Abstract
Conventional photodynamic therapy (PDT) approaches face challenges including limited light penetration, low uptake of photosensitizers by tumors, and lack of oxygen in tumor microenvironments. One promising solution is to internally generate light, photosensitizers, and oxygen. This can be accomplished through endogenous production, such as using bioluminescence as an endogenous light source, synthesizing genetically encodable photosensitizers in situ, and modifying cells genetically to express catalase enzymes. Furthermore, these strategies have been reinforced by the recent rapid advancements in synthetic biology. In this review, we summarize and discuss the approaches to overcome PDT obstacles by means of endogenous production of excitation light, photosensitizers, and oxygen. We envision that as synthetic biology advances, genetically engineered cells could act as precise and targeted “living factories” to produce PDT components, leading to enhanced performance of PDT.
Key words: Photodynamic therapy, Cancer, Biogenesis, Biosynthesis, Engineered bacteria, Phototherapy, Photosensitizers, Catalase
Graphical abstract
Targeted delivery of external light, photosensitizers, and oxygen to tumors is challenging, which hinders the efficacy of photodynamic treatments. Endogenous production of light, photosensitizers, and oxygen in-situ at tumor sites is a promising solution.

1. Introduction
Antitumor treatment with photodynamic therapy (PDT) has been extensively investigated in the past few decades. PDT utilizes photosensitizers that can be excited by light of specific wavelengths. Light-activated photosensitizers generate cytotoxic reactive oxygen species (ROS), thereby inducing apoptosis, necrosis, and autophagy of cells1. PDT has received approval as a non-invasive option for antitumor treatment2. Over the past 30 years, PDT has witnessed a progressive surge in its utilization for the management of diverse solid malignancies3.
ROS are generated via two pathways in PDT. Type I PDT operates in an oxygen (O2)-independent manner, while Type II PDT relies on the presence of molecular oxygen. The Type II pathway is the primary mechanism of most PDT, wherein the energized photosensitizers interact with O2 to produce highly toxic singlet oxygen (1O2, Fig. 1). There are only a limited number of photosensitizers that use Type I mechanism, and most clinically approved PDT use Type II mechanism. Excitation light, photosensitizers, and O2 are the three essential components for Type II PDT. There are several challenges associated with the three components that hinder PDT efficiency in clinical practice, such as the constrained tissue penetration depth of light, the poor water solubility of conventional photosensitizers, and the presence of hypoxia within the tumor microenvironment (TME)4.
Figure 1.
Scheme of the photochemical reaction and three key components involved in type II PDT.
The penetration depth of the excitation light determines the depth of the tumor that can be treated by PDT. In the context of traditional PDT, Ultraviolet–Visible light (UV–Vis) is commonly employed to excite photosensitizers. The light of such wavelength (400–700 nm) has poor tissue penetration, so the PDT efficiency is limited in treating deep-seated tumors5. To overcome this “Achilles’ heel”, different light sources have been explored. For example, upconversion nanoparticles (UCNPs) are applied to PDT to convert near-infrared (NIR) excitation light to UV–Vis light6. There have also been attempts to use X-ray in PDT7. In addition to the external excitation light sources, photosensitizers can be activated by self-luminescence, including chemiluminescence (CL)8 and Cerenkov radiation (CR) luminescence. Such internalized light sources produce excitation light locally at the tumor sites, resolving the tissue penetration limitations of external light. Bioluminescence is a type of chemiluminescence involving luciferase enzymes. The introduction of genes encoding luciferases (Fig. 1) into cancer cells allows for the synthesis of luciferase enzymes, enabling the production of excitation light for in situ PDT. This approach has been validated as a successful strategy for enhancing the performance of PDT9.
The photosensitizers used in PDT are primarily exogenous compounds administered by intravenous injection, subcutaneous injection, and topical application. A plethora of photosensitizers have been synthesized, encompassing both organic and inorganic variants9,10; however, they have some common limitations, such as low tissue selectivity, low water solubility, and low biocompatibility. Endogenous small molecule photosensitizers, such as flavin compounds and porphyrin compounds, can be induced in cancer cells for PDT. Protein photosensitizers can also be expressed endogenously in cancer cells by delivering genes encoding the proteins to target cells11.
Hypoxia represents an inherent attribute exhibited by numerous malignant solid tumors, exerting a profound constraint on the therapeutic effectiveness of oxygen-dependent Type II PDT12. Many strategies have been attempted to alleviate hypoxia at tumor sites13, such as direct delivery of O2 to tumors14 and generation of O2 from MnO2-catalyzed decomposition of hydrogen peroxide (H2O2)15. Recently, biological approaches to relieve tumor hypoxia have been successfully applied to PDT. For instance, the use of catalase to breakdown H2O2 into O2 or the use of photosynthetic microbes to generate O2 in situ enhanced the PDT efficiency16,17.
The reagents and plasmids involved in the above-mentioned strategies need to be efficiently delivered to cancer cells to achieve improved PDT performance. Artificial nanomaterials are commonly used delivery vehicles to enhance PDT efficacy18. In recent years, advancements in synthetic biology have driven the development of engineered bacteria as drug delivery vehicles. Anaerobic bacteria prefer the hypoxic TME and actively colonize and proliferate at tumor sites19. As drug carriers, the accumulation of bacteria in the hypoxic TME is substantially higher than that of conventional abiotic carriers20. In addition, bacteria can stimulate an effective immune response to synergistically promote PDT results. Using the synthetic biology toolkit, bacteria can be further programmed to produce specific drugs or target certain types of cells21.
In this review, we present a comprehensive overview and analysis of the methodologies employed to overcome the constraints associated with antitumor PDT, with a particular emphasis on endogenous approaches. Strategies involving the endogenous production of excitation light, photosensitizers, and O2 are discussed in detail, with particular emphasis on the biogenesis of these components. In light of recent advancements in synthetic biology, the application of genetically engineered bacteria in PDT has also been discussed. Finally, we highlight the potential of engineered bacteria not only as effective drug carriers but also as producers of excitation light, photosensitizers, and O2.
2. Excitation light for PDT
Photosensitizers need to be excited by light with matching wavelengths to generate ROS for antitumor PDT22,23. Upon the absorption of photons, the photosensitizer undergoes a transition from the ground singlet state to a higher-energy excited state, which subsequently initiates a photochemical reaction via two distinct mechanisms. In Type I PDT, the photoactivated photosensitizer engages in direct interactions with adjacent biological substrates, including nucleic acids, proteins, lipids, or water molecules, leading to the production of free radicals. On the other hand, Type II PDT involves the transfer of energy from the excited photosensitizers to molecular oxygen in the vicinity, resulting in the generation of 1O2, which is considered as the primary therapeutic ROS24, 25, 26, 27, 28.
For optimal results, it is imperative to align the emitting wavelength of the light source with the absorption spectrum of the photosensitizer. Furthermore, meticulous modulation of light energy is essential to enhance ROS generation while mitigating potential damage to healthy tissues29. The excitation light wavelength of most photosensitizers falls within the optical window of biological tissues and is therefore not ideal for deep tumor treatment due to limited penetration depth30,31. Numerous strategies have been devised to overcome the aforementioned constraints.
2.1. External light source
To address the issue of light penetration in deep tissues, various wire-based methodologies have been devised to facilitate the transmission of external light. These approaches include the utilization of light-emitting diodes and fiber optics coupled with lasers32. Another promising approach involves the use of light conversion nanomaterials, which can be used to convert external light or radiofrequency signals into localized light internally, enabling wireless phototherapy. UCNPs are nanoparticles that are doped with rare-earth elements and possess the ability to convert NIR radiation to UV–Vis light. When employed in PDT, UCNPs transform externally applied NIR light into localized UV–Vis light, thus enabling the activation of photosensitizers. UCNP-based PDT shows better performance in treating deep-seated tumors33. For instance, a UCNP NaYF4:Yb3+, Er3+ was constructed for PDT application, which can be excited by 980 nm NIR light and emit 660 nm light34,35. The UCNP was coated with mesoporous silica and loaded with the photosensitizer zinc phthalocyanine. The emission light from NIR-activated UCNPs excited zinc phthalocyanine, producing ROS to eliminate cancer cells36. Similar strategies have been explored using different UCNPs and photosensitizers pair37, demonstrating good outcomes in overcoming the poor tissue penetration challenge in conventional PDT38,39. The depth of treatable tumors through NIR-activated PDT can be further enhanced with the adjunctive application of biocompatible implants40.
X-ray is a source of ionizing radiation with no penetration depth limitation in human tissue. X-ray can be used to excite scintillating nanoparticles which emit UV–Vis light for the activation of photosensitizers41. Micheletto et al.42 combined LaF3:Tb3+ with the protein photosensitizer KR (KillerRed) to perform X-ray activated PDT. Under irradiation, the emission light from LaF3:Tb3+ with a wavelength of around 580 nm activates the photosensitizer. X-ray responsive PDT is a promising treatment strategy for deep-seated tumors, but the safety issue of X-ray needs to be addressed with caution.
2.2. Internal light source
Light sources, including Cerenkov radiation luminescence and chemiluminescence, not requiring external irradiation are alternative options for antitumor PDT. CR43 luminescence has a wavelength range of 250–600 nm, which is generated by positron-emitting radionuclides44. Kotagiri et al.45 employed CR luminescence as an internal light source for the activation of the photosensitizer TiO2 nanoparticles. The excited TiO2 nanoparticles generated hydroxyl and superoxide radicals, which effectively impeded tumor proliferation and significantly extended the survival of tumor-bearing mice. Chemiluminescence produces light from chemical reactions, also requiring no external irradiation46. For example, luminol is a commonly used substrate for chemiluminescence, which can luminescence after a reaction with H2O2 catalyzed by horseradish peroxidase. The luminescence from luminol around 450 nm was utilized to excite the photosensitizer Ce6 for PDT. The chemiluminescence induced PDT exhibited in vitro cytotoxicity against human lung cancer cells and inhibited the tumor development in vivo47 (Fig. 2A). Bioluminescence is a type of chemiluminescence observed in living organisms that has been investigated in PDT as well.
Figure 2.
Examples of PDT using chemiluminescence and bioluminescence. (A) Luminol is linked with chlorine e6 (Ce6), and the construct self-assembles into nanoparticles that can be used for chemiluniescence-activated photodynamic therapy (PDT). Reproduced with permission from Ref. 47. Copyright © 2019 Wiley. (B) Polyethyleneimine-modified nano calcium phosphate encapsulating D-luciferin, hypericin, and a plasmid inserted with the Fluc gene is used for bioluminescence-mediated PDT. Reproduced with permission from Ref. 55. Copyright © 2021 Wiley. (C) Renilla luciferase-bound quantum dots (QD-RLuc8) emit 655 nm light due to bioluminescence resonance energy transfer, which can activate Foscan-loaded micelles producing ROS. Reproduced with permission from Ref. 57. Copyright © 2013 Elsevier. (D) QDs were coated with a PEG polymer encapsulating Rluc8. In the presence of coelenterazine, the construct self-illuminates and activates Ce6, leading to cytotoxicity. Reproduced with permission from Ref. 49. Copyright © 2015 Ivyspring. (E) BSA conjugated with both RLuc8.6 and Rose Bengal can produce ROS via BRET in the presence of the luciferin substrate (BSA: bovine serum albumin, RB: Rose Bengal). Reproduced with permission from Ref. 58. Copyright ©2017 Royal Society of Chemistry. (F) A fusion protein of RLuc8.6 and KillerRed is constructed which generates ROS when coelenterazine-h is provided. Injection of the fusion protein and coelenterazine-h resulted in strong luminescence and killed cancer cells in the xenograft mouse model established using MDA-MB-231 cells (RKL: fusion of Rluc8.6 and KillerRed, Co-h: coelenterazine-h). Reproduced with permission from Ref. 59. Copyright © 2020 Wiley.
A typical bioluminescence system is composed of a luciferase enzyme and a luciferin substrate for the enzyme. Bioluminescence systems have been identified across various organisms, including fungi, bacteria, insects, and certain marine organisms. In order to overcome the challenges associated with conventional PDT regarding light delivery, researchers have explored the utilization of bioluminescence as an internal light source alternative. This involves investigating the ability to introduce luciferase proteins or luciferase genes to tumor sites, enabling the endogenous production of light in the presence of luciferins. By ensuring that the bioluminescence spectrum overlaps with the absorption spectrum of the photosensitizer and that they are in close proximity, it becomes possible to excite the photosensitizer and generate ROS for PDT48. Three bioluminescence systems (i.e., FLuc, Rluc, and NLuc) have been tested for PDT49.
2.2.1. Firefly luciferase bioluminescence in PDT
The firefly luciferase (FLuc) bioluminescence system is the first bioluminescence system discovered and is also the most studied and used system. It was initially isolated from the firefly Photinus pyralis. The FLuc protein, with a molecular weight of 61 kDa, catalyzes the biochemical reaction between D-luciferin, O2, and ATP. The Fluc luminescence exhibits a maximum emission wavelength of 558 nm50. Fluc bioluminescence can function as an endogenous light source within tumors, thus solving the problems associated with delivering external light to deep tissues. In 1994, Carpenter et al.51 reported Fluc bioluminescence-mediated PDT using hypericin as the photosensitizer. The PDT effectively inactivated the equine infectious anemia virus, demonstrating that the light from Fluc bioluminescence can induce PDT. In 2003, Fluc bioluminescence induced PDT was reported in cells52. Theodossiou et al.52 transfected the FLuc gene into mouse embryonic fibroblast cells and treated the cells with the photosensitizer Rose Bengal (RB). PDT was induced by the addition of the luciferin substrate, which led to 89% cell death. This study suggested that bioluminescence can be an endogenous light source for antitumor PDT. However, this result was challenged by a study performed by Schipper et al.53 in 2006 showing that the FLuc bioluminescence system cannot produce enough photons to achieve potent photodynamic toxicity. The observed disparity between the outcomes obtained by the two groups may be attributed to the different concentrations of D-luciferin employed in their respective experimental procedures (500 μmol/L by Theodossiou et al.52 and 20 μmol/L by Schipper et al.53). Since then, accumulated efforts have been devoted to improving the performance of Fluc bioluminescence-mediated antitumor PDT.
The poor overlap between the bioluminescent spectrum and the photosensitizer absorption spectrum hinders effectiveness of PDT. Yang et al.54 converted the 558 nm light from Fluc bioluminescence to 635 nm through a BRET (bioluminescence resonance energy transfer) process between Fluc and carbon dots. The wavelength of the emission light from carbon dots overlapped well with the photosensitizer PPIX. The distance between the bioluminescence source and the photosensitizer is another factor limiting PDT efficacy. Confining Fluc proteins and photosensitizers in close proximity contributes to a more efficient energy transfer between them. A polylactic-glycolic acid nanoparticle was loaded with the photosensitizer RB and then bioconjugated with FLuc proteins. The system allowed a more efficient BRET between the Fluc protein and RB, exhibiting enhanced antitumor PDT performance48. Nevertheless, the low delivery efficiency of Fluc protein limits the intensity of bioluminescence. An alternative approach is to deliver the gene encoding Fluc instead of Fluc proteins. A nanomedicine was developed by encapsulating d-luciferin, hypericin, and a plasmid inserted with the Fluc gene in polyethyleneimine-modified nano calcium phosphate55 (Fig. 2B). The nanomedicine collapsed in the lysosome of tumor cells, releasing the plasmid. The Fluc gene carried by the plasmid was abundantly expressed in tumor cells, leading to the accumulation of FLuc proteins. The endogenously expressed Fluc proteins reacted with the released D-luciferin to generate bioluminescence. This strategy can increase the concentration of FLuc protein at tumor sites, thereby enhancing the intensity of bioluminescence and thus stimulating a stronger PDT.
2.2.2. Renilla luciferase bioluminescence in PDT
Renilla luciferase (RLuc) is a 36 kDa protein found in sea pansy Renilla reniformis. RLuc reacts with its enzyme substrate coelenterazine to generate bioluminescence in the presence of O2. Compared to FLuc, RLuc is smaller in size and does not require ATP to luminesce. RLuc bioluminescence exhibits emission at 480 nm. RLuc has been extensively investigated as a luminescent reporter in mammalian cells. RLuc8 and RLuc8.6 are brighter and more stable mutants derived from the original RLuc protein. RLuc8 demonstrates a 4-fold increase in bioluminescent intensity and a marginal redshift in its emission, relative to RLuc. RLuc8.6 has an emission wavelength of 535 nm, which is redshifted by 55 nm relative to RLuc. The RLuc substrate coelenterazine is less soluble and less stable than D-luciferin and is prone to autoxidation56.
To convert the light from RLuc bioluminescence to light with excitation wavelengths of common photosensitizers, quantum dots have been used as the intermediate energy transmitter. The RLuc8 protein has undergone a process of bioconjugation, resulting in its linkage to a quantum dot that emits at 655 nm. When coelenterazine is present, the bioluminescence of RLuc8 activates the fluorescence of the quantum dots, which in turn can stimulate the photosensitizer Foscan for PDT57 (Fig. 2C). The ROS produced led to 50% eradication of A549 tumor cells in vitro, along with a substantial retardation in tumor progression in an in vivo model. Similarly, quantum dots were conjugated with RLuc8 to achieve bioluminescence-mediated excitation of Ce649 (Fig. 2D). The system was therapeutically effective against a variety of cancer cells and metastatic tumors as well. It exhibited better therapeutic effects on deep tumors compared to a system activated by external laser irradiation. RLuc8.6 has redshifted emission that overlaps with the absorption spectrum of the photosensitizer RB. A covalent conjugate of RLuc8.6 and RB was constructed to form a BRET pair58 (Fig. 2E). The conjugate underwent additional coupling with cell-penetrating peptides in order to enhance its internalization efficiency into cancer cells. Upon the introduction of coelenterazine, the conjugate exhibited photodynamic toxicity, resulting in a 75% decrease in viability of colon cancer cells. To avoid the risks for the toxicity of chemical photosensitizers and nanomaterials, KillerRed as a protein photosensitizer was fused with RLuc protein for PDT59 (Fig. 2F). The fusion protein was subsequently conjugated with a leading peptide for the purpose of selectively targeting cancer cells. The resulting protein construct demonstrated the ability to specifically recognize and enter breast cancer cells. It exhibited the capacity to induce PDT-mediated cancer cell death within tumor xenograft mouse models when coelenterazine was present. Notably, these findings highlight that bioluminescence, which is a relatively low-intensity light source, is able to efficiently induce antitumor PDT in vivo.
2.2.3. Luciferase nanoLuc–furimazine bioluminescence in PDT
NanoLuc (NLuc) is a luciferase protein derived from a deep-sea shrimp Oplophorus gracilirostris luciferase after several rounds of mutagenesis. It is smaller than both Fluc and RLuc, with a molecular weight of 19.1 kDa. NLuc catalyzes the reaction involving furimazine, its substrate, leading to the emission of bioluminescent light with a wavelength of 456 nm in the presence of O2. The emission of NLuc is blue-shifted by approximately 20 and 100 nm relative to RLuc and FLuc60. The luminescence produced by NLuc is ∼150-fold greater than that of FLuc or RLuc61. However, the substrate furimazine is more expensive than D-luciferin and coelenterazine. The maximum emission wavelength of NLuc at 460 nm matches the absorption wavelength of the protein photosensitizer miniSOG62,63. The NLuc and miniSOG were encoded in one plasmid and expressed as a fusion protein64. The phototoxicity of miniSOG stimulated by NLuc bioluminescence was comparable with that of miniSOG excited by LED. As the construct is genetically encodable, it can potentially be delivered to any tissue site to treat deep and metastatic tumors.
3. Photosensitizers for PDT
The photosensitizer constitutes a triad of indispensable components for PDT, wherein it absorbs excitation light and subsequently generate cytotoxic ROS65. Photofrin, a photosensitizer authorized by U.S. Food and Drug Admistration (FDA) in 1993, emerged as the first photosensitizing agent approved for application in PDT. Since the approval of photofrin, more than twenty PDT reagents have been commercialized and hundreds more are in clinical trials or preclinical studies66. Photosensitizers are mainly classified into organic photosensitizers and inorganic photosensitizers67,2. The requirements for an ideal photosensitizer include: 1) high stability at room temperature, good solubility and pharmacokinetics, and nontoxicity in the absence of photoactivation; 2) high extinction coefficient within the optical range characterized by minimal tissue absorption; 3) significant singlet oxygen quantum yield and an extended lifetime of trilinear excited state.
3.1. Organic photosensitizer
To date, four generations of organic photosensitizers have been developed. The initial cohort of photosensitizers encompasses chiefly hematoporphyrin and its derivatives as the primary compounds. Photofrin is a mixture of hematoporphyrin derivatives that can be excited by 630-nm light, showing inhibitory effects on breast, colon, prostate, head, and neck cancers68. Most of the 1st generation photosensitizers are faced with limitations such as long metabolic cycles in the human body, high accumulation in skin, and the need for patients to avoid strong light for weeks. The 2nd generation photosensitizers mainly include chlorine, bacteriochlorin analogues, phthalocyanines, and protoporphyrin IX (PPIX) compounds69. Foscan, NPe6, Levulan, and Metvix are 2nd generation photosensitizers that have been clinically approved with inhibitory effects on lung, skin, and bladder cancer. Compared with the 1st generation, 2nd generation photosensitizers are improved in terms of homogeneity, tumor selectivity, quantum yield, and reduced accumulation in the skin70. However, the efficiency of PDT therapy with 2nd generation photosensitizers is hindered by the hydrophobicity and self-aggregation of the reagents in biological media71. The 3rd generation photosensitizers use nanoparticles (e.g., liposomes, micelles, dendrimers, and mesoporous silica) as drug carriers; and the 4th generation are photosensitizers loaded in metal–organic frameworks72. These two types are developed to enhance the uptake by tumors and attenuate the off-target release of photosensitizers. They exhibit promising outcomes in preclinical studies, but their long-term biosafety is still under scrutiny73.
3.2. Inorganic photosensitizer
Compared with organic photosensitizers, inorganic nanoparticle photosensitizers have higher light conversion efficiency, higher 1O2 quantum yield, and better stability. The TiO2 nanoparticle is an inorganic photosensitizer that responds to UV irradiation74. Metallic nanoparticles, including silver, gold, and platinum nanoparticles, exhibit the capability of generating 1O2 when exposed to light irradiation (398–530 nm)75. Additional inorganic photosensitizers encompass nanoparticles based on metal sulfides (such as copper sulfide and molybdenum sulfide)76, as well as nanoparticles based on carbon (including graphene and carbon dots)77. Although with better photostability than most organic photosensitizers, the long-term toxicity and biosafety of inorganic photosensitizers need to be thoroughly investigated by more in vivo studies. Barriers to the clinical use of inorganic photosensitizers mainly include the unclear biocompatability, their slow clearance in bodies, and the tendency to aggregate and bind biomolecules in biological media78.
The organic and inorganic photosensitizers discussed above are chemically synthesized and supplied through injection or oral administration. Efforts have been made to optimize the stability, solubility, and specificity of photosensitizers to ensure that they are not modified or degraded in circulatory systems and that sufficient amounts can be targeted to tumors. Recently, genetically encodable photosensitizers that can be biosynthesized endogenously at tumor sites have received growing attention.
Such photosensitizers are biosynthesized in cells, thus having better biocompatibility than exogenously provided photosensitizers that are chemically synthesized. On-site synthesis of photosensitizers also helps to address the issues related to the stability of PDT reagents during delivery. Using different inducible promoters, the biogenesis of genetically encoded photosensitizers can be precisely regulated temporally and spatially. Genetic tools can be used to modify the photosensitizers, for example, specific localization peptide sequences can be easily fused to protein photosensitizers to target specific organelles of tumor cells. Genetically encodable photosensitizers include protein-based photosensitizers and small molecule-based photosensitizers.
3.3. Genetically encodable protein photosensitizers
3.3.1. Different protein photosensitizers
KillerRed is a dimeric protein with a molecular weight of 54 kDa that originates from anm2CP, a chromoprotein initially identified in hydrozoan species. KillerRed exhibits a structural similarity to GFP and possesses a chromophore composed of the amino acids Gln-Tyr-Gly79. The chromophore absorbs green light (540–580 nm) and emits red fluorescence (maximum emission at 610 nm), meanwhile producing ROS. The KillerRed exhibits a phototoxic effect that is greater than that of other fluorescent proteins by 1000-fold80. KillerRed demonstrated strong inhibitory effects against both bacterial and mammalian cells under green light irradiation. KillerRed proteins mainly produce phototoxicity via Type I PDT, and thus can be applied in hypoxic TME81. Via genetic engineering, fusion proteins of KillerRed with linked localization peptides were constructed to target specific organelles or cellular compartments in cancer cells. It has been observed that the delivery of KillerRed to different organelles leads to varied PDT treatment outcomes. For instance, delivery of KillerRed to mitochondria and lysosomes triggers apoptosis, while PDT induced by KillerRed in the plasma membrane results in necrosis82.
In spite of the efficacious PDT outcomes and the genetic editability exhibited by KillerRed, there exist inherent limitations that hinder the in vivo application of KillerRed. The primary impediment to the in vivo utilization of KillerRed is attributed to the limited tissue penetration depth stemming from the short wavelength of the excitation light. In addition, KillerRed requires dimerization to function, which may interfere with the properties of genetically engineered KillerRed fusion proteins. To address these limitations, KillerRed derivatives (e.g., SuperNova, KillerOrange, and mKillerOrang) containing selected mutations have evolved.
SuperNova is a monomeric phototoxic protein derived from KillerRed containing a total of six mutations. SuperNova exhibits similar optical properties and comparable phototoxicity to KillerRed. SuperNova proteins do not dimerize; therefore SuperNova fusion proteins can be easily designed83,80. KillerOrange is a KillerRed mutant with a blueshifted emission spectrum84. It absorbs light at 455 and 514 nm and emits orange fluorescence, producing ROS with a quantum yield of 0.42. The chromophore of KillerOrange consists of tryptophan instead of the tyrosine in KillerRed. The phototoxicity of KillerOrange was demonstrated in both E. coli cells and HEK 293 cells. It was also observed that when KillerOrange and KillerRed are used simultaneously, the two proteins do not interfere with each other in both bacterial and mammalian cells. A monomeric variant of KillerOrange, mKillerOrange, was constructed by introducing a tyrosine-to-tryptophan mutation in SuperNova85. The mKillerOrange protein has similar optical properties as KillerOrange, but does not require dimerization to function.
MiniSOG (mini Singlet Oxygen Generator) is a protein photosensitizer that has been developed through mutagenesis of the LOV2 domain, which is a component of the blue light photoreceptor known as phototropin 286. MiniSOG is a 15.3 kDa monomeric protein, much smaller than 54 kDa KillerRed, thus allowing easier expression and exerting less influence in the physiology of cells. Unlike KillerRed and KillerOrange, miniSOG uses bound flavin mononucleotide (FMN) as the chromophore, producing green fluorescence (500 nm) and ROS upon irradiation by blue light (448 nm). It was reported later that miniSOG produces singlet oxygen less than originally expected and also generates superoxide via a Type I photochemical reaction87. As a result of the absence of FMN within mammalian cells, the supplementation of FMN from an external source becomes necessary to perform PDT utilizing miniSOG88.
The miniSOG structure was mutated to tailor the optical and photodynamic properties. The targeted mutagenesis of glutamine103 in miniSOG to leucine resulted in a more phototoxic photosensitizer SOPP with a higher 1O2 quantum yield, also named miniSOGQ102L89, 90, 91. miniSOG2 was a miniSOG mutant obtained by directed evolution. Seven mutations were introduced in miniSOG2, four of which are on residues in the binding pocket for FMN. These four mutations substantially enhanced the 1O2 quantum yield of miniSOG2 to 0.4792.
3.3.2. PDT with protein photosensitizers
The localization of the protein photosensitizer is a critical factor that governs the different damage pathways elicited by PDT. In recent times, a variety of promising methodologies have emerged pertaining to the transportation of purified protein photosensitizers or genes encoding such photosensitizers to tumors.
3.3.2.1. Delivery of purified protein photosensitizers
Protein photosensitizers can be expressed in cells, usually bacterial cells, and purified to homogeneity. The purified proteins are then delivered to tumors for PDT treatment. In order to protect the protein photosensitizers and augment the selectivity of transportation, nanocarriers are frequently employed.
KillerRed was expressed and isolated from E. coli cells for PDT in three leukemia cell lines (i.e., K562, NB4, and THP-1). Under irradiation (400–700 nm, 80 mW/cm2, 20 min), KillerRed induced apoptosis of leukemia cells in a concentration-dependent manner93 Liang et al.94 covalently coupled KillerRed to UCNPs to construct a nanoparticle–protein complex responsive to NIR light. Under 980 nm NIR irradiation, the UCNPs emitted green light to excite KillerRed. The KillerRed-mediated PDT efficiently eradicated breast cancer cells in vitro and also demonstrated successful deep tumor therapy in vivo (Fig. 3A). To improve the tumor targeting specificity of KillerRed, a liposome and cancer cell membrane hybrid complex was developed as a delivery vehicle. A fusion protein of KillerRed linked to a membrane-localization signal peptide was expressed in 4T1 cancer cells. The cell membrane was extracted with KillerRed anchored and hybridized with liposomes. The lipo-complex prepared in this study demonstrated a cancer-targeting efficiency that was 3.3 times greater than that of a control liposome. Moreover, it effectively suppressed the growth of primary tumors and inhibited lung metastasis in vivo95. MiniSOG has also been expressed as fusion proteins for correct localization in target cells96. Mironova et al.97 conjugated miniSOG with a peptide capable of selectively binding to the human epidermal growth factor receptor 2 (HER2). The fusion protein specifically targeted HER2-positive cancer cells as it could be internalized via receptor-mediated endocytosis. Upon irradiation with 1 W/cm2 white light for 10 min, the miniSOG derivative exerted strong and highly specific phototoxic effects on HER2-positive breast cancer cells.
Figure 3.
Examples of PDT using protein photosensitizers and small-molecule natural product photosensitizers. (A) UCNPs are employed to enhance the depth of PDT treatment under NIR excitation by coupling with KillerRed through EDC/NHS chemistry (PAA: poly(acrylic acid), EDC: 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide, NHS: N-hydroxysuccinimide). Reproduced with permission from Ref. 94. Copyright © 2017 Elsevier. (B) Virus AAV2 encoding KillerRed is loaded in lactate responsive nanoparticles to achieve tumor microenvironment targeting PDT (AAV2: adeno-associated virus serotype 2). Reproduced with permission from Ref. 98. Copyright © 2018 ACS publications. (C) The fusion of NanoLuc luciferase and miniSOG allows energy transfer from bioluminescence to miniSOG, generating ROS to kill cancer cells. Reproduced with permission from Ref. 103. Copyright © 2022 Springer Nature. (D) A complex consisting of chitosan and poly(γ-glutamic acid) is constructed to enhance the delivery of a plasmid carrying KillerRed gene. KillerRed is expressed intracellularly and induces green light-triggered cell destruction (CS: chitosan, γPGA: poly(γ-glutamic acid)). Reproduced with permission from Ref. 104. Copyright © 2014 Elsevier. (E) 5-ALA loaded PAMAM dendrimers are wrapped with RBCMs and surface modified with DNA aptamer AS1411. 5-ALA released from the resulting materials can be transformed into heme in cells, which forms peroxidase mimics with the AS1411 to produce ROS (5-ALA: 5-aminolevulinic acid, PAMAM: polyamidoamine, RBCMs: red blood cell membrane). Reproduced with permission from Ref. 115. Copyright ©2022 Wiley. (F) A nanocomposite consisting of MnCO3, riboflavin, and pDNA is constructed to treat triple-negative breast cancer. It decomposes under TME, releasing Mn2+, O2, and riboflavin for PDT and CDT, as well as pDNA to suppress the expression of survivin gene for enhanced tumor destruction (Rf: riboflavin, pDNA: plasmid DNA). Reproduced with permission from Ref. 116. Copyright © 2021 Springer Nature.
3.3.2.2. Delivery of genes encoding protein photosensitizers using viral vectors
KillerRed and miniSOG are genetically encodable, thereby their genes can be delivered to tumors enabling endogenous biogenesis of the photosensitizers. This strategy will accumulate more photosensitizers at tumor sites than a single shot of purified proteins, thus allowing sustainable PDT treatment98 (Fig. 3B). Viruses can be genetically engineered to target and eliminate tumor cells. FDA has approved genetically modified viruses as anticancer treatments. Viral vectors have been extensively used to deliver genes into mammalian cells, due to their high transfection efficiency. Both KillerRed and miniSOG have been successfully delivered to tumor cells using viral vectors.
To achieve selective expression of KillerRed in cancer cells, a recombinant adenovirus vector, named TelomeKiller, was constructed by placing the KillerRed gene under the control of a human telomerase reverse transcriptase promoter. This promoter is upregulated in most cancers but not in normal cells. Intratumoral administration of TelomeKiller exhibited potent anti-tumor effects by attenuating the proliferation of human rectal cancer cells and impeding the regional lymph node metastasis in a HCT116 xenograft tumor model, facilitated by the application of green light irradiation99. Intravenous injection of viral vectors faces more challenges, as the immune system tends to clear the viruses. A magnetic field-responsive viral vector was developed by chemically conjugating iron oxide nanoparticles to a recombinant adeno-associated virus carrying the KillerRed gene100. The magnetic vector was administered intravenously through the tail vein of mice bearing xenografted H1975 tumors, and its localization to tumor sites was facilitated by a magnetic field. KillerRed was abundantly expressed in tumors and significantly inhibited tumor growth upon illumination at 561 nm for 20 min.
Lentiviruses can be engineered to be integration vectors that insert the carried gene into chromosomes of target cells to construct stable expression cells. Different miniSOG constructs targeting varied cellular components (i.e., mitochondria, chromatin, and cell membrane) were expressed in tumor cells using lentiviral vectors101. A derivative of miniSOG that specifically targets the cell membrane demonstrated the highest level of photoactivated cytotoxicity against HeLa cells. Nevertheless, the lentiviral vector failed to manifest substantial therapeutic efficacy in murine models, presumably attributed to the limited tissue permeability of blue light and the inefficient delivery of FMN cofactors101. As NLuc bioluminescence overlaps with the absorption of miniSOG, a lentiviral vector hosting a fusion construct of NLuc and miniSOG was developed. The DNA encoding NLuc-miniSOG fusion protein was injected into BT-474 tumor cells bearing mice. The NLuc substrate furimazine and the FMN precursor riboflavin were provided intravenously every day. A tumor growth inhibition coefficient exceeding 67% was achieved after the treatment, suggesting bioluminescence can be an effective light source for in vivo PDT using miniSOG as the photosensitizer102,103 (Fig. 3C).
3.3.2.3. Delivery of genes encoding protein photosensitizers using nonviral vectors
Due to the concern about genotoxicity related to viral vectors, nonviral vectors are also developed to deliver genes for protein photosensitizers. Cationic polymers, such as chitosan, can be carriers for negatively charged DNA molecules. To facilitate the DNA release and improve the transfection efficiency, polyglutamic acid was incorporated into chitosan to deliver a plasmid hosting the KillerRed gene. HEK293 cells transfected with the polymer vehicles could express KillerRed and got inhibited under green light exposure. The effect was negligible after eight days, indicating that cells did not undergo genetic modification104 (Fig. 3D). In addition, guanidine modified pullulan105,106, polyethyleneimine polymer104,107, and DNA tetrahedron108 have also been used to optimize the delivery efficiency of KillerRed gene. Nonviral vectors commonly mediate transient transfection of genes, and thus the expression of protein sensitizers is temporal, and the toxicity associated with the proteins spontaneously decreases over time.
3.4. Genetically encodable small molecule-based photosensitizers
There are a variety of small-molecule photosensitizers discovered that are natural products biosynthesized by plants, microbes, and mammalian cells. For example, the commonly used photosensitizer hypericin can be biosynthesized by species of the genus Hypericum, and some fungi as well. Endogenous small molecules with phototoxicity have also been reported in human cells, such as biogenic 3-hydroxy pyridine derivatives in human skin cells that respond to UV irradiation109, 110, 111. These small-molecule photosensitizers can be endogenously biosynthesized like protein photosensitizers but are smaller in size and better diffused.
PPIX is a ubiquitous natural porphyrin compound found in living organisms. PPIX absorbs light with a wavelength of 380–650 nm and generates ROS to kill cancer cells112, 113, 114. The utilization of the red light for in vivo stimulation of PPIX is a prevalent practice owing to its better tissue permeability characteristics. Verteporfin, an analogue of PPIX, has been approved by FDA as a PDT reagent. Another photosensitizer for PDT that has obtained approval from the FDA is 5-aminolevulinic acid (5-ALA), which acts as the precursor for PPIX. PPIX can be bio-transformed from 5-ALA via a cascade of enzymatic reactions in tumor cells. After supplementation of 5-ALA, PPIX accumulates preferentially at tumor sites. Under laser irradiation, PPIX produces ROS to eradicate tumor cells. Heme is the downstream product of PPIX, which slows the conversion from 5-ALA to PPIX through negative feedback. To alleviate the inhibitory effect of heme, 5-ALA was co-delivered with a DNA aptamer which self-assembled with heme to form peroxidase mimics115 (Fig. 3E). Combination of PPIX PDT and the effects of peroxidase mimics led to 80% regression of tumors in B16F10-tumor-bearing mice.
Riboflavin116 (Fig. 3F), also known as vitamin B2, is a photosensitive natural product biosynthesized in plants and microorganisms (Fig. 3D). Under 430–440 nm light irradiation, riboflavin emits green fluorescence and transfers energy to O2 or H2O2 producing ROS. PDT using riboflavin as the photosensitizer has been tested in 4T1 cells117, SCC-13 cells118, and HCT116 cells119. A crucial shortcoming of riboflavin photosensitizer is that its excitation wavelength falls in a range with poor penetration in tissues. UCNPs were employed for the purpose of converting NIR radiation into localized UV–blue light, to induce the excitation of riboflavin. NIR-activated PDT using riboflavin led to 90% tumor growth inhibition in SK-BR-3 breast tumor xenografts120. Another factor hindering the PDT performance using riboflavin is its limited cellular uptake. The derivative can enter tumor cells more efficiently by conjugating riboflavin with cell-penetrating peptides, thus improving the PDT outcomes121.
In addition to the widely studied biosynthesized photosensitizers PPIX and riboflavin, pterin derivatives and tryptophan derivatives have also been investigated as potential endogenous photosensitizers. Pterins are endogenous photosensitizers that are ubiquitous in all domains of life. They produce singlet oxygen under UV light irradiation. Pterin derivatives have been successfully applied to PDT treatment of pancreatic cancers122. The endogenously synthesized tryptophan-derived photoproduct, 6-formylindolo(3,2-b) carbazole, has been employed in PDT for the eradication of skin cancer cells123,124.
4. Solutions to hypoxia in PDT
Hypoxia is an intrinsic feature of TME that makes tumors resistant to conventional antitumor treatments including chemotherapy, radiotherapy, and PDT125,126. For PDT, especially Type II PDT, the availability of O2 around photosensitizers affects the yield of cytotoxic ROS. Photodynamic reactions result in the depletion of O2, which worsens the issue of hypoxia and ultimately hampers the efficacy of PDT4. Various approaches have been employed to mitigate the hypoxia-related constraints encountered during PDT.
4.1. Non-enzymatic approach
Delivery of exogenous O2 to tumors is a direct approach to alleviating tumor hypoxia. Hyperbaric oxygen therapy increases the level of dissolved O2 in plasma, thereby increasing the amount of O2 transported to tumors and improving the ROS production in PDT127,65. Hemoglobins can bind O2 and therefore have been combined with PDT reagents to deliver O2 for PDT enhancement128.
H2O2 is more abundant in tumors compared to normal tissues, at concentrations of ∼50–100 μmol/L, which can be decomposed to provide O2 in situ. Gold, platinum, and manganese dioxide nanoparticles have been explored as nanozymes to catalyze the transformation of H2O2 to O2. These nanozymes have demonstrated the capability to alleviate tumor hypoxia while augmenting the efficacy of PDT129. Hemin, an iron-containing porphyrin compound, exhibits catalase-like characteristics that enable it to facilitate the breakdown of endogenous H2O2. Hemin has been co-delivered with Ce6 by DNA aptamer nanopolymers130 (Fig. 4A), M1 macrophage vesicles131, and nanographene oxide132 for PDT treatments. Hemin could alleviate the hypoxia via breaking down H2O2 both in vitro and in vivo, significantly improving antitumor PDT performance133. Hemin can be converted to the photosensitizer PPIX after losing the bound iron, and thus hemin can be a photosensitizer precursor as well. A thermally carbonized hemin nanoparticle was prepared to function as the catalase-like nanozyme and photosensitizer. The nanoparticle acted as a catalyst in the conversion of H2O2 into O2 and ⋅OH through Fenton reactions, thereby alleviating hypoxic conditions and generating ROS. Due to the high temperature during preparation, hemin lost the iron ion bound to the porphyrin ring, resulting in a porphyrin derivative that could generate 1O2 upon photoactivation. The nanoparticle derived from hemin demonstrated enhanced inhibitory efficacy against 4T1 breast cancer cells both in vitro and in vivo, surpassing the inhibitory effects of conventional PDT134.
Figure 4.
Examples of PDT that employ strategies to alleviate hypoxia in tumor microenvironment. (A) A coordination polymer is formed using Ca2+ and AS1411 aptamer, with Ce6 and hemin incorporated. Hemin alleviates hypoxia at tumors, improving the performance of PDT mediated by Ce6 (Bcl-2: B-cell lymphoma 2). Reproduced with permission from Ref. 130. Copyright © 2018 ACS publications. (B) GOX, CAT, and photosensitizers are encapsulated with the cancer cell targeting tripeptide Arg-Gly-Asp in nanocarriers to enhance the uptake and accumulation at tumor sites. GOX and CAT generate oxygen from glucose via cascade reactions, alleviating hypoxia at tumor sites. (GOX: glucose oxidase, CAT: catalase). Reproduced with permission from Ref. 137. Copyright © 2022 Elsevier. (C) Nanoparticles containing catalase and MB are coated with BHQ-3 loaded PLGA to quench excited photosensitizers. The catalase reacts with H2O2 present in the tumor microenvironment (TME) to generate O2, which disrupts the PLGA shell leading to the release of BHQ-3 to activate PDT. Additionally, the O2 attenuates hypoxia in TME to enhance PDT performance (MB: methylene blue, BHQ-3: black hole quencher-3, c(RGDfK): pentapeptide targeting cancer cells). Reproduced with permission from Ref. 138. Copyright © 2015 ACS publications. (D) Bacterial outer membrane vesicles are used as carriers for the co-delivery of Ce6 and catalases (OMV-aPDL1: PD-L1 antibody modified-attenuated Salmonella outer membrane vesicles). Reproduced with permission from Ref. 143. Copyright ©2022 DovePress. (E) A pH-responsive nanomedicine, comprising Ce6 core, pDNA-cat, and PEG shell, targets tumor cells and mediates effective PDT and antitumor immunity. Catalase expressed from pDNA-cat alleviates hypoxia and enhances ROS generation (pDNA-cat: plasmid expressing catalase). Reproduced with permission from Ref. 144. Copyright © 2022 Elsevier.
4.2. Enzymatic approach using catalase
Catalase, a natural protein, functions as a catalyst for the enzymatic breakdown of H2O2 into water and oxygen. Its application presents a potential strategy for ameliorating hypoxia within tumors. Intertumoral vascular heterogeneity hampers the diffusion of catalase proteins, therefore various delivery vehicles have been constructed to deliver purified catalase proteins into tumors135. For example, catalase and alginate dialdehyde were assembled onto CaCO3 nanoparticles with RB loaded as the photosensitizer. Catalase decomposed H2O2 to produce O2 in situ, increasing O2 concentration and promoting the production of 1O2, thus enhancing the efficacy of PDT against MCF-7 breast cancer cells. This early work demonstrated that effective delivery of active catalase proteins can be achieved using nanocarriers and can facilitate PDT treatment136. The diffusion distance of 1O2 in cells is limited, and thus precise delivery of catalases to the treatment sites is helpful. The cancer cell targeting tripeptide Arg-Gly-Asp137 (Fig. 4B) and cyclic pentapeptide Arg-Gly-Asp-D-Phe-Lys138 (Fig. 4C) have been encapsulated with catalase proteins in nanocarriers to enhance the uptake and accumulation of catalases at tumor sites. TME responsive delivery vehicles have also been constructed to specifically transport catalases to tumors. Nanomaterials, such as chitosan139, CaCO3140, and metal–organic frameworks141, that are responsive to low pH can disintegrate more rapidly in the acidic TME to release the encapsulated catalases to tumor cells. Catalase proteins coated by cancer cell membrane can be protected from attacks by immune cells and their circulation time in blood can be prolonged to ensure that catalases are effectively retained in tumors142. Bacterial outer membrane vesicles have also been explored as carriers for catalases due to the multiple bacterial components capable of modulating and stimulating tumor immune responses. When bacterial outer membrane vesicles were used to deliver catalases and photosensitizers, PDT was enhanced by the O2 generated by catalases and synergized with immunotherapy induced by the components from bacteria143 (Fig. 4D).
Despite all sorts of vehicles, the catalase proteins accumulated in tumors are much less compared to the total administered dose. Efforts have been undertaken to enhance the accumulation of catalases within tumors by delivering the catalase gene and subsequently expressing the protein in situ at specific tumor locations. A pH sensitive nanomedicine was constructed to internalize the photosensitizer Ce6 and plasmids hosting catalase gene into tumor cells144 (Fig. 4E). The catalase gene expression was driven by a cytomegalovirus (CMV) promoter. Intratumoral injection of the nanomedicine to 4T1 tumor-bearing mice extended their survival to 46 days, compared with less than 31 days when not supplied with catalase plasmids. Delivery of catalase gene instead of purified proteins solved the problem of low catalase delivery efficiency.
Catalases require endogenous H2O2 as the substrate to generate O2. As the reaction catalyzed by catalase will rapidly consume H2O2, gradually no more O2 will be produced therefore the tumor hypoxia is only temporarily relieved. Other alternative strategies have been explored, for instance, the rate of O2 uptake by tumor cells can be reduced by inhibiting the mitochondrial respiration145,146, or photosynthetic microbes that are capable of producing O2 can be used in combination with PDT reagents.
5. Bacteria in PDT
Bacteria including Streptococcus, Salmonella, Escherichia, Clostridium, Bifidobacterium, and Lactococcus have been explored for use in antitumor treatment21,147. Poor vascularization at tumor sites hinders the effective delivery of antitumor drugs and conventional nanomaterial drug carriers148 Anaerobic or facultative anaerobic bacteria can migrate from the bloodstream to tumor areas due to their self-propulsion and preference for the hypoxic TME149,150. Bacteria administered intratumorally or intravenously accumulate substantially more in tumors than in other tissues151. This high targeting ability to tumor tissue makes bacteria promising delivery vehicles for antitumor reagents152. Xiao et al.147 developed a biohybrid antitumor platform from the anaerobic bacterium Bifidobacterium infantis and DOX-loaded bovine serum albumin nanoparticles. The biohybrid exhibited a 4-fold higher cumulative concentration of DOX at the tumor site than in mice treated with drugs not carried by the bacteria. After treatment, the treated group exhibited significantly reduced tumor volume compared to the control groups, and the survival duration of the mice was notably extended (Fig. 5A). Clostridium butyricum has been investigated as an oncolytic bacterium for use in antitumor therapy. It has recently been engineered with an aggregation-induced emission photosensitizer (TPApy) to improve the melanoma treatment. TPApy was incorporated into the peptidoglycan of bacteria and released upon bacterial death. The engineered C. butyricum colonized and proliferated in the hypoxic TME of melanoma, ablating the tumor. Hypoxia was attenuated as the ablation continued, which killed the obligate anaerobe C. butyricum, leading to the release of photosensitizers and ensuing PDT. PDT effectively treated the marginal areas of the tumor and helped achieve complete ablation of malignant melanoma153 (Fig. 5B).
Figure 5.
Examples of PDT using engineered bacteria and schematic representation endogenous production of PDT components by genetically modified bacteria. (A) Bifidobacterium infantis is employed as a carrier to transport DOX to breast tumors. B. infantis loaded with DOX containing nanoparticles targets the hypoxic regions of tumors and increases the drug accumulation, leading to a prolonged survival of tumor-bearing mice (Bif: Bifidobacterium infantis, DOX: doxorubicin). Reproduced with permission from Ref. 147. Copyright © 2022 Springer Nature. (B) C. butyricum is labeled with AIE photosensitizer modified d-Alanine. The labeled bacteria produce singlet oxygen to eliminate cancer cells under light irradiation (AIE: aggregation-induced emission). Reproduced with permission from Ref. 153. Copyright © 2022 Wiley. (C) S. typhimurium is transformed with a plasmid expressing FLuc, which emits bioluminescence in the presence of luciferin serving as the light source for PDT. The engineered bacteria additionally stimulate the immune response to enhance the effectiveness of the antitumor therapy (TAAs: tumor associated antigens, DAMPs/PAMPs: damage/pathogen associated molecular pattern). Reproduced with permission from Ref. 161. Copyright © 2022 Elsevier. (D) S. elongatus hybridized with photosensitizers Ce6 or PPIX exhibit enhanced PDT efficiency due to its capability to produce O2in situ. Reproduced with permission from Ref. 164. Copyright ©Copyright 2020 Wiley. (E) ICG-encapsulated nanoparticles are attached to S. elongatus via amide bonds. Photosynthesis-boosted PDT is achieved using this platform. Reproduced with permission from Ref. 166. Copyright © 2020 Wiley. (F) S. elongatus integrated with UCNPs can convert NIR light to visible light to induce photosynthesis in S. elongatus and to excite Ce6 for PDT. Reproduced with permission from Ref. 168. Copyright © 2021 Springer Nature. (G) Chlorella is co-delivered with perfluorocarbon nanoparticles loaded with Ce6. Upon exposure to laser light, the O2 produced by Chlorella can be absorbed by the nanoparticles to enhance the antitumor PDT (C-Gel: Chlorella-gel, PFC: perfluorocarbon). Reproduced with permission from Ref. 170. Copyright © 2021 Elsevier.
Furthermore, apart from the tumor-targeting ability, the immunogenic nature of bacteria further synergizes to bolster the antitumor effects154. Components of bacteria, such as peptidoglycan and lipopolysaccharide, can be recognized by pattern recognition receptors expressed on dendritic cells, leading to the subsequent initiation of an immune response. For example, nanomedicines encapsulated within outer membrane vesicles derived from Gram-negative bacteria demonstrate enhanced immunostimulatory potential, leading to significant suppression of tumor growth and metastasis155. Synthetic biology tools can genetically modify bacteria to meet the needs of customized antitumor therapies. For instance, virulence factors of bacteria may cause damage and therefore need to be addressed carefully. Attenuated E. coli and Salmonella strains have been constructed by deleting the msbB virulence factor gene, which is responsible for the myristoylation of lipid A. When used for antitumor therapy, such genetically engineered strains are less virulent but retain the ability to colonize tumor sites156. Genetic tools can also empower bacteria with new functionalities that can be used to develop precision medicines. With delicate design, engineered bacteria are capable of biosynthesizing small molecules and proteins with therapeutic activity. Bacteria expressing cytokines and interferons, such as TNFα, IL-2, CCL21, and IFNs, have been constructed for antitumor therapy, demonstrating promising outcomes157, and when combined with PDT can lead to synergistic results158.
There have been several reviews discussing the application of bacteria in antitumor treatments. In the reviewed applications, bacteria are primarily used as drug carriers or therapeutic factories. For detailed reviews on this topic, see Refs. 149, 159 and 160. In this section, we discuss the potential of engineered bacteria as a solution to the limitations of PDT through the endogenous production of light, photosensitizers, and oxygen.
5.1. Endogenous production of light by bacteria
PDT activated by bioluminescence has demonstrated potency in treating deep tumors. Luciferase is a genetically encodable protein that can be heterologously expressed in bacteria. Yang et al.161 recently transformed a plasmid containing the FLuc gene into an attenuated S. typhimurium to construct a bioluminescent strain Luc-S.T.ΔppGpp (Fig. 5C). Bioluminence was enriched in tumors after intratumoral administration of Luc-S.T.ΔppGpp along with luciferin. The bioluminescence generated at tumor sites can activate photosensitizer Ce6 to produce ROS, effectively inhibiting the growth of CT26 cancer cells in mice. The efficacy of PDT activated by bioluminescent Luc-S.T.ΔppGpp surpassed that of PDT activated by externally applied high-energy LED illumination. No evidence of tumor recurrence was observed within a 60-day period. The frequency of effector memory T cells in treated mice was twice as high as in healthy mice, indicating that the treatment induced a potent immune memory effect that could prevent tumor recurrence. Apart from FLuc, alternative luciferases, namely RLuc and NLuc, also hold potential for expression in genetically engineered bacteria.
5.2. Endogenous production of photosensitizers by bacteria
Endogenously biosynthesized photosensitizers can overcome the shortcomings in applications of exogenous chemically synthesized photosensitizers, such as low uptake efficiency and off-target toxicity. Genetically engineered bacteria expressing photosensitizers can swim to tumors and accumulate photosensitizers there. Such bacteria are similar to weapons that can generate explosives automatically and strike targets precisely. Yan et al.162 constructed an engineered bacterium (KR-E. coli) by expressing photosensitizer KillerRed in E. coli. KR-E. coli injected intratumorally or intravenously targeted and proliferated at tumor sites. Within one day, the fluorescence from KillerRed covered the entire tumor, demonstrating that KillerRed was abundantly expressed in the tumor. Upon light irradiation, the KR-E. coli-treated mice underwent efficient PDT, leading to the complete disappearance of tumor within a few days. No tumor recurrence was observed within 2 months. In addition to protein photosensitizers, small-molecule photosensitizers widely used in PDT, such as hypericin and riboflavin, are natural products that can be biosynthesized. Recently, studies have demonstrated the production of hypericin in repurposed Cladosporium fulvum. The gene claM was deleted to accumulate emodin, which is the precursor for hypericin biosynthesis. Heterologous expression of RugG, a substrate-promiscuous fungal P450 monooxygenase, in the mutant strain led to the production of emodin bianthrone derivatives, which can be transformed to hypericin via subsequent nonenzymatic reactions. The engineered C. fulvum strain was capable of biosynthesizing hypericin with a titer of 43.1 mg/L163. We envision that a similar genetic circuit can be constructed in bacteria to produce hypericin inside the tumor for PDT.
5.3. Endogenous production of oxygen by bacteria
Cyanobacteria are prokaryotes that can perform photosynthesis to generate O2. Application of Synechococcus elongatus PCC 7942 (S. elongatus) in PDT with photosensitizer Ce6164 (Fig. 5D) and RB165 demonstrated enhanced production of 1O2 and improved treatment outcomes. S. elongatus has also been combined with nanosized photosensitizers for antitumor PDT17. For example, S. elongatus was engineered by attaching ICG-encapsulated nanoparticles to its surface via amide bonds166 (Fig. 5E). Following the intravenous administration of the engineered bacteria, mice were subjected to illumination using a 660 nm laser to facilitate photosynthesis, and an 808 nm laser to excite ICG. S. elongatus produced O2 under 660 nm irradiation, resulting in the production of more ROS, which completely eliminated the primary tumor. This photosynthesis-boosted PDT not only effectively reverses the hypoxic TME but also strongly triggers a systemic antitumor immune response that prevents tumor relapse and suppresses metastatic progression in murine models of metastatic triple-negative breast cancer. By integrating S. elongatus with UCNPs, the biohybrid can harvest energy from NIR light167. Under NIR irradiation, UCNPs converted NIR light to visible light to induce photosynthesis in S. elongatus and to excite Ce6 for PDT168 (Fig. 5F). Arthrospira platensis (also named as Spirulina platensis) is another cyanobacterium that is biocompatible, safe, and is commercially available as a nutritional supplement. Biohybrids of A. plantensis were constructed by modifying the bacterial cells with magnetic Fe3O4 nanoparticles169. The biohybrids could be guided by magnetic field to target tumors precisely and generated O2 in situ to boost the antitumor PDT effects. No long-term toxicity was observed after the treatment using A. plantensis biohybrids. In addition to cyanobacteria, Chlorella is a genus of single-celled algae that has been developed as O2 suppliers for PDT. Chlorella has a size of 2–10 μm and is capable of photosynthesis. Wang et al.170 established a sustainable PDT platform using Chlorella (the species was not disclosed in the paper) and perfluorocarbon nanoparticles (Fig. 5G). Alginate-encapsulated Chlorella cells were injected into mice harboring CT26 colon cancer in order to generate O2 in response to 660 nm light exposure. Perfluorocarbon nanoparticles that can absorb O2 were used to load and deliver Ce6. With the photosynthesis of Chlorella, the killing rate of tumor cells was enhanced by approximately 63%. Besides O2 producers, Chlorella cells also simulated antitumor immune responses by promoting dendritic cell activation.
In addition to photosynthetic bacteria, other bacteria can be genetically modified to acquire the capability to produce O2 in tumors. As discussed in section 4.2, the catalase enzyme can breakdown endogenous H2O2 at tumor sites to produce O2. Catalase enzymes can be hydrolyzed and cleared by human proteases during delivery, thereby requiring the use of nanocarriers for additional protection. In situ production of catalase enzymes at tumor sites by genetically engineered bacteria helps to solve the stability issue of enzymes during transport. E. coli was engineered by transforming a plasmid containing the catalase gene and further loaded with polydopamine-encapsulating Ce6. Under irradiation, Ce6 was excited to produce ROS, and the PDT performance was enhanced by the O2 produced by the catalase enzyme expressed in E. coli. The system effectively treated malignant tumors and exhibited no significant biotoxicity171. In another similar system, Ce6 was replaced by black phosphorous-derived photosensitizing agents. The catalase enzyme expressed by E. coli enhanced the antitumor PDT mediated by black phosphorous as well172. In these examples, catalase production was achieved via a plasmid-based expression system. The plasmid relied on antibiotic selection to persist within bacterial cells, with the potential for loss over time in the absence of antibiotics. To ensure a sustainable and enduring therapeutic approach, it will be essential to integrate the gene cassette expressing and regulating catalase into the bacterial genome.
6. Conclusions and outlook
PDT is an extensively investigated non-invasive therapy for cancer, involving tripartite fundamental components: excitation light, photosensitizers, and oxygen. Photosensitizers necessitate excitation through light of a specific wavelength and subsequent interaction with O2 to produce ROS. PDT has been a clinically approved treatment option for over three decades, first gaining approval for use in treating bladder cancer. Since then, PDT has demonstrated its efficacy in treating diverse cancers such as breast, head and neck, pancreatic, cervical, and brain cancers. PDT's foremost advantage lies in its non-invasiveness, setting it apart from conventional tumor treatments. For instance, sunlight-based PDT has been approved as an actinic keratosis treatment in Europe. Furthermore, PDT can complement existing therapies, enhancing their effectiveness. PDT has been approved for advanced head and neck squamous cell carcinoma patients when other treatment options are not feasible. Despite its success in numerous clinical trials, PDT has yet to realize its full potential in current clinical practice and is not regarded as the primary standard-of-care treatment for most cancer types. Effective implementation of antitumor PDT encounters diverse obstacles, such as penetration limitations of external light sources, low delivery efficiency and off-target effects of photosensitizers, poor tumor distribution of photosensitizers, and lack of O2 in the hypoxic TME. These obstacles collectively lead to a prominent issue faced by PDT in clinical practice: PDT alone often yields only partial and incomplete tumor destruction, resulting in tumor regrowth and progression. Overcoming this challenge necessitates a multifaceted approach involving the resolution of the aforementioned obstacles and the integration of PDT with complementary treatment modalities.
These obstacles have been the main focus of studies aimed at enhancing the effectiveness of antitumor PDT. For example, X-ray has been used as the excitation light for PDT, which has better tissue penetration but is more restrictive in terms of dose usage. Upconversion nanoparticles can convert external NIR light to localized UV–Vis light for excitation of photosensitizers, but synthesizing them on a large scale is generally challenging and expensive. For the improvement of photosensitizers, different generations of photosensitizers have been developed to enhance their toxicity and solubility profiles. Inorganic and organic nanomaterial delivery vehicles have also been constructed to deliver more photosensitizers to tumors in a targeted manner. Aggregation-induced emission (AIE) photosensitizers represent a class of chemical compounds that have higher photostability and can overcome the aggregation-induced quenching commonly encountered by conventional photosensitizers. Notably, AIE photosensitizers with Type I PDT mechanism have been developed, exhibiting enhanced efficiency within hypoxic tumors173. In order to mitigate hypoxia within tumors, a commonly employed approach entails the administration of catalase enzymes to tumor locations to facilitate the breakdown of endogenous H2O2 into O2. Nanoparticles with catalase-like activity, including gold nanoclusters and MnO2 nanoparticles, can generate O2 in situ to improve PDT performance.
These approaches have shown promising outcomes, however, the three components for PDT still need to be delivered externally, which discourages sustainable treatment and often requires multiple doses to improve efficacy. In this review, we examine the advancements made in an alternative strategy involving the internal production of light, photosensitizers, and oxygen (Table 1). Our emphasis lies in using bioluminescence as the excitation light source, employing encodable proteins and small molecules as photosensitizers, and using enzymes or photosynthetic microbes as sources of oxygen. Bioluminescence can act as an endogenous light source that does not require external light and thus resolves the problem of light transmission. The most commonly used bioluminescence in PDT is produced by firefly luciferases. The luciferase gene was delivered to tumor cells in one shot, and the protein could be expressed thereafter under the control of promoters for ongoing treatment. Similarly, tumor cells were transfected with the gene encoding the catalase enzyme, resulting in the expression of the catalase enzyme that produced O2 endogenously and continuously. Genetically encodable photosensitizers such as KillerRed can be expressed endogenously at tumor sites as well. For example, a plasmid carrying the KillerRed gene was transfected into HEK293 cells with high efficiency. KillerRed protein was expressed and produced ROS under green light, inducing the apoptosis of tumor cells104. This approach avoids problems associated with photosensitizer delivery efficiency and enables continuous in situ production of photosensitizers, thus not requiring repeated drug administration. The excitation spectra of common endogenous photosensitizers and emission spectra of bioluminescence are summarized in Fig. 6.
Table 1.
Recent examples of treatments involving biogenesis of PDT components.
| Type | Photosensitizer source | Light source | Oxygen source | Cell type | Ref. |
|---|---|---|---|---|---|
| Biogenesis of photosensitizer | MiniSOG expressed from lentiviral vector in HeLa cells | External 465 nm laser | – | Cervical cancer (HeLa cells) | 101 |
| MiniSOG expressed from lentiviral vector in SK-BR-3 cells | Bioluminescence of NanoLuc | – | Breast cancer (SK-BR-3 cells) | 102 | |
| MiniSOG expressed from lentiviral vector lentiviral in BT-474 cells | Bioluminescence of NanoLuc | – | Breast cancer (BT-474 cells) | 103 | |
| MiniSOG expressed from plasmid in E. coli | External 465 nm laser | – | Breast cancer (SK-BR-3) | 96 | |
| MiniSOG expressed from plasmid in E. coli | External 458 nm laser | – | Breast cancer (SK-BR-3 cells) | 97 | |
| KillerRed expressed from adenovirus vector in H1975 cells | External 561 nm laser | – | Lung cancer (H1975 cells) | 98 | |
| KillerRed expressed from adenovirus vector in H1299, HCT116, or HT29 cells | External green light | – | Lung cancer and colon cancer (H1299, HCT116, HT29 cells) | 99 | |
| KillerRed expressed from adenovirus vector in MCF7 cells | External 561 nm laser | – | Breast cancer (MCF7 cells) | 100 | |
| KillerRed expressed from plasmid in 4T1 cells | External 532 nm laser | – | Breast cancer (4T1 cells) | 95 | |
| KillerRed expressed from plasmid in E. coli | External 400–780 nm white light | – | Leukemia (K562, NB4, THP1 cells) | 93 | |
| KillerRed expressed from plasmid in E. coli | External 586 nm laser | – | Breast cancer (MDA-MB-231 cells) | 94 | |
| KillerRed expressed from plasmid in E. coli | External green light | – | Ovarian cancer (SKOV-3 cells) | 81 | |
| KillerRed protein delivered in DNA nanostructures | External 610 nm laser | – | Cervical cancer (HeLa cells) | 108 | |
| Biogenesis of light | Hypericin | Firefly luciferase expressed from plasmid in HeLa cells | – | Cervical cancer (HeLa cells) | 55 |
| Rose Bengal | Firefly luciferase expressed from plasmid in 3T3 cells | – | Embryonic fibroblast (3T3 cells) | 52 | |
| Chlorin e6 | Firefly luciferase expressed from plasmid in Salmonellatyphimurium | – | Colon cancer (CT26 cells) | 161 | |
| KillerRed | Renilla luciferase expressed from plasmid in E. coli | – | Breast cancer (MCF7 cells) | 59 | |
| Rose Bengal | Renilla luciferase expressed from plasmid in E. coli | – | Colon cancer (CT26 cells) | 58 | |
| MiniSOG | NanoLuc expressed from a plasmid in E. coli | – | Breast cancer (SK-BR-3 cells) | 62, 63, 64 | |
| Biogenesis of O2 | Chlorin e6 | External 660 nm laser | Photosynthetic Cyanobacteria | Breast cancer (4T1 cells) | 164 |
| Rose Bengal | External 640 nm laser | Photosynthetic Synechococcus elongatus | Breast cancer (4T1 cells) | 165 | |
| Chlorophyll | External 650 nm laser | Photosynthetic Spirulina platensis | Breast cancer (4T1 cells) | 169 | |
| Chlorin e6 | External 660 nm laser | Photosynthetic Chlorella | Colon cancer (CT26 cells) | 170 |
Figure 6.
Schematic diagram showing the excitation spectra of photosensitizers and emission spectra of bioluminescence.
Bacteria have been extensively explored in antitumor therapy owing to their capacity for targeting tumors and eliciting immune responses. Using synthetic biology tools, bacteria can be further engineered to endogenously produce bioluminescence, photosensitizers, and oxygen inside tumors. Bacteria transformed with a plasmid carrying the FLuc gene were successfully applied to in vivo antitumor treatment. Bioluminescence was generated in the presence of luciferin to activate photosensitizers. Such a system requires the luciferase substrates to be supplied externally. Some luciferase substrates have solubility and off-target effect issues, and require multiple doses to enhance therapeutic efficacy. Subsequent efforts can be directed towards the advancement of self-sufficient PDT systems by genetically encoding both luciferase and luciferin, thereby negating the need for exogenous substrates.
Attempts have been performed to heterologously express protein photosensitizers in bacteria for antitumor PDT. Engineered bacteria expressing KillerRed enhanced the accumulation of photosensitizers in tumors and eliminated tumor cells in vivo. Further investigation is needed to explore the excretion and localization of photosensitizers produced by bacteria within tumor sites. Tagging of localization peptide sequences to such protein photosensitizers can help to better target them to tumor cells. The toxicity of endogenous photosensitizers within bacterial cells also needs to be carefully monitored to prevent premature death of bacterial cells prior to treatment. There have been no studies on the employment of engineered bacteria producing small molecule photosensitizers in antitumor PDT. With advances in synthetic biology, heterologous expression of natural products in bacteria is feasible. Small-molecule photosensitizers such as hypericin have been expressed in engineered microbes. In the future, the use of bacteria to produce small-molecule photosensitizers in situ will provide an alternative strategy for the endogenous production of protein photosensitizers.
Endogenous production of light, photosensitizers, and oxygen is a promising strategy to enhance antitumor PDT. As a widely studied antitumor drug delivery vehicle, bacteria can undergo genetic engineering to form an integrated platform for the internal production of the three components of PDT at tumor sites (Fig. 7). This would eliminate the need for exogenous light and ensure a continuous supply of photosensitizers and oxygen during PDT. The rich synthetic biology toolbox offers a wealth of genetic parts and devices to support the design and construction of such bacteria. To maximize the generation of ROS, careful programming of the production and distribution of the three components is crucial. Nonetheless, issues such as engineered bacteria–host interactions, as well as biosafety risks, must be prudently considered. Despite these challenges, the use of engineered bacteria holds great promise for overcoming current limitations of antitumor PDT.
Figure 7.
Schematic representation of the PDT treatment process mediated by the light, photosensitizer and oxygen simultaneously generated by genetically engineered bacteria.
In summary, endogenous production of light, photosensitizers, and oxygen is a promising strategy for overcoming the obstacles in Type II PDT. With the continuous progress of synthetic biology, engineered bacteria emerge as valuable allies in realizing this objective.
Author contributions
Conceptualization, Yihan Wu; Data curation, Lin Yu, Zhen Liu; Project administration, Jinliang Liu, Xiaohui Zhu; Validation, Wei Xu, Kai Jin; Writing – original draft, Lin Yu, Zhen Liu; Writing – review & editing, Kai Jin, Lin Yu, Xiaohui Zhu, Yihan Wu, Yong Zhang. All of the authors have read and approved the final manuscript.
Conflicts of interest
The authors declare no conflicts of interest.
Acknowledgements
This work was supported by the National Natural Science Foundation of China (32000036).
Footnotes
Peer review under the responsibility of Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences.
Contributor Information
Yong Zhang, Email: yozhang@cityu.edu.hk.
Yihan Wu, Email: yihanw@shu.edu.cn.
References
- 1.Li X.S., Lovell J.F., Yoon J., Chen X.Y. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat Rev Clin Oncol. 2020;17:657–674. doi: 10.1038/s41571-020-0410-2. [DOI] [PubMed] [Google Scholar]
- 2.Li X.S., Lee S.Y., Yoon J. Supramolecular photosensitizers rejuvenate photodynamic therapy. Chem Soc Rev. 2018;47:1174–1188. doi: 10.1039/c7cs00594f. [DOI] [PubMed] [Google Scholar]
- 3.Celli J.P., Spring B.Q., Rizvi I., Evans C.L., Samkoe K.S., Verma S., et al. Imaging and photodynamic therapy: mechanisms, monitoring, and optimization. Chem Rev. 2010;110:2795–2838. doi: 10.1021/cr900300p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Fan W.P., Huang P., Chen X.Y. Overcoming the Achilles' heel of photodynamic therapy. Chem Soc Rev. 2016;45:6488–6519. doi: 10.1039/c6cs00616g. [DOI] [PubMed] [Google Scholar]
- 5.Deng X.G., Shao Z.W., Zhao Y.L. Solutions to the drawbacks of photothermal and photodynamic cancer therapy. Adv Sci. 2021;8 doi: 10.1002/advs.202002504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zhang Z., Chen Y.M., Zhang Y. Self-Assembly of upconversion nanoparticles based materials and their emerging applications. Small. 2022;18 doi: 10.1002/smll.202103241. [DOI] [PubMed] [Google Scholar]
- 7.Sun W.J., Zhou Z.J., Pratx G., Chen X.Y., Chen H.M. Nanoscintillator-mediated X-ray induced photodynamic therapy for deep-seated tumors: from concept to biomedical applications. Theranostics. 2020;10:1296–1318. doi: 10.7150/thno.41578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chen M.M., Xu C.H., Zhao W., Chen H.Y., Xu J.J. Single cell imaging of electrochemiluminescence-driven photodynamic therapy. Angew Chem Int Ed Engl. 2022;61 doi: 10.1002/anie.202117401. [DOI] [PubMed] [Google Scholar]
- 9.Zhang Y.T., Hao Y.Q., Chen S., Xu M.T. Photodynamic therapy of cancers with internal light sources: chemiluminescence, bioluminescence, and cerenkov radiation. Front Chem. 2020;8:770. doi: 10.3389/fchem.2020.00770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zheng Q.Y., Liu X.M., Zheng Y.F., Yeung K.W.K., Cui Z.D., Liang Y.Q., et al. The recent progress on metal-organic frameworks for phototherapy. Chem Soc Rev. 2021;50:5086–5125. doi: 10.1039/d1cs00056j. [DOI] [PubMed] [Google Scholar]
- 11.Riani Y.D., Matsuda T., Nagai T. Genetically encoded photosensitizer for destruction of protein or cell function. Adv Exp Med Biol. 2021;1293:265–279. doi: 10.1007/978-981-15-8763-4_16. [DOI] [PubMed] [Google Scholar]
- 12.Wan Y.L., Fu L.H., Li C.Y., Lin J., Huang P. Conquering the hypoxia limitation for photodynamic therapy. Adv Mater. 2021;33 doi: 10.1002/adma.202103978. [DOI] [PubMed] [Google Scholar]
- 13.Sun Y.Y., Zhao D.Y., Wang G., Wang Y., Cao L.L., Sun J., et al. Recent progress of hypoxia-modulated multifunctional nanomedicines to enhance photodynamic therapy: opportunities, challenges, and future development. Acta Pharm Sin B. 2020;10:1382–1396. doi: 10.1016/j.apsb.2020.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Xu T., Ma Y.Y., Yuan Q.L., Hu H.X., Hu X.K., Qian Z.Y., et al. Enhanced ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano. 2020;14:3414–3425. doi: 10.1021/acsnano.9b09426. [DOI] [PubMed] [Google Scholar]
- 15.Liu Y.L., Pan Y.X., Cao W., Xia F.F., Liu B., Niu J.Q., et al. A tumor microenvironment responsive biodegradable CaCO3/MnO2-based nanoplatform for the enhanced photodynamic therapy and improved PD-L1 immunotherapy. Theranostics. 2019;9:6867–6884. doi: 10.7150/thno.37586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Qin X., Wu C., Niu D.C., Qin L.M., Wang X., Wang Q.G., et al. Peroxisome inspired hybrid enzyme nanogels for chemodynamic and photodynamic therapy. Nat Commun. 2021;12:5243. doi: 10.1038/s41467-021-25561-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Qi F.G., Ji P.H., Chen Z.X., Wang L.P., Yao H.L., Huo M.F., et al. Photosynthetic cyanobacteria-hybridized black phosphorus nanosheets for enhanced tumor photodynamic therapy. Small. 2021;17 doi: 10.1002/smll.202102113. [DOI] [PubMed] [Google Scholar]
- 18.Lucky S.S., Soo K.C., Zhang Y. Nanoparticles in photodynamic therapy. Chem Rev. 2015;115:1990–2042. doi: 10.1021/cr5004198. [DOI] [PubMed] [Google Scholar]
- 19.Ganai S., Arenas R.B., Sauer J.P., Bentley B., Forbes N.S. In tumors Salmonella migrate away from vasculature toward the transition zone and induce apoptosis. Cancer Gene Ther. 2011;18:457–466. doi: 10.1038/cgt.2011.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Liu N.N., Jiao N., Tan J.C., Wang Z.L., Wu D.F., Wang A.J., et al. Multi-kingdom microbiota analyses identify bacterial-fungal interactions and biomarkers of colorectal cancer across cohorts. Nat Microbiol. 2022;7:238–250. doi: 10.1038/s41564-021-01030-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhou S.B., Gravekamp C., Bermudes D., Liu K. Tumour-targeting bacteria engineered to fight cancer. Nat Rev Cancer. 2018;18:727–743. doi: 10.1038/s41568-018-0070-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hao Y., Chung C.K., Yu Z.F., Huis In 't Veld R.V., Ossendorp F.A., Ten Dijke P., et al. Combinatorial therapeutic approaches with nanomaterial-based photodynamic cancer therapy. Pharmaceutics. 2022;14:120. doi: 10.3390/pharmaceutics14010120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Akasov R., Khaydukov E.V., Yamada M., Zvyagin A.V., Leelahavanichkul A., Leanse L.G., et al. Nanoparticle enhanced blue light therapy. Adv Drug Deliv Rev. 2022;184 doi: 10.1016/j.addr.2022.114198. [DOI] [PubMed] [Google Scholar]
- 24.Alzeibak R., Mishchenko T.A., Shilyagina N.Y., Balalaeva I.V., Vedunova M.V., Krysko D.V. Targeting immunogenic cancer cell death by photodynamic therapy: past, present and future. J Immunother Cancer. 2021;9 doi: 10.1136/jitc-2020-001926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yang M.Y., Yang T., Mao C.B. Enhancement of photodynamic cancer therapy by physical and chemical factors. Angew Chem Int Ed Engl. 2019;58:14066–14080. doi: 10.1002/anie.201814098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pham T.C., Nguyen V.N., Choi Y., Lee S., Yoon J. Recent strategies to develop innovative photosensitizers for enhanced photodynamic therapy. Chem Rev. 2021;121:13454–13619. doi: 10.1021/acs.chemrev.1c00381. [DOI] [PubMed] [Google Scholar]
- 27.Luby B.M., Walsh C.D., Zheng G. Advanced photosensitizer activation strategies for smarter photodynamic therapy beacons. Angew Chem Int Ed Engl. 2019;58:2558–2569. doi: 10.1002/anie.201805246. [DOI] [PubMed] [Google Scholar]
- 28.Vankayala R., Hwang K.C. Near-infrared-light-activatable nanomaterial-mediated phototheranostic nanomedicines: an emerging paradigm for cancer treatment. Adv Mater. 2018;30 doi: 10.1002/adma.201706320. [DOI] [PubMed] [Google Scholar]
- 29.Brancaleon L., Moseley H. Laser and non-laser light sources for photodynamic therapy. Laser Med Sci. 2002;17:173–186. doi: 10.1007/s101030200027. [DOI] [PubMed] [Google Scholar]
- 30.Xie J.L., Wang Y.W., Choi W.S., Jangili P., Ge Y.Q., Xu Y.J., et al. Overcoming barriers in photodynamic therapy harnessing nano-formulation strategies. Chem Soc Rev. 2021;50:9152–9201. doi: 10.1039/d0cs01370f. [DOI] [PubMed] [Google Scholar]
- 31.Zhu T.C., Sun H.J., Ong Y.H., Morales R.H., Dimofte A., Busch T., et al. Real-time PDT dose dosimetry for pleural photodynamic therapy. Proc SPIE-Int Soc Opt Eng. 2022;11940 doi: 10.1117/12.2612188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Bansal A., Yang F.Y., Xi T., Zhang Y., Ho J.S. In vivo wireless photonic photodynamic therapy. Proc Natl Acad Sci U S A. 2018;115:1469–1474. doi: 10.1073/pnas.1717552115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Tian G., Gu Z.J., Zhou L.J., Yin W.Y., Liu X.X., Yan L., et al. Mn2+ dopant-controlled synthesis of NaYF4:Yb/Er upconversion nanoparticles for in vivo imaging and drug delivery. Adv Mater. 2012;24:1226–1231. doi: 10.1002/adma.201104741. [DOI] [PubMed] [Google Scholar]
- 34.Zhang P., Steelant W., Kumar M., Scholfield M. Versatile photosensitizers for photodynamic therapy at infrared excitation. J Am Chem Soc. 2007;129:4526–4527. doi: 10.1021/ja0700707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Idris N.M., Jayakumar M.K., Bansal A., Zhang Y. Upconversion nanoparticles as versatile light nanotransducers for photoactivation applications. Chem Soc Rev. 2015;44:1449–1478. doi: 10.1039/c4cs00158c. [DOI] [PubMed] [Google Scholar]
- 36.Idris N.M., Gnanasammandhan M.K., Zhang J., Ho P.C., Mahendran R., Zhang Y. In vivo photodynamic therapy using upconversion nanoparticles as remote-controlled nanotransducers. Nat Med. 2012;18:1580–1585. doi: 10.1038/nm.2933. [DOI] [PubMed] [Google Scholar]
- 37.Qiao X.F., Zhou J.C., Xiao J.W., Wang Y.F., Sun L.D., Yan C.H. Triple-functional core-shell structured upconversion luminescent nanoparticles covalently grafted with photosensitizer for luminescent, magnetic resonance imaging and photodynamic therapy in vitro. Nanoscale. 2012;4:4611–4623. doi: 10.1039/c2nr30938f. [DOI] [PubMed] [Google Scholar]
- 38.Gnanasammandhan M.K., Idris N.M., Bansal A., Huang K., Zhang Y. Near-IR photoactivation using mesoporous silica-coated NaYF4:Yb,Er/Tm upconversion nanoparticles. Nat Protoc. 2016;11:688–713. doi: 10.1038/nprot.2016.035. [DOI] [PubMed] [Google Scholar]
- 39.Zhang Z., Jayakumar M.K.G., Zheng X., Shikha S., Zhang Y., Bansal A., et al. Upconversion superballs for programmable photoactivation of therapeutics. Nat Commun. 2019;10:4586. doi: 10.1038/s41467-019-12506-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Teh D.B.L., Bansal A., Chai C., Toh T.B., Tucker R.A.J., Gammad G.G.L., et al. A flexi-PEGDA upconversion implant for wireless brain photodynamic therapy. Adv Mater. 2020;32 doi: 10.1002/adma.202001459. [DOI] [PubMed] [Google Scholar]
- 41.Isikawa M., Guidelli E. Microfluidic synthesis of theranostic nanoparticles with near-infrared scintillation: toward next-generation dosimetry in X-ray-induced photodynamic therapy. ACS Appl Mater Interfaces. 2022;14:324–336. doi: 10.1021/acsami.1c20689. [DOI] [PubMed] [Google Scholar]
- 42.Micheletto M.C., Guidelli É J., Costa-Filho A.J. Interaction of genetically encoded photosensitizers with scintillating nanoparticles for X-ray activated photodynamic therapy. ACS Appl Mater Interfaces. 2021;13:2289–2302. doi: 10.1021/acsami.0c19041. [DOI] [PubMed] [Google Scholar]
- 43.Wang Q., Liu N., Hou Z.Y., Shi J.P., Su X.H., Sun X.L. Radioiodinated persistent luminescence nanoplatform for radiation-induced photodynamic therapy and radiotherapy. Adv Healthcare Mater. 2021;10 doi: 10.1002/adhm.202000802. [DOI] [PubMed] [Google Scholar]
- 44.Ni D., Ferreira C.A., Barnhart T.E., Quach V., Yu B., Jiang D., et al. Magnetic targeting of nanotheranostics enhances cerenkov radiation-induced photodynamic therapy. J Am Chem Soc. 2018;140:14971–14979. doi: 10.1021/jacs.8b09374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kotagiri N., Sudlow G.P., Akers W.J., Achilefu S. Breaking the depth dependency of phototherapy with cerenkov radiation and low-radiance-responsive nanophotosensitizers. Nat Nanotechnol. 2015;10:370–379. doi: 10.1038/nnano.2015.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shah N., Squire J., Guirguis M., Saha D., Hoyt K., Wang K.K., et al. Deep-tissue activation of photonanomedicines: an update and clinical perspectives. Cancers. 2022;14:2004. doi: 10.3390/cancers14082004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Xu X.Q., An H.J., Zhang D.L., Tao H., Dou Y., Li X.H., et al. A self-illuminating nanoparticle for inflammation imaging and cancer therapy. Sci Adv. 2019;5 doi: 10.1126/sciadv.aat2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yang Y.K., Hou W.Y., Liu S.Y., Sun K., Li M.Y., Wu C.F. Biodegradable polymer nanoparticles for photodynamic therapy by bioluminescence resonance energy transfer. Biomacromolecules. 2018;19:201–208. doi: 10.1021/acs.biomac.7b01469. [DOI] [PubMed] [Google Scholar]
- 49.Kim Y.R., Kim S., Choi J.W., Choi S.Y., Lee S.H., Kim H., et al. Bioluminescence-activated deep-tissue photodynamic therapy of cancer. Theranostics. 2015;5:805–817. doi: 10.7150/thno.11520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Li S.F., Ruan Z.Y., Zhang H., Xu H.W. Recent achievements of bioluminescence imaging based on firefly luciferin-luciferase system. Eur J Med Chem. 2021;211 doi: 10.1016/j.ejmech.2020.113111. [DOI] [PubMed] [Google Scholar]
- 51.Carpenter S., Fehr M.J., Kraus G.A., Petrich J.W. Chemiluminescent activation of the antiviral activity of hypericin: a molecular flashlight. Proc Natl Acad Sci U S A. 1994;91:12273–12277. doi: 10.1073/pnas.91.25.12273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Theodossiou T., Hothersall J.S., Woods E.A., Okkenhaug K., Jacobson J., MacRobert A.J. Firefly luciferin-activated rose bengal: in vitro photodynamic therapy by intracellular chemiluminescence in transgenic NIH 3T3 cells. Cancer Res. 2003;63:1818–1821. [PubMed] [Google Scholar]
- 53.Schipper M.L., Patel M.R., Gambhir S.S. Evaluation of firefly luciferase bioluminescence mediated photodynamic toxicity in cancer cells. Mol Imag Biol. 2006;8:218–225. doi: 10.1007/s11307-006-0048-1. [DOI] [PubMed] [Google Scholar]
- 54.Yang K., Wang C.L., Liu C.G., Ding S., Tian F., Li F. Bioluminescence-initiated photodynamic therapy bridged on high-luminescent carbon dots-conjugated protoporphyrin IX. J Mater Sci. 2019;54:3383–3391. [Google Scholar]
- 55.Fan D., Wang T., Hu J.H., Zhou L., Zhou J.H., Wei S.H. Plasmid DNA-based bioluminescence-activated system for photodynamic therapy in cancer treatment. ChemMedChem. 2021;16:1967–1974. doi: 10.1002/cmdc.202000979. [DOI] [PubMed] [Google Scholar]
- 56.Loening A.M., Wu A.M., Gambhir S.S. Red-shifted Renilla reniformis luciferase variants for imaging in living subjects. Nat Methods. 2007;4:641–643. doi: 10.1038/nmeth1070. [DOI] [PubMed] [Google Scholar]
- 57.Hsu C.Y., Chen C.W., Yu H.P., Lin Y.F., Lai P.S. Bioluminescence resonance energy transfer using luciferase-immobilized quantum dots for self-illuminated photodynamic therapy. Biomaterials. 2013;34:1204–1212. doi: 10.1016/j.biomaterials.2012.08.044. [DOI] [PubMed] [Google Scholar]
- 58.Kim S., Jo H., Jeon M., Choi M.G., Hahn S.K., Yun S.H. Luciferase-Rose Bengal conjugates for singlet oxygen generation by bioluminescence resonance energy transfer. Chem Commun. 2017;53:4569–4572. doi: 10.1039/c7cc00041c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kim E.H., Park S., Kim Y.K., Moon M., Park J., Lee K.J., et al. Self-luminescent photodynamic therapy using breast cancer targeted proteins. Sci Adv. 2020;6 doi: 10.1126/sciadv.aba3009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.England C.G., Ehlerding E.B., Cai W.B. NanoLuc: a small luciferase is brightening up the field of bioluminescence. Bioconjugate Chem. 2016;27:1175–1187. doi: 10.1021/acs.bioconjchem.6b00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hall M.P., Unch J., Binkowski B.F., Valley M.P., Butler B.L., Wood M.G., et al. Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone substrate. ACS Chem Biol. 2012;7:1848–1857. doi: 10.1021/cb3002478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Shramova E.I., Proshkina G.M., Chumakov S.P., Khodarovich Y.M., Deyev S.M. Flavoprotein miniSOG cytotoxisity can be induced by bioluminescence resonance energy transfer. Acta Naturae. 2016;8:118–123. [PMC free article] [PubMed] [Google Scholar]
- 63.Shramova E.I., Proshkina G.M., Deyev S.M., Petrov R.V. Death mechanism of breast adenocarcinoma cells caused by bret-induced cytotoxicity of minisog depends on the intracellular localization of the nanoluc-minisog fusion protein. Dokl Biochem Biophys. 2018;482:288–291. doi: 10.1134/S1607672918050150. [DOI] [PubMed] [Google Scholar]
- 64.Proshkina G.M., Shramova E.I., Shilova O.N., Ryabova A.V., Deyev S.M. Phototoxicity of flavoprotein miniSOG induced by bioluminescence resonance energy transfer in genetically encoded system NanoLuc-miniSOG is comparable with its LED-excited phototoxicity. J Photochem Photobiol, B. 2018;188:107–115. doi: 10.1016/j.jphotobiol.2018.09.006. [DOI] [PubMed] [Google Scholar]
- 65.Li J.Q., Huang J.Z., Ao Y.X., Li S.Y., Miao Y., Yu Z.Z., et al. Synergizing upconversion nanophotosensitizers with hyperbaric oxygen to remodel the extracellular matrix for enhanced photodynamic cancer therapy. ACS Appl Mater Interfaces. 2018;10:22985–22996. doi: 10.1021/acsami.8b07090. [DOI] [PubMed] [Google Scholar]
- 66.Tachibana K., Kimura N., Okumura M., Eguchi H., Tachibana S. Enhancement of cell killing of HL-60 cells by ultrasound in the presence of the photosensitizing drug Photofrin II. Cancer Lett. 1993;72:195–199. doi: 10.1016/0304-3835(93)90129-w. [DOI] [PubMed] [Google Scholar]
- 67.Nowak-Stepniowska A., Pergoł P., Padzik-Graczyk A. Photodynamic method of cancer diagnosis and therapy—mechanisms and applications. Postepy Biochem. 2013;59:53–63. [PubMed] [Google Scholar]
- 68.Dolmans D.E., Fukumura D., Jain R.K. Photodynamic therapy for cancer. Nat Rev Cancer. 2003;3:380–387. doi: 10.1038/nrc1071. [DOI] [PubMed] [Google Scholar]
- 69.Kwiatkowski S., Knap B., Przystupski D., Saczko J., Kędzierska E., Knap-Czop K., et al. Photodynamic therapy—mechanisms, photosensitizers and combinations. Biomed Pharmacother. 2018;106:1098–1107. doi: 10.1016/j.biopha.2018.07.049. [DOI] [PubMed] [Google Scholar]
- 70.Yoon I., Li J.Z., Shim Y.K. Advance in photosensitizers and light delivery for photodynamic therapy. Clin Endosc. 2013;46:7–23. doi: 10.5946/ce.2013.46.1.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sternberg E.D., Dolphin D. Second generation photodynamic agents: a review. J Clin Laser Med Surg. 1993;11:233–241. doi: 10.1089/clm.1993.11.233. [DOI] [PubMed] [Google Scholar]
- 72.Kataoka H., Nishie H., Hayashi N., Tanaka M., Nomoto A., Yano S., et al. New photodynamic therapy with next-generation photosensitizers. Ann Transl Med. 2017;5:183. doi: 10.21037/atm.2017.03.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Alves S.R., Calori I.R., Tedesco A.C. Photosensitizer-based metal-organic frameworks for highly effective photodynamic therapy. Mater Sci Eng C. 2021;131 doi: 10.1016/j.msec.2021.112514. [DOI] [PubMed] [Google Scholar]
- 74.Rehman F.U., Zhao C., Jiang H., Wang X. Biomedical applications of nano-titania in theranostics and photodynamic therapy. Biomater Sci. 2016;4:40–54. doi: 10.1039/c5bm00332f. [DOI] [PubMed] [Google Scholar]
- 75.Vankayala R., Sagadevan A., Vijayaraghavan P., Kuo C.L., Hwang K.C. Metal nanoparticles sensitize the formation of singlet oxygen. Angew Chem Int Ed Engl. 2011;50:10640–10644. doi: 10.1002/anie.201105236. [DOI] [PubMed] [Google Scholar]
- 76.Wu X.J., Xie S.J., Zhang H.K., Zhang Q.H., Sels B.F., Wang Y. Metal sulfide photocatalysts for lignocellulose valorization. Adv Mater. 2021;33 doi: 10.1002/adma.202007129. [DOI] [PubMed] [Google Scholar]
- 77.Younis M.R., He G., Qu J., Lin J., Huang P., Xia X.H. Inorganic nanomaterials with intrinsic singlet oxygen generation for photodynamic therapy. Adv Sci. 2021;8 doi: 10.1002/advs.202102587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chen J.J., Zhu Y.F., Wu C.T., Shi J.L. Nanoplatform-based cascade engineering for cancer therapy. Chem Soc Rev. 2020;49:9057–9094. doi: 10.1039/d0cs00607f. [DOI] [PubMed] [Google Scholar]
- 79.Carpentier P., Violot S., Blanchoin L., Bourgeois D. Structural basis for the phototoxicity of the fluorescent protein KillerRed. FEBS Lett. 2009;583:2839–2842. doi: 10.1016/j.febslet.2009.07.041. [DOI] [PubMed] [Google Scholar]
- 80.Bulina M.E., Chudakov D.M., Britanova O.V., Yanushevich Y.G., Staroverov D.B., Chepurnykh T.V., et al. A genetically encoded photosensitizer. Nat Biotechnol. 2006;24:95–99. doi: 10.1038/nbt1175. [DOI] [PubMed] [Google Scholar]
- 81.Serebrovskaya E.O., Edelweiss E.F., Stremovskiy O.A., Lukyanov K.A., Chudakov D.M., Deyev S.M. Targeting cancer cells by using an antireceptor antibody-photosensitizer fusion protein. Proc Natl Acad Sci U S A. 2009;106:9221–9225. doi: 10.1073/pnas.0904140106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Serebrovskaya E.O., Ryumina A.P., Boulina M.E., Shirmanova M.V., Zagaynova E.V., Bogdanova E.A., et al. Phototoxic effects of lysosome-associated genetically encoded photosensitizer KillerRed. J Biomed Opt. 2014;19 doi: 10.1117/1.JBO.19.7.071403. [DOI] [PubMed] [Google Scholar]
- 83.Hilgers F., Bitzenhofer N.L., Ackermann Y., Burmeister A., Grünberger A., Jaeger K.E., et al. Genetically encoded photosensitizers as light-triggered antimicrobial agents. Int J Mol Sci. 2019;20:4608. doi: 10.3390/ijms20184608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Sarkisyan K.S., Zlobovskaya O.A., Gorbachev D.A., Bozhanova N.G., Sharonov G.V., Staroverov D.B., et al. KillerOrange, a genetically encoded photosensitizer activated by blue and green light. PLoS One. 2015;10 doi: 10.1371/journal.pone.0145287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pletneva N.V., Pletnev V.Z., Sarkisyan K.S., Gorbachev D.A., Egorov E.S., Mishin A.S., et al. Crystal structure of phototoxic orange fluorescent proteins with a tryptophan-based chromophore. PLoS One. 2015;10 doi: 10.1371/journal.pone.0145740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Shu X.K., Lev-Ram V., Deerinck T.J., Qi Y.C., Ramko E.B., Davidson M.W., et al. A genetically encoded tag for correlated light and electron microscopy of intact cells, tissues, and organisms. PLoS Biol. 2011;9 doi: 10.1371/journal.pbio.1001041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ruiz-González R., Cortajarena A.L., Mejias S.H., Agut M., Nonell S., Flors C. Singlet oxygen generation by the genetically encoded tag miniSOG. J Am Chem Soc. 2013;135:9564–9567. doi: 10.1021/ja4020524. [DOI] [PubMed] [Google Scholar]
- 88.Kuzichkina E.O., Shilova O.N., Deyev S.M. The Mechanism of fluorescence quenching of protein photosensitizers based on miniSOG during internalization of the HER2 receptor. Acta Naturae. 2018;10:87–94. [PMC free article] [PubMed] [Google Scholar]
- 89.Li Y., Cui Z.J. Photodynamic activation of cholecystokinin 1 receptor with different genetically encoded protein photosensitizers and from varied subcellular sites. Biomolecules. 2020;10:1423. doi: 10.3390/biom10101423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Makhijani K., To T.L., Ruiz-González R., Lafaye C., Royant A., Shu X. Precision optogenetic tool for selective single- and multiple-cell ablation in a live animal model system. Cell Chem Biol. 2017;24:110–119. doi: 10.1016/j.chembiol.2016.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Westberg M., Holmegaard L., Pimenta F.M., Etzerodt M., Ogilby P.R. Rational design of an efficient, genetically encodable, protein-encased singlet oxygen photosensitizer. J Am Chem Soc. 2015;137:1632–1642. doi: 10.1021/ja511940j. [DOI] [PubMed] [Google Scholar]
- 92.Trewin A.J., Berry B.J., Wei A.Y., Bahr L.L., Foster T.H., Wojtovich A.P. Light-induced oxidant production by fluorescent proteins. Free Radic Biol Med. 2018;128:157–164. doi: 10.1016/j.freeradbiomed.2018.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yuan M., Liu C.C., Li J., Ma W.P., Yu X.Z., Zhang P., et al. The effects of photodynamic therapy on leukemia cells mediated by KillerRed, a genetically encoded fluorescent protein photosensitizer. BMC Cancer. 2019;19:934. doi: 10.1186/s12885-019-6124-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Liang L.E., Lu Y.Q., Zhang R., Care A., Ortega T.A., Deyev S.M., et al. Deep-penetrating photodynamic therapy with killerRed mediated by upconversion nanoparticles. Acta Biomater. 2017;51:461–470. doi: 10.1016/j.actbio.2017.01.004. [DOI] [PubMed] [Google Scholar]
- 95.Kim H.Y., Kang M., Choo Y.W., Go S.H., Kwon S.P., Song S.Y., et al. Immunomodulatory lipocomplex functionalized with photosensitizer-embedded cancer cell membrane inhibits tumor growth and metastasis. Nano Lett. 2019;19:5185–5193. doi: 10.1021/acs.nanolett.9b01571. [DOI] [PubMed] [Google Scholar]
- 96.Proshkina G.M., Shilova O.N., Ryabova A.V., Stremovskiy O.A., Deyev S.M. A new anticancer toxin based on HER2/neu-specific DARPin and photoactive flavoprotein miniSOG. Biochimie. 2015;118:116–122. doi: 10.1016/j.biochi.2015.08.013. [DOI] [PubMed] [Google Scholar]
- 97.Mironova K.E., Proshkina G.M., Ryabova A.V., Stremovskiy O.A., Lukyanov S.A., Petrov R.V., et al. Genetically encoded immunophotosensitizer 4D5scFv-miniSOG is a highly selective agent for targeted photokilling of tumor cells in vitro. Theranostics. 2013;3:831–840. doi: 10.7150/thno.6715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Tseng S.J., Kempson I.M., Huang K.Y., Li H.J., Fa Y.C., Ho Y.C., et al. Targeting tumor microenvironment by bioreduction-activated nanoparticles for light-triggered virotherapy. ACS Nano. 2018;12:9894–9902. doi: 10.1021/acsnano.8b02813. [DOI] [PubMed] [Google Scholar]
- 99.Takehara K., Tazawa H., Okada N., Hashimoto Y., Kikuchi S., Kuroda S., et al. Targeted photodynamic virotherapy armed with a genetically encoded photosensitizer. Mol Cancer Therapeut. 2016;15:199–208. doi: 10.1158/1535-7163.MCT-15-0344. [DOI] [PubMed] [Google Scholar]
- 100.Liao Z.X., Kempson I.M., Fa Y.C., Liu M.C., Hsieh L.C., Huang K.Y., et al. Correction to magnetically guided viral transduction of gene-based sensitization for localized photodynamic therapy to overcome multidrug resistance in breast cancer cells. Bioconjugate Chem. 2018;29:2126. doi: 10.1021/acs.bioconjchem.8b00356. [DOI] [PubMed] [Google Scholar]
- 101.Ryumina A.P., Serebrovskaya E.O., Shirmanova M.V., Snopova L.B., Kuznetsova M.M., Turchin I.V., et al. Flavoprotein miniSOG as a genetically encoded photosensitizer for cancer cells. Biochim Biophys Acta. 2013;1830:5059–5067. doi: 10.1016/j.bbagen.2013.07.015. [DOI] [PubMed] [Google Scholar]
- 102.Shramova E.I., Proshkina G.M., Deyev S.M., Petrov R.V. Flavoprotein miniSOG BRET-induced cytotoxicity depends on its intracellular localization. Dokl Biochem Biophys. 2017;474:228–230. doi: 10.1134/S160767291703019X. [DOI] [PubMed] [Google Scholar]
- 103.Shramova E.I., Chumakov S.P., Shipunova V.O., Ryabova A.V., Telegin G.B., Kabashin A.V., et al. Genetically encoded BRET-activated photodynamic therapy for the treatment of deep-seated tumors. Light Sci Appl. 2022;11:38. doi: 10.1038/s41377-022-00729-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Liao Z.X., Li Y.C., Lu H.M., Sung H.W. A genetically-encoded KillerRed protein as an intrinsically generated photosensitizer for photodynamic therapy. Biomaterials. 2014;35:500–508. doi: 10.1016/j.biomaterials.2013.09.075. [DOI] [PubMed] [Google Scholar]
- 105.Yang X.C., Niu Y.L., Zhao N.N., Mao C., Xu F.J. A biocleavable pullulan-based vector via ATRP for liver cell-targeting gene delivery. Biomaterials. 2014;35:3873–3884. doi: 10.1016/j.biomaterials.2014.01.036. [DOI] [PubMed] [Google Scholar]
- 106.Zhou J., Mohamed Wali A.R., Ma S., He Y., Yue D., Tang J.Z., et al. Tailoring the supramolecular structure of guanidinylated pullulan toward enhanced genetic photodynamic therapy. Biomacromolecules. 2018;19:2214–2226. doi: 10.1021/acs.biomac.8b00273. [DOI] [PubMed] [Google Scholar]
- 107.Tseng S.J., Liao Z.X., Kao S.H., Zeng Y.F., Huang K.Y., Li H.J., et al. Highly specific in vivo gene delivery for p53-mediated apoptosis and genetic photodynamic therapies of tumour. Nat Commun. 2015;6:6456. doi: 10.1038/ncomms7456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wu T.T., Liu Q., Cao Y.L., Tian R., Liu J.B., Ding B.Q. Multifunctional double-bundle DNA tetrahedron for efficient regulation of gene expression. ACS Appl Mater Interfaces. 2020;12:32461–32467. doi: 10.1021/acsami.0c08886. [DOI] [PubMed] [Google Scholar]
- 109.Serrano M.P., Vignoni M., Lorente C., Vicendo P., Oliveros E., Thomas A.H. Thymidine radical formation via one-electron transfer oxidation photoinduced by pterin: mechanism and products characterization. Free Radic Biol Med. 2016;96:418–431. doi: 10.1016/j.freeradbiomed.2016.04.196. [DOI] [PubMed] [Google Scholar]
- 110.Justiniano R., Williams J.D., Perer J., Hua A., Lesson J., Park S.L., et al. The B6 -vitamer pyridoxal is a sensitizer of UVA-induced genotoxic stress in human primary keratinocytes and reconstructed epidermis. Photochem Photobiol. 2017;93:990–998. doi: 10.1111/php.12720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tonolli P.N., Chiarelli-Neto O., Santacruz-Perez C., Junqueira H.C., Watanabe I.S., Ravagnani F.G., et al. Lipofuscin generated by UVA turns keratinocytes photosensitive to visible light. J Invest Dermatol. 2017;137:2447–2450. doi: 10.1016/j.jid.2017.06.018. [DOI] [PubMed] [Google Scholar]
- 112.Masuda H., Kimura M., Nishioka A., Kato H., Morita A. Dual wavelength 5-aminolevulinic acid photodynamic therapy using a novel flexible light-emitting diode unit. J Dermatol Sci. 2019;93:109–115. doi: 10.1016/j.jdermsci.2018.12.006. [DOI] [PubMed] [Google Scholar]
- 113.Cox T.M., Alexander G.J., Sarkany R.P. Protoporphyria. Semin Liver Dis. 1998;18:85–93. doi: 10.1055/s-2007-1007144. [DOI] [PubMed] [Google Scholar]
- 114.Yang X., Palasuberniam P., Kraus D., Chen B. Aminolevulinic acid-based tumor detection and therapy: molecular mechanisms and strategies for enhancement. Int J Mol Sci. 2015;16:25865–25880. doi: 10.3390/ijms161025865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Shi J.J., Nie W.M., Zhao X., Yang X.Y., Cheng H., Zhou T.H., et al. An intracellular self-assembly-driven uninterrupted ROS generator augments 5-aminolevulinic-acid-based tumor therapy. Adv Mater. 2022;34 doi: 10.1002/adma.202201049. [DOI] [PubMed] [Google Scholar]
- 116.Li L.H., Chen L.L., Huang L., Ye X.L., Lin Z.F., Wei X.M., et al. Biodegradable mesoporous manganese carbonate nanocomposites for LED light-driven cancer therapy via enhancing photodynamic therapy and attenuating survivin expression. J Nanobiotechnol. 2021;19:310. doi: 10.1186/s12951-021-01057-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Yue J., Li L., Jiang C.Y., Mei Q., Dong W.F., Yan R.H. Riboflavin-based carbon dots with high singlet oxygen generation for photodynamic therapy. J Mater Chem B. 2021;9:7972–7978. doi: 10.1039/d1tb01291f. [DOI] [PubMed] [Google Scholar]
- 118.Juarez A.V., Sosa Ldel V., De Paul A.L., Costa A.P., Farina M., Leal R.B., et al. Riboflavin acetate induces apoptosis in squamous carcinoma cells after photodynamic therapy. J Photochem Photobiol, B. 2015;153:445–454. doi: 10.1016/j.jphotobiol.2015.10.030. [DOI] [PubMed] [Google Scholar]
- 119.Yuan Y., Zhao Y.Y., Chen L.Q., Wu J.S., Chen G.Y., Li S., et al. Selective tumor cell death induced by irradiated riboflavin through recognizing DNA G-T mismatch. Nucleic Acids Res. 2017;45:8676–8683. doi: 10.1093/nar/gkx602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Khaydukov E.V., Mironova K.E., Semchishen V.A., Generalova A.N., Nechaev A.V., Khochenkov D.A., et al. Riboflavin photoactivation by upconversion nanoparticles for cancer treatment. Sci Rep. 2016;6 doi: 10.1038/srep35103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wu C.L., Li Y.Y., Cheng Z.H., Wang P.X., Ma Z.L., Liu K., et al. Cell-penetrating riboflavin conjugate for antitumor photodynamic therapy. Chin Chem Lett. 2022;33:4339–4344. [Google Scholar]
- 122.Yamada H., Arai T., Endo N., Yamashita K., Nonogawa M., Makino K., et al. Photodynamic effects of a novel pterin derivative on a pancreatic cancer cell line. Biochem Biophys Res Commun. 2005;333:763–767. doi: 10.1016/j.bbrc.2005.05.185. [DOI] [PubMed] [Google Scholar]
- 123.Park S.L., Justiniano R., Williams J.D., Cabello C.M., Qiao S., Wondrak G.T. The tryptophan-derived endogenous aryl hydrocarbon receptor ligand 6-formylindolo[3,2-b]carbazole is a nanomolar UVA photosensitizer in epidermal keratinocytes. J Invest Dermatol. 2015;135:1649–1658. doi: 10.1038/jid.2014.503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Justiniano R., de Faria Lopes L., Perer J., Hua A., Park S.L., Jandova J., et al. The endogenous tryptophan-derived photoproduct 6-formylindolo[3,2-b]carbazole (ficz) is a nanomolar photosensitizer that can be harnessed for the photodynamic elimination of skin cancer cells in vitro and in vivo. Photochem Photobiol. 2021;97:180–191. doi: 10.1111/php.13321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Liu J.N., Bu W., Shi J. Chemical design and synthesis of functionalized probes for imaging and treating tumor hypoxia. Chem Rev. 2017;117:6160–6224. doi: 10.1021/acs.chemrev.6b00525. [DOI] [PubMed] [Google Scholar]
- 126.Luoto K.R., Kumareswaran R., Bristow R.G. Tumor hypoxia as a driving force in genetic instability. Genome Integr. 2013;4:5. doi: 10.1186/2041-9414-4-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Tomaselli F., Maier A., Sankin O., Anegg U., Stranzl U., Pinter H., et al. Acute effects of combined photodynamic therapy and hyperbaric oxygenation in lung cancer-a clinical pilot study. Laser Surg Med. 2001;28:399–403. doi: 10.1002/lsm.1067. [DOI] [PubMed] [Google Scholar]
- 128.Ding L., Wu Y.N., Wu M., Zhao Q.F., Li H.S., Liu J.F., et al. Engineered red blood cell biomimetic nanovesicle with oxygen self-supply for near-infrared-II fluorescence-guided synergetic chemo-photodynamic therapy against hypoxic tumors. ACS Appl Mater Interfaces. 2021;13:52435–52449. doi: 10.1021/acsami.1c19096. [DOI] [PubMed] [Google Scholar]
- 129.Yang G.B., Xu L.G., Chao Y., Xu J., Sun X.Q., Wu Y.F., et al. Hollow MnO2 as a tumor-microenvironment-responsive biodegradable nano-platform for combination therapy favoring antitumor immune responses. Nat Commun. 2017;8:902. doi: 10.1038/s41467-017-01050-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Yang Y., Zhu W.J., Feng L.Z., Chao Y., Yi X., Dong Z.L., et al. G-quadruplex-based nanoscale coordination polymers to modulate tumor hypoxia and achieve nuclear-targeted drug delivery for enhanced photodynamic therapy. Nano Lett. 2018;18:6867–6875. doi: 10.1021/acs.nanolett.8b02732. [DOI] [PubMed] [Google Scholar]
- 131.Liu L., Zhang J.Y., Li Z.N., Yang Y., Li L.Y., Zhao Y.Y., et al. Enzyme-loaded catalytic macrophage vesicles with cascade amplification of tumor-targeting for oxygenated photodynamic therapy. Int J Nanomed. 2021;16:7801–7812. doi: 10.2147/IJN.S336333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Sahu A., Min K., Jeon J., Yang H.S., Tae G. Catalytic nanographene oxide with hemin for enhanced photodynamic therapy. J Control Release. 2020;326:442–454. doi: 10.1016/j.jconrel.2020.07.023. [DOI] [PubMed] [Google Scholar]
- 133.Chen J., Chen F., Zhang L., Yang Z.Y., Deng T., Zhao Y.F., et al. Self-assembling porphyrins as a single therapeutic agent for synergistic cancer therapy: a one stone three birds strategy. ACS Appl Mater Interfaces. 2021;13:27856–27867. doi: 10.1021/acsami.1c04868. [DOI] [PubMed] [Google Scholar]
- 134.Lin L.Y., Pang W., Jiang X.Y., Ding S.H., Wei X.B., Gu B.B. Light amplified oxidative stress in tumor microenvironment by carbonized hemin nanoparticles for boosting photodynamic anticancer therapy. Light Sci Appl. 2022;11:47. doi: 10.1038/s41377-021-00704-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Tian W., Wang S.J., Tian Y., Su X.D., Sun H., Tang Y.X., et al. Periodic mesoporous organosilica coupled with chlorin e6 and catalase for enhanced photodynamic therapy to treat triple-negative breast cancer. J Colloid Interface Sci. 2022;610:634–642. doi: 10.1016/j.jcis.2021.11.107. [DOI] [PubMed] [Google Scholar]
- 136.Zhao J., Fei J.B., Du C.L., Cui W., Ma H.C., Li J.B. Assembly of catalase-based bioconjugates for enhanced anticancer efficiency of photodynamic therapy in vitro. Chem Commun. 2013;49:10733–10735. doi: 10.1039/c3cc46969g. [DOI] [PubMed] [Google Scholar]
- 137.Fan X.T., Luo Z., Chen Y., Yeo J.C.C., Li Z.B., Wu Y.L., et al. Oxygen self-supplied enzyme nanogels for tumor targeting with amplified synergistic starvation and photodynamic therapy. Acta Biomater. 2022;142:274–283. doi: 10.1016/j.actbio.2022.01.056. [DOI] [PubMed] [Google Scholar]
- 138.Chen H.C., Tian J.G., He W.J., Guo J.Z. H2O2-activatable and O2-evolving nanoparticles for highly efficient and selective photodynamic therapy against hypoxic tumor cells. J Am Chem Soc. 2015;137:1539–1547. doi: 10.1021/ja511420n. [DOI] [PubMed] [Google Scholar]
- 139.Shen L.Y., Huang Y., Chen D., Qiu F., Ma C., Jin X., et al. pH-Responsive aerobic nanoparticles for effective photodynamic therapy. Theranostics. 2017;7:4537–4550. doi: 10.7150/thno.19546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Zhu J.W., Jiao A.H., Li Q.Z., Lv X.Y., Wang X.R., Song X.J., et al. Mitochondrial Ca2+-overloading by oxygen/glutathione depletion-boosted photodynamic therapy based on a CaCO3 nanoplatform for tumor synergistic therapy. Acta Biomater. 2022;137:252–261. doi: 10.1016/j.actbio.2021.10.016. [DOI] [PubMed] [Google Scholar]
- 141.Cai H.J., Shen T.T., Zhang J., Shan C.F., Jia J.G., Li X., et al. A core‒shell metal-organic-framework (MOF)-based smart nanocomposite for efficient NIR/H2O2-responsive photodynamic therapy against hypoxic tumor cells. J Mater Chem B. 2017;5:2390–2394. doi: 10.1039/c7tb00314e. [DOI] [PubMed] [Google Scholar]
- 142.Li S.Y., Cheng H., Xie B.R., Qiu W.X., Zeng J.Y., Li C.X., et al. Cancer cell membrane camouflaged cascade bioreactor for cancer targeted starvation and photodynamic therapy. ACS Nano. 2017;11:7006–7018. doi: 10.1021/acsnano.7b02533. [DOI] [PubMed] [Google Scholar]
- 143.Zhang J.Y., Li Z.N., Liu L., Li L.Y., Zhang L., Wang Y.K., et al. Self-assembly catalase nanocomplex conveyed by bacterial vesicles for oxygenated photodynamic therapy and tumor immunotherapy. Int J Nanomed. 2022;17:1971–1985. doi: 10.2147/IJN.S353330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Huang J., Xiao Z.C., Chen G.J., Li T., Peng Y., Shuai X.T. A pH-sensitive nanomedicine incorporating catalase gene and photosensitizer augments photodynamic therapy and activates antitumor immunity. Nano Today. 2022;43 [Google Scholar]
- 145.Ashton T.M., Fokas E., Kunz-Schughart L.A., Folkes L.K., Anbalagan S., Huether M., et al. The anti-malarial atovaquone increases radiosensitivity by alleviating tumour hypoxia. Nat Commun. 2016;7 doi: 10.1038/ncomms12308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Shiva S., Brookes P.S., Patel R.P., Anderson P.G., Darley-Usmar V.M. Nitric oxide partitioning into mitochondrial membranes and the control of respiration at cytochrome c oxidase. Proc Natl Acad Sci U S A. 2001;98:7212–7217. doi: 10.1073/pnas.131128898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Xiao S.S., Shi H., Zhang Y., Fan Y., Wang L., Xiang L., et al. Bacteria-driven hypoxia targeting delivery of chemotherapeutic drug proving outcome of breast cancer. J Nanobiotechnol. 2022;20:178. doi: 10.1186/s12951-022-01373-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bejarano L., Jordāo M.J.C., Joyce J.A. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021;11:933–959. doi: 10.1158/2159-8290.CD-20-1808. [DOI] [PubMed] [Google Scholar]
- 149.Duong M.T., Qin Y.H., You S.H., Min J.J. Bacteria-cancer interactions: bacteria-based cancer therapy. Exp Mol Med. 2019;51:1–15. doi: 10.1038/s12276-019-0297-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Huang X.H., Pan J.M., Xu F.N., Shao B.F., Wang Y., Guo X., et al. Bacteria-based cancer immunotherapy. Adv Sci. 2021;8 doi: 10.1002/advs.202003572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Zheng J.H., Nguyen V.H., Jiang S.N., Park S.H., Tan W., Hong S.H., et al. Two-step enhanced cancer immunotherapy with engineered Salmonella typhimurium secreting heterologous flagellin. Sci Transl Med. 2017;9:eaak9537. doi: 10.1126/scitranslmed.aak9537. [DOI] [PubMed] [Google Scholar]
- 152.Lin D.W., Feng X.L., Mai B.J., Li X., Wang F., Liu J.X., et al. Bacterial-based cancer therapy: an emerging toolbox for targeted drug/gene delivery. Biomaterials. 2021;277 doi: 10.1016/j.biomaterials.2021.121124. [DOI] [PubMed] [Google Scholar]
- 153.Shi L.L., Liu X.X., Li Y.Z., Li S., Wu W.B., Gao X.H., et al. Living bacteria-based immuno-photodynamic therapy: metabolic labeling of Clostridium butyricum for eradicating malignant melanoma. Adv Sci. 2022;9 doi: 10.1002/advs.202105807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Greathouse K.L., Stone J.K., Harris C.C. Cancer-type-specific bacteria: freeloaders or partners? Cancer Cell. 2020;38:158–160. doi: 10.1016/j.ccell.2020.06.017. [DOI] [PubMed] [Google Scholar]
- 155.Chen Q., Bai H.Z., Wu W.T., Huang G.J., Li Y., Wu M., et al. Bioengineering bacterial vesicle-coated polymeric nanomedicine for enhanced cancer immunotherapy and metastasis prevention. Nano Lett. 2020;20:11–21. doi: 10.1021/acs.nanolett.9b02182. [DOI] [PubMed] [Google Scholar]
- 156.Stritzker J., Hill P.J., Gentschev I., Szalay A.A. Myristoylation negative msbB-mutants of probiotic E. coli nissle 1917 retain tumor specific colonization properties but show less side effects in immunocompetent mice. Bioeng Bugs. 2010;1:139–145. doi: 10.4161/bbug.1.2.10286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Fan J.X., Li Z.H., Liu X.H., Zheng D.W., Chen Y., Zhang X.Z. Bacteria-mediated tumor therapy utilizing photothermally-controlled TNF-α expression via oral administration. Nano Lett. 2018;18:2373–2380. doi: 10.1021/acs.nanolett.7b05323. [DOI] [PubMed] [Google Scholar]
- 158.Wu C.S., Cui M.H., Cai L., Chen C., Zhu X.H., Wu Y.H., et al. NIR-responsive photodynamic nanosystem combined with antitumor immune optogenetics bacteria for precise synergetic therapy. ACS Appl Mater Interfaces. 2022;14:13094–13106. doi: 10.1021/acsami.2c01138. [DOI] [PubMed] [Google Scholar]
- 159.Sieow B.F., Wun K.S., Yong W.P., Hwang I.Y., Chang M.W. Tweak to treat: reprograming bacteria for cancer treatment. Trends Cancer. 2021;7:447–464. doi: 10.1016/j.trecan.2020.11.004. [DOI] [PubMed] [Google Scholar]
- 160.Yang M.Y., Yang F.W., Chen W.J., Liu S.H., Qiu L.P., Chen J.H. Bacteria-mediated cancer therapies: opportunities and challenges. Biomater Sci. 2021;9:5732–5744. doi: 10.1039/d1bm00634g. [DOI] [PubMed] [Google Scholar]
- 161.Yang Z.J., Zhu Y.J., Dong Z.J., Hao Y., Wang C.J., Li Q.G., et al. Engineering bioluminescent bacteria to boost photodynamic therapy and systemic anti-tumor immunity for synergistic cancer treatment. Biomaterials. 2022;281 doi: 10.1016/j.biomaterials.2021.121332. [DOI] [PubMed] [Google Scholar]
- 162.Yan L.B., Kanada M., Zhang J.Y., Okazaki S., Terakawa S. Photodynamic treatment of tumor with bacteria expressing killerred. PLoS One. 2015;10 doi: 10.1371/journal.pone.0131518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Wu X.M., Guan Q.Y., Han Y.B., Wang X.C., Zhuang W.Y., Tan R.X. Regeneration of phytochemicals by structure-driven organization of microbial biosynthetic steps. Angew Chem Int Ed Engl. 2022;61 doi: 10.1002/anie.202114919. [DOI] [PubMed] [Google Scholar]
- 164.Huo M.F., Wang L.Y., Zhang L.L., Wei C.Y., Chen Y., Shi J.L. Photosynthetic tumor oxygenation by photosensitizer-containing cyanobacteria for enhanced photodynamic therapy. Angew Chem Int Ed Engl. 2020;59:1906–1913. doi: 10.1002/anie.201912824. [DOI] [PubMed] [Google Scholar]
- 165.Zhang X.Y., Zhang Y.Y., Zhang C.N., Yang C., Tian R., Sun T., et al. An injectable hydrogel co-loading with cyanobacteria and upconversion nanoparticles for enhanced photodynamic tumor therapy. Colloids Surf B Biointerfaces. 2021;201 doi: 10.1016/j.colsurfb.2021.111640. [DOI] [PubMed] [Google Scholar]
- 166.Liu L.L., He H.M., Luo Z.Y., Zhou H.M., Cai L.T. In situ photocatalyzed oxygen generation with photosynthetic bacteria to enable robust immunogenic photodynamic therapy in triple-negative breast cancer. Adv Funct Mater. 2020;30 [Google Scholar]
- 167.Wang J., Su Q.F., Lv Q.Y., Cai B., Xiaohalati X., Wang G.B., et al. Oxygen-generating cyanobacteria powered by upconversion-nanoparticles-converted near-infrared light for ischemic stroke treatment. Nano Lett. 2021;21:4654–4665. doi: 10.1021/acs.nanolett.1c00719. [DOI] [PubMed] [Google Scholar]
- 168.Zhang Y.H., Liu H.F., Dai X.Y., Li H., Li Z.H. Cyanobacteria-based near-infrared light-excited self-supplying oxygen system for enhanced photodynamic therapy of hypoxic tumors. Nano Res. 2021;14:667–673. [Google Scholar]
- 169.Zhong D.N., Li W.L., Qi Y.C., He J., Zhou M. Photosynthetic biohybrid nanoswimmers system to alleviate tumor hypoxia for FL/PA/MR imaging-guided enhanced radio-photodynamic synergetic therapy. Adv Funct Mater. 2020;30 [Google Scholar]
- 170.Wang H.R., Guo Y.F., Wang C., Jiang X., Liu H.H., Yuan A., et al. Light-controlled oxygen production and collection for sustainable photodynamic therapy in tumor hypoxia. Biomaterials. 2021;269 doi: 10.1016/j.biomaterials.2020.120621. [DOI] [PubMed] [Google Scholar]
- 171.Deng X.Y., Yang W.B., Shao Z.W., Zhao Y.L. Genetically modified bacteria for targeted phototherapy of tumor. Biomaterials. 2021;272 doi: 10.1016/j.biomaterials.2021.120809. [DOI] [PubMed] [Google Scholar]
- 172.Ding S.J., Liu Z.M., Huang C.Y., Zeng N., Jiang W., Li Q. Novel engineered bacterium/black phosphorus quantum dot hybrid system for hypoxic tumor targeting and efficient photodynamic therapy. ACS Appl Mater Interfaces. 2021;13:10564–10573. doi: 10.1021/acsami.0c20254. [DOI] [PubMed] [Google Scholar]
- 173.Li J.Q., Zhuang Z.Y., Zhao Z.J., Tang B.Z. Type I AIE photosensitizers: mechanism and application. View. 2022;3 [Google Scholar]







