fbpx
Wikipedia

Sonodynamic therapy

Sonodynamic therapy (SDT) is a noninvasive treatment, often used for tumor irradiation, that utilizes a sonosensitizer and the deep penetration of ultrasound to treat lesions of varying depths by reducing target cell number and preventing future tumor growth.[1][2] Many existing cancer treatment strategies cause systemic toxicity or cannot penetrate tissue deep enough to reach the entire tumor; however, emerging ultrasound stimulated therapies could offer an alternative to these treatments with their increased efficiency, greater penetration depth, and reduced side effects. Sonodynamic therapy could be used to treat cancers and other diseases, such as atherosclerosis, and diminish the risk associated with other treatment strategies since it induces cytotoxic effects only when externally stimulated by ultrasound and only at the cancerous region, as opposed to the systemic administration of chemotherapy drugs.[3][4][5]

UV Radiation
Photodynamic Therapy

Reactive oxygen species (ROS) are an essential component of SDT as they provide the cytotoxicity of sonodynamic therapy; they are produced when ultrasound is coupled with a sensitizing drug and molecular oxygen.[1] Without ultrasound, the drug is not toxic. However, once the drug is exposed to ultrasound and molecular oxygen, it becomes toxic.[1] Photodynamic therapy, from which sonodynamic therapy was derived, uses a similar mechanism. Instead of ultrasound, light is used to activate the drug.[1] SDT allows the ultrasound to reach deeper into the tissue (to about 30 centimeters) compared to photodynamic therapy (PDT) since it can be highly focused.[1] This increased penetration depth ultimately means that SDT can be utilized to treat deeper, less accessible tumors and is more cost-effective than PDT.[6][1] Photodynamic therapy can be used in combination with sonodynamic therapy and is expanded upon in the Applications section of this article. Sonodynamic therapy can be used synergistically with other therapeutic methods such as drug-loaded microbubbles, nanoparticles, exosomes, liposomes, and genes for improved efficacy. Currently, SDT does not have any clinical products and acts as an adjuvant for the aforementioned therapeutic methods, but it has been explored for use in atherosclerosis and cancer treatment to reduce tumor size in breast, pancreas, liver, and spinal sarcomas. [7][3] [8][9][10] [11][12][13] [14][15][16]

Mechanism of Action edit

 
Cavitation bubble implosion

The mechanism of action for sonodynamic therapy is the use of low-intensity ultrasound through the use of focused mechanical waves to create a cytotoxic effect. However, SDT itself is non-thermal, non-toxic, and is able to non-invasively penetrate deep into tissue compared to other delivery methods such as photodynamic therapy. SDT is often performed alongside the use of a sonosensitizer such as porphyrin, phthalocyanines, xanthenes, and antitumor drugs.[17] Ultrasound waves are also classified as acoustic waves, and the effect they have on the tissue of application can be described by a process called cavitation. Cavitation occurs as a specific interaction between ultrasound and aqueous surroundings and causes gas bubbles to break upon exposure to particular ultrasonic parameters, thus promoting penetration of the therapeutic into the biological tissues by generating cavities near the edge of the membrane.[18][1] Cavitation can be broken down into stable and inertial cavitation. In stable cavitation, the oscillation of gas bubbles causes the environmental media to intermix.[1] In inertial cavitation, gas bubbles increase in volume and almost reach their resonance volume, swelling before aggressively collapsing.[1] The implosion of vesicles results in a drastic temperature and pressure change, thereby increasing the cell membrane's permeability to various drugs.[1][19] Microbubbles are created by the acoustic waves from the ultrasound that expand and collapse, releasing energy, bringing the sonosensitizer into an excited state, and generating a ROS. The cavitation of this gas bubble can form the ROS with different methodologies such as sonoluminescence and pyrolysis.[1] Apoptosis results from the formation of ROS and mechanical forces of SDT through membrane disruption in a process called lipid peroxidation. Necrosis is also a potential result of SDT.

The influence of sonoluminescence on SDT and ROS has not been fully elaborated within literature.[1] Currently, it is understood that sonoluminescence allows the emission of light upon bubble collapse which can activate sensitizers. A study by Hachimine et al. highlights the use of SDT as a method to activate a low photosensitive sonosensitizer, DCPH-P-Na(I), for cancer that is too deep within the tissue to combat utilizing PDT without skin irritation.[1][20] Pyrolysis raises the surrounding temperature, enhances the cavitation process, breaks down the sensitizer, generating free radicals, and the free radicals interact within their environment to generate ROS.[1] For both methods, the importance of the singlet oxygen compared to the hydroxyl radical to induce cytotoxicity has been highlighted.[1][20][21] While other studies[1][22][23] have found the singlet oxygen to not have a substantial effect. Overall, both of these methodologies lack significant breadth in literature to fully explain their role in ROS formation. However, literature has shown success in their analysis and application.[1][4][24]

Sonoluminescence edit

 
Sonoluminescence acoustics

Two primary mechanisms of ROS generation exist in sonodynamic therapy: sonoluminescence and pyrolysis.[1] Sonoluminescence occurs when ultrasound produces light after irradiating an aqueous solution[1][25] The exact mechanism with which light is produced remains unclear. However, it is suggested that inertial cavitation is a key element for this process.[1][26] Other studies also indicate the potential role of stable cavitation[1][27]

Pyrolysis edit

Pyrolysis is believed to occur when inertial cavitation induces an extreme temperature increase, degrades the sonosensitizers, thus producing free radicals that can react and ultimately produce ROS necessary for SDT.[1][28] The localized temperature increase assists in the inertial cavitation and breakdown of the sonosensitizer in order to create ROS. The pyrolysis within the cavitation bubbles will produce H+ and OH- via weak bonding within the solute molecule.[1][19]

Lipid Peroxidation edit

 
Mechanism of lipid peroxidation.

In addition to chemical methods, mechanical properties of the acoustic wave generated from the ultrasound can assist in initiating cytotoxic effects. This occurs through disruption of the membrane with a hydrophobic sonosensitizer. The mechanical disruption of the membrane causes a process called lipid peroxidation and adjustments to the cell membrane can change cell drug permeability.[1][29] Both sonochemical and sonomechanical methodologies are used to generate ROS and release cargo from vesicles for applications such as tumor targeting.

Apoptosis edit

Low intensity ultrasound has been shown within past literature to induce apoptotic effects within surrounding cells. It has been found that it is not the initial ROS that causes apoptosis within the cells, but the free radicals within the mitochondria. In a study by Honda et al., it was determined that the mitochondria-caspase pathway is responsible for apoptosis through the increase of intracellular calcium.[1][30] Outside of ROS induced apoptosis, cavitation is another factor involved within apoptosis of surrounding cells. Both cavitation types are able to induce apoptosis through damage to the membrane. Conditions such as frequency, duty cycle, pulse, and intensity can be manipulated to optimize cell death conditions such as necrosis, lysis, or apoptosis.[31][24][32]

Autophagy edit

This method of cell death can occur by cell organelles becoming entrapped into autophagosomes that combine with lysosomes. Continuation of this process will lead to cell death and autophagy inhibitors or promoters can be controlled to encourage or discourage cell death and uptake of chemotherapeutics.[1]

Sonosensitizers edit

Sonosensitizers, or sonosensitizing therapeutics, are the primary element of SDT and can be tailored to treat various cancers and generate different effects.[2] These therapeutics, often involving the use of porphyrin or xanthene, will initiate a toxic effect via the ROS upon exposure to ultrasound.

Porphyrin-based sensitizers edit

 
The 18-electron cycle of porphin, the parent structure of porphyrin, highlighted. (Several other choices of atoms, through the pyrrole nitrogens, for example, also give 18-electron cycles.)

Porphyrin-based sensitizers, initially used as a photosensitizer in PDT, are fairly hydrophobic molecules derived from hematoporphyrin.[1] Single oxygen atoms or hydroxyl radicals are produced by porphyrin-based sensitizers upon exposure to ultrasound or light, providing the cytotoxic effects desired with sonodynamic and photodynamic therapies.[1] However, the result of porphyrin-based sensitizers is not as local as desired for sonodynamic therapy since they are also located in non-targeted tissue between the tumor and the ultrasound emitter.[1]

Xanthene-based sensitizers edit

 
Xanthene
 
Rose Bengal

Xanthene-based sensitizers, on the other hand, have shown successful cytotoxicity in vitro by producing reactive oxygen species after being triggered by ultrasound.[1] More research is necessary to improve its potential in vivo performance since it is quickly processed by the liver and cleared from the body.[1] Rose Bengal is a commonly used xanthene-based sonosensitizer.[1]

Additional sensitizers edit

Other sensitizers that have been investigated for their potential in sonodynamic therapy (and have also been used previously in PDT) include acridine orange, methylene blue, curcumin, and indocyanine green.[1] A study by Suzuki et al. used acridine orange, a fluorescent cationic dye that can insert itself into nucleic acids, for treating sarcoma 180 cells with ultrasound and demonstrated that reactive oxygen species are a critical element of SDT considering that their absence decreased the efficacy of SDT.[33] Similar to the previous study, a recent study by Komori et al. utilized ultrasound coupled with methylene blue (a phenothiazine dye commonly used in PDT that exhibits low toxicity) to irradiate sarcoma 180 cells and found that methylene blue was an effective sonosensitizer in decreasing cell viability.[34] Interestingly, curcumin is a spice that also can act as a sensitizer for PDT and SDT.[1] In a study by Waksman et al., curcumin was able to impact macrophages, which are important for development of plaques found in atherosclerosis patients, thus reducing the amount of plaque in an animal model.[35] These findings along with other research indicate that curcumin sensitizers could be used in SDT cancer treatments. Indocyanine green is a dye that absorbs near infrared wavelengths and is another sensitizer that has been shown to reduce cell viability when coupled with ultrasound and/or light.[36] An in vivo study demonstrated that treating a mouse tumor model with indocyanine green coupled with ultrasound and light resulted in a 98% reduction in tumor volume by 27 days after treatment.[36]

Name and structure of additional sensitizers
Name Structure
Phthalocyanine
 
Phthalocyanine
Indocyanine Green
 
Indocyanine Green
Phenothiazine
 
Phenothiazine
Curcumin-keto
 
Curcumin_structure_(Keto)
Curcumin-enol
 
Curcumin-enol

Carriers edit

As aforementioned, sonosensitizers are often used in conjunction with different drug carriers such as microbubbles, nanobubbles, liposomes, and exosomes to improve therapeutic agent concentration and penetration.[18]

Liposomes edit

 
Liposome Drug Incorporation

Liposomes are a common vehicle in drug delivery and specifically for the treatment of cancer. Liposomes contain a phospholipid bilayer. It is prevalent due to its ability to penetrate leaky vasculature and poor lymphatic drainage within tumors for enhanced permeability retention.[37] These drug carriers can encapsulate hydrophobic and lipophilic molecules within their lipid bilayer and can be made naturally or synthetically.[38][39] In addition, liposomes can entrap hydrophilic molecules in their hydrophilic core.[38] Compared to the common cancer treatment chemotherapy, drugs loaded into liposomes allow for decreased systemic toxicity and a potential increase in the efficacy of targeted delivery.[18] Success with liposomes as drug delivery systems has been shown both in vivo and in vitro.[38] A study by Liu et al. showed that liposomes can be used alongside SDT to trigger the release of drugs via oxidation of the lipid components.[40] Another study by Ninomiya et al. utilized nanoemulsion droplets exposed to ultrasonic waves for the formation of larger gas bubbles to disrupt the liposome membrane for drug release. Many properties and elements of liposomes can be altered for their specific purpose and to increase effectiveness, particularly their ability to travel in the blood and interact with cells and tissues in the body.[38] These elements include their diameter, charge, arrangement, as well as the makeup of their membranes.[38] Dai et al. proposed the incorporation of sonosensitizers with liposomes to enhance target specificity.[18] Since SDT stimulates cancerous tissues to absorb and retain sonosentizers followed by activation with extracorporeal ultrasound, Dai et al. investigated the effect of liposome-encapsulated drugs on the efficacy of targeted delivery in SDT. They found that, in addition to its convenience and practicality, SDT is a safe and effective option for treating cancer.[18]

Exosomes edit

Exosomes are nanocarriers that can provide targeted drug delivery of therapeutics to enhance local cytotoxic effects while minimizing any systemic impact. They are acquired from cells and are used for transportation purposes within the cell as membrane-bound vesicles. Advantages of exosomes for drug delivery purposes include their ability to be manipulated and engineered, in addition to their low toxicity and immunogenicity.[41][42] They have also inspired research into non-cell-based treatment methods for various cancers and diseases.[41] Other desirable aspects of exosomes include their overall biocompatibility and stability.[42] A study by Nguyen Cao et al. investigated the use of exosomes for the delivery of indocyanine green (ICG), a sonosensitizer for breast cancer treatment.[43] Significantly increased reactive oxygen species generation was observed in breast cancer cells treated with folic acid-conjugated exosomes.[43] This is one example of a sonosensitizer used to treat a specific cancer using sonodynamic therapy. Another example of exosome-based sonodynamic therapy was illustrated by Liu et al. In this study, exosomes were decorated with porphyrin sensitizers and this system was used with an external ultrasound device to control and target drug delivery through SDT.[40] Liu et al. provided a non-invasive method for treating cancer through extracorporeal activation of exosomes through ultrasound.[40]

Microbubbles edit

 
Mechanisms for Loading Microbubbles with Drug

Due to their ability to oscillate with exposure to low-frequency ultrasound, microbubbles have been used as contrast agents in order to visualize tissues in which the microbubbles have permeated.[44] However, when these microspheres are exposed to higher pressure ultrasound, they can rupture, which could be beneficial for drug delivery purposes.[44] Through SDT, these microbubbles could be selectively bursted at the tumor microenvironment in order to decrease systemic levels of the encapsulated drug and increase therapeutic efficacy. When applying SDT, the increase in acoustic pressure leads to the inertial cavitation, or collapse of the microbubble and local release of the cargo within. The inertial cavitation of the microbubbles when exposed to SDT is also referred to as ultrasound mediated microbubble destruction (UMMD).[45] The shell of microbubbles can be decorated with different components, including polymers, lipids, or proteins depending on their intended purpose.[44] Microbubbles have also been used for the localized release of attached cargo. This cargo is typically chemotherapeutics, antibiotics, or genes.[12] Different drugs can be directly loaded into the microbubble with methods such as conjugation and nanoparticle, liposome loading, and genes. The combination of genes and SDT is referred to as sonotransfection.[12] Examples of outer shell modifications can be seen in a study by McEwan et al. which found that lipid microbubbles showed reduced stability when sonosensitizers were added to their shells.[44] However, attaching the polymer poly lactic-co-glycolic acid (PLGA) to the shell resulted in increased stability compared to the lipid microbubbles without losing other desirable properties such as targeted delivery and selective cytotoxicity.[44] In another study, McEwan et al. investigated the ability of microbubbles carrying oxygen to increase production of reactive oxygen species, which are a necessary component of SDT, in the hypoxic environment of many solid tumors.[46] These microbubbles were stabilized with lipids and a Rose Bengal sonosensitizer was attached to the surface to treat pancreatic cancer.[46] Their work showed that coupling oxygen-loaded microbubbles that are sensitive to ultrasound with sonosensitizing drugs could allow for increased drug activation at the desired target even if hypoxia is present. Examples of therapeutics that have been loaded into microbubbles are gemcitabine, paclitaxel nanoparticles, plasmid DNA and 2,2′-azobis[2-(2-imidazolin-2-yl)propane]dihydrochloride loaded liposomes.[47][45][48][49] Due to the targeting nature of the ligands connected to the microbubble, it allows for the controlled and specific targeting of the desired tissue for treatment. Another study performed by Nesbitt et al. has shown improved tumor reduction when gemcitabine was loaded into the microbubble and applied to a human pancreatic cancer xenograft model with SDT.[48]

Nanobubbles edit

Similar to microbubbles, nanobubbles have shown efficacy in SDT.[50] However, due to their smaller size, nanobubbles are able to reach targets that microbubbles cannot. Nanobubbles can reach deeper tissue and travel past the vasculature. Previous research has demonstrated that nanobubbles are more capable of reaching the tumor since they can permeate endothelial cells and migrate away from the vasculature.[51][50] One study by Nittayacharn et al. developed doxorubicin-loaded nanobubbles and paired them with porphyrin sensitizers to be used in SDT for treatment of breast and ovarian cancer cells in vitro.[50] They found an almost 70% increase in cytotoxicity when using SDT compared to only perfluoropropane nanobubbles filled with iridium(III).[50] Additionally, compared to empty nanobubbles and/or free iridium(III), they observed greatest reactive oxygen species generation in the iridium(III)-nanobubbles exposed to ultrasound.[50] These results demonstrate that nanobubbles loaded with a sonosensitizer and exposed to ultrasound could be a potential effective treatment for cancer using SDT. As with microbubbles, nanobubbles have also shown promise as oxygen-delivering vesicles to enhance the effectiveness of SDT. In order to mitigate hypoxia of target tissue, Owen et al. used a pancreatic cancer rodent model to deliver phospholipid stabilized nanobubbles filled with oxygen.[52] The mice were divided into groups, one that received oxygen-filled nanobubbles prior to injection of a sonosensitizer and one that didn't.[52] A statistically significant difference between the levels of oxygen in the tumors of the two groups was observed, indicating that nanobubbles could be an effective addition to SDT to treat cancers in a hypoxic environment.[52]

Applications edit

Combination with other therapies edit

Sonodynamic therapy can be combined with other therapeutic techniques to enhance treatment efficacy for various types of cancers and diseases. SDT can be combined with photodynamic therapy, chemotherapy, radiation, MRI, and immunotherapy. PDT has often been used in combination with SDT as sonosensitizers are also photosensitive.[1] During initial development of SDT, Umemura et al., have determined that hematoporphyrins were able to initiate cell death similarly to PDT.[21] This is due to SDT being able to initiate sonoluminescence. However, the advantage of SDT over PDT is that it can penetrate deep and precisely into the targeted tissue. In a study by Lui et al., it was shown that using a combination of these two delivery methods results in increased cytotoxicity with sino porphyrin in a metastatic xenograft model.[53] In another example of combining SDT with PDT, Borah et al. investigated the advantage of 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-a (HHPH), a photodynamic therapy drug, as a sonosensitizer and a photosensitizer for treating glioblastoma.[54] Combining these therapies showed increased cell kill/tumor response, possibly caused by synergistic effects.[54]

The goal of a study by Browning et al. was to investigate the potential enhancement of chemoradiation efficacy through combining it with sonodynamic therapy in pancreatic cancer patients. In one model, survival increased with the combination compared to chemoradiation alone. Differences in the results for the two different models could be attributed to variations in tumor organization.[6] The tumors that showed the greatest reduction in size were less vascularized, perhaps making them more vulnerable to SDT.[6] Another study, by Huang et al. used elements of mesoporous organosilica-based nanosystems to fabricate a sonosensitizer to be used with MRI-guided SDT.[55] Increased cell death and inhibiting tumor growth was induced by the sonosensitizers, indicating high SDT efficiency.[55] This shows how SDT can assist with both removal and inhibition of tumor growth.

SDT has also been combined with immunotherapy. A study by Lin et al. aimed to use cascade immuno-sonodynamic therapy to enhance tumor treatment using antibodies.[56] The nanosonosensitizers resulted in high drug loading efficiency and a tumor-specific adaptive immune response. This serves as an example as to how SDT can be coupled with checkpoint blockade immunotherapy to enhance efficiency in cancer treatments. Another study by Yue et al. strived to combine checkpoint-blockade immunotherapy with nanosonosensitizers-augmented noninvasive sonodynamic therapy.[57] Along with inhibiting lung metastasis, this combination promoted an anti-tumor response that prohibited tumor growth. This provides a proof-of-concept for combining SDT with another therapy to enhance treatment effects for the short and long term.

Types of cancers SDT has been shown to treat edit

Cancer Treatment edit

The treatment of many different types of cancers has been investigated using sonodynamic therapy both in vitro and/or in vivo including, glioblastoma, pancreatic, breast, ovarian, lung, prostate, liver, stomach, and colon cancers.[54][6][20][50][52] A study by Gao et al. showed that SDT is capable of inhibiting angiogenesis through the production of ROS. This hindered the proliferation, migration, and invasion of endothelial cells, tumor growth, intratumoral vascularity, and vascular endothelial growth factor expression within the tumor cell in xenograft rat models.[58] Hachimine et al. performed a large in vitro study testing SDT on seventeen different cancer cell lines.[20] The types of cancers included were pancreatic, breast, lung, prostate, liver, stomach, and colon cancers.[20] The most successful treatment was that of lung cancer with 23.4% cell viability post-therapy.[1][20] Qu et al. aimed to develop an “all-in-one” nanosensitizer platform triggered by SDT that combines various diagnostic and therapeutic effects to treat glioblastoma.[59] Apoptosis was successfully induced and mitophagy was inhibited in glioma cells. This is an example of how SDT can be used with a different platform to treat glioblastoma. Borah et al., as mentioned above, also investigated the ability of SDT (and PDT) to treat glioblastoma and found that SDT (combined with PDT) was able to increase the number of tumor cells killed.[54] McEwan et al. and Owen et al. both demonstrated the use of micro/nanobubbles to enhance the oxygen concentration near hypoxic pancreatic tumors, thereby increasing the efficacy of SDT.[46][52]

Breast Cancer edit

 
Morphologic and immunohistochemical features of Triple Negative Breast Cancer

12% of women in the US will be diagnosed with breast cancer.[60] Metastasis and recurrence is a large challenge for deep-seated solid state tumors.[39] SDT is currently being explored as a treatment method for breast cancer, while avoiding the side effects associated with current therapeutic methods.[61] There has been shown success in utilizing SDT in animal and human clinical trials in reduction of tumor size through mitochondrial targeting to initiate apoptosis of tumor cells and autophagy and immune response regulation.[62][39][63][64][24][53][60][65][61] However, there are still complications with proper therapeutic efficacy when used alone.

Glioma edit

 
Glioblastoma

Malignant glioma is an extremely difficult to treat brain tumor that is a leading cause of death worldwide and half of cancer-related deaths.[14] Complications associated with treating glioma include the blood brain barrier (BBB).[14] This protective mechanism for the brain also raises challenges for drug delivery through the tight junctions between endothelial cells, only allowing small lipid-soluble drugs (<400 Da) to permeate.[14] Current delivery methods are surgery and chemotherapy. SDT has been implemented as a method to open the BBB and has shown success in opening tight junctions for delivery. Examples of sonosensitizers that have shown success in glioma treatment are hematopor-phyrin monomethyl ether (HMME), porfimer sodium (Photofrin), di-sulfo-di-phthalimidomethyl phthalolcyaninezinc (ZnPcS2P2), Photolon, 5-aminolevulinic acid (5-ALA), and rose bengal (RB).[14] These have shown to induce effects such as opening of the BBB, improved vascular permeability, and apoptosis of glioma cells.

Prostate Cancer edit

 
Benign prostatic hyperplasia

Prostate cancer is the second cause of cancer and the most common malignancy associated with deaths in men worldwide.[66] Current methods of treatments are invasive resection therapy, radiation therapy, and prostatectomy that can cause complications such as incontinence, impotence, and damage to surrounding organs and tissues.[67][17] Current studies have shown success in using SDT as a stand-alone treatment.[68] SDT uses mitochondria related apoptosis for the reduction of cell viability. SDT for prostate cancer treatment has also been used alongside chemotherapeutics such as docetaxel microbubbles.[17][67][68] This has shown to enhance the effects of docetaxel through a reduction in tumor perfusion and enhanced necrosis and apoptosis.[68] The SDT and docetaxel group showed reduction in tumor growth.[68] Overall, the use of SDT has shown promising results in prostate cancer treatment.

Arterial Diseases edit

Sonodynamic therapy could be used to treat more than just cancers. Atherosclerosis, which is a chronic arterial disease, is another target that has been observed in the literature.[3][5] This disease occurs when fatty plaques aggregate on the inner surface of the artery and could be caused by malfunctions in lipid metabolism.[3] More specifically, atherosclerosis is caused by an increase in endothelial permeability causing low-density lipoprotein particles to become oxidized and undergo sedimentation.[3] These lipoproteins cause an increase in macrophages and lead to intensified plaque build up. As a result, the high influx of macrophages is the target for AS treatment in order to slow plaque build-up.[3] Alongside the relationship between plaque build-up and macrophages, monocyte's differentiation into macrophages exacerbates the aforementioned process in addition to causing inflammation.[3]

 
Normal Artery versus Atherosclerosis

A study by Wang et al. aimed to understand the underlying mechanisms regarding the potential effect of non-lethal SDT on atheroscleroic plaques. It was determined that non-lethal SDT prevents plaque development.[5] A study performed by Jiang et al., showed success in SDT through the reduction of macrophage inflammatory factors such as TNF-alpha, IL-12, and IL-1B. They also showed that SDT could inhibit plaque inflammation in patients with peripheral artery disease and continue to promote positive results for longer than six months.[4] Popular sonosensitizers for AS treatment are protoporphyrin IX (PpIX) and 5-aminolevulinic acid (5-ALA).[69][3] PpIX is often used in PDT and is generated through 5-ALA, a non ultrasound-activated component, through increasing PpIX concentration within a cell. A study by Cheng et al. determined that THP-1 macrophage apoptosis is induced by an increase in PpiX concentration, leading to the production of large amounts of ROS.[70][13][3] The use of SDT for AS treatment has also shown success in promoting the repopulation of vascular smooth muscle cells (VMSCs) through inducing further expression and autophagy to prevent VMSC evolution into plaque-holding macrophages. A study performed by Dan et al. showed the increase in smooth muscle a-actin, smooth muscle 22a, p38 mitogen-activated protein kinase phosphorylation.[71][3] While a study by Geng et al. showed improved VMSC autophagy. Each of these factors contributed to the improved differentiation and development of VMSCs.[3]

In Vitro and In Vivo Work edit

In vitro edit

In vitro experimentation provides great insight and knowledge to characterize the potential of sonosensitizer behavior in vivo. In addition, SDT has shown success through its low intensity allowing increased plasma membrane permeability without cell death.[1] Sonosensitizers have also been used in vitro in applications with different cell lines and to further understand the mechanism of action for cell death. It is currently understood that PDT and SDT have similar mechanisms for free radical generation for inducing apoptosis and necrosis.[1] However, each cell line is unique and can cause cell death with different efficacy.[20][1][72] Some examples of in vitro work include initial studies that were performed by Yumita et al., 1989 who used haematoprophyrin and SDT for mouse sarcoma 180 and rat ascites hepatoma (AH) that showed a relationship between dosage and ultrasound, and microbubbles causing cavitation leading to cell damage without the use of drugs. This study also emphasized the difference in efficacy between cell lines through SDT 180 having less lysis compared to AH-130 cells. Another study by Hachimine et al. emphasized efficacy between cell lines by examining seven different cancers with 17 cell lines total under the use of DCPH-P-NA(I).[1][20] This study revealed that the stomach and lung cancer lines of MKN-28 and LU65A respectively had the highest survival rate, but the stomach and lung cancer lines of RERFLC-KJ and MKN-45 respectively had the lowest survival rates.[20][1] Another study by Honda et al., with U937 and K562 showed that sonication increases the intracellular calcium ion levels and decreases GSH concentration respectively.[30] This increased concentration of calcium plays a significant role in cell death through DNA fragmentation and mitochondrial membrane disruption.[1][30] While a decreased concentration of GSH plays a significant role in allowing the formation of more free radicals.[30][1] A study by Umemura et al., found that ATX-70 versus hematoporphyrin has increased cytotoxic activity.[21][1] Current research typically focuses on using tumor xenograft models to determine the effect of SDT on target cells and delivery efficacy.[1]

In vivo edit

Building upon the study by Umemura et al. and ATX-70, it was found that 24h after administration of the sonosensitizer had improved efficacy when ultrasound was applied compared to immediate administration.[21][1] It was also determined that most ultrasound frequencies range between 1-3 MHz and 0.5-4W/cm^2. Higher frequencies at values such as 20W/cm^2 and 25W/cm^2 resulted in large necrotic lesions.[73][1] This established a relationship between sonosensitizer formulation and ultrasound intensity to necrosis. Other studies have continued to innovate upon this by controlling drug ultrasound interval (DUI) for different sonosensitizers in order to determine the optimal time period to apply the ultrasound for improved efficacy.[58][1] In addition, it has been shown that SDT can disturb surrounding vasculature in tumors.[1][58] This has been shown in studies by Gao et al. with 5-ALA in mice and human umbilical vein endothelial cell lines through inhibition of microvessel density and cell proliferation, migration, and invasion.[58][1]

Challenges and Development edit

 
Ultrasound Imaging vs. Ultrasound Therapy

One of the many advantages of SDT compared to PDT is the ability of SDT to penetrate deeply placed solid tumors allowing a wider treatment range.[1] Despite this fact, there are limitations to SDT that must be overcome or have optimized components in order to expand the effect and application of SDT.[31] SDT does allow for precise activation of the therapeutic, but is limited in the delivery and accumulation of the delivery modality to penetrate deeply into the desired tumor site.[74] This is often accommodated for through delivery vessels such as nanoparticles or liposomes.[1] However, nanomedicine is limited by the enhanced permeability and retention effect and struggles to deliver in targeted abundance depending on the delivery vesicle.[31][74] This can be seen in nanoparticles struggling with non-specific delivery. Future research has been focused on developing high targeting and penetrating nanoparticles for improved delivery and pharmacokinetics.[75][31] Due to the complex nature of tumors and their microenvironments, they are difficult to treat with only one therapy. In order to enhance the oftentimes low production of reactive oxygen species to address the hypoxic tumor environment, SDT can be combined with other therapies, such as PDT, chemotherapy, and immunotherapy to improve patient outcomes.[2][56][54][6] SDT alone does not respond well in hypoxic environments. However, bioreductive therapy could be used to reduce the impact of SDT's limitations regarding hypoxia in the tumor while leaving healthy/normal tissue alone.[2] Sonosensitizers also require continuous high levels of oxygen to create ROS, which is not readily available within a hypoxic tumor microenvironment.[31] However, strategies such as oxygen supplementation and production to supply the required oxygen and enhance cavitation, and glutathione depletion to avoid the reduction of the free radicals produced have been implemented alongside sonosensitizers to supply the required oxygen or reduce the combative function.[76][74] In addition to its relatively low generation of reactive oxygen species, SDT also can cause permanent destruction of normal tissues. This lack of selectivity is caused by ultrasound divergence, resulting in heat and shear that impacts off-target tissues.[2] Although advantages of organic sonosensitizers exist, such as high reproducibility, biocompatibility, production of reactive oxygen species, they also have limitations.[2] Factors that limit the translation of organic sensitizers to clinical applications include low water solubility, sonotoxicity, and targetability as well as high phototoxiticty.[2] Other properties could promote rapid clearance of the drug, which is why various nano and microparticles are used to transport the drug to the desired location.[2] In addition, sonosensitizers in SDT often require increased dosage, and the relationship between therapeutic dosage and toxicity of sonosensitizers has not been properly characterized alongside other variables such as tissue type and acoustic pressure.[31] Inorganic sensitizers produce reactive oxygen species, but in lower concentrations than desirable for SDT, limiting their ability to be used in a clinical setting.[2] Another challenge is reflected in vitro and in vivo work. An example of this can be seen in a study using rose bengal, a xanthene dye.[1] It was found to be successful in vitro, but in vivo showed significantly less efficacy due to liver squestation and clearance.[1] Lastly, there are no current standardized computer simulations to predict the characteristics of different sonosenistizers within tissue, which would provide further insight into how sonosensitizers may behave.[16]

Current Clinical Use edit

 
Photofrin

SDT has been researched most commonly to combat cancers and atherosclerosis such as breast cancer, pancreatic cancer, liver, and spinal sarcomas.[7][3][8][9][13][12][11][10][16][15][14][77] Currently, there are no FDA approved clinical applications of SDT. However, for PDT, Photofrin is an FDA approved hematoporphyrin (PHOTOFRIN®). However, SDT has been used in a clinical trial in combination with PDT to assess for reduction in tumor size in patients with breast cancer.[1] However, it was difficult to determine if SDT PDT or the drug dosage was the primary mechanism of treatment.[1] Another case study expanded on this by using SDT as a standalone treatment with a Gc protein hormone therapy with the use of 5-ALA or chlorin e6 as a sonosensitizer. It was shown that tumor markers significantly decreased during treatment.[1][78]

Future Directions edit

 
Enhanced drug uptake using acoustic targeted drug delivery (ATDD).

The effectiveness of sonodynamic therapy as a cancer treatment is supported by many in vitro and in vivo studies.[1] However, large-scale clinical trials are necessary for translation into the clinical setting. In order to mitigate the limitations aforementioned, new sonosensitizers are being developed and SDT is being combined with other therapies in novel ways. Particularly, organic sonosensitizers with high solubility in water, high sonotoxocity, increased ability to target tumors, and low phototoxicity need to be developed in order to improve the therapeutic efficacy of SDT and allow it to be used for treating cancers.[2] In addition, the mechanisms by which ROS are produced by sonosensitizers upon exposure to ultrasound is yet to be determined, reducing the ability to control its function and outcomes. Ultimately, the synergistic effects of combining SDT with other therapies would allow each to compensate for the limitations of the other, improving their therapeutic efficacy and increasing their ability to destroy tumors.[2]

References edit

  1. ^ a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae af ag ah ai aj ak al am an ao ap aq ar as at au av aw ax ay az ba bb bc bd be bf Costley, David; Mc Ewan, Conor; Fowley, Colin; McHale, Anthony P.; Atchison, Jordan; Nomikou, Nikolitsa; Callan, John F. (17 February 2015). "Treating cancer with sonodynamic therapy: A review". International Journal of Hyperthermia. 31 (2): 107–117. doi:10.3109/02656736.2014.992484. ISSN 0265-6736. PMID 25582025. S2CID 23665143.
  2. ^ a b c d e f g h i j k Xing, Xuejian; Zhao, Shaojing; Xu, Ting; Huang, Li; Zhang, Yi; Lan, Minhuan; Lin, Changwei; Zheng, Xiuli; Wang, Pengfei (15 October 2021). "Advances and perspectives in organic sonosensitizers for sonodynamic therapy". Coordination Chemistry Reviews. 445: 214087. doi:10.1016/j.ccr.2021.214087. ISSN 0010-8545.
  3. ^ a b c d e f g h i j k l Geng, Chi; Zhang, Yunlong; Hidru, Tesfaldet Habtemariam; Zhi, Lianyun; Tao, Mengxing; Zou, Leixin; Chen, Chen; Li, Huihua; Liu, Ying (15 August 2018). "Sonodynamic therapy: A potential treatment for atherosclerosis". Life Sciences. 207: 304–313. doi:10.1016/j.lfs.2018.06.018. ISSN 0024-3205. PMID 29940244. S2CID 49404799.
  4. ^ a b c Jiang, Yongxing; Fan, Jingxue; Li, Yong; Wu, Guodong; Wang, Yuanqi; Yang, Jiemei; Wang, Mengjiao; Cao, Zhengyu; Li, Qiannan; Wang, Hui; Zhang, Zhengyan; Wang, Yu; Li, Bicheng; Sun, Fengyu; Zhang, Haiyu; Zhang, Zhiguo; Li, Kang; Tian, Ye (15 February 2021). "Rapid reduction in plaque inflammation by sonodynamic therapy inpatients with symptomatic femoropopliteal peripheral artery disease:A randomized controlled trial". International Journal of Cardiology. 325: 132–139. doi:10.1016/j.ijcard.2020.09.035. ISSN 0167-5273. PMID 32966832. S2CID 221884358.
  5. ^ a b c Wang, Yu; Wang, Wei; Xu, Haobo; Sun, Yan; Sun, Jing; Jiang, Yongxing; Yao, Jianting; Tian, Ye (2017). "Non-Lethal Sonodynamic Therapy Inhibits Atherosclerotic Plaque Progression in ApoE-/- Mice and Attenuates ox-LDL-mediated Macrophage Impairment by Inducing Heme Oxygenase-1". Cellular Physiology and Biochemistry. 41 (6): 2432–2446. doi:10.1159/000475913. ISSN 1015-8987. PMID 28468003. S2CID 32744546.
  6. ^ a b c d e Browning, Richard J.; Able, Sarah; Ruan, Jia-Ling; Bau, Luca; Allen, Philip D.; Kersemans, Veerle; Wallington, Sheena; Kinchesh, Paul; Smart, Sean; Kartsonaki, Christiana; Kamila, Sukanta; Logan, Keiran; Taylor, Mark A.; McHale, Anthony P.; Callan, John F.; Stride, Eleanor; Vallis, Katherine A. (10 September 2021). "Combining sonodynamic therapy with chemoradiation for the treatment of pancreatic cancer". Journal of Controlled Release. 337: 371–377. doi:10.1016/j.jconrel.2021.07.020. ISSN 0168-3659. PMID 34274382.
  7. ^ a b Fan, Ching-Hsiang; Ting, Chien-Yu; Liu, Hao-Li; Huang, Chiung-Yin; Hsieh, Han-Yi; Yen, Tzu-Chen; Wei, Kuo-Chen; Yeh, Chih-Kuang (1 March 2013). "Antiangiogenic-targeting drug-loaded microbubbles combined with focused ultrasound for glioma treatment". Biomaterials. 34 (8): 2142–2155. doi:10.1016/j.biomaterials.2012.11.048. ISSN 0142-9612. PMID 23246066.
  8. ^ a b Hadi, Marym Mohammad; Nesbitt, Heather; Masood, Hamzah; Sciscione, Fabiola; Patel, Shiv; Ramesh, Bala S.; Emberton, Mark; Callan, John F.; MacRobert, Alexander; McHale, Anthony P.; Nomikou, Nikolitsa (10 January 2021). "Investigating the performance of a novel pH and cathepsin B sensitive, stimulus-responsive nanoparticle for optimised sonodynamic therapy in prostate cancer". Journal of Controlled Release. 329: 76–86. doi:10.1016/j.jconrel.2020.11.040. ISSN 0168-3659. PMC 8551370. PMID 33245955.
  9. ^ a b McHale, Anthony P.; Callan, John F.; Nomikou, Nikolitsa; Fowley, Colin; Callan, Bridgeen (2016). "Sonodynamic Therapy: Concept, Mechanism and Application to Cancer Treatment". Therapeutic Ultrasound. Advances in Experimental Medicine and Biology. Vol. 880. pp. 429–450. doi:10.1007/978-3-319-22536-4_22. ISBN 978-3-319-22535-7. PMID 26486350.
  10. ^ a b Pan, Xueting; Wang, Hongyu; Wang, Shunhao; Sun, Xiao; Wang, Lingjuan; Wang, Weiwei; Shen, Heyun; Liu, Huiyu (1 April 2018). "Sonodynamic therapy (SDT): a novel strategy for cancer nanotheranostics". Science China Life Sciences. 61 (4): 415–426. doi:10.1007/s11427-017-9262-x. ISSN 1869-1889. PMID 29666990. S2CID 4937368.
  11. ^ a b Sun, Shengjie; Wu, Meiying (1 January 2021). "Sonodynamic therapy: Another "light" in tumor treatment by exogenous stimulus". Smart Materials in Medicine. 2: 145–149. doi:10.1016/j.smaim.2021.05.001. ISSN 2590-1834. S2CID 236730960.
  12. ^ a b c d Tachibana, Katsuro; Feril, Loreto B.; Ikeda-Dantsuji, Yurika (1 August 2008). "Sonodynamic therapy". Ultrasonics. 48 (4): 253–259. doi:10.1016/j.ultras.2008.02.003. ISSN 0041-624X. PMID 18433819.
  13. ^ a b c Wan, Guo-Yun; Liu, Yang; Chen, Bo-Wei; Liu, Yuan-Yuan; Wang, Yin-Song; Zhang, Ning (September 2016). "Recent advances of sonodynamic therapy in cancer treatment". Cancer Biology & Medicine. 13 (3): 325–338. doi:10.20892/j.issn.2095-3941.2016.0068. ISSN 2095-3941. PMC 5069838. PMID 27807500.
  14. ^ a b c d e f Wang, Xiaobing; Jia, Yali; Wang, Pan; Liu, Quanhon; Zheng, Hairong (1 July 2017). "Current status and future perspectives of sonodynamic therapy in glioma treatment". Ultrasonics Sonochemistry. 37: 592–599. doi:10.1016/j.ultsonch.2017.02.020. ISSN 1350-4177. PMID 28427672.
  15. ^ a b Wu, Nan; Fan, Ching-Hsiang; Yeh, Chih-Kuang (2 March 2022). "Ultrasound-activated nanomaterials for sonodynamic cancer theranostics". Drug Discovery Today. 27 (6): 1590–1603. doi:10.1016/j.drudis.2022.02.025. ISSN 1359-6446. PMID 35247594. S2CID 247244458.
  16. ^ a b c Zhou, Yiming; Wang, Mengxuan; Dai, Zhifei (30 July 2020). "The molecular design of and challenges relating to sensitizers for cancer sonodynamic therapy". Materials Chemistry Frontiers. 4 (8): 2223–2234. doi:10.1039/D0QM00232A. ISSN 2052-1537. S2CID 225442575.
  17. ^ a b c Yamaguchi, Toshihiro; Kitahara, Shuji; Kusuda, Kaori; Okamoto, Jun; Horise, Yuki; Masamune, Ken; Muragaki, Yoshihiro (8 December 2021). "Current Landscape of Sonodynamic Therapy for Treating Cancer". Cancers. 13 (24): 6184. doi:10.3390/cancers13246184. ISSN 2072-6694. PMC 8699567. PMID 34944804.
  18. ^ a b c d e Dai, Zhi-Jun; Li, Sha; Gao, Jie; Xu, Xiao-Na; Lu, Wang-Feng; Lin, Shuai; Wang, Xi-Jing (1 March 2013). "Sonodynamic therapy (SDT): A novel treatment of cancer based on sonosensitizer liposome as a new drug carrier". Medical Hypotheses. 80 (3): 300–302. doi:10.1016/j.mehy.2012.12.009. ISSN 0306-9877. PMID 23294609.
  19. ^ a b Mišík, Vladimír; Riesz, Peter (25 January 2006). "Free Radical Intermediates in Sonodynamic Therapy". Annals of the New York Academy of Sciences. 899 (1): 335–348. doi:10.1111/j.1749-6632.2000.tb06198.x. PMID 10863551. S2CID 13503189.
  20. ^ a b c d e f g h i Hachimine, Ken; Shibaguchi, Hirotomo; Kuroki, Motomu; Yamada, Hiromi; Kinugasa, Tetsushi; Nakae, Yoshinori; Asano, Ryuji; Sakata, Isao; Yamashita, Yuichi; Shirakusa, Takayuki; Kuroki, Masahide (June 2007). "Sonodynamic therapy of cancer using a novel porphyrin derivative, DCPH-P-Na(I), which is devoid of photosensitivity". Cancer Science. 98 (6): 916–920. doi:10.1111/j.1349-7006.2007.00468.x. ISSN 1347-9032. PMID 17419708. S2CID 25732120.
  21. ^ a b c d Umemura, Shin-ichiro; Yumita, Nagahiko; Nishigaki, Ryuichiro; Umemura, Koshiro (September 1990). "Mechanism of Cell Damage by Ultrasound in Combination with Hematoporphyrin". Japanese Journal of Cancer Research. 81 (9): 962–966. doi:10.1111/j.1349-7006.1990.tb02674.x. PMC 5918111. PMID 2172198.
  22. ^ Hiraoka, Wakako; Honda, Hidemi; Feril, Loreto B.; Kudo, Nobuki; Kondo, Takashi (1 September 2006). "Comparison between sonodynamic effect and photodynamic effect with photosensitizers on free radical formation and cell killing". Ultrasonics Sonochemistry. 13 (6): 535–542. doi:10.1016/j.ultsonch.2005.10.001. ISSN 1350-4177. PMID 16325451.
  23. ^ Miyoshi, Norio; Igarashi, Takashi; Riesz, Peter (1 July 2000). "Evidence against singlet oxygen formation by sonolysis of aqueous oxygen-saturated solutions of Hematoporphyrin and Rose Bengal: The mechanism of sonodynamic therapy". Ultrasonics Sonochemistry. 7 (3): 121–124. doi:10.1016/S1350-4177(99)00042-5. ISSN 1350-4177. PMID 10909730.
  24. ^ a b c Liao, Ai-Ho; Li, Ying-Kai; Lee, Wei-Jiunn; Wu, Ming-Fang; Liu, Hao-Li; Kuo, Min-Liang (1 November 2012). "Estimating the Delivery Efficiency of Drug-Loaded Microbubbles in Cancer Cells with Ultrasound and Bioluminescence Imaging". Ultrasound in Medicine & Biology. 38 (11): 1938–1948. doi:10.1016/j.ultrasmedbio.2012.07.013. ISSN 0301-5629. PMID 22929655.
  25. ^ Pickworth, M J W; Dendy, P P; Leighton, T G; Walton, A J (1 November 1988). "Studies of the cavitational effects of clinical ultrasound by sonoluminescence: 2. Thresholds for sonoluminescence from a therapeutic ultrasound beam and the effect of temperature and duty cycle". Physics in Medicine and Biology. 33 (11): 1249–1260. Bibcode:1988PMB....33.1249P. doi:10.1088/0031-9155/33/11/003. ISSN 0031-9155. S2CID 250766457.
  26. ^ Saksena, T. K.; Nyborg, W. L. (1 September 1970). "Sonoluminescence from Stable Cavitation". The Journal of Chemical Physics. 53 (5): 1722–1734. Bibcode:1970JChPh..53.1722S. doi:10.1063/1.1674249. ISSN 0021-9606.
  27. ^ Gaitan, D. Felipe; Crum, Lawrence A.; Church, Charles C.; Roy, Ronald A. (1 June 1992). "Sonoluminescence and bubble dynamics for a single, stable, cavitation bubble". The Journal of the Acoustical Society of America. 91 (6): 3166–3183. Bibcode:1992ASAJ...91.3166G. doi:10.1121/1.402855. ISSN 0001-4966. S2CID 122235287.
  28. ^ Kessel, David; Lo, Julie; Jeffers, Russell; Brian Fowlkes, J.; Cain, Charles (1 June 1995). "Modes of photodynamic vs. sonodynamic cytotoxicity". Journal of Photochemistry and Photobiology B: Biology. 28 (3): 219–221. doi:10.1016/1011-1344(94)07111-Z. ISSN 1011-1344. PMID 7623187.
  29. ^ Yumita, Nagahiko; Iwase, Yumiko; Nishi, Koji; Ikeda, Toshihiko; Umemura, Shin-Ichiro; Sakata, Isao; Momose, Yasunori (1 June 2010). "Sonodynamically Induced Cell Damage and Membrane Lipid Peroxidation by Novel Porphyrin Derivative, DCPH-P-Na(I)". Anticancer Research. 30 (6): 2241–2246. ISSN 0250-7005. PMID 20651375.
  30. ^ a b c d Honda, Hidemi; Kondo, Takashi; Zhao, Qing-Li; Feril, Loreto B; Kitagawa, Hiroshi (May 2004). "Role of intracellular calcium ions and reactive oxygen species in apoptosis induced by ultrasound". Ultrasound in Medicine & Biology. 30 (5): 683–692. doi:10.1016/j.ultrasmedbio.2004.02.008. PMID 15183235.
  31. ^ a b c d e f Gong, Zhuoran; Dai, Zhifei (12 March 2021). "Design and Challenges of Sonodynamic Therapy System for Cancer Theranostics: From Equipment to Sensitizers". Advanced Science. 8 (10): 2002178. doi:10.1002/advs.202002178. ISSN 2198-3844. PMC 8132157. PMID 34026428.
  32. ^ Araújo Martins, Yugo; Zeferino Pavan, Theo; Fonseca Vianna Lopez, Renata (15 December 2021). "Sonodynamic therapy: Ultrasound parameters and in vitro experimental configurations". International Journal of Pharmaceutics. 610: 121243. doi:10.1016/j.ijpharm.2021.121243. ISSN 0378-5173. PMID 34743959. S2CID 240248013.
  33. ^ Suzuki, Norio; Okada, Kyoji; Chida, Shuichi; Komori, Chiyo; Shimada, Yoichi; Suzuki, Toshio (2007). "Antitumor Effect of Acridine Orange Under Ultrasonic Irradiation In Vitro". Anticancer Research. 27 (6B): 4179–4184. PMID 18225589.
  34. ^ Komori, Chiyo; Okada, Kyoji; Kawamura, Koichi; Chida, Shuichi; Suzuki, Toshio (2009). "The Sonodynamic Antitumor Effect of Methylene Blue on Sarcoma180 Cells In Vitro". Anticancer Research. 29 (6): 2411–2415. PMID 19528509.
  35. ^ Waksman, Ron; McEwan, Pauline E.; Moore, Travis I.; Pakala, Rajbabu; Kolodgie, Frank D.; Hellinga, David G.; Seabron, Rufus C.; Rychnovsky, Steven J.; Vasek, Jeffrey; Scott, Robert W.; Virmani, Renu (16 September 2008). "PhotoPoint Photodynamic Therapy Promotes Stabilization of Atherosclerotic Plaques and Inhibits Plaque Progression". Journal of the American College of Cardiology. 52 (12): 1024–1032. doi:10.1016/j.jacc.2008.06.023. PMID 18786486.
  36. ^ a b Nomikou, Nikolitsa; Sterrett, Christine; Arthur, Ciara; McCaughan, Bridgeen; Callan, John F.; McHale, Anthony P. (August 2012). "The Effects of Ultrasound and Light on Indocyanine-Green-Treated Tumour Cells and Tissues". ChemMedChem. 7 (8): 1465–1471. doi:10.1002/cmdc.201200233. PMID 22715137. S2CID 28851902.
  37. ^ Franco, Marina Santiago; Gomes, Eliza Rocha; Roque, Marjorie Coimbra; Oliveira, Mônica Cristina (2021). "Triggered Drug Release From Liposomes: Exploiting the Outer and Inner Tumor Environment". Frontiers in Oncology. 11: 623760. doi:10.3389/fonc.2021.623760. ISSN 2234-943X. PMC 8008067. PMID 33796461.
  38. ^ a b c d e Bozzuto, Giuseppina; Molinari, Agnese (2 February 2015). "Liposomes as nanomedical devices". International Journal of Nanomedicine. 10: 975–999. doi:10.2147/IJN.S68861. ISSN 1176-9114. PMC 4324542. PMID 25678787.
  39. ^ a b c Chen, Huaqing; Liu, Lanlan; Ma, Aiqing; Yin, Ting; Chen, Ze; Liang, Ruijing; Qiu, Yuzhi; Zheng, Mingbin; Cai, Lintao (1 February 2021). "Noninvasively immunogenic sonodynamic therapy with manganese protoporphyrin liposomes against triple-negative breast cancer". Biomaterials. 269: 120639. doi:10.1016/j.biomaterials.2020.120639. ISSN 0142-9612. PMID 33434714. S2CID 231595969.
  40. ^ a b c Liu, Yichen; Bai, Lianmei; Guo, Kaili; Jia, Yali; Zhang, Kun; Liu, Quanhong; Wang, Pan; Wang, Xiaobing (9 July 2019). "Focused ultrasound-augmented targeting delivery of nanosonosensitizers from homogenous exosomes for enhanced sonodynamic cancer therapy". Theranostics. 9 (18): 5261–5281. doi:10.7150/thno.33183. ISSN 1838-7640. PMC 6691590. PMID 31410214.
  41. ^ a b Liang, Yujie; Duan, Li; Lu, Jianping; Xia, Jiang (1 January 2021). "Engineering exosomes for targeted drug delivery". Theranostics. 11 (7): 3183–3195. doi:10.7150/thno.52570. ISSN 1838-7640. PMC 7847680. PMID 33537081.
  42. ^ a b Patil, Suyash M.; Sawant, Shruti S.; Kunda, Nitesh K. (1 September 2020). "Exosomes as drug delivery systems: A brief overview and progress update". European Journal of Pharmaceutics and Biopharmaceutics. 154: 259–269. doi:10.1016/j.ejpb.2020.07.026. ISSN 0939-6411. PMID 32717385. S2CID 220839948.
  43. ^ a b Nguyen Cao, Thuy Giang; Kang, Ji Hee; You, Jae Young; Kang, Han Chang; Rhee, Won Jong; Ko, Young Tag; Shim, Min Suk (9 June 2021). "Safe and Targeted Sonodynamic Cancer Therapy Using Biocompatible Exosome-Based Nanosonosensitizers". ACS Applied Materials & Interfaces. 13 (22): 25575–25588. doi:10.1021/acsami.0c22883. ISSN 1944-8244. PMID 34033477. S2CID 235204026.
  44. ^ a b c d e McEwan, Conor; Fowley, Colin; Nomikou, Nikolitsa; McCaughan, Bridgeen; McHale, Anthony P.; Callan, John F. (16 December 2014). "Polymeric Microbubbles as Delivery Vehicles for Sensitizers in Sonodynamic Therapy". Langmuir. 30 (49): 14926–14930. doi:10.1021/la503929c. ISSN 0743-7463. PMID 25409533.
  45. ^ a b Xing, Lingxi; Shi, Qiusheng; Zheng, Kailiang; Shen, Ming; Ma, Jing; Li, Fan; Liu, Yang; Lin, Lizhou; Tu, Wenzhi; Duan, Yourong; Du, Lianfang (2016). "Ultrasound-Mediated Microbubble Destruction (UMMD) Facilitates the Delivery of CA19-9 Targeted and Paclitaxel Loaded mPEG-PLGA-PLL Nanoparticles in Pancreatic Cancer". Theranostics. 6 (10): 1573–1587. doi:10.7150/thno.15164. PMC 4955056. PMID 27446491.
  46. ^ a b c McEwan, Conor; Owen, Joshua; Stride, Eleanor; Fowley, Colin; Nesbitt, Heather; Cochrane, David; Coussios, Constantin. C.; Borden, M.; Nomikou, Nikolitsa; McHale, Anthony P.; Callan, John F. (10 April 2015). "Oxygen carrying microbubbles for enhanced sonodynamic therapy of hypoxic tumours". Journal of Controlled Release. 203: 51–56. doi:10.1016/j.jconrel.2015.02.004. ISSN 0168-3659. PMID 25660073.
  47. ^ Lin, Xiahui; Qiu, Yuan; Song, Liang; Chen, Shan; Chen, Xiaofeng; Huang, Guoming; Song, Jibin; Chen, Xiaoyuan; Yang, Huanghao (23 April 2019). "Ultrasound activation of liposomes for enhanced ultrasound imaging and synergistic gas and sonodynamic cancer therapy". Nanoscale Horizons. 4 (3): 747–756. Bibcode:2019NanoH...4..747L. doi:10.1039/C8NH00340H. ISSN 2055-6764. S2CID 104403902.
  48. ^ a b Nesbitt, Heather; Sheng, Yingjie; Kamila, Sukanta; Logan, Keiran; Thomas, Keith; Callan, Bridgeen; Taylor, Mark A.; Love, Mark; O'Rourke, Declan; Kelly, Paul; Beguin, Estelle; Stride, Eleanor; McHale, Anthony P.; Callan, John F. (10 June 2018). "Gemcitabine loaded microbubbles for targeted chemo-sonodynamic therapy of pancreatic cancer". Journal of Controlled Release. 279: 8–16. doi:10.1016/j.jconrel.2018.04.018. ISSN 0168-3659. PMID 29653222. S2CID 4929495.
  49. ^ Shen, Z P; Brayman, A A; Chen, L; Miao, C H (August 2008). "Ultrasound with microbubbles enhances gene expression of plasmid DNA in the liver via intraportal delivery". Gene Therapy. 15 (16): 1147–1155. doi:10.1038/gt.2008.51. PMC 3747825. PMID 18385766.
  50. ^ a b c d e f Nittayacharn, Pinunta; Abenojar, Eric; La Deda, Massimo; Ricciardi, Loredana; Strangi, Giuseppe; Exner, Agata A. (6 March 2021). "Iridium(III) Complex-Loaded Perfluoropropane Nanobubbles for Enhanced Sonodynamic Therapy". Bioconjugate Chemistry. 33 (6): 1057–1068. doi:10.1021/acs.bioconjchem.1c00082. ISSN 1043-1802. PMC 10108504. PMID 33677967. S2CID 232143610.
  51. ^ Perera, Reshani H.; de Leon, Al; Wang, Xinning; Wang, Yu; Ramamurthy, Gopal; Peiris, Pubudu; Abenojar, Eric; Basilion, James P.; Exner, Agata A. (1 August 2020). "Real time ultrasound molecular imaging of prostate cancer with PSMA-targeted nanobubbles". Nanomedicine: Nanotechnology, Biology and Medicine. 28: 102213. doi:10.1016/j.nano.2020.102213. ISSN 1549-9634. PMC 7605099. PMID 32348874.
  52. ^ a b c d e Owen, Joshua; Logan, Keiran; Nesbitt, Heather; Able, Sarah; Vasilyeva, Alexandra; Bluemke, Emma; Kersemans, Veerle; Smart, Sean; Vallis, Katherine A.; McHale, Anthony P.; Callan, John F.; Stride, Eleanor (February 2022). "Orally administered oxygen nanobubbles enhance tumor response to sonodynamic therapy". Nano Select. 3 (2): 394–401. doi:10.1002/nano.202100038. ISSN 2688-4011. S2CID 237906086.
  53. ^ a b Liu, Yichen; Wang, Pan; Liu, Quanhong; Wang, Xiaobing (1 July 2016). "Sinoporphyrin sodium triggered sono-photodynamic effects on breast cancer both in vitro and in vivo". Ultrasonics Sonochemistry. 31: 437–448. doi:10.1016/j.ultsonch.2016.01.038. ISSN 1350-4177. PMID 26964970.
  54. ^ a b c d e Borah, Ballav M.; Cacaccio, Joseph; Durrani, Farukh A.; Bshara, Wiam; Turowski, Steven G.; Spernyak, Joseph A.; Pandey, Ravindra K. (11 December 2020). "Sonodynamic therapy in combination with photodynamic therapy shows enhanced long-term cure of brain tumor". Scientific Reports. 10 (1): 21791. Bibcode:2020NatSR..1021791B. doi:10.1038/s41598-020-78153-0. ISSN 2045-2322. PMC 7732989. PMID 33311561.
  55. ^ a b Huang, Ping; Qian, Xiaoqin; Chen, Yu; Yu, Luodan; Lin, Han; Wang, Liying; Zhu, Yufang; Shi, Jianlin (25 January 2017). "Metalloporphyrin-Encapsulated Biodegradable Nanosystems for Highly Efficient Magnetic Resonance Imaging-Guided Sonodynamic Cancer Therapy". Journal of the American Chemical Society. 139 (3): 1275–1284. doi:10.1021/jacs.6b11846. ISSN 0002-7863. PMID 28024395.
  56. ^ a b Lin, Xiaoning; Huang, Rong; Huang, Yanlin; Wang, Kai; Li, Heng; Bao, Yiheng; Wu, Chaohui; Zhang, Yi; Tian, Xinhua; Wang, Xiaomin (5 March 2021). "Nanosonosensitizer-Augmented Sonodynamic Therapy Combined with Checkpoint Blockade for Cancer Immunotherapy". International Journal of Nanomedicine. 16: 1889–1899. doi:10.2147/IJN.S290796. ISSN 1176-9114. PMC 7943542. PMID 33707944.
  57. ^ Yue, Wenwen; Chen, Liang; Yu, Luodan; Zhou, Bangguo; Yin, Haohao; Ren, Weiwei; Liu, Chang; Guo, Lehang; Zhang, Yifeng; Sun, Liping; Zhang, Kun; Xu, Huixiong; Chen, Yu (2 May 2019). "Checkpoint blockade and nanosonosensitizer-augmented noninvasive sonodynamic therapy combination reduces tumour growth and metastases in mice". Nature Communications. 10 (1): 2025. Bibcode:2019NatCo..10.2025Y. doi:10.1038/s41467-019-09760-3. ISSN 2041-1723. PMC 6497709. PMID 31048681.
  58. ^ a b c d Gao, Zhongxiuzi; Zheng, Jinhua; Yang, Bin; Wang, Zhu; Fan, Haixia; Lv, Yanhong; Li, Haixia; Jia, Limin; Cao, Wenwu (10 July 2013). "Sonodynamic therapy inhibits angiogenesis and tumor growth in a xenograft mouse model". Cancer Letters. 335 (1): 93–99. doi:10.1016/j.canlet.2013.02.006. ISSN 0304-3835. PMID 23402818.
  59. ^ Qu, Fei; Wang, Pan; Zhang, Kun; Shi, Yin; Li, Yixiang; Li, Chengren; Lu, Junhan; Liu, Quanhong; Wang, Xiaobing (2 August 2020). "Manipulation of Mitophagy by "All-in-One" nanosensitizer augments sonodynamic glioma therapy". Autophagy. 16 (8): 1413–1435. doi:10.1080/15548627.2019.1687210. PMC 7480814. PMID 31674265.
  60. ^ a b Waks, Adrienne G.; Winer, Eric P. (22 January 2019). "Breast Cancer Treatment: A Review". JAMA. 321 (3): 288–300. doi:10.1001/jama.2018.19323. ISSN 0098-7484. PMID 30667505. S2CID 58580711.
  61. ^ a b Zuo, Shuting; Zhang, Yan; Wang, Zhenyu; Wang, Jing (5 March 2022). "Mitochondria-Targeted Mesoporous Titanium Dioxide Nanoplatform for Synergistic Nitric Oxide Gas-Sonodynamic Therapy of Breast Cancer". International Journal of Nanomedicine. 17: 989–1002. doi:10.2147/IJN.S348618. PMC 8906874. PMID 35280333.
  62. ^ Alamolhoda, Mahboobeh; Mokhtari-Dizaji, Manijhe (24 June 2015). "Evaluation of fractionated and repeated sonodynamic therapy by using dual frequency for murine model of breast adenocarcinoma". Journal of Therapeutic Ultrasound. 3 (1): 10. doi:10.1186/s40349-015-0031-x. ISSN 2050-5736. PMC 4484850. PMID 26124951.
  63. ^ Feng, Qianhua; Yang, Xuemei; Hao, Yutong; Wang, Ning; Feng, Xuebing; Hou, Lin; Zhang, Zhenzhong (11 September 2019). "Cancer Cell Membrane-Biomimetic Nanoplatform for Enhanced Sonodynamic Therapy on Breast Cancer via Autophagy Regulation Strategy". ACS Applied Materials & Interfaces. 11 (36): 32729–32738. doi:10.1021/acsami.9b10948. ISSN 1944-8244. PMID 31415145. S2CID 201019401.
  64. ^ Huang, Biying; Chen, Sijie; Pei, Wenjing; Xu, Yan; Jiang, Zichao; Niu, Chengcheng; Wang, Long (2020). "Oxygen-Sufficient Nanoplatform for Chemo-Sonodynamic Therapy of Hypoxic Tumors". Frontiers in Chemistry. 8: 358. Bibcode:2020FrCh....8..358H. doi:10.3389/fchem.2020.00358. ISSN 2296-2646. PMC 7199163. PMID 32411675.
  65. ^ Zhang, Nan; Tan, Yang; Yan, Liwei; Zhang, Chunyang; Xu, Ming; Guo, Huanling; Zhuang, Bowen; Zhou, Luyao; Xie, Xiaoyan (6 August 2020). "Modulation of Tumor Hypoxia by pH-Responsive Liposomes to Inhibit Mitochondrial Respiration for Enhancing Sonodynamic Therapy". International Journal of Nanomedicine. 15: 5687–5700. doi:10.2147/IJN.S256038. ISSN 1176-9114. PMC 7418152. PMID 32821097.
  66. ^ Aksel, Mehran; Bozkurt-Girit, Ozlem; Bilgin, Mehmet Dincer (1 September 2020). "Pheophorbide a-mediated sonodynamic, photodynamic and sonophotodynamic therapies against prostate cancer". Photodiagnosis and Photodynamic Therapy. 31: 101909. doi:10.1016/j.pdpdt.2020.101909. ISSN 1572-1000. PMID 32619716. S2CID 220336627.
  67. ^ a b Evans, Andrew J. (January 2018). "Treatment effects in prostate cancer". Modern Pathology. 31 (1): 110–121. doi:10.1038/modpathol.2017.158. ISSN 1530-0285. PMID 29297495. S2CID 21337740.
  68. ^ a b c d Goertz, David E.; Todorova, Margarita; Mortazavi, Omid; Agache, Vlad; Chen, Branson; Karshafian, Raffi; Hynynen, Kullervo (20 December 2012). "Antitumor Effects of Combining Docetaxel (Taxotere) with the Antivascular Action of Ultrasound Stimulated Microbubbles". PLOS ONE. 7 (12): e52307. Bibcode:2012PLoSO...752307G. doi:10.1371/journal.pone.0052307. ISSN 1932-6203. PMC 3527530. PMID 23284980.
  69. ^ Jiang, Yueqing; Kou, Jiayuan; Han, Xiaobo; Li, Xuesong; Zhong, Zhaoyu; Liu, Zhongni; Zheng, Yinghong; Tian, Ye; Yang, Liming (2017). "ROS-Dependent Activation of Autophagy through the PI3K/Akt/mTOR Pathway Is Induced by Hydroxysafflor Yellow A-Sonodynamic Therapy in THP-1 Macrophages". Oxidative Medicine and Cellular Longevity. 2017: 1–16. doi:10.1155/2017/8519169. PMC 5278230. PMID 28191279.
  70. ^ Cheng, Jiali; Sun, Xin; Guo, Shuyuan; Cao, Wei; Chen, Haibo; Jin, Yinghua; Li, Bo; Li, Qiannan; Wang, Huan; Wang, Zhu; Zhou, Qi; Wang, Peng; Zhang, Zhiguo; Cao, Wenwu; Tian, Ye (2013). "Effects of 5-aminolevulinic acid-mediated sonodynamic therapy on macrophages". International Journal of Nanomedicine. 8: 669–676. doi:10.2147/IJN.S39844. ISSN 1176-9114. PMC 3576038. PMID 23426386.
  71. ^ Dan, Juhua; Sun, Xin; Li, Wanlu; Zhang, Yun; Li, Xuesong; Xu, Haobo; Li, Zhitao; Tian, Zhen; Guo, Shuyuan; Yao, Jianting; Gao, Weidong; Tian, Ye (1 June 2015). "5-Aminolevulinic Acid-Mediated Sonodynamic Therapy Promotes Phenotypic Switching from Dedifferentiated to Differentiated Phenotype via Reactive Oxygen Species and p38 Mitogen-Activated Protein Kinase in Vascular Smooth Muscle Cells". Ultrasound in Medicine & Biology. 41 (6): 1681–1689. doi:10.1016/j.ultrasmedbio.2014.12.664. ISSN 0301-5629. PMID 25796412.
  72. ^ Yumita, Nagahiko; Nishigaki, Ryuichiro; Umemura, Koshiro; Umemura, Shin-ichiro (March 1989). "Hematoporphyrin as a Sensitizer of Cell-damaging Effect of Ultrasound". Japanese Journal of Cancer Research. 80 (3): 219–222. doi:10.1111/j.1349-7006.1989.tb02295.x. PMC 5917717. PMID 2470713.
  73. ^ Ohmura, Tadahiro; Fukushima, Takeo; Shibaguchi, Hirotomo; Yoshizawa, Shin; Inoue, Tooru; Kuroki, Masahide; Sasaki, Kazunari; Umemura, Shin-Ichiro (1 July 2011). "Sonodynamic Therapy with 5-Aminolevulinic Acid and Focused Ultrasound for Deep-seated Intracranial Glioma in Rat". Anticancer Research. 31 (7): 2527–2533. ISSN 0250-7005. PMID 21873170.
  74. ^ a b c Zheng, Yilin; Ye, Jinxiang; Li, Ziying; Chen, Haijun; Gao, Yu (August 2021). "Recent progress in sono-photodynamic cancer therapy: From developed new sensitizers to nanotechnology-based efficacy-enhancing strategies". Acta Pharmaceutica Sinica B. 11 (8): 2197–2219. doi:10.1016/j.apsb.2020.12.016. ISSN 2211-3835. PMC 8424231. PMID 34522584.
  75. ^ Choi, Victor; Rajora, Maneesha A.; Zheng, Gang (15 April 2020). "Activating Drugs with Sound: Mechanisms Behind Sonodynamic Therapy and the Role of Nanomedicine". Bioconjugate Chemistry. 31 (4): 967–989. doi:10.1021/acs.bioconjchem.0c00029. ISSN 1043-1802. PMID 32129984. S2CID 212416405.
  76. ^ Cheng, Danling; Wang, Xiaoying; Zhou, Xiaojun; Li, Jingchao (2021). "Nanosonosensitizers With Ultrasound-Induced Reactive Oxygen Species Generation for Cancer Sonodynamic Immunotherapy". Frontiers in Bioengineering and Biotechnology. 9: 761218. doi:10.3389/fbioe.2021.761218. ISSN 2296-4185. PMC 8514668. PMID 34660560.
  77. ^ Lewis, Thomas J.; Mitchell, Doug (June 2008). "The Tumoricidal Effect of Sonodynamic Therapy (SDT) on S-180 Sarcoma in Mice". Integrative Cancer Therapies. 7 (2): 96–102. doi:10.1177/1534735408319065. ISSN 1534-7354. PMID 18550890. S2CID 22303568.
  78. ^ Inui, Toshio; Makita, Kaori; Miura, Hirona; Matsuda, Akiko; Kuchiike, Daisuke; Kubo, Kentaro; Mette, Martin; Uto, Yoshihiro; Nishikata, Takahito; Hori, Hitoshi; Sakamoto, Norihiro (1 August 2014). "Case Report: A Breast Cancer Patient Treated with GcMAF, Sonodynamic Therapy and Hormone Therapy". Anticancer Research. 34 (8): 4589–4593. ISSN 0250-7005. PMID 25075104.

sonodynamic, therapy, noninvasive, treatment, often, used, tumor, irradiation, that, utilizes, sonosensitizer, deep, penetration, ultrasound, treat, lesions, varying, depths, reducing, target, cell, number, preventing, future, tumor, growth, many, existing, ca. Sonodynamic therapy SDT is a noninvasive treatment often used for tumor irradiation that utilizes a sonosensitizer and the deep penetration of ultrasound to treat lesions of varying depths by reducing target cell number and preventing future tumor growth 1 2 Many existing cancer treatment strategies cause systemic toxicity or cannot penetrate tissue deep enough to reach the entire tumor however emerging ultrasound stimulated therapies could offer an alternative to these treatments with their increased efficiency greater penetration depth and reduced side effects Sonodynamic therapy could be used to treat cancers and other diseases such as atherosclerosis and diminish the risk associated with other treatment strategies since it induces cytotoxic effects only when externally stimulated by ultrasound and only at the cancerous region as opposed to the systemic administration of chemotherapy drugs 3 4 5 UV Radiation Photodynamic Therapy Reactive oxygen species ROS are an essential component of SDT as they provide the cytotoxicity of sonodynamic therapy they are produced when ultrasound is coupled with a sensitizing drug and molecular oxygen 1 Without ultrasound the drug is not toxic However once the drug is exposed to ultrasound and molecular oxygen it becomes toxic 1 Photodynamic therapy from which sonodynamic therapy was derived uses a similar mechanism Instead of ultrasound light is used to activate the drug 1 SDT allows the ultrasound to reach deeper into the tissue to about 30 centimeters compared to photodynamic therapy PDT since it can be highly focused 1 This increased penetration depth ultimately means that SDT can be utilized to treat deeper less accessible tumors and is more cost effective than PDT 6 1 Photodynamic therapy can be used in combination with sonodynamic therapy and is expanded upon in the Applications section of this article Sonodynamic therapy can be used synergistically with other therapeutic methods such as drug loaded microbubbles nanoparticles exosomes liposomes and genes for improved efficacy Currently SDT does not have any clinical products and acts as an adjuvant for the aforementioned therapeutic methods but it has been explored for use in atherosclerosis and cancer treatment to reduce tumor size in breast pancreas liver and spinal sarcomas 7 3 8 9 10 11 12 13 14 15 16 Contents 1 Mechanism of Action 1 1 Sonoluminescence 1 2 Pyrolysis 1 3 Lipid Peroxidation 1 4 Apoptosis 1 5 Autophagy 2 Sonosensitizers 2 1 Porphyrin based sensitizers 2 2 Xanthene based sensitizers 2 3 Additional sensitizers 3 Carriers 3 1 Liposomes 3 2 Exosomes 3 3 Microbubbles 3 4 Nanobubbles 4 Applications 4 1 Combination with other therapies 4 2 Types of cancers SDT has been shown to treat 4 2 1 Cancer Treatment 4 2 2 Breast Cancer 4 2 3 Glioma 4 2 4 Prostate Cancer 4 2 5 Arterial Diseases 4 3 In Vitro and In Vivo Work 4 3 1 In vitro 4 3 2 In vivo 5 Challenges and Development 6 Current Clinical Use 7 Future Directions 8 ReferencesMechanism of Action edit nbsp Cavitation bubble implosion The mechanism of action for sonodynamic therapy is the use of low intensity ultrasound through the use of focused mechanical waves to create a cytotoxic effect However SDT itself is non thermal non toxic and is able to non invasively penetrate deep into tissue compared to other delivery methods such as photodynamic therapy SDT is often performed alongside the use of a sonosensitizer such as porphyrin phthalocyanines xanthenes and antitumor drugs 17 Ultrasound waves are also classified as acoustic waves and the effect they have on the tissue of application can be described by a process called cavitation Cavitation occurs as a specific interaction between ultrasound and aqueous surroundings and causes gas bubbles to break upon exposure to particular ultrasonic parameters thus promoting penetration of the therapeutic into the biological tissues by generating cavities near the edge of the membrane 18 1 Cavitation can be broken down into stable and inertial cavitation In stable cavitation the oscillation of gas bubbles causes the environmental media to intermix 1 In inertial cavitation gas bubbles increase in volume and almost reach their resonance volume swelling before aggressively collapsing 1 The implosion of vesicles results in a drastic temperature and pressure change thereby increasing the cell membrane s permeability to various drugs 1 19 Microbubbles are created by the acoustic waves from the ultrasound that expand and collapse releasing energy bringing the sonosensitizer into an excited state and generating a ROS The cavitation of this gas bubble can form the ROS with different methodologies such as sonoluminescence and pyrolysis 1 Apoptosis results from the formation of ROS and mechanical forces of SDT through membrane disruption in a process called lipid peroxidation Necrosis is also a potential result of SDT The influence of sonoluminescence on SDT and ROS has not been fully elaborated within literature 1 Currently it is understood that sonoluminescence allows the emission of light upon bubble collapse which can activate sensitizers A study by Hachimine et al highlights the use of SDT as a method to activate a low photosensitive sonosensitizer DCPH P Na I for cancer that is too deep within the tissue to combat utilizing PDT without skin irritation 1 20 Pyrolysis raises the surrounding temperature enhances the cavitation process breaks down the sensitizer generating free radicals and the free radicals interact within their environment to generate ROS 1 For both methods the importance of the singlet oxygen compared to the hydroxyl radical to induce cytotoxicity has been highlighted 1 20 21 While other studies 1 22 23 have found the singlet oxygen to not have a substantial effect Overall both of these methodologies lack significant breadth in literature to fully explain their role in ROS formation However literature has shown success in their analysis and application 1 4 24 Sonoluminescence edit nbsp Sonoluminescence acoustics Two primary mechanisms of ROS generation exist in sonodynamic therapy sonoluminescence and pyrolysis 1 Sonoluminescence occurs when ultrasound produces light after irradiating an aqueous solution 1 25 The exact mechanism with which light is produced remains unclear However it is suggested that inertial cavitation is a key element for this process 1 26 Other studies also indicate the potential role of stable cavitation 1 27 Pyrolysis edit Pyrolysis is believed to occur when inertial cavitation induces an extreme temperature increase degrades the sonosensitizers thus producing free radicals that can react and ultimately produce ROS necessary for SDT 1 28 The localized temperature increase assists in the inertial cavitation and breakdown of the sonosensitizer in order to create ROS The pyrolysis within the cavitation bubbles will produce H and OH via weak bonding within the solute molecule 1 19 Lipid Peroxidation edit nbsp Mechanism of lipid peroxidation In addition to chemical methods mechanical properties of the acoustic wave generated from the ultrasound can assist in initiating cytotoxic effects This occurs through disruption of the membrane with a hydrophobic sonosensitizer The mechanical disruption of the membrane causes a process called lipid peroxidation and adjustments to the cell membrane can change cell drug permeability 1 29 Both sonochemical and sonomechanical methodologies are used to generate ROS and release cargo from vesicles for applications such as tumor targeting Apoptosis edit Low intensity ultrasound has been shown within past literature to induce apoptotic effects within surrounding cells It has been found that it is not the initial ROS that causes apoptosis within the cells but the free radicals within the mitochondria In a study by Honda et al it was determined that the mitochondria caspase pathway is responsible for apoptosis through the increase of intracellular calcium 1 30 Outside of ROS induced apoptosis cavitation is another factor involved within apoptosis of surrounding cells Both cavitation types are able to induce apoptosis through damage to the membrane Conditions such as frequency duty cycle pulse and intensity can be manipulated to optimize cell death conditions such as necrosis lysis or apoptosis 31 24 32 Autophagy edit This method of cell death can occur by cell organelles becoming entrapped into autophagosomes that combine with lysosomes Continuation of this process will lead to cell death and autophagy inhibitors or promoters can be controlled to encourage or discourage cell death and uptake of chemotherapeutics 1 Sonosensitizers editSonosensitizers or sonosensitizing therapeutics are the primary element of SDT and can be tailored to treat various cancers and generate different effects 2 These therapeutics often involving the use of porphyrin or xanthene will initiate a toxic effect via the ROS upon exposure to ultrasound Porphyrin based sensitizers edit nbsp The 18 electron cycle of porphin the parent structure of porphyrin highlighted Several other choices of atoms through the pyrrole nitrogens for example also give 18 electron cycles Porphyrin based sensitizers initially used as a photosensitizer in PDT are fairly hydrophobic molecules derived from hematoporphyrin 1 Single oxygen atoms or hydroxyl radicals are produced by porphyrin based sensitizers upon exposure to ultrasound or light providing the cytotoxic effects desired with sonodynamic and photodynamic therapies 1 However the result of porphyrin based sensitizers is not as local as desired for sonodynamic therapy since they are also located in non targeted tissue between the tumor and the ultrasound emitter 1 Xanthene based sensitizers edit nbsp Xanthene nbsp Rose Bengal Xanthene based sensitizers on the other hand have shown successful cytotoxicity in vitro by producing reactive oxygen species after being triggered by ultrasound 1 More research is necessary to improve its potential in vivo performance since it is quickly processed by the liver and cleared from the body 1 Rose Bengal is a commonly used xanthene based sonosensitizer 1 Additional sensitizers edit Other sensitizers that have been investigated for their potential in sonodynamic therapy and have also been used previously in PDT include acridine orange methylene blue curcumin and indocyanine green 1 A study by Suzuki et al used acridine orange a fluorescent cationic dye that can insert itself into nucleic acids for treating sarcoma 180 cells with ultrasound and demonstrated that reactive oxygen species are a critical element of SDT considering that their absence decreased the efficacy of SDT 33 Similar to the previous study a recent study by Komori et al utilized ultrasound coupled with methylene blue a phenothiazine dye commonly used in PDT that exhibits low toxicity to irradiate sarcoma 180 cells and found that methylene blue was an effective sonosensitizer in decreasing cell viability 34 Interestingly curcumin is a spice that also can act as a sensitizer for PDT and SDT 1 In a study by Waksman et al curcumin was able to impact macrophages which are important for development of plaques found in atherosclerosis patients thus reducing the amount of plaque in an animal model 35 These findings along with other research indicate that curcumin sensitizers could be used in SDT cancer treatments Indocyanine green is a dye that absorbs near infrared wavelengths and is another sensitizer that has been shown to reduce cell viability when coupled with ultrasound and or light 36 An in vivo study demonstrated that treating a mouse tumor model with indocyanine green coupled with ultrasound and light resulted in a 98 reduction in tumor volume by 27 days after treatment 36 Name and structure of additional sensitizers Name Structure Phthalocyanine nbsp Phthalocyanine Indocyanine Green nbsp Indocyanine Green Phenothiazine nbsp Phenothiazine Curcumin keto nbsp Curcumin structure Keto Curcumin enol nbsp Curcumin enolCarriers editAs aforementioned sonosensitizers are often used in conjunction with different drug carriers such as microbubbles nanobubbles liposomes and exosomes to improve therapeutic agent concentration and penetration 18 Liposomes edit nbsp Liposome Drug Incorporation Liposomes are a common vehicle in drug delivery and specifically for the treatment of cancer Liposomes contain a phospholipid bilayer It is prevalent due to its ability to penetrate leaky vasculature and poor lymphatic drainage within tumors for enhanced permeability retention 37 These drug carriers can encapsulate hydrophobic and lipophilic molecules within their lipid bilayer and can be made naturally or synthetically 38 39 In addition liposomes can entrap hydrophilic molecules in their hydrophilic core 38 Compared to the common cancer treatment chemotherapy drugs loaded into liposomes allow for decreased systemic toxicity and a potential increase in the efficacy of targeted delivery 18 Success with liposomes as drug delivery systems has been shown both in vivo and in vitro 38 A study by Liu et al showed that liposomes can be used alongside SDT to trigger the release of drugs via oxidation of the lipid components 40 Another study by Ninomiya et al utilized nanoemulsion droplets exposed to ultrasonic waves for the formation of larger gas bubbles to disrupt the liposome membrane for drug release Many properties and elements of liposomes can be altered for their specific purpose and to increase effectiveness particularly their ability to travel in the blood and interact with cells and tissues in the body 38 These elements include their diameter charge arrangement as well as the makeup of their membranes 38 Dai et al proposed the incorporation of sonosensitizers with liposomes to enhance target specificity 18 Since SDT stimulates cancerous tissues to absorb and retain sonosentizers followed by activation with extracorporeal ultrasound Dai et al investigated the effect of liposome encapsulated drugs on the efficacy of targeted delivery in SDT They found that in addition to its convenience and practicality SDT is a safe and effective option for treating cancer 18 Exosomes edit Exosomes are nanocarriers that can provide targeted drug delivery of therapeutics to enhance local cytotoxic effects while minimizing any systemic impact They are acquired from cells and are used for transportation purposes within the cell as membrane bound vesicles Advantages of exosomes for drug delivery purposes include their ability to be manipulated and engineered in addition to their low toxicity and immunogenicity 41 42 They have also inspired research into non cell based treatment methods for various cancers and diseases 41 Other desirable aspects of exosomes include their overall biocompatibility and stability 42 A study by Nguyen Cao et al investigated the use of exosomes for the delivery of indocyanine green ICG a sonosensitizer for breast cancer treatment 43 Significantly increased reactive oxygen species generation was observed in breast cancer cells treated with folic acid conjugated exosomes 43 This is one example of a sonosensitizer used to treat a specific cancer using sonodynamic therapy Another example of exosome based sonodynamic therapy was illustrated by Liu et al In this study exosomes were decorated with porphyrin sensitizers and this system was used with an external ultrasound device to control and target drug delivery through SDT 40 Liu et al provided a non invasive method for treating cancer through extracorporeal activation of exosomes through ultrasound 40 Microbubbles edit nbsp Mechanisms for Loading Microbubbles with Drug Due to their ability to oscillate with exposure to low frequency ultrasound microbubbles have been used as contrast agents in order to visualize tissues in which the microbubbles have permeated 44 However when these microspheres are exposed to higher pressure ultrasound they can rupture which could be beneficial for drug delivery purposes 44 Through SDT these microbubbles could be selectively bursted at the tumor microenvironment in order to decrease systemic levels of the encapsulated drug and increase therapeutic efficacy When applying SDT the increase in acoustic pressure leads to the inertial cavitation or collapse of the microbubble and local release of the cargo within The inertial cavitation of the microbubbles when exposed to SDT is also referred to as ultrasound mediated microbubble destruction UMMD 45 The shell of microbubbles can be decorated with different components including polymers lipids or proteins depending on their intended purpose 44 Microbubbles have also been used for the localized release of attached cargo This cargo is typically chemotherapeutics antibiotics or genes 12 Different drugs can be directly loaded into the microbubble with methods such as conjugation and nanoparticle liposome loading and genes The combination of genes and SDT is referred to as sonotransfection 12 Examples of outer shell modifications can be seen in a study by McEwan et al which found that lipid microbubbles showed reduced stability when sonosensitizers were added to their shells 44 However attaching the polymer poly lactic co glycolic acid PLGA to the shell resulted in increased stability compared to the lipid microbubbles without losing other desirable properties such as targeted delivery and selective cytotoxicity 44 In another study McEwan et al investigated the ability of microbubbles carrying oxygen to increase production of reactive oxygen species which are a necessary component of SDT in the hypoxic environment of many solid tumors 46 These microbubbles were stabilized with lipids and a Rose Bengal sonosensitizer was attached to the surface to treat pancreatic cancer 46 Their work showed that coupling oxygen loaded microbubbles that are sensitive to ultrasound with sonosensitizing drugs could allow for increased drug activation at the desired target even if hypoxia is present Examples of therapeutics that have been loaded into microbubbles are gemcitabine paclitaxel nanoparticles plasmid DNA and 2 2 azobis 2 2 imidazolin 2 yl propane dihydrochloride loaded liposomes 47 45 48 49 Due to the targeting nature of the ligands connected to the microbubble it allows for the controlled and specific targeting of the desired tissue for treatment Another study performed by Nesbitt et al has shown improved tumor reduction when gemcitabine was loaded into the microbubble and applied to a human pancreatic cancer xenograft model with SDT 48 Nanobubbles edit Similar to microbubbles nanobubbles have shown efficacy in SDT 50 However due to their smaller size nanobubbles are able to reach targets that microbubbles cannot Nanobubbles can reach deeper tissue and travel past the vasculature Previous research has demonstrated that nanobubbles are more capable of reaching the tumor since they can permeate endothelial cells and migrate away from the vasculature 51 50 One study by Nittayacharn et al developed doxorubicin loaded nanobubbles and paired them with porphyrin sensitizers to be used in SDT for treatment of breast and ovarian cancer cells in vitro 50 They found an almost 70 increase in cytotoxicity when using SDT compared to only perfluoropropane nanobubbles filled with iridium III 50 Additionally compared to empty nanobubbles and or free iridium III they observed greatest reactive oxygen species generation in the iridium III nanobubbles exposed to ultrasound 50 These results demonstrate that nanobubbles loaded with a sonosensitizer and exposed to ultrasound could be a potential effective treatment for cancer using SDT As with microbubbles nanobubbles have also shown promise as oxygen delivering vesicles to enhance the effectiveness of SDT In order to mitigate hypoxia of target tissue Owen et al used a pancreatic cancer rodent model to deliver phospholipid stabilized nanobubbles filled with oxygen 52 The mice were divided into groups one that received oxygen filled nanobubbles prior to injection of a sonosensitizer and one that didn t 52 A statistically significant difference between the levels of oxygen in the tumors of the two groups was observed indicating that nanobubbles could be an effective addition to SDT to treat cancers in a hypoxic environment 52 Applications editCombination with other therapies edit Sonodynamic therapy can be combined with other therapeutic techniques to enhance treatment efficacy for various types of cancers and diseases SDT can be combined with photodynamic therapy chemotherapy radiation MRI and immunotherapy PDT has often been used in combination with SDT as sonosensitizers are also photosensitive 1 During initial development of SDT Umemura et al have determined that hematoporphyrins were able to initiate cell death similarly to PDT 21 This is due to SDT being able to initiate sonoluminescence However the advantage of SDT over PDT is that it can penetrate deep and precisely into the targeted tissue In a study by Lui et al it was shown that using a combination of these two delivery methods results in increased cytotoxicity with sino porphyrin in a metastatic xenograft model 53 In another example of combining SDT with PDT Borah et al investigated the advantage of 2 1 hexyloxyethyl 2 devinyl pyropheophorbide a HHPH a photodynamic therapy drug as a sonosensitizer and a photosensitizer for treating glioblastoma 54 Combining these therapies showed increased cell kill tumor response possibly caused by synergistic effects 54 The goal of a study by Browning et al was to investigate the potential enhancement of chemoradiation efficacy through combining it with sonodynamic therapy in pancreatic cancer patients In one model survival increased with the combination compared to chemoradiation alone Differences in the results for the two different models could be attributed to variations in tumor organization 6 The tumors that showed the greatest reduction in size were less vascularized perhaps making them more vulnerable to SDT 6 Another study by Huang et al used elements of mesoporous organosilica based nanosystems to fabricate a sonosensitizer to be used with MRI guided SDT 55 Increased cell death and inhibiting tumor growth was induced by the sonosensitizers indicating high SDT efficiency 55 This shows how SDT can assist with both removal and inhibition of tumor growth SDT has also been combined with immunotherapy A study by Lin et al aimed to use cascade immuno sonodynamic therapy to enhance tumor treatment using antibodies 56 The nanosonosensitizers resulted in high drug loading efficiency and a tumor specific adaptive immune response This serves as an example as to how SDT can be coupled with checkpoint blockade immunotherapy to enhance efficiency in cancer treatments Another study by Yue et al strived to combine checkpoint blockade immunotherapy with nanosonosensitizers augmented noninvasive sonodynamic therapy 57 Along with inhibiting lung metastasis this combination promoted an anti tumor response that prohibited tumor growth This provides a proof of concept for combining SDT with another therapy to enhance treatment effects for the short and long term Types of cancers SDT has been shown to treat edit Cancer Treatment edit The treatment of many different types of cancers has been investigated using sonodynamic therapy both in vitro and or in vivo including glioblastoma pancreatic breast ovarian lung prostate liver stomach and colon cancers 54 6 20 50 52 A study by Gao et al showed that SDT is capable of inhibiting angiogenesis through the production of ROS This hindered the proliferation migration and invasion of endothelial cells tumor growth intratumoral vascularity and vascular endothelial growth factor expression within the tumor cell in xenograft rat models 58 Hachimine et al performed a large in vitro study testing SDT on seventeen different cancer cell lines 20 The types of cancers included were pancreatic breast lung prostate liver stomach and colon cancers 20 The most successful treatment was that of lung cancer with 23 4 cell viability post therapy 1 20 Qu et al aimed to develop an all in one nanosensitizer platform triggered by SDT that combines various diagnostic and therapeutic effects to treat glioblastoma 59 Apoptosis was successfully induced and mitophagy was inhibited in glioma cells This is an example of how SDT can be used with a different platform to treat glioblastoma Borah et al as mentioned above also investigated the ability of SDT and PDT to treat glioblastoma and found that SDT combined with PDT was able to increase the number of tumor cells killed 54 McEwan et al and Owen et al both demonstrated the use of micro nanobubbles to enhance the oxygen concentration near hypoxic pancreatic tumors thereby increasing the efficacy of SDT 46 52 Breast Cancer edit nbsp Morphologic and immunohistochemical features of Triple Negative Breast Cancer 12 of women in the US will be diagnosed with breast cancer 60 Metastasis and recurrence is a large challenge for deep seated solid state tumors 39 SDT is currently being explored as a treatment method for breast cancer while avoiding the side effects associated with current therapeutic methods 61 There has been shown success in utilizing SDT in animal and human clinical trials in reduction of tumor size through mitochondrial targeting to initiate apoptosis of tumor cells and autophagy and immune response regulation 62 39 63 64 24 53 60 65 61 However there are still complications with proper therapeutic efficacy when used alone Glioma edit nbsp Glioblastoma Malignant glioma is an extremely difficult to treat brain tumor that is a leading cause of death worldwide and half of cancer related deaths 14 Complications associated with treating glioma include the blood brain barrier BBB 14 This protective mechanism for the brain also raises challenges for drug delivery through the tight junctions between endothelial cells only allowing small lipid soluble drugs lt 400 Da to permeate 14 Current delivery methods are surgery and chemotherapy SDT has been implemented as a method to open the BBB and has shown success in opening tight junctions for delivery Examples of sonosensitizers that have shown success in glioma treatment are hematopor phyrin monomethyl ether HMME porfimer sodium Photofrin di sulfo di phthalimidomethyl phthalolcyaninezinc ZnPcS2P2 Photolon 5 aminolevulinic acid 5 ALA and rose bengal RB 14 These have shown to induce effects such as opening of the BBB improved vascular permeability and apoptosis of glioma cells Prostate Cancer edit nbsp Benign prostatic hyperplasia Prostate cancer is the second cause of cancer and the most common malignancy associated with deaths in men worldwide 66 Current methods of treatments are invasive resection therapy radiation therapy and prostatectomy that can cause complications such as incontinence impotence and damage to surrounding organs and tissues 67 17 Current studies have shown success in using SDT as a stand alone treatment 68 SDT uses mitochondria related apoptosis for the reduction of cell viability SDT for prostate cancer treatment has also been used alongside chemotherapeutics such as docetaxel microbubbles 17 67 68 This has shown to enhance the effects of docetaxel through a reduction in tumor perfusion and enhanced necrosis and apoptosis 68 The SDT and docetaxel group showed reduction in tumor growth 68 Overall the use of SDT has shown promising results in prostate cancer treatment Arterial Diseases edit Sonodynamic therapy could be used to treat more than just cancers Atherosclerosis which is a chronic arterial disease is another target that has been observed in the literature 3 5 This disease occurs when fatty plaques aggregate on the inner surface of the artery and could be caused by malfunctions in lipid metabolism 3 More specifically atherosclerosis is caused by an increase in endothelial permeability causing low density lipoprotein particles to become oxidized and undergo sedimentation 3 These lipoproteins cause an increase in macrophages and lead to intensified plaque build up As a result the high influx of macrophages is the target for AS treatment in order to slow plaque build up 3 Alongside the relationship between plaque build up and macrophages monocyte s differentiation into macrophages exacerbates the aforementioned process in addition to causing inflammation 3 nbsp Normal Artery versus Atherosclerosis A study by Wang et al aimed to understand the underlying mechanisms regarding the potential effect of non lethal SDT on atheroscleroic plaques It was determined that non lethal SDT prevents plaque development 5 A study performed by Jiang et al showed success in SDT through the reduction of macrophage inflammatory factors such as TNF alpha IL 12 and IL 1B They also showed that SDT could inhibit plaque inflammation in patients with peripheral artery disease and continue to promote positive results for longer than six months 4 Popular sonosensitizers for AS treatment are protoporphyrin IX PpIX and 5 aminolevulinic acid 5 ALA 69 3 PpIX is often used in PDT and is generated through 5 ALA a non ultrasound activated component through increasing PpIX concentration within a cell A study by Cheng et al determined that THP 1 macrophage apoptosis is induced by an increase in PpiX concentration leading to the production of large amounts of ROS 70 13 3 The use of SDT for AS treatment has also shown success in promoting the repopulation of vascular smooth muscle cells VMSCs through inducing further expression and autophagy to prevent VMSC evolution into plaque holding macrophages A study performed by Dan et al showed the increase in smooth muscle a actin smooth muscle 22a p38 mitogen activated protein kinase phosphorylation 71 3 While a study by Geng et al showed improved VMSC autophagy Each of these factors contributed to the improved differentiation and development of VMSCs 3 In Vitro and In Vivo Work edit In vitro edit In vitro experimentation provides great insight and knowledge to characterize the potential of sonosensitizer behavior in vivo In addition SDT has shown success through its low intensity allowing increased plasma membrane permeability without cell death 1 Sonosensitizers have also been used in vitro in applications with different cell lines and to further understand the mechanism of action for cell death It is currently understood that PDT and SDT have similar mechanisms for free radical generation for inducing apoptosis and necrosis 1 However each cell line is unique and can cause cell death with different efficacy 20 1 72 Some examples of in vitro work include initial studies that were performed by Yumita et al 1989 who used haematoprophyrin and SDT for mouse sarcoma 180 and rat ascites hepatoma AH that showed a relationship between dosage and ultrasound and microbubbles causing cavitation leading to cell damage without the use of drugs This study also emphasized the difference in efficacy between cell lines through SDT 180 having less lysis compared to AH 130 cells Another study by Hachimine et al emphasized efficacy between cell lines by examining seven different cancers with 17 cell lines total under the use of DCPH P NA I 1 20 This study revealed that the stomach and lung cancer lines of MKN 28 and LU65A respectively had the highest survival rate but the stomach and lung cancer lines of RERFLC KJ and MKN 45 respectively had the lowest survival rates 20 1 Another study by Honda et al with U937 and K562 showed that sonication increases the intracellular calcium ion levels and decreases GSH concentration respectively 30 This increased concentration of calcium plays a significant role in cell death through DNA fragmentation and mitochondrial membrane disruption 1 30 While a decreased concentration of GSH plays a significant role in allowing the formation of more free radicals 30 1 A study by Umemura et al found that ATX 70 versus hematoporphyrin has increased cytotoxic activity 21 1 Current research typically focuses on using tumor xenograft models to determine the effect of SDT on target cells and delivery efficacy 1 In vivo edit Building upon the study by Umemura et al and ATX 70 it was found that 24h after administration of the sonosensitizer had improved efficacy when ultrasound was applied compared to immediate administration 21 1 It was also determined that most ultrasound frequencies range between 1 3 MHz and 0 5 4W cm 2 Higher frequencies at values such as 20W cm 2 and 25W cm 2 resulted in large necrotic lesions 73 1 This established a relationship between sonosensitizer formulation and ultrasound intensity to necrosis Other studies have continued to innovate upon this by controlling drug ultrasound interval DUI for different sonosensitizers in order to determine the optimal time period to apply the ultrasound for improved efficacy 58 1 In addition it has been shown that SDT can disturb surrounding vasculature in tumors 1 58 This has been shown in studies by Gao et al with 5 ALA in mice and human umbilical vein endothelial cell lines through inhibition of microvessel density and cell proliferation migration and invasion 58 1 Challenges and Development edit nbsp Ultrasound Imaging vs Ultrasound Therapy One of the many advantages of SDT compared to PDT is the ability of SDT to penetrate deeply placed solid tumors allowing a wider treatment range 1 Despite this fact there are limitations to SDT that must be overcome or have optimized components in order to expand the effect and application of SDT 31 SDT does allow for precise activation of the therapeutic but is limited in the delivery and accumulation of the delivery modality to penetrate deeply into the desired tumor site 74 This is often accommodated for through delivery vessels such as nanoparticles or liposomes 1 However nanomedicine is limited by the enhanced permeability and retention effect and struggles to deliver in targeted abundance depending on the delivery vesicle 31 74 This can be seen in nanoparticles struggling with non specific delivery Future research has been focused on developing high targeting and penetrating nanoparticles for improved delivery and pharmacokinetics 75 31 Due to the complex nature of tumors and their microenvironments they are difficult to treat with only one therapy In order to enhance the oftentimes low production of reactive oxygen species to address the hypoxic tumor environment SDT can be combined with other therapies such as PDT chemotherapy and immunotherapy to improve patient outcomes 2 56 54 6 SDT alone does not respond well in hypoxic environments However bioreductive therapy could be used to reduce the impact of SDT s limitations regarding hypoxia in the tumor while leaving healthy normal tissue alone 2 Sonosensitizers also require continuous high levels of oxygen to create ROS which is not readily available within a hypoxic tumor microenvironment 31 However strategies such as oxygen supplementation and production to supply the required oxygen and enhance cavitation and glutathione depletion to avoid the reduction of the free radicals produced have been implemented alongside sonosensitizers to supply the required oxygen or reduce the combative function 76 74 In addition to its relatively low generation of reactive oxygen species SDT also can cause permanent destruction of normal tissues This lack of selectivity is caused by ultrasound divergence resulting in heat and shear that impacts off target tissues 2 Although advantages of organic sonosensitizers exist such as high reproducibility biocompatibility production of reactive oxygen species they also have limitations 2 Factors that limit the translation of organic sensitizers to clinical applications include low water solubility sonotoxicity and targetability as well as high phototoxiticty 2 Other properties could promote rapid clearance of the drug which is why various nano and microparticles are used to transport the drug to the desired location 2 In addition sonosensitizers in SDT often require increased dosage and the relationship between therapeutic dosage and toxicity of sonosensitizers has not been properly characterized alongside other variables such as tissue type and acoustic pressure 31 Inorganic sensitizers produce reactive oxygen species but in lower concentrations than desirable for SDT limiting their ability to be used in a clinical setting 2 Another challenge is reflected in vitro and in vivo work An example of this can be seen in a study using rose bengal a xanthene dye 1 It was found to be successful in vitro but in vivo showed significantly less efficacy due to liver squestation and clearance 1 Lastly there are no current standardized computer simulations to predict the characteristics of different sonosenistizers within tissue which would provide further insight into how sonosensitizers may behave 16 Current Clinical Use edit nbsp Photofrin SDT has been researched most commonly to combat cancers and atherosclerosis such as breast cancer pancreatic cancer liver and spinal sarcomas 7 3 8 9 13 12 11 10 16 15 14 77 Currently there are no FDA approved clinical applications of SDT However for PDT Photofrin is an FDA approved hematoporphyrin PHOTOFRIN However SDT has been used in a clinical trial in combination with PDT to assess for reduction in tumor size in patients with breast cancer 1 However it was difficult to determine if SDT PDT or the drug dosage was the primary mechanism of treatment 1 Another case study expanded on this by using SDT as a standalone treatment with a Gc protein hormone therapy with the use of 5 ALA or chlorin e6 as a sonosensitizer It was shown that tumor markers significantly decreased during treatment 1 78 Future Directions edit nbsp Enhanced drug uptake using acoustic targeted drug delivery ATDD The effectiveness of sonodynamic therapy as a cancer treatment is supported by many in vitro and in vivo studies 1 However large scale clinical trials are necessary for translation into the clinical setting In order to mitigate the limitations aforementioned new sonosensitizers are being developed and SDT is being combined with other therapies in novel ways Particularly organic sonosensitizers with high solubility in water high sonotoxocity increased ability to target tumors and low phototoxicity need to be developed in order to improve the therapeutic efficacy of SDT and allow it to be used for treating cancers 2 In addition the mechanisms by which ROS are produced by sonosensitizers upon exposure to ultrasound is yet to be determined reducing the ability to control its function and outcomes Ultimately the synergistic effects of combining SDT with other therapies would allow each to compensate for the limitations of the other improving their therapeutic efficacy and increasing their ability to destroy tumors 2 References edit a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae af ag ah ai aj ak al am an ao ap aq ar as at au av aw ax ay az ba bb bc bd be bf Costley David Mc Ewan Conor Fowley Colin McHale Anthony P Atchison Jordan Nomikou Nikolitsa Callan John F 17 February 2015 Treating cancer with sonodynamic therapy A review International Journal of Hyperthermia 31 2 107 117 doi 10 3109 02656736 2014 992484 ISSN 0265 6736 PMID 25582025 S2CID 23665143 a b c d e f g h i j k Xing Xuejian Zhao Shaojing Xu Ting Huang Li Zhang Yi Lan Minhuan Lin Changwei Zheng Xiuli Wang Pengfei 15 October 2021 Advances and perspectives in organic sonosensitizers for sonodynamic therapy Coordination Chemistry Reviews 445 214087 doi 10 1016 j ccr 2021 214087 ISSN 0010 8545 a b c d e f g h i j k l Geng Chi Zhang Yunlong Hidru Tesfaldet Habtemariam Zhi Lianyun Tao Mengxing Zou Leixin Chen Chen Li Huihua Liu Ying 15 August 2018 Sonodynamic therapy A potential treatment for atherosclerosis Life Sciences 207 304 313 doi 10 1016 j lfs 2018 06 018 ISSN 0024 3205 PMID 29940244 S2CID 49404799 a b c Jiang Yongxing Fan Jingxue Li Yong Wu Guodong Wang Yuanqi Yang Jiemei Wang Mengjiao Cao Zhengyu Li Qiannan Wang Hui Zhang Zhengyan Wang Yu Li Bicheng Sun Fengyu Zhang Haiyu Zhang Zhiguo Li Kang Tian Ye 15 February 2021 Rapid reduction in plaque inflammation by sonodynamic therapy inpatients with symptomatic femoropopliteal peripheral artery disease A randomized controlled trial International Journal of Cardiology 325 132 139 doi 10 1016 j ijcard 2020 09 035 ISSN 0167 5273 PMID 32966832 S2CID 221884358 a b c Wang Yu Wang Wei Xu Haobo Sun Yan Sun Jing Jiang Yongxing Yao Jianting Tian Ye 2017 Non Lethal Sonodynamic Therapy Inhibits Atherosclerotic Plaque Progression in ApoE Mice and Attenuates ox LDL mediated Macrophage Impairment by Inducing Heme Oxygenase 1 Cellular Physiology and Biochemistry 41 6 2432 2446 doi 10 1159 000475913 ISSN 1015 8987 PMID 28468003 S2CID 32744546 a b c d e Browning Richard J Able Sarah Ruan Jia Ling Bau Luca Allen Philip D Kersemans Veerle Wallington Sheena Kinchesh Paul Smart Sean Kartsonaki Christiana Kamila Sukanta Logan Keiran Taylor Mark A McHale Anthony P Callan John F Stride Eleanor Vallis Katherine A 10 September 2021 Combining sonodynamic therapy with chemoradiation for the treatment of pancreatic cancer Journal of Controlled Release 337 371 377 doi 10 1016 j jconrel 2021 07 020 ISSN 0168 3659 PMID 34274382 a b Fan Ching Hsiang Ting Chien Yu Liu Hao Li Huang Chiung Yin Hsieh Han Yi Yen Tzu Chen Wei Kuo Chen Yeh Chih Kuang 1 March 2013 Antiangiogenic targeting drug loaded microbubbles combined with focused ultrasound for glioma treatment Biomaterials 34 8 2142 2155 doi 10 1016 j biomaterials 2012 11 048 ISSN 0142 9612 PMID 23246066 a b Hadi Marym Mohammad Nesbitt Heather Masood Hamzah Sciscione Fabiola Patel Shiv Ramesh Bala S Emberton Mark Callan John F MacRobert Alexander McHale Anthony P Nomikou Nikolitsa 10 January 2021 Investigating the performance of a novel pH and cathepsin B sensitive stimulus responsive nanoparticle for optimised sonodynamic therapy in prostate cancer Journal of Controlled Release 329 76 86 doi 10 1016 j jconrel 2020 11 040 ISSN 0168 3659 PMC 8551370 PMID 33245955 a b McHale Anthony P Callan John F Nomikou Nikolitsa Fowley Colin Callan Bridgeen 2016 Sonodynamic Therapy Concept Mechanism and Application to Cancer Treatment Therapeutic Ultrasound Advances in Experimental Medicine and Biology Vol 880 pp 429 450 doi 10 1007 978 3 319 22536 4 22 ISBN 978 3 319 22535 7 PMID 26486350 a b Pan Xueting Wang Hongyu Wang Shunhao Sun Xiao Wang Lingjuan Wang Weiwei Shen Heyun Liu Huiyu 1 April 2018 Sonodynamic therapy SDT a novel strategy for cancer nanotheranostics Science China Life Sciences 61 4 415 426 doi 10 1007 s11427 017 9262 x ISSN 1869 1889 PMID 29666990 S2CID 4937368 a b Sun Shengjie Wu Meiying 1 January 2021 Sonodynamic therapy Another light in tumor treatment by exogenous stimulus Smart Materials in Medicine 2 145 149 doi 10 1016 j smaim 2021 05 001 ISSN 2590 1834 S2CID 236730960 a b c d Tachibana Katsuro Feril Loreto B Ikeda Dantsuji Yurika 1 August 2008 Sonodynamic therapy Ultrasonics 48 4 253 259 doi 10 1016 j ultras 2008 02 003 ISSN 0041 624X PMID 18433819 a b c Wan Guo Yun Liu Yang Chen Bo Wei Liu Yuan Yuan Wang Yin Song Zhang Ning September 2016 Recent advances of sonodynamic therapy in cancer treatment Cancer Biology amp Medicine 13 3 325 338 doi 10 20892 j issn 2095 3941 2016 0068 ISSN 2095 3941 PMC 5069838 PMID 27807500 a b c d e f Wang Xiaobing Jia Yali Wang Pan Liu Quanhon Zheng Hairong 1 July 2017 Current status and future perspectives of sonodynamic therapy in glioma treatment Ultrasonics Sonochemistry 37 592 599 doi 10 1016 j ultsonch 2017 02 020 ISSN 1350 4177 PMID 28427672 a b Wu Nan Fan Ching Hsiang Yeh Chih Kuang 2 March 2022 Ultrasound activated nanomaterials for sonodynamic cancer theranostics Drug Discovery Today 27 6 1590 1603 doi 10 1016 j drudis 2022 02 025 ISSN 1359 6446 PMID 35247594 S2CID 247244458 a b c Zhou Yiming Wang Mengxuan Dai Zhifei 30 July 2020 The molecular design of and challenges relating to sensitizers for cancer sonodynamic therapy Materials Chemistry Frontiers 4 8 2223 2234 doi 10 1039 D0QM00232A ISSN 2052 1537 S2CID 225442575 a b c Yamaguchi Toshihiro Kitahara Shuji Kusuda Kaori Okamoto Jun Horise Yuki Masamune Ken Muragaki Yoshihiro 8 December 2021 Current Landscape of Sonodynamic Therapy for Treating Cancer Cancers 13 24 6184 doi 10 3390 cancers13246184 ISSN 2072 6694 PMC 8699567 PMID 34944804 a b c d e Dai Zhi Jun Li Sha Gao Jie Xu Xiao Na Lu Wang Feng Lin Shuai Wang Xi Jing 1 March 2013 Sonodynamic therapy SDT A novel treatment of cancer based on sonosensitizer liposome as a new drug carrier Medical Hypotheses 80 3 300 302 doi 10 1016 j mehy 2012 12 009 ISSN 0306 9877 PMID 23294609 a b Misik Vladimir Riesz Peter 25 January 2006 Free Radical Intermediates in Sonodynamic Therapy Annals of the New York Academy of Sciences 899 1 335 348 doi 10 1111 j 1749 6632 2000 tb06198 x PMID 10863551 S2CID 13503189 a b c d e f g h i Hachimine Ken Shibaguchi Hirotomo Kuroki Motomu Yamada Hiromi Kinugasa Tetsushi Nakae Yoshinori Asano Ryuji Sakata Isao Yamashita Yuichi Shirakusa Takayuki Kuroki Masahide June 2007 Sonodynamic therapy of cancer using a novel porphyrin derivative DCPH P Na I which is devoid of photosensitivity Cancer Science 98 6 916 920 doi 10 1111 j 1349 7006 2007 00468 x ISSN 1347 9032 PMID 17419708 S2CID 25732120 a b c d Umemura Shin ichiro Yumita Nagahiko Nishigaki Ryuichiro Umemura Koshiro September 1990 Mechanism of Cell Damage by Ultrasound in Combination with Hematoporphyrin Japanese Journal of Cancer Research 81 9 962 966 doi 10 1111 j 1349 7006 1990 tb02674 x PMC 5918111 PMID 2172198 Hiraoka Wakako Honda Hidemi Feril Loreto B Kudo Nobuki Kondo Takashi 1 September 2006 Comparison between sonodynamic effect and photodynamic effect with photosensitizers on free radical formation and cell killing Ultrasonics Sonochemistry 13 6 535 542 doi 10 1016 j ultsonch 2005 10 001 ISSN 1350 4177 PMID 16325451 Miyoshi Norio Igarashi Takashi Riesz Peter 1 July 2000 Evidence against singlet oxygen formation by sonolysis of aqueous oxygen saturated solutions of Hematoporphyrin and Rose Bengal The mechanism of sonodynamic therapy Ultrasonics Sonochemistry 7 3 121 124 doi 10 1016 S1350 4177 99 00042 5 ISSN 1350 4177 PMID 10909730 a b c Liao Ai Ho Li Ying Kai Lee Wei Jiunn Wu Ming Fang Liu Hao Li Kuo Min Liang 1 November 2012 Estimating the Delivery Efficiency of Drug Loaded Microbubbles in Cancer Cells with Ultrasound and Bioluminescence Imaging Ultrasound in Medicine amp Biology 38 11 1938 1948 doi 10 1016 j ultrasmedbio 2012 07 013 ISSN 0301 5629 PMID 22929655 Pickworth M J W Dendy P P Leighton T G Walton A J 1 November 1988 Studies of the cavitational effects of clinical ultrasound by sonoluminescence 2 Thresholds for sonoluminescence from a therapeutic ultrasound beam and the effect of temperature and duty cycle Physics in Medicine and Biology 33 11 1249 1260 Bibcode 1988PMB 33 1249P doi 10 1088 0031 9155 33 11 003 ISSN 0031 9155 S2CID 250766457 Saksena T K Nyborg W L 1 September 1970 Sonoluminescence from Stable Cavitation The Journal of Chemical Physics 53 5 1722 1734 Bibcode 1970JChPh 53 1722S doi 10 1063 1 1674249 ISSN 0021 9606 Gaitan D Felipe Crum Lawrence A Church Charles C Roy Ronald A 1 June 1992 Sonoluminescence and bubble dynamics for a single stable cavitation bubble The Journal of the Acoustical Society of America 91 6 3166 3183 Bibcode 1992ASAJ 91 3166G doi 10 1121 1 402855 ISSN 0001 4966 S2CID 122235287 Kessel David Lo Julie Jeffers Russell Brian Fowlkes J Cain Charles 1 June 1995 Modes of photodynamic vs sonodynamic cytotoxicity Journal of Photochemistry and Photobiology B Biology 28 3 219 221 doi 10 1016 1011 1344 94 07111 Z ISSN 1011 1344 PMID 7623187 Yumita Nagahiko Iwase Yumiko Nishi Koji Ikeda Toshihiko Umemura Shin Ichiro Sakata Isao Momose Yasunori 1 June 2010 Sonodynamically Induced Cell Damage and Membrane Lipid Peroxidation by Novel Porphyrin Derivative DCPH P Na I Anticancer Research 30 6 2241 2246 ISSN 0250 7005 PMID 20651375 a b c d Honda Hidemi Kondo Takashi Zhao Qing Li Feril Loreto B Kitagawa Hiroshi May 2004 Role of intracellular calcium ions and reactive oxygen species in apoptosis induced by ultrasound Ultrasound in Medicine amp Biology 30 5 683 692 doi 10 1016 j ultrasmedbio 2004 02 008 PMID 15183235 a b c d e f Gong Zhuoran Dai Zhifei 12 March 2021 Design and Challenges of Sonodynamic Therapy System for Cancer Theranostics From Equipment to Sensitizers Advanced Science 8 10 2002178 doi 10 1002 advs 202002178 ISSN 2198 3844 PMC 8132157 PMID 34026428 Araujo Martins Yugo Zeferino Pavan Theo Fonseca Vianna Lopez Renata 15 December 2021 Sonodynamic therapy Ultrasound parameters and in vitro experimental configurations International Journal of Pharmaceutics 610 121243 doi 10 1016 j ijpharm 2021 121243 ISSN 0378 5173 PMID 34743959 S2CID 240248013 Suzuki Norio Okada Kyoji Chida Shuichi Komori Chiyo Shimada Yoichi Suzuki Toshio 2007 Antitumor Effect of Acridine Orange Under Ultrasonic Irradiation In Vitro Anticancer Research 27 6B 4179 4184 PMID 18225589 Komori Chiyo Okada Kyoji Kawamura Koichi Chida Shuichi Suzuki Toshio 2009 The Sonodynamic Antitumor Effect of Methylene Blue on Sarcoma180 Cells In Vitro Anticancer Research 29 6 2411 2415 PMID 19528509 Waksman Ron McEwan Pauline E Moore Travis I Pakala Rajbabu Kolodgie Frank D Hellinga David G Seabron Rufus C Rychnovsky Steven J Vasek Jeffrey Scott Robert W Virmani Renu 16 September 2008 PhotoPoint Photodynamic Therapy Promotes Stabilization of Atherosclerotic Plaques and Inhibits Plaque Progression Journal of the American College of Cardiology 52 12 1024 1032 doi 10 1016 j jacc 2008 06 023 PMID 18786486 a b Nomikou Nikolitsa Sterrett Christine Arthur Ciara McCaughan Bridgeen Callan John F McHale Anthony P August 2012 The Effects of Ultrasound and Light on Indocyanine Green Treated Tumour Cells and Tissues ChemMedChem 7 8 1465 1471 doi 10 1002 cmdc 201200233 PMID 22715137 S2CID 28851902 Franco Marina Santiago Gomes Eliza Rocha Roque Marjorie Coimbra Oliveira Monica Cristina 2021 Triggered Drug Release From Liposomes Exploiting the Outer and Inner Tumor Environment Frontiers in Oncology 11 623760 doi 10 3389 fonc 2021 623760 ISSN 2234 943X PMC 8008067 PMID 33796461 a b c d e Bozzuto Giuseppina Molinari Agnese 2 February 2015 Liposomes as nanomedical devices International Journal of Nanomedicine 10 975 999 doi 10 2147 IJN S68861 ISSN 1176 9114 PMC 4324542 PMID 25678787 a b c Chen Huaqing Liu Lanlan Ma Aiqing Yin Ting Chen Ze Liang Ruijing Qiu Yuzhi Zheng Mingbin Cai Lintao 1 February 2021 Noninvasively immunogenic sonodynamic therapy with manganese protoporphyrin liposomes against triple negative breast cancer Biomaterials 269 120639 doi 10 1016 j biomaterials 2020 120639 ISSN 0142 9612 PMID 33434714 S2CID 231595969 a b c Liu Yichen Bai Lianmei Guo Kaili Jia Yali Zhang Kun Liu Quanhong Wang Pan Wang Xiaobing 9 July 2019 Focused ultrasound augmented targeting delivery of nanosonosensitizers from homogenous exosomes for enhanced sonodynamic cancer therapy Theranostics 9 18 5261 5281 doi 10 7150 thno 33183 ISSN 1838 7640 PMC 6691590 PMID 31410214 a b Liang Yujie Duan Li Lu Jianping Xia Jiang 1 January 2021 Engineering exosomes for targeted drug delivery Theranostics 11 7 3183 3195 doi 10 7150 thno 52570 ISSN 1838 7640 PMC 7847680 PMID 33537081 a b Patil Suyash M Sawant Shruti S Kunda Nitesh K 1 September 2020 Exosomes as drug delivery systems A brief overview and progress update European Journal of Pharmaceutics and Biopharmaceutics 154 259 269 doi 10 1016 j ejpb 2020 07 026 ISSN 0939 6411 PMID 32717385 S2CID 220839948 a b Nguyen Cao Thuy Giang Kang Ji Hee You Jae Young Kang Han Chang Rhee Won Jong Ko Young Tag Shim Min Suk 9 June 2021 Safe and Targeted Sonodynamic Cancer Therapy Using Biocompatible Exosome Based Nanosonosensitizers ACS Applied Materials amp Interfaces 13 22 25575 25588 doi 10 1021 acsami 0c22883 ISSN 1944 8244 PMID 34033477 S2CID 235204026 a b c d e McEwan Conor Fowley Colin Nomikou Nikolitsa McCaughan Bridgeen McHale Anthony P Callan John F 16 December 2014 Polymeric Microbubbles as Delivery Vehicles for Sensitizers in Sonodynamic Therapy Langmuir 30 49 14926 14930 doi 10 1021 la503929c ISSN 0743 7463 PMID 25409533 a b Xing Lingxi Shi Qiusheng Zheng Kailiang Shen Ming Ma Jing Li Fan Liu Yang Lin Lizhou Tu Wenzhi Duan Yourong Du Lianfang 2016 Ultrasound Mediated Microbubble Destruction UMMD Facilitates the Delivery of CA19 9 Targeted and Paclitaxel Loaded mPEG PLGA PLL Nanoparticles in Pancreatic Cancer Theranostics 6 10 1573 1587 doi 10 7150 thno 15164 PMC 4955056 PMID 27446491 a b c McEwan Conor Owen Joshua Stride Eleanor Fowley Colin Nesbitt Heather Cochrane David Coussios Constantin C Borden M Nomikou Nikolitsa McHale Anthony P Callan John F 10 April 2015 Oxygen carrying microbubbles for enhanced sonodynamic therapy of hypoxic tumours Journal of Controlled Release 203 51 56 doi 10 1016 j jconrel 2015 02 004 ISSN 0168 3659 PMID 25660073 Lin Xiahui Qiu Yuan Song Liang Chen Shan Chen Xiaofeng Huang Guoming Song Jibin Chen Xiaoyuan Yang Huanghao 23 April 2019 Ultrasound activation of liposomes for enhanced ultrasound imaging and synergistic gas and sonodynamic cancer therapy Nanoscale Horizons 4 3 747 756 Bibcode 2019NanoH 4 747L doi 10 1039 C8NH00340H ISSN 2055 6764 S2CID 104403902 a b Nesbitt Heather Sheng Yingjie Kamila Sukanta Logan Keiran Thomas Keith Callan Bridgeen Taylor Mark A Love Mark O Rourke Declan Kelly Paul Beguin Estelle Stride Eleanor McHale Anthony P Callan John F 10 June 2018 Gemcitabine loaded microbubbles for targeted chemo sonodynamic therapy of pancreatic cancer Journal of Controlled Release 279 8 16 doi 10 1016 j jconrel 2018 04 018 ISSN 0168 3659 PMID 29653222 S2CID 4929495 Shen Z P Brayman A A Chen L Miao C H August 2008 Ultrasound with microbubbles enhances gene expression of plasmid DNA in the liver via intraportal delivery Gene Therapy 15 16 1147 1155 doi 10 1038 gt 2008 51 PMC 3747825 PMID 18385766 a b c d e f Nittayacharn Pinunta Abenojar Eric La Deda Massimo Ricciardi Loredana Strangi Giuseppe Exner Agata A 6 March 2021 Iridium III Complex Loaded Perfluoropropane Nanobubbles for Enhanced Sonodynamic Therapy Bioconjugate Chemistry 33 6 1057 1068 doi 10 1021 acs bioconjchem 1c00082 ISSN 1043 1802 PMC 10108504 PMID 33677967 S2CID 232143610 Perera Reshani H de Leon Al Wang Xinning Wang Yu Ramamurthy Gopal Peiris Pubudu Abenojar Eric Basilion James P Exner Agata A 1 August 2020 Real time ultrasound molecular imaging of prostate cancer with PSMA targeted nanobubbles Nanomedicine Nanotechnology Biology and Medicine 28 102213 doi 10 1016 j nano 2020 102213 ISSN 1549 9634 PMC 7605099 PMID 32348874 a b c d e Owen Joshua Logan Keiran Nesbitt Heather Able Sarah Vasilyeva Alexandra Bluemke Emma Kersemans Veerle Smart Sean Vallis Katherine A McHale Anthony P Callan John F Stride Eleanor February 2022 Orally administered oxygen nanobubbles enhance tumor response to sonodynamic therapy Nano Select 3 2 394 401 doi 10 1002 nano 202100038 ISSN 2688 4011 S2CID 237906086 a b Liu Yichen Wang Pan Liu Quanhong Wang Xiaobing 1 July 2016 Sinoporphyrin sodium triggered sono photodynamic effects on breast cancer both in vitro and in vivo Ultrasonics Sonochemistry 31 437 448 doi 10 1016 j ultsonch 2016 01 038 ISSN 1350 4177 PMID 26964970 a b c d e Borah Ballav M Cacaccio Joseph Durrani Farukh A Bshara Wiam Turowski Steven G Spernyak Joseph A Pandey Ravindra K 11 December 2020 Sonodynamic therapy in combination with photodynamic therapy shows enhanced long term cure of brain tumor Scientific Reports 10 1 21791 Bibcode 2020NatSR 1021791B doi 10 1038 s41598 020 78153 0 ISSN 2045 2322 PMC 7732989 PMID 33311561 a b Huang Ping Qian Xiaoqin Chen Yu Yu Luodan Lin Han Wang Liying Zhu Yufang Shi Jianlin 25 January 2017 Metalloporphyrin Encapsulated Biodegradable Nanosystems for Highly Efficient Magnetic Resonance Imaging Guided Sonodynamic Cancer Therapy Journal of the American Chemical Society 139 3 1275 1284 doi 10 1021 jacs 6b11846 ISSN 0002 7863 PMID 28024395 a b Lin Xiaoning Huang Rong Huang Yanlin Wang Kai Li Heng Bao Yiheng Wu Chaohui Zhang Yi Tian Xinhua Wang Xiaomin 5 March 2021 Nanosonosensitizer Augmented Sonodynamic Therapy Combined with Checkpoint Blockade for Cancer Immunotherapy International Journal of Nanomedicine 16 1889 1899 doi 10 2147 IJN S290796 ISSN 1176 9114 PMC 7943542 PMID 33707944 Yue Wenwen Chen Liang Yu Luodan Zhou Bangguo Yin Haohao Ren Weiwei Liu Chang Guo Lehang Zhang Yifeng Sun Liping Zhang Kun Xu Huixiong Chen Yu 2 May 2019 Checkpoint blockade and nanosonosensitizer augmented noninvasive sonodynamic therapy combination reduces tumour growth and metastases in mice Nature Communications 10 1 2025 Bibcode 2019NatCo 10 2025Y doi 10 1038 s41467 019 09760 3 ISSN 2041 1723 PMC 6497709 PMID 31048681 a b c d Gao Zhongxiuzi Zheng Jinhua Yang Bin Wang Zhu Fan Haixia Lv Yanhong Li Haixia Jia Limin Cao Wenwu 10 July 2013 Sonodynamic therapy inhibits angiogenesis and tumor growth in a xenograft mouse model Cancer Letters 335 1 93 99 doi 10 1016 j canlet 2013 02 006 ISSN 0304 3835 PMID 23402818 Qu Fei Wang Pan Zhang Kun Shi Yin Li Yixiang Li Chengren Lu Junhan Liu Quanhong Wang Xiaobing 2 August 2020 Manipulation of Mitophagy by All in One nanosensitizer augments sonodynamic glioma therapy Autophagy 16 8 1413 1435 doi 10 1080 15548627 2019 1687210 PMC 7480814 PMID 31674265 a b Waks Adrienne G Winer Eric P 22 January 2019 Breast Cancer Treatment A Review JAMA 321 3 288 300 doi 10 1001 jama 2018 19323 ISSN 0098 7484 PMID 30667505 S2CID 58580711 a b Zuo Shuting Zhang Yan Wang Zhenyu Wang Jing 5 March 2022 Mitochondria Targeted Mesoporous Titanium Dioxide Nanoplatform for Synergistic Nitric Oxide Gas Sonodynamic Therapy of Breast Cancer International Journal of Nanomedicine 17 989 1002 doi 10 2147 IJN S348618 PMC 8906874 PMID 35280333 Alamolhoda Mahboobeh Mokhtari Dizaji Manijhe 24 June 2015 Evaluation of fractionated and repeated sonodynamic therapy by using dual frequency for murine model of breast adenocarcinoma Journal of Therapeutic Ultrasound 3 1 10 doi 10 1186 s40349 015 0031 x ISSN 2050 5736 PMC 4484850 PMID 26124951 Feng Qianhua Yang Xuemei Hao Yutong Wang Ning Feng Xuebing Hou Lin Zhang Zhenzhong 11 September 2019 Cancer Cell Membrane Biomimetic Nanoplatform for Enhanced Sonodynamic Therapy on Breast Cancer via Autophagy Regulation Strategy ACS Applied Materials amp Interfaces 11 36 32729 32738 doi 10 1021 acsami 9b10948 ISSN 1944 8244 PMID 31415145 S2CID 201019401 Huang Biying Chen Sijie Pei Wenjing Xu Yan Jiang Zichao Niu Chengcheng Wang Long 2020 Oxygen Sufficient Nanoplatform for Chemo Sonodynamic Therapy of Hypoxic Tumors Frontiers in Chemistry 8 358 Bibcode 2020FrCh 8 358H doi 10 3389 fchem 2020 00358 ISSN 2296 2646 PMC 7199163 PMID 32411675 Zhang Nan Tan Yang Yan Liwei Zhang Chunyang Xu Ming Guo Huanling Zhuang Bowen Zhou Luyao Xie Xiaoyan 6 August 2020 Modulation of Tumor Hypoxia by pH Responsive Liposomes to Inhibit Mitochondrial Respiration for Enhancing Sonodynamic Therapy International Journal of Nanomedicine 15 5687 5700 doi 10 2147 IJN S256038 ISSN 1176 9114 PMC 7418152 PMID 32821097 Aksel Mehran Bozkurt Girit Ozlem Bilgin Mehmet Dincer 1 September 2020 Pheophorbide a mediated sonodynamic photodynamic and sonophotodynamic therapies against prostate cancer Photodiagnosis and Photodynamic Therapy 31 101909 doi 10 1016 j pdpdt 2020 101909 ISSN 1572 1000 PMID 32619716 S2CID 220336627 a b Evans Andrew J January 2018 Treatment effects in prostate cancer Modern Pathology 31 1 110 121 doi 10 1038 modpathol 2017 158 ISSN 1530 0285 PMID 29297495 S2CID 21337740 a b c d Goertz David E Todorova Margarita Mortazavi Omid Agache Vlad Chen Branson Karshafian Raffi Hynynen Kullervo 20 December 2012 Antitumor Effects of Combining Docetaxel Taxotere with the Antivascular Action of Ultrasound Stimulated Microbubbles PLOS ONE 7 12 e52307 Bibcode 2012PLoSO 752307G doi 10 1371 journal pone 0052307 ISSN 1932 6203 PMC 3527530 PMID 23284980 Jiang Yueqing Kou Jiayuan Han Xiaobo Li Xuesong Zhong Zhaoyu Liu Zhongni Zheng Yinghong Tian Ye Yang Liming 2017 ROS Dependent Activation of Autophagy through the PI3K Akt mTOR Pathway Is Induced by Hydroxysafflor Yellow A Sonodynamic Therapy in THP 1 Macrophages Oxidative Medicine and Cellular Longevity 2017 1 16 doi 10 1155 2017 8519169 PMC 5278230 PMID 28191279 Cheng Jiali Sun Xin Guo Shuyuan Cao Wei Chen Haibo Jin Yinghua Li Bo Li Qiannan Wang Huan Wang Zhu Zhou Qi Wang Peng Zhang Zhiguo Cao Wenwu Tian Ye 2013 Effects of 5 aminolevulinic acid mediated sonodynamic therapy on macrophages International Journal of Nanomedicine 8 669 676 doi 10 2147 IJN S39844 ISSN 1176 9114 PMC 3576038 PMID 23426386 Dan Juhua Sun Xin Li Wanlu Zhang Yun Li Xuesong Xu Haobo Li Zhitao Tian Zhen Guo Shuyuan Yao Jianting Gao Weidong Tian Ye 1 June 2015 5 Aminolevulinic Acid Mediated Sonodynamic Therapy Promotes Phenotypic Switching from Dedifferentiated to Differentiated Phenotype via Reactive Oxygen Species and p38 Mitogen Activated Protein Kinase in Vascular Smooth Muscle Cells Ultrasound in Medicine amp Biology 41 6 1681 1689 doi 10 1016 j ultrasmedbio 2014 12 664 ISSN 0301 5629 PMID 25796412 Yumita Nagahiko Nishigaki Ryuichiro Umemura Koshiro Umemura Shin ichiro March 1989 Hematoporphyrin as a Sensitizer of Cell damaging Effect of Ultrasound Japanese Journal of Cancer Research 80 3 219 222 doi 10 1111 j 1349 7006 1989 tb02295 x PMC 5917717 PMID 2470713 Ohmura Tadahiro Fukushima Takeo Shibaguchi Hirotomo Yoshizawa Shin Inoue Tooru Kuroki Masahide Sasaki Kazunari Umemura Shin Ichiro 1 July 2011 Sonodynamic Therapy with 5 Aminolevulinic Acid and Focused Ultrasound for Deep seated Intracranial Glioma in Rat Anticancer Research 31 7 2527 2533 ISSN 0250 7005 PMID 21873170 a b c Zheng Yilin Ye Jinxiang Li Ziying Chen Haijun Gao Yu August 2021 Recent progress in sono photodynamic cancer therapy From developed new sensitizers to nanotechnology based efficacy enhancing strategies Acta Pharmaceutica Sinica B 11 8 2197 2219 doi 10 1016 j apsb 2020 12 016 ISSN 2211 3835 PMC 8424231 PMID 34522584 Choi Victor Rajora Maneesha A Zheng Gang 15 April 2020 Activating Drugs with Sound Mechanisms Behind Sonodynamic Therapy and the Role of Nanomedicine Bioconjugate Chemistry 31 4 967 989 doi 10 1021 acs bioconjchem 0c00029 ISSN 1043 1802 PMID 32129984 S2CID 212416405 Cheng Danling Wang Xiaoying Zhou Xiaojun Li Jingchao 2021 Nanosonosensitizers With Ultrasound Induced Reactive Oxygen Species Generation for Cancer Sonodynamic Immunotherapy Frontiers in Bioengineering and Biotechnology 9 761218 doi 10 3389 fbioe 2021 761218 ISSN 2296 4185 PMC 8514668 PMID 34660560 Lewis Thomas J Mitchell Doug June 2008 The Tumoricidal Effect of Sonodynamic Therapy SDT on S 180 Sarcoma in Mice Integrative Cancer Therapies 7 2 96 102 doi 10 1177 1534735408319065 ISSN 1534 7354 PMID 18550890 S2CID 22303568 Inui Toshio Makita Kaori Miura Hirona Matsuda Akiko Kuchiike Daisuke Kubo Kentaro Mette Martin Uto Yoshihiro Nishikata Takahito Hori Hitoshi Sakamoto Norihiro 1 August 2014 Case Report A Breast Cancer Patient Treated with GcMAF Sonodynamic Therapy and Hormone Therapy Anticancer Research 34 8 4589 4593 ISSN 0250 7005 PMID 25075104 Retrieved from https en wikipedia org w index php title Sonodynamic therapy amp oldid 1203583841, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.