Next Article in Journal
Anti-Proliferative Potential of Quercetin Loaded Polymeric Mixed Micelles on Rat C6 and Human U87MG Glioma Cells
Next Article in Special Issue
Reduction-Responsive Stearyl Alcohol-Cabazitaxel Prodrug Nanoassemblies for Cancer Chemotherapy
Previous Article in Journal
Fabrication and Optimization of Essential-Oil-Loaded Nanoemulsion Using Box–Behnken Design against Staphylococos aureus and Staphylococos epidermidis Isolated from Oral Cavity
Previous Article in Special Issue
A Comparative Study of Quercetin-Loaded Nanocochleates and Liposomes: Formulation, Characterization, Assessment of Degradation and In Vitro Anticancer Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy

1
Department of Urology, Peking University Third Hospital, Beijing 100191, China
2
Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2022, 14(8), 1642; https://doi.org/10.3390/pharmaceutics14081642
Submission received: 17 June 2022 / Revised: 31 July 2022 / Accepted: 4 August 2022 / Published: 6 August 2022
(This article belongs to the Special Issue Recent Developments and Emerging Trends in Nanomedicine)

Abstract

:
Chemotherapy has an essential role not only in advanced solid tumor therapy intervention but also in society’s health at large. Chemoresistance, however, seriously restricts the efficiency and sensitivity of chemotherapeutic agents, representing a significant threat to patients’ quality of life and life expectancy. How to reverse chemoresistance, improve efficacy sensitization response, and reduce adverse side effects need to be tackled urgently. Recently, studies on the effect of ultrasonic microbubble cavitation on enhanced tissue permeability and retention (EPR) have attracted the attention of researchers. Compared with the traditional targeted drug delivery regimen, the microbubble cavitation effect, which can be used to enhance the EPR effect, has the advantages of less trauma, low cost, and good sensitization effect, and has significant application prospects. This article reviews the research progress of ultrasound-mediated microbubble cavitation in the treatment of solid tumors and discusses its mechanism of action to provide new ideas for better treatment strategies.

1. Introduction

Despite the development and research of anti-tumor therapy having made obvious progress, we are compelled to attach great efforts to the drug resistance in patients undergoing systematic chemotherapy [1]. The occurrence of chemotherapy resistance has been directly related to the interaction between genetic factors, cellular heterogeneity, and tumor microenvironment (TME) [2,3,4]. Evidence demonstrated that the overall 5-year survival rate of cancer patients has increased from 49% in 1970 to the current 68% and the overall mortality rate of patients has decreased by 32% aroused by the continuous breakthroughs in medical technology and targeted drugs [5]. Disconcertingly, the gradual improvement in the survival period and mortality of cancer patients promotes the continuous prolongation of the anti-tumor treatment courses, which greatly aggravates the increase in anti-tumor drug resistance and toxic side effects with a serious threat to life quality and prognosis [6,7,8]. Therefore, how to reverse drug resistance, improve side effects, and promote chemotherapy sensitization effects are the key issues to solve urgently.
Most of the anti-tumor drugs and delivery vehicles used in current research do not have tumor selectivity themselves. It is difficult to obtain a satisfactory drug accumulation effect at tumor sites only by inherent EPR effect through passive targeted transport that ultimately leads to inefficient drug efficiency and serious side effects in clinical trials [9]. In recent years, researchers have proposed a variety of methods to facilitate the targeted delivery of drugs to tumor sites. Among them, the ultrasound-mediated microbubble cavitation effect has attracted the attention of many researchers. Using ultrasound to mediate the cavitation effect of microbubbles can enhance tissue and vascular permeability and targeted drug retention, which is an effective way to improve the EPR effect. Compared with the traditional targeted drug delivery scheme, the microbubble cavitation effect has huge advantages due to its smaller trauma, lower cost, and better sensitization effect [10] with significant application prospects. This review summarizes the progress of the ultrasound-mediated microbubble cavitation effect in solid tumor therapy by effectively improving drug delivery obstacles via the EPR effect in TME, discusses the potential mechanisms of microbubble cavitation enhancing the EPR effect, and ultimately describes the new ideas based on the therapeutic research in vitro and in vivo for better therapeutic strategies.

2. EPR Effect in the TME

2.1. Chemotherapy Obstacles Aroused by Aberrant Angiogenesis in TME

Although many chemotherapy agents have ideal effectiveness on tumor cells cultured in vitro, the unique aberrant angiogenesis in TME of tumors in vivo has multiple obstacles that restrict drug efficacy; thus, the provision of traditional chemotherapy is suboptimal [11,12,13,14,15]. The solid TME consists of tumor cells, stromal cells, and secreted cytokines. Compared with normal tissue cells under stable conditions maintained by numerous regulatory factors, tumor cells have characteristics of rapid proliferation, cell cycle disorders, and immune escape [16,17]. Vascular development in TME is necessary for the survival and progression of tumor cells and has an important impact on the remodeling of the TME, and provides a conducive environment for tumor growth and invasion [18,19]. With the rapid proliferation of tumor cells, various angiogenesis regulators are secreted in TME to induce the generation of a neovascular network [18,20]. Vascular development mainly includes three processes: angiogenesis, neovascularization, and vascular remodeling [21,22,23]. Due to the overexpression of pro-angiogenic factors induced by rapid tumor proliferation, tumor neovascularization with abnormal structure and function could not achieve normal oxygen supply of tissue cells leading to a TME surrounded by hypoxia and ischemia [24,25,26]. Remolded TME will further aggravate angiogenesis abnormality and increase intra-tumoral fluid pressure causing reduced blood perfusion supply, eventually inhibiting immune cells from exerting anti-tumor efficiency and impeding chemotherapeutic drug diffusion [25,27,28,29,30]. Meanwhile, imperfect vascularization causing disorder distribution and irregular anastomotic branches combined with discontinuous vascular structure connections in the tumor could induce the lost blood–tumor barrier function, weaken vessel stability, and impair blood perfusion regulation [31,32,33]. In addition, the discontinuous structure between the basement membrane with pericytes and endothelial cells results in leakage and inefficiency in blood perfusion [27,29]. In the central region of the tumor, intra-tumoral high interstitial fluid pressure (IFP) can lead to vascular perfusion decrease, hypoxic state, and lymphatic vessel compression [14,34,35,36]. Thus, immature tumor vessels in TME have characteristics of vascular leakage, decreased blood perfusion, structure connection impairment, barrier dysfunction, and drug diffusion diminishment. Additionally, the increased vascular leakage without effective lymphatic drainage inside the tumor eventually leads to a significant increase in IFP in the tumor, which dramatically hinders the diffusion of therapeutic drugs in the interstitial space [37,38,39,40,41]. In addition, with the rapid proliferation of tumoral cells, the increased dispersal distance between tumoral cells and neovessels further hinders the delivery of chemotherapeutic drugs from blood vessels into interstitium, leading to exacerbating the formation of chemotherapy resistance [42,43,44].

2.2. Therapeutic Strategy Based on the EPR Effect

The TME in solid tumors presents several disorders with vascular tortuosity, angiogenesis, and hypoxia [45,46,47]. Meanwhile, abnormal tissue vasculature, stromal matrix, and interstitial fluid pressure are major limitations preventing drug penetration in solid tumors [34,48]. Therefore, therapeutic agents transported into tumor lesions in the accurate and efficient delivery is crucial for effectively addressing drug delivery in TME which remains a major challenge to date. Fortunately, the lack of vascular supporting tissue in tumors results in leaky vessels and poor lymphatic drainage is a structural basis for enhancing the effect of EPR [49]. The EPR effect in preclinical research with passive targeted delivery systems has shown significant enhancement in anticancer efficacy compared with traditional chemotherapeutics [50,51,52]. However, due to tumor heterogeneity with aberrant vascular permeability, phenotypic mutation, and stromal abnormalities hinder the successful application of EPR-based therapy [53,54,55,56,57]. Therefore, precision therapy and complementary enhancement of EPR to overcome chemotherapy resistance challenges aroused by aberrant angiogenesis in TME are crucial for the development of personalized EPR-mediated solid tumor therapy [58].

3. Ultrasonic Microbubble Cavitation Enhancing the EPR Effect on Tumor Therapy

3.1. Mechanical Effects of Ultrasonic Microbubble Cavitation

The ultrasound-mediated microbubble cavitation effect refers to a series of dynamic processes such as expansion, contraction, oscillation, and violent collapse in tiny bubbles under ultrasound sonication at a specific frequency [59,60,61]. The physical phenomena with high temperature, high pressure, and micro-jet are induced by rapid release of instantaneous energy accompanying ultrasound intervention in bubbles after energy absorption [62,63]. The acoustic cavitation effect is commonly used to improve cancer therapy for cancers such as hepatocellular carcinoma, colon cancer, brain cancers, and so on, in clinical research [64,65,66]. When ultrasound is applied to the current and emerging technique on diagnosis and treatment, the cavitation effect can improve EPR in terms of both drug release and biological effects. During the process of agent release, ultrasound can stimulate the carrier to enhance the efficiency of drug release and distribution in tissues. In terms of biological effects, the use of acoustic energy combined with the cavitation effect of microbubbles is mainly to promote the vascular permeability and extracellular material transport by membranes in TME [67,68,69].
When exposed to an ultrasound field, the liquid around the microbubbles will form acoustic streaming, in which the shear stress generated by the streaming can promote the directional release of the drug from microbubbles and enhance the permeability of the drug in tissues and cell membranes (shear-induced permeability) [68]. These mechanical effects primarily generated by ultrasound may lead to the improvement in permeability and vascular perfusion into solid tumor tissues [70].

3.2. Ultrasonic Microbubble Cavitation Promoting Tumor Therapy by Enhancing the EPR Effect

Microbubbles can present distinct oscillation patterns when acoustic parameters are varied. The stable ultrasonic cavitation effect of microbubbles is usually generated under relatively low peak negative acoustic pressures. When the acoustic pressure amplitudes were further increased, ultrasonic cavitation turned into a violent collapse called inertial cavitation accompanied by shock waves and microstreaming [71]. These effects of cavitation have been extensively applied in various domains of medical applications [72,73,74,75].
Ultrasound-mediated microbubble cavitation can enhance EPR by improving the permeability of the biological barriers in TME through a local controllable cavitation effect, enhance material exchange and transport, and achieve the therapeutic effect of increasing drug concentration in tissue cells (Figure 1). The hydrophobic microbubbles encapsulated by lipid, protein, or polymer shell gas are irradiated by ultrasound to expand, contract, oscillate, and even violently collapse, a process called cavitation. Cavitation can be divided into two forms, namely steady-state cavitation and inertial cavitation. Steady-state cavitation refers to the stable vibration of microbubbles around the resonant diameter at low sound intensities. As the sound intensity increases, the microbubbles expand, contract, or collapse more violently, generating shock waves and microjets near the microbubbles, a process called inertial cavitation [76]. In recent decades, numerous studies have demonstrated that ultrasound-mediated cavitation of microbubbles can facilitate the delivery of anticancer drugs to tumor cells [77,78]. Microbubbles are composed of less than 100 nm layers of polymers, proteins, and lipids covering the surface of a hydrophobic fluorinated gas. To increase tumor tissue specificity, ligands for specific cell surface receptors can be attached to the micro-vesicles. Due to the sound pressure of ultrasonic waves, the microbubbles will shrink and expand periodically. When the sound pressure reaches a certain threshold, the microbubble collapses [79]. The cavitation or explosion of microbubbles will form temporary holes in the cell membrane and blood vessel wall to enhance permeability, providing temporary and reversible channels for the transport of substances, so that therapy agents can enter the cell passively [80,81]. Studies have shown that ultrasound could not only enhance the passive diffusion of drug, but also affect the active transport for enhancing drug uptake. Ultrasound cavitation is thought to induce changes in ion channels and molecular reaction including membrane resealing or gap restoration in different spatial and temporal scales, resulting in increased intracellular Ca2+ concentration and cytoskeletal rearrangement [82]. These changes discussed above could play a crucial role in stimulating the clathrin-mediated endocytosis pathway to promote drug diffusion into cells [83]. In addition, fluid movement caused by cavitation may also facilitate vascular perfusion according to vasodilator expression including nitric oxide induced by increased intracellular Ca2+ concentration and high shear stress from oscillating bubbles, increasing the amount of drug agent uptake by distant tumor cells [83,84,85].
In recent years, many studies have also confirmed that claudins and ZO-1 play an important role in the permeability regulation of biological barriers such as the blood–tumor barrier [86,87,88,89,90]. Studies have shown that there are a series of intercellular junctions between endothelial or epithelial cells, of which tight junctions are the most important. Between mammalian cells, tight junctions are mainly composed of transmembrane proteins (Occludin), claudins, junctional adhesive molecules (JAMs), and cytoplasmic attachment proteins (ZO family) [91,92,93]. Tight junctions widely exist in biological barrier structures such as intestinal mucosal epithelial cells, interstitial cells, the blood–testis barrier, and the blood–brain barrier [94,95,96,97]. Through research, Bae et al. found that the permeability of tight junctions in the barrier structure increased after physical treatment of the biological barriers, and the expression and distribution of the main components of tight junctions changed [98]. Through research, it was found that after intravenous injection of contrast agent microbubbles, low-frequency ultrasound irradiation can significantly increase the drug concentration in the tissue, and 24 h after the ultrasound irradiation, the drug concentration was significantly reduced, and tissue cells were observed by transmission electron microscopy. The gap between them widened and recovered after 24 h. Changes in the spatial structure of tight junctions are temporally consistent with changes in tissue barrier permeability. This indicates that tight junctions play an important role in the regulation of tissue cell permeability. Studies have shown that the tight junction protein Occludin plays an important role in the process of low-frequency ultrasound irradiation combined with contrast agent microbubbles to open the blood–tumor barrier (Figure 2). When the blood–tumor barrier is opened, the expression level of this tight junction protein decreases [99]. This indicates that the tight junction protein Occludin plays an important role in enhancing the permeability of tissue cells by low-frequency ultrasound irradiation combined with contrast agent microbubbles.
Overall, ultrasound can utilize the local microbubble cavitation to enhance the EPR effect for non-invasive targeted therapy of diseases without affecting the surrounding soft tissues. The cavitation effect can achieve the passive agents’ diffusion in tissue through ameliorating the permeability of tissue and vascular barrier by sonoporation and regulate the intercellular tight junction. Similarly, it can also enhance intracellular uptake via stimulating the clathrin-mediated endocytosis pathway induced by ion channels. On the one hand, the cavitation effect can also enhance the drug delivery efficiency by inducing the releasing vasodilator to a certain extent to increase local tissue blood perfusion. The feasibility and potential of this approach might contribute to achieve better targeted delivery in the prospective fast-revolutionizing disease area.

3.3. Application Studies Using Ultrasonic Microbubble Cavitation on Tumor Therapy

Many studies have confirmed that ultrasound-mediated microbubble cavitation can promote drug diffusion and induce tumor-suppressive effects by enhancing EPR through improving permeability and vascular perfusion in vitro (Table 1) and in vivo (Table 2), manifested as tumor growth inhibition, increased tumor cell apoptosis and necrosis, decreased tumor angiogenesis, and decreased expression of tumor-associated proteins. Nevertheless, a profusion of adverse effects has also been reported including hemorrhage, thrombus formation, local burns, tissue necrosis, and various organ toxicities [100,101,102]. Thankfully, most of the serious side effects of ultrasound-assisted therapy including necrosis and hemorrhage, are concentrated in a relatively high intensity focused ultrasound which mainly exerts an acoustic thermal effect rather than cavitation effect. Thus, keeping the ultrasonic waves under a lower intensity level and shorter intervention time could induce the controllable cavitation effect without obvious cell death and thermal damage [103]. Overall, low-intensity ultrasound is a relatively safe non-invasive intervention strategy. It is worthwhile to expect that flexible regulation of ultrasonic intervention parameters and better protocol design can further improve cavitation effect efficacy and potential risk aversion [104].
Meanwhile, complex models are crucial to represent the in vivo TME better which can provide a unique opportunity to study cellular interactions and biophysical mechanisms involved which are difficult to replicate in vitro due to lacking intricate intracellular and intercellular signaling pathways. It is therefore important to use more experimental efforts to comprehend the inherent differences between in vitro and in vivo that will affect microbubble behavior for exploring effective treatment interventions.

4. Challenges and Perspectives of Ultrasonic Microbubble Cavitation in Tumor Therapy

The TME is a complex and variable milieu in which it is a key point of efficacious therapy to overcome obstacles existing in solid tumors for successfully targeting therapeutic agents to the tumor site. In recent years, a variety of tumor-targeted drug delivery systems, including nanogels, liposomes, microbubbles, magnetic nanomaterials, etc., have been researched and developed to enhance tissue permeability and facilitate drug diffusion for antineoplastic therapy. This review focuses on the therapeutic role of ultrasound-mediated microbubble cavitation by enhancing TME permeability and promoting drug diffusion in solid tumors. Although these applications in this sector have realized significant progress, there are still many difficulties and challenges which require further efforts to explore more suitable delivery systems and effective efficacy.
Firstly, the particle size of microbubbles remains a key factor affecting localized drug accumulation and cavitation effects in tumors. Currently, many researchers choose to encapsulate the therapeutic agents inside the microbubble for controllable and targeted release. Studies have shown that liposomes with particle sizes from 100 to 200 nm can accumulate agent concentration four times greater than particle sizes smaller than 50 nm or larger than 300 nm in tumors [118]. Therefore, we need to develop a new process to solve the situation that the cavitation effect is weakened due to the low accumulation of microbubbles around the tumor tissue caused by the unsuitable particle size of microbubbles.
Secondly, with the emergence of multifunctional drug delivery systems, the structure with membrane shells continues to present complex trends. We need to reduce adverse effects of membrane shell material, particle size, and targeted modification type on the cavitation effect in the complex and variable TME, and optimize the best-suited parameters of different drug-loading systems in realizing the cavitation effect for achieving the controllability and stability of the targeted microbubble-loading system between different individuals with finally attaining the standardized application in clinical intervention.
Of course, due to the different research directions of each scholar, there are also differences in the parameters they used. The optimization of the parameters used for the ultrasound-mediated microbubble cavitation effect makes it very important to apply this technology to the clinic. Therefore, we need more efforts to verify the effects of ultrasound intervention time, irradiation time interval, ultrasound frequency, sound intensity, microbubble size, drug dose, and concentration on the therapeutic efficacy of the disease.
Meanwhile, the integration of multiple technologies needs to be carried out, including protein-membrane-targeted modification, photothermal, magnetic field, radiation, free radicals, gene interference, immunotherapy, etc., to comprehensively enhance the anti-tumor efficacy.
Absolutely, the safety of antitumor drugs is also an issue that closer attention should be paid to. Ultrasound-mediated microbubble cavitation accompanied by tissue microvascular damage and thrombosis could cause systematic tissue adverse effects, similarly, the toxicity of intermediate metabolites and degradation products of the microbubble-loading system should also attract our attention. Although many studies have confirmed the high histocompatibility of microbubbles, more research is still needed in the future to further confirm the potential harm caused by long-term accumulation in the body.
At present, there are still many problems to be solved in the treatment of tumors with low-frequency ultrasound combined with microbubbles, but it is undeniable that this technology has shown great clinical application value as a safe, effective, easy-to-operate, and targeted non-invasive treatment method. With the development of technology, this promising non-invasive tumor treatment method will be widely used in clinical practice.

5. Conclusions

The development of tumor therapy relies heavily on the development of non-invasive drug delivery methods to efficiently and selectively deliver drugs to target cells while minimizing the systemic toxicity of drugs, which is especially the focus of future research. Microbubble-mediated ultrasonic cavitation has unique advantages in the study of enhanced drug delivery to improve the efficiency of tumor treatment. The application of ultrasound has certain advantages, such as good tolerance, non-toxicity, easy operation and implementation, and low dependence on instruments and equipment. Many studies have confirmed that thermal and cavitation effects play a major role in various treatments; however, researchers should be aware that the anti-tumor effects of various ultrasound-mediated treatments are produced by a combination of multiple biological effects. Meanwhile, the safety issues generated by ultrasonic cavitation with EPR effects enhancement including toxicity and tissue damage still need to be fully investigated as a priority for future clinical applications.
Although both diagnostic ultrasound and contrast agents are generally recognized as safe and approved for widespread use in clinical diagnosis, the combination of ultrasound and microbubble therapy has some limitations. Recently, several preclinical studies have used small animals to evaluate the safety of ultrasound combined with microbubbles using parameters such as body weight, dietary habits, and mobility [119,120]. However, the specific mechanism of the synergistic effect of ultrasonic cavitation combined with chemotherapeutic drugs is still unclear, the current sample size of relevant clinical studies is small, and the efficacy and safety of combined therapy need more studies with a larger sample size for support. Before the new technology of microbubble-enhanced ultrasonic cavitation combined with drug therapy for tumors is widely used in the clinic, more systematic and comprehensive side effects studies on various treatment options are still needed.
With the further development of molecular targeting technology, the preparation of more new microbubbles, the exploration of new treatment modes, the development of new ultrasonic equipment, and the optimization of new treatment procedures, will provide an opportunity for the treatment of tumors by microbubble-enhanced cavitation combined with drugs. This new technology brings more possibilities and brings more benefits to clinical oncology treatment programs.

Author Contributions

J.H., Z.L., H.X. and X.Z. wrote the manuscript and prepared the figures. H.G. revised the manuscript. J.L. conceived and designed the study, reviewed and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Beijing Natural Science Foundation (L212051, Z200027).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Crucitta, S.; Cucchiara, F.; Mathijssen, R.; Mateo, J.; Jager, A.; Joosse, A.; Passaro, A.; Attili, I.; Petrini, I.; van Schaik, R.; et al. Treatment-driven tumour heterogeneity and drug resistance: Lessons from solid tumours. Cancer Treat Rev. 2022, 104, 102340. [Google Scholar] [CrossRef] [PubMed]
  2. Adhikary, S.; Roy, S.; Chacon, J.; Gadad, S.S.; Das, C. Implications of Enhancer Transcription and eRNAs in Cancer. Cancer Res. 2021, 81, 4174–4182. [Google Scholar] [CrossRef]
  3. Pranzini, E.; Pardella, E.; Paoli, P.; Fendt, S.M.; Taddei, M.L. Metabolic Reprogramming in Anticancer Drug Resistance: A Focus on Amino Acids. Trends Cancer 2021, 7, 682–699. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, Z.; Yue, P.; Lu, T.; Wang, Y.; Wei, Y.; Wei, X. Role of lysosomes in physiological activities, diseases, and therapy. J. Hematol. Oncol. 2021, 14, 79. [Google Scholar] [CrossRef] [PubMed]
  5. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  6. Ding, B.; Lou, W.; Xu, L.; Fan, W. Non-coding RNA in drug resistance of hepatocellular carcinoma. Biosci. Rep. 2018, 38, BSR20180915. [Google Scholar] [CrossRef] [PubMed]
  7. Chen, M.; Juengpanich, S.; Li, S.; Topatana, W.; Lu, Z.; Zheng, Q.; Cao, J.; Hu, J.; Chan, E.; Hou, L.; et al. Bortezomib-Encapsulated Dual Responsive Copolymeric Nanoparticles for Gallbladder Cancer Targeted Therapy. Adv. Sci. 2022, 9, e2103895. [Google Scholar] [CrossRef] [PubMed]
  8. Nam, H.J.; Kim, Y.E.; Moon, B.S.; Kim, H.Y.; Jung, D.; Choi, S.; Jang, J.W.; Nam, D.H.; Cho, H. Azathioprine antagonizes aberrantly elevated lipid metabolism and induces apoptosis in glioblastoma. iScience 2021, 24, 102238. [Google Scholar] [CrossRef]
  9. Yoshikawa, T.; Mori, Y.; Feng, H.; Phan, K.Q.; Kishimura, A.; Kang, J.H.; Mori, T.; Katayama, Y. Rapid and continuous accumulation of nitric oxide-releasing liposomes in tumors to augment the enhanced permeability and retention (EPR) effect. Int. J. Pharm. 2019, 565, 481–487. [Google Scholar] [CrossRef]
  10. Xia, H.; Yang, D.; He, W.; Zhu, X.; Yan, Y.; Liu, Z.; Liu, T.; Yang, J.; Tan, S.; Jiang, J.; et al. Ultrasound-mediated microbubbles cavitation enhanced chemotherapy of advanced prostate cancer by increasing the permeability of blood-prostate barrier. Transl. Oncol. 2021, 14, 101177. [Google Scholar] [CrossRef]
  11. Zhu, D.J.; Li, Y.; Zhang, Z.J.; Xue, Z.Y.; Hua, Z.L.; Luo, X.Y.; Zhao, T.; Lu, C.; Liu, Y.Y. Recent advances of nanotechnology-based tumor vessel-targeting strategies. J. Nanobiotechnol. 2021, 19, 435. [Google Scholar] [CrossRef] [PubMed]
  12. Yu, W.Q.; Lin, R.Y.; He, X.Q.; Yang, X.T.; Zhang, H.L.; Hu, C.; Liu, R.; Huang, Y.; Qin, Y.; Gao, H.L. Self-propelled nanomotor reconstructs tumor microenvironment through synergistic hypoxia alleviation and glycolysis inhibition for promoted anti-metastasis. Acta. Pharm. Sin. B 2021, 11, 2924–2936. [Google Scholar] [CrossRef] [PubMed]
  13. Cheng, Y.Q.; Wang, S.B.; Liu, J.H.; Jin, L.; Liu, Y.; Li, C.Y.; Su, Y.R.; Liu, Y.R.; Sang, X.; Wan, Q.; et al. Modifying the tumour microenvironment and reverting tumour cells: New strategies for treating malignant tumours. Cell Proliferat 2020, 53, e12865. [Google Scholar] [CrossRef]
  14. Majidpoor, J.; Mortezaee, K. Angiogenesis as a hallmark of solid tumors-clinical perspectives. Cell Oncol. 2021, 44, 715–737. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, S.D.; Li, Y.T.; Xing, C.; Ding, C.; Zhang, H.Y.; Chen, L.X.; You, L.; Dai, M.H.; Zhao, Y.P. Tumor microenvironment in chemoresistance, metastasis and immunotherapy of pancreatic cancer. Am. J. Cancer Res. 2020, 10, 1937–1953. [Google Scholar] [PubMed]
  16. Liu, H.Q.; Shi, Y.; Qian, F. Opportunities and delusions regarding drug delivery targeting pancreatic cancer-associated fibroblasts. Adv. Drug Deliver Rev. 2021, 172, 37–51. [Google Scholar] [CrossRef] [PubMed]
  17. Vitale, I.; Shema, E.; Loi, S.; Galluzzi, L. Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat. Med. 2021, 27, 212–224. [Google Scholar] [CrossRef]
  18. Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Zhang, S.; Gong, Z.; Li, X.; Cao, K.; Deng, H.; He, Y.; et al. The role of microenvironment in tumor angiogenesis. J. Exp. Clin. Cancer Res. 2020, 39, 204. [Google Scholar] [CrossRef]
  19. Wei, F.; Wang, D.; Wei, J.; Tang, N.; Tang, L.; Xiong, F.; Guo, C.; Zhou, M.; Li, X.; Li, G.; et al. Metabolic crosstalk in the tumor microenvironment regulates antitumor immunosuppression and immunotherapy resisitance. Cell Mol. Life Sci. 2021, 78, 173–193. [Google Scholar] [CrossRef] [PubMed]
  20. Larionova, I.; Kazakova, E.; Gerashchenko, T.; Kzhyshkowska, J. New Angiogenic Regulators Produced by TAMs: Perspective for Targeting Tumor Angiogenesis. Cancers 2021, 13, 3253. [Google Scholar] [CrossRef]
  21. Herbert, S.P.; Stainier, D.Y. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat. Rev. Mol. Cell Biol. 2011, 12, 551–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Matuszewska, K.; Pereira, M.; Petrik, D.; Lawler, J.; Petrik, J. Normalizing Tumor Vasculature to Reduce Hypoxia, Enhance Perfusion, and Optimize Therapy Uptake. Cancers 2021, 13, 4444. [Google Scholar] [CrossRef]
  23. Meng, Y.M.; Jiang, X.; Zhao, X.B.; Meng, Q.; Wu, S.Q.; Chen, Y.T.; Kong, X.Z.; Qiu, X.Y.; Su, L.P.; Huang, C.; et al. Hexokinase 2-driven glycolysis in pericytes activates their contractility leading to tumor blood vessel abnormalities. Nat. Commun. 2021, 12, 6011. [Google Scholar] [CrossRef] [PubMed]
  24. Ando, Y.; Oh, J.M.; Zhao, W.; Tran, M.; Shen, K.Y. Engineering a Vascularized Hypoxic Tumor Model for Therapeutic Assessment. Cells 2021, 10, 2201. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, W.Z.; Shen, L.S.; Jiang, J.X.; Zhang, L.Y.; Zhang, Z.G.; Pan, J.; Ni, C.; Chen, Z.G. Antiangiogenic therapy reverses the immunosuppressive breast cancer microenvironment. Biomark. Res. 2021, 9, 59. [Google Scholar] [CrossRef]
  26. Sun, C.X.; Gao, S.Q.; Tan, Y.Z.; Zhang, Z.H.; Xu, H.P. Side-Chain Selenium-Grafted Polymers Combining Antiangiogenesis Treatment with Photodynamic Therapy and Chemotherapy. ACS Biomater. Sci. Eng. 2021, 7, 3201–3208. [Google Scholar] [CrossRef] [PubMed]
  27. Martin, J.D.; Miyazaki, T.; Cabral, H. Remodeling tumor microenvironment with nanomedicines. Wires Nanomed. Nanobiotechnol. 2021, 13, e1730. [Google Scholar] [CrossRef] [PubMed]
  28. Koyuncu, B.; Melek, A.; Yilmaz, D.; Tuzer, M.; Unlu, M.B. Chemotherapy response prediction with diffuser elapser network. Sci. Rep. 2022, 12, 1628. [Google Scholar] [CrossRef]
  29. Viallard, C.; Larrivee, B. Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis 2017, 20, 409–426. [Google Scholar] [CrossRef]
  30. Mattheolabakis, G.; Mikelis, C.M. Nanoparticle Delivery and Tumor Vascular Normalization: The Chicken or The Egg? Front. Oncol. 2019, 9, 1227. [Google Scholar] [CrossRef] [Green Version]
  31. Forget, A.; Gianni-Barrera, R.; Uccelli, A.; Sarem, M.; Kohler, E.; Fogli, B.; Muraro, M.G.; Bichet, S.; Aumann, K.; Banfi, A.; et al. Mechanically Defined Microenvironment Promotes Stabilization of Microvasculature, Which Correlates with the Enrichment of a Novel Piezo-1(+) Population of Circulating CD11b(+)/CD115(+) Monocytes. Adv. Mater. 2019, 31, e1808050. [Google Scholar] [CrossRef] [PubMed]
  32. Mpekris, F.; Voutouri, C.; Baish, J.W.; Duda, D.G.; Munn, L.L.; Stylianopoulos, T.; Jain, R.K. Combining microenvironment normalization strategies to improve cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3728–3737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Li, W.; Li, X.; Liu, S.H.; Yang, W.D.; Pan, F.; Yang, X.Y.; Du, B.; Qin, L.; Pan, Y.L. Gold nanoparticles attenuate metastasis by tumor vasculature normalization and epithelial-mesenchymal transition inhibition. Int. J. Nanomed. 2017, 12, 3509–3520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Nakamura, Y.; Mochida, A.; Choyke, P.L.; Kobayashi, H. Nanodrug Delivery: Is the Enhanced Permeability and Retention Effect Sufficient for Curing Cancer? Bioconjug. Chem. 2016, 27, 2225–2238. [Google Scholar] [CrossRef]
  35. Fang, C.; Cen, D.; Wang, Y.F.; Wu, Y.J.; Cai, X.J.; Li, X.; Han, G.R. ZnS@ZIF-8 core-shell nanoparticles incorporated with ICG and TPZ to enable H 2 S-amplified synergistic therapy. Theranostics 2020, 10, 7671–7682. [Google Scholar] [CrossRef] [PubMed]
  36. Papageorgis, P.; Polydorou, C.; Mpekris, F.; Voutouri, C.; Agathokleous, E.; Kapnissi-Christodoulou, C.P.; Stylianopoulos, T. Tranilast-induced stress alleviation in solid tumors improves the efficacy of chemo- and nanotherapeutics in a size-independent manner. Sci. Rep. 2017, 7, 46140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Walker-Samuel, S.; Roberts, T.A.; Ramasawmy, R.; Burrell, J.S.; Johnson, S.P.; Siow, B.M.; Richardson, S.; Calves, M.R.G.C.; Pendse, D.; Robinson, S.P.; et al. Investigating Low-Velocity Fluid Flow in Tumors with Convection-MRI. Cancer Res. 2018, 78, 1859–1872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Qian, X.P.; Ge, L.; Yuan, K.J.; Li, C.; Zhen, X.; Cai, W.B.; Cheng, R.S.; Jiang, X.Q. Targeting and microenvironment-improving of phenylboronic acid-decorated soy protein nanoparticles with different sizes to tumor. Theranostics 2019, 9, 7417–7430. [Google Scholar] [CrossRef] [PubMed]
  39. Duan, L.; Yang, L.; Jin, J.; Yang, F.; Liu, D.; Hu, K.; Wang, Q.X.; Yue, Y.B.; Gu, N. Micro/nano-bubble-assisted ultrasound to enhance the EPR effect and potential theranostic applications. Theranostics 2020, 10, 462–483. [Google Scholar] [CrossRef]
  40. Timbie, K.F.; Afzal, U.; Date, A.; Zhang, C.; Song, J.; Miller, G.W.; Suk, J.S.; Hanes, J.; Price, R.J. MR image-guided delivery of cisplatin-loaded brain-penetrating nanoparticles to invasive glioma with focused ultrasound. J. Control. Release 2017, 263, 120–131. [Google Scholar] [CrossRef]
  41. Haider, T.; Sandha, K.K.; Soni, V.; Gupta, P.N. Recent advances in tumor microenvironment associated therapeutic strategies and evaluation models. Mat. Sci. Eng. C Mater. 2020, 116, 111229. [Google Scholar] [CrossRef] [PubMed]
  42. Zhou, K.; Zhang, J.W.; Wang, Q.Z.; Liu, W.Y.; Liu, J.L.; Yao, L.; Cai, M.M.; Ni, S.Y.; Cai, Q.Y.; Wang, G.J.; et al. Apatinib, a selective VEGFR2 inhibitor, improves the delivery of chemotherapeutic agents to tumors by normalizing tumor vessels in LoVo colon cancer xenograft mice. Acta Pharm. Sin. 2019, 40, 556–562. [Google Scholar] [CrossRef]
  43. Ding, H.Y.; Sun, J.; Song, Y.; Xin, W.X.; Zhu, J.F.; Zhong, L.K.; Chen, Y.B.; Zhang, Y.W.; Tong, Y.H.; Fang, L. Long Distance From Microvessel to Cancer Cell Predicts Poor Prognosis in Non-Small Cell Lung Cancer Patients. Front. Oncol. 2021, 11, 632352. [Google Scholar] [CrossRef]
  44. Mascheroni, P.; Penta, R. The role of the microvascular network structure on diffusion and consumption of anticancer drugs. Int. J. Numer. Methods Biomed. Eng. 2017, 33, e2857. [Google Scholar] [CrossRef] [Green Version]
  45. Papetti, M.; Herman, I.M. Mechanisms of normal and tumor-derived angiogenesis. Am. J. Physiol. Cell Physiol. 2002, 282, C947–C970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ziyad, S.; Iruela-Arispe, M.L. Molecular mechanisms of tumor angiogenesis. Genes Cancer 2011, 2, 1085–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Fukumura, D.; Duda, D.G.; Munn, L.L.; Jain, R.K. Tumor microvasculature and microenvironment: Novel insights through intravital imaging in pre-clinical models. Microcirculation 2010, 17, 206–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Kobayashi, H.; Turkbey, B.; Watanabe, R.; Choyke, P.L. Cancer drug delivery: Considerations in the rational design of nanosized bioconjugates. Bioconjug. Chem. 2014, 25, 2093–2100. [Google Scholar] [CrossRef] [Green Version]
  49. Tosetti, F.; Ferrari, N.; De Flora, S.; Albini, A. Angioprevention’: Angiogenesis is a common and key target for cancer chemopreventive agents. Faseb J. 2002, 16, 2–14. [Google Scholar] [CrossRef] [PubMed]
  50. Torchilin, V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv. Drug Deliv. Rev. 2011, 63, 131–135. [Google Scholar] [CrossRef]
  51. Prabhakar, U.; Maeda, H.; Jain, R.K.; Sevick-Muraca, E.M.; Zamboni, W.; Farokhzad, O.C.; Barry, S.T.; Gabizon, A.; Grodzinski, P.; Blakey, D.C. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. Cancer Res. 2013, 73, 2412–2417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Maeda, H.; Nakamura, H.; Fang, J. The EPR effect for macromolecular drug delivery to solid tumors: Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv. Drug Deliv. Rev. 2013, 65, 71–79. [Google Scholar] [CrossRef] [PubMed]
  53. Jain, R.K.; Stylianopoulos, T. Delivering nanomedicine to solid tumors. Nat. Rev. Clin. Oncol. 2010, 7, 653–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Taurin, S.; Nehoff, H.; Greish, K. Anticancer nanomedicine and tumor vascular permeability; Where is the missing link? J. Control. Release 2012, 164, 265–275. [Google Scholar] [CrossRef]
  55. Kwon, I.K.; Lee, S.C.; Han, B.; Park, K. Analysis on the current status of targeted drug delivery to tumors. J. Control. Release 2012, 164, 108–114. [Google Scholar] [CrossRef] [Green Version]
  56. Bae, Y.H.; Park, K. Targeted drug delivery to tumors: Myths, reality and possibility. J. Control. Release 2011, 153, 198–205. [Google Scholar] [CrossRef] [Green Version]
  57. Maeda, H.; Khatami, M. Analyses of repeated failures in cancer therapy for solid tumors: Poor tumor-selective drug delivery, low therapeutic efficacy and unsustainable costs. Clin. Transl. Med. 2018, 7, 11. [Google Scholar] [CrossRef]
  58. Lammers, T. Smart drug delivery systems: Back to the future vs. clinical reality. Int. J. Pharm. 2013, 454, 527–529. [Google Scholar] [CrossRef] [Green Version]
  59. Inui, A.; Honda, A.; Yamanaka, S.; Ikeno, T.; Yamamoto, K. Effect of ultrasonic frequency and surfactant addition on microcapsule destruction. Ultrason. Sonochem. 2021, 70, 105308. [Google Scholar] [CrossRef]
  60. Li, M.; Lan, B.; Sankin, G.; Zhou, Y.; Liu, W.; Xia, J.; Wang, D.; Trahey, G.; Zhong, P.; Yao, J. Simultaneous Photoacoustic Imaging and Cavitation Mapping in Shockwave Lithotripsy. IEEE Trans. Med. Imaging 2020, 39, 468–477. [Google Scholar] [CrossRef]
  61. Suslick, K.S.; Eddingsaas, N.C.; Flannigan, D.J.; Hopkins, S.D.; Xu, H. The Chemical History of a Bubble. Acc. Chem. Res. 2018, 51, 2169–2178. [Google Scholar] [CrossRef] [PubMed]
  62. Yildirim, A.; Blum, N.T.; Goodwin, A.P. Colloids, nanoparticles, and materials for imaging, delivery, ablation, and theranostics by focused ultrasound (FUS). Theranostics 2019, 9, 2572–2594. [Google Scholar] [CrossRef] [PubMed]
  63. Ye, L.; Zhu, X.; He, Y.; Wei, X. Ultrasonic cavitation damage characteristics of materials and a prediction model of cavitation impact load based on size effect. Ultrason. Sonochem. 2020, 66, 105115. [Google Scholar] [CrossRef] [PubMed]
  64. Daecher, A.; Stanczak, M.; Liu, J.B.; Zhang, J.; Du, S.; Forsberg, F.; Leeper, D.B.; Eisenbrey, J.R. Localized microbubble cavitation-based antivascular therapy for improving HCC treatment response to radiotherapy. Cancer Lett. 2017, 411, 100–105. [Google Scholar] [CrossRef] [PubMed]
  65. Huang, P.T.; You, X.D.; Pan, M.Q.; Li, S.Y.; Zhang, Y.; Zhao, Y.Z.; Wang, M.H.; Hong, Y.R.; Pu, Z.X.; Chen, L.R.; et al. A novel therapeutic strategy using ultrasound mediated microbubbles destruction to treat colon cancer in a mouse model. Cancer Lett. 2013, 335, 183–190. [Google Scholar] [CrossRef]
  66. Wang, F.; Dong, L.; Liang, S.M.; Wei, X.X.; Wang, Y.L.; Chang, L.S.; Guo, K.; Wu, H.W.; Chang, Y.Q.; Yin, Y.L.; et al. Ultrasound-triggered drug delivery for glioma therapy through gambogic acid-loaded nanobubble-microbubble complexes. Biomed. Pharm. 2022, 150, 113042. [Google Scholar] [CrossRef]
  67. Liu, T.; Li, M.; Tang, J.; Li, J.; Zhou, Y.; Liu, Y.; Yang, F.; Gu, N. An acoustic strategy for gold nanoparticle loading in platelets as biomimetic multifunctional carriers. J. Mater. Chem. B 2019, 7, 2138–2144. [Google Scholar] [CrossRef]
  68. Stride, E.; Coussios, C. Nucleation, mapping and control of cavitation for drug delivery. Nat. Rev. Phys. 2019, 1, 495–509. [Google Scholar] [CrossRef]
  69. Athanassiadis, A.G.; Ma, Z.C.; Moreno-Gomez, N.; Melde, K.; Choi, E.; Goyal, R.; Fischer, P. Ultrasound-Responsive Systems as Components for Smart Materials. Chem. Rev. 2022, 122, 5165–5208. [Google Scholar] [CrossRef]
  70. Rapoport, N. Ultrasound-mediated micellar drug delivery. Int. J. Hyperth. 2012, 28, 374–385. [Google Scholar] [CrossRef]
  71. Sankin, G.N.; Simmons, W.N.; Zhu, S.L.; Zhong, P. Shock wave interaction with laser-generated single bubbles. Phys. Rev. Lett. 2005, 95, 034501. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, H.; Kreider, W.; Brayman, A.A.; Bailey, M.R.; Matula, T.J. Blood vessel deformations on microsecond time scales by ultrasonic cavitation. Phys. Rev. Lett. 2011, 106, 034301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Bang, J.H.; Suslick, K.S. Applications of ultrasound to the synthesis of nanostructured materials. Adv. Mater. 2010, 22, 1039–1059. [Google Scholar] [CrossRef] [PubMed]
  74. Fernandez Rivas, D.; Prosperetti, A.; Zijlstra, A.G.; Lohse, D.; Gardeniers, H.J. Efficient sonochemistry through microbubbles generated with micromachined surfaces. Angew. Chem. Int. Ed. Engl. 2010, 49, 9699–9701. [Google Scholar] [CrossRef] [Green Version]
  75. Kwan, J.J.; Graham, S.; Myers, R.; Carlisle, R.; Stride, E.; Coussios, C.C. Ultrasound-induced inertial cavitation from gas-stabilizing nanoparticles. Phys. Rev. E Stat. Nonlin. Soft Matter Phys. 2015, 92, 023019. [Google Scholar] [CrossRef]
  76. Newman, C.M.; Bettinger, T. Gene therapy progress and prospects: Ultrasound for gene transfer. Gene. Ther. 2007, 14, 465–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Escoffre, J.M.; Piron, J.; Novell, A.; Bouakaz, A. Doxorubicin delivery into tumor cells with ultrasound and microbubbles. Mol. Pharm. 2011, 8, 799–806. [Google Scholar] [CrossRef]
  78. Grainger, S.J.; Serna, J.V.; Sunny, S.; Zhou, Y.; Deng, C.X.; El-Sayed, M.E. Pulsed ultrasound enhances nanoparticle penetration into breast cancer spheroids. Mol. Pharm. 2010, 7, 2006–2019. [Google Scholar] [CrossRef] [Green Version]
  79. Marmottant, P.; Bouakaz, A.; de Jong, N.; Quilliet, C. Buckling resistance of solid shell bubbles under ultrasound. J. Acoust. Soc. Am. 2011, 129, 1231–1239. [Google Scholar] [CrossRef] [Green Version]
  80. van Wamel, A.; Kooiman, K.; Harteveld, M.; Emmer, M.; ten Cate, F.J.; Versluis, M.; de Jong, N. Vibrating microbubbles poking individual cells: Drug transfer into cells via sonoporation. J. Control. Release 2006, 112, 149–155. [Google Scholar] [CrossRef]
  81. Mehier-Humbert, S.; Bettinger, T.; Yan, F.; Guy, R.H. Plasma membrane poration induced by ultrasound exposure: Implication for drug delivery. J. Control. Release 2005, 104, 213–222. [Google Scholar] [CrossRef] [PubMed]
  82. Park, J.; Fan, Z.; Deng, C.X. Effects of shear stress cultivation on cell membrane disruption and intracellular calcium concentration in sonoporation of endothelial cells. J. Biomech. 2011, 44, 164–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Qin, P.; Han, T.; Yu, A.C.H.; Xu, L. Mechanistic understanding the bioeffects of ultrasound-driven microbubbles to enhance macromolecule delivery. J. Control. Release 2018, 272, 169–181. [Google Scholar] [CrossRef] [PubMed]
  84. Eggen, S.; Afadzi, M.; Nilssen, E.A.; Haugstad, S.B.; Angelsen, B.; Davies Cde, L. Ultrasound improves the uptake and distribution of liposomal Doxorubicin in prostate cancer xenografts. Ultrasound Med. Biol. 2013, 39, 1255–1266. [Google Scholar] [CrossRef]
  85. Qiu, S.F.; Li, D.X.; Wang, Y.G.; Xiu, J.C.; Lyu, C.Y.; Kutty, S.; Zha, D.G.; Wu, J.F. Ultrasound-Mediated Microbubble Cavitation Transiently Reverses Acute Hindlimb Tissue Ischemia through Augmentation of Microcirculation Perfusion Via the Enos/No Pathway. Ultrasound Med. Biol. 2021, 47, 1014–1023. [Google Scholar] [CrossRef]
  86. Ji, C.; Wang, L.; Dai, R.; Shan, L.; Yang, H.; Zhu, H.; Meng, Q. Hyperthermia exacerbates the effects of cathepsin L on claudin-1 in a blood-brain barrier model in vitro. Brain Res. 2016, 1631, 72–79. [Google Scholar] [CrossRef]
  87. Cai, H.; Liu, W.; Xue, Y.; Shang, X.; Liu, J.; Li, Z.; Wang, P.; Liu, L.; Hu, Y.; Liu, Y. Roundabout 4 regulates blood-tumor barrier permeability through the modulation of ZO-1, Occludin, and Claudin-5 expression. J. Neuropathol. Exp. Neurol. 2015, 74, 25–37. [Google Scholar] [CrossRef] [Green Version]
  88. Ma, J.; Wang, P.; Liu, Y.; Zhao, L.; Li, Z.; Xue, Y. Kruppel-like factor 4 regulates blood-tumor barrier permeability via ZO-1, occludin and claudin-5. J. Cell Physiol. 2014, 229, 916–926. [Google Scholar] [CrossRef]
  89. Wang, F.; Song, X.; Zhou, M.; Wei, L.; Dai, Q.; Li, Z.; Lu, N.; Guo, Q. Wogonin inhibits H2O2-induced vascular permeability through suppressing the phosphorylation of caveolin-1. Toxicology 2013, 305, 10–19. [Google Scholar] [CrossRef]
  90. Martin, T.A.; Mason, M.D.; Jiang, W.G. HGF and the regulation of tight junctions in human prostate cancer cells. Oncol. Rep. 2014, 32, 213–224. [Google Scholar] [CrossRef]
  91. Tang, L.; Zhang, C.Y.; Yang, Q.; Xie, H.; Liu, D.D.; Tian, H.B.; Lu, L.X.; Xu, J.Y.; Li, W.Y.; Xu, G.X.; et al. Melatonin maintains inner blood-retinal barrier via inhibition of p38/TXNIP/NF-kappa B pathway in diabetic retinopathy. J. Cell. Physiol. 2021, 236, 5848–5864. [Google Scholar] [CrossRef] [PubMed]
  92. Liao, J.Z.; Li, Q.W.; Lei, C.Q.; Yu, W.L.; Deng, J.C.; Guo, J.Y.; Han, Q.Y.; Hu, L.M.; Li, Y.; Pan, J.Q.; et al. Toxic effects of copper on the jejunum and colon of pigs: Mechanisms related to gut barrier dysfunction and inflammation influenced by the gut microbiota. Food Funct. 2021, 12, 9642–9657. [Google Scholar] [CrossRef] [PubMed]
  93. Poplawska, M.; Dutta, D.; Jayaram, M.; Chong, N.S.; Salifu, M.; Lim, S.H. Genes modulating intestinal permeability and microbial community are dysregulated in sickle cell disease. Ann. Hematol. 2022, 101, 1009–1013. [Google Scholar] [CrossRef]
  94. Cheng, C.Y.; Mruk, D.D. The Blood-Testis Barrier and Its Implications for Male Contraception. Pharmacol. Rev. 2012, 64, 16–64. [Google Scholar] [CrossRef] [Green Version]
  95. Rose, E.C.; Odle, J.; Blikslager, A.T.; Ziegler, A.L. Probiotics, Prebiotics and Epithelial Tight Junctions: A Promising Approach to Modulate Intestinal Barrier Function. Int. J. Mol. Sci. 2021, 22, 6729. [Google Scholar] [CrossRef] [PubMed]
  96. Kaminsky, L.W.; Al-Sadi, R.; Ma, O.M. IL-1 beta and the Intestinal Epithelial Tight Junction Barrier. Front. Immunol. 2021, 12, 767456. [Google Scholar] [CrossRef]
  97. Sasson, E.; Anzi, S.; Bell, B.; Yakovian, O.; Zorsky, M.; Deutsch, U.; Engelhardt, B.; Sherman, E.; Vatine, G.; Dzikowski, R.; et al. Nano-scale architecture of blood-brain barrier tight-junctions. Elife 2021, 10, e63253. [Google Scholar] [CrossRef]
  98. Bae, M.J.; Lee, Y.M.; Kim, Y.H.; Han, H.S.; Lee, H.J. Utilizing Ultrasound to Transiently Increase Blood-Brain Barrier Permeability, Modulate of the Tight Junction Proteins, and Alter Cytoskeletal Structure. Curr. Neurovasc. Res. 2015, 12, 375–383. [Google Scholar] [CrossRef]
  99. Shang, X.; Wang, P.; Liu, Y.; Zhang, Z.; Xue, Y. Mechanism of low-frequency ultrasound in opening blood-tumor barrier by tight junction. J. Mol. Neurosci. 2011, 43, 364–369. [Google Scholar] [CrossRef]
  100. Cool, S.K.; Geers, B.; Roels, S.; Stremersch, S.; Vanderperren, K.; Saunders, J.H.; De Smedt, S.C.; Demeester, J.; Sanders, N.N. Coupling of drug containing liposomes to microbubbles improves ultrasound triggered drug delivery in mice. J. Control. Release 2013, 172, 885–893. [Google Scholar] [CrossRef]
  101. Goertz, D.E. An overview of the influence of therapeutic ultrasound exposures on the vasculature: High intensity ultrasound and microbubble-mediated bioeffects. Int. J. Hyperth. 2015, 31, 134–144. [Google Scholar] [CrossRef] [PubMed]
  102. Kuijpers, M.J.; Gilio, K.; Reitsma, S.; Nergiz-Unal, R.; Prinzen, L.; Heeneman, S.; Lutgens, E.; van Zandvoort, M.A.; Nieswandt, B.; Egbrink, M.G.; et al. Complementary roles of platelets and coagulation in thrombus formation on plaques acutely ruptured by targeted ultrasound treatment: A novel intravital model. J. Thromb. Haemost 2009, 7, 152–161. [Google Scholar] [CrossRef]
  103. Song, F.Y.; Gao, H.; Li, D.Y.; Petrov, A.V.; Petrov, V.V.; Wen, D.S.; Sukhorukov, G.B. Low intensity focused ultrasound responsive microcapsules for non-ablative ultrafast intracellular release of small molecules. J. Mater. Chem. B 2021, 9, 2384–2393. [Google Scholar] [CrossRef] [PubMed]
  104. Tu, L.; Liao, Z.; Luo, Z.; Wu, Y.L.; Herrmann, A.; Huo, S. Ultrasound-controlled drug release and drug activation for cancer therapy. Exploration 2021, 1, 20210023. [Google Scholar] [CrossRef]
  105. Tinkov, S.; Winter, G.; Coester, C.; Bekeredjian, R. New doxorubicin-loaded phospholipid microbubbles for targeted tumor therapy: Part I--Formulation development and in-vitro characterization. J. Control. Release 2010, 143, 143–150. [Google Scholar] [CrossRef]
  106. Li, F.; Jin, L.; Wang, H.; Wei, F.; Bai, M.; Shi, Q.; Du, L. The dual effect of ultrasound-targeted microbubble destruction in mediating recombinant adeno-associated virus delivery in renal cell carcinoma: Transfection enhancement and tumor inhibition. J. Gene Med. 2014, 16, 28–39. [Google Scholar] [CrossRef]
  107. Haag, P.; Frauscher, F.; Gradl, J.; Seitz, A.; Schafer, G.; Lindner, J.R.; Klibanov, A.L.; Bartsch, G.; Klocker, H.; Eder, I.E. Microbubble-enhanced ultrasound to deliver an antisense oligodeoxynucleotide targeting the human androgen receptor into prostate tumours. J. Steroid. Biochem. Mol. Biol. 2006, 102, 103–113. [Google Scholar] [CrossRef]
  108. Yan, F.; Li, X.; Jin, Q.; Jiang, C.; Zhang, Z.; Ling, T.; Qiu, B.; Zheng, H. Therapeutic ultrasonic microbubbles carrying paclitaxel and LyP-1 peptide: Preparation, characterization and application to ultrasound-assisted chemotherapy in breast cancer cells. Ultrasound Med. Biol. 2011, 37, 768–779. [Google Scholar] [CrossRef]
  109. Cochran, M.C.; Eisenbrey, J.; Ouma, R.O.; Soulen, M.; Wheatley, M.A. Doxorubicin and paclitaxel loaded microbubbles for ultrasound triggered drug delivery. Int. J. Pharm. 2011, 414, 161–170. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, D.S.; Panje, C.; Pysz, M.A.; Paulmurugan, R.; Rosenberg, J.; Gambhir, S.S.; Schneider, M.; Willmann, J.K. Cationic versus neutral microbubbles for ultrasound-mediated gene delivery in cancer. Radiology 2012, 264, 721–732. [Google Scholar] [CrossRef]
  111. Ren, S.T.; Liao, Y.R.; Kang, X.N.; Li, Y.P.; Zhang, H.; Ai, H.; Sun, Q.; Jing, J.; Zhao, X.H.; Tan, L.F.; et al. The antitumor effect of a new docetaxel-loaded microbubble combined with low-frequency ultrasound in vitro: Preparation and parameter analysis. Pharm. Res. 2013, 30, 1574–1585. [Google Scholar] [CrossRef] [PubMed]
  112. Escoffre, J.M.; Mannaris, C.; Geers, B.; Novell, A.; Lentacker, I.; Averkiou, M.; Bouakaz, A. Doxorubicin liposome-loaded microbubbles for contrast imaging and ultrasound-triggered drug delivery. IEEE Trans. Ultrason. Ferroelectr. Freq. Control 2013, 60, 78–87. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, G.; Zhuo, Z.; Xia, H.; Zhang, Y.; He, Y.; Tan, W.; Gao, Y. Investigation into the impact of diagnostic ultrasound with microbubbles on the capillary permeability of rat hepatomas. Ultrasound Med. Biol. 2013, 39, 628–637. [Google Scholar] [CrossRef] [PubMed]
  114. Tang, Q.; He, X.; Liao, H.; He, L.; Wang, Y.; Zhou, D.; Ye, S.; Chen, Q. Ultrasound microbubble contrast agent-mediated suicide gene transfection in the treatment of hepatic cancer. Oncol. Lett. 2012, 4, 970–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Li, P.; Zhu, M.; Xu, Y.; Zhao, Y.; Gao, S.; Liu, Z.; Gao, Y.H. Impact of microbubble enhanced, pulsed, focused ultrasound on tumor circulation of subcutaneous VX2 cancer. Chin. Med. J. 2014, 127, 2605–2611. [Google Scholar]
  116. Lin, C.Y.; Li, J.R.; Tseng, H.C.; Wu, M.F.; Lin, W.L. Enhancement of focused ultrasound with microbubbles on the treatments of anticancer nanodrug in mouse tumors. Nanomedicine 2012, 8, 900–907. [Google Scholar] [CrossRef]
  117. Fokong, S.; Theek, B.; Wu, Z.; Koczera, P.; Appold, L.; Jorge, S.; Resch-Genger, U.; van Zandvoort, M.; Storm, G.; Kiessling, F.; et al. Image-guided, targeted and triggered drug delivery to tumors using polymer-based microbubbles. J. Control. Release 2012, 163, 75–81. [Google Scholar] [CrossRef]
  118. Charrois, G.J.; Allen, T.M. Drug release rate influences the pharmacokinetics, biodistribution, therapeutic activity, and toxicity of pegylated liposomal doxorubicin formulations in murine breast cancer. Biochim. Biophys. Acta 2004, 1663, 167–177. [Google Scholar] [CrossRef]
  119. Pu, C.; Chang, S.; Sun, J.; Zhu, S.; Liu, H.; Zhu, Y.; Wang, Z.; Xu, R.X. Ultrasound-mediated destruction of LHRHa-targeted and paclitaxel-loaded lipid microbubbles for the treatment of intraperitoneal ovarian cancer xenografts. Mol. Pharm. 2014, 11, 49–58. [Google Scholar] [CrossRef] [Green Version]
  120. Zhou, S.; Li, S.; Liu, Z.; Tang, Y.; Wang, Z.; Gong, J.; Liu, C. Ultrasound-targeted microbubble destruction mediated herpes simplex virus-thymidine kinase gene treats hepatoma in mice. J. Exp. Clin. Cancer Res. 2010, 29, 170. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Ultrasound-mediated microbubble cavitation enhances biological barriers’ permeability and material transport. High interstitial pressure aroused by lack of blood perfusion and lymphatic return in solid tumors hinders the uptake and absorption of drug agents in cells. Using ultrasound to mediate the cavitation effect of microbubbles can increase blood–tumor barrier permeability and vascular perfusion, significantly increasing the diffusion of agents and sensitizing chemoresistance.
Figure 1. Ultrasound-mediated microbubble cavitation enhances biological barriers’ permeability and material transport. High interstitial pressure aroused by lack of blood perfusion and lymphatic return in solid tumors hinders the uptake and absorption of drug agents in cells. Using ultrasound to mediate the cavitation effect of microbubbles can increase blood–tumor barrier permeability and vascular perfusion, significantly increasing the diffusion of agents and sensitizing chemoresistance.
Pharmaceutics 14 01642 g001
Figure 2. Ultrasound-mediated microbubble cavitation enhancing tumor–blood barrier permeability. The blood–tumor barrier is composed of vascular endothelial cells, basement membrane, and tumor interstitial cells. Biological barrier regulator proteins include Occludin, Claudin, JAM, ZO, and so on. Ultrasound-mediated microbubble cavitation can significantly increase local tissue tight junction protein permeability and increase drug diffusion, meanwhile, can form vascular microcirculation thrombus by further increasing ultrasound frequency to induce tumor ischemic necrosis.
Figure 2. Ultrasound-mediated microbubble cavitation enhancing tumor–blood barrier permeability. The blood–tumor barrier is composed of vascular endothelial cells, basement membrane, and tumor interstitial cells. Biological barrier regulator proteins include Occludin, Claudin, JAM, ZO, and so on. Ultrasound-mediated microbubble cavitation can significantly increase local tissue tight junction protein permeability and increase drug diffusion, meanwhile, can form vascular microcirculation thrombus by further increasing ultrasound frequency to induce tumor ischemic necrosis.
Pharmaceutics 14 01642 g002
Table 1. Ultrasound-mediated microbubble adjuvant drug therapy in vitro.
Table 1. Ultrasound-mediated microbubble adjuvant drug therapy in vitro.
Cavitation MechanismAuthorsCell TypeComponentInterventionOutcomes after Cavitation Effect
Enhanced permeabilityTinkov et al. [105]Renal carcinoma cellDoxorubicinGroup 1: DOX
Group 2: DOX + MBs
Approximately two-fold enhanced anti-proliferative effect in DOX-loaded MBs.
DOX-loaded MBs with high affinity to the nucleus.
Enhanced permeability
Promoted drug diffusion
Li F. et al. [106]Renal carcinoma cellrAAVGroup 1: rAAV
Group 2: rAAV + MBs
Group 3: rAAV + US
Group 4: rAAV + UTMD
US-mediated MBs inhibit tumor cell proliferation and induce apoptosis.
US-mediated MBs promote viral transfection approximately two-fold.
Enhanced permeability
Promoted drug diffusion
Haag P. et al. [107]Prostate tumor cellODNsGroup 1: MBs
Group 2: ODNs
Group 3: ODNs + MBs
Group 4: ODNs + US
Group 5: ODNs + MBs + US
Best US frequency: 1.75 MHz; best MI: 1.9.
US-mediated MBs inhibit AR protein levels by 36.23%.
US-mediated MBs promote viral transfection approximately 40-fold.
Promoted drug uptakeYan F. et al. [108]Breast cancer cellPaclitaxel and LyP-1 PeptideGroup 1: MBs
Group 2: PTX-loaded MBs
Group 3: Targeted PTX-loaded MBs
Targeted ultrasonic MBs encapsulation rate: 63%.
US for 2 min increased cell uptake approximately seven-fold.
Enhanced permeability
Promoted drug diffusion
Cochran M.C. et al. [109]Breast cancer cellDoxorubicin and paclitaxelGroup 1: MBs
Group 2: MBs + US
Group 3: Drug-loaded MBs
Group 4: Drug-loaded MBs + US
The encapsulation efficiency of PTX and DOX: 72%, 20.5%.
The payload of PTX-loaded MBs is 20 times DOX.
The anti-tumor effect increased by 80.1%.
Promoted drug diffusionWang D.S. et al. [110]Vascular endothelial tumor cellDNAGroup 1: Cationic MBs + US
Group 2: Neutral MBs + US
Group 3: US
Group 4: Cationic MBs
Cationic MBs protect plasmid DNA from degradation.
Cationic MBs promote gene transfection approximately two-fold.
Enhanced permeabilityRen S.T. et al. [111]Colon adenocarcinoma cellDocetaxelGroup 1: DOC
Group 2: DOC + US
Group 3: MBs + US
Group 4: DOC + MBs + US
The maximum encapsulation rate: 54.9%.
The anti-tumor effect increased approximately two-fold.
Enhanced permeability
Promoted drug diffusion
Escoffre J.M. et al. [112]Glioblastoma cellDoxorubicinGroup 1: MBs + US
Group 2: DOX + MBs
Group 3: DOX + MBs + US
US-mediated MBs significantly increased drug uptake.
Tumor cell death with US-mediated MBs was enhanced approximately three-fold.
Abbreviations: MBs, microbubbles; DOX, doxorubicin; US, ultrasound; PTX, paclitaxel; DOC, docetaxel; rAAV, recombinant adeno-associated virus; UTMD, ultrasound-targeted microbubble destruction; ODNs, oligodeoxynucleotides; MI, mechanical index; AR, androgen receptor.
Table 2. Ultrasound-mediated microbubble adjuvant drug therapy in vivo.
Table 2. Ultrasound-mediated microbubble adjuvant drug therapy in vivo.
Cavitation MechanismAuthorsCell TypeComponentInterventionOutcomes after Cavitation Effect
Enhanced permeabilityWang G. et al. [113]Hepatic cancerEvans BlueGroup 1: EB
Group 2: EB + MBs
Group 3: EB + US
Group 4: EB + MBs + US
US-mediated MBs cavitation can increase tumor vascular permeability.
The cavitation effect promotes drug release approximately three-fold.
Enhanced permeabilityTang Q. et al. [114]Hepatic cancerHSV-TK/GCVGroup 1: pEGFP-KDR-TK + pEGFP-C1-AFP-TK
Group 2: pEGFP-KDR-TK + pEGFP-C1-AFP-TK + US
Group 3: pEGFP-KDR-TK + pEGFP-C1-AFP-TK + MBs + US
US-mediated MBs can increase killing effect of HSV-TK/GCV and CD/5-FC systems on vascular and hepatoma cells.
US-mediated MBs can increase tumor vascular permeability and gene transfection efficiency.
Enhanced permeability
Induced tumor necrosis
Li P. et al. [115]Subcutaneous VX2 cancerEvans BlueGroup 1: EB
Group 2: EB + MBs
Group 3: EB + US
Group 4: EB + MBs + US
US-mediated MBs can induce tumor microvasculature disruption resulting in hemorrhage, edema, and thrombosis to cause necrosis.
Enhanced permeabilityCool S.K. et al. [100]No tumorICG-liposomesGroup 1: Drug-MBs + US
Group 2: Drug + MBs + US
Group 3: MBs + US
Group 4: Drug + US
MBs can increase ICG-liposomes loaded approximately three-fold.
US-mediated MBs increase drug release two times more.
US-mediated MBs can cause skin lesions due during microbubble collapse.
Enhanced permeability
Promoted drug diffusion
Enhanced perfusion
Lin C.Y. et al. [116]Colon cancerDOXGroup 1: DOX
Group 2: DOX + MBs
Group 3: DOX + US
Group 4: DOX + MBs + US
US-mediated MBs cavitation can increase tissue permeability and destroy tumor vessels.
US-mediated MBs can increase tumor drug uptake and inhibit growth.
US intervention time should be less than 2 min.
Enhanced permeability
Promoted drug diffusion
Fokong S. et al. [117]Colon cancerRhodamine-B
Coumarin-6
Group 1: MBs-Rhodamine-B
Group 2: MBs-Coumarin-6
Group 3: MBs-Rhodamine-B + US
Group 4: MBs-Coumarin-6 + US
The polymer-based MBs are highly suitable for image-guided, targeted, and triggered drug delivery to tumors and blood vessels.
Enhanced permeability
Induced tumor necrosis
Huang P. et al. [65]Colon cancerNo drugGroup 1: MBs
Group 2: US
Group 3: MBs + US
US-mediated MBs inhibit tumor growth and metastasis.
US-mediated MBs destroy tumor cell nucleus and microvascular.
US-mediated MBs decreases the expression of CD31.
Abbreviations: EB, Evans Blue; MBs, microbubbles; DOX, doxorubicin; US, ultrasound; HSV-TK/GCV, Herpes simplex virus-thymidine kinase/ganciclovir; KDR, kinase insert domain receptor; 5-FC, 5-fluorocytosine; TK, thymidine kinase; pEGFP, enhanced green fluorescent protein plasmid; ICG, indocyanine green; PBCA, poly (butyl cyanoacrylate).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

He, J.; Liu, Z.; Zhu, X.; Xia, H.; Gao, H.; Lu, J. Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy. Pharmaceutics 2022, 14, 1642. https://doi.org/10.3390/pharmaceutics14081642

AMA Style

He J, Liu Z, Zhu X, Xia H, Gao H, Lu J. Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy. Pharmaceutics. 2022; 14(8):1642. https://doi.org/10.3390/pharmaceutics14081642

Chicago/Turabian Style

He, Jide, Zenan Liu, Xuehua Zhu, Haizhui Xia, Huile Gao, and Jian Lu. 2022. "Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy" Pharmaceutics 14, no. 8: 1642. https://doi.org/10.3390/pharmaceutics14081642

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop