1
|
Sahoo GP, Rai VK, Pradhan D, Halder J, Rajwar TK, Mahanty R, Saha I, Mishra A, Dash P, Dash C, Al-Tamimi J, Manoharadas S, Kar B, Ghosh G, Rath G. A doxorubicin loaded chitosan-poloxamer in situ implant for the treatment of breast cancer. RSC Adv 2024; 14:33952-33967. [PMID: 39463476 PMCID: PMC11503159 DOI: 10.1039/d4ra06253a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024] Open
Abstract
Breast cancer is a serious concern for many women worldwide. Drug-loaded implants have shown several benefits over systemic administrations. To provide anti-cancer drugs with controlled release and reduced systemic toxicity, biodegradable in situ implants have attracted a lot of attention. In the present study, we aimed to design and optimize a doxorubicin-loaded chitosan-poloxamer in situ implant for breast cancer treatment. Utilizing Box-Behnken Design and a Quality-by-Design (QbD) methodology, the in situ implant was prepared with chitosan (X1), poloxamer 407 concentration (X2), and stirring time (X3) as the independent variables. It was characterized for its in vitro gelation time, pH, rheology, and morphology, and evaluated based on drug release profile, in vitro cytotoxicity activities, in vitro anti-inflammatory potential, in vitro cellular uptake, and in vivo anti-inflammatory and pharmacokinetics to ensure their therapeutic outcomes. The results revealed that the prepared formulation showed a gelation time of 26 ± 0.2 s with a viscosity of 8312.6 ± 114.2 cPs at 37 °C. The developed formulation showed better cytotoxic activity in MCF-7 cell lines compared to the free drug solution. It demonstrated reduced levels of pro-inflammatory cytokines in RAW 264.7 macrophages. Further, the prepared in situ implant increases the intracellular accumulation of DOX in the MCF-7 cells. The in vivo pharmacokinetic investigations depicted an increase in t 1/2 and a decrease in AUC of the developed formulation resulting in prolonged drug release and there could be a lower drug concentration in the bloodstream than for the free drug. Therefore, the developed in situ implant may offer a viable option for breast cancer treatment.
Collapse
Affiliation(s)
- Guru Prasanna Sahoo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Vineet Kumar Rai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Deepak Pradhan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Jitu Halder
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Tushar Kanti Rajwar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ritu Mahanty
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ivy Saha
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ajit Mishra
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Priyanka Dash
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Chandan Dash
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Jameel Al-Tamimi
- Zoology Department, College of Science, King Saud University P. O. Box 2455 Riyadh Saudi Arabia
| | - Salim Manoharadas
- Department of Botany and Microbiology, College of Science, King Saud University P. O. Box. 2454 Riyadh 11451 Saudi Arabia
| | - Biswakanth Kar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Goutam Ghosh
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| |
Collapse
|
2
|
Hiew TN, Solomos MA, Kafle P, Polyzois H, Zemlyanov DY, Punia A, Smith D, Schenck L, Taylor LS. The importance of surface composition and wettability on the dissolution performance of high drug loading amorphous dispersion formulations. J Pharm Sci 2024:S0022-3549(24)00428-3. [PMID: 39349295 DOI: 10.1016/j.xphs.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/02/2024]
Abstract
One of the limitations with an amorphous solid dispersion (ASD) formulation strategy is low drug loading. Hydrophobic drugs have poor wettability and require a substantial amount of polymer to stabilize the amorphous drug and facilitate release. Using grazoprevir and hypromellose acetate succinate as model drug and polymer respectively, the interplay between particle surface composition, particle wettability, and release performance was investigated. A hierarchical particle approach was used where the surfaces of high drug loading ASDs generated by either solvent evaporation or co-precipitation were further modified with a secondary excipient (i.e., polymer or wetting agent). The surface-modified particles were characterized for drug release, wettability, morphology, and surface composition using two-stage dissolution studies, contact angle measurements, scanning electron microscopy, and X-ray photoelectron spectroscopy, respectively. Despite surface modification with hydrophilic polymers, hierarchical cPAD particles did not consistently exhibit good release performance. Contact angle measurements showed that the secondary excipient had a profound impact on particle wettability. Particles with good wettability showed improved drug release relative to particles that did not wet well, even with similar drug loadings. These observations underscore the intricate interplay between particle wettability and performance in amorphous dispersion formulations and illustrate a promising hierarchical particle approach to formulate high drug loading amorphous dispersions with improved dissolution performance.
Collapse
Affiliation(s)
- Tze Ning Hiew
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Marina A Solomos
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Prapti Kafle
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Hector Polyzois
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ashish Punia
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel Smith
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Luke Schenck
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States.
| |
Collapse
|
3
|
Shobeirean A, Attar H, Varshochian R, Rezvanfar MA. Glatiramer acetate in situ forming gel, a new approach for multiple sclerosis treatment. Daru 2024:10.1007/s40199-024-00532-z. [PMID: 39225953 DOI: 10.1007/s40199-024-00532-z] [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: 03/25/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Glatiramer acetate (GA), a commonly used treatment for multiple sclerosis (MS), requires long-term frequent injections to ensure its effectiveness. This often leads to adverse effects, patient noncompliance, and economic inefficiency. OBJECTIVES In this study, poloxamer, as a thermosensitive polymer modified by chitosan (CS) and hyaluronic acid (HA), was employed to prepare an in situ forming prolonged release formulation of GA to overcome the problems derived from frequent repeated injections and to enhance the patient compliance. METHODS The sol-gel formulation was produced through a cold method and optimized using design of experiments. The final product was characterized in terms of gelation time (GT), rheological behaviors, morphological properties, assay, and drug release kinetics. RESULTS The in vitro release rate of GA during the first 24 h was quite rapid, but then it continued at a slower rate of 0.05 mg ml-1h-1. The in vivo analysis after the subcutaneous injections showed lower levels of IL-5, IL-13, and uric acid (UA) in mice treated with the gel formulation compared with those receiving free GA in the first few days. However, after 10 days, significantly higher concentrations were detected, which continued to increase slowly. CONCLUSION It can be concluded that the designed thermosensitive sol-gel formula is capable of extending the effectiveness of GA and can be considered as a promising sustained release formulation for the treatment of MS.
Collapse
Affiliation(s)
- Anahita Shobeirean
- Department of Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Attar
- Department of Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Reyhaneh Varshochian
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Amin Rezvanfar
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Yang Y, Nan W, Zhang R, Shen S, Wu M, Zhong S, Zhang Y, Cui X. Fabrication of carboxymethyl cellulose-based thermo-sensitive hydrogels and inhibition of corneal neovascularization. Int J Biol Macromol 2024; 261:129933. [PMID: 38309411 DOI: 10.1016/j.ijbiomac.2024.129933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/19/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
Corneal neovascularization (CNV) is a common multifactorial sequela of anterior corneal segment inflammation, which could lead to visual impairment and even blindness. The main treatments available are surgical sutures and invasive drug injections, which could cause serious ocular complications. To solve this problem, a thermo-sensitive drug-loaded hydrogel with high transparency was prepared in this study, which could achieve the sustained-release of drugs without affecting normal vision. In briefly, the thermo-sensitive hydrogel (PFNOCMC) was prepared from oxidized carboxymethyl cellulose (OCMC) and aminated poloxamer 407 (PF127-NH2). The results proved the PFNOCMC hydrogels possess high transparency, suitable gel temperature and time. In the CNV model, the PFNOCMC hydrogel loading bone morphogenetic protein 4 (BMP4) showed significant inhibition of CNV, this is due to the hydrogel allowed the drug to stay longer in the target area. The animal experiments on the ocular surface were carried out, which proved the hydrogel had excellent biocompatibility, and could realize the sustained-release of loaded drugs, and had a significant inhibitory effect on the neovascularization after ocular surface surgery. In conclusion, PFNOCMC hydrogels have great potential as sustained-release drug carriers in the biomedical field and provide a new minimally invasive option for the treatment of neovascular ocular diseases.
Collapse
Affiliation(s)
- Yongyan Yang
- College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Weijin Nan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Ruiting Zhang
- College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Sitong Shen
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Meiliang Wu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Shuangling Zhong
- College of Resources and Environment, Jilin Agricultural University, Changchun 130118, PR China
| | - Yan Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China.
| | - Xuejun Cui
- College of Chemistry, Jilin University, Changchun 130012, PR China; Weihai Institute for Bionics-Jilin University, Weihai 264400, PR China.
| |
Collapse
|
5
|
Hendricksen AT, Ezzatpour S, Pulukuri AJ, Ryan AT, Flanagan TJ, Frantz W, Buchholz DW, Ortega V, Monreal IA, Sahler JM, Nielsen AE, Aguilar HC, Mancini RJ. Thermophobic Trehalose Glycopolymers as Smart C-Type Lectin Receptor Vaccine Adjuvants. Adv Healthc Mater 2023; 12:e2202918. [PMID: 37002787 PMCID: PMC11212414 DOI: 10.1002/adhm.202202918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/06/2023] [Indexed: 04/04/2023]
Abstract
Herein, this work reports the first synthetic vaccine adjuvants that attenuate potency in response to small, 1-2 °C changes in temperature about their lower critical solution temperature (LCST). Adjuvant additives significantly increase vaccine efficacy. However, adjuvants also cause inflammatory side effects, such as pyrexia, which currently limits their use. To address this, a thermophobic vaccine adjuvant engineered to attenuate potency at temperatures correlating to pyrexia is created. Thermophobic adjuvants are synthesized by combining a rationally designed trehalose glycolipid vaccine adjuvant with thermoresponsive poly-N-isoporpylacrylamide (NIPAM) via reversible addition fragmentation chain transfer (RAFT) polymerization. The resulting thermophobic adjuvants exhibit LCSTs near 37 °C, and self-assembled into nanoparticles with temperature-dependent sizes (90-270 nm). Thermophobic adjuvants activate HEK-mMINCLE and other innate immune cell lines as well as primary mouse bone marrow derived dendritic cells (BMDCs) and bone marrow derived macrophages (BMDMs). Inflammatory cytokine production is attenuated under conditions mimicking pyrexia (above the LCST) relative to homeostasis (37 °C) or below the LCST. This thermophobic behavior correlated with decreased adjuvant Rg is observed by DLS, as well as glycolipid-NIPAM shielding interactions are observed by NOESY-NMR. In vivo, thermophobic adjuvants enhance efficacy of a whole inactivated influenza A/California/04/2009 virus vaccine, by increasing neutralizing antibody titers and CD4+ /44+ /62L+ lung and lymph node central memory T cells, as well as providing better protection from morbidity after viral challenge relative to unadjuvanted control vaccine. Together, these results demonstrate the first adjuvants with potency regulated by temperature. This work envisions that with further investigation, this approach can enhance vaccine efficacy while maintaining safety.
Collapse
Affiliation(s)
- Aaron T. Hendricksen
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Anunay J. Pulukuri
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Austin T. Ryan
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Tatum J. Flanagan
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - William Frantz
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - David W. Buchholz
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Victoria Ortega
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Isaac A. Monreal
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Julie M. Sahler
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Amy E. Nielsen
- Astante Therapeutics Inc.120 N Pine Street, Suite 270ASpokaneWA99202USA
| | - Hector C. Aguilar
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Rock J. Mancini
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
- Department of Chemistry and BiochemistryMiami University651 E. High StreetOxfordOH45056USA
| |
Collapse
|
6
|
White JM, Garza A, Griebler JJ, Bates FS, Calabrese MA. Engineering the Structure and Rheological Properties of P407 Hydrogels via Reverse Poloxamer Addition. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:5084-5094. [PMID: 36971824 PMCID: PMC10593112 DOI: 10.1021/acs.langmuir.3c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Aqueous solutions of poloxamer 407 (P407), a commercially available and nontoxic ABA triblock polymer (PEO-PPO-PEO), undergo a solution-to-gel transition with increasing temperature and are promising candidates for injectable therapeutics. The gel transition temperature, modulus, and structure are all dictated by polymer concentration, preventing independent tuning of these properties. Here, we show that addition of BAB reverse poloxamers (RPs) to P407-based solutions dramatically alters the gelation temperature, modulus, and morphology. Gelation temperature and RP localization within the hydrogel are dictated by RP solubility. Highly soluble RPs increase gelation temperature and incorporate primarily into the micelle corona regions. Alternatively, RPs with low aqueous solubility decrease gelation temperature and associate within the micelle core and core-corona interface. These differences in RP localization have significant implications for the hydrogel modulus and microstructure. The ability to tune gelation temperature, modulus, and structure through RP addition allows for the design of thermoresponsive materials with specific properties that are unobtainable with neat P407-based hydrogels.
Collapse
Affiliation(s)
- Joanna M White
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Ave SE, Minneapolis, Minnesota 55455, United States
| | - Ally Garza
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley 1201 W University Drive, Edinburg, Texas 78539, United States
| | - James J Griebler
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, 600 S Mathews Ave, Urbana, Illinois 61801, United States
| | - Frank S Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Ave SE, Minneapolis, Minnesota 55455, United States
| | - Michelle A Calabrese
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Ave SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
7
|
Duong HTT, Yin Y, Le TMD, Jeong JH, Lee DS. Highly Prolonged Release of the Cancer Vaccine and Immunomodulator via a Two-Layer Biodegradable Microneedle for Prophylactic Treatment of Metastatic Cancer. Biomacromolecules 2023; 24:1209-1219. [PMID: 36802451 DOI: 10.1021/acs.biomac.2c01270] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Simultaneous sustained release of cancer vaccines and immunomodulators may effectively trigger durable immune responses and avoid multiple administrations. Here, we established a biodegradable microneedle (bMN) based on a biodegradable copolymer matrix made of polyethylene glycol (PEG) and poly(sulfamethazine ester urethane) (PSMEU). This bMN was applied to the skin and slowly degraded in the epidermis/dermis layers. Then, the complexes composed of a positively charged polymer (DA3), cancer DNA vaccine (pOVA), and toll-like receptor 3 agonist poly(I/C) were synchronously released from the matrix in a pain-free manner. The whole microneedle patch was fabricated with two layers. The basal layer was formed using polyvinyl pyrrolidone/polyvinyl alcohol that could be rapidly dissolved upon applying the microneedle patch to the skin, whereas the microneedle layer was formed by complexes encapsulating biodegradable PEG-PSMEU, which was stuck at the injection site for sustained release of therapeutic agents. According to the results, 10 days is the time for the complexes to be completely released and express specific antigens in antigen-presenting cells in vitro and in vivo. It is noteworthy that this system could successfully elicit cancer-specific humoral immune responses and inhibit metastatic tumors in the lungs after a single shot of immunization.
Collapse
Affiliation(s)
- Huu Thuy Trang Duong
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yue Yin
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Thai Minh Duy Le
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Hoon Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Doo Sung Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
8
|
Gong X, Gao Y, Shu J, Zhang C, Zhao K. Chitosan-Based Nanomaterial as Immune Adjuvant and Delivery Carrier for Vaccines. Vaccines (Basel) 2022; 10:1906. [PMID: 36423002 PMCID: PMC9696061 DOI: 10.3390/vaccines10111906] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 08/26/2023] Open
Abstract
With the support of modern biotechnology, vaccine technology continues to iterate. The safety and efficacy of vaccines are some of the most important areas of development in the field. As a natural substance, chitosan is widely used in numerous fields-such as immune stimulation, drug delivery, wound healing, and antibacterial procedures-due to its good biocompatibility, low toxicity, biodegradability, and adhesion. Chitosan-based nanoparticles (NPs) have attracted extensive attention with respect to vaccine adjuvants and delivery systems due to their excellent properties, which can effectively enhance immune responses. Here, we list the classifications and mechanisms of action of vaccine adjuvants. At the same time, the preparation methods of chitosan, its NPs, and their mechanism of action in the delivery system are introduced. The extensive applications of chitosan and its NPs in protein vaccines and nucleic acid vaccines are also introduced. This paper reviewed the latest research progress of chitosan-based NPs in vaccine adjuvant and drug delivery systems.
Collapse
Affiliation(s)
- Xiaochen Gong
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Yuan Gao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
| | - Jianhong Shu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| | - Chunjing Zhang
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Kai Zhao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| |
Collapse
|
9
|
The influence of different bioadhesive polymers on physicochemical properties of thermoresponsive emulgels containing Amazonian andiroba oil. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
10
|
Ni Y, Zhao W, Cheng W, Deng C, Ying Z, Li L, Wang X, Sun C, Tu J, Jiang L. Lipopeptide liposomes-loaded hydrogel for multistage transdermal chemotherapy of melanoma. J Control Release 2022; 351:245-254. [PMID: 36108811 DOI: 10.1016/j.jconrel.2022.09.014] [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: 04/11/2022] [Revised: 08/04/2022] [Accepted: 09/08/2022] [Indexed: 10/31/2022]
Abstract
Transdermal administration of chemotherapeutics into tumor tissues may be an effective treatment to reduce toxic side effects and improve patient compliance for melanoma. Herein, we report a multistage transdermal drug delivery system for chemotherapy of melanoma. In this system, dendritic lipopeptide (DLP) modified multistage targeted liposomes (Mtlip) were incorporated into the hydrogel matrix to achieve localized and sustained drug release; Ultra-deformability of Mtlip can pass through dense stratum corneum to the epidermis where melanoma is located; Virus-mimicking Mtlip enhances the payload in tumor tissues by high permeability; The positive charged Mtlip can improve cell uptake efficiency and selectively accumulate into mitochondria to increases toxic. The efficacy of this type of multistage targeted liposomes loaded hydrogel in treating melanoma was systematically evaluated both in vitro and in vivo.
Collapse
Affiliation(s)
- Yifei Ni
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Wanzhu Zhao
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Wenjing Cheng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Chengyu Deng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Zhuoyang Ying
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Lei Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Xulei Wang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Chunmeng Sun
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China
| | - Jiasheng Tu
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China.
| | - Lei Jiang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China.
| |
Collapse
|
11
|
Machado AS, Lage DP, Vale DL, Freitas CS, Linhares FP, Cardoso JM, Pereira IA, Ramos FF, Tavares GS, Ludolf F, Oliveira-da-Silva JA, Bandeira RS, Simões AC, Duarte MC, Oliveira JS, Christodoulides M, Chávez-Fumagalli MA, Roatt BM, Martins VT, Coelho EA. A recombinant Leishmania amastigote-specific protein, rLiHyG, with adjuvants, protects against infection with Leishmania infantum. Acta Trop 2022; 230:106412. [PMID: 35305943 DOI: 10.1016/j.actatropica.2022.106412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
Vaccination against visceral leishmaniasis (VL) should be considered as a control measure to protect against disease, and amastigote-specific proteins could help to develop such vaccines, since this parasite form is in contact with the host immune system during the active disease. In this study, a Leishmania amastigote-specific protein, LiHyG, was evaluated as recombinant protein (rLiHyG) as vaccine candidate against Leishmania infantum infection in BALB/c mice. The protein was associated with saponin (rLiHyG/Sap) or Poloxamer 407-based polymeric micelles (rLiHyG/Mic) as adjuvants, and animals receiving saline, saponin or micelle as controls. Immunological and parasitological analyses were performed before (n = 8 per group; as primary endpoint) and after (n = 8 per group; as secondary endpoint) infection. Results showed that, in both endpoints, rLiHyG/Sap and rLiHyG/Mic induced higher levels of IFN-γ, IL-12 and GM-CSF in spleen cell cultures from vaccinated animals, besides elevated presence of IgG2a isotype antibodies. Decreased hepatotoxicity and 'positive lymphoproliferative response were also found after challenge. Such findings reflected in significantly lower levels of parasite load found in their spleens, livers, bone marrows and draining lymph nodes. In conclusion, rLiHyG associated with Th1-type adjuvant could be considered for future studies as vaccine candidate to protect against VL.
Collapse
|
12
|
White JM, Calabrese MA. Impact of small molecule and reverse poloxamer addition on the micellization and gelation mechanisms of poloxamer hydrogels. Colloids Surf A Physicochem Eng Asp 2022; 638. [PMID: 35221534 PMCID: PMC8880963 DOI: 10.1016/j.colsurfa.2021.128246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Poloxamer 407 (P407) is widely used for targeted drug-delivery because it exhibits thermoresponsive gelation behavior near body temperature, stemming from a disorder-to-order transition. Hydrophobic small molecules can be encapsulated within P407; however, these additives often negatively impact the rheological properties and lower the gelation temperatures of the hydrogels, limiting their clinical utility. Here we investigate the impact of adding two BAB reverse poloxamers (RPs), 25R4 and 31R1, on the thermal transitions, rheological properties, and assembled structures of P407 both with and without incorporated small molecules. By employing a combination of differential scanning calorimetry (DSC), rheology, and small-angle x-ray scattering (SAXS), we determine distinct mechanisms for RP incorporation. While 25R4 addition promotes inter-micelle bridge formation, the highly hydrophobic 31R1 co-micellizes with P407. Small molecule addition lowers thermal transition temperatures and increases the micelle size, while RP addition mitigates the decreases in modulus traditionally associated with small molecule incorporation. This fundamental understanding yields new strategies for tuning the mechanical and structural properties of the hydrogels, enabling design of drug-loaded formulations with ideal thermal transitions for a range of clinical applications.
Collapse
Affiliation(s)
- Joanna M White
- University of Minnesota, 421 Washington Ave SE, Minneapolis, 55455, MN, USA
| | | |
Collapse
|
13
|
Villalva DG, França CG, Loh W. Characterization of cubosomes immobilized in hydrogels of hyaluronic acid and their use for diclofenac controlled delivery. Colloids Surf B Biointerfaces 2022; 212:112352. [PMID: 35101824 DOI: 10.1016/j.colsurfb.2022.112352] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/14/2021] [Accepted: 01/18/2022] [Indexed: 11/28/2022]
Abstract
Cubosomes are dispersions of bicontinuous surfactant phases that constitute an assertive option to carry and release drugs and biomolecules, offering high efficiency of entrapment and specificity towards biological targets. This paper reports, for the first time to the best of our knowledge, the immobilization and characterization of cubosomes in chemically cross-linked oxi-hyaluronic acid and the evaluation of their use for controlled delivery of diclofenac, which is chosen as a model drug. Immobilized cubosomes prepared with phytantriol and bearing either negative or positive charges (in this case due to the addition of a cationic surfactant) were characterized by small angle X-ray scattering (SAXS) analysis and high-resolution confocal microscopy, confirming that their internal structure remains unaltered and that they appear uniformly distributed within the hydrogel matrix. Their release properties were assessed, and a limited leaching of the cubosomes from the hydrogel matrix with sustained release of the entrapped diclofenac was confirmed. These results enable the use of immobilized cubosomes as an attractive platform for biomedical applications, significantly extending the already promising features of cubosomes.
Collapse
Affiliation(s)
| | - Carla Giometti França
- Department of Engineering of Materials and Bioprocesses, School of Chemical Engineering, University of Campinas (UNICAMP), 13083-852 Campinas, SP, Brazil
| | - Watson Loh
- Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil.
| |
Collapse
|
14
|
Sha X, Chan L, Fan X, Guo P, Chen T, Liu L, Zhong J. Thermosensitive Tri-Block Polymer Nanoparticle-Hydrogel Composites as Payloads of Natamycin for Antifungal Therapy Against Fusarium Solani. Int J Nanomedicine 2022; 17:1463-1478. [PMID: 35378880 PMCID: PMC8976233 DOI: 10.2147/ijn.s332127] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Fusarium Solani is the principal pathogen associated with fungal keratitis. As a sensitive drug to F. Solani, natamycin (NAT) was limited by the poor penetration and low bioavailability in clinical application. The aim of this study was to develop a new type of tri-block polymer nanoparticle-gel complex (Gel@PLGA-PEI-PEG@NAT) for delivering NAT and evaluate its physicochemical properties, antifungal activity, safety, penetrability, adhesion, and efficacy in treating fungal keratitis. Methods PLGA-PEI-PEG@NAT was prepared and characterized with a nano-particle size analyzer, transmission electron microscopy (TEM), scanning electron microscopy (SEM), and Fourier transform infrared spectroscopy (FTIR). The minimum inhibitory concentration (MIC), cytotoxicity, penetrability of NAT (Natacyn® 5% ophthalmic suspension; Alcon) and PLGA-PEI-PEG@NAT with different concentrations were assessed. The eye surface retention time, ocular irritation, and curative effect of the NAT ophthalmic suspension and Gel@PLGA-PEI-PEG@NAT on a rabbit fungal keratitis model were evaluated. Results PLGA-PEI-PEG@NAT had a particle size of 150 nm, a positive surface charge, and a sustained-release effect. The MIC for F. Solani was 2 μg/mL. A cytotoxicity test and ocular irritation test showed that PLGA-PEI-PEG@NAT and Gel@PLGA-PEI-PEG@NAT had good biocompatibility and no obvious irritation for rabbit corneas. Penetration experiments confirmed that PLGA-PEI-PEG@NAT can successfully enter corneal epithelial cells and through the cornea to enter the anterior chamber. Compared with NAT ophthalmic suspension, Gel@PLGA-PEI-PEG@NAT had stronger cornea permeation at the same concentration. The therapeutic effect and precorneal retention ability of the NAT ophthalmic suspension and Gel@PLGA-PEI-PEG@NAT on the fungal keratitis rabbit model were compared. Gel@PLGA-PEI-PEG@NAT achieved a better therapeutic effect at a lower drug concentration, and its eye surface retention time was significantly longer than that of the NAT ophthalmic suspension. Conclusion Gel@PLGA-PEI-PEG@NAT was shown to be a safe and effective nanodrug delivery system for NAT. It has great potential to improve the cure rate of fungal keratitis, reduce the administration frequency during the treatment process, and improve patient compliance.
Collapse
Affiliation(s)
- Xiaoyuan Sha
- Department of Ophthalmology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, People’s Republic of China
| | - Leung Chan
- Department of Ophthalmology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, People’s Republic of China
| | - Xiaoyi Fan
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Penghao Guo
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Tianfeng Chen
- Department of Ophthalmology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, People’s Republic of China
| | - Lian Liu
- Department of Ophthalmology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, People’s Republic of China
- Correspondence: Lian Liu; Tianfeng Chen, Department of Ophthalmology, The First Affiliated Hospital of Jinan University; Department of Chemistry, Jinan University, 601 Huangpu Road, Guangzhou, 510632, People’s Republic of China, Email ;
| | - Jingxiang Zhong
- Department of Ophthalmology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, People’s Republic of China
- Department of Ophthalmology, The Sixth Affiliated Hospital, Jinan University, Dongguan, People’s Republic of China
| |
Collapse
|
15
|
Tan C, Hosseini SF, Jafari SM. Cubosomes and Hexosomes as Novel Nanocarriers for Bioactive Compounds. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1423-1437. [PMID: 35089018 DOI: 10.1021/acs.jafc.1c06747] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cubosomes and hexosomes are nanostructured liquid crystalline particles, known as biocompatible nanocarriers for drug delivery. In recent years, there has been good interest in using cubosomes and hexosomes for the delivery of bioactive compounds in functional foods. These systems feature thermodynamic stability, encapsulate both hydrophobic and hydrophilic substances, and have a high tolerance to environmental stresses and potential for controlled release. This review outlines the recent advances in cubosomes and hexosomes in the food industry, focusing on their structure, composition, formation mechanisms, and factors influencing phase transformation between cubosomes and hexosomes. The potential applications especially for the bioactive delivery are presented. The integration of cubosomes and hexosomes with other emerging encapsulation technologies such as surface coating, gelation, and incorporation of polymers are also discussed.
Collapse
Affiliation(s)
- Chen Tan
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Seyed Fakhreddin Hosseini
- Department of Seafood Processing, Faculty of Marine Sciences, Tarbiat Modares University, P.O. Box 46414-356, Noor 193954697, Iran
| | - Seid Mahdi Jafari
- Faculty of Food Science and Technology, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan 4913815739, Iran
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, Universidade de Vigo, E-32004 Ourense, Spain
| |
Collapse
|
16
|
Rosseto HC, de Toledo LDAS, Said dos Santos R, de Francisco LMB, Vecchi CF, Esposito E, Cortesi R, Bruschi ML. Design of propolis-loaded film forming systems for topical administration: The effect of acrylic acid derivative polymers. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.114514] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
17
|
Vaccine implants: current status and recent advancements. Emerg Top Life Sci 2020; 4:319-330. [DOI: 10.1042/etls20200164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 01/29/2023]
Abstract
Implants have long been used in the field of drug delivery as controlled release vehicles and are now being investigated as single-shot vaccine technologies. Implants have shown great promise, minimizing the need for multiple immunizations while stimulating potent immune responses with reduced doses of vaccine. Synchronous release of vaccine components from implants over an appropriate period of time is important in order to avoid issues including immune tolerance, sequestration or deletion. Traditionally, implants require surgical implantation and removal, which can be a barrier to their widespread use. Degradable and in situ implants are now being developed that can be administered using minimally invasive subcutaneous or intramuscular injection techniques. Injectable hydrogels remain the most commonly studied approach for sustained vaccine delivery due to their ease of administration and tunable degradation properties. Despite exciting advancements in the field of vaccine implants, few technologies have progressed to clinical trials. To increase the likelihood of clinical translation of vaccine implants, strategic testing of disease-relevant antigens in appropriate species is essential. In this review, the significance of vaccine implants and the different types of implants being developed to deliver vaccines are discussed.
Collapse
|
18
|
Liu Z, Yu L, Gu P, Bo R, Xu S, Wusiman A, Liu J, Hu Y, Wang D. Surface-Engineered Cubosomes Serve as a Novel Vaccine Adjuvant to Modulate Innate Immunity and Improve Adaptive Immunity in vivo. Int J Nanomedicine 2020; 15:8595-8608. [PMID: 33177820 PMCID: PMC7650836 DOI: 10.2147/ijn.s266165] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/29/2020] [Indexed: 12/01/2022] Open
Abstract
Objective Recent studies have revealed the adjuvant activity of cubosomes and their potential utility as an antigen delivery system. In this study, to further enhance the adjuvant activity of cubosomes, two cationic polymers are modified on the surface of cubosomes. Methods Here, we exploit the effects of surface chemistry on the adjuvant activity of Ganoderma lucidum polysaccharide cubosomes (GLPC) by placing two kinds of molecules, that is, cetyltrimethylammonium bromide (CTAB) and poly(diallydimethyl ammonium chloride) (PDDAC), on their surface. Results CTAB- or PDDAC-modified GLPC were found to significantly promote humoral and cellular immune responses, as well as the proliferation of CD3+ CD4+ or CD3+ CD8+T cells through the powerful activation of dendritic cells (DCs). The enhanced immune responses of PDDAC-modified GLPC might be attributed to the maturation of DCs into draining lymph nodes and the activation of spleen and cytokines in serum. Conclusion PDDAC modification is beneficial for enhancing humoral and cellular immune response, suggesting that PDDAC-GLPC-OVA has the ability to be a potential adjuvant for vaccine.
Collapse
Affiliation(s)
- Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Ruonan Bo
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Shuwen Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Adelijiang Wusiman
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yuanliang Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
19
|
Pacifici N, Bolandparvaz A, Lewis JS. Stimuli-Responsive Biomaterials for Vaccines and Immunotherapeutic Applications. ADVANCED THERAPEUTICS 2020; 3:2000129. [PMID: 32838028 PMCID: PMC7435355 DOI: 10.1002/adtp.202000129] [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: 06/04/2020] [Revised: 07/16/2020] [Indexed: 12/26/2022]
Abstract
The immune system is the key target for vaccines and immunotherapeutic approaches aimed at blunting infectious diseases, cancer, autoimmunity, and implant rejection. However, systemwide immunomodulation is undesirable due to the severe side effects that typically accompany such strategies. In order to circumvent these undesired, harmful effects, scientists have turned to tailorable biomaterials that can achieve localized, potent release of immune-modulating agents. Specifically, "stimuli-responsive" biomaterials hold a strong promise for delivery of immunotherapeutic agents to the disease site or disease-relevant tissues with high spatial and temporal accuracy. This review provides an overview of stimuli-responsive biomaterials used for targeted immunomodulation. Stimuli-responsive or "environmentally responsive" materials are customized to specifically react to changes in pH, temperature, enzymes, redox environment, photo-stimulation, molecule-binding, magnetic fields, ultrasound-stimulation, and electric fields. Moreover, the latest generation of this class of materials incorporates elements that allow for response to multiple stimuli. These developments, and other stimuli-responsive materials that are on the horizon, are discussed in the context of controlling immune responses.
Collapse
Affiliation(s)
- Noah Pacifici
- Department of Biomedical Engineering University of California Davis Davis CA 95616 USA
| | - Amir Bolandparvaz
- Department of Biomedical Engineering University of California Davis Davis CA 95616 USA
| | - Jamal S Lewis
- Department of Biomedical Engineering University of California Davis Davis CA 95616 USA
| |
Collapse
|
20
|
Abdeltawab H, Svirskis D, Sharma M. Formulation strategies to modulate drug release from poloxamer based in situ gelling systems. Expert Opin Drug Deliv 2020; 17:495-509. [PMID: 32067500 DOI: 10.1080/17425247.2020.1731469] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Poloxamer based in situ gelling systems offer numerous advantages in drug delivery; however, their application as prolonged-release delivery platforms is limited mainly due to their weak mechanical properties and the interconnected aqueous network causing fast gel erosion and drug diffusion.Area covered: The focus of this review is to provide an insightful discussion on the formulation strategies that can be employed to sustain/prolong the drug release from poloxamer based in situ gelling systems. The review also outlines the formulation factors, influencing drug release from these systems.Expert opinion: The nature, composition, and concentration of poloxamers are the most critical factors in defining the rate of drug release from an in situ gelling matrix. Hydrophobic gel matrices have compact micellar arrangements resulting in slow diffusion and erosion. Depending on the intended clinical application, gel characteristics can be modulated, either by physical blending or by chemical crosslinking with additive materials, to slow release and improve residence time at the administration site. Incorporating drug-loaded particles into poloxamer gels sustains drug release by creating multiple rate-limiting release barriers. Chemical modification of poloxamers appears to be a promising strategy to obtain prolonged sustained release for parenteral application without compromising the rheological properties of the formulation.
Collapse
Affiliation(s)
- Hani Abdeltawab
- School of Pharmacy, Faculty of Medical & Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical & Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Manisha Sharma
- School of Pharmacy, Faculty of Medical & Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
21
|
Bansal A, Gamal W, Wu X, Yang Y, Olson V, D'Souza MJ. Evaluation of an adjuvanted hydrogel-based pDNA nanoparticulate vaccine for rabies prevention and immunocontraception. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2019; 21:102049. [PMID: 31279062 PMCID: PMC11287484 DOI: 10.1016/j.nano.2019.102049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/23/2019] [Accepted: 06/14/2019] [Indexed: 02/05/2023]
Abstract
Immunocontraceptive vaccination is becoming an acceptable strategy in managing animal populations. Mass vaccination of dogs is the most cost-effective and efficient method to control rabies, and combination of rabies vaccination and animal population control will be an added advantage. In this study, we developed an adjuvanted hydrogel-based pDNA nanoparticulate vaccine for rabies protection and immunocontraception. In vivo, we observed an immune response skewed toward a Th2 type, in contrast to the Th1 type in our previous pDNA study. The observation was verified by the IgG2a/IgG1 ratio (<1), and cytokine expression profile of IL-4 and IFN-γ. The humoral immune response is key for rabies protection and a GnRH antibody-based immunocontraception. In mice, anti-GnRH antibody titers were detected 4 weeks after immunization and lasted for 12 weeks, post animal experiment was terminated. The adjuvanted pDNA nanoparticulate vaccine shows promise for future studies evaluating protection from rabies challenge and prevention of animal breeding.
Collapse
Affiliation(s)
- Amit Bansal
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA, USA.
| | - Wael Gamal
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA, USA
| | - Xianfu Wu
- Poxvirus and Rabies Branch, DHCPP, NCEZID, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Yong Yang
- Poxvirus and Rabies Branch, DHCPP, NCEZID, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Victoria Olson
- Poxvirus and Rabies Branch, DHCPP, NCEZID, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Martin J D'Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA, USA
| |
Collapse
|
22
|
von Halling Laier C, Gibson B, Moreno JAS, Rades T, Hook S, Nielsen LH, Boisen A. Microcontainers for protection of oral vaccines, in vitro and in vivo evaluation. J Control Release 2019; 294:91-101. [DOI: 10.1016/j.jconrel.2018.11.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 11/30/2018] [Indexed: 12/21/2022]
|
23
|
Spray dried cubosomes with ovalbumin and Quil-A as a nanoparticulate dry powder vaccine formulation. Int J Pharm 2018; 550:35-44. [DOI: 10.1016/j.ijpharm.2018.08.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 01/30/2023]
|
24
|
Younus M, Hawley A, Boyd BJ, Rizwan SB. Bulk and dispersed aqueous behaviour of an endogenous lipid, selachyl alcohol: Effect of Tween 80 and Pluronic F127 on nanostructure. Colloids Surf B Biointerfaces 2018; 169:135-142. [DOI: 10.1016/j.colsurfb.2018.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/03/2018] [Accepted: 05/06/2018] [Indexed: 11/24/2022]
|
25
|
Bansal A, Wu X, Olson V, D'Souza MJ. Characterization of rabies pDNA nanoparticulate vaccine in poloxamer 407 gel. Int J Pharm 2018; 545:318-328. [PMID: 29746999 DOI: 10.1016/j.ijpharm.2018.05.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 12/18/2022]
Abstract
Plasmid DNA (pDNA) vaccines have the potential for protection against a wide range of diseases including rabies but are rapid in degradation and poor in uptake by antigen-presenting cells. To overcome the limitations, we fabricated a pDNA nanoparticulate vaccine. The negatively charged pDNA was adsorbed onto the surface of cationic PLGA (poly (d, l-lactide-co-glycolide))-chitosan nanoparticles and were used as a delivery vehicle. To create a hydrogel for sustainable vaccine release, we dispersed the pDNA nanoparticles in poloxamer 407 gel which is liquid at 4 °C and turns into soft gels at 37 °C, providing ease of administration and preventing burst release of pDNA. Complete immobilization of pDNA to cationic nanoparticles was achieved at a pDNA to nanoparticles ratio (P/N) of 1/50. Cellular uptake of nanoparticles was both time and concentration dependent and followed a saturation kinetics with Vmax of 11.389 µg/mL h and Km of 139.48 µg/mL. The in vitro release studies showed the nanoparticulate vaccine has a sustained release for up to 24 days. In summary, pDNA PLGA-chitosan nanoparticles were non-cytotoxic, their buffering capacity and cell uptake were enhanced, and sustained the release of pDNA. We expect our pDNA vaccine's potency will be greatly improved in the animal studies.
Collapse
Affiliation(s)
- Amit Bansal
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA.
| | - Xianfu Wu
- Poxvirus and Rabies Branch, DHCPP, NCEZID, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Victoria Olson
- Poxvirus and Rabies Branch, DHCPP, NCEZID, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Martin J D'Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA
| |
Collapse
|
26
|
Bobbala S, Gibson B, Gamble AB, McDowell A, Hook S. Poloxamer 407-chitosan grafted thermoresponsive hydrogels achieve synchronous and sustained release of antigen and adjuvant from single-shot vaccines. Immunol Cell Biol 2018; 96:656-665. [PMID: 29499080 DOI: 10.1111/imcb.12031] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/24/2018] [Accepted: 02/26/2018] [Indexed: 01/22/2023]
Abstract
Sustained-release vaccine delivery systems may enhance the immunogenicity of subunit vaccines and reduce the need for multiple vaccinations. The aim of this study was to develop a thermoresponsive hydrogel using poloxamer 407-chitosan (CP) grafted copolymer as a delivery system for single-shot sustained-release vaccines. The CP copolymer was synthesized using 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide and N-hydroxysuccinimide chemistry. The CP copolymer was a free flowing solution at ambient temperature and transformed rapidly into a gel at body temperature. The hydrogels were loaded with vaccine antigen and adjuvants or the vaccine components were encapsulated in poly (lactic-co-glycolic acid) nanoparticles in order to ensure synchronous release. The CP hydrogels were stable for up to 18 days in vitro. Release of both nanoparticles and the individual components was complete, with release of the individual components being modulated by incorporation into nanoparticles. In vivo, a single dose of CP hydrogel vaccine induced strong, long lasting, cellular and humoral responses that could protect against the development of tumors in a murine melanoma model.
Collapse
Affiliation(s)
- Sharan Bobbala
- School of Pharmacy, University of Otago, Dunedin, New Zealand.,Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Blake Gibson
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Arlene McDowell
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
27
|
Umair S, Pernthaner A, Deng Q, Gibson B, Hook S, Heath D. Preliminary evaluation of a thermosensitive chitosan hydrogel for Echinococcus granulosus vaccine delivery. Vet Parasitol 2017; 236:117-120. [PMID: 28288754 DOI: 10.1016/j.vetpar.2017.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/01/2017] [Accepted: 02/10/2017] [Indexed: 10/20/2022]
Abstract
The EG95 vaccine is effective in protecting grazing animals from infection with Echinococcus granulosus. Six male lambs were used in the study, two were each vaccinated subcutaneously with 50μg EG95/1mg Quil-A, two animals were each vaccinated with 50μg EG95/1mg Quil-A in 1% chitosan thermolabile gel subcutaneously, and two animals served as non-vaccinated controls. Two vaccinations were given at a 7 week interval. Two vaccinations induced a significantly higher antibody titre in the chitosan group compared with the Quil-A only group. The chitosan vaccine group also had a significantly higher antibody titre compared with a positive control sera from vaccinated and challenged sheep. Incorporating the EG95/Quil-A vaccine in a thermo-responsive chitosan sol-gel stimulated, after the second injection, a high level of antibody absorbance which remained high for at least one year. This response was significantly greater than the response to vaccine without the gel.
Collapse
Affiliation(s)
- Saleh Umair
- The Hopkirk Research Institute, AgResearch Ltd, Private Bag 11-008, Palmerston North, New Zealand.
| | - Anton Pernthaner
- The Hopkirk Research Institute, AgResearch Ltd, Private Bag 11-008, Palmerston North, New Zealand
| | - Qing Deng
- The Hopkirk Research Institute, AgResearch Ltd, Private Bag 11-008, Palmerston North, New Zealand
| | - Blake Gibson
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, New Zealand
| | - David Heath
- The Hopkirk Research Institute, AgResearch Ltd, Private Bag 11-008, Palmerston North, New Zealand
| |
Collapse
|
28
|
Yu S, Zhang X, Tan G, Tian L, Liu D, Liu Y, Yang X, Pan W. A novel pH-induced thermosensitive hydrogel composed of carboxymethyl chitosan and poloxamer cross-linked by glutaraldehyde for ophthalmic drug delivery. Carbohydr Polym 2017; 155:208-217. [DOI: 10.1016/j.carbpol.2016.08.073] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/02/2016] [Accepted: 08/24/2016] [Indexed: 11/28/2022]
|
29
|
Vedakumari WS, Ayaz N, Karthick AS, Senthil R, Sastry TP. Quercetin impregnated chitosan–fibrin composite scaffolds as potential wound dressing materials — Fabrication, characterization and in vivo analysis. Eur J Pharm Sci 2017; 97:106-112. [DOI: 10.1016/j.ejps.2016.11.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 12/14/2022]
|
30
|
Díaz AG, Quinteros DA, Gutiérrez SE, Rivero MA, Palma SD, Allemandi DA, Pardo RP, Zylberman V, Goldbaum FA, Estein SM. Immune response induced by conjunctival immunization with polymeric antigen BLSOmp31 using a thermoresponsive and mucoadhesive in situ gel as vaccine delivery system for prevention of ovine brucellosis. Vet Immunol Immunopathol 2016; 178:50-6. [DOI: 10.1016/j.vetimm.2016.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/12/2016] [Accepted: 07/04/2016] [Indexed: 11/15/2022]
|
31
|
Liu Z, Luo L, Zheng S, Niu Y, Bo R, Huang Y, Xing J, Li Z, Wang D. Cubosome nanoparticles potentiate immune properties of immunostimulants. Int J Nanomedicine 2016; 11:3571-83. [PMID: 27536099 PMCID: PMC4973726 DOI: 10.2147/ijn.s110406] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cubosomes have been explored as drug and antigen carriers in the past few years. A few reports have described that cubosomes can enhance the ability of immunostimulants to generate strong immune responses. Polysaccharide (PS), an immunostimulant, has been reported to be a promising adjuvant for vaccines. Herein, we incorporated PS into cubosomes to generate PS-cubosome (Cub-PS) nanoparticles, and Cub-PS was characterized by small-angle X-ray scattering scattering and cryo-field emission scanning electron microscopy. The immunological activity of Cub-PS was compared with that of Cub and PS. The results demonstrated that Cub-PS elicited more potent immune responses than Cub or PS alone. The enhanced immune responses might be attributed to the promotion of antigen transport into draining lymph nodes and efficient dendritic cell activation and memory T-helper cell differentiation in draining lymph nodes. Overall, these findings indicate that cubosomes have the potential to enhance the ability of immunostimulants to generate an immune response.
Collapse
Affiliation(s)
- Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Li Luo
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Sisi Zheng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Yale Niu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Ruonan Bo
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Yee Huang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Jie Xing
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Zhihua Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People's Republic of China
| |
Collapse
|
32
|
Bobbala S, Hook S. Is There an Optimal Formulation and Delivery Strategy for Subunit Vaccines? Pharm Res 2016; 33:2078-97. [DOI: 10.1007/s11095-016-1979-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/21/2016] [Indexed: 12/16/2022]
|
33
|
Bobbala S, Tamboli V, McDowell A, Mitra AK, Hook S. Novel Injectable Pentablock Copolymer Based Thermoresponsive Hydrogels for Sustained Release Vaccines. AAPS JOURNAL 2015; 18:261-9. [PMID: 26589309 DOI: 10.1208/s12248-015-9843-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023]
Abstract
The need for multiple vaccinations to enhance the immunogenicity of subunit vaccines may be reduced by delivering the vaccine over an extended period of time. Here, we report two novel injectable pentablock copolymer based thermoresponsive hydrogels made of polyethyleneglycol-polycaprolactone-polylactide-polycaprolactone-polyethyleneglycol (PEG-PCL-PLA-PCL-PEG) with varying ratios of polycaprolactone (PCL) and polylactide (PLA), as single shot sustained release vaccines. Pentablock copolymer hydrogels were loaded with vaccine-encapsulated poly lactic-co-glycolic acid nanoparticles (PLGA-NP) or with the soluble vaccine components. Incorporation of PLGA-NP into the thermoresponsive hydrogels increased the complex viscosity of the gels, lowered the gelation temperature, and minimized the burst release of antigen and adjuvants. The two pentablock hydrogels stimulated both cellular and humoral responses. The addition of PLGA-NP to the hydrogels sustained immune responses for up to 49 days. The polymer with a higher ratio of PCL to PLA formed a more rigid gel, induced stronger immune responses, and stimulated effective anti-tumor responses in a prophylactic melanoma tumor model.
Collapse
Affiliation(s)
- Sharan Bobbala
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand
| | - Viral Tamboli
- School of Pharmacy, UMKC, Kansas City, Missouri, USA
| | - Arlene McDowell
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand
| | - Ashim K Mitra
- School of Pharmacy, UMKC, Kansas City, Missouri, USA
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
34
|
Neumann S, Young K, Compton B, Anderson R, Painter G, Hook S. Synthetic TRP2 long-peptide and α-galactosylceramide formulated into cationic liposomes elicit CD8+ T-cell responses and prevent tumour progression. Vaccine 2015; 33:5838-5844. [PMID: 26363382 DOI: 10.1016/j.vaccine.2015.08.083] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/28/2015] [Accepted: 08/29/2015] [Indexed: 12/22/2022]
Abstract
The lipid antigen α-galactosylceramide (α-GalCer) is a potent activator of invariant natural killer T-cells (iNKT cells) and can stimulate cytotoxic and anti-tumour immune responses. However optimal responses appear to be induced by α-GalCer when cell-based vaccines are delivered intravenously. Here we investigated if co-delivery of protein and peptide antigens along with α-GalCer in a liposomal formulation could stimulate therapeutic anti-tumour immune responses. Cationic liposomes were inherently immune-stimulatory and induced cytotoxic immune responses when delivered both by intravenous and subcutaneous injection. However, only vaccine delivered intravenously stimulated therapeutic anti-tumour immune responses to a peptide antigen. Surface modification with polyethylene glycol (PEG) did not improve immune responses to either intravenously or subcutaneously delivered vaccines. Immune responses to short and long peptide sequences (CD8 and CD4 epitopes) of the self-antigen tyrosinase-related protein 2 (TRP2) as a vaccine antigen, co-delivered with α-GalCer in either cationic liposomes or PBS were further examined. Enhanced production of IFN-γ, increased cytotoxic T-cell responses and tumour survival were observed when a long TRP2-peptide was delivered with α-GalCer in cationic liposomes.
Collapse
Affiliation(s)
- Silke Neumann
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Katie Young
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Benji Compton
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Regan Anderson
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Gavin Painter
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
35
|
Chen Y, Angelova A, Angelov B, Drechsler M, Garamus VM, Willumeit-Römer R, Zou A. Sterically stabilized spongosomes for multidrug delivery of anticancer nanomedicines. J Mater Chem B 2015; 3:7734-7744. [DOI: 10.1039/c5tb01193k] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
SAXS patterns of drug-loaded lipid nanocarriers stabilized by polysorbate P80 (left); cryo-TEM image of BAI-BJO-spongosomes-2 (right).
Collapse
Affiliation(s)
- Yiyin Chen
- East China University of Science and Technology
- Shanghai
- China
| | - Angelina Angelova
- CNRS UMR8612 Institut Galien Paris-Sud
- Univ Paris Sud
- LabEx LERMIT
- Châtenay-Malabry
- F-92296 France
| | - Borislav Angelov
- Institute of Macromolecular Chemistry
- Academy of Sciences of the Czech Republic
- 16206 Prague
- Czech Republic
| | - Markus Drechsler
- Laboratory for Soft Matter Electron Microscopy
- Bayreuth Institute of Macromolecular Research (BIMF)
- University of Bayreuth
- D-95440 Bayreuth
- Germany
| | - Vasil M. Garamus
- Helmholtz-Zentrum Geesthacht
- Centre for Materials and Coastal Research
- D-21502 Geesthacht
- Germany
| | - Regine Willumeit-Römer
- Helmholtz-Zentrum Geesthacht
- Centre for Materials and Coastal Research
- D-21502 Geesthacht
- Germany
| | - Aihua Zou
- East China University of Science and Technology
- Shanghai
- China
| |
Collapse
|