Open Access

Nanomedicines for high‑intensity focused ultrasound cancer treatment and theranostics (Review)

  • Authors:
    • Qinyun Zheng
    • Bingxin Xia
    • Xianlong Huang
    • Jie Luo
    • Shigen Zhong
    • Xuelin Li
  • View Affiliations

  • Published online on: March 3, 2023     https://doi.org/10.3892/etm.2023.11869
  • Article Number: 170
  • Copyright: © Zheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

High‑intensity focused ultrasound (HIFU) is a promising and representative non‑invasive therapeutic method for treating cancer with a high degree of efficacy. This non‑invasive method induces tumour cell necrosis by increasing the local temperature and mechanical pressure. However, the clinical application of HIFU is limited given its low penetration depth and the incidence of off‑target side effects. With their promising structural adjustability and targeting ability, nanomedicines have been adopted to improve the ablative efficacy of HIFU in the treatment of cancer. By selectively changing the acoustic environment (tissue structure, density and blood supply) of tumour tissue, these nanomedicines may allow for lower HIFU doses and treatment duration, while additionally achieving a higher degree of efficacy. The use of nanomedicines may also enable cancer theranostics of HIFU, allowing for precise cancer therapeutics. The present review aimed to provide an overview of advances in nanomedicines for HIFU cancer treatment and theranostics, stating their current limitations and future perspectives.

1. Introduction

Cancer is one of the most serious fatal diseases, with limited treatment response and unfavourable prognosis (1). Currently, surgery, radiotherapy, chemotherapy and immunotherapy are the primary methods used in clinical practice for management of cancer and, although they have been regarded as the four pillars of cancer therapy (2), these therapeutic modalities have shortcomings. For example, for surgical resection, the incidence of surgical trauma and complications is high and complete removal of all tumour tissue is not guaranteed (3). Tumour radiation resistance and collateral radiation-induced damage to surrounding healthy tissue limit the clinical application of radiotherapy. For chemotherapy and immunotherapy, although new treatment targets and novel drugs are increasingly studied, challenges, such as the low targeting efficacy and intrinsic toxicity of these treatments, remain to be overcome (4). In addition to these four pillars of cancer therapy, more recent studies have focused on developing and improving non-invasive and more patient-friendly modalities with improved treatment efficacy and a lower incidence of side effects: Among these, high-intensity focused ultrasound (HIFU) is a promising approach (5-7).

The identification of the potential of HIFU for clinical therapy dates to the 1950s when it was demonstrated to be an alternative therapeutic procedure for central nervous system disorder (8,9). When HIFU is absorbed by target tissue such as tumour masses, the temperature of the tissue increases to >55˚C, inducing cell death via local coagulative necrosis (10-12) to thermally ablate the tumour mass. HIFU can also induce the generation of small gas bubbles inside the target tissue; sudden collapse of these bubbles results in an increase in the local pressure up to 2-3 kPa, thus causing severe damage to the surrounding tissues (13,14). It has also been shown that HIFU temporarily disrupts the blood-brain barrier (BBB), which aids in delivery of therapeutics into the central nervous system (15). Currently, HIFU has been proven successful in the treatment of numerous diseases such as Parkinson's disease (16,17), essential tremor (18,19), adenomyosis (20) and solid tumour masses (21,22). However, due to the absorption features of HIFU, the penetration of HIFU to deep tumour tissue is severely limited and not sufficient for tumour ablation (23). While increasing HIFU irradiation dosage is a potential strategy to increase efficacy, the collateral damage caused to the surrounding normal tissue would also increase (24). Furthermore, although HIFU primarily results in a focused ablative effect in the targeted tumour mass, off-target collateral damage occurs, resulting in undesired tissue injury and burns, vasospasm and haemorrhaging, impotence, incontinence, formation of atrial-oesophageal fistula and off-site rib necrosis (25,26). Nanoparticles (NPs) are now being adopted to overcome these challenges to improve the clinical value of HIFU (27).

NPs are 10-500 nm in size and have previously been reported to increase therapeutic efficacy whilst decreasing the incidence of side effects (28). These nanomedicines selectively accumulate in tumour tissues to realize a selective and efficient therapeutic effect (Fig. 1A) (29). Furthermore, it has been indicated that the adoption of NPs can effectively change the acoustic environment (tissue structure, density, blood supply and functional state on ultrasonic transmission and energy deposition during HIFU treatment) of tumour tissues (30), making them more sensitive to HIFU, resulting in greater ablative efficacy with the same or lower HIFU irradiation doses. Additionally, since the first report on the combination of HIFU with nanotechnology in 2000(31), it has been recognized that HIFU induces target drug release from platforms such as NPs and liposomes to enhance the ablative efficacy of HIFU and improve safety (32); since then, studies have attempted to design nanomedicines to improve the efficacy of HIFU (33-36) however, progress has not seen clinical translation. Additionally, the promotion of theranostics also highlights novel opportunities in this field. The present review aimed to provide an overview of NPs in combination with HIFU for cancer treatment, including the use of nanomedicines to increase the ablative efficacy of HIFU, achieving greater synergic therapeutic efficacy and theranostics by combining imaging probes and HIFU.

2. Nano-therapeutics for HIFU-based cancer treatment

The combination of NPs and HIFU benefits cancer treatment in multiple ways. By enhancing the permeability and retention (EPR) effect, NPs selectively penetrate tumour tissues and change the acoustic environment. NPs can enhance energy deposition and magnify the thermal, mechanical and cavitation effect via formation of microbubbles through a phase transition (37), resulting in improved HIFU ablation efficacy. Additionally, HIFU can alter vascular permeability and disrupt blockade of overexpressed extracellular matrix, thus enhancing the selective accumulation of NPs into tumour tissue (38,39). In addition, HIFU physically induces formation of cell membrane pores via sonoporation, enabling more effective cellular internalization and accumulation of NPs (Fig. 1B) (40). Furthermore, HIFU disrupts NPs to trigger localized drug release at the target site (41), effectively decreasing the off-target damage to normal tissue.

Lipid-based NPs

Lipid-based NPs such as liposomes and solid lipid NPs, are phospholipid bilayer membranes that carry lipid-soluble drugs with an inner core in which hydrophilic drugs can be loaded (42). When constructed to be thermosensitive, these lipid-based NPs respond to thermal changes caused by HIFU, resulting in release of the loaded therapeutics at the selected lesion site (43). For example, Cha et al (44) and Deng et al (45) constructed liposomes sensitive to low temperatures that contained the chemotherapeutic doxorubicin (DOX). Following induction of hyperthermia caused by HIFU irradiation, these liposomes selectively release encapsulated DOX at the tumour tissue to increase their effective concentration in the tumour cell nuclei, whilst keeping the concentration in the general circulation low, thus effectively decreasing off-target damage to normal tissues. These low temperature-sensitive liposomes can be modified by internalised arginine-glycine-aspartic acid (iRGD) to enhance the targeted delivery of iRGD to cancer and tumour vascular cells (Fig. 2A and C-D) (45). iRGD-modified liposomes allow longer opportunities for HIFU irradiation and shorter HIFU exposure times, effectively decreasing incidence of collateral damage such as skin burns caused by long HIFU exposure.

In addition to triggering targeted drug release, Yang et al (46) used HIFU to disrupt the BBB to increase delivery of nanomedicines to the central nervous system: They formed lipid-polymer hybrid NP to deliver CRISPR/Cas9 for the treatment of drug-resistant glioblastoma. Although the nanoscale size of this delivery platform increases penetration into the central nervous system, this increase is limited due to the effectiveness of the BBB. Thus, nanomedicine platforms bind with microbubbles, which allows HIFU to disrupt the BBB. Irradiation of HIFU induces vibrations of the attached microbubbles, resulting in the generation of a shearing force that results in a temporary disturbance of the BBB. Furthermore, these microbubbles rupture and induce further ‘opening’ of the BBB, which allows increased crossing of nano-delivery platforms to tumour tissue in the brain (46,47). Another study applied lipid-polymer hybrid nanomaterial for treatment of glioblastoma and showed that the presence of HIFU triggered rapid release of loaded drugs, with 47% released in 2 min, effectively increasing the therapeutic efficacy (48).

In addition to use of HIFU as a tool to aid drug delivery, another strategy for applying lipid-based nanomaterials is to form nanobubbles to achieve enhanced tumour ablation efficacy. Microbubbles have long been considered synergistic agents for enhancing HIFU therapeutic efficacy (49-51). However, traditional microbubbles are usually too large for tumour tissue penetration and have short circulation times, limiting their use in cancer treatment (49,52). Thus, forming nano-size bubbles with improved tumour penetration ability and increased stability during circulation is key for improving the efficacy of these therapies. Hamano et al (53) and VanOsdol et al (54) formed nanobubble-based liposomes. These echo-contrast gas or perfluoropentane-containing liposomes were reported to achieve up to 4-5-fold greater drug accumulation and release in tumour tissues compared with nanomedicines or HIFU. Furthermore, these nanobubble-based liposomes not only effectively increased HIFU ablation efficacy, thus reducing irradiation time, but also encapsulated antitumour genes, short interfering RNAs and chemotherapeutics to stimulate a synergetic effect, further increasing their antitumour efficacy.

Perfluorocarbon-containing nanomaterials

Perfluorocarbon-containing nanomaterials are a potential therapy that may solve the size and circulation problems of microbubbles (55). By incorporating liquid fluorocarbons into lipids or polymers, these perfluorocarbon-containing nanomaterials shift from a liquid state at room temperature to a gaseous state when temperature rises or following irradiation of HIFU37). The gas released in tumour tissue further triggers the formation of microbubbles, enhancing the cavitation effect of HIFU ablation (56-58). Since fluorocarbons have already penetrated the deep tumour tissue via the EPR effect when it is in a liquid state with a nano size, the microbubbles created following phase shift no longer exhibit problems of short circulation times and low tumour tissue penetrating rates, effectively increasing the therapeutic efficacy of HIFU ablation. Studies have been designed based on perfluorocarbon-containing nanomaterials applied for HIFU ablation (Fig. 2B). Ashida et al (59) prepared a phase-changing nanodroplet from perfluoro-n-pentane (PFP), perfluoro-n-hexane (PFH), dipalmitoyl-phosphatidylcholine, dipalmitoyl-phosphatidic acid and pegylated dipalmitoyl-phosphatidylethanol amine; use of these novel nanomaterials together with HIFU irradiation resulted in moderate tissue damage compared with histotripsy. This moderate damage is sufficient to suppress tumour growth notably compared with HIFU irradiation alone. In addition, compared with histotripsy, the effect of combination therapy effectively decreases incidence of collateral damage to surrounding normal tissues, reducing the severity of side effects. Furthermore, addition of the chemotherapeutic agent adriamycin further enhanced the tumour-suppressing effects of this combination therapy: Tumour regrowth rate was slowed by 1 week when adriamycin was used during the 30-day observation time. However, the effect of repetitive therapy management with longer observation periods should be assessed to confirm the therapeutic effects of phase-changing perfluorocarbon-containing nanodroplets. The choice of perfluorocarbon is key when constructing HIFU-appliable nanomaterials. Currently, the most commonly used perfluorocarbons are PFP and PFH (60). The boiling temperatures of other perfluorocarbons are usually either too low or high to be applicable for clinical use. As boiling temperatures also affect the phase-shifting temperature of constructed nanomaterials (61,62), there remain challenges before these can be used clinically. As the phase shifting temperature of PFP is lower than that of PFH (>40 vs. >60˚C), PFP may be a better choice for nanomaterial construction, as lower HIFU irradiation doses can be used (63). Zhang et al (64) constructed a poly(lactide-co-glycolic acid) (PLGA) NP that incorporated PFP and hematoporphyrin monomethyl ether (HMME) as synergistic agents (HMME + PFP/PLGA) for HIFU ablation. These agents were further modified by streptavidin as a pre-targeting agent via a two-step biotin-avidin technique. In addition to a lower HIFU irradiation dosage required, the cavitation effect of HIFU, the sonodynamic effect and vascular endothelial growth factor receptor-2 antibody worked together to induce secondary necrosis surrounding the initial HIFU ablation area, resulting in a greater synergetic effect with less collateral damage to the normal tissue. This method highlights the application of perfluorocarbon-containing nanomaterials as HIFU synergetic agents for deep tumour ablation and ablation of tumours with barriers along the HIFU beam path; however, additional studies are needed before this method can be used in clinical practice.

Magnetic nanomaterials

Magnetic nanomaterials, with their unique features such as ease of manipulation using magnets and thermal responsiveness to ultrasound and magnets, have potential as effective sonosensitizers for HIFU cancer therapy (64). Sun et al (65,66), You et al (67), Ho et al (68) and Dibaji et al (69) confirmed that magnetic nanomaterials enhance the HIFU cavitation effect and thus effectively increase tumour tissue destruction efficacy with a lower HIFU exposure dose. According to Devarakonda et al (70), the adoption of magnetic nanomaterials (superparamagnetic iron oxide NPs; 0.047% w/v) halves the HIFU irradiation dose required to obtain 13 mm3 tumour destruction volume, significantly reducing the side effects caused by high HIFU doses (70). They also discovered that the thermal enhancing efficacy of magnetic nanomaterials was higher than that of gold NPs, which are another HIFU hyperthermal candidate, making magnetic NPs clinically preferable (71). However, the mechanism by which magnetic NPs enhance HIFU cancer therapy remains unclear. It has been suggested that magnetic NPs increase attenuation of sound waves in tumour tissue. Thus, when magnetic NPs selectively penetrate the tumour tissues through the EPR effect, a lower HIFU dose is needed to achieve tumour destruction efficacy, with decreased collateral damage to surrounding normal tissues at these lower HIFU irradiation doses (72). Sadeghi-Goughari et al (36,73) discovered that viscous and thermal reaction with medium at the surface of magnetic particles is the primary mechanism that aids conversion of acoustic energy into heat, achieving greater temperature rises with directed HIFU ablation. Additionally, a numerical model was established that could accurately predict and analyse HIFU ablation process when NPs were used, thus providing a novel tool to uncover the detailed mechanism by which magnetic NPs affect HIFU ablation, which is beneficial for future magnetic NP development and potential clinical application.

Bacteria-based targeting

Several studies have indicated that certain anaerobic bacteria species such as Bifidobacterium can colonize and grow into a tumour mass where there is a hypoxic atmosphere due to a lack of sufficient blood supply, whereas in normal tissues, the supply of oxygen would prevent colonization and proliferation of these anaerobic bacteria. Thus, bacteria such as Bifidobacterium have been considered as potential markers that may aid in the identification of tumour masses and facilitate tumour targeting diagnosis and treatment (74-76). When using Bifidobacterium as the targeting agent for tumour tissue, it could be either directly cross-linked to the delivery platform acting as an arch on a ‘bacterial robot’ or allowed to colonize in the tumour mass first, then another Bifidobacterium targeting NP can be utilized to achieve indirect targeting of the tumour (77). Jiang et al (78) compared these targeting strategies and found that when combined with HIFU ablation, the indirect targeting method was more efficient than cross-linking bacteria with NP payloads. This was because agglomeration may occur when preparing this bacteria-containing cross-link nanomedicine, making it larger and thus more difficult for it to penetrate the tumour mass. Furthermore, they also indicated that Bifidobacterium that had colonized a tumour mass in advance triggered activation of macrophages to the phagocytotic active phase, effectively aiding the engulfing and retention of nanotherapeutics inside the tumour tissue as endocytosis is one of the primary mechanisms by which NPs enter cells. Thus, based on these findings, bacteria-based nano strategies for HIFU ablation have focused on bacterial targeting instead of using bacteria-containing NPs (79-81). Additionally, aptamer-CCFM641-5-functionalized PFH-loaded PLGA NPs (82) and polyethyleneimine-modified PLGA NPs loaded with sodium bicarbonate (83) that target Bifidobacterium have been developed to achieve selective delivery of therapeutics into a tumour mass. These two NPs are reported to prolong median survival time of tumour-bearing mice as well as enhance HIFU ablation efficacy. Additionally, since no specific targeting proteins or surface antigens were involved in these studies and anaerobic conditions are present in a variety of tumour types, these nanotherapeutics may be applied regardless of the tumour type. However, these bacteria- and NP-based HIFU ablation methods may not be suitable for tumour masses ≤1-2 mm as the vascular system in a small tumour mass is sufficient to create an oxygen-rich atmosphere (84). These bacteria-based NPs may be used to encapsulate imaging agents for photoacoustic and ultrasound imaging, promoting HIFU cancer theranostics (Fig. 3A) (85); however, further confirmatory studies are required.

Other types of NP

Other NPs, including paclitaxel-loaded thiolated human serum albumin NP-conjugated microbubble complexes (86), heat shock-targeted N-(2-hydroxypropyl) methacrylamide copolymer-docetaxel conjugates (87), Cy5.5-labelled glycol chitosan, nitroxide free radical-generating (88,89), 1,1,2-trichlorotrifluoroethane incorporating pullulan-DOX (90) and phospholipid hydrophobic mesoporous silica NPs (91,92), enhance the efficacy of HIFU ablation with decreased side effects, although additional studies on similar types of NP are required to confirm their effectiveness. The clinical transition of NPs lack systemic examination regarding the safety profile including biocompatibility, biodegradation, tumour accumulation and stability in animals other than mice. Furthermore, establishment of scalable, economical and reproducible synthesis methods for these NPs is also needed before they can be used clinically (93).

3. Nano-based HIFU cancer theranostics

Cancer theranostics (cancer therapy and diagnosis through the packaging of various therapeutic drugs and diagnostic contrast agents) is a relatively new concept in the field of precision medicine. In addition to incorporating therapeutic agents to enhance HIFU ablation efficacy to induce selective therapeutic drug release, NPs can be used to encapsulate diagnostic agents that can enhance the imaging contrast of tumourigenic sites (94-96). This can allow accurate targeting of HIFU ablation, real-time imaging monitoring of ablation procedure and evaluation of the therapeutic response without the need for extra medical tests (Fig. 1C) (97), as well as adjustments of the treatment to maximize the therapeutic efficacy and minimize the collateral damage to the surrounding normal tissue.

Ultrasound-based nano-theranostics for HIFU

HIFU is a type of ultrasound-based treatment method, thus the specific therapeutics used for HIFU have acoustic properties, which makes ultrasounds one of the first choices for monitoring of HIFU. However, given that diagnostic and therapeutic ultrasound have different frequencies (98,99), the acoustic properties of the therapeutics may respond to only one type of ultrasound. Therefore, it is important to design therapeutics that are responsive to both diagnostic and therapeutic ultrasound. Blum et al (100) constructed a polyethylene glycol (PEG)-lipid-shelled microbubble that creates microbubble NPs in the presence of fluorocarbon interiors (C4F10, C5F12 and C6F14) and ultrasound pulses. These microbubble NPs are not detectable by ultrasound, but under HIFU irradiation, the integrated image brightness of NPs on the cadence contrast pulse sequencing mode increases, making them visible on ultrasound scans. The addition of fluorocarbons to this nanosystem also allows enhancement of HIFU efficacy, which was confirmed by complete detachment of breast cancer cells in vitro under HIFU irradiation in presence of these NPs. However, further in vivo studies are required to examine the theranostic efficacy and biosafety profiles of these NPs. Zhu et al (101) examined the in vivo efficacy of nano-theranostics for HIFU ablation. They synthesized a pH-sensitive poly(ethylene glycol) that produced O2 from endogenous H2O2. The generated O2 not only served as a contrast agent for diagnostic ultrasound imaging but served as a synergist agent to enhance HIFU ablation efficacy. Furthermore, they also discovered that this nano-theranostic induced normoxic conditions in the tumour tissues to enhance the chemotherapeutic efficacy of DOX, allowing both theranostics and combination therapy of HIFU and chemotherapy. Li et al (102) designed pentafluoropentane/C9F17-PAsp-ss-camptothecin (CPT) nanodroplets that allowed ultrasound imaging and combination therapy of not only HIFU ablation and chemotherapy but also immunotherapy. The nanodroplets demonstrated an HIFU/glutathione (GSH)-dual responsive drug release profile and successfully delivered the loaded chemotherapeutic CPT into tumour tissue upon HIFU irradiation. These nanodroplets can also generate immunogenic debris following HIFU irradiation and induce maturation of dendritic cells (DCs) via exposure of damage-associated molecular patterns, effectively increasing the infiltration of effector T cells into tumour tissue and thus enhancing the efficacy of tumour immunotherapy. The incorporation of PFP also allows ultrasound monitoring of the whole procedure, making these nanodroplets another promising HIFU-based theranostic candidate. On the other hand, Chen et al (35) focused on HIFU and synthesized PFP-loaded polymer NPs (PFP@Polymer NPs) that were responsive to a dual-frequency HIFU pattern. Compared with single-frequency HIIFU, PFP@Polymer NPs under the irradiation of dual-frequency HIFU (1.1 and 5.0 MHz) were reported to significantly decrease the acoustic intensity threshold needed for ablation from 216.86 to 62.38 W/cm2, thus effectively decreasing collateral damage. Furthermore, these polymer NPs combined with dual-frequency HIFU also demonstrated improved tumour inhibition rates at half the irradiation time of single-frequency HIFU and improved ultrasound contrast-generating quality compared with traditional PFP@BSA nanodroplets. Whether these NPs responsive to dual-frequency HIFU can also encapsulate other therapeutics such as chemotherapeutics or immunotherapeutics to achieve a synergistic theranostic effect remains to be examined; combination therapies are desirable due to potentially improved treatment outcomes and decreased side effects (103,104).

MRI-based nano-theranostics for HIFU

Although ultrasound is often utilized as an imaging tool for HIFU theranostics, MRI is considered an improved imaging tool given its non-invasive nature and high spatial and anatomical resolution (105). Although the majority of HIFU synergists do not have paramagnetic properties that can be seen using an MR scan, certain MR sequences such as MRI thermometry allow for real-time quantification of the local temperature in the tumour tissues (106,107), thus allowing HIFU ablation. Given that magnetic NPs may disrupt the magnetic field when applied for MRI (70), gold NPs are an alternative for MRI-guided HIFU ablation. By using MRI thermometry to evaluate the tissue temperature, Devarakonda et al (108) discovered that the addition of gold NPs significantly enhances the increase in temperature to increase lesion volume compared with HIFU ablation alone. This enhancing effect of gold NPs was also confirmed in vivo (109), although a localized direct injection of NPs into the superficial tumour tissue was used, which is not a method used in clinical practice. Thus, further intravenous injection studies are required to assess the theranostic efficacy of gold NPs and their impact on efficacy of HIFU ablation. Although MRI thermometry can be utilized to evaluate response to treatments, this method of evaluation is indirect (through the measurement of local temperature) and non-selective with unsatisfactory imaging precision due to the lack of involvement of MRI contrast agents (37). Studies have used NPs to combine both MRI contrast and HIFU synergetic agents to achieve MRI-guided HIFU theranostics. Tang et al (39) constructed a temperature-responsive nanoplatform [PFH/DOX@PLGA/Fe3O4-folate (FA)] that achieved HIFU theranostics. The encapsulation of Fe3O4 allowed T2-weighted imaging of the tumour once particles had accumulated into the hepatoma tissue through the EPR effect and active targeting induced by the attached FA. The encapsulation of PFH also permits contrast-enhanced ultrasound imaging of tumour tissue, allowing for a multi-modal imaging profile. In addition, the incorporation of PFH and DOX significantly improved the efficacy of HIFU ablation and allowed enhanced chemotherapeutic efficacy, respectively, evidenced by the strongest in vivo tumour inhibition rate and greatest reduction in tumour volumes among all experimental and control groups. Thus, this nanoplatform could achieve not only multi-modal cancer imaging but also multi-modal treatment. Kuai et al (110) designed a type of perfluorooctyl bromide (PFOB) nanoemulsion that contained MnO2 NPs to allow a combination of computed tomography (CT) and MRI for multi-modal imaging and combination of HIFU ablation and immunotherapy for multi-modal treatment. The use of PFOB not only allowed CT imaging of tumour tissues as it is a desirable CT contrast agent (111,112), but also transformed into microbubbles under HIFU irradiation and enhanced the cavitation effect for stronger HIFU ablation efficacy. The encapsulation of MnO2 also allowed T1-weighted enhanced imaging of tumour tissues instead of T2-weighted enhanced imaging, which is preferable due to difficulties of detecting small negative-contrast lesions on T2-weighted enhanced imaging (113). In addition to the stronger HIFU ablation efficacy, which allowed lower HIFU exposure doses and administration times and thus less collateral damage to the normal tissue, these NPs were also reported to deplete GSH as a result of MnO2-mediated disruption of the antioxidant defence system of tumour tissue and to promote strong immunogenic cell death by inducing maturation of DCs and enhancing activation of CD4+ and CD8+ cells, significantly inhibiting growth of the primary tumour and lung metastasis through combination therapy (114).

Photoacoustic imaging-based nano-theranostics for HIFU

Photoacoustic imaging is a promising biomedical imaging technology that can overcome certain limitations of current ultrasound with its high optical contrast, relatively low cost and portability (115). It can be used to visualize both endogenous and exogenous chromophores with a high spatial resolution (116,117), penetrate >5 cm biological tissue for imaging (118) and is not associated with the potential side effects caused by ionizing radiation. Studies have indicated that photoacoustic imaging can be utilized to image small molecules, including those that are readily extravasated and are present on the cell membrane or intracellularly (119,120). Thus, studies have adopted photoacoustic imaging as the imaging tool for HIFU cancer theranostics. Feng et al (121) constructed an ammonium bicarbonate-containing liposome (Lip-ABC) that could generate microbubbles under HIFU irradiation (122). Through photoacoustic imaging, these liposomes were shown to accumulate in the tumour interstitial space where they generated bubbles to increase cavitation and energy deposition, resulting in higher HIFU ablation rate in a theranostic manner. Gao et al (123) on the other hand designed HMME-loaded CaCO3 NPs (Ca@H) (108). Ca@H NPs responded to the acid tumour microenvironment to produce CO2 and release HMME. These agents may serve as a photoacoustic imaging enhancer for guidance and monitoring of the entire therapeutic process, allowing combination therapy using HIFU ablation and sonodynamic therapy to promote near-complete removal of residual tumour tissue. Although photoacoustic imaging has its diagnostic advantages, its clinical applications are still limited currently (123-125). Thus, several studies have attempted to combine this novel imaging technology with other clinical imaging methods to allow multi-modal imaging of HIFU cancer theranostics. Yan et al (126) and Zhang et al (127) designed NPs that allowed a combination of ultrasound and photoacoustic monitoring. Zhang et al (127) encapsulated the chemotherapeutic DOX in NPs to achieve synergetic therapy of both HIFU ablation and chemotherapy, allowing multi-modal imaging and treatment of cancer theranostics. Both ultrasound and photoacoustic imaging are based on acoustic characteristics of NPs and tumour tissue; this could simplify the design of nanomedicines but risks missing information on the tumour when imaging (128). Thus, studies have combined photoacoustic with other imaging methods. For example, Li et al (129) prepared an F3 (penetrating peptide)-PLGA nanoplatform that could co-deliver sonosensitizer methylene blue and the magnetic resonance contrast agent gadolinium 2-[bis[2-(carboxylatomethyl-(methylcarbamoylmethyl)amino)ethyl]amino]acetate to allow photoacoustic imaging and MRI. This F3-PLGA@MB/Gd platform could further induce a synergistic therapeutic effect via tumour cell apoptosis triggered by HIFU and sonodynamic ultrasound (Fig. 3C-D). Yang et al (130) designed a Fe3O4-shelled and L-arginine-encapsulated PLGA NP that could allow for tri-model imaging (ultrasound, MRI and photoacoustic imaging). These NPs also release nitric oxide as an antitumour gas therapy agent and change the acoustic properties of the tumour tissue to augment HIFU ablation efficacy, realizing synergetic cancer theranostics (Fig. 3B, D and E). Although promising, further clinical trials are required on these nano-based HIFU theranostic methods before they can be translated into clinical practice to benefit patients with cancer.

4. Conclusions and future perspectives

Overall, the combination of nanotechnologies with non-invasive HIFU cancer ablation-based therapies may prove to be a beneficial future treatment. These nanomedicines increase the local HIFU ablation efficacy by enhancing cavitation and changing the acoustic properties of tumour tissue, decrease incidence of collateral damage by allowing for lower HIFU exposure doses and shorter exposure times, achieve a synergetic therapeutic effect by allowing for the concomitant delivery of other therapeutics such as chemotherapeutics, photothermal therapeutics or immunotherapeutics and enable theranostic disease management by allowing monitoring of treatment using single- or multi-modal imaging.

Although progress has been made in this field, challenges remain regarding these HIFU-appliable nanomedicines before they can be used clinically. Although most of these nanomedicines have been reported to exhibit low toxicity in vivo, a degree of hepatotoxicity is observed, often as hepatic fibrosis, particularly in patients with hepatoma (131,132). Thus, it is important to assess and minimise the toxicity and side effects of these nanomedicines. Furthermore, as the majority of the aforementioned nano-based HIFU cancer treatment studies were conducted on small animals, whether the same HIFU dosages used in mice to stimulate these nanomedicines also apply in humans remains to be determined. Additionally, whether the higher HIFU dosages used in clinical practice may hamper therapeutic effects of these nanomedicines and trigger other undesired side effects remain to be assessed (133,134). These issues should be addressed in future studies to improve the value of HIFU-appliable nanomedicines and thus promote their clinical transition.

Acknowledgements

Not applicable.

Funding

Funding: The present study was funded by the Chongqing Science and Technology Bureau ‘Doctor through train’ Scientific Research Project (grant no. CSTB2022BSXM-JCX0082) and the China Postdoctoral Science Foundation (grant no. 2020TQ0212).

Availability of data and materials

Not applicable.

Authors' contributions

QZ conceived, wrote and reviewed the manuscript. BX wrote the manuscript. BX and JL performed the literature review and constructed figures. SZ, XH and XL reviewed the manuscript and agreed to be accountable for all aspects of the work. SZ and XL acquired the funding. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Patel D, Shah Y, Thakkar N, Shah K and Shah M: Implementation of artificial intelligence techniques for cancer detection. Augment Hum Res. 5:1–10. 2020.

2 

Burugu S, Dancsok AR and Nielsen TO: Emerging targets in cancer immunotherapy. Semin Cancer Biol. 52:39–52. 2018.PubMed/NCBI View Article : Google Scholar

3 

Liu S, Zhou X, Yao Y, Shi K, Yu M and Ji F: Resection of the gastric submucosal tumor (G-SMT) originating from the muscularis propria layer: Comparison of efficacy, patients' tolerability, and clinical outcomes between endoscopic full-thickness resection and surgical resection. Surg Endosc. 34:4053–4064. 2020.PubMed/NCBI View Article : Google Scholar

4 

Pirisinu M, Pham TC, Zhang DX, Hong TN, Nguyen LT and Le MT: Extracellular vesicles as natural therapeutic agents and innate drug delivery systems for cancer treatment: Recent advances, current obstacles, and challenges for clinical translation. Semin Cancer Biol. 80:340–355. 2022.PubMed/NCBI View Article : Google Scholar

5 

Westhoff N, Ernst R, Kowalewski KF, Derigs F, Neuberger M, Nörenberg D, Popovic ZV, Ritter M, Stephan Michel M and von Hardenberg J: Medium-term oncological efficacy and patient-reported outcomes after focal high-intensity focused ultrasound: The FOXPRO trial. Eur Urol Focus. 22:S2405–S4569. 2022.PubMed/NCBI View Article : Google Scholar

6 

Li Y, Wang S, Chen L, Feng Y, Shen Z, Chen X, Huang G and Ni Y: Sequential administrations of a vascular-disrupting agent, high-intensity focused ultrasound, and a radioactively labeled necrosis avid compound for eradicating solid malignancies. Technol Cancer Res Treat. 21(15330338221136716)2022.PubMed/NCBI View Article : Google Scholar

7 

Zhong Q, Tang F, Ni T, Chen Y, Liu Y, Wu J, Zhou W, Feng Z, Lu X, Tan S and Zhang Y: Salvage high intensity focused ultrasound for residual or recurrent cervical cancer after definitive chemoradiotherapy. Front Immunol. 13(995930)2022.PubMed/NCBI View Article : Google Scholar

8 

Lindstrom PA: Prefrontal ultrasonic irradiation-a substitute for lobotomy. AMA Arch Neurol Psychiatry. 72:399–425. 1954.PubMed/NCBI View Article : Google Scholar

9 

Fry WJ, Barnard JW, Fry EJ, Krumins RF and Brennan JF: Ultrasonic lesions in the mammalian central nervous system. Science. 122:517–518. 1955.PubMed/NCBI

10 

ter Haar G: Intervention and therapy. Ultrasound Med Biol 26 Suppl. 1:S51–S54. 2000.PubMed/NCBI View Article : Google Scholar

11 

Liu L, Wang T and Lei B: High-intensity focused ultrasound (HIFU) ablation versus surgical interventions for the treatment of symptomatic uterine fibroids: A meta-analysis. Eur Radiol. 32:1195–1204. 2022.PubMed/NCBI View Article : Google Scholar

12 

Wang C, Li Z and Bai J: Bubble-assisted HIFU ablation enabled by calcium peroxide. J Mater Chem B. 10:4442–4451. 2022.PubMed/NCBI View Article : Google Scholar

13 

Chaussy CG and Thüroff S: High-Intensity focused ultrasound for the treatment of prostate cancer: A review. J Endourol. 31(S1):S30–S37. 2017.PubMed/NCBI View Article : Google Scholar

14 

Napoli A, Alfieri G, Scipione R, Leonardi A, Fierro D, Panebianco V, De Nunzio C, Leonardo C and Catalano C: High-intensity focused ultrasound for prostate cancer. Expert Rev Med Devices. 17:427–433. 2020.PubMed/NCBI View Article : Google Scholar

15 

Fishman PS and Fischell JM: Focused ultrasound mediated opening of the blood-brain barrier for neurodegenerative diseases. Front Neurol. 12(749047)2021.PubMed/NCBI View Article : Google Scholar

16 

Martínez-Fernández R, Máñez-Miró JU, Rodríguez-Rojas R, Del Álamo M, Shah BB, Hernández-Fernández F, Pineda-Pardo JA, Monje MHG, Fernández-Rodríguez B, Sperling SA, et al: Randomized trial of focused ultrasound subthalamotomy for Parkinson's Disease. N Engl J Med. 383:2501–2513. 2020.PubMed/NCBI View Article : Google Scholar

17 

Moosa S, Martínez-Fernández R, Elias WJ, Del Alamo M, Eisenberg HM and Fishman PS: The role of high-intensity focused ultrasound as a symptomatic treatment for Parkinson's disease. Mov Disord. 34:1243–1251. 2019.PubMed/NCBI View Article : Google Scholar

18 

Giordano M, Caccavella VM, Zaed I, Foglia Manzillo L, Montano N, Olivi A and Polli FM: Comparison between deep brain stimulation and magnetic resonance-guided focused ultrasound in the treatment of essential tremor: A systematic review and pooled analysis of functional outcomes. J Neurol Neurosurg Psychiatry. 91:1270–1278. 2020.PubMed/NCBI View Article : Google Scholar

19 

Abe K, Horisawa S, Yamaguchi T, Hori H, Yamada K, Kondo K, Furukawa H, Kamada H, Kishima H, Oshino S, et al: Focused ultrasound thalamotomy for refractory essential tremor: A Japanese multicenter single-arm study. Neurosurgery. 88:751–757. 2021.PubMed/NCBI View Article : Google Scholar

20 

Jeng CJ, Ou KY, Long CY, Chuang L and Ker CR: 500 cases of high-intensity focused ultrasound (HIFU) ablated uterine fibroids and adenomyosis. Taiwan J Obstet Gynecol. 59:865–871. 2020.PubMed/NCBI View Article : Google Scholar

21 

Sequeiros RB, Joronen K, Komar G and Koskinen SK: High intensity focused ultrasound (HIFU) in tumor therapy. Duodecim. 133:143–149. 2017.PubMed/NCBI

22 

Marinova M, Wilhelm-Buchstab T and Strunk H: Advanced pancreatic cancer: High-Intensity Focused Ultrasound (HIFU) and other local ablative therapies. RoFo. 191:216–227. 2019.PubMed/NCBI View Article : Google Scholar

23 

Zhou YF: High intensity focused ultrasound in clinical tumor ablation. World J Clin Oncol. 2:8–27. 2011.PubMed/NCBI View Article : Google Scholar

24 

Bachu VS, Kedda J, Suk I, Green JJ and Tyler B: High-Intensity Focused Ultrasound: A Review of Mechanisms and Clinical Applications. Ann Biomed Eng. 49:1975–1991. 2021.PubMed/NCBI View Article : Google Scholar

25 

Izadifar Z, Izadifar Z, Chapman D and Babyn P: An introduction to high intensity focused ultrasound: Systematic review on principles, devices, and clinical applications. J Clin Med. 9(460)2020.PubMed/NCBI View Article : Google Scholar

26 

Miller DL, Smith NB, Bailey MR, Czarnota GJ, Hynynen K and Makin IR: Bioeffects Committee of the American Institute of Ultrasound in Medicine. Overview of therapeutic ultrasound applications and safety considerations. J Ultrasound Med. 31:623–634. 2012.PubMed/NCBI View Article : Google Scholar

27 

Awad NS, Paul V, AlSawaftah NM, Ter Haar G, Allen TM, Pitt WG and Husseini GA: Ultrasound-responsive nanocarriers in cancer treatment: A review. ACS Pharmacol Transl Sci. 4:589–612. 2021.PubMed/NCBI View Article : Google Scholar

28 

Zhang Q, Dai X, Zhang H, Zeng Y, Luo K and Li W: Recent advances in development of nanomedicines for multiple sclerosis diagnosis. Biomed Mater. 16(024101)2021.PubMed/NCBI View Article : Google Scholar

29 

Zeng Y, Li Z, Zhu H, Gu Z, Zhang H and Luo K: Recent advances in nanomedicines for multiple sclerosis therapy. ACS Appl Bio Mater. 3:6571–6597. 2020.PubMed/NCBI View Article : Google Scholar

30 

Zhou Y, Wang Z, Chen Y, Shen H, Luo Z, Li A, Wang Q, Ran H, Li P, Song W, et al: Microbubbles from gas-generating perfluorohexane nanoemulsions for targeted temperature-sensitive ultrasonography and synergistic HIFU ablation of tumors. Adv Mater. 25:4123–4130. 2013.PubMed/NCBI View Article : Google Scholar

31 

Du Y, Lin L, Zhang Z, Tang Y, Ou X, Wang Y and Zou J: Drug-loaded nanoparticles conjugated with genetically engineered bacteria for cancer therapy. Biochem Biophys Res Commun. 606:29–34. 2022.PubMed/NCBI View Article : Google Scholar

32 

Manthe RL, Foy SP, Krishnamurthy N, Sharma B and Labhasetwar V: Tumor ablation and nanotechnology. Mol Pharm. 7:1880–1898. 2010.PubMed/NCBI View Article : Google Scholar

33 

Chen D and Wu J: An in vitro feasibility study of controlled drug release from encapsulated nanometer liposomes using high intensity focused ultrasound. Ultrasonics. 50:744–749. 2010.PubMed/NCBI View Article : Google Scholar

34 

O'Neill BE, Vo H, Angstadt M, Li KP, Quinn T and Frenkel V: Pulsed high intensity focused ultrasound mediated nanoparticle delivery: Mechanisms and efficacy in murine muscle. Ultrasound Med Biol. 35:416–424. 2009.PubMed/NCBI View Article : Google Scholar

35 

Chen J, Nan Z, Zhao Y, Zhang L, Zhu H, Wu D, Zong Y, Lu M, Ilovitsh T, Wan M, et al: Enhanced HIFU Theranostics with dual-frequency-ring focused ultrasound and activatable perfluoropentane-loaded polymer nanoparticles. Micromachines (Basel). 12(1324)2021.PubMed/NCBI View Article : Google Scholar

36 

Sadeghi-Goughari M, Jeon S and Kwon HJ: Analytical and numerical model of high intensity focused ultrasound enhanced with nanoparticles. IEEE Trans Biomed Eng. 67:3083–3093. 2020.PubMed/NCBI View Article : Google Scholar

37 

Chen Y, Chen H and Shi J: Nanobiotechnology promotes noninvasive high-intensity focused ultrasound cancer surgery. Adv Healthc Mater. 4:158–165. 2015.PubMed/NCBI View Article : Google Scholar

38 

Poh S, Chelvam V and Low PS: Comparison of nanoparticle penetration into solid tumors and sites of inflammation: Studies using targeted and nontargeted liposomes. Nanomedicine (Lond). 10:1439–1449. 2015.PubMed/NCBI View Article : Google Scholar

39 

Tang H, Guo Y, Peng L, Fang H, Wang Z, Zheng Y, Ran H and Chen Y: In Vivo targeted, responsive, and synergistic cancer nanotheranostics by magnetic resonance imaging-guided synergistic high-intensity focused ultrasound ablation and chemotherapy. ACS Appl Mater Interfaces. 10:15428–15441. 2018.PubMed/NCBI View Article : Google Scholar

40 

Tharkar P, Varanasi R, Wong WSF, Jin CT and Chrzanowski W: Nano-Enhanced drug delivery and therapeutic ultrasound for cancer treatment and beyond. Front Bioeng Biotechnol. 7(324)2019.PubMed/NCBI View Article : Google Scholar

41 

Wang X, Yan F, Liu X, Wang P, Shao S, Sun Y, Sheng Z, Liu Q, Lovell JF and Zheng H: Enhanced drug delivery using sonoactivatable liposomes with membrane-embedded porphyrins. J Control Release. 286:358–368. 2018.PubMed/NCBI View Article : Google Scholar

42 

Chaudhuri A, Kumar DN, Shaik RA, Eid BG, Abdel-Naim AB, Md S, Ahmad A and Agrawal AK: Lipid-Based nanoparticles as a pivotal delivery approach in triple negative breast cancer (TNBC) therapy. Int J Mol Sci. 23(10068)2022.PubMed/NCBI View Article : Google Scholar

43 

Yudina A and Moonen C: Ultrasound-induced cell permeabilisation and hyperthermia: Strategies for local delivery of compounds with intracellular mode of action. Int J Hyperthermia. 28:311–319. 2012.PubMed/NCBI View Article : Google Scholar

44 

Cha JM, You DG, Choi EJ, Park SJ, Um W, Jeon J, Kim K, Kwon IC, Park JC, Kim HR and Park JH: Improvement of Antitumor Efficacy by Combination of Thermosensitive Liposome with High-Intensity Focused Ultrasound. J Biomed Nanotechnol. 12:1724–1733. 2016.PubMed/NCBI View Article : Google Scholar

45 

Deng Z, Xiao Y, Pan M, Li F, Duan W, Meng L, Liu X, Yan F and Zheng H: Hyperthermia-triggered drug delivery from iRGD-modified temperature-sensitive liposomes enhances the anti-tumor efficacy using high intensity focused ultrasound. J Control Release. 243:333–341. 2016.PubMed/NCBI View Article : Google Scholar

46 

Yang Q, Zhou Y, Chen J, Huang N, Wang Z and Cheng Y: Gene therapy for drug-resistant glioblastoma via lipid-polymer hybrid nanoparticles combined with focused ultrasound. Int J Nanomedicine. 16:185–199. 2021.PubMed/NCBI View Article : Google Scholar

47 

Arsiwala TA, Sprowls SA, Blethen KE, Adkins CE, Saralkar PA, Fladeland RA, Pentz W, Gabriele A, Kielkowski B, Mehta RI, et al: Ultrasound-mediated disruption of the blood tumor barrier for improved therapeutic delivery. Neoplasia. 23:676–691. 2021.PubMed/NCBI View Article : Google Scholar

48 

Luo Z, Jin K, Pang Q, Shen S, Yan Z, Jiang T, Zhu X, Yu L, Pang Z and Jiang X: On-Demand drug release from dual-targeting small nanoparticles triggered by high-intensity focused ultrasound enhanced glioblastoma-targeting therapy. ACS Appl Mater Interfaces. 9:31612–31625. 2017.PubMed/NCBI View Article : Google Scholar

49 

Sokka S, King R and Hynynen K: MRI-guided gas bubble enhanced ultrasound heating in in vivo rabbit thigh. Phys Med Biol. 48:223–241. 2003.PubMed/NCBI View Article : Google Scholar

50 

Clark A, Bonilla S, Suo D, Shapira Y and Averkiou M: Microbubble-Enhanced Heating: Exploring the effect of microbubble concentration and pressure amplitude on high-intensity focused ultrasound treatments. Ultrasound Med Biol. 47:2296–2309. 2021.PubMed/NCBI View Article : Google Scholar

51 

Xin Y, Zhang A, Xu LX and Fowlkes JB: Numerical study of bubble cloud and thermal lesion evolution during acoustic droplet vaporization enhanced HIFU treatment. J Biomech Eng. 144(031007)2022.PubMed/NCBI View Article : Google Scholar

52 

Okita K, Sugiyama K, Takagi S and Matsumto Y: Microbubble behavior in an ultrasound field for high intensity focused ultrasound therapy enhancement. J Acoust Soc Am. 134:1576–1585. 2013.PubMed/NCBI View Article : Google Scholar

53 

Hamano N, Negishi Y, Takatori K, Endo-Takahashi Y, Suzuki R, Maruyama K, Niidome T and Aramaki Y: Combination of bubble liposomes and high-intensity focused ultrasound (HIFU) enhanced antitumor effect by tumor ablation. Biol Pharm Bull. 37:174–177. 2014.PubMed/NCBI View Article : Google Scholar

54 

VanOsdol J, Ektate K, Ramasamy S, Maples D, Collins W, Malayer J and Ranjan A: Sequential HIFU heating and nanobubble encapsulation provide efficient drug penetration from stealth and temperature sensitive liposomes in colon cancer. J Control Release. 247:55–63. 2017.PubMed/NCBI View Article : Google Scholar

55 

Zhou LQ, Li P, Cui XW and Dietrich CF: Ultrasound nanotheranostics in fighting cancer: Advances and prospects. Cancer Lett. 470:204–219. 2020.PubMed/NCBI View Article : Google Scholar

56 

Li K, Liu Y, Zhang S, Xu Y, Jiang J, Yin F, Hu Y, Han B, Ge S, Zhang L and Wang Y: Folate receptor-targeted ultrasonic PFOB nanoparticles: Synthesis, characterization and application in tumor-targeted imaging. Int J Mol Med. 39:1505–1515. 2017.PubMed/NCBI View Article : Google Scholar

57 

Sheeran PS, Matsunaga TO and Dayton PA: Phase change events of volatile liquid perfluorocarbon contrast agents produce unique acoustic signatures. Phys Med Biol. 59:379–401. 2014.PubMed/NCBI View Article : Google Scholar

58 

Picheth G, Houvenagel S, Dejean C, Couture O, Alves de Freitas R, Moine L and Tsapis N: Echogenicity enhancement by end-fluorinated polylactide perfluorohexane nanocapsules: Towards ultrasound-activable nanosystems. Acta Biomater. 64:313–322. 2017.PubMed/NCBI View Article : Google Scholar

59 

Ashida R, Kawabata K, Maruoka T, Asami R, Yoshikawa H, Takakura R, Ioka T, Katayama K and Tanaka S: New approach for local cancer treatment using pulsed high-intensity focused ultrasound and phase-change nanodroplets. J Med Ultrason (2001). 42:457–466. 2015.PubMed/NCBI View Article : Google Scholar

60 

Xu T, Cui Z, Li D, Cao F, Xu J, Zong Y, Wang S, Bouakaz A, Wan M and Zhang S: Cavitation characteristics of flowing low and high boiling-point perfluorocarbon phase-shift nanodroplets during focused ultrasound exposures. Ultrason Sonochem. 65(105060)2020.PubMed/NCBI View Article : Google Scholar

61 

Rapoport N: Phase-shift, stimuli-responsive perfluorocarbon nanodroplets for drug delivery to cancer. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 4:492–510. 2012.PubMed/NCBI View Article : Google Scholar

62 

Sheeran PS and Dayton PA: Phase-change contrast agents for imaging and therapy. Curr Pharm Des. 18:2152–2165. 2012.PubMed/NCBI View Article : Google Scholar

63 

Kwizera EA, Stewart S, Mahmud MM and He X: Magnetic nanoparticle-mediated heating for biomedical applications. J Heat Transfer. 144(030801)2022.PubMed/NCBI View Article : Google Scholar

64 

Zhang Y, Yong L, Luo Y, Ding X, Xu D, Gao X, Yan S, Wang Q, Luo J, Pu D and Zou J: Enhancement of HIFU ablation by sonosensitizer-loading liquid fluorocarbon nanoparticles with pre-targeting in a mouse model. Sci Rep. 9(6982)2019.PubMed/NCBI View Article : Google Scholar

65 

Sun Y, Zheng Y, Ran H, Zhou Y, Shen H, Chen Y, Chen H, Krupka TM, Li A, Li P, et al: Superparamagnetic PLGA-iron oxide microcapsules for dual-modality US/MR imaging and high intensity focused US breast cancer ablation. Biomaterials. 33:5854–5864. 2012.PubMed/NCBI View Article : Google Scholar

66 

Sun Y, Zheng Y, Li P, Wang D, Niu C, Gong Y, Huang R, Wang Z, Wang Z and Ran H: Evaluation of superparamagnetic iron oxide-polymer composite microcapsules for magnetic resonance-guided high-intensity focused ultrasound cancer surgery. BMC Cancer. 14(800)2014.PubMed/NCBI View Article : Google Scholar

67 

You Y, Wang Z, Ran H, Zheng Y, Wang D, Xu J, Wang Z, Chen Y and Li P: Nanoparticle-enhanced synergistic HIFU ablation and transarterial chemoembolization for efficient cancer therapy. Nanoscale. 8:4324–4339. 2016.PubMed/NCBI View Article : Google Scholar

68 

Ho VH, Smith MJ and Slater NK: Effect of magnetite nanoparticle agglomerates on the destruction of tumor spheroids using high intensity focused ultrasound. Ultrasound Med Biol. 37:169–175. 2011.PubMed/NCBI View Article : Google Scholar

69 

Dibaji SAR, Al-Rjoub MF, Myers MR and Banerjee RK: Enhanced heat transfer and thermal dose using magnetic nanoparticles during HIFU thermal ablation-an in-vitro study. J Nanotechnol Eng Med. 4(040902)2014.

70 

Devarakonda SB, Myers MR, Giridhar D, Dibaji SA and Banerjee RK: Enhanced thermal effect using magnetic nano-particles during high-intensity focused ultrasound. PLoS One. 12(e0175093)2017.PubMed/NCBI View Article : Google Scholar

71 

Devarakonda SB, Myers MR and Banerjee RK: Comparison of heat transfer enhancement between magnetic and gold nanoparticles during HIFU sonication. J Biomech Eng. 140:2018.PubMed/NCBI View Article : Google Scholar

72 

Kaczmarek K, Hornowski T, Kubovčíková M, Timko M, Koralewski M and Józefczak A: Heating induced by therapeutic ultrasound in the presence of magnetic nanoparticles. ACS Appl Mater Interfaces. 10:11554–11564. 2018.PubMed/NCBI View Article : Google Scholar

73 

Sadeghi-Goughari M, Jeon S and Kwon HJ: Magnetic nanoparticles-enhanced focused ultrasound heating: Size effect, mechanism, and performance analysis. Nanotechnology. 31(245101)2020.PubMed/NCBI View Article : Google Scholar

74 

Kimura NT, Taniguchi S, Aoki K and Baba T: Selective localization and growth of Bifidobacterium bifidum in mouse tumors following intravenous administration. Cancer Res. 40:2061–2068. 1980.PubMed/NCBI

75 

Li X, Fu GF, Fan YR, Liu WH, Liu XJ, Wang JJ and Xu GX: Bifidobacterium adolescentis as a delivery system of endostatin for cancer gene therapy: Selective inhibitor of angiogenesis and hypoxic tumor growth. Cancer Gene Ther. 10:105–111. 2003.PubMed/NCBI View Article : Google Scholar

76 

Luo CH, Huang CT, Su CH and Yeh CS: Bacteria-Mediated hypoxia-specific delivery of nanoparticles for tumors imaging and therapy. Nano Lett. 16:3493–3499. 2016.PubMed/NCBI View Article : Google Scholar

77 

Xu D, Zou W, Luo Y, Gao X, Jiang B, Wang Y, Jiang F, Xiong J, Chen C, Tang Y, et al: Feasibility between bifidobacteria targeting and changes in the acoustic environment of tumor tissue for synergistic HIFU. Sci Rep. 10(7772)2020.PubMed/NCBI View Article : Google Scholar

78 

Jiang BL, Gao X, Xiong J, Zhu PY, Luo Y, Xu D, Tang Y, Wang YT, Chen C, Yang HY, et al: Experimental study on synergistic effect of HIFU treatment of tumors using Bifidobacterium bound with cationic phase-change nanoparticles. Eur Rev Med Pharmacol Sci. 24:5714–5725. 2020.PubMed/NCBI View Article : Google Scholar

79 

Zhu C, Ji Z, Ma J, Ding Z, Shen J and Wang QW: Recent advances of nanotechnology-facilitated bacteria-based drug and gene delivery systems for cancer treatment. Pharmaceutics. 13(940)2021.PubMed/NCBI View Article : Google Scholar

80 

Yin T, Diao Z, Blum NT, Qiu L, Ma A and Huang P: Engineering bacteria and bionic bacterial derivatives with nanoparticles for cancer therapy. Small. 18(e2104643)2022.PubMed/NCBI View Article : Google Scholar

81 

Kalia VC, Patel SKS, Cho BK, Wood TK and Lee JK: Emerging applications of bacteria as antitumor agents. Semin Cancer Biol. 86(Pt 2):1014–1025. 2022.PubMed/NCBI View Article : Google Scholar

82 

Tang Y, Chen C, Jiang B, Wang L, Jiang F, Wang D, Wang Y, Yang H, Ou X, Du Y, et al: Bifidobacterium bifidum-Mediated specific delivery of nanoparticles for tumor therapy. Int J Nanomedicine. 16:4643–4659. 2021.PubMed/NCBI View Article : Google Scholar

83 

Wang D, Jiang F, Wang L, Tang Y, Zhang Z, Du Y and Zou J: Polyethylenimine (PEI)-modified poly (lactic-co-glycolic) acid (PLGA) nanoparticles conjugated with tumor-homing bacteria facilitate high intensity focused ultrasound-mediated tumor ablation. Biochem Biophys Res Commun. 571:104–109. 2021.PubMed/NCBI View Article : Google Scholar

84 

Tee JK, Yip LX, Tan ES, Santitewagun S, Prasath A, Ke PC, Ho HK and Leong DT: Nanoparticles' interactions with vasculature in diseases. Chem Soc Rev. 48:5381–5407. 2019.PubMed/NCBI View Article : Google Scholar

85 

Wang Y, Chen C, Luo Y, Xiong J, Tang Y, Yang H, Wang L, Jiang F, Gao X, Xu D, et al: Experimental study of tumor therapy mediated by multimodal imaging based on a biological targeting synergistic agent. Int J Nanomedicine. 15:1871–1888. 2020.PubMed/NCBI View Article : Google Scholar

86 

Han H, Lee H, Kim K and Kim H: Effect of high intensity focused ultrasound (HIFU) in conjunction with a nanomedicines-microbubble complex for enhanced drug delivery. J Control Release. 266:75–86. 2017.PubMed/NCBI View Article : Google Scholar

87 

Frazier N, Payne A, Dillon C, Subrahmanyam N and Ghandehari H: Enhanced efficacy of combination heat shock targeted polymer therapeutics with high intensity focused ultrasound. Nanomedicine. 13:1235–1243. 2017.PubMed/NCBI View Article : Google Scholar

88 

Li Q, Zhang J, Li J, Ye H, Li M, Hou W, Li H and Wang Z: Glutathione-Activated NO-/ROS-Generation nanoparticles to modulate the tumor hypoxic microenvironment for enhancing the effect of HIFU-Combined Chemotherapy. ACS Appl Mater Interfaces. 13:26808–26823. 2021.PubMed/NCBI View Article : Google Scholar

89 

Kang Y, Kim J, Park J, Lee YM, Saravanakumar G, Park KM, Choi W, Kim K, Lee E, Kim C and Kim WJ: Tumor vasodilation by N-Heterocyclic carbene-based nitric oxide delivery triggered by high-intensity focused ultrasound and enhanced drug homing to tumor sites for anti-cancer therapy. Biomaterials. 217(119297)2019.PubMed/NCBI View Article : Google Scholar

90 

Li H, Yu C, Zhang J, Li Q, Qiao H, Wang Z and Zeng D: pH-sensitive pullulan-doxorubicin nanoparticles loaded with 1,1,2-trichlorotrifluoroethane as a novel synergist for high intensity focused ultrasound mediated tumor ablation. Int J Pharm. 556:226–235. 2019.PubMed/NCBI View Article : Google Scholar

91 

Yildirim A, Shi D, Roy S, Blum NT, Chattaraj R, Cha JN and Goodwin AP: Nanoparticle-Mediated acoustic cavitation enables high intensity focused ultrasound ablation without tissue heating. ACS Appl Mater Interfaces. 10:36786–36795. 2018.PubMed/NCBI View Article : Google Scholar

92 

Yildirim A, Chattaraj R, Blum NT, Shi D, Kumar K and Goodwin AP: Phospholipid capped mesoporous nanoparticles for targeted high intensity focused ultrasound ablation. Adv Healthc Mater. 6:2017.PubMed/NCBI View Article : Google Scholar

93 

Yildirim A, Blum NT and Goodwin AP: Colloids, nanoparticles, and materials for imaging, delivery, ablation, and theranostics by focused ultrasound (FUS). Theranostics. 9:2572–2594. 2019.PubMed/NCBI View Article : Google Scholar

94 

Jain K and Zhong J: Theranostic applications of nanomaterials. Curr Pharm Des. 28(77)2022.PubMed/NCBI View Article : Google Scholar

95 

Hartl D, de Luca V, Kostikova A, Laramie J, Kennedy S, Ferrero E, Siegel R, Fink M, Ahmed S, Millholland J, et al: Translational precision medicine: An industry perspective. J Transl Med. 19(245)2021.PubMed/NCBI View Article : Google Scholar

96 

Sisodiya SM: Precision medicine and therapies of the future. Epilepsia. 62 (Suppl 2):S90–S105. 2021.PubMed/NCBI View Article : Google Scholar

97 

Li H, Zeng Y, Zhang H, Gu Z, Gong Q and Luo K: Functional gadolinium-based nanoscale systems for cancer theranostics. J Control Release. 329:482–512. 2021.PubMed/NCBI View Article : Google Scholar

98 

Kavros SJ and Coronado R: Diagnostic and therapeutic ultrasound on venous and arterial ulcers: A focused review. Adv Skin Wound Care. 31:55–65. 2018.PubMed/NCBI View Article : Google Scholar

99 

Meng Y, Pople CB, Budiansky D, Li D, Suppiah S, Lim-Fat MJ, Perry J, Sahgal A and Lipsman N: Current state of therapeutic focused ultrasound applications in neuro-oncology. J Neurooncol. 156:49–59. 2022.PubMed/NCBI View Article : Google Scholar

100 

Blum NT, Yildirim A, Chattaraj R and Goodwin AP: Nanoparticles formed by acoustic destruction of microbubbles and their utilization for imaging and effects on therapy by high intensity focused ultrasound. Theranostics. 7:694–702. 2017.PubMed/NCBI View Article : Google Scholar

101 

Zhu J, Li Z, Zhang C, Lin L, Cao S, Che H, Shi X, Wang H and van Hest JCM: Single enzyme loaded nanoparticles for combinational ultrasound-guided focused ultrasound ablation and hypoxia-relieved chemotherapy. Theranostics. 9:8048–8060. 2019.PubMed/NCBI View Article : Google Scholar

102 

Li C, Lu Y, Cheng L, Zhang X, Yue J and Liu J: Combining mechanical high-intensity focused ultrasound ablation with chemotherapy for augmentation of anticancer immune responses. Mol Pharm. 18:2091–2103. 2021.PubMed/NCBI View Article : Google Scholar

103 

Zhu S, Zhang T, Zheng L, Liu H, Song W, Liu D, Li Z and Pan CX: Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol. 14(156)2021.PubMed/NCBI View Article : Google Scholar

104 

Chen J, Tan Q, Yang Z and Jin Y: Engineered extracellular vesicles: Potentials in cancer combination therapy. J Nanobiotechnology. 20(132)2022.PubMed/NCBI View Article : Google Scholar

105 

Barisano G, Sepehrband F, Ma S, Jann K, Cabeen R, Wang DJ, Toga AW and Law M: Clinical 7 T MRI: Are we there yet? A review about magnetic resonance imaging at ultra-high field. Br J Radiol. 92(20180492)2019.PubMed/NCBI View Article : Google Scholar

106 

Lutz NW and Bernard M: Contactless Thermometry by MRI and MRS: Advanced methods for thermotherapy and biomaterials. iScience. 23(101561)2020.PubMed/NCBI View Article : Google Scholar

107 

Sparacia G and Sakai K: Temperature measurement by diffusion-weighted imaging. Magn Reson Imaging Clin N Am. 29:253–261. 2021.PubMed/NCBI View Article : Google Scholar

108 

Devarakonda SB, Myers MR, Lanier M, Dumoulin C and Banerjee RK: Assessment of gold nanoparticle-mediated-enhanced hyperthermia using MR-Guided high-intensity focused ultrasound ablation procedure. Nano Lett. 17:2532–2538. 2017.PubMed/NCBI View Article : Google Scholar

109 

Devarakonda SB, Stringer K, Rao M, Myers M and Banerjee R: Assessment of enhanced thermal effect due to gold nanoparticles during MR-Guided high-intensity focused ultrasound (HIFU) procedures using a mouse-tumor model. ACS Biomater Sci Eng. 5:4102–4111. 2019.PubMed/NCBI View Article : Google Scholar

110 

Kuai X, Zhu Y, Yuan Z, Wang S, Lin L, Ye X, Lu Y, Luo Y, Pang Z, Geng D and Yin B: Perfluorooctyl bromide nanoemulsions holding MnO2 nanoparticles with dual-modality imaging and glutathione depletion enhanced HIFU-eliciting tumor immunogenic cell death. Acta Pharm Sin B. 12:967–981. 2022.PubMed/NCBI View Article : Google Scholar

111 

Mattrey RF: Perfluorooctylbromide: A new contrast agent for CT, sonography, and MR imaging. AJR Am J Roentgenol. 152:247–252. 1989.PubMed/NCBI View Article : Google Scholar

112 

Li X, Sui Z, Li X, Xu W, Guo Q, Sun J and Jing F: Perfluorooctylbromide nanoparticles for ultrasound imaging and drug delivery. Int J Nanomedicine. 13:3053–3067. 2018.PubMed/NCBI View Article : Google Scholar

113 

Pellico J, Ellis CM and Davis JJ: Nanoparticle-based paramagnetic contrast agents for magnetic resonance imaging. Contrast Media Mol Imaging. 2019(1845637)2019.PubMed/NCBI View Article : Google Scholar

114 

Cai X, Zhu Q, Zeng Y, Zeng Q, Chen X and Zhan Y: Manganese oxide nanoparticles As MRI contrast agents in tumor multimodal imaging and therapy. Int J Nanomedicine. 14:8321–8344. 2019.PubMed/NCBI View Article : Google Scholar

115 

Das D, Sharma A, Rajendran P and Pramanik M: Another decade of photoacoustic imaging. Phys Med Biol. 66(5)2021.PubMed/NCBI View Article : Google Scholar

116 

Jacques SL: Optical properties of biological tissues: A review. Phys Med Biol. 58:R37–R61. 2013.PubMed/NCBI View Article : Google Scholar

117 

Wu D, Huang L, Jiang MS and Jiang H: Contrast agents for photoacoustic and thermoacoustic imaging: A review. Int J Mol Sci. 15:23616–23639. 2014.PubMed/NCBI View Article : Google Scholar

118 

Palma-Chavez J, Pfefer TJ, Agrawal A, Jokerst JV and Vogt WC: Review of consensus test methods in medical imaging and current practices in photoacoustic image quality assessment. J Biomed Opt. 26(090901)2021.PubMed/NCBI View Article : Google Scholar

119 

Steinberg I, Huland DM, Vermesh O, Frostig HE, Tummers WS and Gambhir SS: Photoacoustic clinical imaging. Photoacoustics. 14:77–98. 2019.PubMed/NCBI View Article : Google Scholar

120 

Brunker J, Yao JJ, Laufer J and Bohndiek SE: Photoacoustic imaging using genetically encoded reporters: A review. J Biomed Opt. 22:2017.PubMed/NCBI View Article : Google Scholar

121 

Feng G, Hao L, Xu C, Ran H, Zheng Y, Li P, Cao Y, Wang Q, Xia J and Wang Z: High-intensity focused ultrasound-triggered nanoscale bubble-generating liposomes for efficient and safe tumor ablation under photoacoustic imaging monitoring. Int J Nanomedicine. 12:4647–4659. 2017.PubMed/NCBI View Article : Google Scholar

122 

Attia ABE, Balasundaram G, Moothanchery M, Dinish US, Bi R, Ntziachristos V and Olivo M: A review of clinical photoacoustic imaging: Current and future trends. Photoacoustics. 16(100144)2019.PubMed/NCBI View Article : Google Scholar

123 

Gao H, Wang Z, Tan M, Liu W, Zhang L, Huang J, Cao Y, Li P, Wang Z, Wen J, et al: pH-Responsive nanoparticles for enhanced antitumor activity by high-intensity focused ultrasound therapy combined with sonodynamic therapy. Int J Nanomedicine. 17:333–350. 2022.PubMed/NCBI View Article : Google Scholar

124 

Chen Q, Qin W, Qi W and Xi L: Progress of clinical translation of handheld and semi-handheld photoacoustic imaging. Photoacoustics. 22(100264)2021.PubMed/NCBI View Article : Google Scholar

125 

Neprokin A, Broadway C, Myllylä T, Bykov A and Meglinski I: Photoacoustic Imaging in Biomedicine and Life Sciences. Life (Basel). 12(588)2022.PubMed/NCBI View Article : Google Scholar

126 

Yan S, Lu M, Ding X, Chen F, He X, Xu C, Zhou H, Wang Q, Hao L and Zou J: HematoPorphyrin Monomethyl Ether polymer contrast agent for ultrasound/photoacoustic dual-modality imaging-guided synergistic high intensity focused ultrasound (HIFU) therapy. Sci Rep. 6(31833)2016.PubMed/NCBI View Article : Google Scholar

127 

Zhang N, Cai X, Gao W, Wang R, Xu C, Yao Y, Hao L, Sheng D, Chen H, Wang Z and Zheng Y: A multifunctional theranostic nanoagent for dual-mode image-guided HIFU/Chemo-Synergistic cancer therapy. Theranostics. 6:404–417. 2016.PubMed/NCBI View Article : Google Scholar

128 

Park EY, Lee H, Han S, Kim C and Kim J: Photoacoustic imaging systems based on clinical ultrasound platform. Exp Biol Med (Maywood). 247:551–560. 2022.PubMed/NCBI View Article : Google Scholar

129 

Li Y, Hao L, Liu F, Yin L, Yan S, Zhao H, Ding X, Guo Y, Cao Y, Li P, et al: Cell penetrating peptide-modified nanoparticles for tumor targeted imaging and synergistic effect of sonodynamic/HIFU therapy. Int J Nanomedicine. 14:5875–5894. 2019.PubMed/NCBI View Article : Google Scholar

130 

Yang H, Jiang F, Zhang L, Wang L, Luo Y, Li N, Guo Y, Wang Q and Zou J: Multifunctional l-arginine-based magnetic nanoparticles for multiple-synergistic tumor therapy. Biomater Sci. 9:2230–2243. 2021.PubMed/NCBI View Article : Google Scholar

131 

Isoda K, Nagata R, Hasegawa T, Taira Y, Taira I, Shimizu Y, Isama K, Nishimura T and Ishida I: Hepatotoxicity and drug/chemical interaction toxicity of nanoclay particles in mice. Nanoscale Res Lett. 12(199)2017.PubMed/NCBI View Article : Google Scholar

132 

Dai X, Zeng Y, Zhang H, Gu Z, Gong Q and Luo K: Advances on Nanomedicines for Diagnosis and Theranostics of Hepatic Fibrosis. Adv Biomed Res. 1(2000091)2021.

133 

Federau C, Goubran M, Rosenberg J, Henderson J, Halpern CH, Santini V, Wintermark M, Butts Pauly K and Ghanouni P: Transcranial MRI-guided high-intensity focused ultrasound for treatment of essential tremor: A pilot study on the correlation between lesion size, lesion location, thermal dose, and clinical outcome. J Magn Reson Imaging. 48:58–65. 2018.PubMed/NCBI View Article : Google Scholar

134 

Huber PM, Afzal N, Arya M, Boxler S, Dudderidge T, Emberton M, Guillaumier S, Hindley RG, Hosking-Jervis F, Leemann L, et al: An exploratory study of dose escalation vs standard focal high-intensity focused ultrasound for treating nonmetastatic prostate cancer. J Endourol. 34:641–646. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2023
Volume 25 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zheng Q, Xia B, Huang X, Luo J, Zhong S and Li X: Nanomedicines for high‑intensity focused ultrasound cancer treatment and theranostics (Review). Exp Ther Med 25: 170, 2023
APA
Zheng, Q., Xia, B., Huang, X., Luo, J., Zhong, S., & Li, X. (2023). Nanomedicines for high‑intensity focused ultrasound cancer treatment and theranostics (Review). Experimental and Therapeutic Medicine, 25, 170. https://doi.org/10.3892/etm.2023.11869
MLA
Zheng, Q., Xia, B., Huang, X., Luo, J., Zhong, S., Li, X."Nanomedicines for high‑intensity focused ultrasound cancer treatment and theranostics (Review)". Experimental and Therapeutic Medicine 25.4 (2023): 170.
Chicago
Zheng, Q., Xia, B., Huang, X., Luo, J., Zhong, S., Li, X."Nanomedicines for high‑intensity focused ultrasound cancer treatment and theranostics (Review)". Experimental and Therapeutic Medicine 25, no. 4 (2023): 170. https://doi.org/10.3892/etm.2023.11869