Generic placeholder image

Current Biotechnology

Editor-in-Chief

ISSN (Print): 2211-5501
ISSN (Online): 2211-551X

Mini-Review Article

Insights on Bioactive Peptide-loaded Nanoformulations

Author(s): Venkateshwaran Krishnaswami*, Balakrishnan Natarajan, Saravanakumar Arthanari, Jeseeta Suresh and Nirmal Aranvindaraj

Volume 12, Issue 1, 2023

Published on: 19 January, 2023

Page: [3 - 13] Pages: 11

DOI: 10.2174/2211550112666230104123153

Price: $65

conference banner
Abstract

In order to tackle multifactorial illnesses, the importance of bioactive peptides in nano drug delivery systems is emphasised. Multifactorial diseases are primarily brought on by protein misfolding. Therefore, pharmaceutical formulations are recommended to deliver a successful treatment. Difficulties hinder its application in delivering raw peptides with poor bioavailability, absorption, and circulation time, making it a difficult assignment for researchers. Recently, bioactive peptides have become increasingly important in therapy. In addition, several bioactive proteins have poor absorption characteristics in the GIT. These issues can be resolved by creating nano-based peptide-based delivery systems that encapsulate, retain, protect, and transport bioactive peptides. The focus of the proposed review paper is to provide an overview of peptides, commercialization concerns, and their related attempts to develop into novel peptide-based nanoformulations.

Keywords: Peptides, bioactive, drug delivery, nanoformulations, multifactorial illnesses, protein misfolding, bioavailability.

Graphical Abstract
[1]
Lee ACL, Harris JL, Khanna KK, Hong JH. A comprehensive review on current advances in peptide drug development and design. Int J Mol Sci 2019; 20(10): 2383.
[http://dx.doi.org/10.3390/ijms20102383] [PMID: 31091705]
[2]
Otvos Jr L., Wade JD. Current challenges in peptide-baseddrug discovery. Front Chem 2014; 2: 1-4.
[3]
Isidro-Llobet A, Kenworthy MN, Mukherjee S, et al. Sustainability challenges in peptide synthesis and purification: From R&D to production. J Org Chem 2019; 84(8): 4615-28.
[http://dx.doi.org/10.1021/acs.joc.8b03001] [PMID: 30900880]
[4]
Fosgerau K, Hoffmann T. Peptide therapeutics: current status and future directions. Drug Discov Today 2015; 20(1): 122-8.
[http://dx.doi.org/10.1016/j.drudis.2014.10.003] [PMID: 25450771]
[5]
Jeong W, Bu J, Kubiatowicz LJ, Chen SS, Kim Y, Hong S. Peptide-nanoparticle conjugates: a next generation of diagnostic and therapeutic platforms? Nano Converg 2018; 5(1): 38.
[http://dx.doi.org/10.1186/s40580-018-0170-1] [PMID: 30539365]
[6]
Salvalaio M, Rigon L, Belletti D, et al. Targeted polymeric nanoparticles for brain delivery of high molecular weight molecules in lysosomal storage disorders. PLoS One 2016; 11(5): e0156452.
[http://dx.doi.org/10.1371/journal.pone.0156452] [PMID: 27228099]
[7]
Antonow MB, Asbahr ACC, Raddatz P, et al. Liquid formulation containing doxorubicin-loaded lipid-core nanocapsules: Cytotoxicity in human breast cancer cell line and in vitro uptake mechanism. Mater Sci Eng C 2017; 76: 374-82.
[http://dx.doi.org/10.1016/j.msec.2017.03.099] [PMID: 28482541]
[8]
Oliveira CP, Prado WA, Lavayen V, et al. Bromelain-functionalized multiple-wall lipid-core nanocapsules: formulation, chemical structure and antiproliferative effect against human breast cancer cells (MCF-7). Pharm Res 2017; 34(2): 438-52.
[http://dx.doi.org/10.1007/s11095-016-2074-2] [PMID: 27981451]
[9]
Ghezzi M, Pescina S, Padula C, et al. Polymeric micelles in drug delivery: An insight of the techniques for their characterization and assessment in biorelevant conditions. J Control Release 2021; 332: 312-36.
[http://dx.doi.org/10.1016/j.jconrel.2021.02.031] [PMID: 33652113]
[10]
Zhang X, Zhu T, Miao Y, Zhou L, Zhang W. Dual-responsive doxorubicin-loaded nanomicelles for enhanced cancer therapy. J Nanobiotechnology 2020; 18(1): 136.
[http://dx.doi.org/10.1186/s12951-020-00691-6] [PMID: 32972412]
[11]
Ponnusamy C, Sugumaran A, Krishnaswami V, Kandasamy R, Natesan S. Design and development of artemisinin and dexamethasone loaded topical nanodispersion for the effective treatment of age-related macular degeneration. IET Nanobiotechnol 2019; 13(8): 868-74.
[http://dx.doi.org/10.1049/iet-nbt.2019.0130] [PMID: 31625529]
[12]
Kitaoka M, Wakabayashi R, Kamiya N, Goto M. Solid‐in‐oil nanodispersions for transdermal drug delivery systems. Biotechnol J 2016; 11(11): 1375-85.
[http://dx.doi.org/10.1002/biot.201600081] [PMID: 27529824]
[13]
Peng X, Zhou Y, Han K, et al. Characterization of cubosomes as a targeted and sustained transdermal delivery system for capsaicin. Drug Des Devel Ther 2015; 9(9): 4209-18.
[http://dx.doi.org/10.2147/DDDT.S86370] [PMID: 26345516]
[14]
Varghese R, Salvi S, Sood P, Kulkarni B, Kumar D. Cubosomes in cancer drug delivery: A review. Colloid Interface Sci Commun 2022; 46100561.
[http://dx.doi.org/10.1016/j.colcom.2021.100561]
[15]
Almoshari Y. Development, therapeutic evaluation and theranostic applications of cubosomes on cancers: an updated review. Pharmaceutics 2022; 14(3): 600.
[http://dx.doi.org/10.3390/pharmaceutics14030600] [PMID: 35335975]
[16]
Barriga HMG, Holme MN, Stevens MM. Cubosomes: The next generation of smart lipid nanoparticles. Angew Chem Int Ed Engl 2019; 58(10): 2958-78.
[17]
Mason TG, Wilking JN, Meleson K, Chang CB, Graves SM. Nanoemulsions: formation, structure, and physical properties. J Phys Condens Matter 2006; 18(41): R635-66.
[http://dx.doi.org/10.1088/0953-8984/18/41/R01]
[18]
Ramesh Shah R, Shripal Magdum C, Shivagonda Patil S, Shanawaj Niakwade N. Preparation and evaluation of aceclofenac topical microemulsion. Iran J Pharm Res 2010; 9(1): 5-11.
[PMID: 24363700]
[19]
Kumar R, Sinha VR. Preparation and optimization of voriconazole microemulsion for ocular delivery. Colloids Surf B Biointerfaces 2014; 117: 82-8.
[http://dx.doi.org/10.1016/j.colsurfb.2014.02.007] [PMID: 24632034]
[20]
Ghosh P, Murthy R. Microemulsions: a potential drug delivery system. Curr Drug Deliv 2006; 3(2): 167-80.
[http://dx.doi.org/10.2174/156720106776359168] [PMID: 16611003]
[21]
Kale AA, Patravale VB. Development and evaluation of lorazepam microemulsions for parenteral delivery. AAPS PharmSciTech 2008; 9(3): 966-71.
[http://dx.doi.org/10.1208/s12249-008-9131-z] [PMID: 18720016]
[22]
Chaudharishilpa P. Nanosuspension-a novel approaches in drug delivery system Int J Pharm Res Rev 2(12): 30-9.
[23]
Patel M, Shah A, Patel NM, Patel MR, Patel KR. Nanosuspension:a novel approach for drug delivery system. Sci Biosci Res 2011; 1(1): 1-10.
[24]
Kipp JE, Wong JCT, Doty MJ, Werling J, Rebbeck CL, Brynjelsen SUS. U.S. Patent No. 6,884,436, 2005.
[25]
Suchik S, Geetha A. A Novel Technologies for oral delivery of poorly soluable drug. RJPBCS 2010; 1(4): 292-305.
[26]
Kobayashi N, Izumi H, Morimoto Y. Review of toxicity studies of carbon nanotubes. J Occup Health 2017; 59(5): 394-407.
[http://dx.doi.org/10.1539/joh.17-0089-RA] [PMID: 28794394]
[27]
Fujita K, Fukuda M, Endoh S, et al. Pulmonary and pleural inflammation after intratracheal instillation of short single-walled and multi-walled carbon nanotubes. Toxicol Lett 2016; 257: 23-37.
[http://dx.doi.org/10.1016/j.toxlet.2016.05.025] [PMID: 27259835]
[28]
Arnoldussen YJ, Skaug V, Aleksandersen M, et al. Inflammation in the pleural cavity following injection of multi-walled carbon nanotubes is dependent on their characteristics and the presence of IL-1 genes. Nanotoxicology 2018; 12(6): 522-38.
[http://dx.doi.org/10.1080/17435390.2018.1465139] [PMID: 29742950]
[29]
Sierra MI, Rubio L, Bayón GF, et al. DNA methylation changes in human lung epithelia cells exposed to multi-walled carbon nanotubes. Nanotoxicology 2017; 11(7): 857-70.
[http://dx.doi.org/10.1080/17435390.2017.1371350] [PMID: 28901819]
[30]
Ghosh M, Öner D, Poels K, et al. Changes in DNA methylation induced by multi-walled carbon nanotube exposure in the workplace Nanotoxicology 2017; 11(9-10): 1195-1210.
[http://dx.doi.org/10.1080/17435390.2017.1406169] [PMID: 29191063]
[31]
Valdiglesias V, Kiliç G, Costa C, et al. Effects of iron oxide nanoparticles: Cytotoxicity, genotoxicity, developmental toxicity, and neurotoxicity. Environ Mol Mutagen 2015; 56(2): 125-48.
[http://dx.doi.org/10.1002/em.21909] [PMID: 25209650]
[32]
Alarifi S, Ali D, Alkahtani S, Alhader MS. Iron oxide nanoparticles induce oxidative stress, DNA damage, and caspase activation in the human breast cancer cell line. Biol Trace Elem Res 2014; 159(1-3): 416-24.
[http://dx.doi.org/10.1007/s12011-014-9972-0] [PMID: 24748114]
[33]
Yaaghoobi M, Emtiazi G, Roghanian R. A novel approach for aerobic construction of iron oxide nanoparticles by Acinetobactor radio resistens and their effect on red blood cells. Curr Nanosci 2012; 8(2): 286-91.
[http://dx.doi.org/10.2174/157341312800167687]
[34]
Gaharwar US. R P. Iron oxide nanoparticles induced oxidative damage in peripheral blood cells of rat. J Biomed Sci Eng 2015; 8(4): 274-86.
[http://dx.doi.org/10.4236/jbise.2015.84026]
[35]
Zhang L, Xie H, Liu Y, Wang J. Research on nanotoxicity of an iron oxide nanoparticles and potential application Toxicol Open Access 2017; 3(3): 310.
[http://dx.doi.org/10.4172/2476-2067.1000130]
[36]
Hsin YH, Chen CF, Huang S, Shih TS, Lai PS, Chueh PJ. The apoptotic effect of nanosilver is mediated by a ROS- and JNK-dependent mechanism involving the mitochondrial pathway in NIH3T3 cells. Toxicol Lett 2008; 179(3): 130-9.
[http://dx.doi.org/10.1016/j.toxlet.2008.04.015] [PMID: 18547751]
[37]
Mei N, Zhang Y, Chen Y, et al. Silver nanoparticle-induced mutations and oxidative stress in mouse lymphoma cells. Environ Mol Mutagen 2012; 53(6): 409-19.
[http://dx.doi.org/10.1002/em.21698] [PMID: 22576574]
[38]
Kim S, Ryu DY. Silver nanoparticle-induced oxidative stress, genotoxicity and apoptosis in cultured cells and animal tissues. J Appl Toxicol 2013; 33(2): 78-89.
[http://dx.doi.org/10.1002/jat.2792] [PMID: 22936301]
[39]
Vila L, Marcos R, Hernández A. Long-term effects of silver nanoparticles in caco-2 cells. Nanotoxicology 2017; 11(6): 1-10.
[http://dx.doi.org/10.1080/17435390.2017.1355997] [PMID: 28707555]
[40]
Campagnolo L, Massimiani M, Vecchione L, et al. Silver nanoparticles inhaled during pregnancy reach and affect the placenta and the foetus. Nanotoxicology 2017; 11(5): 687-98.
[http://dx.doi.org/10.1080/17435390.2017.1343875] [PMID: 28618895]
[41]
Yamada S, Yamazaki D, Kanda Y. Silver nanoparticles inhibit neural induction in human induced pluripotent stem cells. Nanotoxicology 2018; 12(8): 836-46.
[http://dx.doi.org/10.1080/17435390.2018.1481238] [PMID: 29902946]
[42]
Repar N, Li H, Aguilar JS, Li QQ, Drobne D, Hong Y. Silver nanoparticles induce neurotoxicity in a human embryonic stem cell-derived neuron and astrocyte network. Nanotoxicology 2018; 12(2): 104-16.
[http://dx.doi.org/10.1080/17435390.2018.1425497] [PMID: 29334833]
[43]
Wang F, Chen Y, Wang Y, et al. Ultra-long silver nanowires induced mitotic abnormalities and cytokinetic failure in A549 cells. Nanotoxicology 2019; 13(4): 543-57.
[http://dx.doi.org/10.1080/17435390.2019.1571645] [PMID: 30776941]
[44]
Rodhe Y, Skoglund S, Odnevall Wallinder I, Potácová Z, Möller L. Copper-based nanoparticles induce high toxicity in leukemic HL60 cells. Toxicol In Vitro 2015; 29(7): 1711-9.
[http://dx.doi.org/10.1016/j.tiv.2015.05.020] [PMID: 26028147]
[45]
Lee IC, Ko JW, Park SH, et al. Copper nanoparticles induce early fibrotic changes in the liver via TGF- β/Smad signaling and cause immunosuppressive effects in rats. Nanotoxicology 2018; 12(6): 637-51.
[http://dx.doi.org/10.1080/17435390.2018.1472313] [PMID: 29848140]
[46]
Gagner JE, Lopez MD, Dordick JS, Siegel RW. Effect of gold nanoparticle morphology on adsorbed protein structure and function. Biomaterials 2011; 32(29): 7241-52.
[http://dx.doi.org/10.1016/j.biomaterials.2011.05.091] [PMID: 21705074]
[47]
Pan Y, Neuss S, Leifert A, et al. Size-dependent cytotoxicity of gold nanoparticles. Small 2007; 3(11): 1941-9.
[http://dx.doi.org/10.1002/smll.200700378] [PMID: 17963284]
[48]
Mironava T, Hadjiargyrou M, Simon M, Rafailovich MH. Gold nanoparticles cellular toxicity and recovery: Adipose derived stromal cells. Nanotoxicology 2014; 8(2): 189-201.
[http://dx.doi.org/10.3109/17435390.2013.769128] [PMID: 23330784]
[49]
Akhtar MJ, Ahamed M, Kumar S, et al. Nanotoxicity of pure silica mediated through oxidant generation rather than glutathione depletion in human lung epithelial cells. Toxicology 2010; 276(2): 95-102.
[http://dx.doi.org/10.1016/j.tox.2010.07.010] [PMID: 20654680]
[50]
Xu Z, Chou L, Sun J. Effects of SiO2 nanoparticles on HFL-I activating ROS-mediated apoptosis via p53 pathway. J Appl Toxicol 2012; 32(5): 358-64.
[http://dx.doi.org/10.1002/jat.1710] [PMID: 21766316]
[51]
Hassankhani R, Esmaeillou M, Tehrani AA, Nasirzadeh K, Khadir F, Maadi H. In vivo toxicity of orally administrated silicon dioxide nanoparticles in healthy adult mice. Environ Sci Pollut Res Int 2015; 22(2): 1127-32.
[http://dx.doi.org/10.1007/s11356-014-3413-7] [PMID: 25113834]
[52]
Kim SK, Kim JH, Kim C-S, et al. Immunotoxicity of silicon dioxide nanoparticles with different sizes and electrostatic charge. Int J Nanomedicine 2014; 9 (Suppl. 2): 183-93.
[http://dx.doi.org/10.2147/IJN.S57934] [PMID: 25565836]
[53]
Guo C, Wang J, Yang M, et al. Amorphous silica nanoparticles induce malignant transformation and tumorigenesis of human lung epithelial cells via P53 signaling. Nanotoxicology 2017; 11(9-10): 1176-94.
[http://dx.doi.org/10.1080/17435390.2017.1403658] [PMID: 29164963]
[54]
Guo Z, Martucci NJ, Liu Y, Yoo E, Tako E, Mahler GJ. Silicon dioxide nanoparticle exposure affects small intestine function in an in vitro model. Nanotoxicology 2018; 12(5): 485-508.
[http://dx.doi.org/10.1080/17435390.2018.1463407] [PMID: 29668341]
[55]
Kang SJ, Kim BM, Lee YJ, Hong SH, Chung HW. Titanium dioxide nanoparticles induce apoptosis through the JNK/p38-caspase-8-Bid pathway in phytohemagglutinin-stimulated human lymphocytes. Biochem Biophys Res Commun 2009; 386(4): 682-7.
[http://dx.doi.org/10.1016/j.bbrc.2009.06.097] [PMID: 19555659]
[56]
Liu Y, Xu Z, Li X. Cytotoxicity of titanium dioxide nanoparticles in rat neuroglia cells. Brain Inj 2013; 27(7-8): 934-9.
[http://dx.doi.org/10.3109/02699052.2013.793401] [PMID: 23789867]
[57]
Armand L, Tarantini A, Beal D, et al. Long-term exposure of A549 cells to titanium dioxide nanoparticles induces DNA damage and sensitizes cells towards genotoxic agents. Nanotoxicology 2016; 10(7): 913-23.
[http://dx.doi.org/10.3109/17435390.2016.1141338] [PMID: 26785166]
[58]
Nichols CE, Shepherd DL, Hathaway QA, et al. Reactive oxygen species damage drives cardiac and mitochondrial dysfunction following acute nano-titanium dioxide inhalation exposure. Nanotoxicology 2018; 12(1): 32-48.
[http://dx.doi.org/10.1080/17435390.2017.1416202] [PMID: 29243970]
[59]
Hu H, Li L, Guo Q, et al. RNA sequencing analysis shows that titanium dioxide nanoparticles induce endoplasmic reticulum stress, which has a central role in mediating plasma glucose in mice. Nanotoxicology 2018; 12(4): 341-56.
[http://dx.doi.org/10.1080/17435390.2018.1446560] [PMID: 29510645]
[60]
Siddiqui MA, Alhadlaq HA, Ahmad J, Al-Khedhairy AA, Musarrat J, Ahamed M. Copper oxide nanoparticles induced mitochondria mediated apoptosis in human hepatocarcinoma cells. PLoS One 2013; 8(8): e69534.
[http://dx.doi.org/10.1371/journal.pone.0069534] [PMID: 23940521]
[61]
Sun T, Yan Y, Zhao Y, Guo F, Jiang C. Copper oxide nanoparticles induce autophagic cell death in A549 cells. PLoS One 2012; 7(8): e43442.
[http://dx.doi.org/10.1371/journal.pone.0043442] [PMID: 22916263]
[62]
Chibber S, Ansari SA, Satar R. New vision to CuO, ZnO, and TiO2 nanoparticles: their outcome and effects. J Nanopart Res 2013; 15(4): 1492.
[http://dx.doi.org/10.1007/s11051-013-1492-x]
[63]
Bulcke F, Thiel K, Dringen R. Uptake and toxicity of copper oxide nanoparticles in cultured primary brain astrocytes. Nanotoxicology 2013; 8(7): 1-11.
[http://dx.doi.org/10.3109/17435390.2013.829591] [PMID: 23889294]
[64]
Gosens I, Cassee FR, Zanella M, et al. Organ burden and pulmonary toxicity of nano-sized copper (II) oxide particles after short-term inhalation exposure. Nanotoxicology 2016; 10(8): 1084-95.
[http://dx.doi.org/10.3109/17435390.2016.1172678] [PMID: 27132941]
[65]
Ko JW, Shin NR, Park JW, et al. Copper oxide nanoparticles induce collagen deposition via TGF-β1/Smad3 signaling in human airway epithelial cells. Nanotoxicology 2018; 12(3): 239-50.
[http://dx.doi.org/10.1080/17435390.2018.1432778] [PMID: 29383958]
[66]
Johnson BM, Fraietta JA, Gracias DT, et al. Acute exposure to ZnO nanoparticles induces autophagic immune cell death. Nanotoxicology 2015; 9(6): 737-48.
[http://dx.doi.org/10.3109/17435390.2014.974709] [PMID: 25378273]
[67]
Wang MM, Wang YC, Wang XN, et al. Mutagenicity of ZnO nanoparticles in mammalian cells: Role of physicochemical transformations under the aging process. Nanotoxicology 2015; 9(8): 972-82.
[http://dx.doi.org/10.3109/17435390.2014.992816] [PMID: 25676621]
[68]
Ge W, Zhao Y, Lai FN, et al. Cutaneous applied nano-ZnO reduce the ability of hair follicle stem cells to differentiate. Nanotoxicology 2017; 11(4): 465-74.
[http://dx.doi.org/10.1080/17435390.2017.1310947] [PMID: 28326861]
[69]
Wiesmann N, Kluenker M, Demuth P, Brenner W, Tremel W, Brieger J. Zinc overload mediated by zinc oxide nanoparticles as innovative anti-tumor agent. J Trace Elem Med Biol 2019; 51: 226-34.
[http://dx.doi.org/10.1016/j.jtemb.2018.08.002] [PMID: 30115501]
[70]
Åkerman ME, Chan WCW, Laakkonen P, Bhatia SN, Ruoslahti E. Nanocrystal targeting in vivo. Proc Natl Acad Sci 2002; 99: (20): 12617-21.
[http://dx.doi.org/10.1073/pnas.152463399] [PMID: 12235356]
[71]
Winnik FM, Maysinger D. Quantum dot cytotoxicity and ways to reduce it. Acc Chem Res 2013; 46(3): 672-80.
[http://dx.doi.org/10.1021/ar3000585] [PMID: 22775328]
[72]
O’Hara T, Seddon B, O’Connor A, et al. Quantum dot nanotoxicity investigations using human lung cells and toxor electrochemical enzyme assay methodology. ACS Sens 2017; 2(1): 165-71.
[http://dx.doi.org/10.1021/acssensors.6b00673] [PMID: 28722443]
[73]
Hu Y, Chen Y, Lin L, Zhang J, Lan R, Wu B. Studies on antimicrobial peptide-loaded nanomaterial for root caries restorations to inhibit periodontitis related pathogens in periodontitis care. J Microencapsul 2021; 38(2): 89-99.
[http://dx.doi.org/10.1080/02652048.2020.1842528] [PMID: 33153344]
[74]
Yadav P, Yadav AB. Preparation and characterization of BSA as a model protein loaded chitosan nanoparticles for the development of protein-/peptide-based drug delivery system. Futur. J Pharm Sci 2021; 7: 200.
[75]
Mathew A, Fukuda T, Nagaoka Y, et al. Curcumin loaded-PLGA nanoparticles conjugated with Tet-1 peptide for potential use in Alzheimer’s disease. PLoS One 2012; 7(3): e32616.
[http://dx.doi.org/10.1371/journal.pone.0032616] [PMID: 22403681]
[76]
Tenland E, Pochert A, Krishnan N, et al. Effective delivery of the anti-mycobacterial peptide NZX in mesoporous silica nanoparticles. PLoS One 2019; 14(2): e0212858.
[http://dx.doi.org/10.1371/journal.pone.0212858] [PMID: 30807612]
[77]
Wang Y, Jiang W, Jiang Y, et al. Self-assembled nano-micelles of lactoferrin peptides: Structure, physicochemical properties, and application for encapsulating and delivering curcumin. Food Chem 2022; 387: 132790.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy