Berlin,
Humboldt-Universitat zu Berlin, and Berlin Institute of Health, Center
for Musculoskeletal Surgery, Charitéplatz 1, 10117 Berlin, Germany
| | - Maksim Royzen
- Department
of Chemistry, University at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Jose M. Mejia Oneto
- Shasqi
Inc., 665 Third Street, San Francisco, California 94107, United States
| |
Collapse
|
30
|
Sinha S, Saha ND, Sasmal R, Joshi D, Chandrasekhar S, Bosco MS, Agasti SS. Reversible encapsulations and stimuli-responsive biological delivery from a dynamically assembled cucurbit[7]uril host and nanoparticle guest scaffold. J Mater Chem B 2018; 6:7329-7334. [PMID: 32226626 PMCID: PMC7100906 DOI: 10.1039/c8tb01596a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The positive outcome of any therapeutic molecule requires control over its delivery rate. When delivered without control, administration of large doses is required to stimulate a therapeutic effect, frequently leading to increased toxicity or undesirable side effects. Recent advances introduced "smart" materials that actively release drugs in response to environmental stimuli. Although a variety of endogenous and exogenous triggers are reported, they are either difficult to control or lack tissue penetration depth. We report here a dynamic drug delivery scaffold based on a cucurbit[7]uril (CB[7]) host and benzylammonium functionalized gold nanoparticle (AuNP) guest that utilizes a bioorthogonal small molecule to achieve therapeutic control. In addition to their ability to reach deep tissue, small molecule activation is benefitted by their external controllability. Through cell culture studies we demonstrate that the host-guest supramolecular scaffold provides a nontoxic platform that effectively encapsulates a variety of therapeutic molecules and controls the payload release upon exposure to a high-affinity competitive guest molecule. This study presents a new strategy for controlling drug release rate through the use of competitive interactions of orthogonally presented guest molecules with immediate advantages in dosage control.
Collapse
Affiliation(s)
- Santu Sinha
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Nilanjana Das Saha
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
- Chemistry & Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Ranjan Sasmal
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Divyesh Joshi
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Soumya Chandrasekhar
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
- Chemistry & Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Monica Swetha Bosco
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
- Chemistry & Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Sarit S. Agasti
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
- Chemistry & Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
- School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| |
Collapse
|
31
|
Ultra-long-acting removable drug delivery system for HIV treatment and prevention. Nat Commun 2018; 9:4156. [PMID: 30297889 PMCID: PMC6175887 DOI: 10.1038/s41467-018-06490-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/29/2018] [Indexed: 01/20/2023] Open
Abstract
Non-adherence to medication is an important health care problem, especially in the treatment of chronic conditions. Injectable long-acting (LA) formulations of antiretrovirals (ARVs) represent a viable alternative to improve adherence to HIV/AIDS treatment and prevention. However, the LA-ARV formulations currently in clinical trials cannot be removed after administration even if adverse events occur. Here we show an ultra-LA removable system that delivers drug for up to 9 months and can be safely removed to stop drug delivery. We use two pre-clinical models for HIV transmission and treatment, non-human primates (NHP) and humanized BLT (bone marrow/liver/thymus) mice and show a single dose of subcutaneously administered ultra-LA dolutegravir effectively delivers the drug in both models and show suppression of viremia and protection from multiple high-dose vaginal HIV challenges in BLT mice. This approach represents a potentially effective strategy for the ultra-LA drug delivery with multiple possible therapeutic applications. Long-acting (LA) formulations of antiretroviral (ARV) drugs are an alternative approach to improve adherence for HIV treatment and prevention. Here the authors show a removable biodegradable ultra-LA-ARV drug system that effectively delivers drug, controls viremia and prevents infection in animal models of HIV infection.
Collapse
|
32
|
Im P, Kim J. On-Demand Macroscale Delivery System Based on a Macroporous Cryogel with a High Drug Loading Capacity for Enhanced Cancer Therapy. ACS Biomater Sci Eng 2018; 4:3498-3505. [PMID: 33450796 DOI: 10.1021/acsbiomaterials.8b00911] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The macroscale delivery system has been one of the practical platforms for a controlled delivery system by acting as a local depot close to the target tissue. In this study, we fabricated a macroporous alginate crygel incorporated with gold nanorods (GNRs) for the on-demand release of a chemotherapeutic drug from macroscale materials placed beside the target tumor. The macroporous crygel was prepared by the ice-crystal templating of a covalently crosslinked alginate hydrogel incorporated with GNRs. Mitoxantrone (MX), one of the potent anticancer drugs with a positive charge, was strongly adsorbed on the negative alginate chains of the cryogels. This system enabled a high loading of MX and a successful on-demand release of strongly bound MX from the GNR-loaded macroporous cryogels by near-infrared (NIR) irradiation by the dissociation of the interaction between the alginate backbone and MX. Cell viability after the NIR irradiation of the MX-loaded macroporous cryogel was significantly lower compared to that under no stimuli conditions. The in vivo test showed that repetitive NIR irradiations on the MX-loaded cryogel implanted near the tumor suppressed the tumor volume six times more than that of the control group. This simple approach to fabricate a macroporous cryogel capable of the on-demand release of bioactive cargos could be beneficial in various applications including cell, gene, and the other small molecule delivery systems.
Collapse
|
33
|
Brudno Y, Pezone MJ, Snyder TK, Uzun O, Moody CT, Aizenberg M, Mooney DJ. Replenishable drug depot to combat post-resection cancer recurrence. Biomaterials 2018; 178:373-382. [PMID: 29779862 PMCID: PMC6075722 DOI: 10.1016/j.biomaterials.2018.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 01/05/2023]
Abstract
Local drug presentation made possible by drug-eluting depots has demonstrated benefits in a vast array of diseases, including in cancer, microbial infection and in wound healing. However, locally-eluting depots are single-use systems that cannot be refilled or reused after implantation at inaccessible sites, limiting their clinical utility. New strategies to noninvasively refill drug-eluting depots could dramatically enhance their clinical use. In this report we present a refillable hydrogel depot system based on bioorthogonal click chemistry. The click-modified hydrogel depots capture prodrug refills from the blood and subsequently release active drugs locally in a sustained manner. Capture of the systemically-administered refills serves as an efficient and non-toxic method to repeatedly refill depots. Refillable depots in combination with prodrug refills achieve sustained release at precancerous tumor sites to improve cancer therapy while eliminating systemic side effects. The ability to target tissues without enhanced permeability could allow the use of refillable depots in cancer and many other medical applications.
Collapse
Affiliation(s)
- Yevgeny Brudno
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA; School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, MA 02138, USA; Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, 911 Oval Drive, Raleigh, NC 27695, USA; Lineberger Comprehensive Cancer Center, University of North Carolina - Chapel Hill, 450 West Dr, Chapel Hill, NC 27599, USA
| | - Matthew J Pezone
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Tracy K Snyder
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Oktay Uzun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Christopher T Moody
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, 911 Oval Drive, Raleigh, NC 27695, USA
| | - Michael Aizenberg
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA; School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, MA 02138, USA.
| |
Collapse
|
34
|
Li L, Wang C, Huang Q, Xiao J, Zhang Q, Cheng Y. A degradable hydrogel formed by dendrimer-encapsulated platinum nanoparticles and oxidized dextran for repeated photothermal cancer therapy. J Mater Chem B 2018; 6:2474-2480. [PMID: 32254464 DOI: 10.1039/c8tb00091c] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hydrogels can be used to prolong the retention of photothermal agents in tumors to conduct repeated photothermal therapy (PTT) and thus reduce high-dose-induced toxicity, but the long-term retention of photothermal nanoparticles post-therapy still increases the risk of toxicity. In this work, we developed an injectable and degradable photothermal hydrogel to conduct repeated PTT. The hydrogel was composed of dendrimer-encapsulated platinum nanoparticles (DEPts) crosslinked aldehyde-modified dextran via imine bond formation. The hydrogel represented excellent photothermal effect and good biocompatibility. It was able to remain in tumors for a period of days to allow repeated PTT, leading to complete tumor regression. After treatment, the hydrogel was gradually degraded due to the decomposition of imine bonds. The study developed a practical photothermal hydrogel that allowed repeated PTT and reduced long-term retention-induced toxicity.
Collapse
Affiliation(s)
- Lin Li
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China.
| | | | | | | | | | | |
Collapse
|
35
|
Dimatteo R, Darling NJ, Segura T. In situ forming injectable hydrogels for drug delivery and wound repair. Adv Drug Deliv Rev 2018; 127:167-184. [PMID: 29567395 PMCID: PMC6003852 DOI: 10.1016/j.addr.2018.03.007] [Citation(s) in RCA: 551] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/18/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
Hydrogels have been utilized in regenerative applications for many decades because of their biocompatibility and similarity in structure to the native extracellular matrix. Initially, these materials were formed outside of the patient and implanted using invasive surgical techniques. However, advances in synthetic chemistry and materials science have now provided researchers with a library of techniques whereby hydrogel formation can occur in situ upon delivery through standard needles. This provides an avenue to minimally invasively deliver therapeutic payloads, fill complex tissue defects, and induce the regeneration of damaged portions of the body. In this review, we highlight these injectable therapeutic hydrogel biomaterials in the context of drug delivery and tissue regeneration for skin wound repair.
Collapse
Affiliation(s)
- Robert Dimatteo
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States.
| | - Nicole J Darling
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States.
| | - Tatiana Segura
- Department of Chemical and Biomolecular Engineering, Bioengineering, and Dermatology, School of Medicine, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States.
| |
Collapse
|
36
|
Abstract
Over the last century, there has been a dramatic change in the nature of therapeutic, biologically active molecules available to treat disease. Therapies have evolved from extracted natural products towards rationally designed biomolecules, including small molecules, engineered proteins and nucleic acids. The use of potent drugs which target specific organs, cells or biochemical pathways, necessitates new tools which can enable controlled delivery and dosing of these therapeutics to their biological targets. Here, we review the miniaturisation of drug delivery systems from the macro to nano-scale, focussing on controlled dosing and controlled targeting as two key parameters in drug delivery device design. We describe how the miniaturisation of these devices enables the move from repeated, systemic dosing, to on-demand, targeted delivery of therapeutic drugs and highlight areas of focus for the future.
Collapse
Affiliation(s)
- Derfogail Delcassian
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,b Department of Anaesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , MA , USA.,c Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy , University of Nottingham , Nottingham , UK
| | - Asha K Patel
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,d Division of Cancer and Stem Cells, School of Medicine, and Division of Advanced Materials and Healthcare Technologies, School of Pharmacy , University of Nottingham , Nottingham , UK
| | - Abel B Cortinas
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,e Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Robert Langer
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,e Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA.,f Institute for Medical Engineering and Science , Massachusetts Institute of Technology , Cambridge , MA , USA.,g Media Lab , Massachusetts Institute of Technology , Cambridge , MA , USA
| |
Collapse
|
37
|
Wu JPJ, Cheng B, Roffler SR, Lundy DJ, Yen CYT, Chen P, Lai JJ, Pun SH, Stayton PS, Hsieh PCH. Reloadable multidrug capturing delivery system for targeted ischemic disease treatment. Sci Transl Med 2017; 8:365ra160. [PMID: 27856799 DOI: 10.1126/scitranslmed.aah6228] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 10/01/2016] [Indexed: 12/14/2022]
Abstract
Human clinical trials of protein therapy for ischemic diseases have shown disappointing outcomes so far, mainly because of the poor circulatory half-life of growth factors in circulation and their low uptake and retention by the targeted injury site. The attachment of polyethylene glycol (PEG) extends the circulatory half-lives of protein drugs but reduces their extravasation and retention at the target site. To address this issue, we have developed a drug capture system using a mixture of hyaluronic acid (HA) hydrogel and anti-PEG immunoglobulin M antibodies, which, when injected at a target body site, can capture and retain a variety of systemically injected PEGylated therapeutics at that site. Furthermore, repeated systemic injections permit "reloading" of the capture depot, allowing the use of complex multistage therapies. This study demonstrates this capture system in both murine and porcine models of critical limb ischemia. The results show that the reloadable HA/anti-PEG system has the potential to be clinically applied to patients with ischemic diseases, who require sequential administration of protein drugs for optimal outcomes.
Collapse
Affiliation(s)
- Jasmine P J Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Bill Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - David J Lundy
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | | | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei 115, Taiwan
| | - James J Lai
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan. .,Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.,Institute of Medical Genomics and Proteomics and Department of Surgery, National Taiwan University and Hospital, Taipei 100, Taiwan
| |
Collapse
|
38
|
Chen J, Li S, Zhang Y, Wang W, Zhang X, Zhao Y, Wang Y, Bi H. A Reloadable Self-Healing Hydrogel Enabling Diffusive Transport of C-Dots Across Gel-Gel Interface for Scavenging Reactive Oxygen Species. Adv Healthc Mater 2017; 6. [PMID: 28945014 DOI: 10.1002/adhm.201700746] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/09/2017] [Indexed: 12/20/2022]
Abstract
While reloadable drug delivery platforms are highly prized for the treatment of a broad spectrum of diseases, the gel-gel interface between hydrogels hinders the intergel diffusive transport of drugs and thus limits the application of hydrogels as reloadable depots. Here, this study reports the circumvention of this barrier by employing a self-healing hydrogel prepared from N-carboxyethyl chitosan and sodium alginate dialdehyde, which are cross-linked via a reversible Schiff base linkage. The injectable and bioadhesive hydrogel shows a rapid gelation time of 47 s. The dynamic self-healing process enables the efficient diffusive transport of carbon quantum dots (C-dots) into an adjacent hydrogel, and thus, the C-dots can be used to scavenge reactive oxygen species from a remote inflammation site. Specifically, the diffusive transport of the C-dots in the self-healing hydrogel after three sequential reloading steps is sevenfold greater than that in the non-self-healing counterpart. In vivo, hematoxylin and eosin staining of the murine skin at the injection site shows no apparent symptoms of inflammation in the group treated with the reloadable self-healing hydrogel. The current strategy represents a promising and straightforward route for the design of a reloadable drug delivery system for future use in clinical application.
Collapse
Affiliation(s)
- Jing Chen
- College of Chemistry and Chemical Engineering; Anhui University; Hefei 230601 China
- School of Life Sciences; Hefei Normal University; Hefei 230601 China
| | - Shuya Li
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Diseases School of Life Sciences and Medical Center; University of Science and Technology of China; Hefei 230027 China
| | - Ye Zhang
- College of Chemistry and Chemical Engineering; Anhui University; Hefei 230601 China
| | - Wei Wang
- School of Life Sciences; Hefei Normal University; Hefei 230601 China
| | - Xiang Zhang
- College of Chemistry and Chemical Engineering; Anhui University; Hefei 230601 China
| | - Yangyang Zhao
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Diseases School of Life Sciences and Medical Center; University of Science and Technology of China; Hefei 230027 China
| | - Yucai Wang
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Diseases School of Life Sciences and Medical Center; University of Science and Technology of China; Hefei 230027 China
| | - Hong Bi
- College of Chemistry and Chemical Engineering; Anhui University; Hefei 230601 China
| |
Collapse
|
39
|
Sepantafar M, Maheronnaghsh R, Mohammadi H, Radmanesh F, Hasani-Sadrabadi MM, Ebrahimi M, Baharvand H. Engineered Hydrogels in Cancer Therapy and Diagnosis. Trends Biotechnol 2017; 35:1074-1087. [PMID: 28734545 DOI: 10.1016/j.tibtech.2017.06.015] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023]
Abstract
Over the last decade, numerous investigations have attempted to clarify the intricacies of tumor development to propose effective approaches for cancer treatment. Thanks to the unique properties of hydrogels, researchers have made significant progress in tumor model reconstruction, tumor diagnosis, and associated therapies. Notably, hydrogel-based systems can be adjusted to respond to cancer-specific hallmarks and/or external stimuli. These well-known drug reservoirs can be used as smart carriers for multiple cargos, including both naked and nanoparticle-encapsulated chemotherapeutics, genes, and radioisotopes. Recent works have attempted to specialize hydrogels for cancer research; we comprehensively review this topic for the first time, synthesizing past results and defining paths for future work.
Collapse
Affiliation(s)
- Mohammadmajid Sepantafar
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reihan Maheronnaghsh
- Department of Genetics, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Mohammadi
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300 Nibong Tebal, Penang, Malaysia
| | - Fatemeh Radmanesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Mahdi Hasani-Sadrabadi
- Parker H. Petit Institute for Bioengineering and Bioscience, G.W. Woodruff School of Mechanical Engineering and School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
40
|
Wu CY, Chang CW, Yuan RH, Chiang YC, Chen JT, Kang DY, Chen HY. Multifunctional nanoparticles with controllable dimensions and tripled orthogonal reactivity. NANOSCALE 2017; 9:14787-14791. [PMID: 28952633 DOI: 10.1039/c7nr04771a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Multifunctional nanoparticles featuring three distinct and orthogonal functionalities for performing catalyst-free click reactions of azide-alkyne and maleimide-thiol and atom transfer radical polymerization (ATRP) are fabricated using a simple chemical vapor deposition copolymerization approach with the flexibility to control the particle size and geometry.
Collapse
Affiliation(s)
- Chih-Yu Wu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
41
|
Alginate hydrogel improves anti-angiogenic bevacizumab activity in cancer therapy. Eur J Pharm Biopharm 2017; 119:271-282. [DOI: 10.1016/j.ejpb.2017.06.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 04/09/2017] [Accepted: 06/28/2017] [Indexed: 01/30/2023]
|
42
|
Abstract
The formation of oximes and hydrazones is employed in numerous scientific fields as a simple and versatile conjugation strategy. This imine-forming reaction is applied in fields as diverse as polymer chemistry, biomaterials and hydrogels, dynamic combinatorial chemistry, organic synthesis, and chemical biology. Here we outline chemical developments in this field, with special focus on the past ∼10 years of developments. Recent strategies for installing reactive carbonyl groups and α-nucleophiles into biomolecules are described. The basic chemical properties of reactants and products in this reaction are then reviewed, with an eye to understanding the reaction's mechanism and how reactant structure controls rates and equilibria in the process. Recent work that has uncovered structural features and new mechanisms for speeding the reaction, sometimes by orders of magnitude, is discussed. We describe recent studies that have identified especially fast reacting aldehyde/ketone substrates and structural effects that lead to rapid-reacting α-nucleophiles as well. Among the most effective new strategies has been the development of substituents near the reactive aldehyde group that either transfer protons at the transition state or trap the initially formed tetrahedral intermediates. In addition, the recent development of efficient nucleophilic catalysts for the reaction is outlined, improving greatly upon aniline, the classical catalyst for imine formation. A number of uses of such second- and third-generation catalysts in bioconjugation and in cellular applications are highlighted. While formation of hydrazone and oxime has been traditionally regarded as being limited by slow rates, developments in the past 5 years have resulted in completely overturning this limitation; indeed, the reaction is now one of the fastest and most versatile reactions available for conjugations of biomolecules and biomaterials.
Collapse
Affiliation(s)
- Dominik K Kölmel
- Department of Chemistry, Stanford University , Stanford, California 94305, United States
| | - Eric T Kool
- Department of Chemistry, Stanford University , Stanford, California 94305, United States
| |
Collapse
|
43
|
Nanoformulation-based sequential combination cancer therapy. Adv Drug Deliv Rev 2017; 115:57-81. [PMID: 28412324 DOI: 10.1016/j.addr.2017.04.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 01/07/2023]
Abstract
Although combining two or more treatments is regarded as an indispensable approach for effectively treating cancer, the traditional cocktail-based combination therapies are seriously limited by coordination issues that fail to account for differences in the pharmacokinetics and action sites of each drug. The careful manipulation of dosing regimens, such as by the sequential application of combination treatments, may satisfy the temporal and spatial needs of each drug and achieve successful combination antitumor therapy. Nanotechnology-based carriers might be the best tools for sequential combination therapy, as they can be loaded with multiple cargos and may provide targeted and sustained delivery to target tumor cells. Single nanoformulations capable of sequentially releasing drugs have shown synergistic anticancer activity, such as by sensitizing tumor cells through cascaded drug delivery or remodeling the tumor vasculature and microenvironment to enhance the tumor distribution of nanotherapeutics. This review highlights the use of nanotechnology-based multistage drug delivery for cancer treatment, focusing on the ability of such formulations to enhance antitumor efficacy by applying sequential treatment and modulating dosing regimens, which are challenges currently being faced in the clinic.
Collapse
|
44
|
Yang GZ, Li JJ, Yu DG, He MF, Yang JH, Williams GR. Nanosized sustained-release drug depots fabricated using modified tri-axial electrospinning. Acta Biomater 2017; 53:233-241. [PMID: 28137657 DOI: 10.1016/j.actbio.2017.01.069] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/18/2017] [Accepted: 01/25/2017] [Indexed: 11/15/2022]
Abstract
Nanoscale drug depots, comprising a drug reservoir surrounded by a carrier membrane, are much sought after in contemporary pharmaceutical research. Using cellulose acetate (CA) as a filament-forming polymeric matrix and ferulic acid (FA) as a model drug, nanoscale drug depots in the form of core-shell fibers were designed and fabricated using a modified tri-axial electrospinning process. This employed a solvent mixture as the outer working fluid, as a result of which a robust and continuous preparation process could be achieved. The fiber-based depots had a linear morphology, smooth surfaces, and an average diameter of 0.62±0.07μm. Electron microscopy data showed them to have clear core-shell structures, with the FA encapsulated inside a CA shell. X-ray diffraction and IR spectroscopy results verified that FA was present in the crystalline physical form. In vitro dissolution tests revealed that the fibers were able to provide close to zero-order release over 36h, with no initial burst release and minimal tailing-off. The release properties of the depot systems were much improved over monolithic CA/FA fibers, which exhibited a significant burst release and also considerable tailing-off at the end of the release experiment. Here we thus demonstrate the concept of using modified tri-axial electrospinning to design and develop new types of heterogeneous nanoscale biomaterials. STATEMENT OF SIGNIFICANCE Nanoscale drug depots with a drug reservoir surrounded by a carrier are highly attractive in biomedicine. A cellulose acetate based drug depot was investigated in detail, starting with the design of the nanostructure, and moving through its fabrication using a modified tri-axial electrospinning process and a series of characterizations. The core-shell fiber-based drug depots can provide a more sustained release profile with no initial burst effect and less tailing-off than equivalent monolithic drug-loaded fibers. The drug release mechanisms are also distinct in the two systems. This proof-of-concept work can be further expanded to conceive a series of new structural biomaterials with improved or new functional performance.
Collapse
Affiliation(s)
- Guang-Zhi Yang
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Jiao-Jiao Li
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Deng-Guang Yu
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China.
| | - Mei-Feng He
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Jun-He Yang
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
45
|
Dong X, Chen H, Qin J, Wei C, Liang J, Liu T, Kong D, Lv F. Thermosensitive porphyrin-incorporated hydrogel with four-arm PEG-PCL copolymer (II): doxorubicin loaded hydrogel as a dual fluorescent drug delivery system for simultaneous imaging tracking in vivo. Drug Deliv 2017; 24:641-650. [PMID: 28282993 PMCID: PMC8241078 DOI: 10.1080/10717544.2017.1289570] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Visualization of a drug delivery system could reveal the pharmacokinetic properties, which is essential for the design of a novel drug delivery system. In vivo optical imaging offers an advanced tool to monitor the drug release process and the therapeutic effect by the combination of fluorescence imaging and bioluminescence imaging. Multispectral fluorescence imaging can separate the drug and the carrier without interference. Herein, a dual fluorescent anti-tumor drug delivery system was monitored with the doxorubicin-loaded hydrogel to further explore the application of the porphyrin-incorporated hydrogel with four-arm PEG-PCL copolymer as a drug carrier, based on the beneficial fluorescence and good biocompatibility of the porphyrin incorporated hydrogel. Using nude mice bearing luciferase expressed hepatic tumor as models, the whole process from the drug delivery to the tumor therapeutic effects were real time visualized simultaneously after administration at interval from 0 to 18 d. The imaging results suggest that the fluorescence signals of the drug and the carrier can be separated and unmixed from the drug-loaded hydrogel successfully, avoiding the interference of the fluorescence signals. The tumor growth or inhibition can be real time tracked and analyzed quantitatively by bioluminescence imaging. Noninvasive continuous tracking the in vivo drug delivery process simultaneously is a potential trend for the precise drug delivery and treatment.
Collapse
Affiliation(s)
- Xia Dong
- a Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , PR China and
| | - Hongli Chen
- b School of Life Science and Technology, Xinxiang Medical University , Xinxiang , Henan , PR China
| | - Jingwen Qin
- b School of Life Science and Technology, Xinxiang Medical University , Xinxiang , Henan , PR China
| | - Chang Wei
- a Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , PR China and
| | - Jie Liang
- a Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , PR China and
| | - Tianjun Liu
- a Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , PR China and
| | - Deling Kong
- a Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , PR China and
| | - Feng Lv
- a Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , PR China and
| |
Collapse
|
46
|
Colby AH, Oberlies NH, Pearce CJ, Herrera VLM, Colson YL, Grinstaff MW. Nanoparticle drug-delivery systems for peritoneal cancers: a case study of the design, characterization and development of the expansile nanoparticle. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [PMID: 28185434 DOI: 10.1002/wnan.1451] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/30/2016] [Accepted: 12/17/2016] [Indexed: 12/24/2022]
Abstract
Nanoparticle (NP)-based drug-delivery systems are frequently employed to improve the intravenous administration of chemotherapy; however, few reports explore their application as an intraperitoneal therapy. We developed a pH-responsive expansile nanoparticle (eNP) specifically designed to leverage the intraperitoneal route of administration to treat intraperitoneal malignancies, such as mesothelioma, ovarian, and pancreatic carcinomatoses. This review describes the design, evaluation, and evolution of the eNP technology and, specifically, a Materials-Based Targeting paradigm that is unique among the many active- and passive-targeting strategies currently employed by NP-delivery systems. pH-responsive eNP swelling is responsible for the extended residence at the target tumor site as well as the subsequent improvement in tumoral drug delivery and efficacy observed with paclitaxel-loaded eNPs (PTX-eNPs) compared to the standard clinical formulation of paclitaxel, Taxol®. Superior PTX-eNP efficacy is demonstrated in two different orthotopic models of peritoneal cancer-mesothelioma and ovarian cancer; in a third model-of pancreatic cancer-PTX-eNPs demonstrated comparable efficacy to Taxol with reduced toxicity. Furthermore, the unique structural and responsive characteristics of eNPs enable them to be used in three additional treatment paradigms, including: treatment of lymphatic metastases in breast cancer; use as a highly fluorescent probe to visually guide the resection of peritoneal implants; and, in a two-step delivery paradigm for concentrating separately administered NP and drug at a target site. This case study serves as an important example of using the targeted disease-state's pathophysiology to inform the NP design as well as the method of use of the delivery system. WIREs Nanomed Nanobiotechnol 2017, 9:e1451. doi: 10.1002/wnan.1451 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Aaron H Colby
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, USA.,Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA
| | | | - Victoria L M Herrera
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark W Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, MA, USA
| |
Collapse
|
47
|
Hu J, Quan Y, Lai Y, Zheng Z, Hu Z, Wang X, Dai T, Zhang Q, Cheng Y. A smart aminoglycoside hydrogel with tunable gel degradation, on-demand drug release, and high antibacterial activity. J Control Release 2017; 247:145-152. [DOI: 10.1016/j.jconrel.2017.01.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/05/2016] [Accepted: 01/04/2017] [Indexed: 01/11/2023]
|
48
|
Hu J, Chen Y, Li Y, Zhou Z, Cheng Y. A thermo-degradable hydrogel with light-tunable degradation and drug release. Biomaterials 2017; 112:133-140. [DOI: 10.1016/j.biomaterials.2016.10.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022]
|
49
|
Abstract
Hydrogel delivery systems can leverage therapeutically beneficial outcomes of drug delivery and have found clinical use. Hydrogels can provide spatial and temporal control over the release of various therapeutic agents, including small-molecule drugs, macromolecular drugs and cells. Owing to their tunable physical properties, controllable degradability and capability to protect labile drugs from degradation, hydrogels serve as a platform in which various physiochemical interactions with the encapsulated drugs control their release. In this Review, we cover multiscale mechanisms underlying the design of hydrogel drug delivery systems, focusing on physical and chemical properties of the hydrogel network and the hydrogel-drug interactions across the network, mesh, and molecular (or atomistic) scales. We discuss how different mechanisms interact and can be integrated to exert fine control in time and space over the drug presentation. We also collect experimental release data from the literature, review clinical translation to date of these systems, and present quantitative comparisons between different systems to provide guidelines for the rational design of hydrogel delivery systems.
Collapse
Affiliation(s)
- Jianyu Li
- John A. Paulson School of Engineering and Applied Sciences, and the Wyss Institute for biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, and the Wyss Institute for biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
50
|
Shi J, Zhang H, Jackson J, Shademani A, Chiao M. A robust and refillable magnetic sponge capsule for remotely triggered drug release. J Mater Chem B 2016; 4:7415-7422. [PMID: 32263742 DOI: 10.1039/c6tb02762h] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Numerous different delivery systems have been developed for local administration of drugs. However, their service lives generally depend on the payload depletion time and most of them are designed for one time use due to lack of drug replenishment abilities. To address this issue, a refillable magnetic porous polydimethylsiloxane (PDMS) capsule is proposed for remotely controlled drug delivery applications. An inner cavity is built in the sponge scaffold to provide space for drug storage and the refilling is accomplished by injecting drugs with a syringe. The rapid and reversible deformation of the magnetic porous structure under a magnetic field offers a controlled pumping force to push drugs out of the capsule. In this work, low molecular weight (methylene blue, MB, 320 g mol-1) and high molecular weight (bovine serum albumin, BSA, 67 000 g mol-1) molecules were used as model compounds to test and verify the operational principle. This proof-of-concept study has demonstrated the capability of the refillable porous capsule in controlled drug delivery under external magnetic stimuli.
Collapse
Affiliation(s)
- Jingru Shi
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.
| | | | | | | | | |
Collapse
|