Planta Med 2024; 90(05): 368-379
DOI: 10.1055/a-2257-9194
Biological and Pharmacological Activity
Reviews

Research Progress of Natural Product Photosensitizers in Photodynamic Therapy

Xiaoxia Zhou
1   Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
,
Xufang Ying
2   Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
,
Linjie Wu
2   Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
,
Liqin Liu
1   Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
,
Ying Wang
1   Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
,
Ying He
1   Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
,
Min Han
2   Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
3   Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
4   Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, China
› Institutsangaben
This work was supported by the National Key Research and Development Program of China (No. 2022YFE0107800).
 

Abstract

Photodynamic therapy is a noninvasive cancer treatment that utilizes photosensitizers to generate reactive oxygen species upon light exposure, leading to tumor cell apoptosis. Although photosensitizers have shown efficacy in clinical practice, they are associated with certain disadvantages, such as a certain degree of toxicity and limited availability. Recent studies have shown that natural product photosensitizers offer promising options due to their low toxicity and potential therapeutic effects. In this review, we provide a summary and evaluation of the current clinical photosensitizers that are commonly used and delve into the anticancer potential of natural product photosensitizers like psoralens, quinonoids, chlorophyll derivatives, curcumin, chrysophanol, doxorubicin, tetracyclines, Leguminosae extracts, and Lonicera japonica extract. The emphasis is on their phototoxicity, pharmacological benefits, and effectiveness against different types of diseases. Novel and more effective natural product photosensitizers for future clinical application are yet to be explored in further research. In conclusion, natural product photosensitizers have potential in photodynamic therapy and represent a promising area of research for cancer treatment.


#

Introduction

Photodynamic therapy (PDT), as a novel model for treating tumors, was initially applied in the treatment of skin diseases [1]. In 1975, Kelly et al. [2] demonstrated the efficacy of PDT in treating superficial transitional cell carcinoma of the bladder, following which its application has expanded from skin diseases to tumors. At present, PDT has been approved by the FDA as a curative or palliative treatment for a variety of solid tumors [3], such as glioblastoma [4], [5], [6], oral cancer [7], [8], [9], breast cancer [10], [11], pancreatic cancer [12], [13], and prolonging the survival of inoperable cancer patients and significantly improving their quality of life [14]. In addition, PDT can also effectively treat vascular lesions and microbial infections [15].

To achieve tumor treatment, PDT employs light source of specific wavelengths to activate photosensitizers at the tumor site, where highly toxic reactive oxygen species (ROS) are produced to effectively kill the surrounding tumor cells, resulting in tumor treatment [16]. The mechanisms behind PDT-mediated tumor damage are primarily categorized into three types:

  1. During PDT, a photodynamic reaction occurs, which directly induces apoptosis and necrosis of tumor cells through the production of ROS, particularly singlet oxygen. Additionally, the mass production of ROS will upregulate autophagy, causing autophagic cell death [17].

  2. PDT leads to the destruction of the vascular system associated with the tumor, ultimately resulting in tumor infarction [18], [19].

  3. Numerous proinflammatory factors are released after the apoptosis or necrosis of tumor cells, inducing early inflammatory responses and activating the immune system [20].

Although traditional cancer treatments like surgical resection, chemotherapy, and radiotherapy are beneficial, they still have some drawbacks. For example, surgical resection causes big surgical trauma. Cancers treated with chemotherapy can develop resistance. Radiotherapy has significant side effects, lacks specificity, and is prone to produce systemic toxicity. In comparison, PDT has a host of advantages over the aforementioned treatments, including less trauma, lower toxicity, higher tissue targeting, a wider range of applications, reproducible treatment, and lower long-term morbidity [21], [22]. Moreover, PDT can improve therapeutic effects when combined with traditional treatments. The development of PDT has been an active area of research, with many advances being made in the field. Since 2016, the number of academic papers related to PDT in the Web of Science database has been consistently increasing every year ([Fig. 1]). However, the growth of the field has not been without its challenges. In recent years, some companies have failed, and clinical trials have been unsuccessful, leading to stagnation in the field. While the number of research papers and publications in the field has increased, much of this growth is due to the development of nanotechnology with no clinical applications. Despite these challenges, researchers continue to explore new approaches and technologies to advance the field of PDT and improve its effectiveness as a treatment option. One approach that has shown promise is the search for new photosensitizing agents.

Zoom Image
Fig. 1 PDT-related publications.

The occurrence of photodynamic reactions requires three basic elements: photosensitizers (PSs), appropriate illumination, and tissue oxygen [23], [24]. Initially, the photosensitizer is enriched at the tumor site and mainly concentrated in the organelle, then irradiated with a laser.

Upon absorbing energy, the photosensitizer transitions from the ground singlet state (S0) to the transient excited singlet state (S1), which then converts into a long-lasting excited triplet state (T1) [16], [25]. Photodynamic reactions can be classified into two types: the type I mechanism and the type II mechanism ([Fig. 2]). During photodynamic reactions, hydrogen atom (electron) transfer takes place between the photosensitizer in the excited triplet state and the biological macromolecule or substrate, leading to the creation of free radicals (e.g., superoxide anions, hydroxyl radicals, etc.) [26], [27], which damage the target cell. This reaction is known as the type I mechanism. The type II mechanism involves the photosensitizer in its long-lived excited triplet state directly transferring energy to the oxygen molecule in its triplet ground state. This produces singlet oxygen (1O2), which further interacts with lipids, proteins, and nucleic acids, ultimately resulting in cell necrosis or apoptosis [25], [28]. The two mechanisms occur simultaneously, with the balance between them being determined by factors such as the type of photosensitizer used, the concentration of substrate and oxygen, and the degree of photosensitizer binding to the substrate. The most important single factor is the redox potential of the photosensitizer.

Zoom Image
Fig. 2 Mechanism of photodynamic reactions.

Selecting an appropriate photosensitizer is essential for the success of PDT. The intrinsic properties of the chosen photosensitizer determine its therapeutic efficacy, as it can absorb light of specific wavelengths and trigger photochemical or photophysical reactions [29], [30]. An ideal photosensitizer should be a single well-characterized compound, with a known and constant composition. It ought to effectively generate ROS, selectively accumulate in target tissues, be harmless in the absence of radiation, and be easily eliminated from the body [31], [32], [33], [34]. According to literature reports, over ten types of photosensitizers have been approved for clinical use worldwide ([Table 1]).

Table 1 Photosensitizers approved for clinical use worldwide.

Generation

Photosensitizer

Absorption range

Status

Indication

Refs.

First

Photofrin

~ 630 nm

Approved in Canada (1993), Japan (1994), USA (1996), Russia (1997), Germany (1997)

Lung cancer, esophageal cancer, gastric cancer, cervical cancer, etc.

[35], [36], [37]

Photogem

[38]

Photosan

[39]

HiPorfin

~ 630 nm

Approved in China (1998)

Detect and treat superficial cancer of oral cavity, bladder, bronchus, lung, digestive system and precancerous lesion of vitiligo, port wine stains

[40], [41], [42]

Hemoporfin

480 – 580 nm

Approved in China (2003)

Port wine stains

[43]

Second

Levulan

410 – 635 nm

Approved in USA (1999)

Actinic keratosis, malignant glioma, bladder cancer, Barrettʼs esophagus, vulvar lichen sclerosus

[44], [45], [46], [47], [48]

Metvix/Metvixa

~ 635 nm

Approved in EU (2001), New Zealand (2002), Australia (2003), USA (2004)

Actinic keratosis, nodular basal cell carcinoma, squamous cell carcinoma in situ, Bowenʼs disease

[49], [50], [51]

Hexvix/Cysview

380 – 450 nm

Approved in EU (2001), Sweden (2004), USA (2010)

Detection of bladder cancer

[52]

Visudyne

~ 689 nm

Approved in USA (2000)

Subfoveal choroidal neovascularization caused by age-related macular degeneration, central serous chorioretinopathy, choroidal hemangioma, gastric cancer

[53], [54], [55], [56]

Foscan

652 nm
514 nm

Approved in EU (2001)

Palliative treatment of patients with advanced head and neck cancer, mesothelioma

[57], [58]

Photolon/Fotolon

670 nm
663 nm

Approved in Russia (2002)

Cervical intraepithelial neoplasia, diagnosis and treatment for cutaneous malignant tumor

[59]

Laserphyrin

~ 664 nm

Approved in Japan (2004)

Early-stage endobronchial cancer, prostate cancer, bile duct carcinoma, diagnosis and therapy of malignant glioma, advanced-aged patients suffering from inoperable gastric cancer, oral squamous cell carcinoma, biliary cancer

[60], [61], [62], [63], [64]

Photosense

~ 675 nm

Approved in Russia (2001)

Breast cancer, eye diseases, skin cancer, head and neck tumors

[65], [66], [67], [68]

ICG

795 – 845 nm

Approved in USA

Occult subfoveal choroidal neovascularization caused by age-related macular degeneration, melanomas, periodontal therapy

[69], [70], [71]

Methylene blue

600 – 665 nm

Approved in Canada

Basal cell carcinoma, Kaposiʼs sarcoma, melanoma, virus, fungal infections

[72]

PDT was initially proposed by the German scholar Raab in 1900. In the 1960 s, the first-generation photosensitizers hematoporphyrin derivative (HpD) [73] emerged, ushering in the advance of PDT. At that time, the component of HpD was not fixed, and the proportion of active ingredients was small. In 1972, Diamond and his team [75] purified HpD and obtained the product Photofrin I. In 1983, Dougherty et al. [75] isolated eight components of HPD using reverse-phase HPLC and gel filtration chromatography and identified their biological activity. Photofrin II was created during this process. In 1993, Photofrin was approved for clinical use, mainly for the treatment of esophageal cancer, lung cancer, minimally invasive bronchial cancer, stomach and bladder cancer, cervical dysplasia, and other conditions. To date, it remains the most widely used photosensitizer in clinical practice. Subsequently, Photogem, Photosan, and HiPorfin, which are similar to Photofrin, were launched in different countries. However, due to the shortcomings of the first-generation photosensitizers such as poor stability, shallow penetration depth, and prolonged residence time in vivo, their clinical application was limited [76].

Most of the second-generation photosensitizers are derived from tetrapyrrole structures, mainly including porphyrin derivatives, metallophthalocyanines and polycycloquinones, such as 5-aminolevulinic acid (5-ALA; Levulan), methyl aminolevulinate (MAL; Metvix), hexaminolevulinate (HAL; Hexvix/Cysview), and verteporfin (Visudyne). These are currently available on the market. Notably, 5-ALA and its analogsʼ therapeutic efficacy is attributed to the metabolic conversion to protoporphyrin, which serves as the active photosensitizer in the treatment process. In addition, chlorin photosensitizers like temoporfin (Foscan) and talaporfin (Laserphyrin) are also available. Chlorin e6 (Fotolon) is a representative second-generation porphyrin photosensitizer approved by the FDA [77]. It has been widely used in PDT [78], [79], with initial applications focused on the treatment of skin and mucous membranes, while researchers are currently exploring its effectiveness in the treatment of breast cancer [80]. Photosense is a phthalocyanine photosensitizer approved in Russia in 2001 for the treatment of breast, eye, and skin cancer. There are also specific photosensitizers available, such as the FDA-approved near-infrared fluorescent dye indocyanine green (ICG), for diagnostic cardiology, hepatology, ophthalmology, and fluorescence-guided cancer surgery, as well as for the treatment of melanoma and periodontal diseases. Derived from phenothiazine, methylene blue functions as a photosensitizer with clinical applications in various fields, including basal cell carcinoma, Kaposiʼs sarcoma, melanoma, viral infections, fungal infections, and others. The second-generation photosensitizers are considered safer and more stable than their first-generation counterparts. Their absorption in the higher wavelength area (compared with first-generation photosensitizers) allows them to be reached by deeper penetrating light, resulting in deeper penetration and increased singlet oxygen production [81]. However, their poor water solubility poses significant limitations for intravenous administration.

Chemical modification of third-generation photosensitizers through combination with different factors or groups improves their targeting, water solubility, aggregation, and utilization, while reducing accidental injury to normal skin [29], [82]. Although most third-generation photosensitizers are in the clinical research stage, they are not yet available on the market.

In general, photosensitizers used in clinical practice are often complex and susceptible to biological metabolism. Apart from that, the degree of photosensitization damage is difficult to control, limiting their clinical application to varying degrees. However, partial natural product photosensitizers possess higher clinical translational potential [83], [84], specific biological activities, and selectivity [85], [86], making them a viable option for PDT. Owing to the abundance of natural resources in the Nature Kingdom, some of which are light sensitive, there is an increasing interest in developing new and efficient photosensitizers from natural product extracts. Researchers worldwide have explored the light-sensitive substances in natural products for PDT. The research progress of natural product photosensitizers in PDT is summarized in [Table 2]. The chemical structures of partial natural product sensitizers are illustrated in [Fig. 3].

Table 2 Natural products with photosensitive activity.

Component

Absorption range

Clinical application

Cell lines/animals

Psoralen (PSO)

~320–360 nm

Tumor

Siso tumor line, MCE-1 cell line

Hypericin

~600 nm

Tumor, virus

MCF-7 cell line, colon cancer cells, leukemia cells

Hypocrellin

/

Tumor, virus

Human brain tumor cells, A375-S2 cell line

Chlorophyll derivatives

/

Tumor, acne and other diseases related to sebaceous glands

Mouse S180 transplanted sarcoma, human osteosarcoma, golden hamsters

Curcumin

445 nm

Tumor, bacteria, fungi

MCF-7 cell line, H8 cells, gastric adenocarcinoma cells, cervical cancer cells, liver cancer cells

Chrysophanol

> 430 nm

Microvascular diseases

/

Leguminosae extract

504 nm

Tumor

Ehrlich ascites cancer cells (EAC)

Honeysuckle stem extract

468 nm

Tumor

Ehrlich ascites cancer cells (EAC), S180 solid tumors

Zoom Image
Fig. 3 Chemical structures of partial natural product photosensitizers.

#

Psoralen

Psoralen (PSO), an early clinical application of photosensitizing natural products, is widely distributed in the plant kingdom. It can be sourced from Fraxini Cortex, Fructus Psoraleae, Radix Angelicae Dahuricae, Radix Angelicae Pubescentis, Radix Peucedani, and Fructus Cnidii. PSO can be excited by light with a wavelength of 320 – 360 nm. The absorption of the wavelength of PSO is below 600 nm, which is considered a disadvantage as a photosensitizer. Pharmacological studies have shown that it can improve osteogenetic ability, inhibit cancer cell migration and metastasis, reduce inflammation, improve atherosclerosis in rats, and protect the heart. At present, PSO-mediated PDT is primarily used in the field of oncology. To investigate the potential for growth inhibition of Siso tumor strains in vitro and in vivo, Zhao et al. [87] compared the photosensitization of PSO using different radiation sources. Their findings revealed that while single radiation or in vitro treatment alone proved insufficient in killing tumor cells, the combination of PSO and cobalt 60 irradiation resulted in a significant radiosensitizing enhancement effect. Overall, the study highlights the promising potential of PDT for the treatment of cancer, particularly when used in combination with traditional treatments. PSO has been employed by scholars to enhance its antitumor effect through its photosensitivity properties [88], [89], [90]. Yan et al. [91] proposed that PSO can markedly reduce the rate of lung metastasis in nude mice with a human mucoepidermoid carcinoma cell line (MCE-1 cell line). This is achieved by damaging DNA under the activation of the light source and affecting the expression of lipids and proteins, leading to antitumor effects.


#

Quinonoids

Quinonoids include benzoquinone, anthraquinone, and perylenequinones. Almost all quinonoids are photosensitive, and many of them have antitumor effects. Therefore, current research on quinonoids are chiefly focused on their potential in the field of oncology. Quinonoids that undergo photodynamic reactions are widely distributed in various plants, including hypecorine, fagomine, and cercosporin.

Hypericin

Hypericin (Hyp), a natural photosensitizer isolated from hypericum species, is a polycyclic anthraquinone compound that is easily taken up by tumor cells. It possesses strong photosensitivity properties, low dark toxicity, and high singlet oxygen quantum yields. As a result, it is an effective agent for PDT and can be used to treat a variety of solid tumors [92], [94], [95], [96]. Chen et al. [97] showed that human breast cancer cells (MCF-7) induced by Hyp-PDT (PDT mediated by Hyp) were more sensitive to killer cells (CIK), indicating a positive correlation between Hyp concentration and the effectiveness of CIK. Hu et al. [98] observed that Hyp was capable of inducing S-phase cell cycle arrest and apoptosis, thereby inhibiting the proliferation of colon cancer cells. Zhang et al. [99] proved the effectiveness of Hpy-mediated PDT in inhibiting the proliferation of various leukemia cells and achieving anticancer effects. Chen et al. [100] found that Hpy can inhibit apoptosis caused by infectious bronchial virus (IBV) in chickens. Current research indicates that Hyp can perform photochemical reactions under specific light conditions, and that the ability to kill viruses and tumor cells is more pronounced under these conditions than in the absence of light. These findings suggest that Hpy possesses photochemical reaction characteristics and demonstrates potential for PDT [101], [102].


#

Hypocrellin

Hypocrellin, a secondary metabolite found in a few bamboo parasitic fungi, such as Hypocrella bambusea and Shiraia bambusicola of Hypocreaceae, belongs to the perylenequinones derivative. Its main source is through the intramycelial secondary metabolic synthesis pathway. As a perylenequinones compound, the structure of hypocrellin endows it with certain photodynamic activity and exhibits a promising phototherapeutic effect. It was first isolated from the fleshy fruiting body of the medicinal fungus H. bambusae in 1980 by Wan et al. [103] and was named after its place of origin. In 1991, Kishi et al. [104] isolated hypocrellin from S. bambusicola, another medicinal fungus. Further research has revealed that hypocrellin also has effects such as good photosensitivity to kill tumor cells and inhibition of HIV-1 [105], making it a promising therapeutic option. Compared with the antitumor drug hematoporphyrin derivative, hypocrellin offers certain advantages. It is easily purified, has a high quantum yield of photochemical reactions, and can be eliminated from normal tissues quickly [106]. Hypocrellin A (HA) and hypocrellin B (HB) can promote human brain tumor cell death by producing factors that can inhibit angiogenesis through photodynamic action [107]. The chemical structure of HA may play a key role in inducing apoptosis. HA has been found to produce hydroxyl radicals, which can damage the DNA of tumor cells and induce apoptosis under light conditions. Additionally, HA has been shown to induce apoptosis in human melanoma cells (A375-S2) under light-free conditions and increase expression of Cylin B1 mRNA and stall the cell cycle in the S-phase [108], [109]. However, as a fat-soluble substance, hypocrellin has low water solubility and no specific tissue distribution, which limits its application in PDT. To address this, physical embedding or chemical modification techniques such as embedding with liposomes or cyclodextrin can be used to enhance its water solubility [110]. The photodamage mechanism of hypocrellin involves not only free radicals and singlet oxygen, but also semiquinone radical anion and semiquinone radical cations, which are produced by self-exchange electron transfer of HA and HB under light conditions. In short, hypocrellin causes cell death or apoptosis through a comprehensive multisite, and multi-mechanism photosensitive damage.


#

Cercosporin

Cercosporin is a well-known fungal metabolite recognized for its role in inducing light-activated plant damage through singlet oxygen generation. Mastrangelopoulou et al. [111] examined the photocytotoxicity of cercosporin in human cell lines, including two glioblastoma multiforme lines (T98G and U87) and one breast adenocarcinoma line (MCF7). Upon excitation at 532 nm, cercosporin exhibited remarkable efficiency in producing singlet oxygen. Although cell loading of cercosporin was comparable between MCF7 and U87 cell lines, it was notably elevated by approximately threefold in T98G cells. Cercosporin consistently exhibited subcellular localization within both mitochondria and the endoplasmic reticulum across all cases. Upon irradiation with light around 450 nm, T98G cells displayed heightened susceptibility to cercosporin-mediated PDT, primarily attributed to their elevated uptake of cercosporin. Metabolic analyses conducted prior to and 1 hour after cercosporin-mediated PDT revealed that such treatment led to a pronounced bioenergetic collapse affecting both respiratory and glycolytic activities across all tested cell lines. Cercosporin acts as a potent photosensitizer, ideally suited for superficial PDT due to its short activation wavelength. This makes it especially effective when avoiding perforations is a priority.


#
#

Chlorophyll Derivatives

Chlorophyll derivatives (CPDs) are a new type of photosensitizer recently extracted from natural product silkworm excrement. Apart from those containing magnesium, many derivatives have been artificially synthesized. The term “derivatives” primarily refers to the replacement of the core magnesium atom of chlorophyll with metal ions, including palladium, zinc, copper, nickel, cobalt, iron, and others. This process generates derivatives that are resistant to both light and heat, but which also lose their fluorescence. This synthetic approach significantly broadens the applicability of CPD. Liu et al. [112] conducted a prospective study on the prevention of recurrence of invasive bladder cancer in 32 patients with chlorophyll derivative-PDT (CPD4-PDT) after surgery, and the results demonstrated that compared with the traditionally used HpDs, CPD4 exhibits similar clinical efficacy but with reduced phototoxic side effects. Notably, the time patients need to spend in darkness after CPD4-PDT is shorter, contributing to a more favorable post-treatment experience. Moreover, the photoinactivation effect of human cancer cells in vitro and the photodynamic efficacy of animal transplanted tumors were significantly higher than HPpDs. Cao et al. [113] proposed that CPD4-induced PDT has a direct killing effect on endothelial cells (ECs) and has the characteristics of irreversibility and dose dependence. According to Zhang et al. [114], PDT was found to have an inhibitory effect on mouse S180 transplanted sarcoma. This research provided experimental evidence for the development of new drugs for the treatment of malignant tumors. CPD, represented by methyl pyropheophorbide-α (Mppα), is a chlorin compound with high photosensitivity. It is made of chlorophyll extracted from natural product silkworm excrement and undergoes reprocessing for use in PDT. MPPα-PDT is a common treatment for tumors in clinical practice, including ovarian cancer, nasopharyngeal cancer, and breast cancer, which can induce apoptosis, autophagy, inhibit the growth and reproduction of tumor cells, and is usually associated with endoplasmic reticulum stress pathways. At the same time, studies have indicated that the sensitivity of human osteosarcoma (HOS) cells to MPPα- PDT can be enhanced, and the clinical treatment effect can be improved greatly by silencing the expression of X-box binding protein 1 (XBP1) or blocking the protein kinase RNA-like endoplasmic reticulum kinase (PERK) signaling pathway [115], [116]. In addition to tumors, as evidenced by Wang [117], MPPα-PDT was found to significantly reduce the number, volume, and thickness of sebaceous glands in golden hamsters, providing a basis for the potential use of MPPα-PDT in the treatment of acne and other diseases related to sebaceous glands.


#

Curcumin

Curcuma longa L. is the rhizome of a perennial herb of Zingiberaceae, a natural product that activates blood circulation and resolves stasis, removes masses and alleviates pain, and clears and reduces stagnant heat. Curcumin is a diketone component extracted from the rhizomes of certain plants, such as those in the Zingiberaceae and Araceae families. C. longa L. contains about 3 ~ 6% curcumin, which is a rare pigment with a diketone structure in the plant world and is often used as a seasoning and food dye in daily life. Zeng [118] proved that curcumin has a significant inhibitory effect on human breast cancer MCF-7 cells when exposed to light. This effect increases with higher concentration of curcumin.

In a cell experiment conducted by Mou et al. [119], curcumin was found to have a cytotoxic effect on H8 cells when activated by an excitation wavelength of 445 nm. The study also found that the killing effect of PDT on tumor cells was proportional to the concentration of the drug. Curcumin-mediated PDT has been shown to effectively inhibit and kill gastric adenocarcinoma, cervical cancer, liver cancer, and other tumor cells. In addition, this therapy has a strong antibacterial effect and can inactivate a variety of bacteria and fungi, including Pseudomonas fluorescens and Candida albicans. Curcumin can be used not only as a photosensitizer but also as a sonosensitizer for sonodynamic therapy. Studies have shown that curcumin is more effective in inducing apoptosis under light conditions (photosensitized curcumin) than under no light conditions (non-photosensitized curcumin) [120]. This suggests that curcumin possesses photochemical reaction properties, making it an efficient and low-toxic photosensitizer for inducing apoptosis.

However, there are studies that suggest certain drawbacks associated with curcumin. These include its poor pharmacokinetic/pharmacodynamic (PK/PD) properties, limited efficacy in various disease models, and potential toxic effects observed under specific testing conditions [121]. Consequently, when evaluating the therapeutic effectiveness of curcumin, a comprehensive analysis of its limitations becomes essential.


#

Chrysophanol

Chrysophanol, a natural anthraquinone with a wide range of biological therapeutic potential, is the main active ingredient of the natural products Rheum palmatum L., Polygonum multiflorum Thunb. and Polygonum cuspidatum Sieb. et Zucc. Rao et al. [122]. investigated the photosensitization activity of chrysophanol, which was isolated and purified from R. palmatum L. by electron spin resonance. As discovered in the study, when chrysophanol was irradiated with visible light with a wavelength greater than 430 nm, a semiquinone radical anion could be produced. Furthermore, the addition of dihydrocoenzyme to enhance light can assist chrysophanol in producing singlet oxygen and hydroxyl radicals, suggesting that the photosensitization mechanism of chrysophanol involves both the type I mechanism of electron transfer and type II mechanism of energy transfer. Based on its observed effects, chrysophanol has the potential to be developed as a promising photodynamic agent for the treatment of microvascular diseases.


#

Antibiotics

Tetracyclines

Tetracyclines are widely recognized as established antibiotics but can exhibit phototoxicity as a side effect. Antimicrobial photodynamic inactivation employs harmless light in combination with nontoxic dyes to eliminate microbial cells by generating ROS. Tetracyclines have the capability to function as light-activated antibiotics by binding to bacterial cells and inducing cell death exclusively upon illumination. Bacteria are killed by photoactivation of tetracyclines in the absence of oxygen [123].


#

Doxorubicin

Doxorubicin is in the anthracycline and antitumor antibiotic family of medications. It works in part by interfering with the function of DNA. Researchers exploited the intrinsic photosensitizing properties of doxorubicin to enhance its anticancer activity in leukemia, breast, and epidermoid carcinoma cells, upon irradiation. Light can selectively induce the localized formation of ROS, following photophysical pathways. Upon irradiation, doxorubicin exhibited a concentration-dependent capability to produce peroxides and singlet oxygen. The underlying mechanisms leading to the increase in its cytotoxic activity were intracellular ROS generation and the induction of necrotic cell death. The nuclear localization of doxorubicin represents an added value for its use as a photosensitizer. Employing doxorubicin in photodynamic cancer therapy (PCT), where it functions concurrently as both a chemotherapeutic agent and a photosensitizer, may allow (i) an augmentation of the drugʼs anticancer effects, and (ii) a reduction in its dosage, consequently mitigating dose-related adverse effects [124].


#
#

Other Natural Product Extracts

Leguminosae extract

In a study conducted by Chen et al. [125], the photosensitization properties of an extract from the natural product Leguminosae were analyzed. The results demonstrated that the extract exhibited a significant excitation peak at 504 nm, which is a crucial factor for the occurrence of photosensitization. Experiments have proved that this extract has a significant photodynamic inactivation effect on Ehrlich ascites cancer (EAC) cells. It is capable of mediating light energy to effectively kill tumor cells, indicating its potent photosensitizing activity. Therefore, natural product Leguminosae can be considered an effective natural photosensitizer. There is a dose-effect relationship between Leguminosae extract and the photodynamic inactivation of EAC cells. As the concentration of Leguminosae extract increases, more molecules are excited by photons and undergo photochemical reactions. This leads to increased production of singlet oxygen or free radicals, resulting in a stronger killing effect on EAC cells. While Leguminosae extract itself does exhibit a direct killing effect on EAC cells, this effect is significantly less potent compared to the cell death induced by photodynamic effects.


#

Lonicera japonica extract

To observe the photosensitization of two extracts of Lonicera japonica, 85 221A60 and 85 221A95, Yao and Wu [126] conducted experiments on EAC cells in vitro and photodynamic studies on S180 solid tumors in vivo using a xenon high-pressure lamp as the excitation light source and mouse transplanted tumors as an animal model. Extract 85 221A95 has a significant photodynamic therapeutic effect on Kunming mice with S180 solid tumors and shows a significant photosensitization effect on EAC cells, particularly at a concentration of 500 mg/mL. At this concentration, the mortality rate of EAC cells was over 97%. Extract 85 221A60 has a unique excitation peak at 468 nm and has been shown to have photosensitizing activity in vitro. While it also exhibits a direct killing effect on EAC cells, this effect is significantly lower compared to the mortality rate caused by the photodynamic effects of 85 221A60 on EAC cells. In conclusion, it can be stated that L. japonica contains photosensitizers with promising applications.


#

Other natural products

Liao et al. [127] conducted a study to analyze the fluorescence properties of thirteen natural product extracts, including Cortex Phellodendri Chinensis, Radix Sophorae Flavescentis, Radix Scutellariae, Rhizoma Coptidis, Cortex Fraxini, Fructus Psoraleae, Radix Arnebiae, Radix Peucedani, Radix Angelicae Dahuricae, Herba Lycopi, Rhizoma et Radix Notopterygii, Radix Angelicae Pubescentis, and Radix Sophorae Tonkinensis. The fluorescence excitation and emission wavelengths were measured as well as the cellular fluorescence intensity after being taken up by 823 human gastric cancer cells. Additionally, the distribution and affinity sites of the fluorescent substances were studied using fixed and live-cell staining techniques. The effect of pH on the fluorescence intensity of the cells stained by extracts was also measured. Based on this, further experiments were selectively carried out to test the photosensitive anticancer of 13 natural products. The results showed that the chromosomes of 13 natural products exhibited strong fluorescence due to the presence of fluorescent substances that were partially absorbed by living cells, to a certain extent. However, it should be noted that the fluorescence intensity of approximately half of the extracted sample was affected by pH during uptake and distribution in living cells. This information provided valuable insights into the optimal storage conditions and administration routes for the corresponding natural product. Among the 13 natural products previously mentioned, only Radix Scutellariae, Cortex Fraxini, and Herba Lycopi did not exhibit detectable excitation and emission wavelengths of fluorescence. Several fluorescence detection indicators consistently showed that Cortex Phellodendri Chinensis and Rhizoma Coptidis had strong fluorescence, good cell uptake, and a wide distribution of fluorescent substances. The indicators also revealed that these substances were minimally affected by changes in the pH value and had obvious retention sites within cells. As such, these findings suggested that these substances may have the most significant photosensitivity effect, followed by Radix Arnebiae and Radix Sophorae Flavescentis. The authors concluded that there is potential for further research and attention on natural products such as Rhizoma Coptidis, Cortex Phellodendri Chinensis, Radix Sophorae Flavescentis, Fructus Psoraleae, Radix Arnebiae, and Rhizoma et Radix Notopterygii. These natural products may serve as sources for new photosensitizers with low toxicity and high efficacy.

Scotti et al. [128] screened ten medicinal plants from the Chinese Pharmacopoeia for compounds with known or potential photosensitizing activity, and conducted a detailed evaluation of their chemical composition, pharmacological activity, toxicity, and safety. These ten medicinal plants are Bistortae Rhizoma, Conyzae Herba, Echinopsis Radix, Knoxiae Radix, Polygalae Japonicae Herba, Polygoni Perfoliati Herba, Saururi Herba, Semiaquilegiae Radix, and Trachelospermi Caulis et Folium. These plants contain compounds with antibacterial, anti-inflammatory, wound healing, and photosensitizing activities, such as flavonoids, anthraquinones, and indole alkaloids. Further research and assessment are needed to explore the photosensitizing activity and PDT potential of these compounds.

Research reveals the crucial role of octyl gallate (OG) in PDT. It collaborates with blue light, swiftly eliminating Vibrio parahaemolyticus in both planktonic and biofilm states. Combining with ascorbic acid (AA) significantly enhances its effectiveness, while potassium iodide (KI) boosts efficient sterilization in water. This provides novel strategies for microbial safety and environmental-friendly approaches in the food industry and drinking water treatment [129], [130], [131].

It is worth noting that highly active singlet oxygen is produced within photosynthetic organisms when they absorb more light energy than is required for photosynthesis. This can result in photo-oxidative stress. The singlet oxygen can be quenched by β-carotene and α-tocopherol or react with the D1 protein of photosystem II as a target. If not completely quenched, it can specifically trigger the upregulation of gene expression involved in the molecular defense response of plants to photo-oxidative stress [132]. This implies that evolution has ensured that plants reliant on sunlight for growth do not possess highly active photosensitizers, as they would destroy themselves. Chlorophyll is a good example because there are complex systems designed to protect plants against any singlet oxygen generated as a by-product of photosynthesis.


#
#

Conclusion

To sum up, natural product photosensitizers have shown great potential in the field of PDT. Natural product photosensitizers that are currently available can be classified into several categories, including PSO, quinonoids, CPDs, curcumin, chrysophanol, Leguminosae extracts, and L. japonica extract. The depth of action of photosensitizers is directly related to their absorption wavelength, with longer wavelengths allowing for deeper penetration. In this paper, the depth of action of natural photosensitizers is listed in the following order: PSO, curcumin, Hpy, and hypocrellin. Depending on the nature of the lesion, various photosensitizers may be selected for PDT. Although using natural products as photosensitizers has not yet demonstrated significant advantages in terms of absorption wavelength and safety, some photosensitizers derived from natural sources show higher clinical translational potential due to the diversity and complexity of their molecular structures [83], [84]. Moreover, the specific biological activities and selectivity exhibited by natural products give them more prominent photosensitive characteristics compared to traditional photosensitizers [85], [86]. This is likely to offer valuable insights and serve as a reference for the future development of novel photosensitizers. The main disadvantage of natural product photosensitizers is their short absorption wavelengths. To address those issues, chemical modification can be employed to increase their absorption wavelengths and enhance PDT efficacy. The inherent targeting capability of photosensitizers is limited. Therefore, various strategies have been explored to achieve targeting in PDT. One approach involves conjugating the photosensitizer with targeting molecules such as antibodies, peptides, or ligands that can recognize and bind to specific receptors or biomarkers overexpressed on the surface of target cells. Furthermore, nanotechnology has played a significant role in enhancing the targeting effect of photosensitizers. Nano-sized carriers, such as liposomes, nanoparticles, and micelles, can encapsulate photosensitizers and provide controlled release and improved cellular uptake. These carriers can also be functionalized with targeting ligands to achieve active targeting to specific cells or tissues [133], [134]. Beyond the natural product photosensitizers discussed in this article, there are numerous others that remain unexplored. The discovery of novel and more effective natural product photosensitizers is an essential objective for future research. Further investigation and development of natural product photosensitizers may lead to significant progress and breakthroughs in the field of PDT.


#

Contributorsʼ Statement

Conception and design of the work: X. Zhou, M. Han; Data collection and analysis and interpretation of the data: X. Zhou, X. Ying, L. Wu, L. Liu, Y. Wang, Y. He; Drafting the manuscript: X. Zhou, X. Ying; Critical revision of the manuscript: X. Zhou, X. Ying, L. Wu, M. Han.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

  • References

  • 1 Kato H. [History of photodynamic therapy–past, present and future]. Gan To Kagaku Ryoho 1996; 23: 8-15
  • 2 Kelly JF, Snell AE, Berenbauai MC. Photodynamic destruction of human bladder carcinoma. Br J Cancer 1975; 31: 237-244
  • 3 Pogue BW, Elliott JT, Kanick SC, Davis SC, Samkoe KS, Maytin EV, Pereira SP, Hasan T. Revisiting photodynamic therapy dosimetry: Reductionist & surrogate approaches to facilitate clinical success. Phys Med Biol 2016; 61: R57-R89
  • 4 Mahmoudi K, Garvey KL, Bouras A, Cramer G, Stepp H, Jesu Raj JG, Bozec D, Busch TM, Hadjipanayis CG. 5-Aminolevulinic acid photodynamic therapy for the treatment of high-grade gliomas. J Neurooncol 2019; 141: 595-607
  • 5 Nakano Y, Kitagawa T, Osada Y, Tanaka T, Nishizawa S, Yamamoto J. 5-Aminolevulinic acid suppresses prostaglandin E2 production by murine macrophages and enhances macrophage cytotoxicity against glioma. World Neurosurg 2019; 127: e669-e676
  • 6 Stepp H, Stummer W. 5‐ALA in the management of malignant glioma. Lasers Surg Med 2018; 50: 399-419
  • 7 Fang CY, Chen PY, Ho DC, Tsai LL, Hsieh PL, Lu MY, Yu CC, Yu CH. miR-145 mediates the anti-cancer stemness effect of photodynamic therapy with 5-aminolevulinic acid (ALA) in oral cancer cells. J Formos Med Assoc 2018; 117: 738-742
  • 8 Wang YY, Chen YK, Hu CS, Xiao LY, Huang WL, Chi TC, Cheng KH, Wang YM, Yuan SF. MAL-PDT inhibits oral precancerous cells and lesions via autophagic cell death. Oral Dis 2019; 25: 758-771
  • 9 Qi F, Sun Y, Lv M, Qin F, Cao W, Bi L. Effects of palmatine hydrochloride mediated photodynamic therapy on oral squamous cell carcinoma. Photochem Photobiol Sci 2018; 17: 375-385
  • 10 Aniogo EC, Plackal Adimuriyil George B, Abrahamse H. The role of photodynamic therapy on multidrug resistant breast cancer. Cancer Cell Int 2019; 19: 91
  • 11 Ahn TG, Jung JM, Lee EJ, Choi JH. Effects of cisplatin on photosensitizer-mediated photodynamic therapy in breast tumor-bearing nude mice. Obstet Gynecol Sci 2019; 62: 112-119
  • 12 Huang HC, Mallidi S, Liu J, Chiang CT, Mai Z, Goldschmidt R, Ebrahim-Zadeh N, Rizvi I, Hasan T. Photodynamic therapy synergizes with irinotecan to overcome compensatory mechanisms and improve treatment outcomes in pancreatic cancer. Cancer Res 2016; 76: 1066-1077
  • 13 DeWitt JM, Sandrasegaran K, OʼNeil B, House MG, Zyromski NJ, Sehdev A, Perkins SM, Flynn J, McCranor L, Shahda S. Phase 1 study of EUS-guided photodynamic therapy for locally advanced pancreatic cancer. Gastrointest Endosc 2019; 89: 390-398
  • 14 Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, Hahn SM, Hamblin MR, Juzeniene A, Kessel D, Korbelik M, Moan J, Mroz P, Nowis D, Piette J, Wilson BC, Golab J. Photodynamic therapy of cancer: An update. CA Cancer J Clin 2011; 61: 250-281
  • 15 Li B, Lin L, Lin H, Wilson BC. Photosensitized singlet oxygen generation and detection: Recent advances and future perspectives in cancer photodynamic therapy. J Biophotonics 2016; 9: 1314-1325
  • 16 Gong H, Chao Y, Xiang J, Han X, Song G, Feng L, Liu J, Yang G, Chen Q, Liu Z. Hyaluronidase to enhance nanoparticle-based photodynamic tumor therapy. Nano Lett 2016; 16: 2512-2521
  • 17 Li L, Tan J, Miao Y, Lei P, Zhang Q. ROS and autophagy: Interactions and molecular regulatory mechanisms. Cell Mol Neurobiol 2015; 35: 615-621
  • 18 Thomas E, Colombeau L, Gries M, Peterlini T, Mathieu C, Thomas N, Boura C, Frochot C, Vanderesse R, Lux F, Barberi-Heyob M, Tillement O. Ultrasmall AGuIX theranostic nanoparticles for vascular-targeted interstitial photodynamic therapy of glioblastoma. Int J Nanomedicine 2017; 12: 7075-7088
  • 19 Rosin FC, Barcessat AR, Borges GG, Correa L. Effect of 5-ALA-mediated photodynamic therapy on mast cell and microvessels densities present in oral premalignant lesions induced in rats. J Photochem Photobiol B 2015; 153: 429-434
  • 20 Beltran Hernandez I, Yu Y, Ossendorp F, Korbelik M, Oliveira S. Preclinical and clinical evidence of immune responses triggered in oncologic photodynamic therapy: clinical recommendations. J Clin Med 2020; 9: 333
  • 21 Kawczyk-Krupka A, Bugaj AM, Latos W, Zaremba K, Wawrzyniec K, Sieron A. Photodynamic therapy in colorectal cancer treatment: The state of the art in clinical trials. Photodiagnosis Photodyn Ther 2015; 12: 545-553
  • 22 Wan MT, Lin JY. Current evidence and applications of photodynamic therapy in dermatology. Clin Cosmet Investig Dermatol 2014; 7: 145-163
  • 23 Li Z, Wang C, Cheng L, Gong H, Yin S, Gong Q, Li Y, Liu Z. PEG-functionalized iron oxide nanoclusters loaded with chlorin e6 for targeted, NIR light induced, photodynamic therapy. Biomaterials 2013; 34: 9160-9170
  • 24 Felsher DW. Cancer revoked: Oncogenes as therapeutic targets. Nat Rev Cancer 2003; 3: 375-380
  • 25 Rajendran M. Quinones as photosensitizer for photodynamic therapy: ROS generation, mechanism and detection methods. Photodiagnosis Photodyn Ther 2016; 13: 175-187
  • 26 Mroz P, Hamblin MR. The immunosuppressive side of PDT. Photochem Photobiol Sci 2011; 10: 751-758
  • 27 Baptista MS, Cadet J, Di Mascio P, Ghogare AA, Greer A, Hamblin MR, Lorente C, Nunez SC, Ribeiro MS, Thomas AH, Vignoni M, Yoshimura TM. Type I and type II photosensitized oxidation reactions: Guidelines and mechanistic pathways. Photochem Photobiol 2017; 93: 912-919
  • 28 Lim CK, Heo J, Shin S, Jeong K, Seo YH, Jang WD, Park CR, Park SY, Kim S, Kwon IC. Nanophotosensitizers toward advanced photodynamic therapy of Cancer. Cancer Lett 2013; 334: 176-187
  • 29 Kwiatkowski S, Knap B, Przystupski D, Saczko J, Kedzierska E, Knap-Czop K, Kotlinska J, Michel O, Kotowski K, Kulbacka J. Photodynamic therapy – mechanisms, photosensitizers and combinations. Biomed Pharmacother 2018; 106: 1098-1107
  • 30 Yan K, Zhang Y, Mu C, Xu Q, Jing X, Wang D, Dang D, Meng L, Ma J. Versatile nanoplatforms with enhanced photodynamic therapy: Designs and applications. Theranostics 2020; 10: 7287-7318
  • 31 Martins TD, Lima E, Boto RE, Ferreira D, Fernandes JR, Almeida P, Ferreira LFV, Silva AM, Reis LV. Red and near-infrared absorbing dicyanomethylenesquaraine cyanine dyes: Photophysicochemical properties and anti-tumor photosensitizing effects. Materials (Basel) 2020; 13: 2083
  • 32 Sun J, Kormakov S, Liu Y, Huang Y, Wu D, Yang Z. Recent progress in metal-based nanoparticles mediated photodynamic therapy. Molecules 2018; 23: 1704
  • 33 Montaseri H, Kruger CA, Abrahamse H. Inorganic nanoparticles applied for active targeted photodynamic therapy of breast cancer. Pharmaceutics 2021; 13: 296
  • 34 Kazantzis KT, Koutsonikoli K, Mavroidi B, Zachariadis M, Alexiou P, Pelecanou M, Politopoulos K, Alexandratou E, Sagnou M. Curcumin derivatives as photosensitizers in photodynamic therapy: photophysical properties and in vitro studies with prostate cancer cells. Photochem Photobiol Sci 2020; 19: 193-206
  • 35 Tsukagoshi S. [Porfimer sodium (Photofrin-II)]. Gan To Kagaku Ryoho 1995; 22: 1271-1278
  • 36 Menezes PFC, Bagnato VS, Sibata CH, Imasato H, Perussi JR. Phototocytotoxicity of photogem submitted to photobleaching. Proc. SPIE 5689, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XIV. https://doi.org/10.1117/12.588794. Date last accessed: February 22; 2024
  • 37 Babbar AK, Singh AK, Goel HC, Chauhan UPS, Sharma RK. Evaluation of Tc-99 m-labeled Photosan-3, a hematoporphyrin derivative, as a potential radiopharmaceutical for tumor scintigraphy. Nucl Med Biol 2000; 27: 419-426
  • 38 Lin H, Shen Y, Chen D, Lin L, Li B, Xie S. Determination of singlet oxygen quantum yield of HiPorfin using Singlet Oxygen Sensor Green. Proc. SPIE 2010, 7845, Optics in Health Care and Biomedical Optics IV. https://doi.org/10.1117/12.870108. Date last accessed: February 22; 2024
  • 39 Sun M, Zhou C, Zeng H, Puebla-Osorio N, Damiani E, Chen J, Wang H, Li G, Yin F, Shan L, Zuo D, Liao Y, Wang Z, Zheng L, Hua Y, Cai Z. Hiporfin-mediated photodynamic therapy in preclinical treatment of osteosarcoma. Photochem Photobiol 2015; 91: 533-544
  • 40 Zeng R, Liu C, Li L, Cai X, Chen R, Li Z. Clinical efficacy of HiPorfin photodynamic therapy for advanced obstructive esophageal cancer. Technol Cancer Res Treat 2020; 19: 1533033820930335
  • 41 Banerjee SM, MacRobert AJ, Mosse CA, Periera B, Bown SG, Keshtgar MRS. Photodynamic therapy: Inception to application in breast cancer. Breast 2017; 31: 105-113
  • 42 Hosokawa S, Takahashi G, Sugiyama KI, Takebayashi S, Okamura J, Takizawa Y, Mineta H. Porfimer sodium-mediated photodynamic therapy in patients with head and neck squamous cell carcinoma. Photodiagnosis Photodyn Ther 2020; 29: 101627
  • 43 Tao J, Chen W, Su Y. Method for treating port wine stains. US Patent 2012029045A1, 2012
  • 44 Jeffes EWB. Levulan: The first approved topical photosensitizer for the treatment of actinic keratosis. J Dermatolog Treat 2002; 13: S19-S23
  • 45 Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F. Reulen HJ. ALA-Glioma Study Group. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol 2006; 7: 392-401
  • 46 Kriegmair M, Baumgartner R, Knuchel R, Stepp H, Hofstadter F, Hostetter A. Detection of early bladder cancer by 5-aminolevulinic acid induced porphyrin fluorescence. J Urol 1996; 155: 105-109
  • 47 Gossner L, Stolte M, Sroka R, Rick K, May A, Hahn EG, Ell C. Photodynamic ablation of high-grade dysplasia and early cancer in Barrettʼs esophagus by means of 5-aminolevulinic acid. Gastroenterology 1998; 114: 448-455
  • 48 Hillemanns P, Untch M, Prove F, Baumgartner R, Hillemanns M, Korell M. Photodynamic therapy of vulvar lichen sclerosus with 5-aminolevulinic acid. Obstet Gynecol 1999; 93: 71-74
  • 49 Morton CA. Methyl aminolevulinate: Actinic keratoses and Bowenʼs disease. Dermatol Clin 2007; 25: 81-87
  • 50 Rhodes LE, de Rie M, Enstrom Y, Groves R, Morken T, Goulden V, Wong GAE, Grob JJ, Varma S, Wolf P. Photodynamic therapy using topical methyl aminolevulinate vs. surgery for nodular basal cell carcinoma: results of a multicenter randomized prospective trial. Arch Dermatol 2004; 140: 17-23
  • 51 Szeimies RM, Karrer S, Radakovic-Fijan S, Tanew A, Calzavara-Pinton PG, Zane C, Sidoroff A, Hempel M, Ulrich J, Proebstle T, Meffert H, Mulder M, Salomon D, Dittmar HC, Bauer JW, Kernland K, Braathen L. Photodynamic therapy using topical methyl 5-aminolevulinate compared with cryotherapy for actinic keratosis: A prospective, randomized study. J Am Acad Dermatol 2002; 47: 258-262
  • 52 Lapini A, Minervini A, Masala A, Schips L, Pycha A, Cindolo L, Giannella R, Martini T, Vittori G, Zani D, Bellomo F, Cunico SC. A comparison of hexaminolevulinate (Hexvix) fluorescence cystoscopy and white-light cystoscopy for detection of bladder cancer: Results of the HeRo observational study. Surg Endosc 2012; 26: 3634-3641
  • 53 Senge MO, Brandt JC. Temoporfin (Foscan, 5, 10, 15, 20-tetra(m-hydroxyphenyl)chlorin)–a second-generation photosensitizer. Photochem Photobiol 2011; 87: 1240-1296
  • 54 Friedberg JS, Mick R, Stevenson J, Metz J, Zhu T, Buyske J, Sterman DH, Pass HI, Glatstein E, Hahn SM. A phase I study of Foscan-mediated photodynamic therapy and surgery in patients with mesothelioma. Ann Thorac Surg 2003; 75: 952-959
  • 55 Istomin YP, Lapzevich TP, Chalau VN, Shliakhtsin SV, Trukhachova TV. Photodynamic therapy of cervical intraepithelial neoplasia grades II and III with Photolon. Photodiagnosis Photodyn Ther 2010; 7: 144-151
  • 56 Wang S, Bromley E, Xu L, Chen JC, Keltner L. Talaporfin sodium. Expert Opin Pharmacother 2010; 11: 133-140
  • 57 Akter S, Saito S, Inai M, Honda N, Hazama H, Nishikawa T, Kaneda Y, Awazu K. Efficient photodynamic therapy against drug-resistant prostate cancer using replication-deficient virus particles and talaporfin sodium. Laser Med Sci 2021; 36: 743-750
  • 58 Nonaka T, Nanashima A, Nonaka M, Uehara M, Isomoto H, Nonaka Y, Nagayasu T. Advantages of laserphyrin compared with photofrin in photodynamic therapy for bile duct carcinoma. J Hepatobiliary Pancreat Sci 2011; 18: 592-600
  • 59 Nakamura T, Oinuma T. Usefulness of photodynamic diagnosis and therapy using talaporfin sodium for an advanced-aged patient with inoperable gastric cancer (a secondary publication). Laser Ther 2014; 23: 201-210
  • 60 Ikeda H, Ohba S, Egashira K, Asahina I. The effect of photodynamic therapy with talaporfin sodium, a second-generation photosensitizer, on oral squamous cell carcinoma: A series of eight cases. Photodiagnosis Photodyn Ther 2018; 21: 176-180
  • 61 Iacono P, Da Pozzo S, Varano M, Parravano M. Photodynamic therapy with verteporfin for chronic central serous chorioretinopathy: A review of data and efficacy. Pharmaceuticals (Basel) 2020; 13: 349
  • 62 van Dijk EHC, van Rijssen TJ, Subhi Y, Boon CJF. Photodynamic therapy for chorioretinal diseases: A practical approach. Ophthalmol Ther 2020; 9: 329-342
  • 63 Mae Y, Kanda T, Sugihara T, Takata T, Kinoshita H, Sakaguchi T, Hasegawa T, Tarumoto R, Edano M, Kurumi H, Ikebuchi Y, Kawaguchi K, Isomoto H. Verteporfin-photodynamic therapy is effective on gastric cancer cells. Mol Clin Oncol 2020; 13: 10
  • 64 No authors Photodynamic therapy of subfoveal choroidal neovascularization in age-related macular degeneration with verteporfin – one-year results of 2 randomized clinical trials – TAP report. Treatment of age-related macular degeneration with photodynamic therapy (TAP) Study Group. Arch Ophthalmol 1999; 117: 1329-1345
  • 65 Shevchik SA, Loshchenov MV, Meerovich GA, Budzinskaia MV, Ermakova NA, Kharnas SS, Loshchenov VB. [A device for fluorescence diagnosis and photodynamic therapy of eye diseases, by using photosense]. Vestn Oftalmol 2005; 121: 26-28
  • 66 Vakulovskaya E, Kemov Y, Zalevsky I, Reshetnikov A, Umnova L, Vorozhcsov G. Photodynamic therapy and fluorescent diagnostics of skin cancer with radochlorine and photosense: comparing efficacy and toxicity. Proc. SPIE 2004, 5315, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XIII. https://doi.org/10.1117/12.537787. Date last accessed: February 22; 2024
  • 67 Vakulovskaya EG, Shental VV, Kondratjeva TT. Photodynamic therapy and fluorescent diagnostics of head and neck tumors with photosense. Int J Cancer 2002; 262-262
  • 68 Vakoulovskaya E, Shental V, Oumnova L, Vorozhcsov G. Photodynamic therapy of breast cancer with photosense. Proc. SPIE 2003, 4952, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XII. https://doi.org/10.1117/12.479431. Date last accessed: February 22; 2024
  • 69 Urbanska K, Romanowska-Dixon B, Matuszak Z, Oszajca J, Nowak-Sliwinska P, Stochel G. Indocyanine green as a prospective sensitizer for photodynamic therapy of melanomas. Acta Biochim Pol 2002; 49: 387-391
  • 70 Parker S. The use of diffuse laser photonic energy and indocyanine green photosensitiser as an adjunct to periodontal therapy. Br Dent J 2013; 215: 167-171
  • 71 Costa RA, Farah ME, Cardillo JA, Belfort R. Photodynamic therapy with indocyanine green for occult subfoveal choroidal neovascularization caused by age-related macular degeneration. Curr Eye Res 2001; 23: 271-275
  • 72 Tardivo JP, Del Giglio A, de Oliveira CS, Gabrielli DS, Junqueira HC, Tada DB, Severino D, Turchiello RDF, Baptista MS. Methylene blue in photodynamic therapy: From basic mechanisms to clinical applications. Photodiagnosis Photodyn Ther 2005; 2: 175-191
  • 73 Cengel KA, Simone 2nd CB, Glatstein E. PDT: Whatʼs past is prologue. Cancer Res 2016; 76: 2497-2499
  • 74 Dougherty TJ, Kauffman JE, Goldfarb A, Weishaupt KR, Boyle D, Mittleman A. Photoradiation therapy for the treatment of malignant tumors. Cancer Res 1978; 38: 2628-2635
  • 75 Diamond I, McDonagh A, Wilson C, Granelli S, Nielsen S, Jaenicke R. Photodynamic therapy of malignant tumours. The Lancet 1972; 300: 1175-1177
  • 76 Berns MW, Rettenmaier M, McCoullough J, Coffey J, Wile A, Berman M, Disaia P, Weinstein G. Response of psoriasis to red laser light (630 nm) following systemic injection of hematoporphyrin derivative. Lasers Surg Med 1984; 4: 73-77
  • 77 Adimoolam MG, A V. Nalam MR, Sunkara MV. Chlorin e6 loaded lactoferrin nanoparticles for enhanced photodynamic therapy. J Mater Chem B 2017; 5: 9189-9196
  • 78 Zhu YX, Jia HR, Chen Z, Wu FG. Photosensitizer (PS)/Polyhedral Oligomeric Silsesquioxane (POSS)-crosslinked nanohybrids for enhanced imaging-guided photodynamic cancer therapy. Nanoscale 2017; 9: 12874-12884
  • 79 OʼConnor AE, Gallagher WM, Byrne AT. Porphyrin and nonporphyrin photosensitizers in oncology: Preclinical and clinical advances in photodynamic therapy. Photochem Photobiol 2009; 85: 1053-1074
  • 80 Plonka J, Latocha M, Kusmierz D, Zielinska A. Expression of proapoptotic BAX and TP53 genes and antiapoptotic BCL-2 gene in MCF-7 and T-47D tumour cell cultures of the mammary gland after a photodynamic therapy with photolon. Adv Clin Exp Med 2015; 24: 37-46
  • 81 Copley L, van der Watt P, Wirtz KW, Parker MI, Leaner VD. Photolon, a chlorin e6 derivative, triggers ROS production and light-dependent cell death via necrosis. Int J Biochem Cell Biol 2008; 40: 227-235
  • 82 Chouikrat R, Seve A, Vanderesse R, Benachour H, Barberi-Heyob M, Richeter S, Raehm L, Durand JO, Verelst M, Frochot C. Non polymeric nanoparticles for photodynamic therapy applications: Recent developments. Curr Med Chem 2012; 19: 781-792
  • 83 Li X, Lee S, Yoon J. Supramolecular photosensitizers rejuvenate photodynamic therapy. Chem Soc Rev 2018; 47: 1174-1188
  • 84 Josefsen LB, Boyle RW. Unique diagnostic and therapeutic roles of porphyrins and phthalocyanines in photodynamic therapy, imaging and theranostics. Theranostics 2012; 2: 916-966
  • 85 Abrahamse H, Hamblin MR. New photosensitizers for photodynamic therapy. Biochem J 2016; 473: 347-364
  • 86 Xiao QC, Wu J, Pang X, Jiang Y, Wang P, Leung AW, Gao LQ, Jiang S, Xu CS. Discovery and development of natural products and their derivatives as photosensitizers for photodynamic therapy. Curr Med Chem 2018; 25: 839-860
  • 87 Zhao JB, Cui Q, Wang LG, Wu SH, Yang YC. Irradiation sensitivity-enhancing effect of psoralens on S180 cell line. J Fourth Mil Med Univ 1998; 19: 627-629
  • 88 Carneiro Leite V, Ferreira Santos R, Chen Chen L, Andreu Guillo L. Psoralen derivatives and longwave ultraviolet irradiation are active in vitro against human melanoma cell line. J Photochem Photobiol B 2004; 76: 49-53
  • 89 Plumas J, Drillat P, Jacob MC, Richard MJ, Favrot MC. [Extracorporeal photochemotherapy for treatment of clonal T cell proliferations]. Bull Cancer 2003; 90: 763-770
  • 90 Efferth T, Fabry U, Osieka R. Induction of apoptosis, depletion of glutathione, and DNA damage by extracorporeal photochemotherapy and psoralen with exposure to UV light in vitro . Anticancer Res 2001; 21: 2777-2783
  • 91 Yan XG, Zhou XY, Wu JZ. Study on the photosensitizing properties of the antitumor effect of psoralens. J Int Oncol 1999; 26: 83-85
  • 92 Zheng Y, Yin G, Le V, Zhang A, Chen S, Liang X, Liu J. Photodynamic-therapy activates immune response by disrupting immunity homeostasis of tumor cells, which generates vaccine for cancer therapy. Int J Biol Sci 2016; 12: 120-132
  • 93 Garg AD, Vandenberk L, Koks C, Verschuere T, Boon L, Van Gool SW, Agostinis P. Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell-driven rejection of high-grade glioma. Sci Transl Med 2016; 8: 328ra327
  • 94 Majerník M, Jendželovský R, Babinčák M, Košuth J, Ševc J, Tonelli Gombalová Z, Jendželovská Z, Buríková M, Fedoročko P. Novel insights into the effect of hyperforin and photodynamic therapy with hypericin on chosen angiogenic factors in colorectal micro-tumors created on chorioallantoic membrane. Int J Mol Sci 2019; 20: 3004
  • 95 Kim H, Kim SW, Seok KH, Hwang CW, Ahn JC, Jin JO, Kang HW. Hypericin-assisted photodynamic therapy against anaplastic thyroid cancer. Photodiagnosis Photodyn Ther 2018; 24: 15-21
  • 96 Plenagl N, Duse L, Seitz BS, Goergen N, Pinnapireddy SR, Jedelska J, Brüßler J, Bakowsky U. Photodynamic therapy – hypericin tetraether liposome conjugates and their antitumor and antiangiogenic activity. Drug Deliv 2019; 26: 23-33
  • 97 Chen XM, Li HY, Qu T, Gao M, Xu XH. Inhibitory effects of CIK on breast cancer cells intensified by hypericin mediated photodynamic therapy. Herald Med 2021; 40: 1318-1324
  • 98 Hu J, Song J, Tang Z, Wei S, Chen L, Zhou R. Hypericin-mediated photodynamic therapy inhibits growth of colorectal cancer cells via inducing S phase cell cycle arrest and apoptosis. Eur J Pharmacol 2021; 900: 174071
  • 99 Zhang XQ, Fang JY, Yao J, Chen MY, Song ZW, Xu LL, Zhao TJ. Mechanism of photodynamic therapy mediated by hypericin in inhibiting the proliferation of leukemia cell lines. Chin Pharm J 2018; 53: 967-974
  • 100 Chen HJ, Diao L, Zhang L, Liu HX, Li WQ, Huang XD, Zhang RL, Li GX. Study on antiviral effect of hypericin against infectious bronchitis virus in vitro . Chin Vet Sci 2018; 48: 1415-1422
  • 101 Kamuhabwa AAR, Huygens A, De Witte PAM. Photodynamic therapy of transitional cell carcinoma multicellular tumor spheroids with hypericin. Int J Oncol 2003; 23: 1445-1450
  • 102 Xu CS, Ling RN. Study of hypericin-mediated photodynamic therapy on human nasopharyngeal carcinoma cells. Laser Technology 2005; 29: 395-397
  • 103 Wan XY, Chen YT. A new photochemotherapy drug - Hypocrellin A. Sci Bull 1980; 24: 1148-1149
  • 104 Kishi T, Tahara S, Taniguchi N, Tsuda M, Tanaka C, Takahashi S. New perylenequinones from Shiraia bambusicola . Planta Med 1991; 57: 376-379
  • 105 Hu MM, Cai YJ, Liao XR, Hao ZK, Liu JY. Development of an HPLC method to analyze and prepare elsinochrome C and hypocrellin A in the submerged fermentation broth of Shiria sp SUPER-H168. Biomed Chromatogr 2012; 26: 737-742
  • 106 Diwu ZJ, Haugland RP, Liu J, Lown JW, Miller GG, Moore RB, Brown K, Tulip J, McPhee MS. Photosensitization by anticancer agents 21: New perylene- and aminonaphthoquinones. Free Radic Biol Med 1996; 20: 589-593
  • 107 Deininger MH, Weinschenk T, Morgalla MH, Meyermann R, Schluesener HJ. Release of regulators of angiogenesis following Hypocrellin-A and -B photodynamic therapy of human brain tumor cells. Biochem Biophys Res Commun 2002; 298: 520-530
  • 108 Chen J, Teng LR, Zheng KY, Li TJ, Ma L, Li C, Wu W, Fei XF. Study on the apoptotic molecule mechanism in A375-S2 cell induced by hypocrellin A. Chin Pharm J 2005; 40: 431-434
  • 109 Zhou JH, Xia SQ, Chen JR, Wang XS, Zhang BW, Zhang HJ, Zou P, Ai XC, Zhang JP. Surface binding and improved photodamage of the lanthanum ion complex of hypocrellin A to calf thymus DNA. J Photochem Photobiol A Chem 2004; 165: 143-147
  • 110 Zhao X. Key factors of photoactivated pesticide hypocrellin A against Botrytis cinere [dissertation]. Hangzhou: Zhejiang A&F University; 2015
  • 111 Mastrangelopoulou M, Grigalavicius M, Berg K, Menard M, Theodossiou TA. Cytotoxic and photocytotoxic effects of cercosporin on human tumor cell lines. Photochem Photobiol 2019; 95: 387-396
  • 112 Liu SS, Yang YH, Wang ZS. [Clinical study of CPD4-PDT for the prevention of postoperative recurrence in infiltrative bladder cancer]. Chin J Integr Med 1998; 18: 15-17
  • 113 Cao G, Guo T, Lu CT. Effect of CPD4 photodynamic therapy on endothelial cell in vitro . J Postgrad Med 2004; 17: 114-116
  • 114 Zhang JL, Chen P, Ding K, Dai SG, Wang K, Cao DL, Lin L, Tang GQ. Photodynamic effect of two kinds of CPD photosensitizers on sarcoma S180 transplanted in mice. Tianjin Med J 2006; 34: 705-707
  • 115 Yu HY. Silencing XBP1 expression enhances the sensitivity of human osteosarcoma HOS cells to MPPα-PDT [dissertation]. Chongqing: ChongQing Medical University; 2020
  • 116 Zhong SX. Inhibition of PERK pathway enhances sensitivity of human osteosarcoma HOS cells induced to pyropheophorbide-a methyl ester-mediated photodynamic therapy [dissertation]. Chongqing: ChongQing Medical University; 2019
  • 117 Wang L. Pyropheophorbide a photodynamic therapy in the control of golden hamster sebaceous patches [dissertation]. Nanjing: Nanjing University of Chinese Medicine; 2016
  • 118 Zeng XB. Inhibitive action of curcumin on human breast cancer and observation of the reinforcement effect after curcumin exposure to light [dissertation]. Chongqing: ChongQing Medical University; 2008
  • 119 Mou TL, Liu HM, Pan YS, He GF, Li ZW, Guo J, Pang DC, Zhai Z, Wu ZX. Study on the expression of tumor necrosis factor-α and cysteine protease-8 in cervical cancer xenografts and tumor tissues treated with multidose photodynamic therapy combined with curcumin. Global Tradit Chin Med 2022; 15: 543-549
  • 120 He JA, Hu YZ, Jiang LJ. Photodynamic action of phycobiliproteins: In situ generation of reactive oxygen species. Biochim Biophys Acta Bioenerg 1997; 1320: 165-174
  • 121 Nelson KM, Dahlin JL, Bisson J, Graham J, Pauli GF, Walters MA. The essential medicinal chemistry of curcumin. J Med Chem 2017; 60: 1620-1637
  • 122 Rao J, Xie J, Zhao JQ, Zhu T. Rhein-photodynamic sensitization generates free radicals and singlet oxygen. Sci Sin Chim 2004; 34: 211-217
  • 123 Hamblin MR, Abrahamse H. Tetracyclines: Light-activated antibiotics?. Future Med Chem 2019; 11: 2427-2445
  • 124 Greco G, Ulfo L, Turrini E, Marconi A, Costantini PE, Marforio TD, Mattioli EJ, Di Giosia M, Danielli A, Fimognari C, Calvaresi M. Light-enhanced cytotoxicity of doxorubicin by photoactivation. Cells 2023; 12: 392
  • 125 Chen GQ, Guo LN, Wu QZ. A study of the photosensitizing effect of an extraction from a medicinal herb belongs to leguminosales. Med J Qilu 2001; 16: 279-280
  • 126 Yao CS, Wu QZ. Photosensitizing effect of two extracts from caulis lonicerae: A preliminary study. Chin J Laser Med Surg 2006; 15: 361-364
  • 127 Liao J, Li PP, Wu CJ. Screening new photosensitizers from Chinese medicinal herbs and searching for herbal photodynamic killing effects on human stomach cancer cells. Chin J Integr Med 1997; 17: 726-729
  • 128 Scotti F, Mou L, Huang C, Booker A, Weckerle C, Maake C, Heinrich M. Treating chronic wounds using photoactive metabolites: Data mining the Chinese pharmacopoeia for potential lead species. Planta Med 2021; 87: 1206-1218
  • 129 Shi YG, Lin S, Chen WX, Jiang L, Gu Q, Li DH, Chen YW. Dual-stage blue-light-guided membrane and DNA-targeted photodynamic inactivation using octyl gallate for ultraefficient eradication of planktonic bacteria and sessile biofilms. J Agric Food Chem 2022; 70: 7547-7565
  • 130 Shi YG, Chen WX, Zheng MZ, Zhao YX, Wang YR, Chu YH, Du ST, Shi ZY, Gu Q, Chen JS. Ultraefficient OG-mediated photodynamic inactivation mechanism for ablation of bacteria and biofilms in water augmented by potassium iodide under blue light irradiation. J Agric Food Chem 2023; 71: 13672-13687
  • 131 Zheng MZ, Chen WX, Zhao YX, Fang Q, Wang LG, Tian SY, Shi YG, Chen JS. Ascorbic acid potentiates photodynamic inactivation mediated by octyl gallate and blue light for rapid eradication of planktonic bacteria and biofilms. Date last accessed: February 22 2024 SSRN: https://ssrn.com/abstract=4517458 DOI: 10.2139/ssrn.4517458
  • 132 Krieger-Liszkay A. Singlet oxygen production in photosynthesis. J Exp Bot 2004; 56: 337-346
  • 133 Gierlich P, Mata AI, Donohoe C, Brito RMM, Senge MO, Gomes-da-Silva LC. Ligand-targeted delivery of photosensitizers for cancer treatment. Molecules 2020; 25: 5317
  • 134 Escudero A, Carrillo-Carrión C, Castillejos MC, Romero-Ben E, Rosales-Barrios C, Khiar N. Photodynamic therapy: Photosensitizers and nanostructures. Mater Chem Front 2021; 5: 3788-3812

Correspondence

Min Han, PhD
Institute of Pharmaceutics
Zijingang Campus of Zhejiang University
Yuhangtang Road No. 388
Hangzhou, 310030, Zhejiang Province
China   
Telefon: + 8 65 71 88 20 84 36   
Fax: + 8 65 71 88 20 84 36   

Publikationsverlauf

Eingereicht: 27. April 2023

Angenommen nach Revision: 15. Januar 2024

Artikel online veröffentlicht:
29. Februar 2024

© 2024. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Kato H. [History of photodynamic therapy–past, present and future]. Gan To Kagaku Ryoho 1996; 23: 8-15
  • 2 Kelly JF, Snell AE, Berenbauai MC. Photodynamic destruction of human bladder carcinoma. Br J Cancer 1975; 31: 237-244
  • 3 Pogue BW, Elliott JT, Kanick SC, Davis SC, Samkoe KS, Maytin EV, Pereira SP, Hasan T. Revisiting photodynamic therapy dosimetry: Reductionist & surrogate approaches to facilitate clinical success. Phys Med Biol 2016; 61: R57-R89
  • 4 Mahmoudi K, Garvey KL, Bouras A, Cramer G, Stepp H, Jesu Raj JG, Bozec D, Busch TM, Hadjipanayis CG. 5-Aminolevulinic acid photodynamic therapy for the treatment of high-grade gliomas. J Neurooncol 2019; 141: 595-607
  • 5 Nakano Y, Kitagawa T, Osada Y, Tanaka T, Nishizawa S, Yamamoto J. 5-Aminolevulinic acid suppresses prostaglandin E2 production by murine macrophages and enhances macrophage cytotoxicity against glioma. World Neurosurg 2019; 127: e669-e676
  • 6 Stepp H, Stummer W. 5‐ALA in the management of malignant glioma. Lasers Surg Med 2018; 50: 399-419
  • 7 Fang CY, Chen PY, Ho DC, Tsai LL, Hsieh PL, Lu MY, Yu CC, Yu CH. miR-145 mediates the anti-cancer stemness effect of photodynamic therapy with 5-aminolevulinic acid (ALA) in oral cancer cells. J Formos Med Assoc 2018; 117: 738-742
  • 8 Wang YY, Chen YK, Hu CS, Xiao LY, Huang WL, Chi TC, Cheng KH, Wang YM, Yuan SF. MAL-PDT inhibits oral precancerous cells and lesions via autophagic cell death. Oral Dis 2019; 25: 758-771
  • 9 Qi F, Sun Y, Lv M, Qin F, Cao W, Bi L. Effects of palmatine hydrochloride mediated photodynamic therapy on oral squamous cell carcinoma. Photochem Photobiol Sci 2018; 17: 375-385
  • 10 Aniogo EC, Plackal Adimuriyil George B, Abrahamse H. The role of photodynamic therapy on multidrug resistant breast cancer. Cancer Cell Int 2019; 19: 91
  • 11 Ahn TG, Jung JM, Lee EJ, Choi JH. Effects of cisplatin on photosensitizer-mediated photodynamic therapy in breast tumor-bearing nude mice. Obstet Gynecol Sci 2019; 62: 112-119
  • 12 Huang HC, Mallidi S, Liu J, Chiang CT, Mai Z, Goldschmidt R, Ebrahim-Zadeh N, Rizvi I, Hasan T. Photodynamic therapy synergizes with irinotecan to overcome compensatory mechanisms and improve treatment outcomes in pancreatic cancer. Cancer Res 2016; 76: 1066-1077
  • 13 DeWitt JM, Sandrasegaran K, OʼNeil B, House MG, Zyromski NJ, Sehdev A, Perkins SM, Flynn J, McCranor L, Shahda S. Phase 1 study of EUS-guided photodynamic therapy for locally advanced pancreatic cancer. Gastrointest Endosc 2019; 89: 390-398
  • 14 Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, Hahn SM, Hamblin MR, Juzeniene A, Kessel D, Korbelik M, Moan J, Mroz P, Nowis D, Piette J, Wilson BC, Golab J. Photodynamic therapy of cancer: An update. CA Cancer J Clin 2011; 61: 250-281
  • 15 Li B, Lin L, Lin H, Wilson BC. Photosensitized singlet oxygen generation and detection: Recent advances and future perspectives in cancer photodynamic therapy. J Biophotonics 2016; 9: 1314-1325
  • 16 Gong H, Chao Y, Xiang J, Han X, Song G, Feng L, Liu J, Yang G, Chen Q, Liu Z. Hyaluronidase to enhance nanoparticle-based photodynamic tumor therapy. Nano Lett 2016; 16: 2512-2521
  • 17 Li L, Tan J, Miao Y, Lei P, Zhang Q. ROS and autophagy: Interactions and molecular regulatory mechanisms. Cell Mol Neurobiol 2015; 35: 615-621
  • 18 Thomas E, Colombeau L, Gries M, Peterlini T, Mathieu C, Thomas N, Boura C, Frochot C, Vanderesse R, Lux F, Barberi-Heyob M, Tillement O. Ultrasmall AGuIX theranostic nanoparticles for vascular-targeted interstitial photodynamic therapy of glioblastoma. Int J Nanomedicine 2017; 12: 7075-7088
  • 19 Rosin FC, Barcessat AR, Borges GG, Correa L. Effect of 5-ALA-mediated photodynamic therapy on mast cell and microvessels densities present in oral premalignant lesions induced in rats. J Photochem Photobiol B 2015; 153: 429-434
  • 20 Beltran Hernandez I, Yu Y, Ossendorp F, Korbelik M, Oliveira S. Preclinical and clinical evidence of immune responses triggered in oncologic photodynamic therapy: clinical recommendations. J Clin Med 2020; 9: 333
  • 21 Kawczyk-Krupka A, Bugaj AM, Latos W, Zaremba K, Wawrzyniec K, Sieron A. Photodynamic therapy in colorectal cancer treatment: The state of the art in clinical trials. Photodiagnosis Photodyn Ther 2015; 12: 545-553
  • 22 Wan MT, Lin JY. Current evidence and applications of photodynamic therapy in dermatology. Clin Cosmet Investig Dermatol 2014; 7: 145-163
  • 23 Li Z, Wang C, Cheng L, Gong H, Yin S, Gong Q, Li Y, Liu Z. PEG-functionalized iron oxide nanoclusters loaded with chlorin e6 for targeted, NIR light induced, photodynamic therapy. Biomaterials 2013; 34: 9160-9170
  • 24 Felsher DW. Cancer revoked: Oncogenes as therapeutic targets. Nat Rev Cancer 2003; 3: 375-380
  • 25 Rajendran M. Quinones as photosensitizer for photodynamic therapy: ROS generation, mechanism and detection methods. Photodiagnosis Photodyn Ther 2016; 13: 175-187
  • 26 Mroz P, Hamblin MR. The immunosuppressive side of PDT. Photochem Photobiol Sci 2011; 10: 751-758
  • 27 Baptista MS, Cadet J, Di Mascio P, Ghogare AA, Greer A, Hamblin MR, Lorente C, Nunez SC, Ribeiro MS, Thomas AH, Vignoni M, Yoshimura TM. Type I and type II photosensitized oxidation reactions: Guidelines and mechanistic pathways. Photochem Photobiol 2017; 93: 912-919
  • 28 Lim CK, Heo J, Shin S, Jeong K, Seo YH, Jang WD, Park CR, Park SY, Kim S, Kwon IC. Nanophotosensitizers toward advanced photodynamic therapy of Cancer. Cancer Lett 2013; 334: 176-187
  • 29 Kwiatkowski S, Knap B, Przystupski D, Saczko J, Kedzierska E, Knap-Czop K, Kotlinska J, Michel O, Kotowski K, Kulbacka J. Photodynamic therapy – mechanisms, photosensitizers and combinations. Biomed Pharmacother 2018; 106: 1098-1107
  • 30 Yan K, Zhang Y, Mu C, Xu Q, Jing X, Wang D, Dang D, Meng L, Ma J. Versatile nanoplatforms with enhanced photodynamic therapy: Designs and applications. Theranostics 2020; 10: 7287-7318
  • 31 Martins TD, Lima E, Boto RE, Ferreira D, Fernandes JR, Almeida P, Ferreira LFV, Silva AM, Reis LV. Red and near-infrared absorbing dicyanomethylenesquaraine cyanine dyes: Photophysicochemical properties and anti-tumor photosensitizing effects. Materials (Basel) 2020; 13: 2083
  • 32 Sun J, Kormakov S, Liu Y, Huang Y, Wu D, Yang Z. Recent progress in metal-based nanoparticles mediated photodynamic therapy. Molecules 2018; 23: 1704
  • 33 Montaseri H, Kruger CA, Abrahamse H. Inorganic nanoparticles applied for active targeted photodynamic therapy of breast cancer. Pharmaceutics 2021; 13: 296
  • 34 Kazantzis KT, Koutsonikoli K, Mavroidi B, Zachariadis M, Alexiou P, Pelecanou M, Politopoulos K, Alexandratou E, Sagnou M. Curcumin derivatives as photosensitizers in photodynamic therapy: photophysical properties and in vitro studies with prostate cancer cells. Photochem Photobiol Sci 2020; 19: 193-206
  • 35 Tsukagoshi S. [Porfimer sodium (Photofrin-II)]. Gan To Kagaku Ryoho 1995; 22: 1271-1278
  • 36 Menezes PFC, Bagnato VS, Sibata CH, Imasato H, Perussi JR. Phototocytotoxicity of photogem submitted to photobleaching. Proc. SPIE 5689, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XIV. https://doi.org/10.1117/12.588794. Date last accessed: February 22; 2024
  • 37 Babbar AK, Singh AK, Goel HC, Chauhan UPS, Sharma RK. Evaluation of Tc-99 m-labeled Photosan-3, a hematoporphyrin derivative, as a potential radiopharmaceutical for tumor scintigraphy. Nucl Med Biol 2000; 27: 419-426
  • 38 Lin H, Shen Y, Chen D, Lin L, Li B, Xie S. Determination of singlet oxygen quantum yield of HiPorfin using Singlet Oxygen Sensor Green. Proc. SPIE 2010, 7845, Optics in Health Care and Biomedical Optics IV. https://doi.org/10.1117/12.870108. Date last accessed: February 22; 2024
  • 39 Sun M, Zhou C, Zeng H, Puebla-Osorio N, Damiani E, Chen J, Wang H, Li G, Yin F, Shan L, Zuo D, Liao Y, Wang Z, Zheng L, Hua Y, Cai Z. Hiporfin-mediated photodynamic therapy in preclinical treatment of osteosarcoma. Photochem Photobiol 2015; 91: 533-544
  • 40 Zeng R, Liu C, Li L, Cai X, Chen R, Li Z. Clinical efficacy of HiPorfin photodynamic therapy for advanced obstructive esophageal cancer. Technol Cancer Res Treat 2020; 19: 1533033820930335
  • 41 Banerjee SM, MacRobert AJ, Mosse CA, Periera B, Bown SG, Keshtgar MRS. Photodynamic therapy: Inception to application in breast cancer. Breast 2017; 31: 105-113
  • 42 Hosokawa S, Takahashi G, Sugiyama KI, Takebayashi S, Okamura J, Takizawa Y, Mineta H. Porfimer sodium-mediated photodynamic therapy in patients with head and neck squamous cell carcinoma. Photodiagnosis Photodyn Ther 2020; 29: 101627
  • 43 Tao J, Chen W, Su Y. Method for treating port wine stains. US Patent 2012029045A1, 2012
  • 44 Jeffes EWB. Levulan: The first approved topical photosensitizer for the treatment of actinic keratosis. J Dermatolog Treat 2002; 13: S19-S23
  • 45 Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F. Reulen HJ. ALA-Glioma Study Group. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol 2006; 7: 392-401
  • 46 Kriegmair M, Baumgartner R, Knuchel R, Stepp H, Hofstadter F, Hostetter A. Detection of early bladder cancer by 5-aminolevulinic acid induced porphyrin fluorescence. J Urol 1996; 155: 105-109
  • 47 Gossner L, Stolte M, Sroka R, Rick K, May A, Hahn EG, Ell C. Photodynamic ablation of high-grade dysplasia and early cancer in Barrettʼs esophagus by means of 5-aminolevulinic acid. Gastroenterology 1998; 114: 448-455
  • 48 Hillemanns P, Untch M, Prove F, Baumgartner R, Hillemanns M, Korell M. Photodynamic therapy of vulvar lichen sclerosus with 5-aminolevulinic acid. Obstet Gynecol 1999; 93: 71-74
  • 49 Morton CA. Methyl aminolevulinate: Actinic keratoses and Bowenʼs disease. Dermatol Clin 2007; 25: 81-87
  • 50 Rhodes LE, de Rie M, Enstrom Y, Groves R, Morken T, Goulden V, Wong GAE, Grob JJ, Varma S, Wolf P. Photodynamic therapy using topical methyl aminolevulinate vs. surgery for nodular basal cell carcinoma: results of a multicenter randomized prospective trial. Arch Dermatol 2004; 140: 17-23
  • 51 Szeimies RM, Karrer S, Radakovic-Fijan S, Tanew A, Calzavara-Pinton PG, Zane C, Sidoroff A, Hempel M, Ulrich J, Proebstle T, Meffert H, Mulder M, Salomon D, Dittmar HC, Bauer JW, Kernland K, Braathen L. Photodynamic therapy using topical methyl 5-aminolevulinate compared with cryotherapy for actinic keratosis: A prospective, randomized study. J Am Acad Dermatol 2002; 47: 258-262
  • 52 Lapini A, Minervini A, Masala A, Schips L, Pycha A, Cindolo L, Giannella R, Martini T, Vittori G, Zani D, Bellomo F, Cunico SC. A comparison of hexaminolevulinate (Hexvix) fluorescence cystoscopy and white-light cystoscopy for detection of bladder cancer: Results of the HeRo observational study. Surg Endosc 2012; 26: 3634-3641
  • 53 Senge MO, Brandt JC. Temoporfin (Foscan, 5, 10, 15, 20-tetra(m-hydroxyphenyl)chlorin)–a second-generation photosensitizer. Photochem Photobiol 2011; 87: 1240-1296
  • 54 Friedberg JS, Mick R, Stevenson J, Metz J, Zhu T, Buyske J, Sterman DH, Pass HI, Glatstein E, Hahn SM. A phase I study of Foscan-mediated photodynamic therapy and surgery in patients with mesothelioma. Ann Thorac Surg 2003; 75: 952-959
  • 55 Istomin YP, Lapzevich TP, Chalau VN, Shliakhtsin SV, Trukhachova TV. Photodynamic therapy of cervical intraepithelial neoplasia grades II and III with Photolon. Photodiagnosis Photodyn Ther 2010; 7: 144-151
  • 56 Wang S, Bromley E, Xu L, Chen JC, Keltner L. Talaporfin sodium. Expert Opin Pharmacother 2010; 11: 133-140
  • 57 Akter S, Saito S, Inai M, Honda N, Hazama H, Nishikawa T, Kaneda Y, Awazu K. Efficient photodynamic therapy against drug-resistant prostate cancer using replication-deficient virus particles and talaporfin sodium. Laser Med Sci 2021; 36: 743-750
  • 58 Nonaka T, Nanashima A, Nonaka M, Uehara M, Isomoto H, Nonaka Y, Nagayasu T. Advantages of laserphyrin compared with photofrin in photodynamic therapy for bile duct carcinoma. J Hepatobiliary Pancreat Sci 2011; 18: 592-600
  • 59 Nakamura T, Oinuma T. Usefulness of photodynamic diagnosis and therapy using talaporfin sodium for an advanced-aged patient with inoperable gastric cancer (a secondary publication). Laser Ther 2014; 23: 201-210
  • 60 Ikeda H, Ohba S, Egashira K, Asahina I. The effect of photodynamic therapy with talaporfin sodium, a second-generation photosensitizer, on oral squamous cell carcinoma: A series of eight cases. Photodiagnosis Photodyn Ther 2018; 21: 176-180
  • 61 Iacono P, Da Pozzo S, Varano M, Parravano M. Photodynamic therapy with verteporfin for chronic central serous chorioretinopathy: A review of data and efficacy. Pharmaceuticals (Basel) 2020; 13: 349
  • 62 van Dijk EHC, van Rijssen TJ, Subhi Y, Boon CJF. Photodynamic therapy for chorioretinal diseases: A practical approach. Ophthalmol Ther 2020; 9: 329-342
  • 63 Mae Y, Kanda T, Sugihara T, Takata T, Kinoshita H, Sakaguchi T, Hasegawa T, Tarumoto R, Edano M, Kurumi H, Ikebuchi Y, Kawaguchi K, Isomoto H. Verteporfin-photodynamic therapy is effective on gastric cancer cells. Mol Clin Oncol 2020; 13: 10
  • 64 No authors Photodynamic therapy of subfoveal choroidal neovascularization in age-related macular degeneration with verteporfin – one-year results of 2 randomized clinical trials – TAP report. Treatment of age-related macular degeneration with photodynamic therapy (TAP) Study Group. Arch Ophthalmol 1999; 117: 1329-1345
  • 65 Shevchik SA, Loshchenov MV, Meerovich GA, Budzinskaia MV, Ermakova NA, Kharnas SS, Loshchenov VB. [A device for fluorescence diagnosis and photodynamic therapy of eye diseases, by using photosense]. Vestn Oftalmol 2005; 121: 26-28
  • 66 Vakulovskaya E, Kemov Y, Zalevsky I, Reshetnikov A, Umnova L, Vorozhcsov G. Photodynamic therapy and fluorescent diagnostics of skin cancer with radochlorine and photosense: comparing efficacy and toxicity. Proc. SPIE 2004, 5315, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XIII. https://doi.org/10.1117/12.537787. Date last accessed: February 22; 2024
  • 67 Vakulovskaya EG, Shental VV, Kondratjeva TT. Photodynamic therapy and fluorescent diagnostics of head and neck tumors with photosense. Int J Cancer 2002; 262-262
  • 68 Vakoulovskaya E, Shental V, Oumnova L, Vorozhcsov G. Photodynamic therapy of breast cancer with photosense. Proc. SPIE 2003, 4952, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XII. https://doi.org/10.1117/12.479431. Date last accessed: February 22; 2024
  • 69 Urbanska K, Romanowska-Dixon B, Matuszak Z, Oszajca J, Nowak-Sliwinska P, Stochel G. Indocyanine green as a prospective sensitizer for photodynamic therapy of melanomas. Acta Biochim Pol 2002; 49: 387-391
  • 70 Parker S. The use of diffuse laser photonic energy and indocyanine green photosensitiser as an adjunct to periodontal therapy. Br Dent J 2013; 215: 167-171
  • 71 Costa RA, Farah ME, Cardillo JA, Belfort R. Photodynamic therapy with indocyanine green for occult subfoveal choroidal neovascularization caused by age-related macular degeneration. Curr Eye Res 2001; 23: 271-275
  • 72 Tardivo JP, Del Giglio A, de Oliveira CS, Gabrielli DS, Junqueira HC, Tada DB, Severino D, Turchiello RDF, Baptista MS. Methylene blue in photodynamic therapy: From basic mechanisms to clinical applications. Photodiagnosis Photodyn Ther 2005; 2: 175-191
  • 73 Cengel KA, Simone 2nd CB, Glatstein E. PDT: Whatʼs past is prologue. Cancer Res 2016; 76: 2497-2499
  • 74 Dougherty TJ, Kauffman JE, Goldfarb A, Weishaupt KR, Boyle D, Mittleman A. Photoradiation therapy for the treatment of malignant tumors. Cancer Res 1978; 38: 2628-2635
  • 75 Diamond I, McDonagh A, Wilson C, Granelli S, Nielsen S, Jaenicke R. Photodynamic therapy of malignant tumours. The Lancet 1972; 300: 1175-1177
  • 76 Berns MW, Rettenmaier M, McCoullough J, Coffey J, Wile A, Berman M, Disaia P, Weinstein G. Response of psoriasis to red laser light (630 nm) following systemic injection of hematoporphyrin derivative. Lasers Surg Med 1984; 4: 73-77
  • 77 Adimoolam MG, A V. Nalam MR, Sunkara MV. Chlorin e6 loaded lactoferrin nanoparticles for enhanced photodynamic therapy. J Mater Chem B 2017; 5: 9189-9196
  • 78 Zhu YX, Jia HR, Chen Z, Wu FG. Photosensitizer (PS)/Polyhedral Oligomeric Silsesquioxane (POSS)-crosslinked nanohybrids for enhanced imaging-guided photodynamic cancer therapy. Nanoscale 2017; 9: 12874-12884
  • 79 OʼConnor AE, Gallagher WM, Byrne AT. Porphyrin and nonporphyrin photosensitizers in oncology: Preclinical and clinical advances in photodynamic therapy. Photochem Photobiol 2009; 85: 1053-1074
  • 80 Plonka J, Latocha M, Kusmierz D, Zielinska A. Expression of proapoptotic BAX and TP53 genes and antiapoptotic BCL-2 gene in MCF-7 and T-47D tumour cell cultures of the mammary gland after a photodynamic therapy with photolon. Adv Clin Exp Med 2015; 24: 37-46
  • 81 Copley L, van der Watt P, Wirtz KW, Parker MI, Leaner VD. Photolon, a chlorin e6 derivative, triggers ROS production and light-dependent cell death via necrosis. Int J Biochem Cell Biol 2008; 40: 227-235
  • 82 Chouikrat R, Seve A, Vanderesse R, Benachour H, Barberi-Heyob M, Richeter S, Raehm L, Durand JO, Verelst M, Frochot C. Non polymeric nanoparticles for photodynamic therapy applications: Recent developments. Curr Med Chem 2012; 19: 781-792
  • 83 Li X, Lee S, Yoon J. Supramolecular photosensitizers rejuvenate photodynamic therapy. Chem Soc Rev 2018; 47: 1174-1188
  • 84 Josefsen LB, Boyle RW. Unique diagnostic and therapeutic roles of porphyrins and phthalocyanines in photodynamic therapy, imaging and theranostics. Theranostics 2012; 2: 916-966
  • 85 Abrahamse H, Hamblin MR. New photosensitizers for photodynamic therapy. Biochem J 2016; 473: 347-364
  • 86 Xiao QC, Wu J, Pang X, Jiang Y, Wang P, Leung AW, Gao LQ, Jiang S, Xu CS. Discovery and development of natural products and their derivatives as photosensitizers for photodynamic therapy. Curr Med Chem 2018; 25: 839-860
  • 87 Zhao JB, Cui Q, Wang LG, Wu SH, Yang YC. Irradiation sensitivity-enhancing effect of psoralens on S180 cell line. J Fourth Mil Med Univ 1998; 19: 627-629
  • 88 Carneiro Leite V, Ferreira Santos R, Chen Chen L, Andreu Guillo L. Psoralen derivatives and longwave ultraviolet irradiation are active in vitro against human melanoma cell line. J Photochem Photobiol B 2004; 76: 49-53
  • 89 Plumas J, Drillat P, Jacob MC, Richard MJ, Favrot MC. [Extracorporeal photochemotherapy for treatment of clonal T cell proliferations]. Bull Cancer 2003; 90: 763-770
  • 90 Efferth T, Fabry U, Osieka R. Induction of apoptosis, depletion of glutathione, and DNA damage by extracorporeal photochemotherapy and psoralen with exposure to UV light in vitro . Anticancer Res 2001; 21: 2777-2783
  • 91 Yan XG, Zhou XY, Wu JZ. Study on the photosensitizing properties of the antitumor effect of psoralens. J Int Oncol 1999; 26: 83-85
  • 92 Zheng Y, Yin G, Le V, Zhang A, Chen S, Liang X, Liu J. Photodynamic-therapy activates immune response by disrupting immunity homeostasis of tumor cells, which generates vaccine for cancer therapy. Int J Biol Sci 2016; 12: 120-132
  • 93 Garg AD, Vandenberk L, Koks C, Verschuere T, Boon L, Van Gool SW, Agostinis P. Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell-driven rejection of high-grade glioma. Sci Transl Med 2016; 8: 328ra327
  • 94 Majerník M, Jendželovský R, Babinčák M, Košuth J, Ševc J, Tonelli Gombalová Z, Jendželovská Z, Buríková M, Fedoročko P. Novel insights into the effect of hyperforin and photodynamic therapy with hypericin on chosen angiogenic factors in colorectal micro-tumors created on chorioallantoic membrane. Int J Mol Sci 2019; 20: 3004
  • 95 Kim H, Kim SW, Seok KH, Hwang CW, Ahn JC, Jin JO, Kang HW. Hypericin-assisted photodynamic therapy against anaplastic thyroid cancer. Photodiagnosis Photodyn Ther 2018; 24: 15-21
  • 96 Plenagl N, Duse L, Seitz BS, Goergen N, Pinnapireddy SR, Jedelska J, Brüßler J, Bakowsky U. Photodynamic therapy – hypericin tetraether liposome conjugates and their antitumor and antiangiogenic activity. Drug Deliv 2019; 26: 23-33
  • 97 Chen XM, Li HY, Qu T, Gao M, Xu XH. Inhibitory effects of CIK on breast cancer cells intensified by hypericin mediated photodynamic therapy. Herald Med 2021; 40: 1318-1324
  • 98 Hu J, Song J, Tang Z, Wei S, Chen L, Zhou R. Hypericin-mediated photodynamic therapy inhibits growth of colorectal cancer cells via inducing S phase cell cycle arrest and apoptosis. Eur J Pharmacol 2021; 900: 174071
  • 99 Zhang XQ, Fang JY, Yao J, Chen MY, Song ZW, Xu LL, Zhao TJ. Mechanism of photodynamic therapy mediated by hypericin in inhibiting the proliferation of leukemia cell lines. Chin Pharm J 2018; 53: 967-974
  • 100 Chen HJ, Diao L, Zhang L, Liu HX, Li WQ, Huang XD, Zhang RL, Li GX. Study on antiviral effect of hypericin against infectious bronchitis virus in vitro . Chin Vet Sci 2018; 48: 1415-1422
  • 101 Kamuhabwa AAR, Huygens A, De Witte PAM. Photodynamic therapy of transitional cell carcinoma multicellular tumor spheroids with hypericin. Int J Oncol 2003; 23: 1445-1450
  • 102 Xu CS, Ling RN. Study of hypericin-mediated photodynamic therapy on human nasopharyngeal carcinoma cells. Laser Technology 2005; 29: 395-397
  • 103 Wan XY, Chen YT. A new photochemotherapy drug - Hypocrellin A. Sci Bull 1980; 24: 1148-1149
  • 104 Kishi T, Tahara S, Taniguchi N, Tsuda M, Tanaka C, Takahashi S. New perylenequinones from Shiraia bambusicola . Planta Med 1991; 57: 376-379
  • 105 Hu MM, Cai YJ, Liao XR, Hao ZK, Liu JY. Development of an HPLC method to analyze and prepare elsinochrome C and hypocrellin A in the submerged fermentation broth of Shiria sp SUPER-H168. Biomed Chromatogr 2012; 26: 737-742
  • 106 Diwu ZJ, Haugland RP, Liu J, Lown JW, Miller GG, Moore RB, Brown K, Tulip J, McPhee MS. Photosensitization by anticancer agents 21: New perylene- and aminonaphthoquinones. Free Radic Biol Med 1996; 20: 589-593
  • 107 Deininger MH, Weinschenk T, Morgalla MH, Meyermann R, Schluesener HJ. Release of regulators of angiogenesis following Hypocrellin-A and -B photodynamic therapy of human brain tumor cells. Biochem Biophys Res Commun 2002; 298: 520-530
  • 108 Chen J, Teng LR, Zheng KY, Li TJ, Ma L, Li C, Wu W, Fei XF. Study on the apoptotic molecule mechanism in A375-S2 cell induced by hypocrellin A. Chin Pharm J 2005; 40: 431-434
  • 109 Zhou JH, Xia SQ, Chen JR, Wang XS, Zhang BW, Zhang HJ, Zou P, Ai XC, Zhang JP. Surface binding and improved photodamage of the lanthanum ion complex of hypocrellin A to calf thymus DNA. J Photochem Photobiol A Chem 2004; 165: 143-147
  • 110 Zhao X. Key factors of photoactivated pesticide hypocrellin A against Botrytis cinere [dissertation]. Hangzhou: Zhejiang A&F University; 2015
  • 111 Mastrangelopoulou M, Grigalavicius M, Berg K, Menard M, Theodossiou TA. Cytotoxic and photocytotoxic effects of cercosporin on human tumor cell lines. Photochem Photobiol 2019; 95: 387-396
  • 112 Liu SS, Yang YH, Wang ZS. [Clinical study of CPD4-PDT for the prevention of postoperative recurrence in infiltrative bladder cancer]. Chin J Integr Med 1998; 18: 15-17
  • 113 Cao G, Guo T, Lu CT. Effect of CPD4 photodynamic therapy on endothelial cell in vitro . J Postgrad Med 2004; 17: 114-116
  • 114 Zhang JL, Chen P, Ding K, Dai SG, Wang K, Cao DL, Lin L, Tang GQ. Photodynamic effect of two kinds of CPD photosensitizers on sarcoma S180 transplanted in mice. Tianjin Med J 2006; 34: 705-707
  • 115 Yu HY. Silencing XBP1 expression enhances the sensitivity of human osteosarcoma HOS cells to MPPα-PDT [dissertation]. Chongqing: ChongQing Medical University; 2020
  • 116 Zhong SX. Inhibition of PERK pathway enhances sensitivity of human osteosarcoma HOS cells induced to pyropheophorbide-a methyl ester-mediated photodynamic therapy [dissertation]. Chongqing: ChongQing Medical University; 2019
  • 117 Wang L. Pyropheophorbide a photodynamic therapy in the control of golden hamster sebaceous patches [dissertation]. Nanjing: Nanjing University of Chinese Medicine; 2016
  • 118 Zeng XB. Inhibitive action of curcumin on human breast cancer and observation of the reinforcement effect after curcumin exposure to light [dissertation]. Chongqing: ChongQing Medical University; 2008
  • 119 Mou TL, Liu HM, Pan YS, He GF, Li ZW, Guo J, Pang DC, Zhai Z, Wu ZX. Study on the expression of tumor necrosis factor-α and cysteine protease-8 in cervical cancer xenografts and tumor tissues treated with multidose photodynamic therapy combined with curcumin. Global Tradit Chin Med 2022; 15: 543-549
  • 120 He JA, Hu YZ, Jiang LJ. Photodynamic action of phycobiliproteins: In situ generation of reactive oxygen species. Biochim Biophys Acta Bioenerg 1997; 1320: 165-174
  • 121 Nelson KM, Dahlin JL, Bisson J, Graham J, Pauli GF, Walters MA. The essential medicinal chemistry of curcumin. J Med Chem 2017; 60: 1620-1637
  • 122 Rao J, Xie J, Zhao JQ, Zhu T. Rhein-photodynamic sensitization generates free radicals and singlet oxygen. Sci Sin Chim 2004; 34: 211-217
  • 123 Hamblin MR, Abrahamse H. Tetracyclines: Light-activated antibiotics?. Future Med Chem 2019; 11: 2427-2445
  • 124 Greco G, Ulfo L, Turrini E, Marconi A, Costantini PE, Marforio TD, Mattioli EJ, Di Giosia M, Danielli A, Fimognari C, Calvaresi M. Light-enhanced cytotoxicity of doxorubicin by photoactivation. Cells 2023; 12: 392
  • 125 Chen GQ, Guo LN, Wu QZ. A study of the photosensitizing effect of an extraction from a medicinal herb belongs to leguminosales. Med J Qilu 2001; 16: 279-280
  • 126 Yao CS, Wu QZ. Photosensitizing effect of two extracts from caulis lonicerae: A preliminary study. Chin J Laser Med Surg 2006; 15: 361-364
  • 127 Liao J, Li PP, Wu CJ. Screening new photosensitizers from Chinese medicinal herbs and searching for herbal photodynamic killing effects on human stomach cancer cells. Chin J Integr Med 1997; 17: 726-729
  • 128 Scotti F, Mou L, Huang C, Booker A, Weckerle C, Maake C, Heinrich M. Treating chronic wounds using photoactive metabolites: Data mining the Chinese pharmacopoeia for potential lead species. Planta Med 2021; 87: 1206-1218
  • 129 Shi YG, Lin S, Chen WX, Jiang L, Gu Q, Li DH, Chen YW. Dual-stage blue-light-guided membrane and DNA-targeted photodynamic inactivation using octyl gallate for ultraefficient eradication of planktonic bacteria and sessile biofilms. J Agric Food Chem 2022; 70: 7547-7565
  • 130 Shi YG, Chen WX, Zheng MZ, Zhao YX, Wang YR, Chu YH, Du ST, Shi ZY, Gu Q, Chen JS. Ultraefficient OG-mediated photodynamic inactivation mechanism for ablation of bacteria and biofilms in water augmented by potassium iodide under blue light irradiation. J Agric Food Chem 2023; 71: 13672-13687
  • 131 Zheng MZ, Chen WX, Zhao YX, Fang Q, Wang LG, Tian SY, Shi YG, Chen JS. Ascorbic acid potentiates photodynamic inactivation mediated by octyl gallate and blue light for rapid eradication of planktonic bacteria and biofilms. Date last accessed: February 22 2024 SSRN: https://ssrn.com/abstract=4517458 DOI: 10.2139/ssrn.4517458
  • 132 Krieger-Liszkay A. Singlet oxygen production in photosynthesis. J Exp Bot 2004; 56: 337-346
  • 133 Gierlich P, Mata AI, Donohoe C, Brito RMM, Senge MO, Gomes-da-Silva LC. Ligand-targeted delivery of photosensitizers for cancer treatment. Molecules 2020; 25: 5317
  • 134 Escudero A, Carrillo-Carrión C, Castillejos MC, Romero-Ben E, Rosales-Barrios C, Khiar N. Photodynamic therapy: Photosensitizers and nanostructures. Mater Chem Front 2021; 5: 3788-3812

Zoom Image
Fig. 1 PDT-related publications.
Zoom Image
Fig. 2 Mechanism of photodynamic reactions.
Zoom Image
Fig. 3 Chemical structures of partial natural product photosensitizers.