1
|
Chang J, Shin K, Lewis JM, Suh HW, Lee J, Damsky W, Xu S, Bosenberg M, Saltzman WM, Girardi M. Enhanced Intratumoral Delivery of Immunomodulator Monophosphoryl Lipid A through Hyperbranched Polyglycerol-Coated Biodegradable Nanoparticles. J Invest Dermatol 2024:S0022-202X(24)01983-3. [PMID: 39122142 DOI: 10.1016/j.jid.2024.07.019] [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: 03/12/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Immunomodulatory agents have significant potential to enhance cancer treatment but have demonstrated limited efficacy beyond the preclinical setting owing to poor pharmacokinetics and toxicity associated with systemic administration. Conversely, when locally delivered, immunomodulatory agents require repeated administration to optimize immune stimulation. To overcome these challenges, we encapsulated the toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) within hyperbranched polyglycerol-coated biodegradable nanoparticles (NPs) engineered for gradual drug release from the NP core, resulting in a more persistent stimulation of antitumor immune responses while minimizing systemic side effects. In a model of malignant melanoma, we demonstrate that hyperbranched polyglycerol-NP encapsulation significantly improves the antitumor efficacy of MPLA by enhancing its ability to remodel the tumor microenvironment. Relative to free MPLA, hyperbranched polyglycerol-coated NP-encapsulated MPLA significantly increased the NK cell- and cytotoxic T-cell-mediated antitumor immune response and tuned the tumor-draining lymph nodes toward a T helper 1 response. Furthermore, when combined with local delivery of a chemotherapeutic agent, hyperbranched polyglycerol-NP-MPLA induces the conversion of an immunosuppressive tumor microenvironment to immunogenic tumor microenvironment and significantly improves survival.
Collapse
Affiliation(s)
- Jungsoo Chang
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Julia M Lewis
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hee Won Suh
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Joohyung Lee
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Suzanne Xu
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marcus Bosenberg
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - W Mark Saltzman
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Department of Chemical & Environmental Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Girardi
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
2
|
Liang S, Xiao L, Chen T, Roa P, Cocco E, Peng Z, Yu L, Wu M, Liu J, Zhao X, Deng W, Wang X, Zhao C, Deng Y, Mai Y. Injectable Nanocomposite Hydrogels Improve Intraperitoneal Co-delivery of Chemotherapeutics and Immune Checkpoint Inhibitors for Enhanced Peritoneal Metastasis Therapy. ACS NANO 2024; 18:18963-18979. [PMID: 39004822 DOI: 10.1021/acsnano.4c02312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Intraperitoneal co-delivery of chemotherapeutic drugs (CDs) and immune checkpoint inhibitors (ICIs) brings hope to improve treatment outcomes in patients with peritoneal metastasis from ovarian cancer (OC). However, current intraperitoneal drug delivery systems face issues such as rapid drug clearance from lymphatic drainage, heterogeneous drug distribution, and uncontrolled release of therapeutic agents into the peritoneal cavity. Herein, we developed an injectable nanohydrogel by combining carboxymethyl chitosan (CMCS) with bioadhesive nanoparticles (BNPs) based on polylactic acid-hyperbranched polyglycerol. This system enables the codelivery of CD and ICI into the intraperitoneal space to extend drug retention. The nanohydrogel is formed by cross-linking of aldehyde groups on BNPs with amine groups on CMCS via reversible Schiff base bonds, with CD and ICI loaded separately into BNPs and CMCS network. BNP/CMCS nanohydrogel maintained the activity of the biomolecules and released drugs in a sustained manner over a 7 day period. The adhesive property, through the formation of Schiff bases with peritoneal tissues, confers BNPs with an extended residence time in the peritoneal cavity after being released from the nanohydrogel. In a mouse model, BNP/CMCS nanohydrogel loaded with paclitaxel (PTX) and anti-PD-1 antibodies (αPD-1) significantly suppressed peritoneal metastasis of OC compared to all other tested groups. In addition, no systemic toxicity of nanohydrogel-loaded PTX and αPD-1 was observed during the treatment, which supports potential translational applications of this delivery system.
Collapse
Affiliation(s)
- Shu Liang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen 518020, China
| | - Tian Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Paola Roa
- Department of Biochemistry and Molecular Biology/Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine, Miami, Florida 33136, United States
| | - Emiliano Cocco
- Department of Biochemistry and Molecular Biology/Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine, Miami, Florida 33136, United States
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Jie Liu
- ISCTE Business School, BRU-IUL, University Institute of Lisbon, Avenida das Armadas, Lisbon 1649-026, Portugal
| | - Xizhe Zhao
- Department of Chemistry, College of Staten Island, City University of New York, New York, New York 10314, United States
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Xiongjun Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Chao Zhao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama 35487, United States
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| |
Collapse
|
3
|
Liu J, Zhou L, Cong H, Hu J, Tang J. Resveratrol-loaded microemulsion based thermosensitive hydrogel for potential topical treatment of the vaginal inflammation. J Drug Target 2024; 32:404-412. [PMID: 38288679 DOI: 10.1080/1061186x.2024.2310879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/22/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Vaginal inflammation is a prevalent gynecological condition. If left untreated, it can potentially spread to the urinary and reproductive systems. METHODS In this study, we propose a resveratrol-loaded microemulsion-based thermosensitive hydrogel (Res-Me-Tsgel) and compare it with a chitosan hydrogel-based Res-Me-Cogel. We characterized the different characters of Res-Me-Tsgel. The safety of Res-Me-Tsgel was also evaluated in vitro and in vivo. Finally, we measured the retention of Res in the vagina after drug administration. RESULTS The Res-Me-Tsgel we prepared is a transparent liquid solution at room temperature that rapidly forms a gel at 37oC. Compared to Res solution and Res-Me, both Res-Me-Cogel and Res-Me-Tsgel demonstrate superior sustained release properties. Both in vitro and in vivo studies confirm the excellent biosafety profile of Res-Me-Cogel and Res-Me-Tsgel. Vaginal administration of these formulations in rats results in prolonged retention of resveratrol within the vagina. Notably, due to its improved flow into vaginal folds after administration, the retention of Resveratrol was approximately three times higher for the Res-Me-Tsgel group compared to the Res-Me-Cogel group at 24 h post-administration. Overall, these findings highlight the potential application of Res-Me-Tsgel as an effective means for vaginal inflammation. CONCLUSIONS We developed a novel micromulsion based thermosensitive hydrogel for the delivery of Res. The sustained release of Res and favorable vaginal retention from Res-Me-Tsgel make them promise as a potential candidate for local intravaginal therapy.
Collapse
Affiliation(s)
- Jiaxin Liu
- School of Pharmacy, Harbin Medical University, Harbin, China
| | - Liuqi Zhou
- School of Pharmacy, Harbin Medical University, Harbin, China
| | - Huijing Cong
- School of Pharmacy, Harbin Medical University, Harbin, China
| | - Jing Hu
- School of Pharmacy, Harbin Medical University, Harbin, China
| | - Jingling Tang
- School of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Farooq U, Mirza MA, Alshetaili A, Mohapatra S, Jain P, Hassan N, Iqbal Z, Ali A. In silico and in vitro assessment of an optimized QbD-guided myoinositol and metformin-loaded mucus-penetrating particle-based gel for the amelioration of PCOS. NANOSCALE ADVANCES 2024; 6:648-668. [PMID: 38235090 PMCID: PMC10791119 DOI: 10.1039/d3na00943b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/29/2023] [Indexed: 01/19/2024]
Abstract
Polycystic ovarian syndrome (PCOS) is a multi-factorial endocrine disorder affecting women of reproductive age. However, its high prevalence and the unsuccessful translation of conventional modalities have made PCOS a pharmaco-therapeutic challenge. In the present study, we explored bi-formulations (comprising metformin-loaded mucus-penetrating nanoparticles, MTF-MPPs, and myoinositol-loaded mucus-penetrating particles, MI-MPPs) incorporated in a carbomer gel tailored for intravaginal administration. For the development and optimization of the MPPs-gel, a QbD (quality by design) approach was employed, including the initial and final risk assessment, central composite design of experts, and method validation. The optimized MTF-MPPs and MI-MPPs possessed an optimum nanometric particle size (195.0 nm and 178.8 nm, respectively) and a PDI of 0.150 and 0.123, respectively, together with a negligible negative zeta potential (-5.19 mV and -6.19 mV, respectively) through the vaginal mucus. It was observed that the MPPs are small and monodisperse with a neutral surface charge. It was observed that the MPPs-gel formulations released approximately 69.86 ± 4.65% of MTF and 67.14 ± 5.74% of MI within 120 h (5 days), which was observed to be sustained unlike MFT-MI-gel with approximately 94.89 ± 4.17% of MTF and 90.91 ± 15% of MI drugs released within 12 h. The confocal microscopy study of rhodamine-loaded MPPs indicated that they possessed a high fluorescence intensity at a depth of 15 μm, while as the penetration trajectory in the vaginal tissue increased to 35 μm, their intensity was reduced, appearing to be more prominent in the blood vessels. The analyzed data of MPPs-gel suggest that the optimized MPPs-gel formulation has potential to reach the targeted area via the uterovaginal mucosa, which has a wide network of blood vessels. Subsequently, in vivo studies were conducted and the results revealed that the proposed MPPs-gel formulation could regulate the estrous cycle of the reproductive system compared to the conventional formulation. Moreover, the formulation significantly reduced the weight of the ovaries compared to the control and conventional vaginal gel. Biochemical estimation showed improved insulin and sex hormone levels. Thus, the obtained data revealed that the deep penetration and deposition of MTF and MI on the targeted area through intravaginal delivery resulted in better therapeutic effects than the conventional vaginal gel. The obtained results confirmed the amelioration of PCOS upon treatment using the prepared MPPs-gel formulation. According to the relevant evaluation studies, it was concluded that MPPs-gel was retained in the vaginal cavity for systemic effects. Also, the sustained and non-irritating therapeutic effect meets the safety aspects. This work serves as a promising strategy for intravaginal drug delivery.
Collapse
Affiliation(s)
- Uzma Farooq
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard New Delhi 110062 India +91-9899571726 +91-9811733016
| | - Mohd Aamir Mirza
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard New Delhi 110062 India +91-9899571726 +91-9811733016
| | - Abdullah Alshetaili
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University Alkharj Saudi Arabia
| | - Sradhanjali Mohapatra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard New Delhi 110062 India +91-9899571726 +91-9811733016
| | - Pooja Jain
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard New Delhi 110062 India +91-9899571726 +91-9811733016
| | - Nazia Hassan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard New Delhi 110062 India +91-9899571726 +91-9811733016
| | - Zeenat Iqbal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard New Delhi 110062 India +91-9899571726 +91-9811733016
| | - Asgar Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard New Delhi 110062 India +91-9899571726 +91-9811733016
| |
Collapse
|
5
|
Khang M, Lee JH, Lee T, Suh HW, Lee S, Cavaliere A, Rushing A, Geraldo LH, Belitzky E, Rossano S, de Feyter HM, Shin K, Huttner A, Roussel MF, Thomas JL, Carson RE, Marquez-Nostra B, Bindra RS, Saltzman WM. Intrathecal delivery of nanoparticle PARP inhibitor to the cerebrospinal fluid for the treatment of metastatic medulloblastoma. Sci Transl Med 2023; 15:eadi1617. [PMID: 37910601 PMCID: PMC11078331 DOI: 10.1126/scitranslmed.adi1617] [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: 04/06/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
The morbidity associated with pediatric medulloblastoma, in particular in patients who develop leptomeningeal metastases, remains high in the absence of effective therapies. Administration of substances directly into the cerebrospinal fluid (CSF) is one approach to circumvent the blood-brain barrier and focus delivery of drugs to the site of tumor. However, high rates of CSF turnover prevent adequate drug accumulation and lead to rapid systemic clearance and toxicity. Here, we show that PLA-HPG nanoparticles, made with a single-emulsion, solvent evaporation process, can encapsulate talazoparib, a PARP inhibitor (BMN-673). These degradable polymer nanoparticles improve the therapeutic index when delivered intrathecally and lead to sustained drug retention in the tumor as measured with PET imaging and fluorescence microscopy. We demonstrate that administration of these particles into the CSF, alone or in combination with systemically administered temozolomide, is a highly effective therapy for tumor regression and prevention of leptomeningeal spread in xenograft mouse models of medulloblastoma. These results provide a rationale for harnessing nanoparticles for the delivery of drugs limited by brain penetration and therapeutic index and demonstrate important advantages in tolerability and efficacy for encapsulated drugs delivered locoregionally.
Collapse
Affiliation(s)
- Minsoo Khang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Ju Hyun Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Teresa Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Supum Lee
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Alessandra Cavaliere
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Amy Rushing
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Luiz H. Geraldo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT 06510, USA
| | - Erika Belitzky
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Samantha Rossano
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Henk M. de Feyter
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Martine F. Roussel
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 38103, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France
| | - Richard E. Carson
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | | | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale University, New Haven, CT 06520, USA
| | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT 06510, USA
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT 06511, USA
- Department of Dermatology, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
6
|
Avlani D, Kumar A, H N S. Development of Dispersible Vaginal Tablets of Tenofovir Loaded Mucoadhesive Chitosan Microparticles for Anti-HIV Pre-Exposure Prophylaxis. Mol Pharm 2023; 20:5006-5018. [PMID: 37656937 DOI: 10.1021/acs.molpharmaceut.3c00288] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Tenofovir disoproxil fumarate (TDF)-loaded bioadhesive chitosan microparticles (CM) were developed by an emulsification internal gelation technique. Among different batches produced, ECH-4 was found to display a high % entrapment efficiency (68.93 ± 1.76%) and sustained drug release of 88.05 ± 0.38% at 24 h. Solid state characterization of ECH-4 employing DSC and PXRD indicated that the TDF existed in an amorphous state as a solid-solid solution in chitosan. Scanning electron microscopy revealed CM of ECH-4 was spherical in shape with a rough surface topography. Laser scattering analysis using Malvern Master sizer indicated that particle size of ECH-4 was in the range of 0.52 ± 0.10 μm to 284.79 ± 21.42 μm with a surface-mean diameter of 12.41 ± 0.06 μm. Ex vivo mucoadhesion studies using rabbit mucosa as a substrate indicated that 10.34 ± 2.08% of CM of ECH-4 was retained at the end of 24 h. The microparticles of ECH-4 were incorporated into dispersible tablets (DT-TCM) intended for intravaginal administration, in view to arrest the pre-exposure transmission of HIV during sexual intercourse. In vitro release from the dispersible tablet (F3) into simulated vaginal fluid (pH 4.5) displayed a sustained release profile of TDF as 89.98 ± 1.61% of TDF was released at 24 h. The in vitro dissolution profile of the DT-TCM was found to be similar to that of TDF loaded CM with the values of f1 (difference factor) and f2 (similarity factor) being 1.52 and 78.02, respectively. Therefore, DT-TCM would be a promising novel drug delivery platform for pre-exposure prophylaxis against HIV.
Collapse
Affiliation(s)
- Dhruti Avlani
- Department of Pharmaceutics, Dr. Prabhakar B Kore Basic Science Research Center, Off-campus, KLE College of Pharmacy (A constituent unit of KAHER-Belagavi), Rajajinagar, Bengaluru 560010 Karnataka, India
| | - Avichal Kumar
- Department of Pharmaceutics, Dr. Prabhakar B Kore Basic Science Research Center, Off-campus, KLE College of Pharmacy (A constituent unit of KAHER-Belagavi), Rajajinagar, Bengaluru 560010 Karnataka, India
| | - Shivakumar H N
- Department of Pharmaceutics, Dr. Prabhakar B Kore Basic Science Research Center, Off-campus, KLE College of Pharmacy (A constituent unit of KAHER-Belagavi), Rajajinagar, Bengaluru 560010 Karnataka, India
| |
Collapse
|
7
|
Chang J, Yu B, Saltzman WM, Girardi M. Nanoparticles as a Therapeutic Delivery System for Skin Cancer Prevention and Treatment. JID INNOVATIONS 2023; 3:100197. [PMID: 37205301 PMCID: PMC10186617 DOI: 10.1016/j.xjidi.2023.100197] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/14/2023] [Accepted: 01/30/2023] [Indexed: 03/18/2023] Open
Abstract
The use of nanoparticles (NPs) as a therapeutic delivery system has expanded markedly over the past decade, particularly regarding applications targeting the skin. The delivery of NP-based therapeutics to the skin requires special consideration owing to its role as both a physical and immunologic barrier, and specific technologies must not only take into consideration the target but also the pathway of delivery. The unique challenge this poses has been met with the development of a wide panel of NP-based technologies meant to precisely address these considerations. In this review article, we describe the application of NP-based technologies for drug delivery targeting the skin, summarize the types of NPs, and discuss the current landscape of NPs for skin cancer prevention and skin cancer treatment as well as future directions within these applications.
Collapse
Affiliation(s)
- Jungsoo Chang
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Beverly Yu
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - W. Mark Saltzman
- Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Michael Girardi
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Xie Y, Ye J, Ouyang Y, Gong J, Li C, Deng Y, Mai Y, Liu Y, Deng W. Microneedle-Assisted Topical Delivery of Idebenone-Loaded Bioadhesive Nanoparticles Protect against UV-Induced Skin Damage. Biomedicines 2023; 11:1649. [PMID: 37371744 DOI: 10.3390/biomedicines11061649] [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: 04/30/2023] [Revised: 05/27/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Ultraviolet (UV) radiation can penetrate the basal layer of the skin and induce profound alterations in the underlying dermal tissues, including skin pigmentation, oxidative stress, photoaging, glycation, and skin cancer. Idebenone (IDB), an effective antioxidant that suppresses melanin biosynthesis and glycation, can protect the skin from UV-induced damage, accounting for its use in commercial anti-aging formulations. Ideally, IDB formulations should retain IDB inside the skin for a sufficient period, despite disturbances such as sweating or swimming. Herein, we present an IDB topical formulation based on Tris (tris(hydroxymethyl)-aminomethane)-modified bioadhesive nanoparticles (Tris-BNPs) and microneedle-assisted delivery. We found that Tris-BNPs loaded with IDB (IDB/Tris-BNPs) effectively reached the basal layer of the skin and were retained for at least 4 days with a slow and continuous drug release profile, unlike non-bioadhesive nanoparticles (NNPs) and bioadhesive nanoparticles (BNPs) of similar sizes (ranging from 120-142 nm) and zeta-potentials (above -20 mV), which experienced a significant reduction in concentration within 24 h. Notably, IDB/Tris-BNPs showed superior performance against UV-induced damage relative to IDB/NNPs and IDB/BNPs. This effect was demonstrated by lower levels of reactive oxygen species and advanced glycation end-products in skin tissues, as well as suppressed melanogenesis. Therefore, the proposed IDB delivery strategy provided long-term protective effects against UV-induced skin damage.
Collapse
Affiliation(s)
- Yuan Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Jingping Ye
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yaqi Ouyang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Jianing Gong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Chujie Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
9
|
Yu L, Yu M, Chen W, Sun S, Huang W, Wang T, Peng Z, Luo Z, Fang Y, Li Y, Deng Y, Wu M, Tao W. In Situ Separable Nanovaccines with Stealthy Bioadhesive Capability for Durable Cancer Immunotherapy. J Am Chem Soc 2023. [PMID: 36930579 DOI: 10.1021/jacs.2c12986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Because of tumor heterogeneity and the immunosuppressive tumor microenvironment, most cancer vaccines typically do not elicit robust antitumor immunological responses in clinical trials. In this paper, we report findings about a bioadhesive nanoparticle (BNP)-based separable cancer vaccine, FeSHK@B-ovalbumin (OVA), to target multi-epitope antigens and exert effective cancer immunotherapy. After the FeSHK@B-OVA "nanorocket" initiates the "satellite-rocket separation" procedure in the acidic tumor microenvironment, the FeSHK@B "launch vehicle" can amplify intracellular oxidative stress persistently. This procedure allows for bioadhesiveness-mediated prolonged drug retention within the tumor tissue and triggers the immunogenic death of tumor cells that transforms the primary tumors into antigen depots, which acts synergistically with the OVA "satellite" to trigger robust antigen-specific antitumor immunity. The cooperation of these two immunostimulants not only efficiently inhibits the primary tumor growth and provokes durable antigen-specific immune activation in vivo but also activates a long-term and robust immune memory effect to resist tumor rechallenge and metastasis. These results highlight the enormous potential of FeSHK@B-OVA to serve as an excellent therapeutic and prophylactic cancer nanovaccine. By leveraging the antigen depots in situ and the synergistic effect among multi-epitope antigens, such a nanovaccine strategy with stealthy bioadhesion may offer a straightforward and efficient approach to developing various cancer vaccines for different types of tumors.
Collapse
Affiliation(s)
- Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Wenxin Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Tianqi Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Zewen Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Yixuan Fang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
10
|
Patel R, Yadav BK, Patel G. Progresses in Nano-Enabled Platforms for the Treatment of Vaginal Disorders. RECENT PATENTS ON NANOTECHNOLOGY 2023; 17:208-227. [PMID: 35762539 DOI: 10.2174/1872210516666220628150447] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The most common vaginal disorders are within the uterus. According to the latest statistics, vaginal disorders occur in 50% to 60% of females. Although curative treatments rely on surgical therapy, still first-line treatment is a non invasive drug. Conventional therapies are available in the oral and parenteral route, leading to nonspecific targeting, which can cause dose-related side effects. Vaginal disorders are localized uterine disorders in which intrauterine delivery via the vaginal site is deemed the preferable route to mitigate clinical drug delivery limitations. OBJECTIVE This study emphasizes the progress of site-specific and controlled delivery of therapeutics in the treatment of vaginal disorders and systemic adverse effects as well as the therapeutic efficacy. METHODS Related research reports and patents associated with topics are collected, utilized, and summarized the key findings. RESULTS The comprehensive literature study and patents like (US 9393216 B2), (JP6672370B2), and (WO2018041268A1) indicated that nanocarriers are effective above traditional treatments and have some significant efficacy with novelty. CONCLUSION Nowadays, site-specific and controlled delivery of therapeutics for the treatment of vaginal disorders is essential to prevent systemic adverse effects and therapeutic efficacy would be more effective. Nanocarriers have therefore been used to bypass the problems associated with traditional delivery systems for the vaginal disorder.
Collapse
Affiliation(s)
- Riya Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat 388421, India
| | - Bindu Kumari Yadav
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat 388421, India
| | - Gayatri Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat 388421, India
| |
Collapse
|
11
|
Thurman AR, Ouattara LA, Yousefieh N, Anderson PL, Bushman LR, Fang X, Hanif H, Clark M, Singh O, Doncel GF. A phase I study to assess safety, pharmacokinetics, and pharmacodynamics of a vaginal insert containing tenofovir alafenamide and elvitegravir. Front Cell Infect Microbiol 2023; 13:1130101. [PMID: 37153145 PMCID: PMC10154607 DOI: 10.3389/fcimb.2023.1130101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
Background New multi-purpose prevention technology (MPT) products are needed to prevent human immunodeficiency virus (HIV) and herpes simplex virus type 2 (HSV2). In this study, we evaluated a fast-dissolve insert that may be used vaginally or rectally for prevention of infection. Objective To describe the safety, acceptability, multi-compartment pharmacokinetics (PK), and in vitro modeled pharmacodynamics (PD) after a single vaginal dose of an insert containing tenofovir alafenamide (TAF) and elvitegravir (EVG) in healthy women. Methods This was a Phase I, open-label, study. Women (n=16) applied one TAF (20mg)/EVG (16mg) vaginal insert and were randomized (1:1) to sample collection time groups for up to 7 days post dosing. Safety was assessed by treatment-emergent adverse events (TEAEs). EVG, TAF and tenofovir (TFV) concentrations were measured in plasma, vaginal fluid and tissue, and TFV-diphosphate (TFV-DP) concentration in vaginal tissue. PD was modeled in vitro by quantifying the change in inhibitory activity of vaginal fluid and vaginal tissue against HIV and HSV2 from baseline to after treatment. Acceptability data was collected by a quantitative survey at baseline and post treatment. Results The TAF/EVG insert was safe, with all TEAEs graded as mild, and acceptable to participants. Systemic plasma exposure was low, consistent with topical delivery, while high mucosal levels were detected, with median TFV vaginal fluid concentrations exceeding 200,000 ng/mL and 1,000 ng/mL for up to 24 hours and 7 days post dosing, respectively. All participants had vaginal tissue EVG concentrations of > 1 ng/mg at 4 and 24 hours post dosing. The majority had tissue TFV-DP concentrations exceeding 1000 fmol/mg by 24 - 72 hours post dosing. Vaginal fluid inhibition of HIV-1 and HSV-2 in vitro significantly increased from baseline and was similarly high at 4 and 24 hours post dosing. Consistent with high tissue TFV-DP concentrations, p24 HIV antigen production from ectocervical tissues infected ex vivo with HIV-1 significantly decreased from baseline at 4 hours post dosing. HSV-2 production from tissue also decreased post treatment. Conclusions A single dose of TAF/EVG inserts met PK benchmarks, with PK data supporting an extended window of high mucosal protection. PD modeling supports mucosal protection against both HIV-1 and HSV-2. The inserts were safe and highly acceptable. Clinical trial registration ClinicalTrials.gov, identifier NCT03762772.
Collapse
Affiliation(s)
- Andrea R. Thurman
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
- *Correspondence: Andrea R. Thurman,
| | - Louise A. Ouattara
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Nazita Yousefieh
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Peter L. Anderson
- University of Colorado, Colorado Antiviral Pharmacology Lab, School of Pharmacy, Anschutz Medical Campus, Aurora, CO, United States
| | - Lane R. Bushman
- University of Colorado, Colorado Antiviral Pharmacology Lab, School of Pharmacy, Anschutz Medical Campus, Aurora, CO, United States
| | - Xi Fang
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Homaira Hanif
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Meredith Clark
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Onkar Singh
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| | - Gustavo F. Doncel
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, VA, United States
| |
Collapse
|
12
|
Cheng X, Xie Q, Sun Y. Advances in nanomaterial-based targeted drug delivery systems. Front Bioeng Biotechnol 2023; 11:1177151. [PMID: 37122851 PMCID: PMC10133513 DOI: 10.3389/fbioe.2023.1177151] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Nanomaterial-based drug delivery systems (NBDDS) are widely used to improve the safety and therapeutic efficacy of encapsulated drugs due to their unique physicochemical and biological properties. By combining therapeutic drugs with nanoparticles using rational targeting pathways, nano-targeted delivery systems were created to overcome the main drawbacks of conventional drug treatment, including insufficient stability and solubility, lack of transmembrane transport, short circulation time, and undesirable toxic effects. Herein, we reviewed the recent developments in different targeting design strategies and therapeutic approaches employing various nanomaterial-based systems. We also discussed the challenges and perspectives of smart systems in precisely targeting different intravascular and extravascular diseases.
Collapse
|
13
|
Mai Y, Ouyang Y, Yu M, Qin Y, Girardi M, Saltzman WM, Cocco E, Zhao C, Yu L, Jia Y, Xiao L, Dou L, Deng W, Liu Y, Xie J, Deng Y. Topical formulation based on disease-specific nanoparticles for single-dose cure of psoriasis. J Control Release 2022; 349:354-366. [PMID: 35817278 DOI: 10.1016/j.jconrel.2022.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/09/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
First-line treatments for mild to moderate psoriasis are typically topical formulations containing corticosteroids, however, the therapeutic efficacy of these formulations is compromised by limited penetration and skin retention. Even more challenging, off-target corticosteroids are known to adversely affect healthy skin, including induction of epidermal and dermal atrophy. Here, we report a nanoparticle-based topical formulation that cures psoriasis in a single dose, but leaves healthy skin intact. Specifically, we developed tris(hydroxymethyl)aminomethane-modified bioadhesive nanoparticles (Tris-BNPs) that exploit the high permeability characteristic of psoriasis to penetrate only psoriatic skin but not the healthy skin. Furthermore, as Tris-BNPs diffuse and penetrate into the epidermis, the Tris molecules slowly diffuse away, exposing the aldehyde groups of BNPs, which can bind to amine groups present within lesional skin, leading to long local retention of BNPs in lesions of psoriatic skin. The accumulated BNPs within lesions release corticosteroids over a ~ 3 day period to maintain local drug concentration above the therapeutic level. In addition to deeper penetration and longer retention compared with commercial psoriasis treatments, the topical applied Tris-BNPs were not affected by sweating, humidity, or active wiping due to their preferential accumulation between the stratum corneum and the basal cells of the epidermis. Overall, Tris-BNP as a topical formulation hold promise to overcome the limitations of current psoriasis treatment.
Collapse
Affiliation(s)
- Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yaqi Ouyang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yujia Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Michael Girardi
- Department of Dermatology, Yale University, 333 Cedar Street, New Haven, CT 06520, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, 55 Prospect Street, New Haven, CT 06511, USA
| | - Emiliano Cocco
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Liu Dou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China.
| | - Julin Xie
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
14
|
Lin H, Jia Y, Kong X, Wang S, Liu X, Liu Y, Deng Y. In Vivo Evaluation of Cefuroxime Axetil-Loaded Bioadhesive Nanoparticles to Treat Haemophilus influenzae-Induced Otitis Media. Front Bioeng Biotechnol 2022; 10:884797. [PMID: 35573224 PMCID: PMC9099258 DOI: 10.3389/fbioe.2022.884797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
Otitis media (OM) is a common disease in children. One of the most common pathogens causing OM is non-typeable Haemophilus influenzae (NTHi). NTHi in the middle ear can be successfully eradicated by a regimen of oral antibiotics sustained for 7–10 days (e.g., cefuroxime axetil 250 mg/day for patients aged 3 months to 2 years and 500 mg/day for patients ages ≥2 years). However, lack of compliance is relevant to treatment failure or early relapse. In order to overcome these challenges, we have developed antibiotics-loaded bioadhesive nanoparticles (BNPs) that can adhere to the epidermis of the middle ear after local administration and significantly prolong the release time of antibiotics in the middle ear. Compared with oral administration of CA, local delivery of free antibiotic cefuroxime axetil (CA), and CA loaded non-bioadhesive nanoparticles (CA/NNPs), BNPs loaded with cefuroxime axetil (CA/BNPs) showed significantly longer retention time in the middle ear, resulting in continuous release of the drug and higher therapeutic efficacy against OM with only a single dosage. CA concentrations were maintained above the minimum inhibitory concentration (MIC) for NTHi throughout 7 days’ treatment. NTHi OM in a mouse model was successfully eradicated without causing tissue toxicity. CA/BNPs minimize systemic drug exposure through local administration, as demonstrated by undetectable levels in the blood.
Collapse
Affiliation(s)
| | | | | | | | | | - Yang Liu
- *Correspondence: Yang Liu, ; Yang Deng,
| | - Yang Deng
- *Correspondence: Yang Liu, ; Yang Deng,
| |
Collapse
|
15
|
Watchorn J, Clasky AJ, Prakash G, Johnston IAE, Chen PZ, Gu FX. Untangling Mucosal Drug Delivery: Engineering, Designing, and Testing Nanoparticles to Overcome the Mucus Barrier. ACS Biomater Sci Eng 2022; 8:1396-1426. [PMID: 35294187 DOI: 10.1021/acsbiomaterials.2c00047] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mucus is a complex viscoelastic gel and acts as a barrier covering much of the soft tissue in the human body. High vascularization and accessibility have motivated drug delivery to various mucosal surfaces; however, these benefits are hindered by the mucus layer. To overcome the mucus barrier, many nanomedicines have been developed, with the goal of improving the efficacy and bioavailability of drug payloads. Two major nanoparticle-based strategies have emerged to facilitate mucosal drug delivery, namely, mucoadhesion and mucopenetration. Generally, mucoadhesive nanoparticles promote interactions with mucus for immobilization and sustained drug release, whereas mucopenetrating nanoparticles diffuse through the mucus and enhance drug uptake. The choice of strategy depends on many factors pertaining to the structural and compositional characteristics of the target mucus and mucosa. While there have been promising results in preclinical studies, mucus-nanoparticle interactions remain poorly understood, thus limiting effective clinical translation. This article reviews nanomedicines designed with mucoadhesive or mucopenetrating properties for mucosal delivery, explores the influence of site-dependent physiological variation among mucosal surfaces on efficacy, transport, and bioavailability, and discusses the techniques and models used to investigate mucus-nanoparticle interactions. The effects of non-homeostatic perturbations on protein corona formation, mucus composition, and nanoparticle performance are discussed in the context of mucosal delivery. The complexity of the mucosal barrier necessitates consideration of the interplay between nanoparticle design, tissue-specific differences in mucus structure and composition, and homeostatic or disease-related changes to the mucus barrier to develop effective nanomedicines for mucosal delivery.
Collapse
Affiliation(s)
- Jeffrey Watchorn
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Aaron J Clasky
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Gayatri Prakash
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Ian A E Johnston
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Paul Z Chen
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Frank X Gu
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada.,Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
16
|
Engineering drug delivery systems to overcome the vaginal mucosal barrier: Current understanding and research agenda of mucoadhesive formulations of vaginal delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
17
|
Tong X, Patel SK, Li J, Patton D, Xu E, Anderson PL, Parikh U, Sweeney Y, Strizki J, Hillier SL, Rohan LC. Development and Evaluation of Nanoparticles-in-Film Technology to Achieve Extended In Vivo Exposure of MK-2048 for HIV Prevention. Polymers (Basel) 2022; 14:polym14061196. [PMID: 35335526 PMCID: PMC8955144 DOI: 10.3390/polym14061196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/05/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022] Open
Abstract
MK-2048 is a second-generation integrase inhibitor active against HIV, which has been applied vaginally using ring formulations. In this work, a nanoparticle-in-film technology was developed as a discrete pre-exposure prophylactic product option against HIV for an extended duration of use. A film platform loaded with poly (lactic-co-glycolic acid) nanoparticles (PNP) encapsulating MK-2048 was engineered. MK-2048 PNPs were loaded into films that were manufactured via the solvent casting method. Physicochemical and mechanical properties, in vitro efficacy, Lactobacillus compatibility, in vitro and ex vivo permeability, and in vivo pharmacokinetics in macaques were evaluated. PNPs with a mean diameter of 382.2 nm and −15.2 mV zeta potential were obtained with 95.2% drug encapsulation efficiency. PNP films showed comparable in vitro efficacy to free MK-2048 (IC50 0.46 vs. 0.54 nM) and were found to have no impact on Lactobacillus. MK-2048 encapsulated in PNPs showed an increase in permeability (>4-fold) compared to the free MK-2048 in MDCKII cell lines. Furthermore, PNPs had higher ectocervical tissue permeability (1.7-fold) compared to free MK-2048. PNP films showed sustained drug levels for at least 3 weeks in the macaque vaginal fluid. This work demonstrates the synergy of integrating nanomedicine and polymeric film technology to achieve sustained vaginal drug delivery.
Collapse
Affiliation(s)
- Xin Tong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.T.); (S.K.P.); (J.L.); (E.X.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA; (U.P.); (S.L.H.)
| | - Sravan Kumar Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.T.); (S.K.P.); (J.L.); (E.X.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA; (U.P.); (S.L.H.)
| | - Jing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.T.); (S.K.P.); (J.L.); (E.X.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA; (U.P.); (S.L.H.)
| | - Dorothy Patton
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (D.P.); (Y.S.)
| | - Elaine Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.T.); (S.K.P.); (J.L.); (E.X.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA; (U.P.); (S.L.H.)
| | - Peter L. Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO 80045, USA;
| | - Urvi Parikh
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA; (U.P.); (S.L.H.)
| | - Yvonne Sweeney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (D.P.); (Y.S.)
| | - Julie Strizki
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA;
| | - Sharon L. Hillier
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA; (U.P.); (S.L.H.)
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lisa C. Rohan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.T.); (S.K.P.); (J.L.); (E.X.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA; (U.P.); (S.L.H.)
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence: ; Tel.: +1-412-641-6108
| |
Collapse
|
18
|
Bioadhesive Nanoparticles for Local Drug Delivery. Int J Mol Sci 2022; 23:ijms23042370. [PMID: 35216484 PMCID: PMC8874699 DOI: 10.3390/ijms23042370] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 12/18/2022] Open
Abstract
Local drug delivery is an effective strategy for achieving direct and instant therapeutic effects. Current clinical treatments have fallen short and are limited by traditional technologies. Bioadhesive nanoparticles (NPs), however, may be a promising carrier for optimized local drug delivery, offering prolonged drug retention time and steadily maintained therapeutic concentrations. In addition, the possibility of clinical applications of this platform are abundant, as most polymers used for bioadhesion are both biodegradable and biocompatible. This review highlights the major advances in the investigations of polymer-based bioadhesive nanoparticles and their innumerable applications in local drug delivery.
Collapse
|
19
|
Yan X, Huang Z, Wu Y, Yu Z, Yang K, Chen Z, Wang W, Hu H, Wang Z. Sequential loading of inclusion complex/nanoparticles improves the gastric retention of Vladimiriae Radix essential oil to promote the protection of acute gastric mucosal injury. Int J Pharm 2021; 610:121234. [PMID: 34718092 DOI: 10.1016/j.ijpharm.2021.121234] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/03/2021] [Accepted: 10/23/2021] [Indexed: 12/22/2022]
Abstract
The essential oil from Vladimiriae Radix (VEO) is a medicinal natural product with anti-ulcer activity. A novel gastroretentive drug delivery system was developed by preparing the hydroxypropyl-β-cyclodextrin (HP-β-CD) inclusion complex incorporated into chitosan nanoparticles (V-CD/NPs), to improve the bioavailability of VEO and its protective effect on gastric mucosa. The optimum preparation process of V-CD/NPs was obtained by Plackett-Burman and Box-Behnken response surface methodology. The resulting V-CD/NPs gained a suitable positive potential and small particle size, and showed stability in simulated gastric fluid, whose morphology and in vitro drug release profiles had a pH-sensitivity. Besides, V-CD/NPs was proved to strongly bind with mucin, and in vivo imaging revealed that it could be retained in the stomach for more than 8 h. The results of drug concentration in gastric tissues showed that the sequential loading of inclusion complex/nanoparticles promoted the local absorption of VEO in gastric tissues, which was favorable to reach the effective therapeutic concentration in the lesioned mucosa area. In comparison to VEO and V-CD, the callback effect of V-CD/NPs on 1L-1β, 1L-6, TNF-α, NF-κB, MDA and SOD was comparable to cimetidine, and V-CD/NPs outperformed in gastric mucosal protection. Therefore, the gastroretentive drug delivery system developed in our study effectively enhanced the anti-ulcer activity of VEO, which could be a promising strategy for the prevention and treatment of the acute gastric mucosal injury.
Collapse
Affiliation(s)
- Xiaomin Yan
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Zecheng Huang
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Yuyi Wu
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Ziwei Yu
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Ke Yang
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Ziqiang Chen
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Wenjun Wang
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Huiling Hu
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China.
| | - Zhanguo Wang
- Holistic Integrative Medicine Industry Collaborative Innovation Research Center, Qiang Medicine Standard Research Promotion Base and Collaborative Innovation Research Center, School of Preclinical Medicine, Chengdu University, Sichuan, Chengdu 610106, China.
| |
Collapse
|
20
|
das Neves J, Notario-Pérez F, Sarmento B. Women-specific routes of administration for drugs: A critical overview. Adv Drug Deliv Rev 2021; 176:113865. [PMID: 34280514 DOI: 10.1016/j.addr.2021.113865] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/19/2022]
Abstract
The woman's body presents a number of unique anatomical features that can constitute valuable routes for the administration of drugs, either for local or systemic action. These are associated with genitalia (vaginal, endocervical, intrauterine, intrafallopian and intraovarian routes), changes occurring during pregnancy (extra-amniotic, intra-amniotic and intraplacental routes) and the female breast (breast intraductal route). While the vaginal administration of drug products is common, other routes have limited clinical application and are fairly unknown even for scientists involved in drug delivery science. Understanding the possibilities and limitations of women-specific routes is of key importance for the development of new preventative, diagnostic and therapeutic strategies that will ultimately contribute to the advancement of women's health. This article provides an overview on women-specific routes for the administration of drugs, focusing on aspects such as biological features pertaining to drug delivery, relevance in current clinical practice, available drug dosage forms/delivery systems and administration techniques, as well as recent trends in the field.
Collapse
|
21
|
Nanotechnology-based approaches for emerging and re-emerging viruses: Special emphasis on COVID-19. Microb Pathog 2021; 156:104908. [PMID: 33932543 PMCID: PMC8079947 DOI: 10.1016/j.micpath.2021.104908] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022]
Abstract
In recent decades, the major concern of emerging and re-emerging viral diseases has become an increasingly important area of public health concern, and it is of significance to anticipate future pandemic that would inevitably threaten human lives. The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a newly emerged virus that causes mild to severe pneumonia. Coronavirus disease (COVID-19) became a very much concerned issue worldwide after its super-spread across the globe and emerging viral diseases have not got specific and reliable diagnostic and treatments. As the COVID-19 pandemic brings about a massive life-loss across the globe, there is an unmet need to discover a promising and typically effective diagnosis and treatment to prevent super-spreading and mortality from being decreased or even eliminated. This study was carried out to overview nanotechnology-based diagnostic and treatment approaches for emerging and re-emerging viruses with the current treatment of the disease and shed light on nanotechnology's remarkable potential to provide more effective treatment and prevention to a special focus on recently emerged coronavirus.
Collapse
|
22
|
Recent Advances in Nanomaterials for Dermal and Transdermal Applications. COLLOIDS AND INTERFACES 2021. [DOI: 10.3390/colloids5010018] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The stratum corneum, the most superficial layer of the skin, protects the body against environmental hazards and presents a highly selective barrier for the passage of drugs and cosmetic products deeper into the skin and across the skin. Nanomaterials can effectively increase the permeation of active molecules across the stratum corneum and enable their penetration into deeper skin layers, often by interacting with the skin and creating the distinct sites with elevated local concentration, acting as reservoirs. The flux of the molecules from these reservoirs can be either limited to the underlying skin layers (for topical drug and cosmeceutical delivery) or extended across all the sublayers of the epidermis to the blood vessels of the dermis (for transdermal delivery). The type of the nanocarrier and the physicochemical nature of the active substance are among the factors that determine the final skin permeation pattern and the stability of the penetrant in the cutaneous environment. The most widely employed types of nanomaterials for dermal and transdermal applications include solid lipid nanoparticles, nanovesicular carriers, microemulsions, nanoemulsions, and polymeric nanoparticles. The recent advances in the area of nanomaterial-assisted dermal and transdermal delivery are highlighted in this review.
Collapse
|
23
|
Nonsurgical treatment of skin cancer with local delivery of bioadhesive nanoparticles. Proc Natl Acad Sci U S A 2021; 118:2020575118. [PMID: 33526595 DOI: 10.1073/pnas.2020575118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Keratinocyte-derived carcinomas, including squamous cell carcinoma (SCC), comprise the most common malignancies. Surgical excision is the therapeutic standard but is not always clinically feasible, and currently available alternatives are limited to superficial tumors. To address the need for a nonsurgical treatment for nodular skin cancers like SCC, we developed a bioadhesive nanoparticle (BNP) drug delivery system composed of biodegradable polymer, poly(lactic acid)-hyperbranched polyglycerol (PLA-HPG), encapsulating camptothecin (CPT). Nanoparticles (NPs) of PLA-HPG are nonadhesive NPs (NNPs), which are stealthy in their native state, but we have previously shown that conversion of the vicinal diols of HPG to aldehydes conferred NPs the ability to form strong covalent bonds with amine-rich surfaces. Herein, we show that these BNPs have significantly enhanced binding to SCC tumor cell surfaces and matrix proteins, thereby significantly enhancing the therapeutic efficacy of intratumoral drug delivery. Tumor injection of BNP-CPT resulted in tumor retention of CPT at ∼50% at 10 d postinjection, while CPT was undetectable in NNP-CPT or free (intralipid) CPT-injected tumors at that time. BNP-CPT also significantly reduced tumor burden, with a portion (∼20%) of BNP-CPT-treated established tumors showing histologic cure. Larger, more fully established PDV SCC tumors treated with a combination of BNP-CPT and immunostimulating CpG oligodeoxynucleotides exhibited enhanced survival relative to controls, revealing the potential for BNP delivery to be used along with local tumor immunotherapy. Taken together, these results indicate that percutaneous delivery of a chemotherapeutic agent via BNPs, with or without adjuvant immunostimulation, represents a viable, nonsurgical alternative for treating cutaneous malignancy.
Collapse
|
24
|
Shandilya R, Pathak N, Lohiya NK, Sharma RS, Mishra PK. Nanotechnology in reproductive medicine: Opportunities for clinical translation. Clin Exp Reprod Med 2020; 47:245-262. [PMID: 33227186 PMCID: PMC7711096 DOI: 10.5653/cerm.2020.03650] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
In recent years, nanotechnology has revolutionized global healthcare and has been predicted to exert a remarkable effect on clinical medicine. In this context, the clinical use of nanomaterials for cancer diagnosis, fertility preservation, and the management of infertility and other pathologies linked to pubertal development, menopause, sexually transmitted infections, and HIV (human immunodeficiency virus) has substantial promise to fill the existing lacunae in reproductive healthcare. Of late, a number of clinical trials involving the use of nanoparticles for the early detection of reproductive tract infections and cancers, targeted drug delivery, and cellular therapeutics have been conducted. However, most of these trials of nanoengineering are still at a nascent stage, and better synergy between pharmaceutics, chemistry, and cutting-edge molecular sciences is needed for effective translation of these interventions from bench to bedside. To bridge the gap between translational outcome and product development, strategic partnerships with the insight and ability to anticipate challenges, as well as an in-depth understanding of the molecular pathways involved, are highly essential. Such amalgamations would overcome the regulatory gauntlet and technical hurdles, thereby facilitating the effective clinical translation of these nano-based tools and technologies. The present review comprehensively focuses on emerging applications of nanotechnology, which holds enormous promise for improved therapeutics and early diagnosis of various human reproductive tract diseases and conditions.
Collapse
Affiliation(s)
- Ruchita Shandilya
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Neelam Pathak
- School of Life Sciences, University of Rajasthan, Jaipur, India
| | | | - Radhey Shyam Sharma
- Division of Reproductive Biology, Maternal and Child Health, Indian Council of Medical Research, New Delhi, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
25
|
Mucus-penetrating PEGylated polysuccinimide-based nanocarrier for intravaginal delivery of siRNA battling sexually transmitted infections. Colloids Surf B Biointerfaces 2020; 196:111287. [PMID: 32768985 DOI: 10.1016/j.colsurfb.2020.111287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 11/21/2022]
Abstract
Intravaginal delivery of siRNA for prevention of sexually transmitted infections faces obstacles such as the acidic environment and vaginal mucus barrier. To achieve effective protection and delivery of siRNA, we developed a polysuccinimide (PSI)-based nanocarrier (PSI-PEG-API-PMA, PPAP) by conjugating methoxy polyethylene glycol amine (Me-PEG-NH2, Mw 5000), 1-(3-aminopropyl)imidazole (API), and 1-pyrenemethylamine hydrochloride (PMA) to PSI. PPAP demonstrated a spherical self-assembled nanostructure before and after encapsulation of a model siRNA. Variable electrostatic interaction between API and siRNA at acidic vs. neutral pH accomplished significantly lower burst release at pH 4.2 (4 ± 1%) than pH 7.0 (26 ± 5%) within 1 h. PEGylation enabled siRNA-PPAP to achieve higher mucus penetration efficiency (64 ± 17%) than free siRNA (27 ± 5%) for 24 h. Moreover, in vitro study showed minimal toxicity, successful internalization of siRNA-PPAP in HeLa cells and improved gene knockdown (97.5 ± 0.4%). Overall, PPAP is promising for developing preventative treatments for battling sexually transmitted infections.
Collapse
|
26
|
Qi L, Zhu Q, Cao D, Liu T, Zhu KR, Chang K, Gao Q. Preparation and Properties of Stereocomplex of Poly(lactic acid) and Its Amphiphilic Copolymers Containing Glucose Groups. Polymers (Basel) 2020; 12:E760. [PMID: 32244536 PMCID: PMC7240496 DOI: 10.3390/polym12040760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/22/2020] [Accepted: 03/26/2020] [Indexed: 12/24/2022] Open
Abstract
The stereocomplex of poly(lactic acid) containing glucose groups (sc-PLAG) was prepared by solution blending from equal amounts of poly(l-lactic acid) (PLLA) and poly(d-lactic acid-co-glucose) (PDLAG), which were synthesized from l- and d-lactic acid and glucose by melt polycondensation. The methods, including 1H nuclear magnetic resonance spectroscopy (1H NMR), gel permeation chromatography (GPC), differential scanning calorimetry (DSC), X-ray diffraction (XRD), fourier transform infrared spectroscopy (FT-IR), thermogravimetric analysis (TGA), polarizing microscope (POM), scanning electron microscope (SEM), transmission electron microscope (TEM), and contact angle were used to determine the effects of the stereocomplexation of enantiomeric poly(lactic acid) (PLA) units, the amphiphilicity due to glucose residues and lactic acid units, and the interaction of glucose residues with lactic units on the crystallization performance, hydrophilicity, thermal stability, and morphology of samples. The results showed PDLAG was multi-armed, and partial OH groups of glucose residues in PDLAG might remain unreacted. The molecular weight (Mw), dispersity (Ɖ), and glucose proportion in the chain of PDLAG thereby had significant effects on sc-PLAG. There were the stereocomplexation of enantiomeric lactic units and the amphiphilic self-assembly of PDLAG in sc-PLAG, which resulted in glucose groups mainly in the surface phase and lactic units in the bulk phase. The sc-PLAG only possessed the stereocomplex crystal owing to the interaction between nearly equimolar of l-lactic units of PLLA and d-lactic units of PDLAG, and had no homo-crystallites of l- or d-lactic units, which improved the melting temperature (Tm) of sc-PLAG about 50 °C higher than that of PLLA. Glucose groups in sc-PLAG played an important role by forming heterogeneous nucleation, promoting amphiphilic self-assembly, and affecting the ordered arrangement of lactic units. The glass transition temperature (Tg), the melting temperature (Tm), crystallinity, crystallization rate, and water absorption of sc-PLAG showed similar changes with the increased glucose content in feeding. All these parameters increased at first, and the maximum appeared as glucose content in feeding about 2%, such as the maximum crystallinity of 48.8% and the maximum water absorption ratio being 11.7%. When glucose content in feeding continued increasing, all these performances showed a downward trend due to the decrease of arrangement regularity of lactic acid chains caused by glucose groups. Moreover, the contact angle of sc-PLAG decreased gradually with the increased glucose content in feeding to obtain the minimum 77.5° as the glucose content in feeding being 5%, while that of PLLA was 85.0°. The sc-PLAG possessed a regular microsphere structure, and its microspheres with a diameter of about 200 nm could be observed. In conclusion, sc-PLAG containing proper glucose amount could effectively enhance the crystallinity, hydrophilicity, and thermal stability of PLA material, which is useful for drug delivery, a scaffold for tissue engineering, and other applications of biomedicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qinwei Gao
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China; (L.Q.); (Q.Z.); (D.C.); (T.L.); (K.R.Z.); (K.C.)
| |
Collapse
|
27
|
Gong Y, Chowdhury P, Nagesh PKB, Rahman MA, Zhi K, Yallapu MM, Kumar S. Novel elvitegravir nanoformulation for drug delivery across the blood-brain barrier to achieve HIV-1 suppression in the CNS macrophages. Sci Rep 2020; 10:3835. [PMID: 32123217 PMCID: PMC7052245 DOI: 10.1038/s41598-020-60684-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023] Open
Abstract
The use of antiretroviral therapy (ART) has remarkably decreased the morbidity associated with HIV-1 infection, however, the prevalence of HIV-1-associated neurocognitive disorders (HAND) is still increasing. The blood-brain barrier (BBB) is the major impediment for penetration of antiretroviral drugs, causing therapeutics to reach only suboptimal level to the brain. Conventional antiretroviral drug regimens are not sufficient to improve the treatment outcomes of HAND. In our recent report, we have developed a poloxamer-PLGA nanoformulation loaded with elvitegravir (EVG), a commonly used antiretroviral drug. The nanoformulated EVG is capable of elevating intracellular drug uptake and simultaneously enhance viral suppression in HIV-1-infected macrophages. In this work, we identified the clinical parameters including stability, biocompatibility, protein corona, cellular internalization pathway of EVG nanoformulation for its potential clinical translation. We further assessed the ability of this EVG nanoformulation to cross the in vitro BBB model and suppress the HIV-1 in macrophage cells. Compared with EVG native drug, our EVG nanoformulation demonstrated an improved BBB model penetration cross the in vitro BBB model and an enhanced HIV-1 suppression in HIV-1-infected human monocyte-derived macrophages after crossing the BBB model without altering the BBB model integrity. Overall, this is an innovative and optimized treatment strategy that has a potential for therapeutic interventions in reducing HAND.
Collapse
Affiliation(s)
- Yuqing Gong
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Prashanth K B Nagesh
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Mohammad A Rahman
- National Institute of Environmental Health Sciences, Durham, NC, 27703, USA
| | - Kaining Zhi
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Department of Microbiology and Immunology, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA.
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
28
|
Tyo KM, Lasnik AB, Zhang L, Mahmoud M, Jenson AB, Fuqua JL, Palmer KE, Steinbach-Rankins JM. Sustained-release Griffithsin nanoparticle-fiber composites against HIV-1 and HSV-2 infections. J Control Release 2020; 321:84-99. [PMID: 32035194 DOI: 10.1016/j.jconrel.2020.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023]
Abstract
Human immunodeficiency virus (HIV-1) and herpes simplex virus 2 (HSV-2) affect hundreds of millions of people worldwide. The antiviral lectin, Griffithsin (GRFT), has been shown to be both safe and efficacious against HSV-2 and HIV-1 infections in vivo. The goal of this work was to develop a multilayered nanoparticle (NP)-electrospun fiber (EF) composite to provide sustained-release of GRFT, and to examine its safety and efficacy in a murine model of lethal HSV-2 infection. Composites were fabricated from polycaprolactone (PCL) fibers surrounding polyethylene oxide (PEO) fibers that incorporated methoxy poly(ethylene glycol)-b-poly(lactide-co-glycolide) (mPEG-PLGA) GRFT NPs. GRFT loading and release were determined via ELISA, showing that NP-EF composites achieved high GRFT loading, and provided sustained-release of GRFT for up to 90 d. The in vitro efficacy of GRFT NP-EFs was assessed using HIV-1 pseudovirus assays, demonstrating complete in vitro protection against HIV-1 infection. Additionally, sustained-release NP-EFs, administered 24 h prior to infection, prevented against a lethal dose of HSV-2 infection in a murine model. In parallel, histology and cytokine expression from murine reproductive tracts and vaginal lavages collected 24 and 72 h post-administration were similar to untreated mice, suggesting that NP-EF composites may be a promising and safe sustained-delivery platform to prevent HSV-2 infection. Future work will evaluate the ability to provide prolonged protection against multiple virus challenges, and different administration times with respect to infection.
Collapse
Affiliation(s)
- Kevin M Tyo
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, KY, United States; Center for Predictive Medicine, Louisville, KY, United States
| | - Amanda B Lasnik
- Center for Predictive Medicine, Louisville, KY, United States
| | - Longyun Zhang
- Center for Predictive Medicine, Louisville, KY, United States; Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY, United States
| | - Mohamed Mahmoud
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, KY, United States; Center for Predictive Medicine, Louisville, KY, United States
| | - Alfred B Jenson
- James Graham Brown Cancer Center, University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States
| | - Joshua L Fuqua
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, KY, United States; Center for Predictive Medicine, Louisville, KY, United States
| | - Kenneth E Palmer
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, KY, United States; Center for Predictive Medicine, Louisville, KY, United States; James Graham Brown Cancer Center, University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States; Department of Microbiology and Immunology, School of Medicine, University of Louisville, KY, United States
| | - Jill M Steinbach-Rankins
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, KY, United States; Center for Predictive Medicine, Louisville, KY, United States; James Graham Brown Cancer Center, University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States; Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY, United States; Department of Microbiology and Immunology, School of Medicine, University of Louisville, KY, United States.
| |
Collapse
|
29
|
Recent developments in functionalized polymer nanoparticles for efficient drug delivery system. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.nanoso.2019.100397] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
30
|
Topical Inserts: A Versatile Delivery Form for HIV Prevention. Pharmaceutics 2019; 11:pharmaceutics11080374. [PMID: 31374941 PMCID: PMC6723036 DOI: 10.3390/pharmaceutics11080374] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/21/2019] [Accepted: 07/23/2019] [Indexed: 11/16/2022] Open
Abstract
The development of topical inserts for the prevention of sexually transmitted infections (STIs), particularly human immunodeficiency virus (HIV), represents a promising alternative to oral and parenteral pre-exposure prophylaxis (PrEP) dosage forms. They may be used for vaginal and/or rectal administration of a variety of agents with antiviral activity. Topical inserts deliver drugs to the portal of viral entry, i.e., the genital or rectal mucosa, with low systemic exposure, and therefore are safer and have fewer side effects than systemic PrEP agents. They may dissolve fast, releasing the active drugs within minutes of insertion, or slowly for long-acting drug delivery. Furthermore, they are user-friendly being easy to administer, discreet and highly portable. They are also economical and easy to manufacture at scale and to distribute, with excellent stability and shelf-life. Altogether, topical inserts represent a particularly promising form of drug delivery for HIV and STI prevention. Highlighted within this review are end-user acceptability research dedicated to understanding preferred attributes for this form of drug delivery, advantages and disadvantages of the formulation platform options, considerations for their development, clinical assessment of select placebo prototypes, future directions, and the potential impact of this dosage form on the HIV prevention landscape.
Collapse
|
31
|
Rossi S, Vigani B, Sandri G, Bonferoni MC, Caramella CM, Ferrari F. Recent advances in the mucus-interacting approach for vaginal drug delivery: from mucoadhesive to mucus-penetrating nanoparticles. Expert Opin Drug Deliv 2019; 16:777-781. [DOI: 10.1080/17425247.2019.1645117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Silvia Rossi
- Department of Drug Science, University of Pavia, Pavia, Italy
| | - Barbara Vigani
- Department of Drug Science, University of Pavia, Pavia, Italy
| | | | | | | | - Franca Ferrari
- Department of Drug Science, University of Pavia, Pavia, Italy
| |
Collapse
|
32
|
Gong Y, Chowdhury P, Nagesh PKB, Cory TJ, Dezfuli C, Kodidela S, Singh A, Yallapu MM, Kumar S. Nanotechnology approaches for delivery of cytochrome P450 substrates in HIV treatment. Expert Opin Drug Deliv 2019; 16:869-882. [PMID: 31328582 DOI: 10.1080/17425247.2019.1646725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Introduction: Antiretroviral therapy (ART) has led to a significant reduction in HIV-1 morbidity and mortality. Many antiretroviral drugs (ARVs) are metabolized by cytochrome P450 (CYP) pathway, and the majority of these drugs are also either CYP inhibitors or inducers and few possess both activities. These CYP substrates, when used for HIV treatment in the conventional dosage form, have limitations such as low systemic bioavailability, potential drug-drug interactions, and short half-lives. Thus, an alternative mode of delivery is needed in contrast to conventional ARVs. Areas covered: In this review, we summarized the limitations of conventional ARVs in HIV treatment, especially for ARVs which are CYP substrates. We also discussed the preclinical and clinical studies using the nanotechnology strategy to overcome the limitations of these CYP substrates. The preclinical studies and clinical studies published from 2000 to February 2019 were discussed. Expert opinion: Since preclinical and clinical studies for prevention and treatment of HIV using nanotechnology approaches have shown considerable promise in recent years, nanotechnology could become an alternative strategy for daily oral therapy as a future treatment.
Collapse
Affiliation(s)
- Yuqing Gong
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Pallabita Chowdhury
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Prashanth K B Nagesh
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Theodore J Cory
- b Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Chelsea Dezfuli
- b Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Sunitha Kodidela
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Ajay Singh
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Murali M Yallapu
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Santosh Kumar
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| |
Collapse
|
33
|
Tyo KM, Minooei F, Curry KC, NeCamp SM, Graves DL, Fried JR, Steinbach-Rankins JM. Relating Advanced Electrospun Fiber Architectures to the Temporal Release of Active Agents to Meet the Needs of Next-Generation Intravaginal Delivery Applications. Pharmaceutics 2019; 11:E160. [PMID: 30987206 PMCID: PMC6523330 DOI: 10.3390/pharmaceutics11040160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/28/2019] [Accepted: 03/30/2019] [Indexed: 02/07/2023] Open
Abstract
Electrospun fibers have emerged as a relatively new delivery platform to improve active agent retention and delivery for intravaginal applications. While uniaxial fibers have been explored in a variety of applications including intravaginal delivery, the consideration of more advanced fiber architectures may offer new options to improve delivery to the female reproductive tract. In this review, we summarize the advancements of electrospun coaxial, multilayered, and nanoparticle-fiber architectures utilized in other applications and discuss how different material combinations within these architectures provide varied durations of release, here categorized as either transient (within 24 h), short-term (24 h to one week), or sustained (beyond one week). We seek to systematically relate material type and fiber architecture to active agent release kinetics. Last, we explore how lessons derived from these architectures may be applied to address the needs of future intravaginal delivery platforms for a given prophylactic or therapeutic application. The overall goal of this review is to provide a summary of different fiber architectures that have been useful for active agent delivery and to provide guidelines for the development of new formulations that exhibit release kinetics relevant to the time frames and the diversity of active agents needed in next-generation multipurpose applications.
Collapse
Affiliation(s)
- Kevin M Tyo
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA.
- Center for Predictive Medicine, Louisville, KY 40202, USA.
| | - Farnaz Minooei
- Department of Chemical Engineering, University of Louisville, Louisville, KY 40292, USA.
| | - Keegan C Curry
- Department of Biology, University of Louisville, Louisville, KY 40292, USA.
| | - Sarah M NeCamp
- Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY 40292, USA.
| | - Danielle L Graves
- Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY 40292, USA.
| | - Joel R Fried
- Department of Chemical Engineering, University of Louisville, Louisville, KY 40292, USA.
| | - Jill M Steinbach-Rankins
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA.
- Center for Predictive Medicine, Louisville, KY 40202, USA.
- Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY 40292, USA.
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
34
|
Mesquita L, Galante J, Nunes R, Sarmento B, das Neves J. Pharmaceutical Vehicles for Vaginal and Rectal Administration of Anti-HIV Microbicide Nanosystems. Pharmaceutics 2019; 11:pharmaceutics11030145. [PMID: 30917532 PMCID: PMC6472048 DOI: 10.3390/pharmaceutics11030145] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 12/27/2022] Open
Abstract
Prevention strategies play a key role in the fight against HIV/AIDS. Vaginal and rectal microbicides hold great promise in tackling sexual transmission of HIV-1, but effective and safe products are yet to be approved and made available to those in need. While most efforts have been placed in finding and testing suitable active drug candidates to be used in microbicide development, the last decade also saw considerable advances in the design of adequate carrier systems and formulations that could lead to products presenting enhanced performance in protecting from infection. One strategy demonstrating great potential encompasses the use of nanosystems, either with intrinsic antiviral activity or acting as carriers for promising microbicide drug candidates. Polymeric nanoparticles, in particular, have been shown to be able to enhance mucosal distribution and retention of promising antiretroviral compounds. One important aspect in the development of nanotechnology-based microbicides relates to the design of pharmaceutical vehicles that allow not only convenient vaginal and/or rectal administration, but also preserve or even enhance the performance of nanosystems. In this manuscript, we revise relevant work concerning the selection of vaginal/rectal dosage forms and vehicle formulation development for the administration of microbicide nanosystems. We also pinpoint major gaps in the field and provide pertinent hints for future work.
Collapse
Affiliation(s)
- Letícia Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Joana Galante
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
| | - Rute Nunes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Bruno Sarmento
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, 4585-116 Gandra, Portugal.
| | - José das Neves
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, 4585-116 Gandra, Portugal.
| |
Collapse
|
35
|
Xu Y, Lu S, Liu Q, Hong Y, Xu B, Ping Q, Jin X, Shen Y, Webster TJ, Rao Y. Preparation, intestinal segment stability, and mucoadhesion properties of novel thymopentin-loaded chitosan derivatives coated with poly (n-butyl) cyanoacrylate nanoparticles. Int J Nanomedicine 2019; 14:1659-1668. [PMID: 30880971 PMCID: PMC6404670 DOI: 10.2147/ijn.s194529] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background In order to develop a promising carrier for the oral delivery of proteins and peptide drugs, a novel bioadhesive nanocarrier of chitosan (CTS) derivatives coated with poly (n-butyl) cyanoacrylate nanoparticles (PBCA-NPs) was prepared in this study. Methods Three different thymopentin (TP5)-loaded nanoparticles were prepared in the present study. TP5-PBCA-NPs were developed by modifying an emulsion polymerization method, and CTS and chitosan-glutathione (CG) derivative-coated PBCA nanoparticles were obtained from the electrostatic interactions between CTS or CG with negatively charged PBCA nanoparticles. Results The particle sizes of TP5-PBCA-NPs, TP5-CTS-PBCA-NPs, and TP5-CG-PBCA-NPs were 212.3±6.9, 274.6±8.2, and 310.4±7.5 nm, respectively, while the respective zeta potentials were -22.6±0.76, 23.3±1.2, and 34.6±1.6 mV with encapsulation efficiencies of 79.37%±2.15%, 74.21%±2.13%, and 72.65%±1.48%, respectively. An everted intestinal ring method indicated that drug stability was remarkably improved after incorporation into the nanoparticles, especially the CG-coated nanoparticles. The mucus layer retention rates for CTS- and CG-coated nanoparticles were 1.43 and 1.83 times that of the uncoated nanoparticles, respectively, using ex vivo mucosa. The in vivo mucoadhesion study illustrated that the transfer of uncoated PBCA-NPs from the stomach to the intestine was faster than that of CTS-PBCA-NPs and CG-PBCA-NPs, while the CG-PBCA-NPs presented the best intestinal retentive characteristic. Conclusion In summary, this study demonstrated the feasibility and benefit of orally delivering peptide drugs using novel CTS derivative-coated nanoparticles with optimal stability and bioadhesive properties.
Collapse
Affiliation(s)
- Ying Xu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Shengzhe Lu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yun Hong
- Department of Pharmacy, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China,
| | - Bohui Xu
- Department of Pharmacy, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Qineng Ping
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuefeng Jin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Shen
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA,
| | - Yuefeng Rao
- Department of Pharmacy, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China, .,Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,
| |
Collapse
|
36
|
Rai M, Jamil B. Nanoformulations: A Valuable Tool in the Therapy of Viral Diseases Attacking Humans and Animals. Nanotheranostics 2019. [PMCID: PMC7121811 DOI: 10.1007/978-3-030-29768-8_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Various viruses can be considered as one of the most frequent causes of human diseases, from mild illnesses to really serious sicknesses that end fatally. Numerous viruses are also pathogenic to animals and plants, and many of them, mutating, become pathogenic also to humans. Several cases of affecting humans by originally animal viruses have been confirmed. Viral infections cause significant morbidity and mortality in humans, the increase of which is caused by general immunosuppression of the world population, changes in climate, and overall globalization. In spite of the fact that the pharmaceutical industry pays great attention to human viral infections, many of clinically used antivirals demonstrate also increased toxicity against human cells, limited bioavailability, and thus, not entirely suitable therapeutic profile. In addition, due to resistance, a combination of antivirals is needed for life-threatening infections. Thus, the development of new antiviral agents is of great importance for the control of virus spread. On the other hand, the discovery and development of structurally new antivirals represent risks. Therefore, another strategy is being developed, namely the reformulation of existing antivirals into nanoformulations and investigation of various metal and metalloid nanoparticles with respect to their diagnostic, prophylactic, and therapeutic antiviral applications. This chapter is focused on nanoscale materials/formulations with the potential to be used for the treatment or inhibition of the spread of viral diseases caused by human immunodeficiency virus, influenza A viruses (subtypes H3N2 and H1N1), avian influenza and swine influenza viruses, respiratory syncytial virus, herpes simplex virus, hepatitis B and C viruses, Ebola and Marburg viruses, Newcastle disease virus, dengue and Zika viruses, and pseudorabies virus. Effective antiviral long-lasting and target-selective nanoformulations developed for oral, intravenous, intramuscular, intranasal, intrarectal, intravaginal, and intradermal applications are discussed. Benefits of nanoparticle-based vaccination formulations with the potential to secure cross protection against divergent viruses are outlined as well.
Collapse
Affiliation(s)
- Mahendra Rai
- Department of Biotechnology, Nanobiotechnology Laboratory, Amravati, Maharashtra, India, Department of Chemistry, Federal University of Piauí, Teresina, Piauí Brazil
| | - Bushra Jamil
- Department of DMLS, University of Lahore, Islamabad, Pakistan
| |
Collapse
|
37
|
Kozlowski PA, Aldovini A. Mucosal Vaccine Approaches for Prevention of HIV and SIV Transmission. CURRENT IMMUNOLOGY REVIEWS 2019; 15:102-122. [PMID: 31452652 PMCID: PMC6709706 DOI: 10.2174/1573395514666180605092054] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Optimal protective immunity to HIV will likely require that plasma cells, memory B cells and memory T cells be stationed in mucosal tissues at portals of viral entry. Mucosal vaccine administration is more effective than parenteral vaccine delivery for this purpose. The challenge has been to achieve efficient vaccine uptake at mucosal surfaces, and to identify safe and effective adjuvants, especially for mucosally administered HIV envelope protein immunogens. Here, we discuss strategies used to deliver potential HIV vaccine candidates in the intestine, respiratory tract, and male and female genital tract of humans and nonhuman primates. We also review mucosal adjuvants, including Toll-like receptor agonists, which may adjuvant both mucosal humoral and cellular immune responses to HIV protein immunogens.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Anna Aldovini
- Department of Medicine, and Harvard Medical School, Boston Children’s Hospital, Department of Pediatrics, Boston MA, 02115, USA
| |
Collapse
|
38
|
Suh H, Lewis J, Fong L, Ramseier JY, Carlson K, Peng Z, Yin ES, Saltzman WM, Girardi M. Biodegradable bioadhesive nanoparticle incorporation of broad-spectrum organic sunscreen agents. Bioeng Transl Med 2019; 4:129-140. [PMID: 30680324 PMCID: PMC6336670 DOI: 10.1002/btm2.10092] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 11/17/2022] Open
Abstract
Conventional emulsion-based sunscreen formulations are limited by postapplication epicutaneous penetration that increases the risk of allergic dermatitis, cellular damage, and filter photodegradation upon ultraviolet radiation (UVR) exposure. Encapsulation of the UVB filter padimate O within bioadhesive biodegradable nanoparticles (BNPs) composed of poly(d,l-lactic acid)-hyperbranched polyglycerol was previously shown to enhance UVR protection while preventing skin absorption. Herein, we assess the capacity of BNP co-incorporation of avobenzone and octocrylene to provide broad-spectrum UVR protection. The ratio of UV filters within nanoparticles (NPs) was optimized for filter-filter stabilization upon UV irradiation and maximum drug loading. In vitro water-resistance test showed significant particle retention at 85% over 3 hr. In a pilot clinical study, protection against UVR-induced erythema of BNPs was found to be comparable to the FDA standard P2. Thus, sunscreen formulations utilizing BNP incorporation of a combination of organic filters may offer key safety and performance advantages.
Collapse
Affiliation(s)
- Hee‐Won Suh
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Julia Lewis
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - Linda Fong
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Julie Ying Ramseier
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - Kacie Carlson
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - Zheng‐Hong Peng
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Emily Sara Yin
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - W. Mark Saltzman
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Michael Girardi
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| |
Collapse
|
39
|
Han J, Ma B, Liu H, Wang T, Wang F, Xie C, Li M, Liu H, Ge S. Hydroxyapatite nanowires modified polylactic acid membrane plays barrier/osteoinduction dual roles and promotes bone regeneration in a rat mandible defect model. J Biomed Mater Res A 2018; 106:3099-3110. [DOI: 10.1002/jbm.a.36502] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/13/2018] [Accepted: 07/02/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Jing Han
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration; School of Stomatology, Shandong University; Jinan 250012 China
- Department of Periodontology; School of Stomatology, Shandong University; Jinan 250012 China
| | - Baojin Ma
- State Key Laboratory of Crystal Materials; Shandong University; Jinan 250100 China
| | - Hongrui Liu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration; School of Stomatology, Shandong University; Jinan 250012 China
- Department of Bone Metabolism; School of Stomatology Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration; Jinan 250100 China
| | - Ting Wang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration; School of Stomatology, Shandong University; Jinan 250012 China
- Department of Periodontology; School of Stomatology, Shandong University; Jinan 250012 China
| | - Fang Wang
- Department of Periodontology; Yantai Stomatological Hospital; Yantai 264001 China
| | - Chengjia Xie
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration; School of Stomatology, Shandong University; Jinan 250012 China
- Department of Periodontology; School of Stomatology, Shandong University; Jinan 250012 China
| | - Minqi Li
- Department of Bone Metabolism; School of Stomatology Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration; Jinan 250100 China
| | - Hong Liu
- State Key Laboratory of Crystal Materials; Shandong University; Jinan 250100 China
| | - Shaohua Ge
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration; School of Stomatology, Shandong University; Jinan 250012 China
- Department of Periodontology; School of Stomatology, Shandong University; Jinan 250012 China
| |
Collapse
|
40
|
Monroe M, Flexner C, Cui H. Harnessing nanostructured systems for improved treatment and prevention of HIV disease. Bioeng Transl Med 2018; 3:102-123. [PMID: 30065966 PMCID: PMC6063869 DOI: 10.1002/btm2.10096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022] Open
Abstract
Combination antiretroviral therapy effectively controls human immunodeficiency virus (HIV) viral replication, delaying the progression to acquired immune deficiency syndrome and improving and extending quality of life of patients. However, the inability of antiretroviral therapeutics to target latent virus and their poor penetration of viral reserve tissues result in the need for continued treatment for the life of the patient. Side effects from long-term antiretroviral use and the development of drug resistance due to patient noncompliance are also continuing problems. Nanostructured systems of antiretroviral therapeutics have the potential to improve targeted delivery to viral reservoirs, reduce drug toxicity, and increase dosing intervals, thereby improving treatment outcomes and enhancing patient adherence. Despite these advantages, very few nanostructured antiretroviral delivery systems have made it to clinical trials due to challenges in preclinical and clinical development. In this context, we review the current challenges in HIV disease management, and the recent progress in leveraging the unique performance of nanostructured systems in therapeutic delivery for improved treatment and prevention of this incurable human disease.
Collapse
Affiliation(s)
- Maya Monroe
- Dept. of Chemical and Biomolecular Engineering The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218.,Institute for NanoBioTechnology The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218
| | - Charles Flexner
- Div. of Clinical Pharmacology and Infectious Diseases Johns Hopkins University School of Medicine and Bloomberg School of Public Health Baltimore MD 21205
| | - Honggang Cui
- Dept. of Chemical and Biomolecular Engineering The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218.,Institute for NanoBioTechnology The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218.,Dept. of Oncology, Sidney Kimmel Comprehensive Cancer Center The Johns Hopkins University School of Medicine Baltimore MD 21205.,Center for Nanomedicine The Wilmer Eye Institute, The Johns Hopkins University School of Medicine Baltimore MD 21231
| |
Collapse
|
41
|
Viswanadh MK, Muthu MS. Targeted bioadhesive nanomedicine: an effective approach for synergistic drug delivery to cancers. Nanomedicine (Lond) 2018; 13:1401-1403. [DOI: 10.2217/nnm-2018-0114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Matte Kasi Viswanadh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
42
|
Approaches in Polymeric Nanoparticles for Vaginal Drug Delivery: A Review of the State of the Art. Int J Mol Sci 2018; 19:ijms19061549. [PMID: 29882846 PMCID: PMC6032388 DOI: 10.3390/ijms19061549] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/20/2022] Open
Abstract
The vagina is a region of administration with a high contact surface to obtain local or systemic effects. This anatomical area represents special interest for government health systems for different sexually transmitted infections. However, the chemical changes of the vagina, as well as its abundant mucus in continuous exchange, act as a barrier and a challenge for the development of new drugs. For these purposes, the development of new pharmaceutical forms based on nanoparticles has been shown to offer various advantages, such as bioadhesion, easy penetration of the mucosa, and controlled release, in addition to decreasing the adverse effects of conventional pharmaceutical forms. In order to obtain nanoparticles for vaginal administration, the use of polymers of natural and synthetic origin including biodegradable and non-biodegradable systems have gained great interest both in nanospheres and in nanocapsules. The main aim of this review is to provide an overview of the development of nanotechnology for vaginal drug release, analyzing the different compositions of polymeric nanoparticles, and emphasizing new trends in each of the sections presented. At the end of this review, a section analyzes the properties of the vehicles employed for the administration of nanoparticles and discusses how to take advantage of the properties that they offer. This review aims to be a reference guide for new formulators interested in the vaginal route.
Collapse
|
43
|
Fenton OS, Olafson KN, Pillai PS, Mitchell MJ, Langer R. Advances in Biomaterials for Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705328. [PMID: 29736981 PMCID: PMC6261797 DOI: 10.1002/adma.201705328] [Citation(s) in RCA: 464] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/12/2018] [Indexed: 04/14/2023]
Abstract
Advances in biomaterials for drug delivery are enabling significant progress in biology and medicine. Multidisciplinary collaborations between physical scientists, engineers, biologists, and clinicians generate innovative strategies and materials to treat a range of diseases. Specifically, recent advances include major breakthroughs in materials for cancer immunotherapy, autoimmune diseases, and genome editing. Here, strategies for the design and implementation of biomaterials for drug delivery are reviewed. A brief history of the biomaterials field is first established, and then commentary on RNA delivery, responsive materials development, and immunomodulation are provided. Current challenges associated with these areas as well as opportunities to address long-standing problems in biology and medicine are discussed throughout.
Collapse
Affiliation(s)
- Owen S Fenton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Katy N Olafson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Padmini S Pillai
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, School of Engineering and Applied Science, Philadelphia, PA, 19104, USA
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|