Open access peer-reviewed chapter

Phenazines and Photoactive Formulations: Promising Photodrugs for Photodynamic Therapy

Written By

Ranulfo Combuca da Silva Junior, Katieli da Silva Souza Campanholi, Flávia Amanda Pedroso de Morais, Laura Adriane de Moraes Pinto, Fabiana dos Santos Rando, Magali Soares dos Santos Pozza and Wilker Caetano

Submitted: 09 December 2022 Reviewed: 17 February 2023 Published: 20 March 2023

DOI: 10.5772/intechopen.110588

From the Edited Volume

Dyes and Pigments - Insights and Applications

Edited by Brajesh Kumar

Chapter metrics overview

105 Chapter Downloads

View Full Metrics

Abstract

Photodynamic Therapy (PDT) is a therapeutic modality that can be applied with many photosensitizing compounds (PS). Photosensitization has shown promising results in damage against abnormal cell growth as cancer and inactivating a broad spectrum of microorganisms with no reported microbial resistance. Photodynamic processes occur by the light action at the appropriate wavelength in the presence of a PS that will be excited by the energy absorbed from the light source, where the interaction with the oxygen present in the cell will generate reactive oxygen species (ROS). The potential of phenazines as a photosensitizer is reviewed in this chapter as a practical guide to the future development of formulations that are effective for cancer treatment and microorganism control. Here we mainly summarize articles about phenazines from 2005 to 2021 when we performed a systematic search in the Science Direct, PubMed, Google Scholar, Web of Science, and Scopus databases. The carrier systems formed by micellar copolymers type Pluronic® have demonstrated effectiveness in incorporating several PS, ensuring its monomeric form for PDT applications. The fundamentals of the photosensitization mechanism are discussed. Studies have shown the beneficial impact of an appropriate incorporation technique to enhance the cellular uptake of phenazines compounds.

Keywords

  • phenazines
  • photodynamic therapy
  • photosensitizing agents
  • nanoplatform
  • micelles

1. Introduction

Success in treating diseases such as cancer or microbial diseases directly depends on the therapeutic modality applied. Given the side effects presented and the limitation in the efficiency of traditional procedures (surgery, chemotherapy, and radiotherapy), other alternatives are constantly being proposed in oncology [1]. Among the most promising modalities, Photodynamic Therapy (PDT) stands out as it does not present serious side effects and does not have limited efficiency [2]. PDT and antimicrobial Photodynamic Therapy (aPDT) are medical modality that has high specificity and selectivity in the treatment of infections caused by a virus, bacteria, protozoa, and fungi, as well as several cardiovascular, dermatological, and other diseases related to abnormal cell growth as cancer [2, 3, 4, 5].

The PDT efficiency directly depends on a photosensitizing compound (PS) with ideal properties for the photophysical and photochemical processes that leads to the formation of singlet oxygen (1O2) and/or reactive oxygen species (ROS) that cause cell damage [2, 6]. PDT aims at the localized damage of living tissue with abnormal cell growth through its necrosis or infeasibility [7]. Likely targets of PDT are mitochondria, plasma membrane and other cell organelles, tumor cell nucleus, and blood vessels [8, 9]. This selectivity occurs due to the high concentration of lipoprotein receptors in neoplastic cells, where PS accumulates preferentially in diseased tissues, forming intravascular complexes with low-density proteins (LDL) [10].

The 1O2 is a free radical produced during PDT and other related therapies. It is made when light activates the photosensitive compound administered to the patient. The 1O2 action mechanism is based on its ability to cause damage to DNA, proteins, and other cellular molecules, leading to cell death [2]. Lipid peroxidation occurs when 1O2 reacts with lipids in cell membranes, causing the formation of secondary free radicals and damaging the cell membrane structure. DNA damage can arise when 1O2 reacts with DNA nucleotides, causing damage and interfering with replication and gene expression. The 1O2 stops energy production when reacting with the enzymes of the respiratory cell chain and leads to cell death. The 1O2 can react with cellular ribosomes and interrupt protein synthesis, causing cell death. The 1O2 is an essential agent in photodynamic therapy and other radiation therapies, as these action mechanisms lead to the destruction of cancer cells and a reduction in the size of tumors [2].

Among the main characteristics of adequate PS is their low toxicity in the dark, light absorption between 400 and 850 nm (therapeutic window), high molar absorptivity values, and considerable formation of ROS inherent in the technique [11].

The development of promising drugs requires science to improve investigations based on the action of a compound or the joint effort of two or more drugs, thus synergy [12]. The first generation of phototherapeutic agents used in PDT is based on mixtures of porphyrin derivatives [7]. The search for PS with better optical and pharmacokinetic characteristics gave rise to the second generation of PS, similar to porphyrin molecules such as benzoporphyrins, chlorins, texapyrins, phthalocyanines, and naphthalocyanines [13]. Some of those PS compounds are already approved by the Food and Drug Administration (FDA/USA) for clinical applications using PDT. In addition, some countries are already approved, for example, Photofrin®, Levulan Kerasticks®, and Visudyne® (Verteporfin) [14].

Due to advances in research, studies, and treatments based on PDT, there is a great demand for new naturally occurring or synthetic PS that are biocompatible and have adequate properties [15, 16]. Over time, hematoporphyrin derivatives have been replaced by various PS compounds [17]. The third generation of PS is based on formulations with different drug delivery systems (DDS) that provide better solubility in physiological media, more favorable pharmacokinetics, and allow the use of light sources with more remarkable penetration power into tissues.

Due to economic and environmental considerations, PS compounds obtained from abundant raw materials attract more interest than those prepared by complex chemical routes [15, 18]. Studies involving naphthodianthrones (hypericin), phenothiazines (methylene blue and toluidine blue), phthalocyanines, chlorins (chlorophyll A), xanthenes (rose bengal), and curcuminoids (curcumin) stand out in the literature [14]. These PS compounds are extensively studied in vitro, in vivo, and in photodiagnostic aspects [19, 20, 21].

This particular chapter presents explicitly an overview of compounds of the phenazine class (Figure 1), in which promising photoactive drugs such as neutral red (NR) [22], phenosafranin (PhS) [23], and safranine-O (Sf) [24], still poor explored for PDT applications.

Figure 1.

Phenazines compounds incorporated into drug delivery system have great potential for photodynamic therapy applications.

Advertisement

2. Search strategy

The strategy we performed a systematic literature search in Science Direct, PubMed, Google Scholar, Web of Science, and Scopus databases using the combinations of the term “phenazines” with the following: “photodynamic therapy,” “aPDT or antimicrobial Photodynamic Therapy, or PACT or photodynamic inactivation or PDI,” “Animal studies or in vitro studies involving phenazines”. Peer-reviewed articles published in English from 2005 to 2021 were included to compile this chapter. We have also scanned references for relevant articles.

Advertisement

3. Photosensitization mechanism

The combination of a PS compound with molecular oxygen (3O2) and visible light of the adequate wavelength generates ROS that causes cell components to oxidize and lead to death [2, 25]. In addition, PS reacts with neighboring molecules by electron or hydrogen transfer leading to the production of free radicals or by energy transfer to oxygen, inducing the production of 1O2 [2, 26].

The photochemical processes originate from the interaction of light with matter which, by absorbing energy with adequate wavelength, allows the promotion of an electron from the ground state, called HOMO (Highest Occupied Molecular Orbital), to the excited state LUMO (Lowest Unoccupied Molecular Orbital), of higher energy [27]. The excited state is unstable, and in it, the molecule can suffer chemical processes (rearrangements or fragmentation of the molecule) or physical processes (deexcitation) [27]. According to the Molecular Orbital Theory, oxygen in the ground state and excited can assume different forms of occupation of molecular orbitals anti-binders, as shown in Figure 2.

Figure 2.

Electronic distribution in the antibonding molecular orbitals for the oxygen electronic states. It was adapted from Lakowicz [27].

In the ground state, molecular oxygen has two unpaired electrons in the doubly degenerate antibonding orbitals, π*x, and π*y. These electrons have the same spin, resulting in a maximum multiplicity and, thus, the lowest state oxygen energy. Therefore, the ground state of molecular oxygen is a triplet, which has the spectroscopic term 3g. The excited state of oxygen that has all valence electrons paired is singlet oxygen. Singlet oxygen has two forms with distinct symmetries, one of smaller energy 1g, doubly degenerate (1x and 1y; 92.4 kJ mol−1), and another one with higher energy (1Σ; 159.6 kJ mol−1). The second excited state of oxygen has a short lifetime since the transition to the 1g state is allowed by spin. The different symmetry of the 1g species with respect to the ground state and the spin prohibition of the 1g3g transition ensures that the 1g species has a long enough lifetime to allow oxidation of organic molecules [7].

The photochemical processes that occur in the excited state of photoactive molecules can be represented by the Jablonski diagram (Figure 3) [27, 28]. Among these processes, internal conversion (IC), fluorescence, intersystem crossing (ISC), and phosphorescence stand out [27, 29].

Figure 3.

Jablonski diagram: Electron transfer scheme by type I and type II mechanisms with singlet oxygen, superoxide anions, and hydroxyls production. It was adapted from Lakowicz [27].

After molecule excitation to the second excited state, the relaxation process for the first excited state can occur by IC (where the internal energy is lost by collision) and then return to the ground state via fluorescence emission. Deactivation can also arise by ISC between vibrational levels of the same energy and electronic states of different multiplicities. The decay from the triplet state to the singlet fundamental can then occur by phosphorescence emission or collisions [27, 29]. For PDT, it is interesting that the PS is preferably in the excited triplet state, as it will have the same spin multiplicity of 3O2 favoring the photochemical processes of producing 1O2 and ROS. The interaction with biological substrates in PDT can occur by two mechanisms: Type I, where photo-oxidation occurs through the transfer of electrons between the triplet state of PS and the substrate forming radical ions that react with oxygen in the ground state resulting in ROS. The Type II mechanism involves the energy transfer from the triplet excited state of PS to molecular oxygen generating 1O2 [2, 28].

The literature reports the use of PDT in treating primary carcinomas or metastases in the head and neck regions, such as the oral cavity, pharynx, and larynx [30]. However, after tumor recession, secondary diseases such as those caused by bacteria may manifest. Photodynamic treatment circumvents this problem as PDT is not restricted to microorganisms.

With the increasing cases of acquired resistance by bacteria against antibiotics, the search for the control of microorganisms via PDT also attracts interest from the scientific community [31]. As a result, PDT has been widely applied in the microbiological area, standing out as a promising technique in microorganism inactivation [32]. Furthermore, PDT offers advantages over the usual antimicrobial agents, triggering rapid cell death and unlikely development of resistance by the microorganism [33].

Advertisement

4. Photosensitizer formulation

Several PS can be used for different applications because of their particular properties. Therefore, it is necessary to know the essential characteristics of each PS to use its therapeutic properties better. The most cited PS compounds in the literature present, for the most part, structures containing aromatic rings or conjugated systems, which give them high hydrophobicity. In this sense, efforts in developing strategies in the formulation of PS administration stand out. However, even PS with a small structure presents a self-aggregation problem, making it challenging to apply in aqueous media.

Incorporating PS compounds into nanostructured systems aims to minimize the effects of self-aggregation in aqueous media, protect PS against degradation and elimination by the organism, and facilitate the biotransport. In some cases, the adequately incorporated PS assists the vectorization of the nanostructured system, increasing the bioavailability of the photoactive drug at the application site (third generation), promoting a controlled release in the regions (tissues and organs) to be treated [34]. In addition, they decrease side effects (toxicity) and microbial resistance. In this sense, several carrier systems are generally constituted by colloidal dispersion systems, such as copolymeric micelles [35], liposomal vesicles [36], dendrimers [37], cyclodextrins [38, 39], polymer-DNA complexes (polyplexes) [40], nanogels [41], nanotubes [42], nanosuspensions [43], nanocrystals [44], solid lipid nanoparticles [45], metallic or ceramic nanoparticles [46], among other nanoscale materials for medical use are being widely studied [47, 48, 49]. Figure 4 illustrates the most commonly found nanoparticulate carrier systems in the literature for carrying PS [50].

Figure 4.

Exemplification of some carrier systems currently used. I was adapted from Senapati et al. [50].

All these carrier systems have the advantage of protecting PS as a common characteristic. In addition, they contribute to keeping the PS monomeric form ideal for application in PDT [48, 51]. The leading site of photodynamic action is the cell membranes; therefore, it is of great importance that the carrier system is similar or presents a specific interaction with it. The PS efficiency in PDT applications depends on the microenvironment in which the PS is located so that there are no changes in its physicochemical and photophysical properties and in its interaction with living biological systems [36]. Parameters such as bioaccessibility, passive transport, and permeation of the drug into the membrane are of great importance in developing new drugs for PDT applications [11, 52]. Micellar and liposomal environments, for example, are reasonable models of cellular environments and provide useful information about PS molecules against organized systems and biomimetics [5, 41, 52, 53].

Since the cell membrane is a complex system composed of a lipid bilayer consisting of several types of phospholipids, cholesterol, glycolipids, and proteins, the copolymer micelles are good carrier systems for biomimetic the cell membrane [54]. Different micellar microdomains, called hydrophobic and hydrophilic, allow for estimating the drug partition tendency according to the region where it was solubilized. Compared to membrane models, polymeric micelles have the following advantages: minimal toxicity, narrow size distribution, longer residence time in the circulatory system, improved bioavailability, and more excellent stability of the incorporated photosensitizer [54].

Advertisement

5. Polymeric nanoparticles: copolymeric micellar systems in drug formulation

Most dyes, including phenazines, are characterized by forming aggregates in an aqueous solution harming their application. For this case, using DDS systems such as copolymeric micelles contributes to stabilization and solubilization, which maintains the PS characteristics inherent in PDT application [55]. In addition, these compounds are readily adsorbed by anionic micelles due to their distinct and amphiphilic character [56]. Therefore, studying the interaction of dyes with micellar systems is essential for biological applications since PS-associated biopolymers provide high drug efficacy with reduced toxicity [57].

Polymeric nanoparticles have been extensively applied in the pharmaceutical industry for drug formulation [58, 59, 60]. Currently, research is directed toward studying the association of photoactive molecules with nanostructured systems aiming for a specific carrier for controlled release in the regions (tissues and organs) to be treated [58]. Colloidal copolymers stand out, forming micelles (triblock systems) and effectively stabilizing hydrophobic molecules in aqueous media, such as PS, that remain in the form of monomers [16].

Polymeric colloidal systems are excellent for drug delivery and have been widely explained in several studies [48, 53]. These carrier systems can be classified as anionic, cationic, zwitterionic, and nonionic (according to their state of charge) [6]. Micelles are amphiphilic structures composed of a hydrophobic and a hydrophilic region (Figure 5A) [52]. In the aqueous phase, the micelles keep the hydrophobic portion facing the inside of the structure (lipophilic core) [61].

Figure 5.

(A) General monomer structure of polymeric micelle, and (B) general structure of the copolymeric monomer (Pluronic® class).

Micelles simulate the interface of a biomembrane and provide a biomimetic environment for the study of specific interactions, as well as the penetration and location of PS in the intracellular domain [4, 5]. Furthermore, studies have shown that PS incorporation into micellar systems can reduce toxicity, target PS to specific sites and improve permeation and bioavailability in topical applications [11, 41]. Given these particularities, the biotransport and delivery of the drug in the target tissue become a particular challenge in PDT [4].

Several studies point to the use of surfactants of the Pluronic® class (or Poloxamers) [62]. The copolymer unimers (Figure 5B) consist of triblock molecules of repeating units of oxyethylene (EO – hydrophilic) and oxypropylene (PO – hydrophobic) with the following configuration: (EO)X(PO)Y(EO)X [63]. This group of copolymers is non-toxic, biocompatible, and has binding sites suitable for the solubilization of hydrophobic drugs [63]. In addition, triblock copolymers have a low critical micellar concentration (CMC, 10−6 – 10−7 mol L−1) and the micelles formed to have high thermodynamic and kinetic stability, which guarantees a slow destructuring for unimers when they are exposed to an environment where the concentration is below the CMC [64, 65]. Another factor to be considered for studies with polymeric surfactants is the temperature. The micellization process will only occur above a specific critical micellar temperature (CMT), which, in turn, is a function of the surfactant concentration [64, 65]. In addition, there is evidence that temperature increases also cause an increase in the aggregation number (Nag) of the copolymers [64]. Therefore, to be effective, the polymer used to obtain the micelle must present CMT compatible with the conditions necessary for the application [65].

Due to their favorable properties, the triblock copolymers of the Pluronic® class are potentially useful for drug delivery and controlled release systems [4, 53]. Furthermore, compared to other membrane models, copolymeric micelles have the following advantages: minimal toxicity, narrow size distribution, longer residence time at the application site, improved bioavailability, and more excellent stability of the incorporated PS [54]. These characteristics allow us to describe the micellar copolymer system as promising for clinical applications [66, 67].

Advertisement

6. Phenazines compound as photosensitizers or photoactive drugs

Phenazines (C12H8N2) are heterocyclic aromatic nitrogenous compounds and the most important backbone is a pyrazine ring (1,4-diazobenzene) with two annulated benzenes (Figure 6) [68].

Figure 6.

General chemical structure of phenazines compounds.

This compound class is classified as redox-active secondary metabolites and pigmented. They are produced by fluorescent Pseudomonas species and have antimicrobial, antiparasitic, neuroprotective, insecticidal, anti-inflammatory, and anticancer activity [69].

Phenazines can undergo redox cycling in the presence of various reducing agents and molecular oxygen, which leads to the accumulation of superoxide (O2) and hydrogen peroxide (H2O2), causing oxidative cell injury or death [68]. This property can be better explored considering the photochemical/photophysical potential of phenazines in PDT applications.

Natural phenazines are isolated from marine and terrestrial microorganisms, including (Pseudomonas ssp., Streptomyces ssp., and Actinomycete ssp.), which generates an infinity of synthetic derivatives [69]. Pseudomonas aeruginosa is a Gram-negative bacterium most studied for its ability to produce phenazine-active pigments, such as pyocyanin, phenazine-1 carboxyamide, and pyorubrins [68]. Furthermore, P. aeruginosa can survive in varied environments such as soil and water and colonize plant and animal tissues [70].

The literature describes the antioxidant, anti-inflammatory, and non-cytotoxic properties of the red and yellow phenazynic pigments produced by P. aeruginosa. These pigments confer great potential for application in the pharmaceutical and cosmetic industry [71, 72]. Phenazine derivatives differ in their chemical and physical properties based on the type of functional group position present, and they can also be used against the proliferation of various cancer cell lines [73, 74, 75, 76]. There are more than 100 different compounds of natural origin and over 6000 synthetic compounds. Many of these compounds have been investigated as potential anticancer agents and microorganism control [77]. Some in vitro studies using phenazine derivatives are shown in Table 1.

Phenazines derivativesObtainingApplicationsReference
2-chloroacetylamino-7(8)-nitrophenazine N5,N10-dioxide
2-amino-7(8)-(1,3-dioxol-2-yl)phenazine
N5,N10-dioxide(2),2-chloroacetylamino-7(8)-(1,3-dioxol-2-yl)phenazine N5,N10-dioxide
2-amino-7(8)-methoxyphenazine N5,N10-dioxide
Cytotoxins of 2-amino-or2-hydroxyphenazine5,10-dioxide derivativesin vitro antitumoral effect against Caco-2 cells[78]
phenazine 5,10-dioxide derivativesDerivative from N-oxides containing heterocyclesin vitro growth inhibitors of T. cruzi[79]
phenazine-1-carboxamideObtained from the Pseudomona aeruginosaantibacterial activity of a Pseudomonas aeruginosa-derived compound against methicillin-resistant S. aureus (MRSA) strains[80]
phenazin-1-ol
phenazine-1-carboxylic acid
2-heptyl-3-hydroxyl-4(1H)-quinolone
phenazine-1-carboxamide
Producing from Pseudomona aeruginosa fluorescentMedically important fungi: Aspergillus flavus MTCC 183, Candida albicans MTCC 277, Candida tropicalis MTCC 184, Cryptococcus gastricus MTCC 1715, and Trichophyton rubrum MTCC 296.
Agriculturally important fungi: Fusarium oxysporum MTCC 284, Rhizoctonia solani MTCC 4634, and Penicillium expansum MTCC 2006
[81]
phenazine 1-carboxamideProduced by Pseudomonas strain MCC2142fungi such as Candida albicans, Candida glabrata, Cryptococcus neoformans, Fusarium oxysporum, Aspergillus fumigatus, Aspergillus niger and Benjaminiella poitrasii[82]
Pontemazines A and BIsolated from the culture broth of Streptomyces sp. UT1123neuronal cell protective effect on
glutamate-induced mouse hippocampal HT-22 cell damage
[83]
A series of 2,3,7-trisubstituted phenazinesIntroduction of carboxylic or carboxamide group in 2,3-dialkoxy-phenazinein vitro on human pancreatic (MiaPaCa-2) cell lines[74]
imidazo[4,5-b]phenazine-2-thione
methylthio
ethyl 1-aryl-3H-[1, 2, 4]triazolo[2,3-a]imidazo[4,5-b]phenazines
ethyl (2Z)-3-aminophenazin-2-yl)amino](phenylhydrazono) ethanoate
pyrazino[2,3-b]phenazine
[1, 4]diazepino[2,3-b]phenazine
2,3-dibenzoylaminophenazine
1H-Imidazo[4,5-b]phenazine
4-[(E)-(3-amino phenazin-2-yl)diazenyl]
2,3-Diaminophenazine was used as a precursorall compounds were tested as inhibitors of the proliferation of human lung carcinoma and colorectal cancer cell lines through inhibition of Tyrosine Kinases[73]
benzo[a]phenazin derivativesTreatment of 2-fluoro-nitrobenzene or 2-chloro-3-nitropyridinefour human cancer cell lines (HL-60, K-562, HeLa, and A549)[84]
16-(4-ethoxyphenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(4-Ethoxyphenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(2,5-Dimethylphenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(Benzo[d][1, 3]dioxol-5-yl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
3,3-Dimethyl-16-(o-tolyl)-2,3,4,16-tetrahydro-1H-benzo-[a]chromeno[2,3-c]phenazin-1-one
16-(3-Bromophenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(2-Bromophenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(3-Hydroxyphenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-]phenazin-1-one
16-(3-Methoxyphenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-]phenazin-1-one
4-(3,3-Dimethyl-1-oxo-2,3,4,16-tetrahydro-1H-benzo-[a]chromeno[2,3-c]phenazin-16-yl)benzonitrile
16-(4-Fluorophenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(2-Methoxyphenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-]phenazin-1-one
16-(1H-Indol-3-yl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
3,3-Dimethyl-16-(3,4,5-trimethoxyphenyl)-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
3,3-Dimethyl-16-(thiophen-2-yl)-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
3,3-Dimethyl-16-(3-nitrophenyl)-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(3-Fluorophenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
3,3-Dimethyl-16-(4-nitrophenyl)-2,3,4,16-tetrahydro-1Hbenzo[a]chromeno[2,3-c]phenazin-1-one
16-(4-(Dimethylamino)phenyl)-3,3-dimethyl-2,3,4,16-tetrahydro-1H-benzo[a]chromeno[2,3-c]phenazin-1-one
16-(4-Ethoxyphenyl)-2,3,4,16-tetrahydro-1H-benzo[a]-chromeno[2,3-c]phenazin-1-one16-(4-Isopropylphenyl)-2,3,4,16-tetrahydro-1H-benzo-[a]chromeno[2,3-c]phenazin-1-one
16-(2-Chlorophenyl)-2,3,4,16-tetrahydro-1H-benzo[a]-chromeno[2,3-c]phenazin-1-one
16-(2-Methoxyphenyl)-2,3,4,16-tetrahydro-1H-benzo-[a]chromeno[2,3-c]phenazin-1-one
16-(3-Methoxyphenyl)-2,3,4,16-tetrahydro-1H-benzo-[a]chromeno[2,3-c]phenazin-1-one
16-(3-Chlorophenyl)-2,3,4,16-tetrahydro-1H-benzo-[a]chromeno[2,3-c]phenazin-1-one
16-(3-Fluorophenyl)-2,3,4,16-tetrahydro-1H-benzo-[a]chromeno[2,3-c]phenazin-1-one
Synthesis by condensation reaction of benzo[a]phenazine-5-olantioxidant and anticancer activities against HeLa and SK-BR-3 cell lines[75]
Phenazine-1-carboxamideIsolated from Pseudomonas sp. strain PUP6cytotoxic activity against lung (A549) and breast (MDA-MB-231) cancer cell lines[85]
phenazine-1,6-dicarboxylic acid
phenazine-1-carboxylic acid
Produced by Lactococcus BSN307 strainantifungal activity against Aspergillus niger, Penicillium chrysogenum as well
as Fusarium oxysporum
cytotoxicity against cancer cell lines HeLa and MCF-7 and normal H9c2 cells
[86]
phenazine-1-carboxamideIsolated from the bacterium Pantoea agglomerans naturally present in soilcytotoxicity on the cancer cell lines A549, HeLa, and SW480[87]
Pyrano[3,2-a]phenazine derivatives:
3-amino-10-((1-aryl-1H-1,2,3-triazol-5-yl)methyl)-20-oxospiro[benzo[a] pyrano[2,3-]phenazine-1,30-indoline]-2-carbonitrile
Synthesized from 2-amino-3-hydroxyphenazinecytotoxicity against HCT116, MCF7, HepG2 and A549 cancer cell lines in vitro[76]
RiminophenazineSynthesized from clofazimine and TMP-phenazinesin vitro antiplasmodial, cytotoxic, and oxidative activities against HeLa cells[88]
N-((tert-Butyldimethylsilyl)oxy)-9-chlorophenazine-1- carboxamide
9-Chloro-N-hydroxyphenazine-1-carboxamide
9-Chlorophenazine-1-carboxamide
9-Chloro-N-cyanophenazine-1-carboxamide
9-Chloro-N-(1H-tetrazol-5-yl)phenazine-1-carboxamide
9-Chlorophenazine-1-carbonitrile
2-((2,5-Dichlorophenyl)amino)-3-nitrobenzoic acid
6,9-Dichlorophenazine-1-carboxylic acid
Methyl 6,9-dichlorophenazine-1-carboxylate
6,9-Dichlorophenazine-1-carboxamide
6,9-Dichloro-N-(methylsulfonyl)phenazine-1-carboxamide
9-Bromo-6-methoxyphenazine-1-carboxylic acid
7-Bromo-9-methoxyphenazine-1-carboxylic acid
6,9-Dimethoxyphenazine-1-carboxylic acid
9-Bromo-6-methoxy-N-(methylsulfonyl)phenazine-1-carboxamide
9-Bromo-N-cyano-6-methoxyphenazine-1-carboxamide
Methyl 7-bromo-9-methoxyphenazine-1-carboxylate
9-Bromo-7-methoxyphenazine-1-carboxylic acid (25) and
7-methoxyphenazine-1-carboxylic acid
Methyl 9-bromo-7-methoxyphenazine-1-carboxylate
Methyl 7-methoxyphenazine-1-carboxylate
N-(Methylsulfonyl)phenazine-1-carboxamide
N-Methyl-N-(methylsulfonyl)phenazine-1-carboxamide
N-Cyano-9-fluorophenazine-1-carboxamide
9-Fluoro-N-(methylsulfonyl)phenazine-1-carboxamide
9-Methyl-N-(methylsulfonyl)phenazine-1-carboxamide
Methyl 9-methoxyphenazine-1-carboxylate
9-Chloro-N-methyl-N-(methylsulfonyl)phenazine-1-carboxamide
Chloro-substituted phenazines containing acid bioisosteresin vitro antimicrobial activity against Gram-positive (methicillin-resistant Staphylococcus aureus, MRSA) and Gram-negative (Escherichia coli) bacteria[89]
phenazine analogue (CPUL1)Synthesized from 2-amino-3-hydroxyphenazineantitumor activities in initial stage of Hep G2 cells[90]

Table 1.

Studies of phenazines derivatives compounds.

Most studies involving compounds of the phenazines class have been reported promisingly in the dark [23]; however, the photodynamic potential cannot be disregarded. The literature presents scattered articles concerning different studies of phenazines derivative compounds performed in vitro, but without continuity or emphasis on the photophysical potential of phenazines, as well as in vitro and in vivo studies. Further, this group of compounds has also been used to develop color-emitting materials [91] or fluorescent biosensors [92].

Udumula et al. [93] described the synthesis and biological evaluation of two phenazines natural products and a series of phenazines that show promising activities against methicillin-resistant Staphylococcus aureus associated with the CA-MRSA community with low minimum inhibitory concentration (MIC) values in the micromolar range [93]. The most active compound also showed good IC50 values against Human Keratinocyte Cell (HaCat) [93]. In work proposed by Krishnaiah et al. [89], the authors synthesized a series of phenazines, and the in vitro antimicrobial activity was evaluated against Gram-positive bacteria (methicillin-resistant Staphylococcus aureus, MRSA) and Gram-negative bacteria (Escherichia coli) [89]. These studies have indicated that the molecules do not disrupt bacterial membranes, and activity is not directly linked to the reactive oxygen species generation [89]. Therefore, despite a large number of studies on this compound class, this review highlights the great photodynamic potential of compounds of this class that can be used in the control of microorganisms and cancer treatment [94].

Neutral Red (NR) cationic dyes have great potential as photosensitizers. The exciting characteristic of NR as a PS is its water solubility compared with the majority of photosensitive and absorption bands located in the range of 550–700 nm [22]. Concerning with few phenazines studies as photodrugs explored in the literature (Table 2), for instance, the NR phenazines compound, when incorporated into Gold Nanoparticles, contributed to the in vitro tumor cell lines reduction [22], or when in aqueous medium significantly reduced in vitro S. aureus colonies in photoinactivation assays [98, 103].

CompoundDelivery systemApplicationIllumination conditionEffectReference
SfAqueous solution
(1 mMdm−3)
Bactericidal effect of serum combined with the action of PSWhite non mutagenic light (5Wcm−2)/90 minThe strains are sensitive to the photodynamic action[24]
SfAqueous solution
(in PBS 0.7 μg mL−1)
Stimulated production of ROS in the inner space of mitochondriaLED at λ = 509 ± 5 nm (29 Wm−2)/0.5–15 minDamage induced apoptosis[95]
PhSSingle-wall carbon nanotubes modifiedBHK-21 cell line (from mouse fibroblasts)Visible light irradiation (Hg lamp)Low toxicity in the dark and higher toxicity in the presence of light[42]
SfBovine Serum Albumin pH 7.4 (in Tris–HCl-NaOH buffer solution)Study of the damage caused by ROS in bovine serum albuminControllable Serial-Ultrasonic apparatus (frequency 59 kHz and power 50 WDamage of Bovine Serum Albumin under ultrasonic irradiation in the presence of Sf.[96]
SfAqueous solution (PBS containing 7% ethanol)Oral-pathogenic species (Gram-positive and Gram-negative)Laser Light (20 J cm−2)Significant antibacterial impact on different oral pathogenic species[97]
SfGold nanoparticles incorporated into a copolymer emulsionStaphylococcus aureus and Escherichia coli,28 W white light sourceThe bacterial count reduced (>4 ± 0.3 log kill)[43]
NR Monobrominated NRAqueous solution
(in PBS pH 7.4)
Staphylococcus aureusLight dose 7.6–30.2 J cm−2Photoantimicrobial effect (>3 log10) killing[98]
SfAqueous solutionHyalomma dromedariiSpot white-light source (power 100 W)Reduction of ovipositing, eggs per female, tickets laying viable eggs and hatched eggs[99]
NRGold Nanoparticles and Sodium thioglycolateNIH-3 T3 fibroblast (noncancerous) and 4 T1 tumor cell linesTwin Flex Laser LED MM optics λ = 440 nm; 220 mWReduction of cell viability[22]
PhS-Chlorambucil conjugateEncapsulated liposomes (DMPC)HeLa (human, cervical carcinoma)Excellent cell contrast facilitating its use as a theranostic anti-cancer drug[23]
SfPluronic® F127 and P123 4% (w/V)In vitro: Staphylococcus aureus, Escherichia coli, Streptococcus agalactiae, Corynebacterium bovis
In vivo: prevent mastitis in Dutch dairy cows.
In vitro studies: Green LED λ = 520 nm (7.2 mW cm−2)
In vivo studies: Green LED λ = 520 nm (12.7 mW cm−2)
In vitro studies of the Sf-F127 and Sf-P123 systems proved to be efficient in inactivating the bacteria that cause bovine mastitis. In vivo studies prevent bovine mastitis[33]
NR Monobrominated NRAqueous solution
(in PBS pH 7.4)
Staphylococcus aureusParathom lamp (OSRAM-5 W) 8.4 mW cm−2 (15–30 min)2–3 log of killing[98]
SfStimuli-responsive hydrogel by F127 Pluronic® and Carbopol (C934P)Mastitis treatmentIn vitro studies: Green LED λ = 520 nm (7.2 mW cm−2)
In vivo studies: Green LED λ = 520 (12.7 mW cm−2)
In vitro: efficiency in the inactivation of pathogens that cause mastitis. In vivo: Sf is highly efficient for mastitis treatment.[100]
SfPluronic® F127 4% (w/V)In vitro: Staphylococcus aureus, Escherichia coliGreen LED λ = 520
(7.2 mWcm−2)
The bacteria were sensitive to the photodynamic action of Sf[101]
PhS polyhedral oligomeric silsesquioxaneAqueous solution (in PBS pH 7.4)Staphylococcus aureus and Escherichia coliLED-based light source λ = 522 nm (10.6 mW cm−2)Bacterial cells were eradicated by ROS produced upon irradiation[102]

Table 2.

Studies of phenazine class compounds in photodynamic applications.

Phenosafranin (PhS), another phenazines class compound, has the potential for use as a PS in PDT. It is well known that PhS absorbs strongly at 503–530 nm. Additionally, this dye presents a good photodynamic effect, namely low toxicity in the dark and higher toxicity under optical excitation [23]. PhS was incorporated into single-wall carbon nanotubes and showed satisfactory photodynamic effects against the BHK-21 cell line [42]. Furthermore, when encapsulated in liposomes (DMPC), PhS exhibited excellent in vitro activity against Human Cervical Carcinoma (HeLa) [23]. Another application for PhS found in the literature is in vitro photodynamic inactivation of S. aureus and E. coli. The bacterial cells were eradicated by ROS produced upon irradiation [102].

Sf, phenazine compound can also be used as a sensitizer in electron transfer reactions in a homogeneous medium [104, 105, 106], semiconductors [106], polymeric medium [104] and as probes in the reverse micellar system [107, 108] due to its absorbs band in 500–550 nm [56, 109]. In addition, the antimicrobial photodynamic ability of Sf is exploited for the inactivation of microorganisms, such as Staphylococcus aureus and Escherichia coli [24, 43], Shigella flexneri, Bacillus subtilis [24], periodontopathogenic bacteria (biofilms in periodontal treatment) [97], and mitochondrial oxidation [95].

Among the compounds of this class, the most cited for PDT applications is Sf, which presents some photophysical studies with properties already defined. Sf has properties that allow its use as a PS in PDT, such as considerable singlet oxygen quantum yield (ΦΔ1O2) [33], an amphiphilic character that confers more significant interaction with biological substrates, does not present toxicity to healthy cells and generates reactive oxygen species (ROS), which inactivate microorganisms [101]. Although Sf presents several favorable properties to be applied in PDT, it has been poorly explored for this purpose. For example, spectroscopic studies on the interaction of Sf in liposomes L-egg lecithin phosphatidylcholine (PC) showed interaction. Still, the photodynamic effects of this formulation were not explored [57].

Sf incorporated into a silica matrix as a heterogeneous delivery system indicated that Sf has the potential for oxidation with singlet oxygen production [110]. Recent studies report Sf is incorporated into copolymer micelles [101]. This formulation ensured the Sf monomerization and preserved its physicochemical and photodynamic properties [33]. The use of triblock copolymers in the Sf incorporation guaranteed better results in PDT applications when compared to aqueous solutions [33].

Some authors evaluated the bactericidal effect of a serum combined with the action of Sf [24]. The authors did not use carrier systems, considering only the implications observed for the PS, not taking into account the self-aggregation processes characteristic of a PS in an aqueous medium. The photoefficacy of Sf in aqueous media was evaluated as an acaricide against female Hyalomma dromedarii ticks using in vitro immersion bioassays [99].

Similarly, Sasnauskiene et al. [95] evaluated the stimulated production of ROS by Sf in the inner space of mitochondria. The authors do not consider the self-aggregating effects resulting from the gradual release of PS in an aqueous medium [95].

Li et al. [96] explored the damage of bovine serum albumin (BSA) caused by Sf under ultrasonic irradiation [96]. The authors used Sf in an aqueous medium, a condition in which Sf tends to form self-aggregates that drastically reduce the 1O2 formation. Therefore, we propose the use of low-cost biocarrier systems, Pluronic® class. This triblock copolymer was used to solubilize Sf and maintain its photophysical properties [33]. Such attributes favored the prevention and treatment of mastitis via PDT [33, 100, 101].

Table 2 presents some photochemotherapeutic antimicrobial studies of phenazines found in the literature.

Table 2 shows a few amounts of work on this class of compounds and the way performed majority in vitro, which the scientific community can still better explore for applications in PDT. Combining the biocompatibility of the copolymer micelles and the non-toxicity of phenazines compounds, the continuity of pre-clinical studies for developing new photoactive-based phenazine formulations has been motivated for applications in different species of animals. These formulations can treat a wide range of diseases associated with different types of pathogenic microorganisms such as bacteria, fungi, viruses, and parasites. In this way, recent research has still been developed for the first time in vivo. It is in the submission phase of (unpublished) promising results concerning the prevention and treatment of mastitis. This review seeks to address the lack of literature on the approach of phenazines as a potential PS for PDT application. It is hoped that this work could offer some valuable information in developing new types of DDS systems.

Advertisement

7. Conclusion

Several products are used to control microorganisms and treat diseases like cancer; however, they present disadvantages such as cost and lack of effectiveness against resistant microorganisms. Photosensitization has recently gained attention benefiting from the use of a wide range of PS associated with a light source. The oxygen species produced by photoexcitation of a PS attack cancer cells or microorganisms non-selectively. Phenazine compounds showed promising phototoxicity, and their applicability has still been poorly investigated in the prevention and treatment of diseases caused by microorganisms and diseases related to abnormal cell growth, such as cancer. Furthermore, incorporating PS into polymeric micelles produced efficient, biocompatible formulations with better stability than aqueous systems.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Appendices and nomenclature

PDTPhotodynamic Therapy
aPDTantimicrobial Photodynamic Therapy
PSphotosensitizing compounds
ROSreactive oxygen species
3O2molecular oxygen
1O2singlet oxygen
O2−superoxide
H2O2hydrogen peroxide
ΦΔ1O2singlet oxygen quantum yield
LDLlow-density proteins
FDAFood and Drug Administration
DDSdrug delivery systems
ICinternal conversion
ISCintersystem crossing
CMCcritical micellar concentration
CMTcritical micellar temperature
HOMOHighest Occupied Molecular Orbital
LUMOLowest Unoccupied Molecular Orbital

References

  1. 1. Simplicio FI, Maionchi F, Hioka N. Terapia fotodinâmica: aspectos farmacológicos, aplicações e avanços recentes no desenvolvimento de medicamentos. Química Nova. 2002;25:801-807. DOI: 10.1590/S0100-40422002000500016
  2. 2. Dougherty TJ, Gomer CJ, Henderson BW, Jori G, Kessel D, Korbelik M, et al. Photodynamic therapy. JNCI: Journal of the national cancer institute. 1998;90:889-905. DOI: 10.1093/JNCI/90.12.889
  3. 3. Bonin E, dos Santos AR, da Silva AF, Ribeiro LH, Favero ME, Campanerut-Sá PAZ, et al. Photodynamic inactivation of foodborne bacteria by eosin Y. Journal of Applied Microbiology. 2018;124:1617-1628. DOI: 10.1111/jam.13727
  4. 4. de Morais FAP, Gonçalves RS, Braga G, Calori IR, Pereira PCS, Batistela VR, et al. Stable dipalmitoylphosphatidylcholine liposomes coated with an F127 copolymer for hypericin loading and delivery. ACS Applied Nano Materials. 2020;3:4530-4541. DOI: 10.1021/acsanm.0c00386
  5. 5. de Moeais FAP, Gonçalves RS, Campanholi KS, França BM, Capeloto OA, Lazarin-Bidoia D, et al. Photophysical characterization of hypericin-loaded in micellar, liposomal and copolymer-lipid nanostructures based F127 and DPPC liposomes. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscop. 2021;248:119173-112021. DOI: 10.1016/J.saa.2020.1419173
  6. 6. de Souza PCP, Costa PFA, Pellosi DS, Clori IR, Vilsinski BH, Estevão BM, et al. Photophysical properties and interaction studies of rose Bengal derivatives with biomimetic systems based in micellar aqueous solutions. Journal of Molecular Liquids. 2017;230:674-685. DOI: 10.1016/j.molliq.2017.01.055
  7. 7. Machado AEDH. Terapia fotodinâmica: princípios, potencial de aplicação e perspectivas. Química Nova. 2000;23:237-243. DOI: 10.1590/S0100-40422000000200015
  8. 8. Deeb D, Xu YX, Jiang H, Gao X, Janakiraman N, Chapman RA, et al. Curcumin (diferuloyl-methane) enhances tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in lncap prostate cancer cells1. Molecular Cancer Therapeutics. 2003;2:95-103
  9. 9. Zhu TC, Finaly JC. Prostate PDT dosimetry. Photodiagnosis and Photodynamic Therapy. 2006;3:234-246. DOI: 10.1016/j.pdpdt.2006.08.002
  10. 10. Bastos MM, Boechat N, Gomes ATPC, Neves AG, Cavaleiro J. O uso de porfirinas em terapia fotodinâmica no tratamento da leishmaniose cutânea. Revista Virtual de Química. 2012;4:257-267. DOI: 10.5935/1984-6835.20120021
  11. 11. de Oliveira EL, Ferreira SBS, Castro-Hoshino LV, Campanholi KSS, Calori IR, de Morais FAP, et al. Thermoresponsive hydrogel-loading Aluminum chloride Phthalocyanine as a drug release platform for topical administration in photodynamic therapy. Langmuir. 2021;37:3202-3213. DOI: 10.1021/acs.langmuir.1c00148
  12. 12. Woodcock J, Griffin JP, Behrman RE. Development of novel combination therapies. The New England Journal of Medicine. 2011;364:985-987. DOI: 10.1056/NEjMp1101548
  13. 13. de Oliveira KT, Souza JM, Globo NRS, Assis FF, Brocksom TJ. Conceitos fundamentais e Aplicações de Fotossensibilizadores do tipo Porfirinas, clorinas e Ftalocianinas em Terapias Fotônicas. Revista Virtual de Química. 2015;7:310-335. DOI: 10.5935/1984-6835.20150016
  14. 14. Ormond AB, Freeman HS. Dye sensitizers for photodynamic therapy. Materials. 2013;6:817-840. DOI: 10.3390/ma6030817
  15. 15. Yoon I, Li JZ, Shim YK. Advance in photosensitizers and light delivery for photodynamic therapy. Clinical Endoscopy. 2013;46:7-23. DOI: 10.5946/ce.2013.46.1.7
  16. 16. Braga G, Campanholi KSS, Ferreira SBS, Calori IR, Oliveira JH, Bruschi ML, et al. Tautomeric and aggregational dynamics of curcumin-supersaturated pluronic nanocarriers. ACS Applied Polymer Materials. 2020;2:4493-4511. DOI: 10.1021/acsapm.0c00589
  17. 17. Melo WC, Perussi JR. Comparando inativação fotodinâmica e antimicrobianos. Revista de Ciências Farmacêuticas Básica e Aplicada. 2012;33:331-340
  18. 18. Campanholi KSS, Zanqui AB, de Morais FAP, Jaski JM, Gonçalves RS, Silva-Junior RC, et al. Obtaining phytotherapeutic chlorophyll extracts using pressurized liquid technology. The Journal of Supercritical Fluids. 2022;180:105457. DOI: 10.1016/j.supflu.2021.105457
  19. 19. Byvaltsev VA, Bardonova LA, Onaka NR, Polkin RA, Ochkal SV, Shepelev VV, et al. Acridine orange: A review of novel applications for surgical cancer imaging and therapy. Frontiers in Oncology. 2019;9:925. DOI: 10.3389/fonc.2019.00925
  20. 20. Carobeli LR, Meirelles LEF, Damke GMZF, Damke E, Souza MVF, Mari NL, et al. Phthalocyanine and its formulations: A promising photosensitizer for cervical Cancer phototherapy. Pharmaceutics. 2021;13:2057. DOI: 10.3390/pharmaceutics13122057
  21. 21. Wiench R, Skaba D, Matys J, Grzech-Lesniak K. Efficacy of toluidine blue-mediated antimicrobial photodynamic therapy on Candida spp. A systematic review. Antibiotics. 2021;10:349. DOI: 10.3390/antibiotics10040349
  22. 22. Verissimo TV, Santos NT, Silva JR, Azevedo RB, Gomes AJ, Lunardi CN. In vitro cytotoxicity and phototoxicity of surface-modified gold nanoparticles associated with neutral red as a potential drug delivery system in phototherapy. Materials Science and Engineering: C. 2016;65:199-204. DOI: 10.1016/j.msec.2016.04.030
  23. 23. Miksa B, Sierant M, Slorupska E, Michalski A, Kazmierski S, Steinke U, et al. Chlorambucil labelled with the phenosafranin scaffold as a new chemotherapeutic for imaging and cancer treatment. Colloids and Surfaces. B, Biointerfaces. 2017;159:820-828. DOI: 10.1016/J.COLSURFB.2017.08.045
  24. 24. Jankowski A, Jankowski S, Mironczyk A, Niedbach J. The action of photosensitizers and serum in a bactericidal process. II. The effects of dyes: Hypericin, eosin Y and saphranine O. Polish Journal of Microbiology. 2005;54:323-330
  25. 25. Vilsinsk BH, Witt MA, Barbosa PM, Montanha MC, Nunes CS, Bellettini IC, et al. Formulation of chloroaluminum phthalocyanine incorporated into PS-b-PAA diblock copolymer nanomicelles. Journal of Molecular Liquids. 2018;271:949-958. DOI: 10.1016/j.molliq.2018.09.034
  26. 26. Rossin ARS, de Oliveira EL, de Morais FAP, Silva-Junior RC, Scheid DT, Caetano W, et al. Terapia Fotodinâmica em Eletrofiação: Revisão de técnicas e aplicações. Química Nova. 2020;43:613-622. DOI: 10.21577/0100-4042.20170524
  27. 27. Lakowicz JR. Principles of Fluorescence Spectroscopy. Boston, US: Springer Science + Business Media, LLC; 2006. ISBN: 978-0-387-46312-4
  28. 28. Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, et al. Photodynamic therapy of cancer: An update. CA: A Cancer Journal for Clinicians. 2011;61:250-281. DOI: 10.3322/CAAC.20114
  29. 29. Dai T, Huang Y-Y, Hamblin MR. Photodynamic therapy for localized infections state of the art. Photodiagnosis and Photodynamic Therapy. 2009;6:170-188. DOI: 10.1016/j.pdpdt.2009.10.008
  30. 30. Kubler AC. Photodynamic therapy. Medical Laser Application. 2005;20:37-45. DOI: 10.1016/j.mla.2005.02.001
  31. 31. Kharkwal GB, Sharma SK, Huang Y-Y, Dai T, Hamblin MR. Photodynamic therapy for infections: Clinical applications. Lasers in Surgery and Medicine. 2011;43:755-767. DOI: 10.1002/LSM.21080
  32. 32. Saraiva BB, Rodrigues BM, Silva-Junior RC, Scapim MRS, Lancheros CAC, Nakamura CV, et al. Photodynamic inactivation of Pseudomonas fluorescens in Minas Frescal cheese using curcumin as a photosensitizer. LWT. 2021;151:112143. DOI: 10.1016/j.lwt.2021.112143
  33. 33. da Silva-Junior RC, Campanholi KSS, de Morais FAP, Pozza MSS, Santos GT, Hioka N, et al. Development and applications of safranine-loaded Pluronic® F127 and P123 photoactive nanocarriers for prevention of bovine mastitis: In vitro and in vivo studies. Dyes and Pigments. 2019;167:204-215. DOI: 10.1016/j.dyepig.2019.04.037
  34. 34. Kataoka K, Harada A, Nagaski Y. Block copolymer micelles for drug delivery: Design, characterization and biological significance. Advanced Drug Delivery Reviews. 2012;64:37-48. DOI: 10.1016/j.addr.2012.09.013
  35. 35. Rijcken CJF, Hofman JW, van Zeeland F, Hennink WE, van Nostrum CF. Photosensitiser-loaded biodegradable polymeric micelles: Preparation, characterisation and in vitro PDT efficacy. Journal of Controlled Release. 2007;124:144-153. DOI: 10.1016/j.conrel.2007.09.002
  36. 36. de Morais FAP, Emuno-Junior A, Gonçalves RS, Cesar GB, Miranda N, Vilsinski BH, et al. Hypericin photodynamic activity. Part III: in vitro evaluation in different nanocarriers against trypomastigotes of Trypanosoma cruzi. Photochemical & Photobiological Sciences. 2019;18:487-494. DOI: 10.1039/c8pp00444g
  37. 37. Li Y, Jang WD, Nishiyama N, Kishimura A, Kawauchi S, Morimoto Y, et al. Dendrimer generation effects on photodynamic efficacy of dendrimer porphyrins and dendrimer-loaded supramolecular nanocarriers. Chemistry of Materials. 2007;19:5557-5562. DOI: 10.1021/cm071451m
  38. 38. Mihoub AB, Larue L, Moussaron A, Youssef Z, Colombeau L, Baros F, et al. Use of cyclodextrins in anticancer photodynamic therapy treatment. Molecules. 2018;23:1936. DOI: 10.339/molecules23081936
  39. 39. Semeraro P, Chimient G, Altamura E, Fini P, Rizzi V, Cosma P. Chlorophyll a in cyclodextrin supramolecular complexes as a natural photosensitizer for photodynamic therapy (PDT) applications. Materials Science and Engineering: C. 2018;85:47-56. DOI: 10.1016/j.msec.2017.12.012
  40. 40. Wang J, Liu L, Chen J, Deng M, Feng X, Chen L. Supramolecular nanoplatforms via cyclodextrin host-guest recognition for synergistic gene-photodynamic therapy. European Polymer Journal. 2019;118:222-230. DOI: 10.1016/jeurpolymj.2019.04.051
  41. 41. Campanholi KSS, Silva-Junior RC, Silva IC, Santos RS, Vecchi CF, Bruschi ML, et al. Stimulus-responsive phototherapeutic micellar platform of rose Bengal B: A new perspective for the treatment of wounds. Journal of Drug Delivery Science and Technology. 2021;2021:102739. DOI: 10.1016/j.jddst.2021.102739
  42. 42. do Nascimento GM, Oliveira RC, Pradie NA, PRG L, Worfel PR, Martinez GR, et al. Single-wall carbon nanotubes modified with organic dyes: Synthesis, characterization and potential cytotoxic effects. Journal of Photochemistry and Photobiology A: Chemistry. 2010;211:99-107. DOI: 10.1016/j.jphotochem.2010.01.019
  43. 43. Beech SJ, Noimark S, Page K, Noor N, Allan E, Parkin IP. Incorporation of crystal violet, methylene blue and safranin O into a copolymer emulsion; the development of a novel antimicrobial paint. RSC Advances. 2015;5:26364-26375. DOI: 10.1039/c5ra01673h
  44. 44. Drogat N, Granet R, Morvan CL, Begaud-Grimaud G, Krausz P, Sol V. Chlorin-PEI-labeled cellulose nanocrystals: Synthesis, characterization and potential application in PDT. Bioorganic & Medicinal Chemistry Letters. 2012;22:3648-3652. DOI: 10.1016/j.bmcl.2012.04.044
  45. 45. Jiang S, Zhu R, He X, Wang J, Wang M, Qian Y, et al. Enhanced photocytotoxicity of curcumin delivered by solid lipid nanoparticles. International Journal of Nanomedicine. 2017;12:167-178. DOI: 10.2147/ijn.s123107
  46. 46. Bechet D, Couleaud P, Frochot C, Virot ML, Guillemin F, Barberi-Heyob M. Nanoparticles as vehicles for delivery of photodynamic therapy agents. Trends in Biotechnology. 2008;26:612-621. DOI: 10.1016/j.tibtech.2008.07.007
  47. 47. Malmsten M. Surfactants and Polymers in Drug Delivery. Boca Raton, US: CRC Press; 2002. DOI: 10.1201/9780824743758
  48. 48. van Nostrum CF. Polymeric micelles to deliver photosensitizers for photodynamic therapy. Advanced Drug Delivery Reviews. 2004;56:9-16. DOI: 10.1016/J.ADDR.2003.07.013
  49. 49. Batrakova EV, Kabavov AV. Pluronic block copolymers: Evolution of drug delivery concept from inert nanocarriers to biological response modifiers. Journal of Controlled Release. 2008;130:98-106. DOI: 10.1016/j.jconrel.2008.04.013
  50. 50. Senapati S, Mahanta AK, Kumar S, Maiti P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduction and Targeted Therapy. 2018;3:1-19. DOI: 10.1038/s41392-017-0004-3
  51. 51. Lima AM, Pizzol CD, Monteiro FBF, Creczynski-Pasa TB, Andrade GP, Ribeiro AO, et al. Hypericin encapsulated in solid lipid nanoparticles: Phototoxicity and photodynamic efficiency. Journal of Photochemistry and Photobiology B: Biology. 2013;125:146-154. DOI: 10.1016/j.jphotobiol.2013.05.010
  52. 52. Campanholi KSS, Braga G, Rocha NL, Francisco LMB, Oliveira EL, Bruschi ML, et al. Biomedical platform development of a chlorophyll-based extract for topic photodynamic therapy: Mechanical and spectroscopic properties. Langmuir. 2018;34:8230-8244. DOI: 10.1021/acs.langmuir.8b00658
  53. 53. Gerola A, Costa PF, de Morais FA, Tsubone TA, Calared AO, Nakamura CV, et al. Liposome and polymeric micelle-based delivery systems for chlorophylls: Photodamage effects on Staphylococcus aureus. Colloids and Surfaces B: Biointerfaces. 2019;177:487-495. DOI: 10.1016/j.colsurfb.2019.02.032
  54. 54. George A. Advances in biomimetics. BoD–Books on Demand. Rijeka, Croatia: InTech; 2011. ISBN 978-953-307-191-6
  55. 55. Neuman MG, Gessner F, Oliveira VA. Formation of mixed dimers in solutions of basic dyes. Journal of the Chemical Society, Faraday Transactions. 1990;86:3551-3555. DOI: 10.1039/ft9908603551
  56. 56. Pastre IA, Neumann MG. Protonation of the triplet state of safranine in the presence of cationic micelles and polyelectrolytes. Journal of Photochemistry and Photobiology, A: Chemistry. 1994;79:115-119. DOI: 10.1016/1010-6030(93)03744-2
  57. 57. Bozkurt E, Bayraktutan T, Mahmut A, Toprak M. Spectroscopic studies on the interaction of fluorescein and safranine T in PC liposomes. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy. 2013;101:31-35. DOI: 10.1016/j.saa.2012.09.082
  58. 58. Junqueira MV, Borghi-Pangoni FB, Ferreira SBS, Rabello BR, Hioka N, Bruschi ML. Functional polymeric systems as delivery vehicles for methylene blue in photodynamic therapy. Langmuir. 2016;32:19-27. DOI: 10.1021/acs.langmuir.5b02039
  59. 59. Ferreira SBDS, Silva JB, Junqueira MV, Borghi-PangoniaRaquel FB, Gomes RG, Bruschi ML, et al. The importance of the relationship between mechanical analyses and rheometry of mucoadhesive thermoresponsive polymeric materials for biomedical applications. Journal of the Mechanical Behavior of Biomedical Materials. 2017;74:142-153. DOI: 10.1016/j.jmbbm.2017.05.040
  60. 60. Trigo-Gutierrez JK, Zanatta GC, Ortega ALM, Balastegui MIC, Sanita PV, Pavarina AC, et al. Encapsulation of curcumin in polymeric nanoparticles for antimicrobial Photodynamic Therapy. PLoS One. 2017;12:e0187418. DOI: 10.1371;journal.pone.0187418
  61. 61. Gerola AP, de Morais FAP, Costa PFA, Kimura E, Caetano W, Hioka N. Characterization of chlorophyll derivatives in micelles of polymeric surfactants aiming photodynamic applications. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy. 2017;173:213-221. DOI: 10.1016/j.saa.2016.09.019
  62. 62. Vukicevic M, Hegge AB, Vulic P, Tonnesen H. Poloxamer-based curcumin solid dispersions for ex tempore preparation of supersaturated solutions intended for antimicrobial photodynamic therapy. Pharmaceutical Development and Technology. 2015;20:863-871. DOI: 10.3109/10837450.2014.930489
  63. 63. Costa PFA, Gerola AP, Pereira PCS, Souza BS, Caetano W, Fiedler HD, et al. Chlorophylls B formulated in nanostructured colloidal solutions: Interaction, spectroscopic, and photophysical studies. Journal of Molecular Liquids. 2019;274:393-401. DOI: 10.1016/j.molliq.2018.10.143
  64. 64. Alexandridis P, Holzwarth JF, Hatton TA. Micellization of poly (ethylene oxide)-poly (propylene oxide)-poly (ethylene oxide) triblock copolymers in aqueous solutions: Thermodynamics of copolymer association. Macromolecules. 1994;27:2414-2425
  65. 65. Freitas CF, Rocha NL, Pereverzieff IS, Batistela VR, Malacarne LC, Hioka N, et al. Potential of triblock copolymers Pluronic® P-84 and F-108 with erythrosine B and its synthetic ester derivatives for photodynamic applications. Journal of Molecular Liquids. 2021;322:114904. DOI: 10.1016/j.molliq.2020.114904
  66. 66. Alizadeh AM, Sadeghizadeh M, Najafi F, Ardestani SK, Erfani-Moghadam V, Khanik M, et al. Encapsulation of curcumin in diblock copolymer micelles for cancer therapy. BioMed Research International. 2015;2015:1-13. DOI: 10.1155/2015/824746
  67. 67. Obata M, Masuda S, Takahashi M, Yazaki K, Hirohara S. Effect of the hydrophobic segment of an amphiphilic block copolymer on micelle formation, zinc phthalocyanine loading, and photodynamic activity. European Polymer Journal. 2021;147:10325. DOI: 10.1016/j.eurpolymj.2021.110325
  68. 68. Mavrodi DV, Bonsall RF, Delaney SM, Soule MJ, Philips G, Thomashow LS. Functional analysis of genes for biosynthesis of pyocyanin and phenazine-1-carboxamide from Pseudomonas aeruginosa PAO1. Journal of Bacteriology. 2001;183:6454-6465. DOI: 10.1128/jb.183.21.6454-6465.2001
  69. 69. Yan J, Liu W, Cai J, Wang Y, Li D, Hua H, et al. Advances in Pphenazines over the past decade: Review of their pharmacological activities, mechanisms of action, biosynthetic pathways and synthetic strategies. Marine Drugs. 2021;19:610. DOI: 10.3390/md19110610
  70. 70. Stover CK, Pham XQ , Erwin AL, Mizoguchi SD, Warrener P, Hickey MJ, et al. Complete genome sequence of Pseudomonas aeruginosa PAO1, an opportunistic pathogen. Nature. 2000;406:959-964
  71. 71. Abu EA, Su S, Sallans L, Boissy RE, Greatens A, Heineman WR, et al. Cyclic voltammetric, fluorescence and biological analysis of purified aeruginosin a, a secreted red pigment of Pseudomonas aeruginosa PAO1. Microbiology. 2013;159:1736-1747. DOI: 10.1099/mic.0.065235-0
  72. 72. Kennedy RK, Veena V, Naik PR, Lakshmi P, Krishna R, Sudharani S, et al. Phenazine-1-carboxamide (PCN) from Pseudomonas sp. strain PUP6 selectively induced apoptosis in lung (A549) and breast (MDA MB-231) cancer cells by inhibition of antiapoptotic Bcl-2 family proteins. Apoptosis. 2015;20:858-868. DOI: 1007/s10495-015-1118-0
  73. 73. Mahan AM, Ragab SS, Hashem A, Ali MM, Nada AA. Synthesis and antiproliferative activity of novel polynuclear heterocyclic compounds derived from 2, 3-diaminophenazine. European Journal of Medicinal Chemistry. 2015;90:568-576. DOI: 10.1016/j.ejmech.2013.12.007
  74. 74. Moris MA, Andrieu C, Rocchi P, Seillan C, Acunzo J, Brunel F, et al. 2, 3-Dialkoxyphenazines as anticancer agents. Tetrahedron Letters. 2015;56:2695-2698. DOI: 10.1016/j.tetlet.2015.04.003
  75. 75. Reddy MV, Valasani KR, Lim KT, Jeong YT. Tetrame thylguanidiniumchlorosulfonate ionic liquid (TMG IL): An efficient reusable catalyst for the synthesis of tetrahydro-1 H-benzo [a] chromeno [2, 3-c] phenazin-1-ones under solvent-free conditions and evaluation for their in vitro bioassay activity. New Journal of Chemistry. 2015;39:9931-9941. DOI: 10.1039/C5NJ01866H
  76. 76. Lu Y, Wang L, Wang X, Xi T, Liao J, Wang Z, et al. Design, combinatorial synthesis and biological evaluations of novel 3-amino-1′-((1-aryl-1H-1, 2, 3-triazol-5-yl) methyl)-2′-oxospiro [benzo [a] pyrano [2, 3-c] phenazine-1, 3′-indoline]-2-carbonitrile antitumor hybrid molecules. European Journal of Medicinal Chemistry. 2017;135:125-141. DOI: 10.1016/j.ejmech.2017.04.040
  77. 77. Cimmino A, Evidente A, Mathieu V, Andolfi A, Lefranc F, Kornienco A, et al. Phenazines and cancer. Natural Product Reports. 2012;29:487-501. DOI: 10.1039/c2np00079b
  78. 78. Pachon OG, Azqueta A, Lavaggi ML, Cerani AL, Creppy E, Collins A, et al. Antitumoral effect of phenazine N 5, N 10-dioxide derivatives on Caco-2 cells. Chemical Research in Toxicology. 2008;21:1578-1585. DOI: 10.1021/tx80002k
  79. 79. Lavaggi ML, Aguirre G, Boiani L, Orelli L, Garcia B, Cerecetto H, et al. Pyrimido [1, 2-a] quinoxaline 6-oxide and phenazine 5, 10-dioxide derivatives and related compounds as growth inhibitors of Trypanosoma cruzi. European Journal of Medicinal Chemistry. 2008;43:1737-1741. DOI: 10.1016/j.ejmech.2007.10.031
  80. 80. Cardozo VF, Oliveira AG, Nishio EK, Perugini MRE, Andrade CGTJ, Silveira WD, et al. Antibacterial activity of extracellular compounds produced by a Pseudomonas strain against methicillin-resistant Staphylococcus aureus (MRSA) strains. Annals of Clinical Microbiology and Antimicrobials. 2013;12:1-8
  81. 81. Gorantla JN, Kumar SN, Nisha GV, Sumandu AS, Dileep C, Sudaresan A, et al. Purification and characterization of antifungal phenazines from a fluorescent Pseudomonas strain FPO4 against medically important fungi. Journal de mycologie médicale. 2014;24:185-192. DOI: 10.1016/j.mycmed. 2014.02.003
  82. 82. Tupe SG, Kulkarni RR, Shirazi F, Sant DG, Joshi SP, Deshpande MV. Possible mechanism of antifungal phenazine-1-carboxamide from Pseudomonas sp. against dimorphic fungi B enjaminiella poitrasii and human pathogen Candidda albicans. Journal of Applied Microbiology. 2015;118:39-48. DOI: 10.1111/jam.12675
  83. 83. Cha JW, Lee S, Kim MC, Thida M, Lee JW, Park JS, et al. Pontemazines a and B, phenazine derivatives containing a methylamine linkage from Streptomyces sp. UT1123 and their protective effect to HT-22 neuronal cells. Bioorganic & Medicinal Chemistry Letters. 2015;25:5083-5086. DOI: 10.1016/j.bmcl. 2015.10.019
  84. 84. Yao BL, Mai YW, Chen SB, Xie HT, Yao PF, Ou TM, et al. Design, synthesis and biological evaluation of novel 7-alkylamino substituted benzo [a] phenazin derivatives as dual topoisomerase I/II inhibitors. European Journal of Medicinal Chemistry. 2015;92:540-553. DOI: 10.1016/j.ejmech.2015.01.024
  85. 85. Kennedy RK, Naik PR, Venna V, Lakshmi BS, Lakshmi P, Krishna R, et al. 5-methyl phenazine-1-carboxylic acid: A novel bioactive metabolite by a rhizosphere soil bacterium that exhibits potent antimicrobial and anticancer activities. Chemico-Biological Interactions. 2015;231:71-82. DOI: 10.1016/j.cbi. 2015.03.002
  86. 86. Varsha KK, Nishant G, Sneha SM, Shilpa G, Devendra L, Priya S, et al. Antifungal, anticancer and aminopeptidase inhibitory potential of a phenazine compound produced by Lactococcus BSN307. Indian Journal of Microbiology. 2016;56:411-416. DOI: 10.1007/s12088-016-0597-1
  87. 87. Ali HM, Shikh MS, Salem MZ. Isolation of bioactive phenazine-1-carboxamide from the soil bacterium Pantoea agglomerans and study of its anticancer potency on different cancer cell lines. Journal of AOAC International. 2016;99:1233-1239. DOI: 10.5740/jaoacint.16-0090
  88. 88. Makgatho E, Mbajiorgu E. In vitro investigation of clofazimine analogues for antiplasmodial, cytotoxic and pro-oxidative activities. African Health Sciences. 2017;17:191-198. DOI: 10.4314/ahs.v17i1.24
  89. 89. Krishnaiaha M, Almeida NR, Udumula V, Song Z, Chhonker YS, Abdelmoaty MM, et al. Synthesis, biological evaluation, and metabolic stability of phenazine derivatives as antibacterial agents. European Journal of Medicinal Chemistry. 2018;143:936-947. DOI: 10.1016/j.ejmech.2017.11.026
  90. 90. Liao J, Wang L, Wu Z, Wan Z, Chen J, Zhong Y, et al. Identification of phenazine analogue as a novel scaffold for thioredoxin reductase I inhibitors against Hep G2 cancer cell lines. Journal of Enzyme Inhibition and Medicinal Chemistry. 2019;34:1158-1163. DOI: 10.1080/14756366.2019.1624541
  91. 91. Gu P-Y, Zhao Y, He JH, Zhang J, Wang C, Xu Q-F, et al. Synthesis, physical properties, and light-emitting diode performance of phenazine-based derivatives with three, five, and nine fused six-membered rings. The Journal of organic chemistry. 2015;80:3030-3035. DOI: 10.1021/jo5027707
  92. 92. Liu X, Yang Z, Xu W, Chu Y, Yang J, Yan Y, et al. Fine tuning of pyridinium-functionalized dibenzo [a, c] phenazine near-infrared AIE fluorescent biosensors for the detection of lipopolysaccharide, bacterial imaging and photodynamic antibacterial therapy. Journal of Materials Chemistry C. 2019;7:12509-12517. DOI: 10.1039/C9TC04427B
  93. 93. Udumula V, Endres JL, Harper CN, Jaramillo L, Zhong HA, Bayles KW, et al. Simple synthesis of endophenazine G and other phenazines and their evaluation as anti-methicillin-resistant Staphylococcus aureus agents. European Journal of Medicinal Chemistry. 2017;125:710-721. DOI: 10.1016/j.ejmech.2016.09.079
  94. 94. Elhadyv HA, Mekawy RE, Fadda A. Valuable chemistry of phenazine derivatives: Synthesis, reactions and, applications. European Journal of Medicinal Chemistry. 2020;12:2894-2926. DOI: 10.1080/10406638.2020.1833051
  95. 95. Sasnauskiene A, Kadziauskas J, Vezelyte N, Jonusiene V, Kirveliene V. Apoptosis, autophagy and cell cycle arrest following photodamage to mitochondrial interior. Apoptosis. 2009;14:276-286. DOI: 10.1007/s10495-008-0292-8
  96. 96. Li J, Wang Z, Wang J, Gao J, Zou M, Li Y, et al. Spectroscopic investigation on the sonodynamic activity of Safranine T to bovine serum albumin damage. Journal of Luminescence. 2012;132:282-288. DOI: 10.1016/jlumin.2011.09.014
  97. 97. Voos AC, Krans DMD, Tonndorf-martini S, Dipl-Ing AV, Sigusch H, Staudte H, et al. Photodynamic antimicrobial effect of safranine O on an ex vivo periodontal biofilm. Lasers in Surgery and Medicine. 2014;46:235-243. DOI: 10.1002/Ism.22217
  98. 98. Urrutia MN, Ortiz CS, Alovero FL. Mechanistic insight into the photodynamic effect mediated by neutral red and a new Azine compound in Staphylococcus aureus cells. Chemistry & Biodiversity. 2019;16:e1900262. DOI: 10.1002/cbdv.201900262
  99. 99. Khater H, Hendawy N, Govindarajan M, Marugan K, Banelli G. Photosensitizers in the fight against ticks: Safranin as a novel photodynamic fluorescent acaricide to control the camel tick Hyalomma dromedarii (Ixodidae). Parasitology Research. 2016;15:3747-3758. DOI: 10.1007/s00436-016-5136-9
  100. 100. da Silva-Junior RC, Campanholi KSS, de Morais FAP, Pozza MSS, Castro-Hoshino LV, Baesso ML, et al. Photothermal stimuli-responsive hydrogel containing Safranine for mastitis treatment in veterinary using phototherapy. ACS Applied Bio Materials. 2020;4:581-596. DOI: 10.1021/acsabm.0.c01143
  101. 101. da Silva-Junior RC, Campanholi KSS, Rando FS, Gonçalves RS, Lazarin-Bidóia D, de Morais FAP, et al. Antimicrobial photoinactivation approach based on Safranine-O loaded F127 copolymeric micelles for control of gram-negative and gram-positive bacteria. Dyes and Pigments. 2021;197:109900. DOI: 10.1016/j.dyepog.2021.109900
  102. 102. Rozga-Wijas K, Bak-Sypien I, Turecka K, Narajczyk M, Waleron K. Cationic Phenosafranin photosensitizers based on polyhedral Oligomeric Silsesquioxanes for inactivation of gram-positive and gram-negative Bacteria. International Journal of Molecular Sciences. 2021;22:13373. DOI: 10.3390/ijms222413373
  103. 103. Urrutia MN, Alovero FL, Ortiz CS. New azine compounds as photoantimicrobial agents against Staphylococcus aureus. Dyes and Pigments. 2015;116:27-35. DOI: 10.1016/j.dyepig.2014.12.021
  104. 104. Gopidas K, Kamat PV. Photophysics and photochemistry of phenosafranin dye in aqueous and acetonitrile solutions. Journal of Photochemistry and Photobiology, A: Chemistry. 1989;48:291-301. DOI: 10.1016/1010-6030(89)87010-8
  105. 105. Jockusch S, Tempo H-J, Schnabel W, Turro NJ. Photoinduced energy and electron transfer between ketone triplets and organic dyes. The Journal of Physical Chemistry A. 1997;101:440-445. DOI: 10.1021/jp961744v
  106. 106. Jayanthi SS, Ramamurthy P. Excited singlet state reaction of phenosafranine with electron donors Role of the heavy-atom effect in triplet induction. Journal of the Chemical Society, Faraday Transactions. 1997;94:1675-1679. DOI: 10.1039/A800455B
  107. 107. Chaudhuri R, Sengupta PK, Mukherjee KR. Luminescence behaviour of phenosafranin in reverse micelles of AOT in n-heptane. Journal of Photochemistry and Photobiology, A: Chemistry. 1997;108:261-265. DOI: 10.1016/S1010-6030(97)00091-9
  108. 108. Broglia MF, Bertolotti SG, Previtali CM. Proton and electron transfer in the excited state quenching of phenosafranine by aliphatic amines. Photochemistry and Photobiology. 2007;83:535-541. DOI: 10.1562/2006-07-31-ra-989
  109. 109. Bertolotti SG, Previtali CM. The excited states quenching of safranine T by p-benzoquinones in polar solvents. Journal of Photochemistry and Photobiology, A: Chemistry. 1997;103:115-119. DOI: 10.1016/s1010-6030(97)85300-2
  110. 110. Albiter E, Alfaro S, Valenzuela MA. Photosensitized oxidation of 9, 10-dimethylanthracene with singlet oxygen by using a safranin O/silica composite under visible light. Photochemical & Photobiological Sciences. 2015;14:597-602. DOI: 10.1039/c4pp00261j

Written By

Ranulfo Combuca da Silva Junior, Katieli da Silva Souza Campanholi, Flávia Amanda Pedroso de Morais, Laura Adriane de Moraes Pinto, Fabiana dos Santos Rando, Magali Soares dos Santos Pozza and Wilker Caetano

Submitted: 09 December 2022 Reviewed: 17 February 2023 Published: 20 March 2023