Next Article in Journal
Anti-Inflammatory Effect of Xanthones from Hypericum beanii on Macrophage RAW 264.7 Cells through Reduced NO Production and TNF-α, IL-1β, IL-6, and COX-2 Expression
Previous Article in Journal
Synthesis of N-(3-Acyloxyacyl)glycines, Small Molecules with Potential Role in Gut Microbiome-Endocannabinoidome Communication
Previous Article in Special Issue
Metformin-Loaded Chitosan Hydrogels Suppress Bladder Tumor Growth in an Orthotopic Mouse Model via Intravesical Administration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Progress in Photothermal, Photodynamic and Sonodynamic Cancer Therapy: Through the cGAS-STING Pathway to Efficacy-Enhancing Strategies

by
Kelan Fang
1,2,†,
Huiling Zhang
1,3,†,
Qinghong Kong
1,2,
Yunli Ma
1,
Tianchan Xiong
1,2,
Tengyao Qin
1,2,
Sanhua Li
1,2,* and
Xinting Zhu
1,2,*
1
Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
2
College of Basic Medicine, Zunyi Medical University, Zunyi 563000, China
3
Department of Medicine and Pharmacy, Shizhen College of Guizhou University of Traditional Chinese Medicine, Guiyang 550000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2024, 29(15), 3704; https://doi.org/10.3390/molecules29153704
Submission received: 5 June 2024 / Revised: 19 July 2024 / Accepted: 29 July 2024 / Published: 5 August 2024

Abstract

:
Photothermal, photodynamic and sonodynamic cancer therapies offer opportunities for precise tumor ablation and reduce side effects. The cyclic guanylate adenylate synthase-stimulator of interferon genes (cGAS-STING) pathway has been considered a potential target to stimulate the immune system in patients and achieve a sustained immune response. Combining photothermal, photodynamic and sonodynamic therapies with cGAS-STING agonists represents a newly developed cancer treatment demonstrating noticeable innovation in its impact on the immune system. Recent reviews have concentrated on diverse materials and their function in cancer therapy. In this review, we focus on the molecular mechanism of photothermal, photodynamic and sonodynamic cancer therapies and the connected role of cGAS-STING agonists in treating cancer.

1. Introduction

Photothermal, photodynamic and sonodynamic cancer therapies are physical strategies that have demonstrated prominent anticancer efficacy [1,2,3]. These cancer therapy methods integrate light, thermal and acoustic modalities in a single platform to reinforce their therapeutic effects. Photothermal therapy (PTT) uses photothermal agents (PTAs) to convert light into heat. The heat generated by PTAs can induce hyperthermia, leading to cell death and stimulating the immune system. Favorable results in experiments concerning tumors such as breast cancer [4] and cervical cancer [5] showed their potential to be an adjuvant therapy in the clinic. Clinical trials also showed that PTT-induced hyperthermia has great potential in cancer therapy [6]. Like PTT, photodynamic therapy (PDT) uses photosensitizers (PS) to mediate energy transmission and generate toxic residues, such as reactive oxygen species (ROS). ROS can disrupt DNA and protein structure in cells, leading to organelle destruction and cell death [7]. This process can stimulate the immune system and lead to immunogenic cell death (ICD) [8]. PDT can be used in treating many superficial diseases including but not limited to premalignant conditions and tumors in the skin, digestive system and urinary system [9]. It is an important non-invasive therapy in the clinic. Due to the limited penetration ability of light, sonodynamic therapy (SDT) offers an alternative to PTT and has been demonstrated to have a wide range of applications in different solid tumors such as hepatocarcinoma, glioma and melanoma [10,11]. SDT utilizes sonosensitizers and ultrasound to produce ROS which lead to cancer cell destruction [12,13] {Das, 2024 #181}. SDT plays a vital role in the therapy of deep tumors, as ultrasound can penetrate into deeper tissues. Radiotherapy, chemotherapy and immune therapy are also extensively used to prevent tumor recurrence after surgical removal [14,15,16]. In recent years, immune therapy has flourished as it aims at reactivating patients’ innate immune system against cancer [17,18]. When combined with other therapeutic interventions, immune therapy may provide a silver line against cancer.
The cGAS-STING pathway is considered a potential target for stimulating immune system in patients and achieving a sustained immune response. With intracytoplasmic DNA, cGAS-STING agonist can increase the production of interleukin 6 (IL-6), tumor necrosis factor (TNF) and interferon (IFN) [19] and play a regulatory role in inflammation and tumor treatment [20,21,22]. Deng et al. found that injecting cyclic dinucleotide (CDN) intratumorally after radiotherapy can suppress tumor growth [23]. The cGAS-STING signaling pathway has been extensively studied and adopted for tumor immune therapy [24]. However, administrating a cGAS-STING agonist at a high concentration can lead to significant side effects [25]. It is necessary to find a method to precisely control cGAS-STING activation. Moreover, the effectiveness of cGAS-STING agonists depends on the level of STING expression [26]. When myeloid cells are depleted, the activity of cGAS-STING agonists can be attenuated [27]. In addition, cytokine induced by cGAS-STING can elicit a carcinogenetic impact on epithelial cells [28], and the prolonged overexpression of STING can disrupt the homeostasis in the endoplasmic reticulum (ER), causing ER stress in T cells and leading T-cell death [29]. Recently, Chen et al. reported the use of chitosan hydrogels in photothermal therapy to precisely control the release of a STING agonist in the tumor microenvironment (TME), inducing stable tumor immunity [30]. Yu et al. used a photosensitizer, MHI148, and a STING agonist, 2′3′-cGAMP, to achieve diagnosis and therapeutic effects [31]. Jiang et al. designed a material that used ultrasound to produce singlet oxygen and release a STING agonist to activate tumor immunity [32]. By combining PTT, PDT and SDT, the intrinsic drawbacks of cGAS-STING apoptosis can be mitigated. These synergistic therapies cover a variety of tumors and have the possibility of long-term immunity. The synergistic therapy of non-invasive therapy and immunotherapy is in line with the future direction of tumor therapy. Especially the cGAS-STING pathway holds great promise for immune therapy by mobilizing the patients’ innate immune system to inhibit and eliminate tumors [33]. Some of the STING agonists have reached clinical trials and we list them in Table 1. The reported trials are all in phase I or a combination of phase I and phase II. In five trials, two have been terminated. Three trials are recruiting. The results of E7766 showed dose-limiting toxicities and serious adverse events when the dose escalation came to 600 mcg.
Previous reviews focused on development in photothermal, photodynamic and sonodynamic cancer therapies. For PTT and PDT, most reviews were oriented from a material perspective. Liu’s review provided a general overview of the characteristics of different nanomaterials used in phototherapy and the methods scientists developed to enhance the outcomes [34]. Yang et al. reported on nanomaterials used in SDT and their combination with immune therapies targeting the PD-1/PD-L1 pathway [35]. Moreover, there were limited reviews that concentrated on the application of the cGAS-STING pathway in sonodynamic therapy. Regarding the cGAS-STING pathway, most reviews concentrated on the drug development, different ways to regulate it and its clinical prospects in various diseases [36,37,38]. In this review, we illustrate the combination of the cGAS-STING pathway with photothermal, photodynamic and sonodynamic physical methods in cancer therapy. Relationship and current development of the cGAS-STING pathway to individual cancer therapy method will be discussed in the following sections. We hope this review will provide new insights into the synergistic effects of cGAS-STING agonists and new leads for clinical cancer therapy.

2. cGAS-STING Signaling Pathway with Photothermal Therapy

Photothermal therapy (PTT) is a strategy that transfers photoenergy into heat to ablate tumors through hyperthermal effects. Hyperthermia can inhibit tumor repair and facilitate tumor immunity. Song et al. reported that hyperthermia could downregulate oxygenation in tumors, leading to a suppressive environment for the tumor to repair. For normal tissues, the hyperthermia effects were negligible [39,40]. Azocar et al. reported that hyperthermia increased the sensitivity of human natural killer cells [41]. In 2007 and 2021, Ostberg et al. and Pan et al. reported that hyperthermia can promote the anti-tumor effects of NK cells [42,43]. Burd et al. reported that steady heat could cause vascular changes and induce tumor apoptosis [44]. Hyperthermia generated in PTT is mediated by PTAs. PTAs are usually introduced into cells through nanoparticles, as nano PTAs have a higher permeability and can be combined with other therapeutic materials [45]. The surface of PTAs can be modified to control tissue persistence and toxicity [46]. Poursalehi et al. designed chemically modified gold nanoparticles loaded with Doxorubicin, which demonstrated 99% cellular uptake after 3 h [47]. Huilgol et al. reported that the combination of hyperthermia with radiation can significantly improve the response rate of patients and extend median survival [48]. Tang et al. reported using a photosensitive dimer to target the tumor membrane and achieve a synergistic therapy of PTT and PDT [49]. Cell death is followed by the release of tumor-associated antigens (TAAs) and damage-associated molecular patterns (DAMPs). However, TAAs and DAMPs may not be effectively taken up by dendritic cells (DCs) nor induce a T-cell response, as these processes are inhibited in the TME [50].
Adding immune therapy can enhance the anti-tumor effects of photothermal therapy (PTT) through activating immune system within the TME. The STING pathway is one of the promising options for such a combination. By combining PTT with a STING agonist, the maturation of DCs can be strengthened and cytotoxic T cells can be activated. Cyclic-GMP-AMP (cGAMP) can directly bind to STING and activate the STING pathway [51]. Ishikawa et al. demonstrated that STING is located in the upstream of TBK1/IRF3 and plays an important role in IFN-β expression and innate immune activation [52]. Jiang et al. reported that cGAMP-mediated STING/STAT3 can inhibit the activity, proliferation and invasion of tumor cells and inhibit tumor progression through upregulating IL-2, TNF-α and IFN-γ by cGAMP and downregulating CXCL8, BCL-2 and VEGFA to inhibit angiogenesis [53]. Koshy et al. used liposomes as a delivery vehicle for cGAMP to improve the immune response and achieve immune memory in mice. The injection of liposome-delivered cGAMP could inhibit the growth of metastatic tumors while free drug displayed a limited effect [54]. The theragnostic thermosensitive liposome (PLDD) also demonstrated great potential. Long et al. developed a theragnostic thermosensitive liposome (PLDD) using a D-A-D conjugated oligomer (DTTB) and 5,6-dimethylxanthenone-4-acetic acid (DMXAA)Long [55]. In this combination, DTTB is a newly developed photothermal agent that has demonstrated a high quantum yield and is being evaluated for the pharmaceutical properties of nanomedicine. The PEGylation of DTTB can dramatically improve its blood circulation time and tumor accumulation [56]. Demonstrated by Corrales et al., DMXAA, as a STING agonist, can lead to tumor regression and stimulate strong anti-tumor immunity [57]. However, it showed a limited effect in clinical trials because it turned out to be agonist to mouse STING but not human STING [58,59]. The combination of the photothermal agent DTTB with the STING agonist DMXAA constituted a nanoplatform. When exposed to second near-infrared (NIR-Ⅱ) fluorescence, it could generate heat and lead to ICD, subsequently releasing DMXAA from the thermosensitive liposome. This approach has shown considerable anti-tumor efficiency and biosafety in tumor therapy [55]. Apart from liposomes, Chen et al. used hydrogel to combine the STING agonist DMXAA and the PTA indocyanine green (ICD) (Figure 1A). This hydrogel improved the intensity of ICD (Figure 1B). Combining with DMXAA further improved the anti-tumor effect (Figure 1C) [30]. Ma et al. used the STING agonist diABZIs and the PTA Croconaine dye IR1024 to develop a nanomedicine named STING agonist-based photo-immuno-thernostic nanomedicine (SAPTN) (Figure 1D). SAPTN could stimulate the tumor compared to normal tissue (Figure 1E) and lead to sustained anti-tumor immunity when rechallenged by the same tumor cells (Figure 1F) [60]. In addition, metal ions are reported to stimulate the cGAS-STING pathway; for instance, Mn2+ has been demonstrated to activate the immune system through this pathway. Wang’s team found that Mn2+ can enhance the sensitivity of cGAS, thereby improving the ability to respond to dsDNA in the cytoplasm. Their study showed that even at low concentration of dsDNA, Mn2+ could promote the synthesis of the second messenger cGAMP and enhance the affinity between cGAMP and STING [61]. Mn2+ and MoO42+ were reported to form a nanoparticle named MMP NDs. MMP NDs could induce tumor cell ferroptosis directly or through reducing the glutathione accumulated in tumor cells as well as activating the cGAS-STING pathway. Furthermore, MMP NDs could stimulate IFN-γ secretion by CD8+ T cells and inhibit the expression of GPX4 which promotes ferroptosis [62]. Lin et al. developed a polydopamine-manganese-based nanomaterial [63]. In this report, polydopamine acts as a photothermal agent, and Mn2+ can be released with glutathione to produce hydroxyl radicals (·OH) and stimulate the cGAS-STING pathway. This results in the suppression of 86.7% of tumor cells and the production of more cytotoxic T cells compared to the negative immune regulator Treg cells. Moreover, in the experiments of Xia et al. and Zheng et al., another PTA—Prussian blue—was combined with manganese and proved to be effective in colon and breast tumor models [64,65]. These experiments represent that the combination of PTT and STING agonists is a potential strategy for future cancer treatment. Synergistic therapies of photothermal therapy and cGAS-STING agonists in recent studies are listed Table 2 [30,55,56,57,60,63,64,65,66,67,68,69,70,71].
Before photothermal therapy (PTT) can be used clinically, several obstacles must be addressed. First, light attenuation restricts the usage of PTT, especially for internal tumors, where the therapeutic effect of PPT is limited. Additionally, the photosensitizers’ pharmacologic processes, for example, toxicity or metabolism, still require further study. Third, Cherukula et al. reported that the effect of PTT might be limited to 7 days, accompanied by side effects of increased immune tolerance in the tumor microenvironment. But they also found possible therapeutic targets to solve this question [72]. In the experiment of Yue et al., they use an agent named TMP195 to repolarize immunosuppressive tumor-associated macrophages, which also provides a novel way to solve this question [73].

3. cGAS-STING Signaling Pathway with Photodynamic Therapy

Photodynamic therapy (PDT) has been clinically approved for over 200 years, commonly used for superficial cancers and in situ cancers such as esophageal cancer [74], skin cancer [75] and gynecologic malignant diseases [76]. For instance, Barrette’s esophagus (BE) is known to progress to high-grade dysplasia (HGD) and adenocarcinoma [77]. In Japan, PTT is applied for esophageal squamous cell carcinoma (ESCC) treatment. The American College of Gastroenterology strongly recommends ablative therapy for residual BE in patients with EMR specimens demonstrating HGD or intramucosal carcinoma. A randomized phase III trial held by 30 centers and 485 patients reported that PDT, when combined with chemotherapy, increases the complete ablation of HGD and reduces the likelihood of adenocarcinoma development [78]. PDT utilizes a specific wavelength of light to generate toxic production through photosensitizers (PS) with tolerable pain. Two pathways contribute to cytotoxicity: the Type I pathway directly transfers electrons from the PS to the oxygen molecule, producing superoxide anions such as free radicals and reactive oxygen species; the Type II pathway involves energy transfer from the PS for singlet oxygen generation [79,80]. Singlet oxygen can penetrate cellular membranes, disrupting protein and DNA. It can also generate damage-associated molecular patterns and mediate the immune response [81]. PDT primarily affects tumor cells, vasculature and the immune system [82]. Shi et al. reported the use of a light-emitting diode for convenient, cost-effective and accurate PDT in gastrointestinal cancer treatment. Their point LED-PDT can offer sufficient light density and induce tumor apoptosis and necrosis [83]. Zhang et al. reported a nano delivery system named R6RGD-CMβCD-se-se-Ce6/LND (RCC/LND NPS), comprising the photosensitizer chlorin e6 (Ce6) and the chemotherapeutic lonidamine (LND). This nanomaterial can disrupt the tumor extracellular matrix (ECM), weaken anoikis resistance in triple-negative breast cancer and activate apoptotic pathways [84]. After injection, the developed photosensitizers accumulate in vasculature tissue [85]. The PS vasculature accumulation and damage can lead to an oxygen-deficient environment, inhibiting the growth of the tumor. The photodynamic reaction can ablate the tumor as well as strengthen tumor immunity [86]. This process includes innate immunity and adaptive immunity. Immune cells, for example, neutrophils, macrophages, NK cells, dendritic cells and T cells, are involved in the immune response [87]. The destruction of tumor tissues creates an inflammatory environment and releases cytokines, leading to dendritic cell (DC) accumulation and maturation. Mature DCs then phagocytose tumor cells, return to lymph nodes, present antigens to CD8+ T cells and activate T-cell migration to the tumor [88].
Studies have found that a synergistic therapy of photodynamic therapy (PDT) and a STING agonist is viable. The cGAS-STING signaling pathway, activated during DNA damage from ultraviolet irradiation or cisplatin treatment, induces cell apoptosis and is associated with inflammation and cell senescence [89,90]. Jiang et al. further illuminated the connection between cGAS and cell apoptosis, showing that cGAS inhibits the DNA damage repair process. After DNA damage, cGAS is recruited to double-strand breaks, interacting with PARP1 to hinder the formation of the PARP1–Timeless complex. This inhibits homologous recombination, promotes tumor occurrence, accelerates genomic instability and micronucleus formation and eventually leads to cell death [32,91]. Since PDT can cause DNA damage, adding a STING activator can promote apoptosis in tumor cells. ADU-S100 is a potent synthetic cyclic dinucleotide STING agonist [92]. But a recent study showed its limited clinical effect [93]. Hao et al. reported that using PDT and the STING agonist ADU-S100 can amplify the immune reaction then obtain a systemic immune response and immune memory [94]. Increasing antigen presentation and the repolarization of bone marrow-derived macrophages from the M1 to M2 phenotype have been observed. Both in vivo and in vitro experiments show that this combination can hugely increase the anti-tumor effects compared to monotherapy and demonstrates tumor resistance when rechallenged with tumor cells. SR-717 is a non-nucleotide STING agonist mimicking the structure of cGAMP and can induce a conformation change of STING to activate it [95]. Zhou et al. developed a nanoparticle using a polymeric metal−organic framework (PMOF) containing the photosensitizer Meso-tetra(carboxyphenyl) porphyrin (TCPP). The formulated nanoparticle SR@PMOF combined PMOF and the STING agonist SR-717. After irradiation, 1O2 produced by PDT can destroy the structure and release SR-717 into the tumor (Figure 2A). The synergistic effect of photodynamic therapy and a STING agonist can strengthen antigen presentation and the infiltration of CD8+ T cells and suppress the growth of primary tumors (Figure 2B) and distant tumors (Figure 2C) [96]. Yu et al. further demonstrated that a synergistic therapy of photodynamic therapy and a STING agonist can promote an inflammatory response and tumor suppression (Figure 2D–F) [31]. In Table 2, we list the agents used in the synergistic therapy of photodynamic therapy and a cGAS-STING agonist in recent studies [31,90,93,94,95,96,97,98,99].
Although photodynamic therapy (PDT) has been used in the clinic for a long time, it is still not widely utilized. Issues still needs to be handled. As oxygen is important in the therapy, the destruction of the vasculature can cause oxygen-deficiency in tumor tissues and influence the therapeutic effect [85]. In addition, in clinical settings, adverse effects of PDT are reported in the majority of people [78].

4. cGAS-STING Signaling Pathway with Sonodynamic Therapy

Ultrasound is widely used in the clinic for diagnosis and therapy due to its excellent tissue penetration and ability to accumulate sufficient energy for thermal effects [100]. For tumor treatment, sonodynamic therapy (SDT) has more advantages over PDT as light has a limited penetration for deeper tissue. SDT can be categorized based on the intensity of the ultrasound. Low-intensity ultrasound, for instance, can be employed in physiotherapy to heat specific structures. Low-intensity ultrasound (0.51 W/cm2, 1.0 MHz, 10 min) was reported for use on mouse squamous cell carcinoma (SCC), showing an anti-tumor effect [101]. High-intensity ultrasound (ranging from 103 to 104 W cm−2 with frequencies from 0.5 to 10.0 MHz) of 1.5 MHz is most commonly used for cancer treatment. Similar to photosensitizers, sonosensitizers can be activated by ultrasound to produce reactive intermediates that release free radicals. In addition, cavitation is also the main theory regarded as the mechanism of SDT. In the process of cavitation, bubbles are created when irradiated by ultrasound. These bubbles can expand and collapse at different phases. When these bubbles collapse, the temperature and pressure increase and oxidants are formed [102]. The resulting oxidants can induce the generation of ROS and the leakage of mitochondrial DNA in cytosol, damaging tumor tissues and vasculature [103]. In addition, cavitation can produce shock waves and shear stress to directly cause mechanical damage to cancer cells [104]. Wood et al. reported that low-intensity SDT could cause an antivascular effect in tumor tissues, creating an ischemic environment and leading to cell death [105,106]. SDT has been used to ablate tumors at a high temperature from 60 °C to about 95 °C in prostate cancer, hepatic cancer and esophageal cancer [105]. SDT uses sonosensitizers that harness ultrasound energy to eliminate the biofilm of cancer cells. These sonosensitizers also possess imaging capabilities, aiding in the precise delineation of target areas [107]. Wang et al. combined SDT with photodynamic therapy in a clinical trial for patients with advanced breast cancer, observing cancer degradation [108].
In sonodynamic therapy (SDT), cell apoptosis triggers the release of tumor-derived DNA. However, the immune system in TME is suppressive. This issue can be addressed by synergistic therapy with cGAS-STING agonists. cGAMP and other STING agonists can directly activate DC and enhance the presentation of related antigens to CD8+ T cells in vitro, thus promoting the activation of CD8+ T cells and their killing effect on tumor cells. cGAMP and other STING agonists can directly activate DCs and enhance the presentation of related antigens to CD8+ T cells in vitro [54], thus promoting the activation of CD8+ T cells and their killing effect on tumor cells. The use of a STING agonist can enhance the activation, cytotoxicity and anti-tumor effects of natural killer (NK) cells independently of CD8+ T cells, thus improving the clearance of tumors resistant to CD8+ T cells [109]. Using a STING agonist can revive the silenced immune system in “cold” tumors, turning them into “hot” tumors by inducing the infiltration of CD8+ T cells to activate anti-tumor immunity, thereby improving tumor clearance [110]. Reported by Lei et al., a cobalt-based nanoagonist was combined with the mitochondria-targeting ligand triphenyl phosphonium (TPP). TPP can target mitochondria and induce the leakage of mitochondrial DNA with the involvement of SDT. In the presence of cobalt, STING activation is more effective. And controllable activation of the immune system can be achieved, even in bone and metastatic tumors [111] (Figure 3). Jiang et al. and Yu et al. used a semiconducting polymer to combine with an orally available agonist agent MSA-2 [112,113,114]. Yu et al. designed polymeric STING pro-agonists to target the tumor microenvironment with elevated glutathione expression and the improved release of MSA-2 [113]. Lu et al. use the clinically approved sonosensitizer and STING agonist SR-717 [95,115]. An anaerobic microorganism of Bifidobacteria Longum (BiL) was designed, demonstrated sono-sensitivity in anaerobic environments and was named HMME@BiL. HMME@BiL demonstrated high efficacy and selectivity and good biocompatibility against malignant tumors [116]. Tian et al. used zinc oxide, zinc ions as sonosensitizer and a STING agonist (PZnO@DOX) [117,118,119]. PZnO@DOX was proved to modulate the immunogenic cell death induced by chemotherapy [119]. We summarize the synergistic therapy of sonodynamic therapy and cGAS-STING agonists of recent studies in Table 2 [95,112,113,114,115,116,117,118,119,120].
While scientists have reported cases using sonodynamic therapy (SDT) with other therapies, a systemic analysis of the therapeutic effects of SDT on cancer and the immune system is still required. Pain could occur in patients when energy accumulates in deep organs especially in the bones [108]. Moreover, no entirely suitable sonosensitizer has been developed for clinical use.
Table 2. Overview of synergistic therapy of PDT, PTT and SDT with cGAS-STING agonists.
Table 2. Overview of synergistic therapy of PDT, PTT and SDT with cGAS-STING agonists.
Therapy References
Photothermal TherapyPhotothermal Transduction AgentsSTING agonist
Indocyanine green DMXAA[30,57,66]
Croconaine dye IR1024 DiABZIs[60,67,68]
DTTB DMXAA[55,56,57]
Prussian blueManganese[64,65,69,70]
PolydopamineManganese[63,70,71]
Photodynamic TherapyPhotosensitizersSTING agonist
VerteporfinADU-S100[93,94,97]
Meso-tetra(carboxyphenyl) porphyrin (TCPP)SR-717[95,96,98]
MHI1482′3′-cGAMP[31,90,99]
Sonodynamic TherapySonosensitizersSTING agonist
Semiconducting polymerMSA-2[112,113,114,120]
Hematoporphyrin monomethyl ether SR-717[95,115,116]
Zinc oxideZinc ions[117,118,119]

5. Summary and Outlook

Recent progress has emerged with promising solutions to advance photothermal, photodynamic and sonodynamic cancer therapy. Addressing the challenges of limited light penetration and oxygen deficiency in the tumor environment, Tian et al. utilized a singlet oxygen battery (SOB) to release ROS independently of oxygen and light and to control the release within the tumor [121]. Shaw et al. also reported that by modulating the concentration and distribution of nanoparticles and the range of irradiation, thermal damage can be achieved for tumors at depths of up to 9 mm [122]. But we still have a long way to go in clinical cancer therapy, particularly in the aspects of therapeutic efficacy, material safety, minimal side effects, etc. In addition, some agents in PTT, PDT and SDT have reached clinical trials, and we have listed some of them in Table 3. For photothermal therapy, only one reported trial has reached phase III. For photothermal therapy, the safety issue still needs more clinical confirmation. For photodynamic therapy, four of them have reached phase II and one has reached phase IV. Three trials are recruiting and four have not started to recruit. For sonodynamic therapy, three trials have reached phase I and two trials have reached phase II. Three out of five clinic trials are under active recruiting.
We hope this review will provide a glimpse into a synergistic effect with cGAS-STING agonists and their potential in clinic application. Combining photothermal, photodynamic and sonodynamic therapeutic methods with cGAS-STING agonists can help to overcome the blockade of the immune-suppressive environment within tumors. In turn, those therapeutic methods can strengthen the stimulation of the cGAS-STING pathway as they can induce cell death and the release of tumor-associated antigen. In addition, side effects caused by cGAS-STING agonists can be accurately controlled by synergistic application of these methods. These three kinds of synergistic therapy can be applied in various tumors and lead to an efficient, persistent systemic anti-tumor effect compared to monotherapy. They shine a light on a new direction of clinical application along with chances and challenges. In conclusion, the combination of photothermal, photodynamic and sonodynamic therapeutic methods with cGAS-STING agonists could offer more precise and safe approaches with broad future prospects for cancer therapy.

Author Contributions

Conceptualization, X.Z. and S.L.; writing and original draft preparation, K.F. and H.Z.; writing—review and editing, visualization, K.F., H.Z., Q.K., Y.M., T.X., T.Q., S.L. and X.Z.; supervision, X.Z. and S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China grant number 82260760, the Science and Technology Foundation of Guizhou Province grant number QKHJC-ZK2021YB571, the Project of Education Department of Guizhou Province grant number QJHKY2021219 and the Innovation and Entrepreneurship Project for College Students of ZMU grant number 202310661250, 202210661144X.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Fan, W.; Yung, B.; Huang, P.; Chen, X. Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem. Rev. 2017, 117, 13566–13638. [Google Scholar] [CrossRef]
  2. Li, Y.; Qi, H.; Geng, Y.; Li, L.; Cai, X. Research progress of organic photothermal agents delivery and synergistic therapy systems. Colloids Surf. Biointerfaces 2024, 234, 113743. [Google Scholar] [CrossRef] [PubMed]
  3. Son, S.; Kim, J.H.; Wang, X.; Zhang, C.; Yoon, S.A.; Shin, J.; Sharma, A.; Lee, M.H.; Cheng, L.; Wu, J.; et al. Multifunctional sonosensitizers in sonodynamic cancer therapy. Chem. Soc. Rev. 2020, 49, 3244–3261. [Google Scholar] [CrossRef] [PubMed]
  4. Kang, X.; Sun, T.; Zhang, L.; Zhou, C.; Xu, Z.; Du, M.; Xiao, S.; Liu, Y.; Gong, M.; Zhang, D. Synergistic Theranostics of Magnetic Resonance Imaging and Photothermal Therapy of Breast Cancer Based on the Janus Nanostructures Fe3O4-Au(shell)-PEG. Int. J. Nanomed. 2021, 16, 6383–6394. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, Q.; Guo, Q.; Chen, Q.; Zhao, X.; Pennycook, S.J.; Chen, H. Highly Efficient 2D NIR-II Photothermal Agent with Fenton Catalytic Activity for Cancer Synergistic Photothermal-Chemodynamic Therapy. Adv. Sci. 2020, 7, 1902576. [Google Scholar] [CrossRef] [PubMed]
  6. van der Zee, J.; González González, D.; van Rhoon, G.C.; van Dijk, J.D.; van Putten, W.L.; Hart, A.A. Comparison of radiotherapy alone with radiotherapy plus hyperthermia in locally advanced pelvic tumours: A prospective, randomised, multicentre trial. Dutch Deep Hyperthermia Group. Lancet 2000, 355, 1119–1125. [Google Scholar] [CrossRef] [PubMed]
  7. Ming, L.; Cheng, K.; Chen, Y.; Yang, R.; Chen, D. Enhancement of tumor lethality of ROS in photodynamic therapy. Cancer Med. 2021, 10, 257–268. [Google Scholar] [CrossRef] [PubMed]
  8. Galluzzi, L.; Vitale, I.; Warren, S.; Adjemian, S.; Agostinis, P.; Martinez, A.B.; Chan, T.A.; Coukos, G.; Demaria, S.; Deutsch, E.; et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J. Immunother. Cancer 2020, 8, e000337. [Google Scholar] [CrossRef]
  9. Agostinis, P.; Berg, K.; Cengel, K.A.; Foster, T.H.; Girotti, A.W.; Gollnick, S.O.; Hahn, S.M.; Hamblin, M.R.; Juzeniene, A.; Kessel, D.; et al. Photodynamic therapy of cancer: An update. CA Cancer J. Clin. 2011, 61, 250–281. [Google Scholar] [CrossRef]
  10. Hu, C.; Hou, B.; Xie, S. Application of nanosonosensitizer materials in cancer sono-dynamic therapy. RSC Adv. 2022, 12, 22722–22747. [Google Scholar] [CrossRef]
  11. Kong, C.; Chen, X. Combined Photodynamic and Photothermal Therapy and Immunotherapy for Cancer Treatment: A Review. Int. J. Nanomed. 2022, 17, 6427–6446. [Google Scholar] [CrossRef] [PubMed]
  12. Su, X.; Wang, P.; Wang, X.; Guo, L.; Li, S.; Liu, Q. Involvement of MAPK activation and ROS generation in human leukemia U937 cells undergoing apoptosis in response to sonodynamic therapy. Int. J. Radiat. Biol. 2013, 89, 915–927. [Google Scholar] [CrossRef] [PubMed]
  13. Das, M.; Pandey, V.; Jajoria, K.; Bhatia, D.; Gupta, I.; Shekhar, H. Glycosylated Porphyrin Derivatives for Sonodynamic Therapy: ROS Generation and Cytotoxicity Studies in Breast Cancer Cells. ACS Omega 2024, 9, 1196–1205. [Google Scholar] [CrossRef] [PubMed]
  14. Socinski, M.A. Incorporating Immunotherapy Into the Treatment of Non-Small Cell Lung Cancer: Practical Guidance for the Clinic. Semin. Oncol. 2015, 42, S19–S28. [Google Scholar] [CrossRef] [PubMed]
  15. Park, R.; Saeed, A. Immunotherapy in Colorectal Cancer—Finding the Achilles’ Heel. NEJM Evid. 2024, 3, EVIDra2300353. [Google Scholar] [CrossRef] [PubMed]
  16. Rios-Hoyo, A.; Arriola, E. Immunotherapy and brain metastasis in lung cancer: Connecting bench side science to the clinic. Front. Immunol. 2023, 14, 1221097. [Google Scholar] [CrossRef] [PubMed]
  17. Fenton, G.A.; Mitchell, D.A. Cellular Cancer Immunotherapy Development and Manufacturing in the Clinic. Clin. Cancer Res. 2023, 29, 843–857. [Google Scholar] [CrossRef] [PubMed]
  18. Yi, M.; Zheng, X.; Niu, M.; Zhu, S.; Ge, H.; Wu, K. Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol. Cancer 2022, 21, 28. [Google Scholar] [CrossRef]
  19. Dunphy, G.; Flannery, S.M.; Almine, J.F.; Connolly, D.J.; Paulus, C.; Jonsson, K.L.; Jakobsen, M.R.; Nevels, M.M.; Bowie, A.G.; Unterholzner, L. Non-canonical Activation of the DNA Sensing Adaptor STING by ATM and IFI16 Mediates NF-kappaB Signaling after Nuclear DNA Damage. Mol. Cell 2018, 71, 745–760.E5. [Google Scholar] [CrossRef]
  20. Li, X.D.; Wu, J.; Gao, D.; Wang, H.; Sun, L.; Chen, Z.J. Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 2013, 341, 1390–1394. [Google Scholar] [CrossRef]
  21. Kemp, M.G.; Lindsey-Boltz, L.A.; Sancar, A. UV Light Potentiates STING (Stimulator of Interferon Genes)-dependent Innate Immune Signaling through Deregulation of ULK1 (Unc51-like Kinase 1). J. Biol. Chem. 2015, 290, 12184–12194. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, M.Z.; Cheng, W.C.; Chen, S.F.; Nieh, S.; O’Connor, C.; Liu, C.L.; Tsai, W.W.; Wu, C.J.; Martin, L.; Lin, Y.S.; et al. miR-25/93 mediates hypoxia-induced immunosuppression by repressing cGAS. Nat. Cell Biol. 2017, 19, 1286–1296. [Google Scholar] [CrossRef] [PubMed]
  23. Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [PubMed]
  24. Pu, F.; Chen, F.; Liu, J.; Zhang, Z.; Shao, Z. Immune Regulation of the cGAS-STING Signaling Pathway in the Tumor Microenvironment and Its Clinical Application. OncoTargets Ther. 2021, 14, 1501–1516. [Google Scholar] [CrossRef] [PubMed]
  25. Garland, K.M.; Sheehy, T.L.; Wilson, J.T. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem. Rev. 2022, 122, 5977–6039. [Google Scholar] [CrossRef] [PubMed]
  26. Czapla, J.; Drzyzga, A.; Matuszczak, S.; Cichon, T.; Rusin, M.; Jarosz-Biej, M.; Pilny, E.; Smolarczyk, R. Antitumor effect of anti-vascular therapy with STING agonist depends on the tumor microenvironment context. Front. Oncol. 2023, 13, 1249524. [Google Scholar] [CrossRef] [PubMed]
  27. Xu, T.; Dai, J.; Tang, L.; Sun, L.; Si, L.; Guo, J. Systemic administration of STING agonist promotes myeloid cells maturation and antitumor immunity through regulating hematopoietic stem and progenitor cell fate. Cancer Immunol. Immunother. 2023, 72, 3491–3505. [Google Scholar] [CrossRef] [PubMed]
  28. Ahn, J.; Xia, T.; Konno, H.; Konno, K.; Ruiz, P.; Barber, G.N. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 2014, 5, 5166. [Google Scholar] [CrossRef] [PubMed]
  29. Wu, J.; Chen, Y.J.; Dobbs, N.; Sakai, T.; Liou, J.; Miner, J.J.; Yan, N. STING-mediated disruption of calcium homeostasis chronically activates ER stress and primes T cell death. J. Exp. Med. 2019, 216, 867–883. [Google Scholar] [CrossRef]
  30. Chen, C.; Hu, M.; Cao, Y.; Zhu, B.; Chen, J.; Li, Y.; Shao, J.; Zhou, S.; Shan, P.; Zheng, C.; et al. Combination of a STING Agonist and Photothermal Therapy Using Chitosan Hydrogels for Cancer Immunotherapy. Biomacromolecules 2023, 24, 2790–2803. [Google Scholar] [CrossRef]
  31. Yu, H.; Chen, Q.; Zheng, M.; Wang, R.; Wang, H.; Cheng, L.; Hu, Y.; Dai, M.; Du, C.; Luo, W.; et al. Combination of MHI148 Targeted Photodynamic Therapy and STING Activation Inhibits Tumor Metastasis and Recurrence. ACS Appl. Mater. 2024, 16, 29672–29685. [Google Scholar] [CrossRef] [PubMed]
  32. Jiang, H.; Xue, X.; Panda, S.; Kawale, A.; Hooy, R.M.; Liang, F.; Sohn, J.; Sung, P.; Gekara, N.O. Chromatin-bound cGAS is an inhibitor of DNA repair and hence accelerates genome destabilization and cell death. EMBO J. 2019, 38, e102718. [Google Scholar] [CrossRef] [PubMed]
  33. Jie, C.; Li, R.; Cheng, Y.; Wang, Z.; Wu, Q.; Xie, C. Prospects and feasibility of synergistic therapy with radiotherapy, immunotherapy, and DNA methyltransferase inhibitors in non-small cell lung cancer. Front. Immunol. 2023, 14, 1122352. [Google Scholar] [CrossRef]
  34. Liu, Y.; Bhattarai, P.; Dai, Z.; Chen, X. Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer. Chem. Soc. Rev. 2019, 48, 2053–2108. [Google Scholar] [CrossRef]
  35. Yang, N.; Li, J.; Yu, S.; Xia, G.; Li, D.; Yuan, L.; Wang, Q.; Ding, L.; Fan, Z.; Li, J. Application of Nanomaterial-Based Sonodynamic Therapy in Tumor Therapy. Pharmaceutics 2024, 16, 603. [Google Scholar] [CrossRef]
  36. An, C.; Li, Z.; Chen, Y.; Huang, S.; Yang, F.; Hu, Y.; Xu, T.; Zhang, C.; Ge, S. The cGAS-STING pathway in cardiovascular diseases: From basic research to clinical perspectives. Cell Biosci. 2024, 14, 58. [Google Scholar] [CrossRef]
  37. Xie, F.; Zhu, Q. The regulation of cGAS-STING signaling by RNA virus-derived components. Viro. J. 2024, 21, 101. [Google Scholar] [CrossRef] [PubMed]
  38. Colangelo, N.W.; Gerber, N.K.; Vatner, R.E.; Cooper, B.T. Harnessing the cGAS-STING pathway to potentiate radiation therapy: Current approaches and future directions. Front. Pharmacol. 2024, 15, 1383000. [Google Scholar] [CrossRef]
  39. Song, C.W. Effect of Local Hyperthermia on Blood Flow and Microenvironment: A Review. Cancer Res. 1984, 44, 4721s–4730s. [Google Scholar]
  40. Song, C.W.; Rhee, J.G.; Levitt, S.H. Blood flow in normal tissues and tumors during hyperthermia. J. Natl. Cancer Inst. 1980, 64, 119–124. [Google Scholar] [CrossRef]
  41. Azocar, J.; Yunis, E.J.; Essex, M. Sensitivity of human natural killer cells to hyperthermia. Lancet 1982, 319, 16–17. [Google Scholar] [CrossRef]
  42. Ostberg, J.R.; Dayanc, B.E.; Yuan, M.; Oflazoglu, E.; Repasky, E.A. Enhancement of natural killer (NK) cell cytotoxicity by fever-range thermal stress is dependent on NKG2D function and is associated with plasma membrane NKG2D clustering and increased expression of MICA on target cells. J. Leukoc. Biol. 2007, 82, 1322–1331. [Google Scholar] [CrossRef] [PubMed]
  43. Pan, J.; Xu, Y.; Wu, Q.; Hu, P.; Shi, J. Mild Magnetic Hyperthermia-Activated Innate Immunity for Liver Cancer Therapy. J. Am. Chem. Soc. 2021, 143, 8116–8128. [Google Scholar] [CrossRef] [PubMed]
  44. Burd, R.; Dziedzic, T.S.; Xu, Y.; Caligiuri, M.A.; Subjeck, J.R.; Repasky, E.A. Tumor cell apoptosis, lymphocyte recruitment and tumor vascular changes are induced by low temperature, long duration (fever-like) whole body hyperthermia. J. Cell. Physiol. 1998, 177, 137–147. [Google Scholar] [CrossRef]
  45. Nag, S.; Mitra, O.; Tripathi, G.; Adur, I.; Mohanto, S.; Nama, M.; Samanta, S.; Gowda, B.H.J.; Subramaniyan, V.; Sundararajan, V.; et al. Nanomaterials-assisted photothermal therapy for breast cancer: State-of-the-art advances and future perspectives. Photodiagnosis Photodyn. Ther. 2024, 45, 103959. [Google Scholar] [CrossRef] [PubMed]
  46. Pramanik, S.; Mohanto, S.; Manne, R.; Rajendran, R.R.; Deepak, A.; Edapully, S.J.; Patil, T.; Katari, O. Nanoparticle-Based Drug Delivery System: The Magic Bullet for the Treatment of Chronic Pulmonary Diseases. Mol. Pharm. 2021, 18, 3671–3718. [Google Scholar] [CrossRef] [PubMed]
  47. Poursalehi, Z.; Salehi, R.; Samadi, N.; Rasta, S.H.; Mansoori, B.; Majdi, H. A simple strategy for chemo-photothermal ablation of breast cancer cells by novel smart gold nanoparticles. Photodiagnosis Photodyn. Ther. 2019, 28, 25–37. [Google Scholar] [CrossRef] [PubMed]
  48. Huilgol, N.G.; Gupta, S.; Sridhar, C.R. Hyperthermia with radiation in the treatment of locally advanced head and neck cancer: A report of randomized trial. J. Cancer Res. Ther. 2010, 6, 492–496. [Google Scholar] [CrossRef]
  49. Tang, Y.; Bisoyi, H.K.; Chen, X.M.; Liu, Z.; Chen, X.; Zhang, S.; Li, Q. Pyroptosis-Mediated Synergistic Photodynamic and Photothermal Immunotherapy Enabled by a Tumor-Membrane-Targeted Photosensitive Dimer. Adv. Mater. 2023, 35, e2300232. [Google Scholar] [CrossRef]
  50. Del Prete, A.; Salvi, V.; Soriani, A.; Laffranchi, M.; Sozio, F.; Bosisio, D.; Sozzani, S. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell. Mol. Immunol. 2023, 20, 432–447. [Google Scholar] [CrossRef]
  51. Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef] [PubMed]
  52. Ishikawa, H.; Barber, G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 2008, 455, 674–678. [Google Scholar] [CrossRef] [PubMed]
  53. Jiang, X.; Liu, G.; Hu, Z.; Chen, G.; Chen, J.; Lv, Z. cGAMP inhibits tumor growth in colorectal cancer metastasis through the STING/STAT3 axis in a zebrafish xenograft model. Fish Shellfish. Immunol. 2019, 95, 220–226. [Google Scholar] [CrossRef] [PubMed]
  54. Koshy, S.T.; Cheung, A.S.; Gu, L.; Graveline, A.R.; Mooney, D.J. Liposomal Delivery Enhances Immune Activation by STING Agonists for Cancer Immunotherapy. Adv. Biosyst. 2017, 1, 2366–7478. [Google Scholar] [CrossRef] [PubMed]
  55. Long, Q.; Yang, Y.; Liao, F.; Chen, H.; He, D.; Li, S.; Li, P.; Guo, W.; Xiao, Y. NIR-II fluorescence and PA imaging guided activation of STING pathway in photothermal therapy for boosting cancer immunotherapy by theranostic thermosensitive liposomes. J. Mater. Chem. B 2023, 11, 8528–8540. [Google Scholar] [CrossRef] [PubMed]
  56. Li, S.; Chen, H.; Liu, H.; Liu, L.; Yuan, Y.; Mao, C.; Zhang, W.; Zhang, X.; Guo, W.; Lee, C.-S.; et al. In Vivo Real-Time Pharmaceutical Evaluations of Near-Infrared II Fluorescent Nanomedicine Bound Polyethylene Glycol Ligands for Tumor Photothermal Ablation. ACS Nano 2020, 14, 13681–13690. [Google Scholar] [CrossRef] [PubMed]
  57. Corrales, L.; Glickman, L.H.; McWhirter, S.M.; Kanne, D.B.; Sivick, K.E.; Katibah, G.E.; Woo, S.-R.; Lemmens, E.; Banda, T.; Leong, J.J.; et al. Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell Rep. 2015, 11, 1018–1030. [Google Scholar] [CrossRef] [PubMed]
  58. Lara, P.N., Jr.; Douillard, J.Y.; Nakagawa, K.; von Pawel, J.; McKeage, M.J.; Albert, I.; Losonczy, G.; Reck, M.; Heo, D.S.; Fan, X.; et al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. J. Clin. Oncol. 2011, 29, 2965–2971. [Google Scholar] [CrossRef] [PubMed]
  59. Conlon, J.; Burdette, D.L.; Sharma, S.; Bhat, N.; Thompson, M.; Jiang, Z.; Rathinam, V.A.; Monks, B.; Jin, T.; Xiao, T.S.; et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. J. Immuno. 2013, 190, 5216–5225. [Google Scholar] [CrossRef]
  60. Ma, W.; Sun, R.; Tang, L.; Li, Z.; Lin, L.; Mai, Z.; Chen, G.; Yu, Z. Bioactivable STING Nanoagonists to Synergize NIR-II Mild Photothermal Therapy Primed Robust and Long-Term Anticancer Immunity. Adv. Mater. 2023, 35, e2303149. [Google Scholar] [CrossRef]
  61. Wang, C.; Guan, Y.; Lv, M.; Zhang, R.; Guo, Z.; Wei, X.; Du, X.; Yang, J.; Li, T.; Wan, Y.; et al. Manganese Increases the Sensitivity of the cGAS-STING Pathway for Double-Stranded DNA and Is Required for the Host Defense against DNA Viruses. Immunity 2018, 48, 675–687.E7. [Google Scholar] [CrossRef] [PubMed]
  62. Lei, H.; Li, Q.; Li, G.; Wang, T.; Lv, X.; Pei, Z.; Gao, X.; Yang, N.; Gong, F.; Yang, Y.; et al. Manganese molybdate nanodots with dual amplification of STING activation for “cycle” treatment of metalloimmunotherapy. Bioact. Mater. 2024, 31, 53–62. [Google Scholar] [CrossRef] [PubMed]
  63. Lin, H.; Jiang, C.; Wang, B.; Wang, Y.; Shangguan, Z.; Wu, Y.; Wang, X.; Huang, Y.; Wang, L.; Chen, P.; et al. Glutathione degradable manganese-doped polydopamine nanoparticles for photothermal therapy and cGAS-STING activated immunotherapy of lung tumor. J. Colloid Interface Sci. 2024, 663, 167–176. [Google Scholar] [CrossRef] [PubMed]
  64. Xia, J.; Wang, L.; Shen, T.; Li, P.; Zhu, P.; Xie, S.; Chen, Z.; Zhou, F.; Zhang, J.; Ling, J.; et al. Integrated manganese (III)-doped nanosystem for optimizing photothermal ablation: Amplifying hyperthermia-induced STING pathway and enhancing antitumor immunity. Acta Biomater. 2023, 155, 601–617. [Google Scholar] [CrossRef] [PubMed]
  65. Zheng, Y.; Chen, J.; Song, X.R.; Chang, M.Q.; Feng, W.; Huang, H.; Jia, C.X.; Ding, L.; Chen, Y.; Wu, R. Manganese-enriched photonic/catalytic nanomedicine augments synergistic anti-TNBC photothermal/nanocatalytic/immuno-therapy via activating cGAS-STING pathway. Biomaterials 2023, 293, 121988. [Google Scholar] [CrossRef] [PubMed]
  66. Güney Akkurt, M.; Gülsoy, M. Polylactide nanoparticles encapsulating indocyanine green for photothermal therapy of prostate cancer cells. Photodiagnosis Photodyn. Ther. 2022, 37, 102693. [Google Scholar] [CrossRef] [PubMed]
  67. McGarraugh, H.H.; Liu, W.; Matthews, B.P.; Smith, B.D. Croconaine Rotaxane Dye with 984 nm Absorption: Wavelength-Selective Photothermal Heating. Eur. J. Org. Chem. 2019, 2019, 3489–3494. [Google Scholar] [CrossRef] [PubMed]
  68. Ramanjulu, J.M.; Pesiridis, G.S.; Yang, J.; Concha, N.; Singhaus, R.; Zhang, S.Y.; Tran, J.L.; Moore, P.; Lehmann, S.; Eberl, H.C.; et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature 2018, 564, 439–443. [Google Scholar] [CrossRef] [PubMed]
  69. Tang, K.; Li, X.; Hu, Y.; Zhang, X.; Lu, N.; Fang, Q.; Shao, J.; Li, S.; Xiu, W.; Song, Y.; et al. Recent advances in Prussian blue-based photothermal therapy in cancer treatment. Biomater. Sci. 2023, 11, 4411–4429. [Google Scholar] [CrossRef]
  70. Lv, M.; Chen, M.; Zhang, R.; Zhang, W.; Wang, C.; Zhang, Y.; Wei, X.; Guan, Y.; Liu, J.; Feng, K.; et al. Manganese is critical for antitumor immune responses via cGAS-STING and improves the efficacy of clinical immunotherapy. Cell Res. 2020, 30, 966–979. [Google Scholar] [CrossRef]
  71. Qiu, J.; Shi, Y.; Xia, Y. Polydopamine Nanobottles with Photothermal Capability for Controlled Release and Related Applications. Adv. Mater. 2021, 33, e2104729. [Google Scholar] [CrossRef] [PubMed]
  72. Cherukula, K.; Park, M.S.; Sontyana, A.G.; Mathew, A.P.; Vijayan, V.; Bae, W.K.; Park, I.K. Role of Immunosuppressive Microenvironment in Acquiring Immunotolerance Post-Photothermal Therapy. J. Korean Med. Sci. 2019, 34, e272. [Google Scholar] [CrossRef] [PubMed]
  73. Yue, Y.; Li, F.; Li, Y.; Wang, Y.; Guo, X.; Cheng, Z.; Li, N.; Ma, X.; Nie, G.; Zhao, X. Biomimetic Nanoparticles Carrying a Repolarization Agent of Tumor-Associated Macrophages for Remodeling of the Inflammatory Microenvironment Following Photothermal Therapy. ACS Nano 2021, 15, 15166–15179. [Google Scholar] [CrossRef] [PubMed]
  74. Yano, T.; Hatogai, K.; Morimoto, H.; Yoda, Y.; Kaneko, K. Photodynamic therapy for esophageal cancer. Ann. Transl. Med. 2014, 2, 29. [Google Scholar] [CrossRef]
  75. Valli, F.; García Vior, M.C.; Roguin, L.P.; Marino, J. Crosstalk between oxidative stress-induced apoptotic and autophagic signaling pathways in Zn(II) phthalocyanine photodynamic therapy of melanoma. Free Radic. Biol. Med. 2020, 152, 743–754. [Google Scholar] [CrossRef] [PubMed]
  76. Matoba, Y.; Banno, K.; Kisu, I.; Aoki, D. Clinical application of photodynamic diagnosis and photodynamic therapy for gynecologic malignant diseases: A review. Photodiagnosis Photodyn. Ther. 2018, 24, 52–57. [Google Scholar] [CrossRef] [PubMed]
  77. Yoshida, K.; Suzuki, S.; Mimura, S.; Ichii, M.; Sakai, H.; Shimao, H.; Kato, H.; Ito, Y.; Hiki, Y.; Hayashi, K.; et al. Photodynamic therapy for superficial esophageal cancer: A phase III study using PHE and excimer dye laser. Gan Kagaku Ryoho. Cancer Chemother. 1993, 20, 2063–2066. [Google Scholar]
  78. Overholt, B.F.; Lightdale, C.J.; Wang, K.K.; Canto, M.I.; Burdick, S.; Haggitt, R.C.; Bronner, M.P.; Taylor, S.L.; Grace, M.G.; Depot, M. Photodynamic therapy with porfimer sodium for ablation of high-grade dysplasia in Barrett’s esophagus: International, partially blinded, randomized phase III trial. Gastrointest. Endosc. 2005, 62, 488–498. [Google Scholar] [CrossRef]
  79. Bilski, P.; Motten, A.G.; Bilska, M.; Chignell, C.F. The photooxidation of diethylhydroxylamine by rose bengal in micellar and nonmicellar aqueous solutions. Photochem. Photobiol. 1993, 58, 11–18. [Google Scholar] [CrossRef]
  80. Ma, J.; Jiang, L. Photogeneration of singlet oxygen (1O2) and free radicals (Sen·−, O·−2) by tetra-brominated hypocrellin B derivative. Free Radic. Res. 2001, 35, 767–777. [Google Scholar] [CrossRef]
  81. Maeding, N.; Verwanger, T.; Krammer, B. Boosting Tumor-Specific Immunity Using PDT. Cancers 2016, 8, 91. [Google Scholar] [CrossRef]
  82. Korbelik, M. Induction of tumor immunity by photodynamic therapy. J. Clin. Laser Med. Surg. 1996, 14, 329–334. [Google Scholar] [CrossRef] [PubMed]
  83. Shi, X.; Yin, H.; Dong, X.; Li, H.; Li, Y. Photodynamic therapy with light-emitting diode arrays producing different light fields induces apoptosis and necrosis in gastrointestinal cancer. Front. Oncol. 2022, 12, 1062666. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, T.; Wang, X.; Wang, D.; Lei, M.; Hu, Y.; Chen, Z.; Li, Y.; Luo, Y.; Zhang, L.; Zhu, Y. Synergistic effects of photodynamic therapy and chemotherapy: Activating the intrinsic/extrinsic apoptotic pathway of anoikis for triple-negative breast cancer treatment. Biomater. Adv. 2024, 160, 213859. [Google Scholar] [CrossRef]
  85. Fingar, V.H. Vascular effects of photodynamic therapy. J. Clin. Laser Med. Surg. 1996, 14, 323–328. [Google Scholar] [CrossRef]
  86. Donohoe, C.; Senge, M.O.; Arnaut, L.G.; Gomes-da-Silva, L.C. Cell death in photodynamic therapy: From oxidative stress to anti-tumor immunity. Biochim. Et Biophys. Acta Rev. Cancer 2019, 1872, 188308. [Google Scholar] [CrossRef] [PubMed]
  87. Falk-Mahapatra, R.; Gollnick, S.O. Photodynamic Therapy and Immunity: An Update. Photochem. Photobiol. 2020, 96, 550–559. [Google Scholar] [CrossRef] [PubMed]
  88. Castano, A.P.; Mroz, P.; Hamblin, M.R. Photodynamic therapy and anti-tumour immunity. Nat. Rev. Cancer 2006, 6, 535–545. [Google Scholar] [CrossRef] [PubMed]
  89. Li, C.; Liu, W.; Wang, F.; Hayashi, T.; Mizuno, K.; Hattori, S.; Fujisaki, H.; Ikejima, T. DNA damage-triggered activation of cGAS-STING pathway induces apoptosis in human keratinocyte HaCaT cells. Mol. Immunol. 2021, 131, 180–190. [Google Scholar] [CrossRef]
  90. Li, T.; Chen, Z.J. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 2018, 215, 1287–1299. [Google Scholar] [CrossRef]
  91. Liu, H.; Zhang, H.; Wu, X.; Ma, D.; Wu, J.; Wang, L.; Jiang, Y.; Fei, Y.; Zhu, C.; Tan, R.; et al. Nuclear cGAS suppresses DNA repair and promotes tumorigenesis. Nature 2018, 563, 131–136. [Google Scholar] [CrossRef]
  92. Glickman, L.H.; Kanne, D.B.; Kasibhatla, S.; Li, J.; Pferdekamper, A.C.; Gauthier, K.S.; Deng, W.; Desbien, A.L.; Katibah, G.E.; Leong, J.J.; et al. Abstract 1445: STING activation in the tumor microenvironment with a synthetic human STING-activating cyclic dinucleotide leads to potent anti-tumor immunity. Cancer Res. 2016, 76, 1445. [Google Scholar] [CrossRef]
  93. Meric-Bernstam, F.; Sweis, R.F.; Hodi, F.S.; Messersmith, W.A.; Andtbacka, R.H.I.; Ingham, M.; Lewis, N.; Chen, X.; Pelletier, M.; Chen, X.; et al. Phase I Dose-Escalation Trial of MIW815 (ADU-S100), an Intratumoral STING Agonist, in Patients with Advanced/Metastatic Solid Tumors or Lymphomas. Clin. Cancer Res. 2022, 28, 677–688. [Google Scholar] [CrossRef] [PubMed]
  94. Hao, Y.; Ma, S.; Gu, Z.; Haghparast, A.; Schomann, T.; Yu, Z.; He, Y.; Dong, X.; Cruz, L.J.; Ten Dijke, P. Combination of photodynamic therapy and stimulator of interferon genes (STING) agonist inhibits colorectal tumor growth and recurrence. Cancer Commun. 2023, 43, 513–518. [Google Scholar] [CrossRef] [PubMed]
  95. Chin, E.N.; Yu, C.; Vartabedian, V.F.; Jia, Y.; Kumar, M.; Gamo, A.M.; Vernier, W.; Ali, S.H.; Kissai, M.; Lazar, D.C.; et al. Antitumor activity of a systemic STING-activating non-nucleotide cGAMP mimetic. Science 2020, 369, 993–999. [Google Scholar] [CrossRef] [PubMed]
  96. Zhou, Q.; Dutta, D.; Cao, Y.; Ge, Z. Oxidation-Responsive PolyMOF Nanoparticles for Combination Photodynamic-Immunotherapy with Enhanced STING Activation. ACS Nano 2023, 17, 9374–9387. [Google Scholar] [CrossRef] [PubMed]
  97. Battaglia Parodi, M.; La Spina, C.; Berchicci, L.; Petruzzi, G.; Bandello, F. Photosensitizers and Photodynamic Therapy: Verteporfin. Dev. Ophthalmol. 2016, 55, 330–336. [Google Scholar] [CrossRef]
  98. Chitgupi, U.; Lovell, J.F.; Rajendiran, V. Assessing Photosensitizer Targeting Using Meso-Tetra(Carboxyphenyl) Porphyrin. Molecules 2018, 23, 892. [Google Scholar] [CrossRef]
  99. Yang, X.; Shi, C.; Tong, R.; Qian, W.; Zhau, H.E.; Wang, R.; Zhu, G.; Cheng, J.; Yang, V.W.; Cheng, T.; et al. Near IR heptamethine cyanine dye-mediated cancer imaging. Clin. Cancer Res. 2010, 16, 2833–2844. [Google Scholar] [CrossRef]
  100. Rosenthal, I.; Sostaric, J.Z.; Riesz, P. Sonodynamic therapy—A review of the synergistic effects of drugs and ultrasound. Ultrason. Sonochem. 2004, 11, 349–363. [Google Scholar] [CrossRef]
  101. Jin, Z.H.; Miyoshi, N.; Ishiguro, K.; Umemura, S.; Kawabata, K.; Yumita, N.; Sakata, I.; Takaoka, K.; Udagawa, T.; Nakajima, S.; et al. Combination effect of photodynamic and sonodynamic therapy on experimental skin squamous cell carcinoma in C3H/HeN mice. J. Dermatol. 2000, 27, 294–306. [Google Scholar] [CrossRef] [PubMed]
  102. Yasui, K. Production of O Radicals from Cavitation Bubbles under Ultrasound. Molecules 2022, 27, 4788. [Google Scholar] [CrossRef] [PubMed]
  103. Xu, M.; Zhou, L.; Zheng, L.; Zhou, Q.; Liu, K.; Mao, Y.; Song, S. Sonodynamic therapy-derived multimodal synergistic cancer therapy. Cancer Lett. 2021, 497, 229–242. [Google Scholar] [CrossRef]
  104. Yang, Y.; Huang, J.; Liu, M.; Qiu, Y.; Chen, Q.; Zhao, T.; Xiao, Z.; Yang, Y.; Jiang, Y.; Huang, Q.; et al. Emerging Sonodynamic Therapy-Based Nanomedicines for Cancer Immunotherapy. Adv. Sci. 2023, 10, e2204365. [Google Scholar] [CrossRef]
  105. Wood, A.K.; Ansaloni, S.; Ziemer, L.S.; Lee, W.M.; Feldman, M.D.; Sehgal, C.M. The antivascular action of physiotherapy ultrasound on murine tumors. Ultrasound Med. Biol. 2005, 31, 1403–1410. [Google Scholar] [CrossRef] [PubMed]
  106. Wood, A.K.; Bunte, R.M.; Price, H.E.; Deitz, M.S.; Tsai, J.H.; Lee, W.M.; Sehgal, C.M. The disruption of murine tumor neovasculature by low-intensity ultrasound-comparison between 1- and 3-MHz sonication frequencies. Acad. Radiol. 2008, 15, 1133–1141. [Google Scholar] [CrossRef]
  107. Guo, J.; Pan, X.; Wang, C.; Liu, H. Molecular Imaging-Guided Sonodynamic Therapy. Bioconjugate Chem. 2022, 33, 993–1010. [Google Scholar] [CrossRef]
  108. Wang, X.; Zhang, W.; Xu, Z.; Luo, Y.; Mitchell, D.; Moss, R.W. Sonodynamic and Photodynamic Therapy in Advanced Breast Carcinoma: A Report of 3 Cases. Integr. Cancer Ther. 2009, 8, 283–287. [Google Scholar] [CrossRef]
  109. Nicolai, C.J.; Wolf, N.; Chang, I.C.; Kirn, G.; Marcus, A.; Ndubaku, C.O.; McWhirter, S.M.; Raulet, D.H. NK cells mediate clearance of CD8+ T cell-resistant tumors in response to STING agonists. Sci. Immunol. 2020, 5, eaaz2738. [Google Scholar] [CrossRef]
  110. Pantelidou, C.; Sonzogni, O.; De Oliveria Taveira, M.; Mehta, A.K.; Kothari, A.; Wang, D.; Visal, T.; Li, M.K.; Pinto, J.; Castrillon, J.A.; et al. PARP Inhibitor Efficacy Depends on CD8+ T-cell Recruitment via Intratumoral STING Pathway Activation in BRCA-Deficient Models of Triple-Negative Breast Cancer. Cancer Discov. 2019, 9, 722–737. [Google Scholar] [CrossRef]
  111. Lei, J.; Zhang, W.; Ma, L.; He, Y.; Liang, H.; Zhang, X.; Li, G.; Feng, X.; Tan, L.; Yang, C. Sonodynamic amplification of cGAS-STING activation by cobalt-based nanoagonist against bone and metastatic tumor. Biomaterials 2023, 302, 122295. [Google Scholar] [CrossRef]
  112. Pan, B.-S.; Perera, S.A.; Piesvaux, J.A.; Presland, J.P.; Schroeder, G.K.; Cumming, J.N.; Trotter, B.W.; Altman, M.D.; Buevich, A.V.; Cash, B.; et al. An orally available non-nucleotide STING agonist with antitumor activity. Science 2020, 369, eaba6098. [Google Scholar] [CrossRef] [PubMed]
  113. Yu, J.; He, S.; Zhang, C.; Xu, C.; Huang, J.; Xu, M.; Pu, K. Polymeric STING Pro-agonists for Tumor-Specific Sonodynamic Immunotherapy. Angew. Chem. Int. Ed. 2023, 62, e202307272. [Google Scholar] [CrossRef]
  114. Jiang, J.; Zhang, M.; Lyu, T.; Chen, L.; Wu, M.; Li, R.; Li, H.; Wang, X.; Jiang, X.; Zhen, X. Sono-Driven STING Activation using Semiconducting Polymeric Nanoagonists for Precision Sono-Immunotherapy of Head and Neck Squamous Cell Carcinoma. Adv. Mater. 2023, 35, e2300854. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, Y.; Bi, L.; Hu, Z.; Cao, W.; Zhuang, D. Hematoporphyrin monomethyl ether-mediated sonodynamic therapy induces A-253 cell apoptosis. Oncol. Lett. 2020, 19, 3223–3228. [Google Scholar] [CrossRef] [PubMed]
  116. Lu, D.; Wang, L.; Wang, L.; An, L.; Huo, M.; Xu, H.; Shi, J. Probiotic Engineering and Targeted Sonoimmuno-Therapy Augmented by STING Agonist. Adv. Sci. 2022, 9, e2201711. [Google Scholar] [CrossRef] [PubMed]
  117. Liu, Y.; Wang, Y.; Zhen, W.; Wang, Y.; Zhang, S.; Zhao, Y.; Song, S.; Wu, Z.; Zhang, H. Defect modified zinc oxide with augmenting sonodynamic reactive oxygen species generation. Biomaterials 2020, 251, 120075. [Google Scholar] [CrossRef] [PubMed]
  118. Rozenberg, J.M.; Kamynina, M.; Sorokin, M.; Zolotovskaia, M.; Koroleva, E.; Kremenchutckaya, K.; Gudkov, A.; Buzdin, A.; Borisov, N. The Role of the Metabolism of Zinc and Manganese Ions in Human Cancerogenesis. Biomedicines 2022, 10, 1072. [Google Scholar] [CrossRef] [PubMed]
  119. Tian, Y.; Tian, H.; Li, B.; Feng, C.; Dai, Y. An Ultrasound-Triggered STING Pathway Nanoagonist for Enhanced Chemotherapy-Induced Immunogenic Cell Death. Small 2024, 20, e2309850. [Google Scholar] [CrossRef] [PubMed]
  120. Wang, F.; Dong, G.; Ding, M.; Yu, N.; Sheng, C.; Li, J. Dual-Programmable Semiconducting Polymer NanoPROTACs for Deep-Tissue Sonodynamic-Ferroptosis Activatable Immunotherapy. Small 2024, 20, e2306378. [Google Scholar] [CrossRef] [PubMed]
  121. Tian, J.; Li, B.; Wu, C.; Li, Z.; Tang, H.; Song, W.; Qi, G.B.; Tang, Y.; Ping, Y.; Liu, B. Programmable Singlet Oxygen Battery for Automated Photodynamic Therapy Enabled by Pyridone-Pyridine Tautomer Engineering. J. Am. Chem. Soc. 2024, 146, 16458–16468. [Google Scholar] [CrossRef] [PubMed]
  122. Shaw, A.K.; Khurana, D.; Soni, S. Assessment of thermal damage for plasmonic photothermal therapy of subsurface tumors. Phys. Eng. Sci. Med. 2024; online ahead of print. [Google Scholar] [CrossRef]
Figure 1. The principles and results of two synergistic therapies of photothermal therapy and STING agonist and their effect on tumor in vivo. (A) Schematic illustration of a synergistic therapy of the photothermal transduction agent Indocyanine green (ICG) with the STING agonist DMXAA [30]. (B) Temperature development after the injection of ICG of different forms in vivo, the color represents different temperature, and have been shown in the legend [30]. (C) Tumor growth in vivo after photothermal therapy and a STING agonist alone and synergistic therapy. * p < 0.05 vs. control, *** p < 0.001 vs. control [30]. Copyright 2023 American Chemical Society. (D) Schematic illustration of a synergistic therapy of the photothermal transduction agent croconaine dye with the STING agonist diABZIs using SAPTNs [60]. (E) The images of drug distribution in vivo after the injection of SAPTNs with irradiation alone or with irradiation and the myeloperoxidase inhibitor PF1355, the color represents different temperature and the connection can be seen in the legend [60]. (F) Average volume of tumor reinjected in vivo after no treatment in the untreated group and SAPTNs plus PTT group treated by a synergistic therapy of PTT and a STING agonist using SAPTNs. *** p < 0.001 vs. control [60]. Copyright 2024 John Wiley and Sons.
Figure 1. The principles and results of two synergistic therapies of photothermal therapy and STING agonist and their effect on tumor in vivo. (A) Schematic illustration of a synergistic therapy of the photothermal transduction agent Indocyanine green (ICG) with the STING agonist DMXAA [30]. (B) Temperature development after the injection of ICG of different forms in vivo, the color represents different temperature, and have been shown in the legend [30]. (C) Tumor growth in vivo after photothermal therapy and a STING agonist alone and synergistic therapy. * p < 0.05 vs. control, *** p < 0.001 vs. control [30]. Copyright 2023 American Chemical Society. (D) Schematic illustration of a synergistic therapy of the photothermal transduction agent croconaine dye with the STING agonist diABZIs using SAPTNs [60]. (E) The images of drug distribution in vivo after the injection of SAPTNs with irradiation alone or with irradiation and the myeloperoxidase inhibitor PF1355, the color represents different temperature and the connection can be seen in the legend [60]. (F) Average volume of tumor reinjected in vivo after no treatment in the untreated group and SAPTNs plus PTT group treated by a synergistic therapy of PTT and a STING agonist using SAPTNs. *** p < 0.001 vs. control [60]. Copyright 2024 John Wiley and Sons.
Molecules 29 03704 g001
Figure 2. The principle and result of two synergistic therapies of photodynamic therapy and STING agonist on primary tumors and distant tumors in vivo. (A) Schematic illustration of a synergistic therapy of the photosensitizer Meso-tetra(carboxyphenyl) porphyrin (TCPP) and STING agonist SR-717 [96]. Volume growth of a primary tumor (B) and distant tumor (C) when treated with a photodynamic material polymeric metal−organic framework (PMOF) and SR-717 alone and a synergistic therapy of PDT and a STING agonist by using a nanoparticles called SR@PMOF combining PMOF and a STING agonist (SR-717), without light irradiation (-L) or with light irradiation (+L). ** p < 0.01 vs. control, *** p < 0.001 vs. control [96]. Copyright 2023 American Chemical Society. (D) Schematic illustration of a synergistic therapy of a nanoparticle named GM@P, consisting of a hydrophobic shell encapsulating the photosensitizer MHI148 and the STING agonist 2′3′-cGAMP [31]. Tumor growth of primary tumors (E) and distant tumors (F) after photothermal therapy using a nanoparticle with MHI148 (M@P) alone without irradiation (Group B: M@P), with irradiation (Group D: M@P + light irradiation), with a free STING agonist and irradiation (group E: M@P + light irradiation + 2′3′-cGAMP), a STING agonist alone (Group C: 2′3′-cGAMP), and a synergistic therapy of GM@P (Group F: GM@P + light irradiation) and a control group (Group A). * p < 0.05 vs. control, ** p < 0.01 vs. control [31]. Copyright 2024 American Chemical Society.
Figure 2. The principle and result of two synergistic therapies of photodynamic therapy and STING agonist on primary tumors and distant tumors in vivo. (A) Schematic illustration of a synergistic therapy of the photosensitizer Meso-tetra(carboxyphenyl) porphyrin (TCPP) and STING agonist SR-717 [96]. Volume growth of a primary tumor (B) and distant tumor (C) when treated with a photodynamic material polymeric metal−organic framework (PMOF) and SR-717 alone and a synergistic therapy of PDT and a STING agonist by using a nanoparticles called SR@PMOF combining PMOF and a STING agonist (SR-717), without light irradiation (-L) or with light irradiation (+L). ** p < 0.01 vs. control, *** p < 0.001 vs. control [96]. Copyright 2023 American Chemical Society. (D) Schematic illustration of a synergistic therapy of a nanoparticle named GM@P, consisting of a hydrophobic shell encapsulating the photosensitizer MHI148 and the STING agonist 2′3′-cGAMP [31]. Tumor growth of primary tumors (E) and distant tumors (F) after photothermal therapy using a nanoparticle with MHI148 (M@P) alone without irradiation (Group B: M@P), with irradiation (Group D: M@P + light irradiation), with a free STING agonist and irradiation (group E: M@P + light irradiation + 2′3′-cGAMP), a STING agonist alone (Group C: 2′3′-cGAMP), and a synergistic therapy of GM@P (Group F: GM@P + light irradiation) and a control group (Group A). * p < 0.05 vs. control, ** p < 0.01 vs. control [31]. Copyright 2024 American Chemical Society.
Molecules 29 03704 g002
Figure 3. The principle of a synergistic therapy of sonodynamic therapy and a cGAS-STING agonist using a material consisting of triphenyl phosphonium (TPP) and sonodynamic cobalt organic frame-work nanosheets (TPP@CoTCPP) and some experimental results. (A) Graphic illustration of TPP@CoTCPP [111]. (B) Comparison of the volume of ipsilateral and contralateral tumor in vivo in different groups treated by sonodynamic therapy and a STING agonist alone and a synergistic therapy of sonodynamic therapy and a cGAS-STING agonist. ns indicates not significant, ** p < 0.01 vs. control [111]. (C) Tumor volume of sonodynamic therapy alone and synergistic therapy [111]. Copyright 2023 Elsevier.
Figure 3. The principle of a synergistic therapy of sonodynamic therapy and a cGAS-STING agonist using a material consisting of triphenyl phosphonium (TPP) and sonodynamic cobalt organic frame-work nanosheets (TPP@CoTCPP) and some experimental results. (A) Graphic illustration of TPP@CoTCPP [111]. (B) Comparison of the volume of ipsilateral and contralateral tumor in vivo in different groups treated by sonodynamic therapy and a STING agonist alone and a synergistic therapy of sonodynamic therapy and a cGAS-STING agonist. ns indicates not significant, ** p < 0.01 vs. control [111]. (C) Tumor volume of sonodynamic therapy alone and synergistic therapy [111]. Copyright 2023 Elsevier.
Molecules 29 03704 g003
Table 1. Summary of clinical trials of a STING agonist published on the NIH website (https://clinicaltrials.gov/ by 13 July 2024).
Table 1. Summary of clinical trials of a STING agonist published on the NIH website (https://clinicaltrials.gov/ by 13 July 2024).
Clinical Trials IDStudy StartDrugSponsorStudy StatusStudy Phase
NCT041441402020-02E7766Eisai Inc. (Tokyo, Japan)TerminatedPhase I/Ib
NCT046095792020-11SNX281Stingthera, Inc. (Boston, MA, USA)TerminatedPhase I
NCT050702472022-04TAK-500Takeda (Tokyo, Japan)RecruitingPhase I/II
NCT053879282022-06KL340399Sichuan Kelun Pharmaceutical Research Institute Co., Ltd. (Chengdu, China)RecruitingPhase I
NCT060216262023-08CRD3874-SIMemorial Sloan Kettering Cancer Center (New York, NY, USA)RecruitingPhase I
Table 3. Summary of photothermal, photodynamic and sonodynamic therapies in clinical trials published on the NIH website (https://clinicaltrials.gov/ by 13 July 2024).
Table 3. Summary of photothermal, photodynamic and sonodynamic therapies in clinical trials published on the NIH website (https://clinicaltrials.gov/ by 13 July 2024).
TherapyClinical Trials IDStudy StartReport TitleSponsorStudy StatusStudy Phase
Photothermal TherapyNCT016794702012-10Efficacy Study of AuroLase Therapy in Subjects with Primary and/or Metastatic Lung TumorsNanospectra Biosciences, Inc. (Houston, TX, USA)TerminatedNot Applicable
NCT032024462016-06Randomized Clinical Trial Evaluating the Use of the Laser-Assisted Immunotherapy (LIT/inCVAX) in Advanced Breast CancerEske Corporation S.A.C (Lima, Peru)TerminatedPhase III
Photodynamic therapyNCT053860562022-12Pembrolizumab and Photodynamic Therapy in Previously Treated Metastatic Esophageal Squamous Cell CarcinomaPeking University (Beijing, China)Not Yet RecruitingPhase II
NCT055512992023-02Treatment of Non-resectable Bile Duct Cancer with Radiofrequency Ablation or Photodynamic Therapy (CARP)University of Leipzig (Leipzig, Germany)RecruitingPhase IV
NCT057364062024-02A Dose-escalation Clinical Study of Intraoperative Photodynamic Therapy of GlioblastomaHemerion Therapeutics (Villeneuve d’Ascq, France)RecruitingPhase I/II
NCT053749152024-02Efficacy and Safety Study of REM-001 Photodynamic Therapy for Treatment of Cutaneous Metastatic Breast Cancer (CMBC)Kintara Therapeutics, Inc. (San Diego, CA, USA)RecruitingPhase II
NCT063811542024-06Photoradiation with Verteporfin to Facilitate Immunologic Activity of Pembrolizumab in Unresectable, Locally Advance or Metastatic Pancreatic CancerMayo Clinic (Scottsdale, AZ, USA)Not Yet RecruitingPhase II
NCT063066382024-07Interstitial Photodynamic Therapy Following Palliative Radiotherapy in Treating Patients with Inoperable Malignant Central Airway ObstructionRoswell Park Cancer Institute (Buffalo, NY, USA)Not Yet RecruitingPhase I/II
NCT064372882024-07Hematoporphyrin Photodynamic Therapy for Esophageal CancerSun Yat-sen University (Guangzhou, China)Not Yet RecruitingPhase II
Sonodynamic therapyNCT045596852021-03Study of Sonodynamic Therapy in Participants with Recurrent High-Grade GliomaNader Sanai (Phoenix, AZ, USA)RecruitingEarly Phase I
NCT053624092022-06Study to Evaluate 5-ALA Combined with CV01 Delivery of Ultrasound in Recurrent High Grade GliomaAlpheus Medical, Inc. (Chanhassen, MN, USA)Active, Not RecruitingPhase I
NCT051235342022-08A Phase 2 Study of Sonodynamic Therapy Using SONALA-001 and Exablate 4000 Type 2.0 in Patients With DIPGSonALAsense, Inc. (Berkeley, CA, USA)RecruitingPhase II
NCT048459192023-02Sonodynamic Therapy with ExAblate System in Glioblastoma Patients (Sonic ALA)Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta (Milan, Italy)Not Yet RecruitingPhase II
NCT060397092024-01Sonodynamic Therapy in Patients with Recurrent GBM (GBM 001)Shayan Moosa, MD (Charlottesville, VA, USA)RecruitingPhase I
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fang, K.; Zhang, H.; Kong, Q.; Ma, Y.; Xiong, T.; Qin, T.; Li, S.; Zhu, X. Recent Progress in Photothermal, Photodynamic and Sonodynamic Cancer Therapy: Through the cGAS-STING Pathway to Efficacy-Enhancing Strategies. Molecules 2024, 29, 3704. https://doi.org/10.3390/molecules29153704

AMA Style

Fang K, Zhang H, Kong Q, Ma Y, Xiong T, Qin T, Li S, Zhu X. Recent Progress in Photothermal, Photodynamic and Sonodynamic Cancer Therapy: Through the cGAS-STING Pathway to Efficacy-Enhancing Strategies. Molecules. 2024; 29(15):3704. https://doi.org/10.3390/molecules29153704

Chicago/Turabian Style

Fang, Kelan, Huiling Zhang, Qinghong Kong, Yunli Ma, Tianchan Xiong, Tengyao Qin, Sanhua Li, and Xinting Zhu. 2024. "Recent Progress in Photothermal, Photodynamic and Sonodynamic Cancer Therapy: Through the cGAS-STING Pathway to Efficacy-Enhancing Strategies" Molecules 29, no. 15: 3704. https://doi.org/10.3390/molecules29153704

Article Metrics

Back to TopTop