1
|
Mohizin A, Sung B. Biophysical simulation of transcutaneous drug delivery for the rational design of hollow microneedle-based insulin infusion. Comput Biol Med 2025; 193:110365. [PMID: 40403633 DOI: 10.1016/j.compbiomed.2025.110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 05/24/2025]
Abstract
Drug delivery through the skin using miniaturised needles is a promising microtechnology-based strategy for precise control of drug release dynamics. Active injection of therapeutic insulin through hollow microneedles has recently been implemented as patch-type microsystems in which liquid dispensers and drug reservoirs are integrated with the microneedle array. Nevertheless, little is known about the relationship of the micro-infusion parameters, such as the microfluid flow rate, geometry and opening size of hollow microneedle, and needle insertion depth, to the biomechanics of viable skin multilayers and its associated pain perception, and to insulin absorption kinetics by microcirculation. In this study, we developed a novel computational modelling method that encapsulates numerical analysis modules to simulate (i) insulin infusion processes by intradermal and subcutaneous injection, (ii) skin tissue deformation and pain sensation during micro-infusion, and (iii) microfluid transport and pharmacokinetics. Our model predicts that the convection-diffusion behaviour of the insulin solution in the tissue matrix can be dramatically affected by the different target skin layers, that is, the dermis and hypodermis. Furthermore, this could be extended to the differences in insulin absorption kinetics depending on the skin layer where insulin is injected. We found that coupling between the biomicrofluidics of insulin infusion and the biomechanics of heterogeneous skin tissues could result in a prominent variation in plasma insulin concentration as a function of time. In addition, a numerical relationship between the pain perception level and microflow characteristics at the opening of the needle tip was obtained. Our model could serve as a versatile tool for designing skin drug delivery microdevices, by considering the quantitative interactions of the multifaceted biophysical mechanisms involved in microneedle-based drug infusion.
Collapse
Affiliation(s)
- Abdul Mohizin
- Biosensor Group, KIST Europe Forschungsgesellschaft mbH, 66123, Saarbrücken, Germany
| | - Baeckkyoung Sung
- Biosensor Group, KIST Europe Forschungsgesellschaft mbH, 66123, Saarbrücken, Germany; Division of Energy & Environment Technology, University of Science & Technology, 34113, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Kim JY, Um YS, Na YG, Kim DE, Song YH, Hwang S, Jin M, Kim J, Baek SK, Baek JS, Lee HK, Cho CW. Buccal application of microneedles coated with an optimized hydrogel containing naproxen and dexamethasone. Drug Deliv Transl Res 2025:10.1007/s13346-025-01870-4. [PMID: 40325306 DOI: 10.1007/s13346-025-01870-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Inflammation and impaired bone regeneration are major challenges in oral and maxillofacial surgery, necessitating the development of effective drug delivery systems. This study aimed to develop a hydrogel-based microneedle (MN) system for the controlled release of anti-inflammatory and osteogenic drugs. A hydrogel loaded with naproxen sodium (NAS) and dexamethasone sodium phosphate (DEX) using poloxamer 407 (NDgel) was prepared using a low-temperature method and optimized via the Box-Behnken design. The optimized hydrogel exhibited a gelation temperature of 30.87 ± 0.64℃, pH 7.92 ± 0.12, and viscosity 87.47 ± 5.66 cP. Physicochemical evaluations, including differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FT-IR), confirmed that NAS and DEX were incorporated in an amorphous form. The hydrogel was coated onto microneedles (NDgMN) via a dip-coating method and dried. In vitro drug release studies in artificial saliva showed NAS and DEX release rates of 21.7 ± 5.8% and 19.0 ± 1.8%, respectively, after 5 min. The NDgMN exhibited significantly enhanced permeability, with 48.5% and 48.7% permeability for NAS and DEX after 48 h, compared to 31.0% and 28.8% for the hydrogel alone. The IC50 values of the drug solution and drug-containing gel were 123 µg/mL and 203.2 µg/mL, respectively. NDgel demonstrated concentration-dependent inhibition of nitrogen oxide (NO) production at 1-1000 µg/mL, and alkaline phosphatase (ALP) activity assays revealed a 1.2-fold increase at concentrations above 50 µg/mL. These findings suggest that hydrogel-coated MNs have potential as a novel drug delivery system for reducing inflammation and promoting osteocyte differentiation due to their enhanced permeability and bioactivity.
Collapse
Affiliation(s)
- Joo-Young Kim
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea
| | - Yun-Sik Um
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea
| | - Young-Guk Na
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea
| | - Da-Eun Kim
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea
| | - Yo Han Song
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea
| | - Suyeon Hwang
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea
| | - Minki Jin
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Jooyoung Kim
- QuadMedicine R&D Centre, QuadMedicine, Inc, Seongnam, Gyeonggi, 13209, South Korea
| | - Seung-Ki Baek
- QuadMedicine R&D Centre, QuadMedicine, Inc, Seongnam, Gyeonggi, 13209, South Korea
| | - Jong-Suep Baek
- Department of Bio‑Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Department of Bio‑Functional Materia, Kangwon National University, Samcheok, 25949, Republic of Korea.
| | - Hong-Ki Lee
- College of Veterinary Medicine, Chungbuk National University, Chungbuk, 28644, Republic of Korea.
| | - Cheong-Weon Cho
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 31434, South Korea.
| |
Collapse
|
3
|
Lin H, Liu J, Hou Y, Yu Z, Hong J, Yu J, Chen Y, Hu J, Xia D. Microneedle patch with pure drug tips for delivery of liraglutide: pharmacokinetics in rats and minipigs. Drug Deliv Transl Res 2025; 15:216-230. [PMID: 38619705 DOI: 10.1007/s13346-024-01582-1] [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] [Accepted: 03/16/2024] [Indexed: 04/16/2024]
Abstract
Transdermal delivery of peptide drugs is almost impossible with conventional penetration enhancers because of epidermal barrier function. Microneedle (MN) patches can bypass the epidermal barrier and have been developed for trans- and intradermal delivery of peptide drugs and vaccines. However, dissolving MN patches are limited by low drug loading capacities due to their small size and admixture of drug and water-soluble excipients. Furthermore, few in vivo pharmacokinetic studies, especially in large animals such as pigs, have been performed to assess post-application systemic drug exposure. Here, we developed a dissolving MN patch with pure liraglutide at the needle tips. The MN patch could load up to 2.21 ± 0.14 mg of liraglutide in a patch size of 0.9 cm2, which was nearly two orders of magnitude higher than that obtained with conventional MN patches of the same size. Raman imaging confirmed that liraglutide was localized at the MN tips. The MN had sufficient mechanical strength to penetrate the epidermis and could deliver up to 0.93 ± 0.04 mg of liraglutide into skin with a dosing variability of less than 6.8%. The MN patch delivery enabled faster absorption of liraglutide than that provided by subcutaneous (S.C.) injection, and achieved relative bioavailability of 69.8% and 46.3% compared to S.C. injection in rats and minipigs, respectively. The MN patch also exhibited similar patterns of anti-hyperglycemic effect in diabetic rats and individual variability in pharmacokinetic parameters as S.C. injection. The liraglutide MN application was well tolerated; no skin irritation was observed in minipigs except for mild erythema occurring within 4 h after once daily administration for 7 days at the same site. Our preclinical study suggests that MN patch with pure drug needle tips might offer a safe and effective alternative to S.C. injection for administration of liraglutide.
Collapse
Affiliation(s)
- Hongbing Lin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jinbin Liu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yulin Hou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhiyan Yu
- Dongguan HEC Biopharmaceutical R&D Co., Ltd., Dongguan, China
| | - Juan Hong
- Dongguan HEC Biopharmaceutical R&D Co., Ltd., Dongguan, China
| | - Jianghong Yu
- Dongguan HEC Biopharmaceutical R&D Co., Ltd., Dongguan, China
| | - Yu Chen
- Dongguan HEC Biopharmaceutical R&D Co., Ltd., Dongguan, China
| | - Jingwen Hu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Dengning Xia
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
4
|
Fu W, Li Q, Sheng J, Wu H, Ma M, Zhang Y. Whole-Cell Vaccine Preparation Through Prussian Blue Nanoparticles-Elicited Immunogenic Cell Death and Loading in Gel Microneedles Patches. Gels 2024; 10:838. [PMID: 39727596 DOI: 10.3390/gels10120838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Tumor whole-cell vaccines are designed to introduce a wide range of tumor-associated antigens into the body to counteract the immunosuppression caused by tumors. In cases of lymphoma of which the specific antigen is not yet determined, the tumor whole-cell vaccine offers distinct advantages. However, there is still a lack of research on an effective preparation method for the lymphoma whole-cell vaccine. To solve this challenge, we prepared a whole-cell vaccine derived from non-Hodgkin B-cell lymphoma (A20) via the photothermal effect mediated by Prussian blue nanoparticles (PBNPs). The immune activation effect of this vaccine against lymphoma was verified at the cellular level. The PBNPs-treated A20 cells underwent immunogenic cell death (ICD), causing the loss of their ability to form tumors while retaining their ability to trigger an immune response. A20 cells that experienced ICD were further ultrasonically crushed to prepare the A20 whole-cell vaccine with exposed antigens and enhanced immunogenicity. The A20 whole-cell vaccine was able to activate the dendritic cells (DCs) to present antigens to T cells and trigger specific immune responses against lymphoma. Whole-cell vaccines are primarily administered through direct injection, a method that often results in low delivery efficiency and poor patient compliance. Comparatively, the microneedle patch system provides intradermal delivery, offering enhanced lymphatic absorption and improved patient adherence due to its minimally invasive approach. Thus, we developed a porous microneedle patch system for whole-cell vaccine delivery using Gelatin Methacryloyl (GelMA) hydrogel and n-arm-poly(lactic-co-glycolic acid) (n-arm-PLGA). This whole-cell vaccine combined with porous gel microneedle patch delivery system has the potential to become a simple immunotherapy method with controllable production and represents a promising new direction for the treatment of lymphoma.
Collapse
Affiliation(s)
- Wenxin Fu
- State Key Laboratory of Digital Medical Engineering, Basic Medicine Research and Innovation Center of Ministry of Education, Southeast University, Nanjing 211102, China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Qianqian Li
- State Key Laboratory of Digital Medical Engineering, Basic Medicine Research and Innovation Center of Ministry of Education, Southeast University, Nanjing 211102, China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Jingyi Sheng
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Haoan Wu
- State Key Laboratory of Digital Medical Engineering, Basic Medicine Research and Innovation Center of Ministry of Education, Southeast University, Nanjing 211102, China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ming Ma
- State Key Laboratory of Digital Medical Engineering, Basic Medicine Research and Innovation Center of Ministry of Education, Southeast University, Nanjing 211102, China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Basic Medicine Research and Innovation Center of Ministry of Education, Southeast University, Nanjing 211102, China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
5
|
Koenitz L, Crean A, Vucen S. Pharmacokinetic differences between subcutaneous injection and intradermal microneedle delivery of protein therapeutics. Eur J Pharm Biopharm 2024; 204:114517. [PMID: 39349073 DOI: 10.1016/j.ejpb.2024.114517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
Protein therapeutics are essential in the treatment of various diseases, but most of them require parenteral administration. Since intravenous and subcutaneous injections are associated with discomfort and pain, other routes have been investigated including intradermal microneedle delivery. Microneedles are shorter than hypodermic needles and therefore minimize contact with pain receptors in deeper skin layers. But the differences in anatomical and physiological characteristics of dermis and subcutis can potentially result in varying protein penetration through the skin, absorption, and metabolism. This review summarizes pharmacokinetic studies that compare the administration of protein therapeutics by subcutaneous injections and different types of microneedles intradermally including hollow, dissolvable, coated, and hydrogel-forming microneedles. Across animal and human studies, hollow microneedle delivery resulted in quicker and higher peak plasma levels of proteins and comparable bioavailability to subcutaneous injections potentially due to the extensive network of lymphatic and blood vessels in the dermis. In case of dissolvable and coated microneedles, drug release kinetics depend on component materials. The dissolution of polymer excipients can slow the release and permeation of protein therapeutics at the administration site and thereby delay absorption. The understanding of drug penetration through different skin layers, its absorption into blood capillaries or lymphatics, and dermal metabolism remains limited. Additionally, the effects of these processes on the differences in pharmacokinetic profiles of proteins following intradermal microneedle administration are not well understood. Greater insights are required for the development of the next generation of intradermal microneedle biotherapeutics.
Collapse
Affiliation(s)
- Laura Koenitz
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland.
| | - Abina Crean
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| | - Sonja Vucen
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
6
|
Ertas YN, Ertas D, Erdem A, Segujja F, Dulchavsky S, Ashammakhi N. Diagnostic, Therapeutic, and Theranostic Multifunctional Microneedles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308479. [PMID: 38385813 DOI: 10.1002/smll.202308479] [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: 09/23/2023] [Revised: 01/04/2024] [Indexed: 02/23/2024]
Abstract
Microneedles (MNs) have maintained their popularity in therapeutic and diagnostic medical applications throughout the past decade. MNs are originally designed to gently puncture the stratum corneum layer of the skin and have lately evolved into intelligent devices with functions including bodily fluid extraction, biosensing, and drug administration. MNs offer limited invasiveness, ease of application, and minimal discomfort. Initially manufactured solely from metals, MNs are now available in polymer-based varieties. MNs can be used to create systems that deliver drugs and chemicals uniformly, collect bodily fluids, and are stimulus-sensitive. Although these advancements are favorable in terms of biocompatibility and production costs, they are insufficient for the therapeutic use of MNs. This is the first comprehensive review that discusses individual MN functions toward the evolution and development of smart and multifunctional MNs for a variety of novel and impactful future applications. The study examines fabrication techniques, application purposes, and experimental details of MN constructs that perform multiple functions concurrently, including sensing, drug-molecule release, sampling, and remote communication capabilities. It is highly likely that in the near future, MN-based smart devices will be a useful and important component of standard medical practice for different applications.
Collapse
Affiliation(s)
- Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, 38039, Türkiye
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, 06800, Türkiye
| | - Derya Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye
| | - Ahmet Erdem
- Department of Biomedical Engineering, Kocaeli University, Umuttepe Campus, Kocaeli, 41380, Türkiye
- Department of Chemistry, Kocaeli University, Umuttepe Campus, Kocaeli, 41380, Türkiye
| | - Farouk Segujja
- Department of Biomedical Engineering, Kocaeli University, Umuttepe Campus, Kocaeli, 41380, Türkiye
| | - Scott Dulchavsky
- Department of Surgery, Henry Ford Health, Detroit, MI, 48201, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Colleges of Engineering and Human Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
7
|
Chang Z, Wu Y, Hu P, Jiang J, Quan G, Wu C, Pan X, Huang Z. The Necessity to Investigate In Vivo Fate of Nanoparticle-Loaded Dissolving Microneedles. Pharmaceutics 2024; 16:286. [PMID: 38399340 PMCID: PMC10892231 DOI: 10.3390/pharmaceutics16020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Transdermal drug delivery systems are rapidly gaining prominence and have found widespread application in the treatment of numerous diseases. However, they encounter the challenge of a low transdermal absorption rate. Microneedles can overcome the stratum corneum barrier to enhance the transdermal absorption rate. Among various types of microneedles, nanoparticle-loaded dissolving microneedles (DMNs) present a unique combination of advantages, leveraging the strengths of DMNs (high payload, good mechanical properties, and easy fabrication) and nanocarriers (satisfactory solubilization capacity and a controlled release profile). Consequently, they hold considerable clinical application potential in the precision medicine era. Despite this promise, no nanoparticle-loaded DMN products have been approved thus far. The lack of understanding regarding their in vivo fate represents a critical bottleneck impeding the clinical translation of relevant products. This review aims to elucidate the current research status of the in vivo fate of nanoparticle-loaded DMNs and elaborate the necessity to investigate the in vivo fate of nanoparticle-loaded DMNs from diverse aspects. Furthermore, it offers insights into potential entry points for research into the in vivo fate of nanoparticle-loaded DMNs, aiming to foster further advancements in this field.
Collapse
Affiliation(s)
- Ziyao Chang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (Y.W.); (X.P.)
| | - Yuhuan Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (Y.W.); (X.P.)
| | - Ping Hu
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Junhuang Jiang
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (Y.W.); (X.P.)
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| |
Collapse
|
8
|
Babalola KT, Arora M, Ganugula R, Agarwal SK, Mohan C, Kumar MNVR. Leveraging Lymphatic System Targeting in Systemic Lupus Erythematosus for Improved Clinical Outcomes. Pharmacol Rev 2024; 76:228-250. [PMID: 38351070 PMCID: PMC10877736 DOI: 10.1124/pharmrev.123.000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 02/16/2024] Open
Abstract
The role of advanced drug delivery strategies in drug repositioning and minimizing drug attrition rates, when applied early in drug discovery, is poised to increase the translational impact of various therapeutic strategies in disease prevention and treatment. In this context, drug delivery to the lymphatic system is gaining prominence not only to improve the systemic bioavailability of various pharmaceutical drugs but also to target certain specific diseases associated with the lymphatic system. Although the role of the lymphatic system in lupus is known, very little is done to target drugs to yield improved clinical benefits. In this review, we discuss recent advances in drug delivery strategies to treat lupus, the various routes of drug administration leading to improved lymph node bioavailability, and the available technologies applied in other areas that can be adapted to lupus treatment. Moreover, this review also presents some recent findings that demonstrate the promise of lymphatic targeting in a preclinical setting, offering renewed hope for certain pharmaceutical drugs that are limited by efficacy in their conventional dosage forms. These findings underscore the potential and feasibility of such lymphatic drug-targeting approaches to enhance therapeutic efficacy in lupus and minimize off-target effects of the pharmaceutical drugs. SIGNIFICANCE STATEMENT: The World Health Organization estimates that there are currently 5 million humans living with some form of lupus. With limited success in lupus drug discovery, turning to effective delivery strategies with existing drug molecules, as well as those in the early stage of discovery, could lead to better clinical outcomes. After all, effective delivery strategies have been proven to improve treatment outcomes.
Collapse
Affiliation(s)
- K T Babalola
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - M Arora
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - R Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - S K Agarwal
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - C Mohan
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - M N V Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
9
|
Ramöller IK, Volpe-Zanutto F, Vora LK, Abbate MTA, Hutton ARJ, McKenna PE, Peng K, Tekko IA, Sabri A, McAlister E, McCarthy HO, Paredes AJ, Donnelly RF. Intradermal delivery of the antiretroviral drugs cabotegravir and rilpivirine by dissolving microarray patches: Investigation of lymphatic uptake. J Control Release 2024; 366:548-566. [PMID: 38211640 DOI: 10.1016/j.jconrel.2024.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
The lymphatic system possesses the main viral replication sites in the body following viral infection. Unfortunately, current antiretroviral agents penetrate the lymph nodes insufficiently when administered orally and, therefore, cannot access the lymphatic system sufficiently to interrupt this viral replication. For this reason, novel drug delivery systems aimed at enhancing the lymphatic uptake of antiretroviral drugs are highly desirable. Dissolving polymeric microarray patches (MAPs) may help to target the lymph intradermally. MAPs are intradermal drug delivery systems used to deliver many types of compounds. The present work describes a novel work investigating the lymphatic uptake of two anti-HIV drugs: cabotegravir (CAB) and rilpivirine (RPV) when delivered intradermally using dissolving MAPs containing nanocrystals of both drugs. Maps were formulated using NCs obtained by solvent-free milling technique. The polymers used to prepare the NCs of both drugs were PVA 10 Kda and PVP 58 Kda. Both NCs were submitted to the lyophilization process and reconstituted with deionized water to form the first layer of drug casting. Backing layers were developed for short application times and effective skin deposition. In vivo biodistribution profiles of RPV and CAB after MAP skin application were investigated and compared with the commercial intramuscular injection using rats. After a single application of RPV MAPs, a higher concentration of RPV was delivered to the axillary lymph nodes (AL) (Cmax 2466 ng/g - Tmax 3 days) when compared with RPV IM injection (18 ng/g - Tmax 1 day), while CAB MAPs delivered slightly lower amounts of drug to the AL (5808 ng/g in 3 days) when compared with CAB IM injection (9225 ng/g in 10 days). However, CAB MAPs delivered 7726 ng/g (Tmax 7 days) to the external lumbar lymph nodes, which was statistically equivalent to IM delivery (Cmax 8282 ng/g - Tmax 7 days). This work provides strong evidence that MAPs were able to enhance the delivery of CAB and RPV to the lymphatic system compared to the IM delivery route.
Collapse
Affiliation(s)
- Inken K Ramöller
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Fabiana Volpe-Zanutto
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom; Faculty of Pharmaceutical Sciences, R. Cândido Portinari, 200 - Cidade Universitária, University of Campinas, Campinas, SP 13083-871, Brazil
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Marco T A Abbate
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Aaron R J Hutton
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Peter E McKenna
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Ke Peng
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Ismaiel A Tekko
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom; Faculty of Pharmacy, Aleppo University, Syria
| | - Akmal Sabri
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Emma McAlister
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Alejandro J Paredes
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom.
| |
Collapse
|
10
|
Nayan MU, Panja S, Sultana A, Zaman LA, Vora LK, Sillman B, Gendelman HE, Edagwa B. Polymer Delivery Systems for Long-Acting Antiretroviral Drugs. Pharmaceutics 2024; 16:183. [PMID: 38399244 PMCID: PMC10892262 DOI: 10.3390/pharmaceutics16020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The success of long-acting (LA) drug delivery systems (DDSs) is linked to their biocompatible polymers. These are used for extended therapeutic release. For treatment or prevention of human immune deficiency virus type one (HIV-1) infection, LA DDSs hold promise for improved regimen adherence and reduced toxicities. Current examples include Cabenuva, Apretude, and Sunlenca. Each is safe and effective. Alternative promising DDSs include implants, prodrugs, vaginal rings, and microarray patches. Each can further meet patients' needs. We posit that the physicochemical properties of the formulation chemical design can optimize drug release profiles. We posit that the strategic design of LA DDS polymers will further improve controlled drug release to simplify dosing schedules and improve regimen adherence.
Collapse
Affiliation(s)
- Mohammad Ullah Nayan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Sudipta Panja
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Ashrafi Sultana
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Lubaba A. Zaman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Lalitkumar K. Vora
- School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK;
| | - Brady Sillman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| |
Collapse
|
11
|
Futaki M, Inamura K, Hashimoto M, Motegi S, Itakura S, Sugibayashi K, Todo H. Effects of Intradermal Administration Volume Using a Hollow Microneedle on the Pharmacokinetics of Fluorescein Isothiocyanate Dextran (M.W. 4,000). Pharm Res 2023; 40:1953-1963. [PMID: 37165148 DOI: 10.1007/s11095-023-03529-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/21/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE Hollow microneedles (hMNs) have been gaining attention as a tool to enable the intradermal (i.d.) administration of pharmaceutical products. However, few reports have examined the effect of administration volume on distribution in the skin and pharmacokinetics parameters after i.d. injection. In the present study, a model middle molecular weight compound, fluorescein isothiocyanate dextran (M.W. 4,000, FD-4), was selected, and blood concentration-time profiles after i.d. and subcutaneous (s.c.) injections with different administration volumes were compared. METHODS FD-4 solution was injected i.d. using a hMN or injected s.c. with a 27 G needle. Pharmacokinetics and dermatokinetics of FD-4 were analyzed using a compartment model. The skin distribution of iodine, as an X ray tracer, was used to evaluate drug disposition. RESULTS With the administered drug assumed to be absorbed from the broad injection site into blood vessels in the upper and lower dermis by rapid (krapid) and slow (kslow) first-order absorption rate constants, respectively, better agreement of observed and theoretical values was obtained. Furthermore, the fraction, F, of the administered dose absorbed with krapid decreased with the increase in injection volume after i.d. injection, although the pharmacokinetics parameters were almost the same regardless of administration volume after s.c. injection. CONCLUSION The drug distribution in the skin may be related to the obtained pharmacokinetics parameters suggested that the number of needles in the MN system and the total administration volume should be considered in designing hMN systems. The present results provide useful information that may support effective drug delivery with hMNs.
Collapse
Affiliation(s)
- Mika Futaki
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Kazuya Inamura
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Miyu Hashimoto
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Shunsaku Motegi
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Shoko Itakura
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Kenji Sugibayashi
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
- Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba-Ken, 283-8555, Japan
| | - Hiroaki Todo
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan.
| |
Collapse
|
12
|
Reyna D, Bejster I, Chadderdon A, Harteg C, Kurnia Anjani Q, Hidayat Bin Sabri A, Brown AN, Drusano GL, Westover J, Bart Tarbet E, Vora LK, Donnelly RF, Lipka E. A five-day treatment course of zanamivir for the flu with a single, self-administered, painless microneedle array patch: Revolutionizing delivery of poorly membrane-permeable therapeutics. Int J Pharm 2023; 641:123081. [PMID: 37230371 PMCID: PMC10347771 DOI: 10.1016/j.ijpharm.2023.123081] [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: 02/17/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Seasonal influenza virus infections cause a substantial number of deaths each year. While zanamivir (ZAN) is efficacious against oseltamivir-resistant influenza strains, the efficacy of the drug is limited by its route of administration, oral inhalation. Herein, we present the development of a hydrogel-forming microneedle array (MA) in combination with ZAN reservoirs for treating seasonal influenza. The MA was fabricated from Gantrez® S-97 crosslinked with PEG 10,000. Various reservoir formulations included ZAN hydrate, ZAN hydrochloric acid (HCl), CarraDres™, gelatin, trehalose, and/or alginate. In vitro permeation studies with a lyophilized reservoir consisting of ZAN HCl, gelatin, and trehalose resulted in rapid and high delivery of up to 33 mg of ZAN across the skin with delivery efficiency of up to ≈75% by 24 h. Pharmacokinetics studies in rats and pigs demonstrated that a single administration of a MA in combination with a CarraDres™ ZAN HCl reservoir offered a simple and minimally invasive delivery of ZAN into the systemic circulation. In pigs, efficacious plasma and lung steady-state levels of ∼120 ng/mL were reached within 2 h and sustained between 50 and 250 ng/mL over 5 days. MA-enabled delivery of ZAN could enable a larger number of patients to be reached during an influenza outbreak.
Collapse
Affiliation(s)
- Dawn Reyna
- TSRL, Inc., 540 Avis Dr., Suite A, Ann Arbor, MI 48108, USA
| | - Ian Bejster
- TSRL, Inc., 540 Avis Dr., Suite A, Ann Arbor, MI 48108, USA
| | | | - Cheryl Harteg
- TSRL, Inc., 540 Avis Dr., Suite A, Ann Arbor, MI 48108, USA
| | | | | | - Ashley N Brown
- College of Medicine, University of Florida, Orlando, FL 32827, USA
| | - George L Drusano
- College of Medicine, University of Florida, Orlando, FL 32827, USA
| | - Jonna Westover
- Institute for Antiviral Research, Utah State University, 5600 Old Main Hill, Logan, Utah 84322, USA
| | - E Bart Tarbet
- Institute for Antiviral Research, Utah State University, 5600 Old Main Hill, Logan, Utah 84322, USA
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Elke Lipka
- TSRL, Inc., 540 Avis Dr., Suite A, Ann Arbor, MI 48108, USA.
| |
Collapse
|
13
|
McNamee M, Wong S, Guy O, Sharma S. Microneedle technology for potential SARS-CoV-2 vaccine delivery. Expert Opin Drug Deliv 2023:1-16. [PMID: 37128730 DOI: 10.1080/17425247.2023.2209718] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
INTRODUCTION Microneedle fabrication was conceptualised in the 1970s as devices for painless transdermal drug delivery. The last two decades have seen considerable research and financial investment in this area with SARS-CoV-2 and other vaccines catalysing their application to in vivo intradermal vaccine delivery. Microneedle arrays have been fabricated in different shapes, geometries, formats, and out of different materials. AREAS COVERED The recent pandemic has offered microneedle platforms the opportunity to be employed as a vehicle for SARS-CoV-2 vaccine administration. The various modes of vaccination delivery and the potential of microneedle arrays-based vaccines will be presented, with a specific focus placed on recent SARS-CoV-2 research. The advantages of microneedle-based vaccine administration, in addition to the major hurdles to their en masse implementation, will be examined. EXPERT OPINION Considering the widely acknowledged disadvantages of current vaccine delivery, such as anxiety, pain, and the requirement for professional administration, a large shift in this research sphere is imminent. The SARS-CoV-2 pandemic has catalysed the development of alternate vaccination platforms, working to avoid the requirement for mass vaccination centres. As microneedle vaccine patches are transitioning through clinical study phases, research will be required to ready this technology for a more mass production environment.
Collapse
Affiliation(s)
- Megan McNamee
- School of Engineering and Applied Sciences, Faculty of Science and Engineering , Fabian Way, Bay Campus, Swansea University, Swansea SA1 8EN, UK
| | - Shuyi Wong
- School of Engineering and Applied Sciences, Faculty of Science and Engineering , Fabian Way, Bay Campus, Swansea University, Swansea SA1 8EN, UK
| | - Owen Guy
- School of Engineering and Applied Sciences, Faculty of Science and Engineering , Fabian Way, Bay Campus, Swansea University, Swansea SA1 8EN, UK
| | - Sanjiv Sharma
- School of Engineering and Applied Sciences, Faculty of Science and Engineering , Fabian Way, Bay Campus, Swansea University, Swansea SA1 8EN, UK
| |
Collapse
|
14
|
Nguyen HX, Nguyen CN. Microneedle-Mediated Transdermal Delivery of Biopharmaceuticals. Pharmaceutics 2023; 15:277. [PMID: 36678906 PMCID: PMC9864466 DOI: 10.3390/pharmaceutics15010277] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Transdermal delivery provides numerous benefits over conventional routes of administration. However, this strategy is generally limited to a few molecules with specific physicochemical properties (low molecular weight, high potency, and moderate lipophilicity) due to the barrier function of the stratum corneum layer. Researchers have developed several physical enhancement techniques to expand the applications of the transdermal field; among these, microneedle technology has recently emerged as a promising platform to deliver therapeutic agents of any size into and across the skin. Typically, hydrophilic biomolecules cannot penetrate the skin by passive diffusion. Microneedle insertion disrupts skin integrity and compromises its protective function, thus creating pathways (microchannels) for enhanced permeation of macromolecules. Microneedles not only improve stability but also enhance skin delivery of various biomolecules. Academic institutions and industrial companies have invested substantial resources in the development of microneedle systems for biopharmaceutical delivery. This review article summarizes the most recent research to provide a comprehensive discussion about microneedle-mediated delivery of macromolecules, covering various topics from the introduction of the skin, transdermal delivery, microneedles, and biopharmaceuticals (current status, conventional administration, and stability issues), to different microneedle types, clinical trials, safety and acceptability of microneedles, manufacturing and regulatory issues, and the future of microneedle technology.
Collapse
Affiliation(s)
- Hiep X. Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Chien N. Nguyen
- National Institute of Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi 100000, Vietnam
- Faculty of Pharmaceutics and Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi 100000, Vietnam
| |
Collapse
|
15
|
Grabowski T, Derlacz R, Burmańczuk A. Göttingen pigs as a potential model for natalizumab pharmacokinetics, pharmacodynamics, and immunogenicity evaluation. Biomed Pharmacother 2022; 156:113926. [DOI: 10.1016/j.biopha.2022.113926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
|
16
|
Khadria A, Paavola CD, Maslov K, Valenzuela FA, Sperry AE, Cox AL, Cao R, Shi J, Brown-Augsburger PL, Lozano E, Blankenship RL, Majumdar R, Bradley SA, Beals JM, Oladipupo SS, Wang LV. Photoacoustic imaging reveals mechanisms of rapid-acting insulin formulations dynamics at the injection site. Mol Metab 2022; 62:101522. [PMID: 35671972 PMCID: PMC9207296 DOI: 10.1016/j.molmet.2022.101522] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Ultra-rapid insulin formulations control postprandial hyperglycemia; however, inadequate understanding of injection site absorption mechanisms is limiting further advancement. We used photoacoustic imaging to investigate the injection site dynamics of dye-labeled insulin lispro in the Humalog® and Lyumjev® formulations using the murine ear cutaneous model and correlated it with results from unlabeled insulin lispro in pig subcutaneous injection model. METHODS We employed dual-wavelength optical-resolution photoacoustic microscopy to study the absorption and diffusion of the near-infrared dye-labeled insulin lispro in the Humalog and Lyumjev formulations in mouse ears. We mathematically modeled the experimental data to calculate the absorption rate constants and diffusion coefficients. We studied the pharmacokinetics of the unlabeled insulin lispro in both the Humalog and Lyumjev formulations as well as a formulation lacking both the zinc and phenolic preservative in pigs. The association state of insulin lispro in each of the formulations was characterized using SV-AUC and NMR spectroscopy. RESULTS Through experiments using murine and swine models, we show that the hexamer dissociation rate of insulin lispro is not the absorption rate-limiting step. We demonstrated that the excipients in the Lyumjev formulation produce local tissue expansion and speed both insulin diffusion and microvascular absorption. We also show that the diffusion of insulin lispro at the injection site drives its initial absorption; however, the rate at which the insulin lispro crosses the blood vessels is its overall absorption rate-limiting step. CONCLUSIONS This study provides insights into injection site dynamics of insulin lispro and the impact of formulation excipients. It also demonstrates photoacoustic microscopy as a promising tool for studying protein therapeutics. The results from this study address critical questions around the subcutaneous behavior of insulin lispro and the formulation excipients, which could be useful to make faster and better controlled insulin formulations in the future.
Collapse
Affiliation(s)
- Anjul Khadria
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Chad D Paavola
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Konstantin Maslov
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Francisco A Valenzuela
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Andrea E Sperry
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Amy L Cox
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Rui Cao
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Junhui Shi
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Emmanuel Lozano
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ross L Blankenship
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ranajoy Majumdar
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Scott A Bradley
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - John M Beals
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Biotechnology Center, San Diego, CA, 92121, USA.
| | - Sunday S Oladipupo
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| | - Lihong V Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA; Caltech Optical Imaging Laboratory, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
17
|
Russell PS, Velivolu R, Maldonado Zimbrón VE, Hong J, Kavianinia I, Hickey AJR, Windsor JA, Phillips ARJ. Fluorescent Tracers for In Vivo Imaging of Lymphatic Targets. Front Pharmacol 2022; 13:952581. [PMID: 35935839 PMCID: PMC9355481 DOI: 10.3389/fphar.2022.952581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
The lymphatic system continues to gain importance in a range of conditions, and therefore, imaging of lymphatic vessels is becoming more widespread for research, diagnosis, and treatment. Fluorescent lymphatic imaging offers advantages over other methods in that it is affordable, has higher resolution, and does not require radiation exposure. However, because the lymphatic system is a one-way drainage system, the successful delivery of fluorescent tracers to lymphatic vessels represents a unique challenge. Each fluorescent tracer used for lymphatic imaging has distinct characteristics, including size, shape, charge, weight, conjugates, excitation/emission wavelength, stability, and quantum yield. These characteristics in combination with the properties of the target tissue affect the uptake of the dye into lymphatic vessels and the fluorescence quality. Here, we review the characteristics of visible wavelength and near-infrared fluorescent tracers used for in vivo lymphatic imaging and describe the various techniques used to specifically target them to lymphatic vessels for high-quality lymphatic imaging in both clinical and pre-clinical applications. We also discuss potential areas of future research to improve the lymphatic fluorescent tracer design.
Collapse
Affiliation(s)
- P. S. Russell
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - R. Velivolu
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - V. E. Maldonado Zimbrón
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - J. Hong
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - I. Kavianinia
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
- School of Chemical Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - A. J. R. Hickey
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - J. A. Windsor
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - A. R. J. Phillips
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
Jet injectors: Perspectives for small volume delivery with lasers. Adv Drug Deliv Rev 2022; 182:114109. [PMID: 34998902 DOI: 10.1016/j.addr.2021.114109] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/16/2021] [Accepted: 12/29/2021] [Indexed: 12/27/2022]
Abstract
Needle-free jet injectors have been proposed as an alternative to injections with hypodermic needles. Currently, a handful of commercial needle-free jet injectors already exist. However, these injectors are designed for specific injections, typically limited to large injection volumes into the deeper layers beneath the skin. There is growing evidence of advantages when delivering small volumes into the superficial skin layers, namely the epidermis and dermis. Injections such as vaccines and insulin would benefit from delivery into these superficial layers. Furthermore, the same technology for small volume needle-free injections can serve (medical) tattooing as well as other personalized medicine treatments. The research dedicated to needle-free jet injectors actuated by laser energy has increased in the last decade. In this case, the absorption of the optical energy by the liquid results in an explosively growing bubble. This bubble displaces the rest of the liquid, resulting in a fast microfluidic jet which can penetrate the skin. This technique allows for precise control over volumes (pL to µL) and penetration depths (µm to mm). Furthermore, these injections can be tuned without changing the device, by varying parameters such as laser power, beam diameter and filling level of the liquid container. Despite the published research on the working principles and capabilities of individual laser-actuated jet injectors, a thorough overview encompassing all of them is lacking. In this perspective, we will discuss the current status of laser-based jet injectors and contrast their advantages and limitations, as well as their potential and challenges.
Collapse
|
19
|
Ono N, Iibuchi T, Todo H, Itakura S, Adachi H, Sugibayashi K. Enhancement of skin permeation of fluorescein isothiocyanate-dextran 4 kDa (FD4) and insulin by thermalporation. Eur J Pharm Sci 2021; 170:106096. [PMID: 34929301 DOI: 10.1016/j.ejps.2021.106096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/18/2021] [Accepted: 12/09/2021] [Indexed: 11/03/2022]
Abstract
Thermalporation has gained attention as a physical means to enhance skin permeation by creating micropores in the primary skin barrier, stratum corneum, which allows much higher permeation of middle and high molecular weight biopharmaceuticals. In the present study, a PassPort® system (PS) was used as a thermalporation device, and the obtained change in permeation resistance of drugs was evaluated using a parallel skin permeation-resistance model. In addition, the blood concentration-time profile after topical application of insulin was also investigated with the PS treatment. Fluorescein isothiocyanate-dextran (FD-4) and insulin were used as model middle molecular weight drugs. Micropores created by the PS treatment were measured using an optical microscope. An in vitro skin permeation and an in vivo pharmacokinetics experiments were done with FD-4 and insulin, respectively. Barrier function recovery after the PS treatment was evaluated with changes in the electrical skin resistance. About 960-fold higher skin permeation of FD-4 was observed by PSs treatment (4 milliseconds (ms), 200 micropores/cm2). A gradually increased blood concentration of insulin was observed by the PSs treatment, and the relative bioavailability of insulin was 21.1% compared with subcutaneous injection. Skin resistance value was dramatically decreased immediately after the PS treatment, but its value was turned into the initial one by 12 h. The thermalporation is effective for improving skin permeation of FD-4 and transdermal absorption of insulin. These results suggested that the PS treatment may be utilized to increase the skin permeation of topically applied FD-4 and insulin.
Collapse
Affiliation(s)
- Naoto Ono
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Tomoya Iibuchi
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Hiroaki Todo
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan.
| | - Shoko Itakura
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Hirotoshi Adachi
- PassPort Technologies, Inc., 5580 Morehouse Drive, Suite 120, San Diego, CA 92121, USA
| | - Kenji Sugibayashi
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| |
Collapse
|
20
|
Transdermal Drug Delivery in the Pig Skin. Pharmaceutics 2021; 13:pharmaceutics13122016. [PMID: 34959299 PMCID: PMC8707795 DOI: 10.3390/pharmaceutics13122016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/04/2022] Open
Abstract
Transdermal delivery can be accomplished through various mechanisms including formulation optimization, epidermal stratum corneum barrier disruption, or directly by removing the stratum corneum layer. Microneedling, electroporation, a combination of both and also the intradermal injection known as mesotherapy have proved efficacy in epidermal-barrier disruption. Here we analyzed the effects of these methods of epidermal-barrier disruption in the structure of the skin and the absorption of four compounds with different characteristics and properties (ketoprofen, biotin, caffein, and procaine). Swine skin (Pietrain x Durox) was used as a human analogue, both having similar structure and pharmacological release. They were biopsied at different intervals, up to 2 weeks after application. High-pressure liquid chromatography and brightfield microscopy were performed, conducting a biometric analysis and measuring histological structure and vascular status. The performed experiments led to different results in the function of the studied molecules: ketoprofen and biotin had the best concentrations with intradermal injections, while delivery methods for obtaining procaine and caffein maximum concentrations changed on the basis of the lapsed time. The studied techniques did not produce significant histological alterations after their application, except for an observed increase in Langerhans cells and melanocytes after applying electroporation, and an epidermal thinning after using microneedles, with variable results regarding dermal thickness. Although all the studied barrier disruptors can accomplish transdermal delivery, the best disruptor is dependent on the particular molecule.
Collapse
|
21
|
Grabowski T, Burmańczuk A, Derlacz R, Stefaniak T, Rząsa A, Borkowski J. Ustekinumab pharmacokinetics after subcutaneous administration in swine model. J Vet Sci 2021; 22:e47. [PMID: 34423596 PMCID: PMC8460463 DOI: 10.4142/jvs.2021.22.e47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/22/2021] [Accepted: 05/23/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Due to multiple similarities in the structure and physiology of human and pig skin, the pig model is extremely useful for biological drug testing after subcutaneous administration. Knowledge of the differences between subcutaneous injection sites could have a significant impact on the absorption phase and pharmacokinetic profiles of biological drugs. OBJECTIVES This study aimed to analyze the impact of administration site on pharmacokinetics and selected biochemical and hematological parameters after a single subcutaneous administration of ustekinumab in pigs. Drug concentrations in blood plasma were analyzed by enzyme-linked immunosorbent assay. Pharmacokinetic analyses were performed based on raw data using Phoenix WinNonlin 8.1 software and ThothPro v 4.1. METHODS The study included 12 healthy, female, large white piglets. Each group received a single dose of ustekinumab given as a 1 mg/kg subcutaneous injection into the internal part of the inguinal fold or the external part of the inguinal fold. RESULTS The differences in absorption rate between the internal and external parts of the inguinal fold were not significant. However, the time of maximal concentration, clearance, area under the curve calculated between zero and mean residence time and mean residence time between groups were substantially different (p > 0.05). The relative bioavailability after administration of ustekinumab into the external part of the inguinal fold was 40.36% lower than after administration of ustekinumab into the internal part of the inguinal fold. CONCLUSIONS Healthy breeding pigs are a relevant model to study the pharmacokinetic profile of subcutaneously administered ustekinumab.
Collapse
Affiliation(s)
| | - Artur Burmańczuk
- Sub-Department of Pharmacology, Toxicology and Environmental Protection, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland.
| | | | - Tadeusz Stefaniak
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wrocław, Poland
| | - Anna Rząsa
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wrocław, Poland
| | - Jacek Borkowski
- Department of Physiology and Biochemistry, University School of Physical Education in Wroclaw, 51-612 Wrocław, Poland
| |
Collapse
|
22
|
Tessier N, Moawad F, Amri N, Brambilla D, Martel C. Focus on the Lymphatic Route to Optimize Drug Delivery in Cardiovascular Medicine. Pharmaceutics 2021; 13:1200. [PMID: 34452161 PMCID: PMC8398144 DOI: 10.3390/pharmaceutics13081200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022] Open
Abstract
While oral agents have been the gold standard for cardiovascular disease therapy, the new generation of treatments is switching to other administration options that offer reduced dosing frequency and more efficacy. The lymphatic network is a unidirectional and low-pressure vascular system that is responsible for the absorption of interstitial fluids, molecules, and cells from the peripheral tissue, including the skin and the intestines. Targeting the lymphatic route for drug delivery employing traditional or new technologies and drug formulations is exponentially gaining attention in the quest to avoid the hepatic first-pass effect. The present review will give an overview of the current knowledge on the involvement of the lymphatic vessels in drug delivery in the context of cardiovascular disease.
Collapse
Affiliation(s)
- Nolwenn Tessier
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Montreal Heart Institute Research Center, Montreal, QC H1T 1C8, Canada
| | - Fatma Moawad
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Pharmaceutics, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Nada Amri
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Montreal Heart Institute Research Center, Montreal, QC H1T 1C8, Canada
| | - Davide Brambilla
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Montreal Heart Institute Research Center, Montreal, QC H1T 1C8, Canada
| |
Collapse
|
23
|
Qin L, Zhang H, Zhou Y, Umeshappa CS, Gao H. Nanovaccine-Based Strategies to Overcome Challenges in the Whole Vaccination Cascade for Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006000. [PMID: 33768693 DOI: 10.1002/smll.202006000] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/26/2020] [Indexed: 06/12/2023]
Abstract
Nanovaccine-based immunotherapy (NBI) has received greater attention recently for its potential to prime tumor-specific immunity and establish a long-term immune memory that prevents tumor recurrence. Despite encouraging results in the recent studies, there are still numerous challenges to be tackled for eliciting potent antitumor immunity using NBI strategies. Based on the principles that govern immune response, here it is proposed that these challenges need to be addressed at the five critical cascading events: Loading tumor-specific antigens by nanoscale drug delivery systems (L); Draining tumor antigens to lymph nodes (D); Internalization by dendritic cells (DCs) (I); Maturation of DCs by costimulatory signaling (M); and Presenting tumor-peptide-major histocompatibility complexes to T cells (P) (LDIMP cascade in short). This review provides a detailed and objective overview of emerging NBI strategies to improve the efficacy of nanovaccines in each step of the LDIMP cascade. It is concluded that the balance between each step must be optimized by delicate designing and modification of nanovaccines and by combining with complementary approaches to provide a synergistic immunity in the fight against cancer.
Collapse
Affiliation(s)
- Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Chongqing Vocational College of Transportation, Chongqing, 400715, China
| | - Huilin Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Channakeshava Sokke Umeshappa
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
24
|
Abstract
The current situation, heavily influenced by the ongoing pandemic, puts vaccines back into the spotlight. However, the conventional and traditional vaccines present disadvantages, particularly related to immunogenicity, stability, and storage of the final product. Often, such products require the maintenance of a “cold chain,” impacting the costs, the availability, and the distribution of vaccines. Here, after a recall of the mode of action of vaccines and the types of vaccines currently available, we analyze the past, present, and future of vaccine formulation. The past focuses on conventional formulations, the present discusses the use of nanoparticles for vaccine delivery and as adjuvants, while the future presents microneedle patches as alternative formulation and administration route. Finally, we compare the advantages and disadvantages of injectable solutions, nanovaccines, and microneedles in terms of efficacy, stability, and patient-friendly design. Different approaches to vaccine formulation development, the conventional vaccine formulations from the past, the current development of lipid nanoparticles as vaccines, and the near future microneedles formulations are discussed in this review. ![]()
Collapse
|
25
|
Vora LK, Moffatt K, Tekko IA, Paredes AJ, Volpe-Zanutto F, Mishra D, Peng K, Raj Singh Thakur R, Donnelly RF. Microneedle array systems for long-acting drug delivery. Eur J Pharm Biopharm 2021; 159:44-76. [DOI: 10.1016/j.ejpb.2020.12.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/25/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022]
|
26
|
Permana AD, Nainu F, Moffatt K, Larrañeta E, Donnelly RF. Recent advances in combination of microneedles and nanomedicines for lymphatic targeted drug delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1690. [PMID: 33401339 DOI: 10.1002/wnan.1690] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Numerous diseases have been reported to affect the lymphatic system. As such, several strategies have been developed to deliver chemotherapeutics to this specific network of tissues and associated organs. Nanotechnology has been exploited as one of the main approaches to improve the lymphatic uptake of drugs. Different nanoparticle approaches utilized for both active and passive targeting of the lymphatic system are discussed here. Specifically, due to the rich abundance of lymphatic capillaries in the dermis, particular attention is given to this route of administration, as intradermal administration could potentially result in higher lymphatic uptake compared to other routes of administration. Recently, progress in microneedle research has attracted particular attention as an alternative for the use of conventional hypodermic injections. The benefits of microneedles, when compared to intradermal injection, are subsequently highlighted. Importantly, microneedles exhibit particular benefit in relation to therapeutic targeting of the lymphatic system, especially when combined with nanoparticles, which are further discussed. However, despite the apparent benefits provided by this combination approach, further comprehensive preclinical and clinical studies are now necessary to realize the potential extent of this dual-delivery platform, further taking into consideration eventual usability and acceptability in the intended patient end-users. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Kurtis Moffatt
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | | | | |
Collapse
|
27
|
Jana BA, Shinde U, Wadhwani A. Synthetic enzyme-based nanoparticles act as smart catalyst for glucose responsive release of insulin. J Biotechnol 2020; 324:1-6. [PMID: 32987063 DOI: 10.1016/j.jbiotec.2020.09.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Earlier studies have attempted to create electronic free insulin delivery systems using different glucose sensing mechanism, no successful clinical translation as hitherto been made. This study aimed to assess the faster responsiveness of the insulin release from this enzyme based nanoparticles which is a self-regulated insulin delivery system constructed by loading with insulin, enzyme glucose oxidase into hyaluronic acid and 2-nitroimidazole forming enzyme-based nanoparticles which works in accordance to the blood glucose level. MATERIALS AND METHOD Enzyme-based nanoparticles were prepared by ionic gelation method. Insulin content in the nanoparticles kept for stability study was estimated by human insulin enzyme based immunosorbent assay. In in-vitro studies; different concentrations of glucose were taken and the release study of insulin was recorded. RESULTS This enzyme-based nanoparticles were having average diameter of around 193 nm and stability studies showed that nanoparticles were stable upto 30 days at 4 °C. In-vitro studies showed the release of insulin from nanoparticle conjugates which was effectively correlated with the external glucose concentration created where different concentrations of glucose taken thus facilitating the stabilization of blood glucose levels in the hyperglycemia state which was achieved within 10 min. (400 mg/dL) wherein drug release rate remarkably increased in hyperglycemia state and no specific changes or small amount of release was observed in normoglycemia state (100 mg/dL). CONCLUSION Overall, this preliminary study of this enzyme-based nanoparticles formulation showed excellent rapid responsiveness towards hyperglycemia which might act as a potential biomimetic system in triggering the release of insulin in sustained manner.
Collapse
Affiliation(s)
- Baishali A Jana
- Department of Pharmaceutical Biotechnology, JSS Academy of Higher Education & Research - JSS College of Pharmacy, Ooty, 643001, Tamil Nadu, India.
| | - Ujwala Shinde
- Department of Pharmaceutics, Bombay College of Pharmacy, Mumbai, 400098, Maharashtra, India.
| | - Ashish Wadhwani
- Department of Pharmaceutical Biotechnology, JSS Academy of Higher Education & Research - JSS College of Pharmacy, Ooty, 643001, Tamil Nadu, India.
| |
Collapse
|
28
|
Long LY, Zhang J, Yang Z, Guo Y, Hu X, Wang Y. Transdermal delivery of peptide and protein drugs: Strategies, advantages and disadvantages. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Cao J, Su J, An M, Yang Y, Zhang Y, Zuo J, Zhang N, Zhao Y. Novel DEK-Targeting Aptamer Delivered by a Hydrogel Microneedle Attenuates Collagen-Induced Arthritis. Mol Pharm 2020; 18:305-316. [PMID: 33253580 DOI: 10.1021/acs.molpharmaceut.0c00954] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
DEK protein is critical to the formation of neutrophil extracellular traps (NETs) in rheumatoid arthritis (RA). Blocking DEK using the aptamer DTA via articular injection has been shown to have robust anti-inflammatory efficacy in a previous study. However, DTA is prone to nuclease degradation and renal clearance in vivo. RA is a systemic disease that involves multiple joints, and local injection is impractical in clinical settings. In this study, DTA was modified with methoxy groups on all deoxyribose sugar units and inverted deoxythymidine on the 3' end (DTA4) to enhance its stability against nuclease. DTA4 is stable for 72 h in 90% mouse serum and maintains a high binding affinity to DEK. DTA4 effectively inhibits the formation of NETs and the migration of HUVECs in vitro. DTA4 was then modified with cholesterol on its 5' end to form DTA6. DTA6 dramatically reduces DEK expression in inflammatory RAW264.7 cells. A hydrogel microneedle (hMN) was then fabricated for the transdermal delivery of DTA6. The hMN maintains morphological integrity after absorbing the aptamer solution, effectively pierces the skin, and rapidly releases DTA6 into the dermis. The DTA6-loaded hMN significantly attenuates inflammation and protects joints from cartilage/bone erosion in collagen-induced arthritis (CIA) mice.
Collapse
Affiliation(s)
- Jian Cao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Jingjing Su
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Mengchen An
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Yang Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Yi Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Jing Zuo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou 450001, Henan, P. R. China.,Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Henan Province, Zhengzhou 450001, Henan, P. R. China
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou 450001, Henan, P. R. China.,Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Henan Province, Zhengzhou 450001, Henan, P. R. China
| |
Collapse
|
30
|
Li Z, Biswas A, Finkelstein J, Grein S, Kapoor Y, Milewski M, Queisser G. Modeling Drug Absorption from the Dermis after an Injection. J Pharm Sci 2020; 110:1279-1291.e1. [PMID: 33248056 DOI: 10.1016/j.xphs.2020.10.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 11/19/2022]
Abstract
A dermal absorption model for small and macromolecules was previously proposed by Ibrahim et al. This model estimated absorption of therapeutics from the dermal tissue based on their molecular size and protein binding through blood and lymphatics. Blood absorption followed a two-pore theory and the lymphatic absorption was limited by the constant lymphatic flow rate. Current work builds on this steady-state concept by modeling the absorption from the dermis immediately after an injection is given (unsteady state). An injection in the dermis creates a localized pressure gradient which resolves itself over time. This phenomenon is captured in the model to estimate the impact of injection volume on the absorption rate constant. Blood absorption follows the two-pore theory but is time-dependent and the lymphatic absorption is determined based on valve opening and pressure driven convective flow, returning to steady-state as the molecule is absorbed. A direct comparison of the steady-state analysis, experimental data and the current model is made. The results indicate that accounting for the localized time-varying pressure can better predict the experimental absorption rate constants. This work significantly improves the existing understanding of macromolecule uptake from the interstitial fluid following intradermal injection.
Collapse
Affiliation(s)
- Zhi Li
- Temple University, Department of Mathematics, 1805 N Broad Street, Philadelphia, PA 19122, USA
| | - Abhijit Biswas
- Temple University, Department of Mathematics, 1805 N Broad Street, Philadelphia, PA 19122, USA
| | - Joshua Finkelstein
- Temple University, Department of Mathematics, 1805 N Broad Street, Philadelphia, PA 19122, USA
| | - Stephan Grein
- Temple University, Department of Mathematics, 1805 N Broad Street, Philadelphia, PA 19122, USA
| | - Yash Kapoor
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, 07033, USA.
| | - Mikolaj Milewski
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, 07033, USA.
| | - Gillian Queisser
- Temple University, Department of Mathematics, 1805 N Broad Street, Philadelphia, PA 19122, USA.
| |
Collapse
|
31
|
Zero-order drug delivery: State of the art and future prospects. J Control Release 2020; 327:834-856. [PMID: 32931897 DOI: 10.1016/j.jconrel.2020.09.020] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/21/2023]
Abstract
Pharmaceutical drugs are an important part of the global healthcare system, with some estimates suggesting over 50% of the world's population takes at least one medication per day. Most drugs are delivered as immediate-release formulations that lead to a rapid increase in systemic drug concentration. Although these formulations have historically played an important role, they can be limited by poor patient compliance, adverse side effects, low bioavailability, or undesirable pharmacokinetics. Drug delivery systems featuring first-order release kinetics have been able to improve pharmacokinetics but are not ideal for drugs with short biological half-lives or small therapeutic windows. Zero-order drug delivery systems have the potential to overcome the issues facing immediate-release and first-order systems by releasing drug at a constant rate, thereby maintaining drug concentrations within the therapeutic window for an extended period of time. This release profile can be used to limit adverse side effects, reduce dosing frequency, and potentially improve patient compliance. This review covers strategies being employed to attain zero-order release or alter traditionally first-order release kinetics to achieve more consistent release before discussing opportunities for improving device performance based on emerging materials and fabrication methods.
Collapse
|
32
|
Abdallah M, Müllertz OO, Styles IK, Mörsdorf A, Quinn JF, Whittaker MR, Trevaskis NL. Lymphatic targeting by albumin-hitchhiking: Applications and optimisation. J Control Release 2020; 327:117-128. [PMID: 32771478 DOI: 10.1016/j.jconrel.2020.07.046] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
The lymphatic system plays an integral role in the development and progression of a range of disease conditions, which has impelled medical researchers and clinicians to design, develop and utilize advanced lymphatic drug delivery systems. Following interstitial administration, most therapeutics and molecules are cleared from tissues via the draining blood capillaries. Macromolecules and delivery systems >20 kDa in size or 10-100 nm in diameter are, however, transported from the interstitium via draining lymphatic vessels as they are too large to cross the blood capillary endothelium. Lymphatic uptake of small molecules can be promoted by two general approaches: administration in association with synthetic macromolecular constructs, or through hitchhiking on endogenous cells or macromolecular carriers that are transported from tissues via the lymphatics. In this paper we review the latter approach where molecules are targeted to lymph by hitchhiking on endogenous albumin transport pathways after subcutaneous, intramuscular or intradermal injection. We describe the properties of the lymphatic system and albumin that are relevant to lymphatic targeting, the characteristics of drugs and delivery systems designed to hitchhike on albumin trafficking pathways and how to further optimise these properties, and finally the current applications and potential future directions for albumin-hitchhiking approaches to target the lymphatics.
Collapse
Affiliation(s)
- Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Olivia O Müllertz
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia
| | - Alexander Mörsdorf
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - John F Quinn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Michael R Whittaker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia.
| |
Collapse
|
33
|
Microneedle Mediated Transdermal Delivery of Protein, Peptide and Antibody Based Therapeutics: Current Status and Future Considerations. Pharm Res 2020; 37:117. [PMID: 32488611 PMCID: PMC7266419 DOI: 10.1007/s11095-020-02844-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/21/2020] [Indexed: 02/08/2023]
Abstract
The success of protein, peptide and antibody based therapies is evident - the biopharmaceuticals market is predicted to reach $388 billion by 2024 [1], and more than half of the current top 20 blockbuster drugs are biopharmaceuticals. However, the intrinsic properties of biopharmaceuticals has restricted the routes available for successful drug delivery. While providing 100% bioavailability, the intravenous route is often associated with pain and needle phobia from a patient perspective, which may translate as a reluctance to receive necessary treatment. Several non-invasive strategies have since emerged to overcome these limitations. One such strategy involves the use of microneedles (MNs), which are able to painlessly penetrate the stratum corneum barrier to dramatically increase transdermal drug delivery of numerous drugs. This review reports the wealth of studies that aim to enhance transdermal delivery of biopharmaceutics using MNs. The true potential of MNs as a drug delivery device for biopharmaceuticals will not only rely on acceptance from prescribers, patients and the regulatory authorities, but the ability to upscale MN manufacture in a cost-effective manner and the long term safety of MN application. Thus, the current barriers to clinical translation of MNs, and how these barriers may be overcome are also discussed.
Collapse
|
34
|
Preparation of enzyme based polymeric biomimetic nanoparticle for the controlled release of insulin. SENSING AND BIO-SENSING RESEARCH 2020. [DOI: 10.1016/j.sbsr.2020.100342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
35
|
Chen SX, Ma M, Xue F, Shen S, Chen Q, Kuang Y, Liang K, Wang X, Chen H. Construction of microneedle-assisted co-delivery platform and its combining photodynamic/immunotherapy. J Control Release 2020; 324:218-227. [PMID: 32387551 DOI: 10.1016/j.jconrel.2020.05.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
Despite advances in photodynamic therapy (PDT) for treating superficial tumor, the prospect of this monotherapy remains challenges in the context of systemic phototoxicity and poor efficacy. In this work, a physiologically self-degradable microneedle (MN)-assisted platform is developed for combining PDT and immunotherapy via controlled co-delivery of photosensitizer (PS) and checkpoint inhibitor anti-CTLA4 antibody (aCTLA4), which generates synergistic reinforcement outcome while reducing side effects. MN is composed of biocompatible hyaluronic acid integrated with the pH-sensitive dextran nanoparticles, which is fabricated to simultaneously encapsulate hydrophobic (Zinc Phthalocyanine) and hydrophilic agents (aCTLA4) via a double emulsion method. This co-loading carrier can aggregate effectively around topical tumor by microneedle-assisted transdermal delivery. In vivo studies using 4T1 mouse models, PDT firstly exerts its effect to killing tumor and triggers the immune responses, subsequently, facilitating the immunotherapy with immune checkpoint inhibitor (aCTLA4). The possible mechanism and systemic effects of the combined therapy are investigated, which demonstrate that this co-administration platform can be a promising tool for focal cancer treatment.
Collapse
Affiliation(s)
- Shi-Xiong Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, d, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ming Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, d, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Fengfeng Xue
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, d, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shuzhan Shen
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, PR China
| | - Qian Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, d, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yichen Kuang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, d, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Kaicheng Liang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, d, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, PR China
| | - Hangrong Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, d, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
36
|
Hutton ARJ, McCrudden MTC, Larrañeta E, Donnelly RF. Influence of molecular weight on transdermal delivery of model macromolecules using hydrogel-forming microneedles: potential to enhance the administration of novel low molecular weight biotherapeutics. J Mater Chem B 2020; 8:4202-4209. [PMID: 32292995 DOI: 10.1039/d0tb00021c] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
With a view to improve the current monoclonal antibody-based therapies dominating the pharmaceutical market, low molecular weight (MW) protein-based macromolecules, such as recombinant antibody fragments, typically within the range of 10-70 kDa, have been developed. Previously, our group successfully delivered Avastin®, a monoclonal antibody (mAb) across the skin using hydrogel-forming microneedles (MN). However, it is thought that this delivery system can be further enhanced using novel, lower MW biomolecules. To address this perception, in the current study, FITC-dextran of different MWs (10, 70 and 150 kDa) was used to model the transdermal delivery of low MW biotherapeutics and mAbs with MWs of approximately 150 kDa. Conversely, fluorescein sodium was the compound selected to model hydrophilic, low MW drugs. As expected, fluorescein sodium produced the greatest cumulative permeation (637.4 ± 42.69 μg). The amounts of FITC-dextran 10 kDa and 150 kDa which permeated across neonatal porcine skin in vitro were 462.17 ± 65.85 μg and 213.54 ± 15.19 μg after 24 h, respectively. The results collated here suggest that the delivery of emerging novel biotherapeutics, via'super swelling' hydrogel-forming MNs, have the potential to result in greater permeation across human skin, compared to the delivery of mAbs delivered via the same route.
Collapse
Affiliation(s)
- Aaron R J Hutton
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | | | | | | |
Collapse
|
37
|
Controlled release of a model hydrophilic high molecular weight compound from injectable non-lamellar liquid crystal formulations containing different types of phospholipids. Int J Pharm 2020; 577:118944. [DOI: 10.1016/j.ijpharm.2019.118944] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/18/2019] [Accepted: 12/06/2019] [Indexed: 11/21/2022]
|
38
|
Stabilization and Transdermal Delivery of an Investigational Peptide Using MicroCor® Solid-State Dissolving Microstructure Arrays. J Pharm Sci 2020; 109:1288-1296. [DOI: 10.1016/j.xphs.2019.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
|
39
|
Sabri AH, Kim Y, Marlow M, Scurr DJ, Segal J, Banga AK, Kagan L, Lee JB. Intradermal and transdermal drug delivery using microneedles - Fabrication, performance evaluation and application to lymphatic delivery. Adv Drug Deliv Rev 2020; 153:195-215. [PMID: 31634516 DOI: 10.1016/j.addr.2019.10.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/26/2019] [Accepted: 10/15/2019] [Indexed: 12/20/2022]
Abstract
The progress in microneedle research is evidenced by the transition from simple 'poke and patch' solid microneedles fabricated from silicon and stainless steel to the development of bioresponsive systems such as hydrogel-forming and dissolving microneedles. In this review, we provide an outline on various microneedle fabrication techniques which are currently employed. As a range of factors, including materials, geometry and design of the microneedles, affect the performance, it is important to understand the relationships between them and the resulting delivery of therapeutics. Accordingly, there is a need for appropriate methodologies and techniques for characterization and evaluation of microneedle performance, which will also be discussed. As the research expands, it has been observed that therapeutics delivered via microneedles has gained expedited access to the lymphatics, which makes them a favorable delivery method for targeting the lymphatic system. Such opportunity is valuable in the area of vaccination and treatment of lymphatic disorders, which is the final focus of the review.
Collapse
|
40
|
Abstract
Skin cancer, as the most physically accessible malignancy, allows for the greatest variety in treatment innovation. The last 2 decades have seen striking increases in the life expectancies of those diagnosed with malignant melanoma. However, many cases remain in which disease prevails against standard treatment, and those patients rely on continuing ingenuity. Drugs that can be injected directly into patients' tumors have become increasingly promising, not least for the reduction in side effects observed. Intratumoral therapy encompasses a wide array of agents, from chemotherapeutic drugs to cancer vaccines. While each show some efficacy, those agents which regulate the immune system likely have the greatest potential for preventing disease progression or recurrence. Recent research has highlighted the importance of the presence of cytotoxic T cells and of keeping regulatory T cells in check. Thus, manipulating the tumor microenvironment is a need in skin cancer therapy, which intratumoral delivery can potentially address. In order to find the best approach to each person's disease, more studies are needed to test intralesional agents in combination with currently approved therapies and with each other.
Collapse
|
41
|
Yang W, Jin BH, Chen YJ, Cao C, Zhu JZ, Zhao YZ, Yu XC, Li FZ. The involvement of perivascular spaces or tissues in the facial intradermal brain-targeted delivery. Drug Deliv 2019; 26:393-403. [PMID: 30929532 PMCID: PMC6450571 DOI: 10.1080/10717544.2019.1587044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Our previous work indicates the lymphatic network and perivascular spaces or tissues might be involved in the facial intradermal brain-targeted delivery of Evans blue (EB). In this article, we presented the detailed involvement of both, and the linkage between lymphatic network and perivascular spaces or tissues. The in-vivo imaging, the trigeminal transection and immunohistochemistry were used. In-vivo imaging indicated intradermal injection in the mystacial pad (i.d.) delivered EB into the brain at 2-, 6- and 24 h, while intranasal injection (i.n.) delivered EB into the rostral head and intravenous injection (i.v.) diffused EB weakly into the brain. Trigeminal perineurial and epineurial EB occurred along the perivascular spaces or tissues and along brain vessels. EB diffused into the lymphatic vessels and submandibular lymph nodes. Moreover, perineurial and epineurial EB co-located or overlaid with Lyve1 immuno-reactivity and VEGF antibody, and lymphatic network connected with perivascular spaces or tissues, suggesting lymphatic system-perivascular spaces might involve in the EB delivery with i.d. The trigeminal transection reduced the trigeminal epineurial and perineurial EB and brain EB along vessels. EB diffused in the fasciculus and the perineurium, blood and lymphatic vessels in the mystacial pad, mystacial EB overlaid VEGF or Lyve1 antibody. In summary, the dermal-trigeminal-brain perivascular spaces or tissues and the linkage to the lymphatic network mediated the intradermal brain-targeted delivery.
Collapse
Affiliation(s)
- Wei Yang
- a College of Pharmaceutical Science , Zhejiang Chinese Medical University , Hangzhou , China.,b School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang Province , China
| | - Bing-Hui Jin
- b School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang Province , China
| | - Ya-Jing Chen
- b School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang Province , China
| | - Chang Cao
- b School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang Province , China
| | - Jia-Zhen Zhu
- a College of Pharmaceutical Science , Zhejiang Chinese Medical University , Hangzhou , China
| | - Ying-Zheng Zhao
- b School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang Province , China
| | - Xi-Chong Yu
- b School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang Province , China
| | - Fan-Zhu Li
- a College of Pharmaceutical Science , Zhejiang Chinese Medical University , Hangzhou , China
| |
Collapse
|
42
|
Cao J, Zhang N, Wang Z, Su J, Yang J, Han J, Zhao Y. Microneedle-Assisted Transdermal Delivery of Etanercept for Rheumatoid Arthritis Treatment. Pharmaceutics 2019; 11:E235. [PMID: 31096705 PMCID: PMC6572071 DOI: 10.3390/pharmaceutics11050235] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a complicated autoimmune disease. The clinical applications of etanercept (EN), a TNF-α inhibitor, can efficiently halt the development of RA. EN is mainly administrated by subcutaneous injection, which may cause low compliance, side effects, and infection risk. In this study, a hyaluronic acid crosslinked microneedle system (MN) was constructed as the transdermal alternative to deliver EN. We describe the formulation, fabrication, characterization, and transdermal insertion study of MN. In vitro bioactivity of EN was conducted and analyzed by dynamic light scattering and circular dichroism spectrum. In vivo evaluation of MN was studied on adjuvant-induced arthritis mice. The MN possessed sufficient mechanical strength, good biocompatibility, little influence on the bioactivity of EN, and high anti-inflammatory efficacy. This work represents a successful example of delivering macromolecule therapeutic treatment by MN for RA treatment. The transdermal delivery of EN by MN offers a new treatment option for RA patients.
Collapse
Affiliation(s)
- Jian Cao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou 450001, China.
| | - Ziyi Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| | - Jingjing Su
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| | - Jing Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| | - Jiabing Han
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou 450001, China.
| |
Collapse
|
43
|
Sun X, Zeng L, Huang Y. Transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination. J Gene Med 2019; 21:e3089. [PMID: 30958606 DOI: 10.1002/jgm.3089] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/17/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
Therapeutic vaccination is a promising strategy for the immunotherapy of cancers. It eradicates cancer cells by evoking and strengthening the patient's own immune system. Because of the easy access and sophisticated immune networks, the skin becomes an ideal target organ for vaccination. Genetic vaccines have been widely investigated, with the advantages of the delivery of multiple antigens and a lower cost for production compared to protein/peptide vaccines. This review summarizes the advances made with respect to the transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination and also gives a brief description of the immunological milieu of the skin and the importance of dendritic cell-targeting in vaccine delivery, as well as the technologies that aim to facilitate antigen delivery and modulate antigen-presenting cells, thus improving cellular responses. The applications of genetic vaccines encoding tumor antigens delivered through the skin route, both in preclinical and clinical trials, are outlined.
Collapse
Affiliation(s)
- Xiaoyi Sun
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Linghui Zeng
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
44
|
Rini C, Roberts BC, Morel D, Klug R, Selvage B, Pettis RJ. Evaluating the Impact of Human Factors and Pen Needle Design on Insulin Pen Injection. J Diabetes Sci Technol 2019; 13:533-545. [PMID: 30880448 PMCID: PMC6501541 DOI: 10.1177/1932296819836987] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Limited published data exists quantifying the influence of human factors (HF) and pen needle (PN) design on delivery outcomes of pen injection systems. This preclinical in vivo study examines the impact of PN hub design and applied force against the skin during injection on needle penetration depth (NPD). METHOD To precisely locate injection depth, PN injections (20 µl; 2 IU, U-100 volume equivalent) of iodinated contrast agent were administered to the flank of Yorkshire swine across a range of clinically relevant application forces against the skin (0.25, 0.75, 1.25, and 2.0 lbf). The NPD, representing in vivo needle tip depth in SC tissue, from four 32 G × 4 mm PN devices (BD Nano™ 2nd Gen and three commercial posted-hub PN devices; n = 75/device/force, 1200 total) was measured by fluoroscopic imaging of the resulting depot. RESULTS The reengineered hub design more closely achieved the 4 mm target NPD with significantly less variability ( P = .006) than commercial posted-hub PN devices across the range of applied injection forces. Calculations of IM (intramuscular) injection risk completed through in silico probability model, using NPD and average human tissue thickness measurements, displayed a commensurate reduction (~2-8x) compared to conventional PN hub designs. CONCLUSIONS Quantifiable differences in injection depth were observed between identical labeled length PN devices indicating that hub design features, coupled with aspects of variable injection technique, may influence injection depth accuracy and consistency. The reengineered hub design may reduce the impact of unintended individual technique differences by improving target injection depth consistency and reducing IM injection potential.
Collapse
Affiliation(s)
- Christopher Rini
- BD Technologies and Innovation, Research Triangle Park, NC, USA
- Christopher Rini, MS, BD Technologies and Innovation, 21 Davis Dr, Research Triangle Park, NC 27709, USA.
| | | | | | - Rick Klug
- BD Technologies and Innovation, Research Triangle Park, NC, USA
| | | | | |
Collapse
|
45
|
Cote B, Rao D, Alany RG, Kwon GS, Alani AW. Lymphatic changes in cancer and drug delivery to the lymphatics in solid tumors. Adv Drug Deliv Rev 2019; 144:16-34. [PMID: 31461662 DOI: 10.1016/j.addr.2019.08.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023]
Abstract
Although many solid tumors use the lymphatic system to metastasize, there are few treatment options that directly target cancer present in the lymphatic system, and those that do are highly invasive, uncomfortable, and/or have limitations. In this review we provide a brief overview of lymphatic function and anatomy, discusses changes that befall the lymphatics in cancer and the mechanisms by which these changes occur, and highlight limitations of lymphatic drug delivery. We then go on to summarize relevant techniques and new research for targeting cancer populations in the lymphatics and enhancing drug delivery intralymphatically, including intralymphatic injections, isolated limb perfusion, passive nano drug delivery systems, and actively targeted nanomedicine.
Collapse
|
46
|
Liu X, Wang C, Liu Z. Protein-Engineered Biomaterials for Cancer Theranostics. Adv Healthc Mater 2018; 7:e1800913. [PMID: 30260583 DOI: 10.1002/adhm.201800913] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/19/2018] [Indexed: 12/18/2022]
Abstract
Proteins are an important class of biomaterials promising a variety of applications such as drug delivery, and imaging or therapy, owing to their biodegradability, biocompatibility, as well as inherent biological activities acting as enzymes, recognizing molecules, or therapeutics by themselves. Over the few past decades, different types of proteins with desired properties have been widely explored for biomedical applications. Many therapeutic proteins have now entered clinical use. This review therefore summarizes various strategies in the engineering of biomaterials for delivery of therapeutic proteins, as well as the recent development of protein-based biomaterials for cancer theranostics.
Collapse
Affiliation(s)
- Xiaowen Liu
- Pharmacology; Department of Basic Medical Sciences; Faculty of Medical Science; Jinan University; Guangzhou Guangdong 510632 China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM); Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices; Soochow University; Suzhou Jiangsu 215123 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM); Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices; Soochow University; Suzhou Jiangsu 215123 China
| |
Collapse
|
47
|
Qi F, Wu J, Li H, Ma G. Recent research and development of PLGA/PLA microspheres/nanoparticles: A review in scientific and industrial aspects. Front Chem Sci Eng 2018. [DOI: 10.1007/s11705-018-1729-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
48
|
Ye Y, Yu J, Wen D, Kahkoska AR, Gu Z. Polymeric microneedles for transdermal protein delivery. Adv Drug Deliv Rev 2018; 127:106-118. [PMID: 29408182 PMCID: PMC6020694 DOI: 10.1016/j.addr.2018.01.015] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 12/24/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
The intrinsic properties of therapeutic proteins generally present a major impediment for transdermal delivery, including their relatively large molecule size and susceptibility to degradation. One solution is to utilize microneedles (MNs), which are capable of painlessly traversing the stratum corneum and directly translocating protein drugs into the systematic circulation. MNs can be designed to incorporate appropriate structural materials as well as therapeutics or formulations with tailored physicochemical properties. This platform technique has been applied to deliver drugs both locally and systemically in applications ranging from vaccination to diabetes and cancer therapy. This review surveys the current design and use of polymeric MNs for transdermal protein delivery. The clinical potential and future translation of MNs are also discussed.
Collapse
Affiliation(s)
- Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jicheng Yu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Di Wen
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anna R Kahkoska
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
49
|
Yu J, Zhang Y, Kahkoska AR, Gu Z. Bioresponsive transcutaneous patches. Curr Opin Biotechnol 2017; 48:28-32. [PMID: 28292673 PMCID: PMC6053065 DOI: 10.1016/j.copbio.2017.03.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022]
Abstract
Transdermal drug delivery systems that utilize transcutaneous patches of arrayed microneedles have attracted increasing interest in medical practice as an alternative method to hypodermic injection. Over the past ten years, research has focused on leveraging physiological signals associated with diseases or skin-specific tissues to create bioresponsive patches that release drug directly in response to an internally-generated stimulus. This review surveys the recent advances in the development and use of bioresponsive transcutaneous patches for on-demand smart and precise drug delivery, exploiting different physiological signals including pH, serum glucose levels, and enzyme activity. The clinical potential of these devices, including challenges and opportunities, is also discussed.
Collapse
Affiliation(s)
- Jicheng Yu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuqi Zhang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anna R Kahkoska
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
50
|
Collins DS, Kourtis LC, Thyagarajapuram NR, Sirkar R, Kapur S, Harrison MW, Bryan DJ, Jones GB, Wright JM. Optimizing the Bioavailability of Subcutaneously Administered Biotherapeutics Through Mechanochemical Drivers. Pharm Res 2017; 34:2000-2011. [PMID: 28707164 PMCID: PMC5579144 DOI: 10.1007/s11095-017-2229-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/07/2017] [Indexed: 01/09/2023]
Abstract
The subcutaneous route offers myriad benefits for the administration of biotherapeutics in both acute and chronic diseases, including convenience, cost effectiveness and the potential for automation through closed-loop systems. Recent advances in parenteral administration devices and the use of additives which enhance drug dispersion have generated substantial additional interest in IV to SQ switching studies. Designing pre-clinical and clinical studies using SQ mediated delivery however requires deep understanding of complex inter-related physiologies and transport pathways governing the interstitial matrix, vascular system and lymphatic channels. This expert review will highlight key structural features which contribute to transport and biodistribution in the subcutaneous space and also assess the impact of drug formulations. Based on the rapidly growing interest in the SQ delivery route, a number of potential areas for future development are highlighted, which are likely to allow continued evolution and innovation in this important area.
Collapse
Affiliation(s)
- D S Collins
- Eli Lilly Innovation Center, 450 Kendall Street, Cambridge, Massachusetts, 02142, USA
| | - L C Kourtis
- Eli Lilly Innovation Center, 450 Kendall Street, Cambridge, Massachusetts, 02142, USA
| | - N R Thyagarajapuram
- Eli Lilly Innovation Center, 450 Kendall Street, Cambridge, Massachusetts, 02142, USA
| | - R Sirkar
- Eli Lilly Innovation Center, 450 Kendall Street, Cambridge, Massachusetts, 02142, USA
| | - S Kapur
- Eli Lilly Innovation Center, 450 Kendall Street, Cambridge, Massachusetts, 02142, USA
| | - M W Harrison
- Eli Lilly Innovation Center, 450 Kendall Street, Cambridge, Massachusetts, 02142, USA
| | - D J Bryan
- Division of Plastic and Reconstructive Surgery, Lahey Hospital and Medical Center, Burlington, Massachusetts, 01805, USA
| | - G B Jones
- Clinical & Translational Science Institute, Tufts University Medical Center, 800 Washington St, Boston, Massachusetts, 02111, USA.
| | - J M Wright
- Eli Lilly Innovation Center, 450 Kendall Street, Cambridge, Massachusetts, 02142, USA
| |
Collapse
|