Acessibilidade / Reportar erro

Nanobubbles: A Novel Targeted Drug Delivery System

Abstract

Nanobubbles are nanometer size bubbles having different constituents of varying physicochemical characteristic for the inner core and outer shell. Nanobubbles are mainly fabricated to improve the stability, bioavailability and improve the biodistribution of the delivered drug to the specific targeted site. Their small sizes bubbles allow the possibility of extravasation from blood vessels into the surrounding tissues and ultrasound-targeted site-specific release with minimal invasiveness. Nanobubbles are developing as important contrast agents for imaging and carriers for drug delivery at targeted region. Sonication is the primary method for preparation of nanobubbles followed by thin-layer evaporation, high shear emulsification, mechanical agitation and coacervation or coalescence. With exposure to ultrasound/extracorporeal shock waves, the drug is liberated from the nanobubbles into the target cells. This review paper is an effort to reveal the different formulation development techniques briefly and varying shell and core content for developing nanobubbles.

Keywords:
Nanobubbles; Target drug delivery; Gene therapy; Thrombolysis

INTRODUCTION

Nanobubbles are gas carrying concavities in aqueous solution with the size range of below 1µm. Bubbles are round, globular particles with a shell and a gas-filled core structure that enables them to perceptibly dynamic properties (Unger et al., 2004Unger EC, Porter T, Culp W, Labell R, Matsunaga T, Zutshi R. Therapeutic applications of lipid-coated microbubbles. Adv Drug Deliv Rev . 2004;56(9):1291-1314.; Ferrara, 2008Ferrara KW. Driving delivery vehicles with ultrasound. Adv Drug Deliv Rev. 2008;60(10):1097-1102.; Sirsi, Borden, 2009Sirsi S, Borden M. Microbubble compositions, properties and biomedical applications. Bubble Sci Eng Technol. 2009;1(1-2):3-17.). The shell mostly composed of lipids, polymers, proteins, surfactant and polyelectrolyte multilayer, whereas core base can be charged up with different gases, such as perfluorocarbon, carbon dioxide, sulfur hexafluoride and air (Figure 1) (Cavalli, Soster, Argenziano, 2016Cavalli R, Soster M, Argenziano M. Nanobubbles: a promising efficienft tool for therapeutic delivery. Ther Deliv. 2016;7(2):117-138.). Their shell constitution can ensure a significant impact on the half-life of a bubble, as it regulates the interchange of gas from the shell core to the surrounding medium. Indeed, shell thickness and elasticity are the contributing parameters in overall nanobubble stability (Delalande et al., 2012Delalande A, Postema M, Mignet N, Midoux P, Pichon C. Ultrasound and microbubble-assisted gene delivery: recent advances and ongoing challenges. Ther Deliv . 2012;3(10):1199-1215.). A variation on chemical components of ultrasound contrast agents (UCA), acoustic pressure, fluid viscosity, ambient temperature and pressure, gas diffusivity, and size distribution can all affect echogenicity and stability of UCA which affects shell properties and stability of UCA which in turn affect the threshold of nonlinear bubble activity (Perera et al., 2018Perera R, Nittayacharn P, Cooley M, Jung O, Exner AA. Ultrasound contrast agents and delivery systems in cancer detection and therapy. Adv Cancer Res . 2018;139:57-84.).

FIGURE 1
Schematic representation of nanobubble structure.

These nanobubbles (NBs) are sub-micron sized and they are under development with the aim to extend stability, bioavailability and to augment the biodistribution of the drug to be delivered at the targeted pathological region. Devilish of Microbubbles is not possible from the bloodstream due to its large size (Bisazza et al., 2011Bisazza A, Civra A, Donalisio M, Lembo D, Cavalli R. The in vitro characterization of dextran-based nanobubbles as possible DNA transfection agents. Soft Matter. 2011;7(22):10590-10593.; Nomikou, McHale, 2012Nomikou N, McHale AP. Microbubble-enhanced ultrasound-mediated gene transfer-towards the development of targeted gene therapy for cancer. Int J Hyperthermia. 2012;28(4):300-310.; Cavalli, Bisazza, Lembo, 2013Cavalli R, Bisazza A, Lembo D. Micro-and nanobubbles: a versatile non-viral platform for gene delivery. Int J Pharm . 2013;456(2):437-445.). Nanobubbles pave the route for extravasation from blood vessels into surrounding tissues, thus improving delivery efficiency and localization (Cavalli, Bisazza, Lembo, 2013Cavalli R, Bisazza A, Lembo D. Micro-and nanobubbles: a versatile non-viral platform for gene delivery. Int J Pharm . 2013;456(2):437-445.). The fabrication and initial application of nanobubbles have shown promising results in recent years, with improved stability and loading compared to microbubbles (MBs) due to the size range between 400-800 nm in diameter, as well as the achievement of system extravasation. Conversely, MBs are unable to leave the vasculature, even in solid tumors, which often have leaky vasculature and poor lymphatic drainage which leads to extravasation and retention of macromolecules, also known as the EPR effect (enhanced permeability and retention) (Maeda et al., 2000Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release . 2000;65(1-2):271-284.; Oeffinger, Wheatley, 2004Oeffinger BE, Wheatley MA. Development and characterization of a nano-scale contrast agent. Ultrasonics. 2004;42(1-9):343-347.).

In the view of enhanced nuclear envelope (NE) rupture, optical methods specifically, laser induced ‘Photoblation technique’ has shown an inflict damage or minute mechanical damage to the NE using vapour nanobubble (VNB)-mediated photoporation. This technique exploits the optical properties of sensitizing plasmonic nanoparticles such as gold nanoparticles (AUNPs) in significant enhancement of efficiency and throughput. Nanobubbles were initially fabricated as contrast agents, ultrasonic energy reflectors, inducing oscillations asymmetrically in the provision of exposure to acoustic fields in given diameters (Houthaeve et al., 2018Houthaeve G, Xiong R, Robijns J, Luyckx B, Beulque Y, Brans T, et al. Targeted Perturbation of Nuclear Envelope Integrity with Vapor Nanobubble-Mediated Photoporation. ACS Nano. 2018;12(8):7791-7802.).

Ultrasound imaging has become a successful and well established diagnostic technique for clinical decision making (Frinking et al., 2000Frinking PJ, Bouakaz A, Kirkhorn J, Ten Cate FJ, De Jong N. Ultrasound contrast imaging: current and new potential methods. Ultrasound Med Biol. 2000;26(6):965-975.). It is due to ultrasound imaging is non-invasive, less expensive and provides real-time images of soft tissue structures such as heart and blood flow without the use of ionizing radiation. Ultrasound beam reduces the toxic side effect of anti-cancer drugs used in cancer treatment (Liu, Miyoshi, Nakamura, 2006Liu Y, Miyoshi H, Nakamura M. Encapsulated ultrasound microbubbles: therapeutic application in drug/gene delivery. J Control Release . 2006;114(1):89-99.; Husseini, Pitt, 2008Husseini GA, Pitt WG. The use of ultrasound and micelles in cancer treatment. J Nanosci Nanotechnol . 2008;8(5):2205-2215.). The delivery of drugs to organs and tissues are affected by two main ultrasound effects including the cavitation and sonoporation effects. The cavitation effect results in the reduction of bubble size, while the sonoporation effect leads to uptake of the reduced bubble (Ayodele et al., 2017Ayodele AT, Valizadeh A, Adabi M, Esnaashari SS, Madani F, Khosravani M, et al. Ultrasound nanobubbles and their applications as theranostic agents in cancer therapy: A review. Biointerface Res Appl Chem. 2017;7(6):2253-2262.). Therapeutic ultrasound was considered a tool for hyperthermia or thermal ablation of tumors. Ultrasound-assisted drug delivery system can help, the drug to penetrate through various tissues, change in permeability or absorption of the drug into the cells and tissue, converting specific drug into a therapeutically active form its inactive state (Wu, 1998Wu J. Temperature rise generated by ultrasound in the presence of contrast agent. Ultrasound Med Biol . 1998;24(2):267-274.; Husseini, Runyan, Pitt, 2002Husseini GA, Pitt WG. The use of ultrasound and micelles in cancer treatment. J Nanosci Nanotechnol . 2008;8(5):2205-2215.; Smith et al., 2003Smith NB, Lee S, Maione E, Roy RB, McElligott S, Shung KK. Ultrasound-mediated transdermal transport of insulin in vitro through human skin using novel transducer designs. Ultrasound Med Biol . 2003;29(2):311-317.; Dijkmans et al., 2004Dijkmans P, Juffermans L, Musters R, van Wamel A, Ten Cate F, van Gilst W, et al. Microbubbles and ultrasound: from diagnosis to therapy. Eur J Echocardiogr. 2004;5(4):245-246.).

A wide variety of applications for drug delivery with ultrasound are under investigation in numerous areas (Takeuchi, Sato, Kawashima, 2002Takeuchi S, Sato T, Kawashima N. Nonlinear response of microbubbles coated with surfactant membrane developed as ultrasound contrast agent-experimental study and numerical calculations. Colloids Surf B . 2002;24(3-4):207-216.). Recent advances in nanobubbles formulation in the treatment of specific targeted tissues with the application of US energy are summarized in the following the Table I.

TABLE I
Recent advances in nanobubbles formulation in the treatment of specific targeted tissues with the application of US energy

PRINCIPLE

Nanobubbles have the advantage of modifying the normal water characteristics. These bubbles are minute or more specific nanoscopic than normal bubbles and do not float and have reduced buoyancy as the diameter is small. Due to its longevity, physical stability, they do not readily dissolve. With established Laplace pressure equation, the pressure difference within the inner and the outer side of a droplet (or a bubble) can be denoted as:

Δ P = P i n n e r - P o u t e r = 2 σ r

Where Pinner and P outer are the pressures inner and outer of the bubble respectively with interfacial tension "σ" and "r" as the radius of the bubble. The US energy is responsible in activating liquid to the gas phase inside the nanobubbles through the Acoustic Droplet Vaporization (ADV) mechanism (Kripfgans et al., 2004Kripfgans OD, Fabiilli ML, Carson PL, Fowlkes JB. On the acoustic vaporization of micrometer-sized droplets. J Acoust Soc Am. 2004;116(1):272-281.). The Laplace equation of pressure is inversely related to the bubble size and hence small sized bubbles contribute to high-pressure values in comparison with larger ones. The bubble gets shrinks with an increase in Laplace pressure as inner gas quits the core, navigated by the pressure gradient and hence acceleration of gas intemperance rate, followed with the shrinkage of the bubble until the rupturing of the system. As the surface tension is responsible for accelerating the pressure for dissolution of the bubble by diffusion, dissolution rate of the bubble in vivo is solely controlled by Laplace pressure (Van Liew, Raychaudhuri, 1997Van Liew HD, Raychaudhuri S. Stabilized bubbles in the body: pressure-radius relationships and the limits to stabilization. J Appl Physiol. 1997;82(6):2045-2053.).

APPLICATIONS OF NANOBUBBLE

Targeted nanobubble

Nanobubbles are also compliant for modifications at surface to augment the signals and selectivity about tumor, and suppress nonspecific toxicity. Targeting nanobubbles through modifications at surface is also considered an interesting theranostic approach. A novel in targeted nanobubble system is a most favourable tool in diagnosis and evaluation for Her-2-positive breast cancer treatment response. Thin-film hydration and sonication method was used in fabrication of PEGylated phospholipid shell NBs to which the covalent bondage is established by Herceptin molecules. The NBs-Her linkage provides long-acting contrast enhancement, without toxic effects as proved in earlier performed vitro and in vivo experiments. Importantly, it is proven with efficient penetration in tumor and were retained longer in Her-2-expressing tumors by in vivo results (Jiang et al., 2016Jiang Q, Hao S, Xiao X, Yao J, Ou B, Zhao Z, et al. Production and characterization of a novel long-acting Herceptin-targeted nanobubble contrast agent specific for Her-2-positive breast cancers. Breast Cancer. 2016;23(3):445-455.).

Drug Delivery

Nanobubbles drug delivery is potentially being exploited in the delivery of anticancer drugs like doxorubicin in both in vitro and in vivo. These NBs reach and get assembled at the site of a tumor followed by their amalgamation to form microbubbles. Further, these microbubbles undergo distortion at the target site with intense ultrasound and hence release of the drug is achieved which results in accumulating the higher proportion of drug within the targeted cells with increased efficacy and reduced toxicity. Further exploration is necessary for these method's utility while treating various malignancies.

A polymeric nanobubble system to increase the doxorubicin sensitivity of cancer cells associated with US. The systems developed include perfluorocarbon nanodroplets stabilized by a biodegradable block copolymer wall (PEG-PLLA or PEG-PCL). With increase in temperature at about to physiological temperatures, the nanodroplets get converted into nano/microbubbles. As per the received sonic wave signals by tumor-directed ultrasound, cavitations occurs in doxorubicin-loaded nanobubbles and finally get collapse, to release the encapsulated drug and increasing the effectiveness in tumor chemotherapy in vivo (Gao et al., 2008Gao Z, Kennedy AM, Christensen DA, Rapoport NY. Drug-loaded nano/microbubbles for combining ultrasonography and targeted chemotherapy. Ultrasonics. 2008;48(4):260-270.).

Nanobubbles have also been used for treatment of Parkinson’s disease. The authors demonstrated that nanobubbles can be used to delivery apomorphine, a particularly beneficial but unstable drug for treating Parkinson’s disease, through the blood barrier (Hwang et al., 2009Hwang TL, Lin YK, Chi CH, Huang TH, Fang JY. Development and evaluation of perfluorocarbon nanobubbles for apomorphine delivery. J Pharm Sci. 2009;98(10):3735-3747.).

The approach like ‘Photoablation technique’ can be used to promote the influx of various macromolecules that are too large to migrate passively through the NE. Thus, by providing unprecedented control over nuclear compartmentalization, nuclear photoporation offers a powerful tool over nanobubble based drug delivery applications (Houthaeve et al., 2018Houthaeve G, Xiong R, Robijns J, Luyckx B, Beulque Y, Brans T, et al. Targeted Perturbation of Nuclear Envelope Integrity with Vapor Nanobubble-Mediated Photoporation. ACS Nano. 2018;12(8):7791-7802.).

Another approach viz. Sonoporation technology is having an application in the transfer of drug or gene into the cell or tissue in the targeted region (Fan, Kumon, Deng, 2014Fan Z, Kumon RE, Deng CX. Mechanisms of microbubble-facilitated sonoporation for drug and gene delivery. Ther Deliv . 2014;5(4):467-486.). Presently, they are also under investigations in relation with gene and gas along with drug delivery (Marxer et al., 2011Marxer EEJ, Brüßler J, Becker A, Schümmelfeder J, Schubert R, Nimsky C, et al. Development and characterization of new nanoscaled ultrasound active lipid dispersions as contrast agents. Eur J Pharm Biopharm . 2011;77(3):430-437.; O’Neill, Rapoport, 2011O’Neill BE, Rapoport N. Phase-shift, stimuli-responsive drug carriers for targeted delivery. Ther Deliv . 2011;2(9):1165-1187.).

(Suzuki et al., 2005Suzuki M, Koshiyama K, Shinohara F, Mori S, Ono M, Tomita Y, et al. Nanobubbles enhanced drug susceptibility of cancer cells using ultrasound. International Congress Series. Elsevier, 2005, p. 338-339.) reported on the use of nanobubbles combined with US to permeabilise four types cancer cells and potentiate the cytotoxic effect of anticancer drugs (cisplatin and 5-FU). Cell sensitivity to cisplatin and 5-FU was effectively increased with nanobubbles and ultrasound.

Oxygen Delivery

Nanobubble can be utilized to create supersaturated fluids for oxygen delivery. In a previous study, it has been demonstrated and proved that oxygen nanobubbles are most effective therapy in reversing the hypoxia (Khan et al., 2018bKhan MS, Hwang J, Seo Y, Shin K, Lee K, Park C, et al. Engineering oxygen nanobubbles for the effective reversal of hypoxia. Artif Cells Nanomed Biotechnol. 2018b;46(sup3):S318-S327. ). It also demonstrated that oxygen nanobubble could be prominent therapy in down regulation of an intermediate protein, hypoxia-inducible factor-1α (HIF-1α), with significant role in chemotherapy. Higher oxygen availability also favours the therapeutic efficacy of radiotherapy and photodynamic therapy (Khan et al., 2018aKhan MS, Hwang J, Lee K, Choi Y, Kim K, Koo HJ, et al. Oxygen-carrying micro/nanobubbles: Composition, synthesis techniques and potential prospects in photo-triggered theranostics. Molecules. 2018a;23(9):2210.) and therefore, oxygen nanobubbles have many potential therapeutic applications.

(Matsuki et al., 2014Matsuki N, Ishikawa T, Ichiba S, Shiba N, Ujike Y, Yamaguchi T. Oxygen supersaturated fluid using fine micro/nanobubbles. Int J Nanomed . 2014;9:4495.) demonstrated that oxygen-filled nanobubbles can generate fluids with supersaturated oxygen that would not sufficiently accommodate the same to be safely infused into blood vessels. A normal saline solution containing oxygen-filled nanobubbles was found effective treatment in improvement of blood oxygenation. Thus, the use of oxygen-filled nanobubbles would be potentially effective and method of choice to improve blood oxygenation in disorders, ischemic heart diseases (IHD), anticancer chemoradiation therapies and in control of hypoxia related infection.

Theranostic Delivery

Another application is the development of a system that provides imaging support to therapeutic treatment. This approach, which is called theranostic, appears to be particularly effective in improving treatment of cancer or other important diseases at an early stage, as it can image the pathological tissues while also monitoring delivery kinetics and biodistribution of a drug, thereby obtaining important benefits in terms of tuning therapy and doses, and reducing adverse side effects (Shi, 2009Shi D. Integrated multifunctional nanosystems for medical diagnosis and treatment. Adv Funct Mater. 2009;19(21):3356-3373.; Kelkar, Reineke, 2011Kelkar SS, Reineke TM. Theranostics: combining imaging and therapy. Bioconjug Chem . 2011;22(10):1879-1903. ; Lammers et al., 2011Lammers T, Aime S, Hennink WE, Storm G, Kiessling F. Theranostic nanomedicine. Acc Chem Res. 2011;44(10):1029-1038. ; Terreno, Uggeri, Aime, 2012Terreno E, Uggeri F, Aime S. Image guided therapy: the advent of theranostic agents. J Control Rel . 2012;161(2):328-337. ).

Lipid nanobubbles for Ultrasound imaging detection have been proposed as an in vivo contrast enhanced system for imaging, and as a tumor drug delivery (Yin et al., 2012Yin T, Wang P, Zheng R, Zheng B, Cheng D, Zhang X, et al. Nanobubbles for enhanced ultrasound imaging of tumors. Int J Nanomed . 2012;7:895. ). Notably, the use of MRI detectable microbubbles for theranostic purposes has already been proposed. Moreover, PLGA micro/nanobubbles loaded with Gd-DTPA were fabricated as multimodal contrast agents for both MRI and US (Ao et al., 2010Ao M, Wang Z, Ran H, Guo D, Yu J, Li A, et al. Gd-DTPA-loaded PLGA microbubbles as both ultrasound contrast agent and MRI contrast agent-A feasibility research. J Biomed Mater Res Part B. 2010;93(2):551-556. ).

Gene therapy

Liposome-based nanobubbles and microbubbles are also under investigation for effective gene-based drug delivery with the application of non-viral vectors. Researches also have shown that ultrasound energy fused nanobubbles have shown improvement of gene transfer in diversified studies (Negishi et al., 2008Negishi Y, Endo Y, Fukuyama T, Suzuki R, Takizawa T, Omata D, et al. Delivery of siRNA into the cytoplasm by liposomal bubbles and ultrasound. J Control Release . 2008;132(2):124-130.; Suzuki et al., 2008Suzuki R, Takizawa T, Negishi Y, Utoguchi N, Maruyama K. Effective gene delivery with novel liposomal bubbles and ultrasonic destruction technology. Int J Pharm . 2008;354(1-2):49-55. ).

Plasmid DNA encoding TNF-α, encapsulated in lipid NBs containing octafluoropropane (C3F8) gas, can be transfected to tumor cells in a spatial and temporal manner, as triggered by US exposure. This results in local production of TNF-α and thus causes antitumor action, including p53-dependent apoptosis activation, decrease in tumor vessel density and suppression of tumor size, but which reduces the acute toxic effects (Watanabe et al., 2010Watanabe Y, Horie S, Funaki Y, Kikuchi Y, Yamazaki H, Ishii K, et al. Delivery of Na/I symporter gene into skeletal muscle using nanobubbles and ultrasound: visualization of gene expression by PET. J Nucl Med. 2010;51(6):951-958. ).

Thrombolysis

Various studies also indicate the application of nanobubbles in the removal of the clot in vascular tissues with induction of ultrasound. This process is widely known as sonothrombolysis. It is a non-invasive method causing less damage to endothelium (Iverson et al., 2008Iverson N, Plourde N, Chnari E, Nackman GB, Moghe PV. Convergence of nanotechnology and cardiovascular medicine. BioDrugs. 2008;22(1):1-10. ).

ADVANTAGES OF NANOBUBBLES

Till the date, chemotherapy is routinely being used as the prime modality in treatment of malignant neoplasms and substantial therapy in improving the rate of survival in case of cancer patients. In spite of that, further assessment is absolutely required to determine the efficacy of chemotherapeutic drugs to maximize drug toxicity in treating the cancer cells. The frequently found the associated adverse effect in such case is systemic toxicity. Moreover, the local delivery of chemotherapeutic drugs may minimise toxicity with increasing therapeutic dose at targeted sites and by lowering the plasma levels of circulating drugs. Ultrasound-targeted nano/microbubble destruction (UTN/MD) therapy has been widely used due to its non-invasiveness and targetability, as an effective drug delivery system (Zhou et al., 2019Zhou X, Guo L, Shi D, Duan S, Li J. Biocompatible chitosan nanobubbles for ultrasound-mediated targeted delivery of doxorubicin. Nanoscale Res Lett. 2019;14(1):24. ).

The ultrasound (US)-targeted nanobubble destruction (UTND) method has become a new trend as targeted drug delivery system to solid tumors which imminently lowers systemic drug exposure and enhances therapeutic efficacy. Thus, UTND has multiple significant advantages when compared with other drug delivery systems, Nanobubbles can be formulated easily by modified emulsification processes and employed as US contrast agents in visualising tumors (Tian et al., 2018Tian Y, Liu Z, Zhang L, Zhang J, Han X, Wang Q, et al. Apatinib-loaded lipid nanobubbles combined with ultrasound-targeted nanobubble destruction for synergistic treatment of HepG2 cells in vitro. OncoTargets Ther. 2018;11:4785. ).

Nanobubbles being the contrast agents may disrupt blood vessels and thus enhances the site-specific delivery of drugs. This could be the most effective approach employing the EPR effect in passive targeting of tumors. Moreover, NBs in combination with US could promote acoustic cavitation, improving cell membrane stimulation and ultimately permeabilization in drug uptake by tumor cells. The extensive use of the retention (EPR) effect with enhanced permeability in delivering drug at target tumor site is the motto that drug delivery by theranostics therapy involves. The EPR effect reasonably makes accumulate various types of nanomedicines in solid tumors, demonstrated by different studies. However, EPR is blamed as phenomenon with high variation due to tumor heterogeneity that ultimately results in delivering low drugs in clinical trials (Duan et al., 2020Duan L, Yang L, Jin J, Yang F, Liu D, Hu K, et al. Micro/nano-bubble-assisted ultrasound to enhance the EPR effect and potential theranostic applications. Theranostics. 2020;10(2):462-483. ).

The earlier studies have thrown light on nontargeted NBs that are easily accumulated in the reticulo-endothelial system, causing lower drug concentration at the tumor site. To reduce systemic toxicity and increase therapeutic efficacy, it is crucial to construct drug-loaded and targeted NBs, carrying antibodies and peptides as tumor-specific ligands (Tian et al., 2018Tian Y, Liu Z, Zhang L, Zhang J, Han X, Wang Q, et al. Apatinib-loaded lipid nanobubbles combined with ultrasound-targeted nanobubble destruction for synergistic treatment of HepG2 cells in vitro. OncoTargets Ther. 2018;11:4785. ).

The formulations with echogenic bubble have wide applications in diagnosis of disease and as therapeutics. Therefore, nanobubbles were formulated and the evaluation of contrast agent was performed in determining property of nanosized bubble in ultrasonic imaging. Drug loaded NBs exhibit excellent ability to achieve ultrasound enhancement and effective drug loading/targeting. The release of drug from drug loaded-NBs at targeted site assure better efficacy with promotion of Ultrasound irradiation. The drug-loaded NBs could promote drug delivery to cells significantly and the process be analyzed with sigmoidal type pharmacokinetic curve. It further can be concluded that the formulation of nanobubble is most promising approach for both drug delivery enhancing. as well as ultrasound imaging (Wang et al., 2010Wang Y, Li X, Zhou Y, Huang P, Xu Y. Preparation of nanobubbles for ultrasound imaging and intracelluar drug delivery. Int J Pharm . 2010;384(1-2):148-153. ).

DISADVANTAGES OF NANOBUBBLES

The major drawback of the lipid-encapsulated NBs as a drug delivery vehicle is its low payload efficacy. To combat this, an oil shell can be incorporated to the interior of the lipid monolayer to enhance payload efficacy. The shell ingredients of NBs such as polydimethylsiloxane, Tween 80, polyethylene glycol stearate or polyvinyl pyrrolidone that were commonly added, reinforce the shell avoiding any gas escape and thus prolonging the liposome gas bubbles lifetime in the bloodstream. The avidin-biotin interaction method is frequently used to tether antibodies to micro/ nanobubbles, leading to the development of a molecular targeting US imaging agent. However, avidin still has limitations due to its strong immunogenic character (Hamano et al., 2019Hamano N, Kamoshida S, Kikkawa Y, Yano Y, Kobayashi T, Endo-Takahashi Y, et al. Development of antibody-modified nanobubbles using Fc-region-binding polypeptides for ultrasound imaging. Pharmaceutics. 2019;11(6):283. ).

The use of cationic peptides or cell-penetratinge peptide (CPP) can enable ease for cellular internalization of therapeutic agents that assigned for interaction between positively charged carrier and the negatively charged plasma membrane. A CPP/cargo combination may inhibit the endocytosis pathway and translocate directly into the cells without consuming energy. Use of such CPPs is non-specific functional molecule that can penetrate any cell upon encountering it (Xie et al., 2016Xie X, Lin W, Liu H, Deng J, Chen Y, Liu H, et al. Ultrasound-responsive nanobubbles contained with peptide-camptothecin conjugates for targeted drug delivery. Drug Deliv. 2016;23(8):2756-2764. ).

Interaction of charged components of drugs could be easily coupled electrostatically with the shell when anionic/cationic lipids or polymers viz. electrostatic loading is the premature release of the drug cargo in the body after the intravenous injection. Indeed, blood components, like serum albumin could interact with the nanobubble shell favoring the displacement of the drug (Zelphati et al., 1998Zelphati O, Uyechi LS, Barron LG, Szoka Jr FC. Effect of serum components on the physico-chemical properties of cationic lipid/oligonucleotide complexes and on their interactions with cells. Biochim Biophys Acta. 1998;1390(2):119-133.).

NBs are Ultrasound-responsive drug delivery systems and the actual molecular mechanism behind the DNA damage induced by URDDS remains elusive and subtle and needs detailed study. (Furusawa et al., 2012Furusawa Y, Fujiwara Y, Hassan MA, Tabuchi Y, Morita A, Enomoto A, et al. Inhibition of DNA-dependent protein kinase promotes ultrasound-induced cell death including apoptosis in human leukemia cells. Cancer Lett. 2012;322(1):107-112. ) found that Akt, a substrate of ataxia telangiectasia-mutated and DNA-dependent protein kinase (ATM/DNA-PK) when exposed to ultrasound, phosphorylated to the active form in U937 and Molt-4 cell lines without p53 (Zhao et al., 2013Zhao YZ, Du LN, Lu CT, Jin YG, Ge SP. Potential and problems in ultrasound-responsive drug delivery systems. Int J Nanomed . 2013;8:1621. ).

Most of ultrasound-responsive materials require an ultrasound-responsive core (gaseous, PFC, or gas-generating). These ultrasound-responsive cores consume a lot of space in ultrasound-responsive materials (microbubbles, nanobubbles, or droplets), which makes lower drug/gene-loaded contents, and decrease the amount of drug/gene delivered to diseased tissues, and eventually lead to limited therapeutic efficacy. NBs induce cavitation by using ultrasound intensity in order to promote effective release of drugs/genes from nanomaterials base. But high intensity ultrasound may cause damage to neighbouring healthy cells. The rapid collapse of bubbles and hence rapid release of the drug/gene loaded in the bubbles is caused by high-intensity ultrasound waves induction, that may or may not meet the specifications for sustained release of certain drugs (e.g., insulin) (Cai et al., 2020Cai X, Jiang Y, Lin M, Zhang J, Guo H, Yang F, et al. Ultrasound-Responsive Materials for Drug/Gene Delivery. Front Pharmacol. 2020;10:1650. ).

NBs employ US-energy and most important shortcoming of this energy is that it strongly attenuated or resisted by bone. The larger surface area phase arrays and other information gathered from sophisticated imaging techniques may rectify the ultrasound distortion of waves produced by the skull in exposing the brain tissue by means of trans-skull ultrasound (Zhao et al., 2013Zhao YZ, Du LN, Lu CT, Jin YG, Ge SP. Potential and problems in ultrasound-responsive drug delivery systems. Int J Nanomed . 2013;8:1621. ).

Yet, the ultrasonic parameters are the noticeable issues. Any biological tissues may get damaged by low- and high-frequency ultrasound, when heat related to induced sonication higher and the pore generation on cell membranes is not reversible (Mehier-Humbert et al., 2005Mehier-Humbert S, Bettinger T, Yan F, Guy RH. Plasma membrane poration induced by ultrasound exposure: implication for drug delivery. J Control Release . 2005;104(1):213-222. ). Hence, the duration and intensity of ultrasound sonication should be controlled. It also invented that pulsed-focused and ultrasound induced opening of the blood-brain barrier which also was accompanied by increased expression of heat-shock protein 70 (HsP70), interleukin-1, interleukin-18, tumor necrosis factor-α, and inflammation of brain tissues. It ultimate suggests that use of ultrasound-responsive materials in drug/gene delivery to the brain system should be performed with utmost care and extreme caution (Cai et al., 2020Cai X, Jiang Y, Lin M, Zhang J, Guo H, Yang F, et al. Ultrasound-Responsive Materials for Drug/Gene Delivery. Front Pharmacol. 2020;10:1650. ).

CLINICAL TRIALS EXAMPLES

Micro/ Nanobubbles have been applied in drug delivery in clinical trials as proven ultrasound-responsive materials (Hynynen et al., 2001Hynynen K, Pomeroy O, Smith DN, Huber PE, McDannold NJ, Kettenbach J, et al. MR imaging-guided focused ultrasound surgery of fibroadenomas in the breast: a feasibility study. Radiology. 2001;219(1):176-185. ; Dimcevski et al., 2016Dimcevski G, Kotopoulis S, Bjånes T, Hoem D, Schjøtt J, Gjertsen BT, et al. A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer. J Control Release. 2016;243:172-181. ; He et al., 2016He X, Wu DF, Ji J, Ling WP, Chen Xl, Chen YX. Ultrasound microbubble-carried PNA targeting to c-myc mRNA inhibits the proliferation of rabbit iliac arterious smooth muscle cells and intimal hyperplasia. Drug Delivery. 2016;23(7):2482-2487. ). The controllability of delivering the cargo like drugs and gene materials with ultrasonic switch and visualization of treatment was promoted by results of confirmed clinical trials.

MB/NBs as ultrasound contrast agents have been used in clinical for decades (Stride, 2015Stride E. Physical principles of microbubbles for ultrasound imaging and therapy. Translational Neurosonology. Karger Publishers. 2015, p. 11-22. ). Extensive pre-clinical studies also assure that the combination of microbubbles and ultrasound may result in enhanced drug delivery or gene expression at specific and selective sites (Martin, Dayton, 2013Martin KH, Dayton PA. Current status and prospects for microbubbles in ultrasound theranostics. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2013;5(4):329-345.). Preclinical study in xenograft mouse models demonstrates that the anti-tumor effects of doxorubicin can be enhanced and at the same time the drug-induced heart damage in ATC be prevented by the combined treatment with doxorubicin-loaded glycol chitosan NBs and ESWs (Marano et al., 2017Marano F, Frairia R, Rinella L, Argenziano M, Bussolati B, Grange C, et al. Combining doxorubicin-nanobubbles and shockwaves for anaplastic thyroid cancer treatment: preclinical study in a xenograft mouse model. Endocr Relat Cancer. 2017;24(6):275-286. ).

Several pre-clinical and clinical trials reported that apatinib, a novel and selective inhibitor of VEGF receptor 2, was efficient and safer for patients with advanced HCC as the first line therapy. A phase I study was undertaken for patients bearing advanced solid tumors, and turned with the maximum-tolerated dose of 850 mg once daily. In phase II study, patients with metastatic gastric cancer displayed that apatinib improved PFS obvious in comparing with placebo. The adverse effects included hypertension, fatigue and hand-foot syndrome, which were well tackled (Kou et al., 2017Kou P, Zhang Y, Shao W, Zhu H, Zhang J, Wang H, et al. Significant efficacy and well safety of apatinib in an advanced liver cancer patient: a case report and literature review. Oncotarget. 2017;8(12):20510.). In the recent clinical trial, (Wang et al., 2019Wang IL, Hsiao CY, Li YH, Meng FB, Huang CC, Chen YM. Nanobubbles water curcumin extract reduces injury risks on drop jumps in women: a pilot study. Evid Based Complement Alternat Med. 2019;2019:8647587. ) examined the beneficial effects of nanobubbles water applied on curcumin extract enhancing explosiveness without a training program. Nanobubbles water curcumin extract (NCE) supplementation may be helpful for athletes with antifatigue effect and in performing explosiveness or heavy -type exercises including overall physiological protective effects. More specifically, it reduces injury risks in case of women performing drop jumps.

FABRICATION OF NANOBUBBLE

Nanobubbles are fabricated predominantly by sonication, mechanical agitation, coacervation or coalescence thin-layer evaporation, and high shear emulsification techniques. The different processes involved in the construction of nanobubbles are given schematically illustrated in Figures 2, 3 and 4. Nanobubbles are currently under investigation in their application in targeted delivery of drugs, proteins and nucleic acids subjected to ultrasound exposure. The drug of choice is loaded or encapsulated in the inner core and with irradiation of ultrasound/extracorporeal shock waves the drug liberated from the nanobubbles into the target cells.

FIGURE 2
Mechanical Agitation Method.

FIGURE 3
High Shear Emulsification Method.

FIGURE 4
Probe-Type Sonication Method.

Different nanobubble formulations reported earlier are discussed and categorized here, based on inner gas used, there in vitro/in vivo behavior, interaction with ultrasound/extracorporeal shock waves (ESWs) used for delivery of drugs to target region, and their use for imaging, targeting and controlled the release.

Core Gas Based Nanobubbles

The various gases have employed in the preparation of gas filed nanobubble such as air, perfluorocarbon and sulfur hexafluoride. As per previous studies reported in the microbubbles designing, first-generation microbubbles were nothing but air-filled micro-cavities, with mean diameters of 1-8 μm which make them compatible to pass through the pulmonary capillaries. However, such air-charged microbubbles expelled from the circulatory system very rapidly cause of low resistance at different arterial pressure gradients and due to a greater extent of air solubility in blood (Kabalnov et al., 1998Kabalnov A, Klein D, Pelura T, Schutt E, Weers J. Dissolution of multicomponent microbubbles in the bloodstream: 1. Theory. Ultrasound Med Biol. 1998;24(5):739-749.). Therefore, different divergent agents as second-generation agents more specifically hydrophobic gases with high molecular weight viz. perfluorocarbons or sulfur hexafluoride has investigated. In this review, various gas-filled nanobubbles are discussed in Table II shown examples of various nanobubble used for drug delivery.

TABLE II
Various nanobubble used for drug delivery

Perfluorocarbon-based nanobubbles for drug delivery

Lipid-based nanobubbles

Apomorphine loaded on lipid-based nanobubble

Apomorphine was prepared using a perfluorocarbon and lipid-based a nanobubble which is aimed for the sustained type drug delivery. Drugs like apomorphine are known for inherent instability, low oral bioavailability and with a very short half-life which was required a suitable drug delivery carrier for targeting this drug at the specific site. These nanobubbles helped in the safety and enhanced stability of apomorphine (Li, Danhof, Bouwstra, 2001Li GL, Danhof M, Bouwstra JA. Iontophoretic delivery of apomorphine in vitro: physicochemic considerations. Pharm Res . 2001;18(11):1509-1513. ; Subramony, 2006Subramony JA. Apomorphine in dopaminergic therapy. Mol Pharm. 2006;3(4):380-385. ; Hwang et al., 2009Hwang TL, Lin YK, Chi CH, Huang TH, Fang JY. Development and evaluation of perfluorocarbon nanobubbles for apomorphine delivery. J Pharm Sci. 2009;98(10):3735-3747.). Apomorphine microbubbles are unstable with the mean diameter of 1-6 µm which is too large for their intravascular applications (Suzuki et al., 2007Suzuki R, Takizawa T, Negishi Y, Hagisawa K, Tanaka K, Sawamura K, et al. Gene delivery by combination of novel liposomal bubbles with perfluoropropane and ultrasound. J Control Rel. 2007;117(1):130-136. ). The fabrication of nanobubble is depicted in Figure 2. The increased oil and perfluoropentane content ratio result in an enlarged size nanobubbles while an additional increase in the emulsifier content like cholesterol aids in lowering the particle size of the proposed nanobubbles. The increased value of oil and perfluoropentane helps in minimizing the surface charges of nanobubbles while cholesterol has zero effect on surface charges. Cholesterol helps in minimizing surface tension in contrast to oil and perfluoropentane which does not change the surface tension significantly. The nanobubbles loaded with high perfluorocarbon (PNs) protect the apomorphine base. However, no specific effect was studied in perfluorocarbon nanobubbles having high oil content in comparison with free form. High oil content PNs shows very moderate drug delivery rate, in comparison with the non-ultrasound effect and hence drug release percentage is decreased. Incorporation of ultrasound increased 3.9-fold of drug release when compared to the other formulation at the end of the experiment (8th hours). The clinical connection of this is to avoid dose repeatability of apomorphine which could be much favorable in those patients who require per day 10 to 15 injections (Ugwoke et al., 2000Ugwoke MI, Kaufmann G, Verbeke N, Kinget R. Intranasal bioavailability of apomorphine from carboxymethylcellulose-based drug delivery systems. Int J Pharm . 2000;202(1-2):125-131. ). The current studies also prove that the high percentage of oil or perfluoropentane system indicated lower drug release profiles in comparison to the low percentage oil and perfluoropentane in case of the apomorphine HCl and also for its base form. The hemolysis percentage decreases with the increased percentage of oil and perfluoropentane since almost of all phospholipids get occupied at the interface with the emulsified form within the inner phase containing a large volume with a consideration of the hemolysis results, the developed PNs in present study work to be considered as biologically safe and with minimal toxicological risk.

Lipid and polymer-based nanobubbles

Doxorubicin

Doxorubicin was prepared using perfluorocarbon and glycol Chitosan-based on nanobubbles was aimed for the controlled pattern of drug release at the targeted site. Nanobubbles filled with perfluorocarbon gas have used previously for preparation of contrast agent in consideration of their high stability and inert biological nature. Furthermore, its lower water solubility extent reduces the dissolution rate of gas from the inner bubble core into the blood circulation, and hence ultimately stabilizes the system with increasing the in vivo lifetime of the bubbles. Recent studies also have shown that the chitosan shelled perfluoropentane cored NBs have recently designed as agents for theranostic in nano-formulations (Cavalli et al., 2015Cavalli R, Argenziano M, Vigna E, Giustetto P, Torres E, Aime S, et al. Preparation and in vitro characterization of chitosan nanobubbles as theranostic agents. Colloids Surf B. 2015;129:39-46. ; Marano et al., 2016Marano F, Argenziano M, Frairia R, Adamini A, Bosco O, Rinella L, et al. Doxorubicin-loaded nanobubbles combined with extracorporeal shock waves: basis for a new drug delivery tool in anaplastic thyroid cancer. Thyroid. 2016a;26(5):705-716. ).

These nanobubbles, with amalgamation of doxorubicin in anaplastic thyroid cancer (ATC) Cell lines and enhances its anti-tumor activity. In this method, extracorporeal shock waves (ESWs) are widely employed in the delivery of drug at the target site. Extracorporeal shock waves (ESWs) are nothing but acoustic waves which are used frequently in urology for lithotripsy (Jocham, Chaussy, Schmiedt, 1986Jocham D, Chaussy C, Schmiedt E. Extracorporeal shock wave lithotripsy. Urol Int. 1986;41(5):357-368. ). ESWs have to be concentrated with high precision in detail to estimate it’s permeability through the plasma membranes (Constantinides et al., 2000Constantinides PP, Lambert KJ, Tustian AK, Schneider B, Lalji S, Ma W, et al. Formulation development and antitumor activity of a filter-sterilizable emulsion of paclitaxel. Pharm Res. 2000;17(2):175-182. ; Fang et al., 2006Fang JY, Lee WR, Shen SC, Huang YL. Effect of liposome encapsulation of tea catechins on their accumulation in basal cell carcinomas. J Dermatol Sci. 2006;42(2):101-109. ; Ingram, Priston, Sewell, 2006Ingram WM, Priston MJ, Sewell GJ. Improved assay for R (−)-apomorphine with application to clinical pharmacokinetic studies in Parkinson's disease. J Chromatogr B. 2006;831(1-2):1-7.). Doxorubicin also interferes with normal cells causing several adverse effects, among which cardiotoxicity is prominent. Notably, the severity of these effects and their incidence are dose-dependent (Carvalho et al., 2009Carvalho C, Santos RX, Cardoso S, Correia S, Oliveira PJ, Santos MS, et al. Doxorubicin: the good, the bad and the ugly effect. Curr Med Chem. 2009;16(25):3267-3285. ; Chatterjee et al., 2010Chatterjee K, Zhang J, Honbo N, Karliner JS. Doxorubicin cardiomyopathy. Cardiology. 2010;115(2):155-162. ). These nanobubbles help in reducing the cumulative dose and targeting the drug at the tumor region. Doxorubicin-loaded nanobubble preparation as shown in Figure 3. For tuning the formulation, two different types of fluorescent glycol chitosan nanobubbles formulation was carried out.

The empty nanobubbles and the drug-loaded nanobubbles presented an average diameter lower than 500 nm with a positively charged surface. Doxorubicin-loaded nanobubbles do not significantly alter these values, in the presence or absence of an ESW treatment when compared to a doxorubicin hydrochloride aqueous solution, used in the therapy. After 3 hours, only 9.2 % of the drug was released in the absence of ESW with 40.3 % of free doxorubicin, also no initial burst effect was observed showing NBs stability. Interestingly, doxorubicin was released to a greater extent following ESW treatment with 500 pulses at 0.59 mJ/mm2. Notably, after 3 hours, ESWs generated an increase of about 70 % of free doxorubicin nanobubbles without ESW treatment. Empty Nanobubbles affected cell viability both CAL-62 and 8305C cell lines and Glycol Chitosan Nanobubbles shells had no significant effect on cell viability. Cytotoxic effect of doxorubicin in both CAL-62 and 8305C cell lines only at levels hardly obtainable in patient receiving the doxorubicin dose of 60 mg/m2 with a GI50 at 48 hours of 1.57 µM in CAL-62 and 3.93 µM in 8305C cells, ESW treatment increased the cytotoxic effect of the free drug with decrease in GI50 in both cell lines.

The combination of ESWs with doxorubicin-loaded nanobubbles had the highest cytotoxic effect. The cytotoxicity of CAL-62 treated with negative nanobubbles in combination with free doxorubicin and ESWs treatment was considered as more cytotoxic when compared to the free drug. However, combining ESWs with free doxorubicin plus empty nanobubbles was less effective than using ESWs with doxorubicin loaded into Nanobubbles. Empty nanobubbles were found to be safe in rat cardiomyocytes H9C2 cells as they entered the cells at a level comparable to that observed in cancer cells. The drug-loaded nanobubbles enhance the overall cytotoxic effect in comparison to the free drug. Mechanistically, ESWs triggered the intracellular release of drugs from NBs out-turned with the highest nuclear drug content.

Prednisolone phosphate and a Gd (III) complex

In this case, prednisolone is being loaded in chitosan nanobubble using perfluoropentane as an inert gas. This stable theranostic chitosan-based nanobubble contains the MR imaging gadolinium diethylenetriaminepentaacetic acid (Gd-DOTP) along with the drug (Prednisolone phosphate) companions have helped to reduce adverse effects of anticancer drugs and achieve prolonged drug release. The earlier research studies suggested that the lipid nanobubbles for US imaging detection were an application for the in vivo distinct enhanced imaging of tumor type tissues in essence of drug delivery field (Yin et al., 2012Yin T, Wang P, Zheng R, Zheng B, Cheng D, Zhang X, et al. Nanobubbles for enhanced ultrasound imaging of tumors. Int J Nanomed . 2012;7:895. ; Cavalli et al., 2015Cavalli R, Argenziano M, Vigna E, Giustetto P, Torres E, Aime S, et al. Preparation and in vitro characterization of chitosan nanobubbles as theranostic agents. Colloids Surf B. 2015;129:39-46. ). The MRI detectable MB’s are having application for theranostic purposes has already been proposed. Gd-DTPA loaded microbubbles of PLGA were fabricated as contrast agents for several modes both US and MRI (Ao et al., 2010Ao M, Wang Z, Ran H, Guo D, Yu J, Li A, et al. Gd-DTPA-loaded PLGA microbubbles as both ultrasound contrast agent and MRI contrast agent-A feasibility research. J Biomed Mater Res Part B. 2010;93(2):551-556. ). Albumin-based microbubbles charged with decafluorobutane gas and accommodating Gd-DTPA linked covalently with Human Serum Albumin (HSA) as a model protein drug was employed to enhance the diagnosis of aggravated sites within the vascular vessels (Anderson et al., 2012Anderson DR, Duryee MJ, Garvin RP, Boska MD, Thiele GM, Klassen LW. A method for the making and utility of gadolinium-labeled albumin microbubbles. Magn Reson Imaging. 2012;30(1):96-103. ).

The preparation of theranostic nanobubbles is depicted in Figure 3. In order of tuning the sizes of formulation, two distinct co-surfactants was tested- tetradecylphosphoric acid (C14) and palmitic acid (C16). Prednisolone phosphate (PLP) and Gd-DOTP do possess distinct physicochemical characteristics and may have futuristic application in various areas of the nanobubble system. Being an amphiphilic molecule, PLP is detected mostly at the interface with the perfluoropentane core and the negative charged Gd-DOTP complex is bound electrostatically to the cationic surface of the nanobubble. Chitosan was a preferred as an essential component of the shell of nanobubble due to its better biocompatibility and the polycationic character that makes it as a unique compound to generate stable supramolecular ion pairs along with anions (Dash et al., 2011Dash M, Chiellini F, Ottenbrite R, Chiellini E. Chitosan-A versatile semi-synthetic polymer in biomedical applications. Prog Polym Sci. 2011;36(8):981-1014. ). Among two non-toxic co-bound slightly stronger to palmitic acid-based nanobubbles. All the preparations showed polydispersity index values ranging from 0.12 to 0.27, with a narrow dispersion observed in C16 containing nanobubbles.

Interestingly, C16 preparations showed a slightly smaller size (around 10%) than the C14 containing system, regardless of the presence of Gd-DOTP. Further additional experimental support to determine the effective binding of Gd-DOTP to the chitosan NBs were obtained by determining the zeta-potential. Instead, the presence of the anionic Gd-complex causes in overall neutralisation of partial charge of the bubbles, with a minimization of the zeta potential of ca. 30%; as the Gd-DOTP complex is being neutralized with help of specific positives charges of the NBs, and thus the residual charges responsible for the established stability possessed by the nanobubbles.

Based on the enhanced imaging performance of MRI/US, smaller size, and improved polydispersity index, palmitic acid-based chitosan nanobubbles was the formulation of choice for loading PLP. Pluronic F68 is a non-ionic copolymer type amphiphile based on ethylene and propylene oxide units extensively employed in pharmaceuticals. The presence of Pluronic F68 in the formulation is to stop the NBs aggregation through steric stabilization that has been dominated by the effect of solvation. The chitosan-based nanobubble formulations disclosed good physical stability with incubation with the plasma at 37ºC for 3 hours, without deducing morphology or size changes and aggregation phenomena. This behavior is in proportionality with the presence of perfluoropentane core which would be still in a liquid state. This stabilization is a response of the reduced size of the nanodroplets and the existence of a phospholipid monolayer at the interface of the system.

The insonation parameters are not having any role in disrupting the nanobubble structure, as disclosed by maintaining low and controlled release kinetics. Preferably, the physical stability of the theranostic NBs could be visualized even after 5 minutes of insonation. The existence of lipid monolayer would be the significant factor for maintaining the structural preservation of the overall system after insonation.

Vancomycin

Vancomycin was loaded in dextran sulfate shell and perfluoropentane cored nanobubbles. This nanobubble was aimed at local delivery with the potential treatment of skin infections, with prolonged release at the site of the target region and had better stability. Previous studies reported the vancomycin with poor absorption via gastrointestinal tract along with a poor oral bioavailability which was often recommended with low intravenous infusion as another feasibility for drug administration, even though vancomycin instability in aqueous solutions at 37°C was a significant obstacle in rapid suppression in drug efficacy (Mawhinney et al., 1992Mawhinney W, Adair C, Gorman S, McClurg B. Stability of vancomycin hydrochloride in peritoneal dialysis solution. Am J Health Syst Pharm. 1992;49(1):137-139. ; Raverdy et al., 2013Raverdy V, Ampe E, Hecq JD, Tulkens PM. Stability and compatibility of vancomycin for administration by continuous infusion. J Antimicrob Chemother. 2013;68(5):1179-1182. ; Argenziano et al., 2017Argenziano M, Banche G, Luganini A, Finesso N, Allizond V, Gulino GR, et al. Vancomycin-loaded nanobubbles: A new platform for controlled antibiotic delivery against methicillin-resistant Staphylococcus aureus infections. Int J Pharm. 2017;523(1):176-188.). Following parenteral administration displays a slow mode of action, a complex concentration-time profile, and a low penetration of tissues (Vandecasteele, De Vriese, Tacconelli, 2012Vandecasteele S, De Vriese A, Tacconelli E. The pharmacokinetics and pharmacodynamics of vancomycin in clinical practice: evidence and uncertainties. J Antimicrob Chemother . 2012;68(4):743-748. ). Moreover, systemic Vancomycin (Vm) administration might be the reason for several adverse effects (Vidal, González, Fuente, 1992Vidal C, González AQ, Fuente R. Toxic epidermal necrolysis due to vancomycin. Annals Allergy. 1992;68(4):345-347. ). Vm topical application that would be much safer than the systemic administration is currently in the limit by several factors such as skin barrier properties and poor drug permeability (Giandalia et al., 2001Giandalia G, De Caro V, Cordone L, Giannola LI. Trehalose-hydroxyethylcellulose microspheres containing vancomycin for topical drug delivery. Eur J Pharm Biopharm. 2001;52(1):83-89.).

The vancomycin nanobubble preparation is as depicted in Figure 3. The Empty fluorescent NBs and drug-loaded nanobubbles have fabricated by the addition of 6-coumarin to the perfluoropentane core. Alternatively, fluorescent vancomycin formulated in a chemical reaction between fluorescein isothiocyanate (FITC) and vancomycin. All formulation bubbles sizes indicate around 300 nm as a value of average diameters rather polydispersity index is included in between 0.22 to 0.25 with zeta potentials ranged between -34.5 mV (Empty Nanobubble) to -29 mV (Vancomycin loaded nanobubbles). The loading of vancomycin in the Nanobubble structure does not affect the viscosity of the formulations. The Nanobubbles has loaded with vancomycin and encapsulation efficiency determined was 86% along with loading capacity of 29%. The stability of the empty nanobubbles and drug-loaded nanobubbles formulation has determined on a long-term basis with no significant variation up to six months. Vancomycin loaded nanobubbles also confirmed with significant toxic effect and viability of HaCaT cell lines was not affected by any formulation significantly. The Vancomycin loaded nanobubbles displayed slow and steady prolonged release drug kinetics when compared with vancomycin based aqueous solution, with only 16% of drug release from VmLNBs after 6 hours. The drug-loaded nanobubbles showed more effectivity and enhanced efficacy than vancomycin alone in killing MRSA (methicillin-resistant Staphylococcus aureus), with vancomycin loaded nanobubble the antibacterial activity is effectively sustained overtimes as a result of prolonged drug release.

Taxanes loaded on Glycol-chitosan Nanobubbles

Taxanes (Paclitaxel and Docetaxel) were loaded in perfluoropentane inner core and glycol chitosan shell based nanobubbles was aimed for prolonged-release and to reduce side effect. Previous studies reported the paclitaxel and docetaxel is the potentially treat for castration-resistant prostate cancer (CRPC), but unfortunately this shows little effect on prolonging survival (Darshan et al., 2011Darshan MS, Loftus MS, Thadani-Mulero M, Levy BP, Escuin D, Zhou XK, et al. Taxane-induced blockade to nuclear accumulation of the androgen receptor predicts clinical responses in metastatic prostate cancer. Cancer Res. 2011;71(18):6019-6029. ; Marano et al., 2016Marano F, Argenziano M, Frairia R, Adamini A, Bosco O, Rinella L, et al. Doxorubicin-loaded nanobubbles combined with extracorporeal shock waves: basis for a new drug delivery tool in anaplastic thyroid cancer. Thyroid. 2016a;26(5):705-716. ; Shiota, Eto, 2016Shiota M, Eto M. Current status of primary pharmacotherapy and future perspectives toward upfront therapy for metastatic hormone‐sensitive prostate cancer. Int J Urol. 2016;23(5):360-369. ) and is complicated with several side effects (Schutz, Buzaid, Sartor, 2014Schutz FA, Buzaid AC, Sartor O. Taxanes in the management of metastatic castration-resistant prostate cancer: efficacy and management of toxicity. Crit Rev Oncol Hematol. 2014;91(3):248-256. ). These nanobubbles fabricated in the treatment of CRPC. In this method, extracorporeal shock waves (ESWs) are being used for delivery of the drug in the target region. Taxanes nanobubbles preparation as shown in Figure 3. The Fluorescent nanobubbles without any drug have obtained by the addition of green fluorescent 6-coumarin to the perfluoropentane core.

All formulation nanobubbles present an average diameter of about 300 nm along with a positive surface charge. The drug loading slightly affected the zeta potential values. The Glycol chitosan nanobubbles were able to incorporate the two taxanes in the core to a higher extent, paclitaxel were loaded into the nanobubble system with approximately 94% encapsulation efficiency and the loading capacity of about 5.8% approximately and the docetaxel was also loaded into the nanobubble system with an encapsulated efficiency of about 86% and the loading capacity of about 5.2% respectively. The release profiles of both drug loaded nanobubbles found prolonged duration in vitro release kinetics of the two taxanes from the drug-loaded nanobubbles has previously demonstrated and no initial burst effect has determined for both the drug formulations.

The paclitaxel results showed that about 15% and 22% of docetaxel has released from the nanobubbles within the first 8 hours. The stability of drug loaded nanobubbles formulation has confirmed by long-term and there was no significant variation up to three months after the fabrication of these formulations. The combination of ESWs and NBs enhances paclitaxel cytotoxicity, thus decreasing the drug GI50 of 52% in PC3 cells and 61% in DU145 cells, respectively. The similar effect was obtained in the docetaxel activity; that is, the drug GI50 is 48% in PC3 cells and 40% in DU145 cells. The Nanobubbles also works as a stable taxane reservoir in the CRPC cells. The ESWs increases the release of drug from nanobubbles, leading to a higher cytotoxicity and anti-migration effect.

Polymer-based nanobubbles

Doxorubicin-loaded on PLGA

Doxorubicin was prepared using Perfluoropropane gas and PLGA, such nanobubbles are aimed for the controlled release of certain drugs at the target site and had better stability. These nanobubble increase ultrasound imaging of the tumor and improve its therapeutic effect. Nanobubble was formulated using a double emulsion (W/O/W) technique. The doxorubicin nanobubble formulation sketch is as shown in Figure 4.

A doxorubicin nanobubble increases the concentration of the drug in targeted tissues or tumors to achieve the tumorolytic effect. The perfluoropropane gas enhances the accuracy of ultrasound imaging and hence drug release. The average size of nanoparticles was 500 nm with the potential of -23 mV exhibit better stability. The encapsulation efficiency of the Doxorubicin nanobubbles was 78.6 % while the efficiency of drug loading was 7.4 %. Different researches on the drug release pattern from nanobubbles with or without ultrasonic effect concluded that the ultrasound is necessary for the drug release and also contribute to the controlled release effect for Doxorubicin nanobubbles. In vivo studies of the ultrasound imaging system on the VX2 liver cancer cells, in comparison to saline, Doxorubicin nanobubbles could augment the ultrasonic imaging remarkably at the tumor site.

After administration of the Doxorubicin nanobubble drug in the ear vein, the nanobubbles could quickly attain the tumor area and reach the peak after 17 seconds. The result confirmed that the Doxorubicin nanobubbles are used to strengthen the ultrasound imaging and release of drug in the targeted tissue. The drug loaded into PLGA helps to minimize its toxic effect on the body remarkably, thus improving the therapeutic efficacy of a tumor at a greater extent (Meng et al., 2016Meng M, Gao J, Wu C, Zhou X, Zang X, Lin X, et al. Doxorubicin nanobubble for combining ultrasonography and targeted chemotherapy of rabbit with VX2 liver tumor. Tumor Biol. 2016;37(7):8673-8680. ).

Air-based nanobubbles for drug delivery

Doxorubicin-loaded on Poly (lactic-co-glycolic acid) Nanobubbles

The Doxorubicin was prepared using Poly (lactic-co-glycolic acid) and such nanobubble is under investigation of the sustained type of drug delivery. These nanobubbles help in improving specific targeting, increased delivery to the site of interest and decreases the incidence and intensity of the side effects (Betancourt, Brown, Brannon-Peppas, 2007Betancourt T, Brown B, Brannon-Peppas L. Doxorubicin-loaded PLGA nanoparticles by nanoprecipitation: preparation, characterization and in vitro evaluation. Nanomedicine. 2007;2(2):219-32.; Tang et al., 2007Tang N, Du G, Wang N, Liu C, Hang H, Liang W. Improving penetration in tumors with nanoassemblies of phospholipids and doxorubicin. J Natl Cancer Inst. 2007;99(13):1004-1015.; Wang et al., 2011Wang H, Zhao Y, Wu Y, Hu Yl, Nan K, Nie G, et al. Enhanced anti-tumor efficacy by co-delivery of doxorubicin and paclitaxel with amphiphilic methoxy PEG-PLGA copolymer nanoparticles. Biomaterials. 2011;32(32):8281-8290. ; Deng et al., 2014Deng L, Li L, Yang H, Li L, Zhao F, Wu C, et al. Development and optimization of doxorubicin loaded poly (lactic-co-glycolic acid) nanobubbles for drug delivery into HeLa cells. J Nanosci Nanotechnol. 2014;14(4):2947-2954. ). The doxorubicin loaded into PLGA nanoparticles is a feasible approach to decrease chemotherapy cytotoxicity to healthy tissues, which help to control undesirable adverse effects viz. impaired cardiac function (Lin, Ng, Wang, 2005Lin R, Ng LS, Wang CH. In vitro study of anticancer drug doxorubicin in PLGA-based microparticles. Biomaterials. 2005;26(21):4476-4485. ; Jain et al., 2011Jain AK, Swarnakar NK, Das M, Godugu C, Singh RP, Rao PR, et al. Augmented anticancer efficacy of doxorubicin-loaded polymeric nanoparticles after oral administration in a breast cancer induced animal model. Mol Pharm. 2011;8(4):1140-1151. ). The drug routinely loaded employing double emulsion technique the preparation is as illustrated in Figure 4 without loading the gas used for this method.

The Doxorubicin free PLGA Nanobubbles do possess a negatively charged surface and smaller diameter ranges between 531 nm and 258 nm by controlling the ultrasound amplitude from 15% to 45%. The low polydispersity of Doxorubicin free PLGA Nanobubbles suggests a narrowed distribution of size. The doxorubicin free PLGA Nanobubbles are round, spheroid in shape with a smoother surface and a form of a dispersion.

The 45% ultrasound amplitude was selected to fabricate doxorubicin loaded with PLGA Nanobubbles. The doxorubicin concentration varies from 0 to 65 mg DOX/g PLGA, and other parameters for instance Poly (vinyl alcohol) concentration, sonication duration and ultrasound amplitude are kept constant. The diameter ranges from 258 nm to 317 nm. The initial DOX concentration has minimal effect on the particle size, surface charge and polydispersity. The drug payload and encapsulated efficiency are almost increased with doxorubicin concentration. There was a relative saturation when doxorubicin concentration was beyond 52 mg DOX/g PLGA. In actual the drug content in the PLGA NBs was interfered by drug-polymer interactions and the drug miscibility in the polymer.

The maximum drug-polymer miscibility would result in the higher drug incorporation (Panyam et al., 2004Panyam J, Williams D, Dash A, Leslie‐Pelecky D, Labhasetwar V. Solid‐state solubility influences encapsulation and release of hydrophobic drugs from PLGA/PLA nanoparticles. J Pharm Sci . 2004;93(7):1804-1814. ; Manchanda et al., 2010Manchanda R, Fernandez-Fernandez A, Nagesetti A, McGoron AJ. Preparation and characterization of a polymeric (PLGA) nanoparticulate drug delivery system with simultaneous incorporation of chemotherapeutic and thermo-optical agents. Colloids Surf B . 2010;75(1):260-267. ). The drug payload and encapsulation efficiencies are 3.6% and 70.9% respectively. The drug-loaded PLGA NBs are uniform and globular sphere with a smooth surfaced and in dispersion with water. The mean diameter is approximately 270 nm with narrow size distribution. The Stability studies were performed by re-dissolving the drug-loaded PLGA NBs in PBS at 37ºC for one month to assess their size change. The doxorubicin loaded nanobubbles were devoid of any remarkable changes in the average diameter within one month.

The advantage of this drug delivery system with the nanobubbles is the controlled drug release, which improves the drug bioavailability and reduces the side effects and toxicity of drugs in the normal tissues by the EPR (enhanced permeation and retention) effect (Santander-Ortega et al., 2010Santander-Ortega MJ, Csaba N, González L, Bastos-González D, Ortega-Vinuesa JL, Alonso MJ. Protein-loaded PLGA-PEO blend nanoparticles: encapsulation, release and degradation characteristics. Colloid Polym Sci. 2010;288(2):141-150. ; Jia et al., 2012Jia Y, Yuan M, Yuan H, Huang X, Sui X, Cui X, et al. Co-encapsulation of magnetic Fe3O4 nanoparticles and doxorubicin into biodegradable PLGA nanocarriers for intratumoral drug delivery. Int J Nanomed . 2012;7:1697. ). The cell viability has shown higher cytotoxicity to kill HeLa cells when compared to the free DOX at the same DOX concentration.

Sulfur hexafluoride based nanobubbles for drug delivery

Coumarin-6 loaded on lipid-based nanobubble

Coumarin-6 model drug was loaded into the lipid-based nanobubble. The nanobubbles are under investigation of ultrasound imaging function and cellular delivery property. The contrast agent nanobubble preparation as depicted in Figure 2. In this method, coumarin-6 was selected as a model drug for cellular testing. The Emulsion, liposomes and chitosan nanoparticle were also made ready for the cell test. These can refer as the control agents for nanobubbles. The Emulsion is formulated using the technique of preparation of bubbles without introducing the gas. The fabrication of liposomes extensively employs "film" technique (Sezer, Baş, Akbuğa, 2004Sezer AD, Baş AL, Akbuğa J. Encapsulation of enrofloxacin in liposomes I: preparation and in vitro characterization of LUV. J Liposome Res. 2004;14(1-2):77-86.; Wang et al., 2010Wang Y, Li X, Zhou Y, Huang P, Xu Y. Preparation of nanobubbles for ultrasound imaging and intracelluar drug delivery. Int J Pharm . 2010;384(1-2):148-153. ). The Chitosan nanoparticle (CNP) was developed through a crosslinking mechanism. In all the preparation, the overall concentration of coumarin-6 and lipid were 20 µg/ml and 3 mg/ml maintained respectively.

The nanobubbles are bright spots present in the focus region and the range of size of bubble used to be between 333 to 246 nm. The high ratio of tween 80 ensures improvement in the acoustic backscatter feature of bubbles. The nanobubbles multiplied from 3.002 to 16.110 as tween 80 changed from 0% to 3% while an excessive quantity of cholesterol blamed for negative contribution to the acoustic bubble property. When the lipid was around 2.5mg/ml, the bubbles represent the ultimate acoustic backscatter profile among the formulations tested. On comparison of overall pharmacokinetic task part within chitosan nanoparticles, emulsion and liposome, the nanobubbles scored with the highest final concentration (plateau concentration) 29.780 ng/ml while the most substantial value of t 1/2 found was 35.4 min.

CONCLUSION

Nanobubbles are novel systems of drug delivery specifically designed for targeting the drug to affected sites upon exposing to ultrasound/ESWs. Nanobubbles are gas composed hollow, spherical pouches or cavities in aqueous solution with size range below 1µm. Bubbles are sphere-shaped particles with a shell and core structure filled with gas to give the acoustically active properties. Nanobubbles help in reducing the cumulative frequency of dose and targets specifically at to site of interest. Nanobubbles formulation for drugs with gas entrapped in different core shields helps for sustained delivery, reduced frequency of administration and enhances bioavailability and efficacy in comparison with conventional dosage forms. The nature of gas like perfluorocarbon contributes for enhancing the stability of bubbles to augment release kinetics of the drug against air bubbles which gets dissolved quickly in the bloodstream before achieving actual therapeutic window.

ACKNOWLEDGEMENT

The authors are grateful to the Department of Pharmacy, Annamalai University, Chidambaram, Tamilnadu for their valuable support for this paper.

REFERENCES

  • Anderson DR, Duryee MJ, Garvin RP, Boska MD, Thiele GM, Klassen LW. A method for the making and utility of gadolinium-labeled albumin microbubbles. Magn Reson Imaging. 2012;30(1):96-103.
  • Ao M, Wang Z, Ran H, Guo D, Yu J, Li A, et al. Gd-DTPA-loaded PLGA microbubbles as both ultrasound contrast agent and MRI contrast agent-A feasibility research. J Biomed Mater Res Part B. 2010;93(2):551-556.
  • Argenziano M, Banche G, Luganini A, Finesso N, Allizond V, Gulino GR, et al. Vancomycin-loaded nanobubbles: A new platform for controlled antibiotic delivery against methicillin-resistant Staphylococcus aureus infections. Int J Pharm. 2017;523(1):176-188.
  • Ayodele AT, Valizadeh A, Adabi M, Esnaashari SS, Madani F, Khosravani M, et al. Ultrasound nanobubbles and their applications as theranostic agents in cancer therapy: A review. Biointerface Res Appl Chem. 2017;7(6):2253-2262.
  • Betancourt T, Brown B, Brannon-Peppas L. Doxorubicin-loaded PLGA nanoparticles by nanoprecipitation: preparation, characterization and in vitro evaluation. Nanomedicine. 2007;2(2):219-32.
  • Bhandari P, Novikova G, Goergen CJ, Irudayaraj J. Ultrasound beam steering of oxygen nanobubbles for enhanced bladder cancer therapy. Sci Rep. 2018;8(1):3112.
  • Bhandari PN, Cui Y, Elzey BD, Goergen CJ, Long CM, Irudayaraj J. Oxygen nanobubbles revert hypoxia by methylation programming. Sci Rep . 2017;7(1):9268.
  • Bisazza A, Civra A, Donalisio M, Lembo D, Cavalli R. The in vitro characterization of dextran-based nanobubbles as possible DNA transfection agents. Soft Matter. 2011;7(22):10590-10593.
  • Bosca F, Bielecki PA, Exner AA, Barge A. Porphyrin-Loaded Pluronic Nanobubbles: A New US-Activated Agent for Future Theranostic Applications. Bioconjug Chem. 2018;29(2):234-240.
  • Cai X, Jiang Y, Lin M, Zhang J, Guo H, Yang F, et al. Ultrasound-Responsive Materials for Drug/Gene Delivery. Front Pharmacol. 2020;10:1650.
  • Carvalho C, Santos RX, Cardoso S, Correia S, Oliveira PJ, Santos MS, et al. Doxorubicin: the good, the bad and the ugly effect. Curr Med Chem. 2009;16(25):3267-3285.
  • Cavalli R, Argenziano M, Vigna E, Giustetto P, Torres E, Aime S, et al. Preparation and in vitro characterization of chitosan nanobubbles as theranostic agents. Colloids Surf B. 2015;129:39-46.
  • Cavalli R, Bisazza A, Lembo D. Micro-and nanobubbles: a versatile non-viral platform for gene delivery. Int J Pharm . 2013;456(2):437-445.
  • Cavalli R, Bisazza A, Trotta M, Argenziano M, Civra A, Donalisio M, et al. New chitosan nanobubbles for ultrasound-mediated gene delivery: preparation and in vitro characterization. Int J Nanomed. 2012;7:3309.
  • Cavalli R, Soster M, Argenziano M. Nanobubbles: a promising efficienft tool for therapeutic delivery. Ther Deliv. 2016;7(2):117-138.
  • Chatterjee K, Zhang J, Honbo N, Karliner JS. Doxorubicin cardiomyopathy. Cardiology. 2010;115(2):155-162.
  • Constantinides PP, Lambert KJ, Tustian AK, Schneider B, Lalji S, Ma W, et al. Formulation development and antitumor activity of a filter-sterilizable emulsion of paclitaxel. Pharm Res. 2000;17(2):175-182.
  • Darshan MS, Loftus MS, Thadani-Mulero M, Levy BP, Escuin D, Zhou XK, et al. Taxane-induced blockade to nuclear accumulation of the androgen receptor predicts clinical responses in metastatic prostate cancer. Cancer Res. 2011;71(18):6019-6029.
  • Dash M, Chiellini F, Ottenbrite R, Chiellini E. Chitosan-A versatile semi-synthetic polymer in biomedical applications. Prog Polym Sci. 2011;36(8):981-1014.
  • Delalande A, Postema M, Mignet N, Midoux P, Pichon C. Ultrasound and microbubble-assisted gene delivery: recent advances and ongoing challenges. Ther Deliv . 2012;3(10):1199-1215.
  • Deng L, Li L, Yang H, Li L, Zhao F, Wu C, et al. Development and optimization of doxorubicin loaded poly (lactic-co-glycolic acid) nanobubbles for drug delivery into HeLa cells. J Nanosci Nanotechnol. 2014;14(4):2947-2954.
  • Dijkmans P, Juffermans L, Musters R, van Wamel A, Ten Cate F, van Gilst W, et al. Microbubbles and ultrasound: from diagnosis to therapy. Eur J Echocardiogr. 2004;5(4):245-246.
  • Dimcevski G, Kotopoulis S, Bjånes T, Hoem D, Schjøtt J, Gjertsen BT, et al. A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer. J Control Release. 2016;243:172-181.
  • Ding S, Patel N, Zhang H. Ciclosporin A as a Reversal Agent Against Concurrent Multidrug Resistance in Tumors with Nanobubbles. J Biomed Nanotechnol. 2018;14(1):190-197.
  • Duan L, Yang L, Jin J, Yang F, Liu D, Hu K, et al. Micro/nano-bubble-assisted ultrasound to enhance the EPR effect and potential theranostic applications. Theranostics. 2020;10(2):462-483.
  • Fan Z, Kumon RE, Deng CX. Mechanisms of microbubble-facilitated sonoporation for drug and gene delivery. Ther Deliv . 2014;5(4):467-486.
  • Fang JY, Lee WR, Shen SC, Huang YL. Effect of liposome encapsulation of tea catechins on their accumulation in basal cell carcinomas. J Dermatol Sci. 2006;42(2):101-109.
  • Ferrara KW. Driving delivery vehicles with ultrasound. Adv Drug Deliv Rev. 2008;60(10):1097-1102.
  • Frinking PJ, Bouakaz A, Kirkhorn J, Ten Cate FJ, De Jong N. Ultrasound contrast imaging: current and new potential methods. Ultrasound Med Biol. 2000;26(6):965-975.
  • Furusawa Y, Fujiwara Y, Hassan MA, Tabuchi Y, Morita A, Enomoto A, et al. Inhibition of DNA-dependent protein kinase promotes ultrasound-induced cell death including apoptosis in human leukemia cells. Cancer Lett. 2012;322(1):107-112.
  • Gao Z, Kennedy AM, Christensen DA, Rapoport NY. Drug-loaded nano/microbubbles for combining ultrasonography and targeted chemotherapy. Ultrasonics. 2008;48(4):260-270.
  • Giandalia G, De Caro V, Cordone L, Giannola LI. Trehalose-hydroxyethylcellulose microspheres containing vancomycin for topical drug delivery. Eur J Pharm Biopharm. 2001;52(1):83-89.
  • Hamano N, Kamoshida S, Kikkawa Y, Yano Y, Kobayashi T, Endo-Takahashi Y, et al. Development of antibody-modified nanobubbles using Fc-region-binding polypeptides for ultrasound imaging. Pharmaceutics. 2019;11(6):283.
  • He X, Wu DF, Ji J, Ling WP, Chen Xl, Chen YX. Ultrasound microbubble-carried PNA targeting to c-myc mRNA inhibits the proliferation of rabbit iliac arterious smooth muscle cells and intimal hyperplasia. Drug Delivery. 2016;23(7):2482-2487.
  • Houthaeve G, Xiong R, Robijns J, Luyckx B, Beulque Y, Brans T, et al. Targeted Perturbation of Nuclear Envelope Integrity with Vapor Nanobubble-Mediated Photoporation. ACS Nano. 2018;12(8):7791-7802.
  • Husseini GA, Pitt WG. The use of ultrasound and micelles in cancer treatment. J Nanosci Nanotechnol . 2008;8(5):2205-2215.
  • Husseini GA, Runyan CM, Pitt WG. Investigating the mechanism of acoustically activated uptake of drugs from Pluronic micelles. BMC Cancer. 2002;2(1):20.
  • Hwang TL, Lin YK, Chi CH, Huang TH, Fang JY. Development and evaluation of perfluorocarbon nanobubbles for apomorphine delivery. J Pharm Sci. 2009;98(10):3735-3747.
  • Hynynen K, Pomeroy O, Smith DN, Huber PE, McDannold NJ, Kettenbach J, et al. MR imaging-guided focused ultrasound surgery of fibroadenomas in the breast: a feasibility study. Radiology. 2001;219(1):176-185.
  • Ingram WM, Priston MJ, Sewell GJ. Improved assay for R (−)-apomorphine with application to clinical pharmacokinetic studies in Parkinson's disease. J Chromatogr B. 2006;831(1-2):1-7.
  • Iverson N, Plourde N, Chnari E, Nackman GB, Moghe PV. Convergence of nanotechnology and cardiovascular medicine. BioDrugs. 2008;22(1):1-10.
  • Jain AK, Swarnakar NK, Das M, Godugu C, Singh RP, Rao PR, et al. Augmented anticancer efficacy of doxorubicin-loaded polymeric nanoparticles after oral administration in a breast cancer induced animal model. Mol Pharm. 2011;8(4):1140-1151.
  • Jia Y, Yuan M, Yuan H, Huang X, Sui X, Cui X, et al. Co-encapsulation of magnetic Fe3O4 nanoparticles and doxorubicin into biodegradable PLGA nanocarriers for intratumoral drug delivery. Int J Nanomed . 2012;7:1697.
  • Jiang Q, Hao S, Xiao X, Yao J, Ou B, Zhao Z, et al. Production and characterization of a novel long-acting Herceptin-targeted nanobubble contrast agent specific for Her-2-positive breast cancers. Breast Cancer. 2016;23(3):445-455.
  • Jocham D, Chaussy C, Schmiedt E. Extracorporeal shock wave lithotripsy. Urol Int. 1986;41(5):357-368.
  • Kabalnov A, Klein D, Pelura T, Schutt E, Weers J. Dissolution of multicomponent microbubbles in the bloodstream: 1. Theory. Ultrasound Med Biol. 1998;24(5):739-749.
  • Kelkar SS, Reineke TM. Theranostics: combining imaging and therapy. Bioconjug Chem . 2011;22(10):1879-1903.
  • Khan MS, Hwang J, Lee K, Choi Y, Kim K, Koo HJ, et al. Oxygen-carrying micro/nanobubbles: Composition, synthesis techniques and potential prospects in photo-triggered theranostics. Molecules. 2018a;23(9):2210.
  • Khan MS, Hwang J, Seo Y, Shin K, Lee K, Park C, et al. Engineering oxygen nanobubbles for the effective reversal of hypoxia. Artif Cells Nanomed Biotechnol. 2018b;46(sup3):S318-S327.
  • Kou P, Zhang Y, Shao W, Zhu H, Zhang J, Wang H, et al. Significant efficacy and well safety of apatinib in an advanced liver cancer patient: a case report and literature review. Oncotarget. 2017;8(12):20510.
  • Kripfgans OD, Fabiilli ML, Carson PL, Fowlkes JB. On the acoustic vaporization of micrometer-sized droplets. J Acoust Soc Am. 2004;116(1):272-281.
  • Lammers T, Aime S, Hennink WE, Storm G, Kiessling F. Theranostic nanomedicine. Acc Chem Res. 2011;44(10):1029-1038.
  • Li GL, Danhof M, Bouwstra JA. Iontophoretic delivery of apomorphine in vitro: physicochemic considerations. Pharm Res . 2001;18(11):1509-1513.
  • Lin R, Ng LS, Wang CH. In vitro study of anticancer drug doxorubicin in PLGA-based microparticles. Biomaterials. 2005;26(21):4476-4485.
  • Liu Y, Miyoshi H, Nakamura M. Encapsulated ultrasound microbubbles: therapeutic application in drug/gene delivery. J Control Release . 2006;114(1):89-99.
  • Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release . 2000;65(1-2):271-284.
  • Manchanda R, Fernandez-Fernandez A, Nagesetti A, McGoron AJ. Preparation and characterization of a polymeric (PLGA) nanoparticulate drug delivery system with simultaneous incorporation of chemotherapeutic and thermo-optical agents. Colloids Surf B . 2010;75(1):260-267.
  • Marano F, Argenziano M, Frairia R, Adamini A, Bosco O, Rinella L, et al. Doxorubicin-loaded nanobubbles combined with extracorporeal shock waves: basis for a new drug delivery tool in anaplastic thyroid cancer. Thyroid. 2016a;26(5):705-716.
  • Marano F, Frairia R, Rinella L, Argenziano M, Bussolati B, Grange C, et al. Combining doxorubicin-nanobubbles and shockwaves for anaplastic thyroid cancer treatment: preclinical study in a xenograft mouse model. Endocr Relat Cancer. 2017;24(6):275-286.
  • Marano F, Rinella L, Argenziano M, Cavalli R, Sassi F, D’Amelio P, et al. Targeting Taxanes to Castration-Resistant Prostate Cancer Cells by Nanobubbles and Extracorporeal Shock Waves. PloS ONE. 2016b;11(12):e0168553.
  • Martin KH, Dayton PA. Current status and prospects for microbubbles in ultrasound theranostics. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2013;5(4):329-345.
  • Marxer EEJ, Brüßler J, Becker A, Schümmelfeder J, Schubert R, Nimsky C, et al. Development and characterization of new nanoscaled ultrasound active lipid dispersions as contrast agents. Eur J Pharm Biopharm . 2011;77(3):430-437.
  • Matsuki N, Ishikawa T, Ichiba S, Shiba N, Ujike Y, Yamaguchi T. Oxygen supersaturated fluid using fine micro/nanobubbles. Int J Nanomed . 2014;9:4495.
  • Mawhinney W, Adair C, Gorman S, McClurg B. Stability of vancomycin hydrochloride in peritoneal dialysis solution. Am J Health Syst Pharm. 1992;49(1):137-139.
  • Mehier-Humbert S, Bettinger T, Yan F, Guy RH. Plasma membrane poration induced by ultrasound exposure: implication for drug delivery. J Control Release . 2005;104(1):213-222.
  • Meng M, Gao J, Wu C, Zhou X, Zang X, Lin X, et al. Doxorubicin nanobubble for combining ultrasonography and targeted chemotherapy of rabbit with VX2 liver tumor. Tumor Biol. 2016;37(7):8673-8680.
  • Misra SK, Ghoshal G, Jensen TW, Ray PS, Burdette EC, Pan D. Bi-modal cancer treatment utilizing therapeutic ultrasound and an engineered therapeutic nanobubble. RSC Adv. 2015;5(78):63839-63845.
  • Negishi Y, Endo Y, Fukuyama T, Suzuki R, Takizawa T, Omata D, et al. Delivery of siRNA into the cytoplasm by liposomal bubbles and ultrasound. J Control Release . 2008;132(2):124-130.
  • Nomikou N, McHale AP. Microbubble-enhanced ultrasound-mediated gene transfer-towards the development of targeted gene therapy for cancer. Int J Hyperthermia. 2012;28(4):300-310.
  • O’Neill BE, Rapoport N. Phase-shift, stimuli-responsive drug carriers for targeted delivery. Ther Deliv . 2011;2(9):1165-1187.
  • Oeffinger BE, Wheatley MA. Development and characterization of a nano-scale contrast agent. Ultrasonics. 2004;42(1-9):343-347.
  • Panyam J, Williams D, Dash A, Leslie‐Pelecky D, Labhasetwar V. Solid‐state solubility influences encapsulation and release of hydrophobic drugs from PLGA/PLA nanoparticles. J Pharm Sci . 2004;93(7):1804-1814.
  • Perera R, Nittayacharn P, Cooley M, Jung O, Exner AA. Ultrasound contrast agents and delivery systems in cancer detection and therapy. Adv Cancer Res . 2018;139:57-84.
  • Raverdy V, Ampe E, Hecq JD, Tulkens PM. Stability and compatibility of vancomycin for administration by continuous infusion. J Antimicrob Chemother. 2013;68(5):1179-1182.
  • Sanlier SH, Ak G, Yılmaz H, Unal A, Bozkaya UF, Taniyan G, et al. Development Of Ultrasound-Triggered And Magnetic Targeted Nanobubble System For Dual-Drug Delivery . J Pharm Sci . 2018;108(3): DOI:10.1016/j.xphs.2018.10.030.
    » https://doi.org/10.1016/j.xphs.2018.10.030
  • Santander-Ortega MJ, Csaba N, González L, Bastos-González D, Ortega-Vinuesa JL, Alonso MJ. Protein-loaded PLGA-PEO blend nanoparticles: encapsulation, release and degradation characteristics. Colloid Polym Sci. 2010;288(2):141-150.
  • Schutz FA, Buzaid AC, Sartor O. Taxanes in the management of metastatic castration-resistant prostate cancer: efficacy and management of toxicity. Crit Rev Oncol Hematol. 2014;91(3):248-256.
  • Sezer AD, Baş AL, Akbuğa J. Encapsulation of enrofloxacin in liposomes I: preparation and in vitro characterization of LUV. J Liposome Res. 2004;14(1-2):77-86.
  • Shen J, Zhuo N, Xu S, Song Z, Hu Z, Hao J, et al. Resveratrol delivery by ultrasound-mediated nanobubbles targeting nucleus pulposus cells. Nanomedicine . 2018;13(12):1433-1446.
  • Shi D. Integrated multifunctional nanosystems for medical diagnosis and treatment. Adv Funct Mater. 2009;19(21):3356-3373.
  • Shiota M, Eto M. Current status of primary pharmacotherapy and future perspectives toward upfront therapy for metastatic hormone‐sensitive prostate cancer. Int J Urol. 2016;23(5):360-369.
  • Sirsi S, Borden M. Microbubble compositions, properties and biomedical applications. Bubble Sci Eng Technol. 2009;1(1-2):3-17.
  • Smith NB, Lee S, Maione E, Roy RB, McElligott S, Shung KK. Ultrasound-mediated transdermal transport of insulin in vitro through human skin using novel transducer designs. Ultrasound Med Biol . 2003;29(2):311-317.
  • Stride E. Physical principles of microbubbles for ultrasound imaging and therapy. Translational Neurosonology. Karger Publishers. 2015, p. 11-22.
  • Subramony JA. Apomorphine in dopaminergic therapy. Mol Pharm. 2006;3(4):380-385.
  • Suzuki M, Koshiyama K, Shinohara F, Mori S, Ono M, Tomita Y, et al. Nanobubbles enhanced drug susceptibility of cancer cells using ultrasound. International Congress Series. Elsevier, 2005, p. 338-339.
  • Suzuki R, Takizawa T, Negishi Y, Hagisawa K, Tanaka K, Sawamura K, et al. Gene delivery by combination of novel liposomal bubbles with perfluoropropane and ultrasound. J Control Rel. 2007;117(1):130-136.
  • Suzuki R, Takizawa T, Negishi Y, Utoguchi N, Maruyama K. Effective gene delivery with novel liposomal bubbles and ultrasonic destruction technology. Int J Pharm . 2008;354(1-2):49-55.
  • Takeuchi S, Sato T, Kawashima N. Nonlinear response of microbubbles coated with surfactant membrane developed as ultrasound contrast agent-experimental study and numerical calculations. Colloids Surf B . 2002;24(3-4):207-216.
  • Tang N, Du G, Wang N, Liu C, Hang H, Liang W. Improving penetration in tumors with nanoassemblies of phospholipids and doxorubicin. J Natl Cancer Inst. 2007;99(13):1004-1015.
  • Terreno E, Uggeri F, Aime S. Image guided therapy: the advent of theranostic agents. J Control Rel . 2012;161(2):328-337.
  • Tian Y, Liu Z, Zhang L, Zhang J, Han X, Wang Q, et al. Apatinib-loaded lipid nanobubbles combined with ultrasound-targeted nanobubble destruction for synergistic treatment of HepG2 cells in vitro. OncoTargets Ther. 2018;11:4785.
  • Ugwoke MI, Kaufmann G, Verbeke N, Kinget R. Intranasal bioavailability of apomorphine from carboxymethylcellulose-based drug delivery systems. Int J Pharm . 2000;202(1-2):125-131.
  • Unger EC, Porter T, Culp W, Labell R, Matsunaga T, Zutshi R. Therapeutic applications of lipid-coated microbubbles. Adv Drug Deliv Rev . 2004;56(9):1291-1314.
  • Van Liew HD, Raychaudhuri S. Stabilized bubbles in the body: pressure-radius relationships and the limits to stabilization. J Appl Physiol. 1997;82(6):2045-2053.
  • Vandecasteele S, De Vriese A, Tacconelli E. The pharmacokinetics and pharmacodynamics of vancomycin in clinical practice: evidence and uncertainties. J Antimicrob Chemother . 2012;68(4):743-748.
  • Vidal C, González AQ, Fuente R. Toxic epidermal necrolysis due to vancomycin. Annals Allergy. 1992;68(4):345-347.
  • Wang H, Zhao Y, Wu Y, Hu Yl, Nan K, Nie G, et al. Enhanced anti-tumor efficacy by co-delivery of doxorubicin and paclitaxel with amphiphilic methoxy PEG-PLGA copolymer nanoparticles. Biomaterials. 2011;32(32):8281-8290.
  • Wang IL, Hsiao CY, Li YH, Meng FB, Huang CC, Chen YM. Nanobubbles water curcumin extract reduces injury risks on drop jumps in women: a pilot study. Evid Based Complement Alternat Med. 2019;2019:8647587.
  • Wang Y, Li X, Zhou Y, Huang P, Xu Y. Preparation of nanobubbles for ultrasound imaging and intracelluar drug delivery. Int J Pharm . 2010;384(1-2):148-153.
  • Watanabe Y, Horie S, Funaki Y, Kikuchi Y, Yamazaki H, Ishii K, et al. Delivery of Na/I symporter gene into skeletal muscle using nanobubbles and ultrasound: visualization of gene expression by PET. J Nucl Med. 2010;51(6):951-958.
  • Wu J. Temperature rise generated by ultrasound in the presence of contrast agent. Ultrasound Med Biol . 1998;24(2):267-274.
  • Xie X, Lin W, Liu H, Deng J, Chen Y, Liu H, et al. Ultrasound-responsive nanobubbles contained with peptide-camptothecin conjugates for targeted drug delivery. Drug Deliv. 2016;23(8):2756-2764.
  • Yang H, Cai W, Xu L, Lv X, Qiao Y, Li P, et al. Nanobubble-Affibody: Novel ultrasound contrast agents for targeted molecular ultrasound imaging of tumor. Biomaterials. 2015a;37:279-288.
  • Yang H, Deng L, Li T, Shen X, Yan J, Zuo L, et al. Multifunctional PLGA nanobubbles as theranostic agents: combining doxorubicin and P-gp siRNA co-delivery into human breast cancer cells and ultrasound cellular imaging. J Biomed Nanotechnol . 2015b;11(12):2124-2136.
  • Yin T, Wang P, Li J, Wang Y, Zheng B, Zheng R, et al. Tumor-penetrating codelivery of siRNA and paclitaxel with ultrasound-responsive nanobubbles hetero-assembled from polymeric micelles and liposomes. Biomaterials. 2014;35(22):5932-5943.
  • Yin T, Wang P, Li J, Zheng R, Zheng B, Cheng D, et al. Ultrasound-sensitive siRNA-loaded nanobubbles formed by hetero-assembly of polymeric micelles and liposomes and their therapeutic effect in gliomas. Biomaterials. 2013;34(18):4532-4543.
  • Yin T, Wang P, Zheng R, Zheng B, Cheng D, Zhang X, et al. Nanobubbles for enhanced ultrasound imaging of tumors. Int J Nanomed . 2012;7:895.
  • Zelphati O, Uyechi LS, Barron LG, Szoka Jr FC. Effect of serum components on the physico-chemical properties of cationic lipid/oligonucleotide complexes and on their interactions with cells. Biochim Biophys Acta. 1998;1390(2):119-133.
  • Zhang X, Zheng Y, Wang Z, Huang S, Chen Y, Jiang W, et al. Methotrexate-loaded PLGA nanobubbles for ultrasound imaging and Synergistic Targeted therapy of residual tumor during HIFU ablation. Biomaterials. 2014;35(19):5148-5161.
  • Zhao YZ, Du LN, Lu CT, Jin YG, Ge SP. Potential and problems in ultrasound-responsive drug delivery systems. Int J Nanomed . 2013;8:1621.
  • Zhou X, Guo L, Shi D, Duan S, Li J. Biocompatible chitosan nanobubbles for ultrasound-mediated targeted delivery of doxorubicin. Nanoscale Res Lett. 2019;14(1):24.

Publication Dates

  • Publication in this collection
    08 July 2022
  • Date of issue
    2022

History

  • Received
    14 Aug 2019
  • Accepted
    17 Oct 2020
Universidade de São Paulo, Faculdade de Ciências Farmacêuticas Av. Prof. Lineu Prestes, n. 580, 05508-000 S. Paulo/SP Brasil, Tel.: (55 11) 3091-3824 - São Paulo - SP - Brazil
E-mail: bjps@usp.br