Antimicrobial photodynamic therapy

Last updated

Antimicrobial photodynamic therapy (aPDT), also referred to as photodynamic inactivation (PDI), photodisinfection (PD), or photodynamic antimicrobial chemotherapy (PACT), is a photochemical antimicrobial method that has been studied for over a century. [1] Supported by in vitro, in vivo and clinical studies, aPDT offers a treatment option for broad-spectrum infections, particularly in the context of rising antimicrobial resistance. [2] [3] Its multi-target mode of action allows aPDT to be a viable therapeutic strategy against drug-resistant microorganisms. [4] The procedure involves the application of photosensitizing compounds, also called photoantimicrobials, which, upon activation by light, generate reactive oxygen species (ROS). These ROS lead to the oxidation of cellular components of a wide array of microbes, including pathogenic bacteria, fungi, protozoa, algae, and viruses. [5]

Contents

Historical perspective

In the early 20th century, decades before the first chemical antibiotics were developed, Dr. Niels Finsen discovered that blue light could be used to treat skin infections. [6] In the following years, Finsen's phototherapy was used in many European medical institutions as a topical antimicrobial. [7] In 1903, the Nobel Prize committee awarded him for his work in Physiology/Medicine, "in recognition of his contribution to the treatment of diseases, especially lupus vulgaris, with concentrated light radiation, whereby he has opened a new avenue for medical science". [8]

Similarly, in the beginning of the 20th century, Oscar Raab, a German medical student supervised by Professor Herman Von Tappeiner, accidentally made a scientific observation of the antimicrobial effects of light-activated dyes. [7] While conducting experiments on the viability of motile protozoa, Raab noticed that fluorescent dyes, like some acridine and xanthene dyes, could kill stained microbes when sunlight was directed onto the stained samples. These effects were particularly pronounced during the summer, when sunlight is brightest. This chance observation highlighted the ability of certain fluorescent compounds, now termed "photosensitizers" (PS), to artificially induce light sensitivity in microorganisms and enhance the known antimicrobial effects of sunlight. Shortly thereafter, Von Tappeiner and Jodlbauer discovered that oxygen was crucial for light-mediated reactions, leading to the creation of the term "photodynamische wirkung" (photodynamic effect). [7]

However, it wasn't until the 1970s that researchers began to systematically explore the potential of photodynamic therapy for medical applications. [9] Since then, significant progress has been made in understanding the underlying mechanisms and optimizing the efficacy of photodynamic therapy (PDT) for treatment of cancers and age-related macular degeneration. Today, the branch of PDT focused on killing microbial cells is considered as an option to prevent and treat infectious diseases in a manner that avoids the emergence of antimicrobial drug-resistance. [5]

Mechanism of action

Illustrative scheme of photodynamic reactions. The photosensitizer absorbs light and is promoted from its ground singlet state ( PS) to an excited singlet state ( PS*). Alternatively, the photosensitizer can convert to an excited triplet state ( PS*) by intersystem crossing. This is a longer-living state that allow sufficient time for chemical reactions to occur. A photosensitizer in PS* state can return to ground state ( PS) either by emitting phosphorescence, or by photochemical reactions that occur through transfer of charges or energy. These photochemical reactions can locally generate cytotoxic reactive oxygen species (ROS) via the Type I or II photodynamic reactions. In a cellular microenvironment, these ROS have a short lifespan (<10ms), and react with and destroy biomolecules, such as proteins, carbohydrates, nucleic acids, and lipids, very close (<1mm) to the production site. Type I: Charges, such as electrons, are transferred to surrounding substrates (R), forming radicals (R ) due to the presence of the unpaired electron that was received. Molecular oxygen (O2) participates directly or indirectly in this reaction pathway forming the radical anion known as superoxide (O2 ). The superoxide radical can be further reduced to form hydrogen peroxide (H2O2), which can also be reduced to form highly reactive free hydroxyl radicals (HO ) via Fenton-like reactions. Type II: Energy is transferred to ground state triplet molecular oxygen ( O2), creating singlet oxygen ( O2*), an excited form of oxygen that is much more reactive than its ground state triplet counterpart. PS = Ground Singlet State of Photosensitizer; PS* = First Excited Singlet State of Photosensitizer; PS* = First Excited Triplet State of Photosensitizer; ISC = Intersystem Crossing; O2 = Ground State Triplet Oxygen; O2 = Excited State Singlet Oxygen. Photodynamic reaction.jpg
Illustrative scheme of photodynamic reactions. The photosensitizer absorbs light and is promoted from its ground singlet state ( PS) to an excited singlet state ( PS*). Alternatively, the photosensitizer can convert to an excited triplet state ( PS*) by intersystem crossing. This is a longer-living state that allow sufficient time for chemical reactions to occur. A photosensitizer in PS* state can return to ground state ( PS) either by emitting phosphorescence, or by photochemical reactions that occur through transfer of charges or energy. These photochemical reactions can locally generate cytotoxic reactive oxygen species (ROS) via the Type I or II photodynamic reactions. In a cellular microenvironment, these ROS have a short lifespan (<10μs), and react with and destroy biomolecules, such as proteins, carbohydrates, nucleic acids, and lipids, very close (<1μm) to the production site. Type I: Charges, such as electrons, are transferred to surrounding substrates (R), forming radicals (R ) due to the presence of the unpaired electron that was received. Molecular oxygen (O2) participates directly or indirectly in this reaction pathway forming the radical anion known as superoxide (O2 ). The superoxide radical can be further reduced to form hydrogen peroxide (H2O2), which can also be reduced to form highly reactive free hydroxyl radicals (HO ) via Fenton-like reactions. Type II: Energy is transferred to ground state triplet molecular oxygen ( O2), creating singlet oxygen ( O2*), an excited form of oxygen that is much more reactive than its ground state triplet counterpart. PS = Ground Singlet State of Photosensitizer; PS* = First Excited Singlet State of Photosensitizer; PS* = First Excited Triplet State of Photosensitizer; ISC = Intersystem Crossing; O2 = Ground State Triplet Oxygen; O2 = Excited State Singlet Oxygen.

The photochemical principle underlying antimicrobial photodynamic therapy involves the activation of a photosensitizer, a light-sensitive compound that can locally generate reactive products, such as radicals and reactive oxygen species (ROS), upon exposure to specific wavelengths of light. [10] An ideal photosensitizer selectively accumulates in the target microbial cells, where it remains inactive and non-toxic until it is activated by irradiation with light of a specific wavelength. This activation promotes the photosensitizer molecules to a short-lived excited state that possesses different chemical reactivity relative to its ground-state counterpart. When the photosensitizer molecule is in an excited triplet state, it can induce local Type 1 photodynamic reactions by direct contact with molecular oxygen, inorganic ions or biological targets. [11] [12] These redox reactions (Type 1) involve charge transfers, by donation of electron (e) or Hydrogen ion (H+), to form radicals and ROS, such as anion radical superoxide, hydrogen peroxide and hydroxyl radicals. [10] The excited triplet-state photosensitizer can also transfer energy to molecular triplet-state oxygen producing singlet oxygen via Type 2 photodynamic reactions. [10] The photoinduced burst of active reactants affect cellular redox regulations and can cause oxidative damage to vital structures made of proteins, lipids, carbohydrates and nucleic acids, leading to localized cellular death. [13] [11]

Efficacy against drug-resistant pathogens

The efficacy of antimicrobial photodynamic therapy, using various distinct photosensitizers, has been studied since the 1990s. [9] [7] Most studies have yielded positive outcomes, often achieving disinfection levels, as defined by infection control guidelines, exceeding 5 log10 (99.999%) of microbial inactivation. [14] Over the past decade, a collection of novel photoantimicrobials has been developed, exhibiting improved efficiencies in antimicrobial photodynamic action against various bacterial species. [5] These studies have primarily focused on the inactivation of planktonic cultures, which are free-floating bacterial cells. This method serves as a convenient approach for high-throughput antimicrobial screening of multiple compounds, such as evaluating whether minor chemical modifications to a given photosensitizer can enhance antimicrobial efficacy. [15] However, when present in biofilms, microbial populations can exhibit distinct characteristics compared to their planktonic counterparts, including significantly higher tolerance towards antimicrobials (up to 1,000-fold). [16] Among the various factors contributing to this enhanced tolerance is the biofilm matrix composed extracellular polymeric substance (EPS). The EPS can shield constituent bacteria from antimicrobials through dual mechanisms: 1) by impeding the penetration of antimicrobial substances throughout the biofilm due to interactions between positively charged agents and negatively charged EPS residues, and also by 2) redox processes and π-π interactions involving aromatic surfaces generally acting to dismute the incoming active substance. EPS must be considered in the rational design of antimicrobial photosensitizers, because the densely cross linked matrix may also obstruct diffusion of photosensitizer into deeper biofilm layers. [17]

The multi-target mechanisms of aPDT avoid antimicrobial resistance, which continues to be a major global health concern. [18] [19] [20] The likelihood of developing resistance in pathogens is higher for antimicrobial strategies that have a specific target structure, following the key-lock principle, embodied in many antibiotics or antiseptics. [21] [22] In such cases, pathogens can evade the antimicrobial challenge through specific mutations, upregulation of efflux pumps, or production of enzymes that deactivate antimicrobials. In contrast, aPDT acts through a variety of non-specific oxidative mechanisms targeting multiple structures and pathways simultaneously, making the technique far less prone to resistance. [13] The possibility of bacteria developing tolerance to aPDT has therefore been deemed highly unlikely. [4] Several studies have demonstrated the efficacy of aPDT against various drug-resistant pathogens, including the World Health Organization (WHO) priority pathogens, such as Staphylococcus aureus , Pseudomonas aeruginosa , Klebsiella pneumoniae , Acinetobacter baumannii , Enterococcus faecium , Candida auris , Escherichia coli and many others. [20] [23]

Light sources

Light is required to excite the photosensitizer, which leads to the photochemical production of ROS. To efficiently transfer photon energy to the electron structure of the photosensitizer, the wavelength of the light source must be matched to the absorption spectrum of the photosensitizer. Different light sources have been used in aPDT, such as lamps (e.g. tungsten filament, Xenon arc and fluorescent lamps), lasers and light emitting diodes (LEDs). Lamps typically emit white light, but a filter can be used to select the appropriate wavelength to be absorbed by the photosensitizer and to avoid undesired thermal effects. In contrast, lasers are monochromatic light sources that can be easily coupled to optical fibers to access non-surface regions. LEDs are also monochromatic light sources, although their spectral emission bands are wider than those of lasers. However, the coupling of LEDs and optical fibers is not efficient, resulting in significant loss of light. More recently, organic LEDs (OLEDs) have been used in aPDT as wearable light sources because they can be made to be more flexible, thinner, and lighter than conventional LEDs. Sunlight can also serve as a source of light for aPDT; however, exact illumination parameters may be difficult to precisely reproduce. [24]

Light dosimetry

aPDT results depend on the interplay of three physical quantities: irradiance, radiant exposure and exposure time. Irradiance is defined as the optical power of the light source in Watts, divided by the area of tissue illumination conventionally described in square meters or centimeters (W/m2 or W/cm2). The irradiance, as a photodynamic parameter, is limited by the onset of adverse thermal factors in exposed tissue, or by degradative consequences to the sensitizer itself (commonly referred to as "photobleaching"). Radiant exposure is given by the product of irradiance and exposure time in seconds, divided by the illuminated area (J/cm2), and is commonly termed the light dose. This parameter is often limited by acceptable treatment times because lengthy treatment times can be unacceptable in a point-of-care setting. Fluence is a different physical quantity often used by aPDT practitioners, which considers the backscattering flux of light-tissue interaction causing re-entry of photons back into the treated area. [25]

Photosensitizers

Photodynamic action relies on absorption of electromagnetic radiation by the photosensitizing compound and conversion of this energy into redox chemical reactions or transfer to ground-state oxygen, producing the highly oxidizing species, singlet oxygen. [10] Consequently, the photosensitizer can be considered a photocatalyst, but it is also true that the sensitizer directly interacts with target moieties such as microbes to establish, for example, molecular targeting. This explains why not all photosensitizers are useful as photoantimicrobials. [26]

The most effective photosensitizer molecules carry a positive charge (cationic). [26] This promotes electrostatic attraction with negatively charged groups found on microbial cell surfaces (e.g. phosphate, carboxylate, sulfate), thus ensuring that during illumination, production of reactive oxygen species occurs in close contact with the targeted cellular population. Consequently, negatively charged photosensitizers are less effective, particularly against gram-negative bacteria l cells that carry a strongly negative zeta potential. [1]

The most widely employed photosensitizer in clinical practice is the phenothiazine derivative, methylene blue, which carries a +1 charge. [27] Methylene blue is also favored due to its long record of safe use in patients, both in surgical staining and the systemic treatment of methemoglobinemia. [28] Many other photosensitizers have been suggested, from various chemical classes, such as porphyrins, phthalocyanines and xanthenes, but the requirement for cationic nature and proven safety for human/animal use represents a high barrier to new chemical entity development. [5]

Molecular frameworks most often used as photosensitizers for antimicrobial photodynamic therapy. The examples listed in this figure include: methylene blue (MB, phenothiazine), crystal violet (CV, triarylmethane), porphyrins, phtalocyanines, riboflavin (Vitamin B2), rose bengal (RB, halogenated xanthene), chlorins and curcumin. Photosensitizer molecules.jpg
Molecular frameworks most often used as photosensitizers for antimicrobial photodynamic therapy. The examples listed in this figure include: methylene blue (MB, phenothiazine), crystal violet (CV, triarylmethane), porphyrins, phtalocyanines, riboflavin (Vitamin B2), rose bengal (RB, halogenated xanthene), chlorins and curcumin.

aPDT Enhancement by inorganic salts and gold nanoparticles

It was discovered in 2015 that the addition of inorganic salts can potentiate aPDT by several orders of magnitude, [29] and may even allow oxygen-independent photoinactivation to take place. [30] Potassium iodide (KI) is the most relevant example. Other inorganic salts such as potassium thiocyanate (KSCN), potassium selenocyanate (KSeCN), potassium bromide (KBr), sodium nitrite (NaNO2) and even sodium azide (NaN3, toxic) have also been shown to increase the killing of a broad range of pathogens by up to one million times. [31]

The addition of KI at concentrations up to 100 mM allows gram-negative bacteria to be killed by photosensitizers, which have no effect on their own, and this was shown to be effective in several animal models of localized infections. [32] KI was shown to be effective in human AIDS patients with oral candidiasis who were treated with methylene blue aPDT. [33] Oral consumption of saturated KI solution (4-6 g KI/day) is a standard treatment for some deep fungal infections of the skin.

The photochemical mechanisms of action are complex. [34] KI can react with singlet oxygen to form free molecular iodine plus hydrogen peroxide, which show synergistic and long-lived antimicrobial effects, as well as forming short-lived, reactive iodine radicals. Type 1 photosensitizers can carry out direct electron transfer to form iodine radicals, even in the absence of oxygen. KSCN reacts with singlet oxygen to form sulfur trioxide radicals, while KSeCN forms semi-stable selenocyanogen. KBr reacts with TiO2 photocatalysis to form hypobromite, while NaNO2 reacts with singlet oxygen to form unstable peroxynitrate. NaN3 quenches singlet oxygen so it can only react by electron transfer to form azide radicals. Relatively high concentrations of salts are necessary to trap the short-lived reactive species produced during aPDT.

The presence of gold nanoparticles is able to enhance the antimicrobial effectiveness of photosensitizers such as toludine blue. [35] Covalently linking nanoparticles to a photosensitizer also results in enhanced antimicrobial activity. [36] The gold nanoparticles have two roles: firstly they enhance the light capture of the dye and secondly they help direct the decay pathway for the dye, encouraging a non-radiative process through the formation of excess bactericidal radical species.

Incorporation of photosensitizers into polymers

Photosensitizers can be incorporated into polymers resulting in materials that can kill microbes on their surfaces when activated by visible light. [37] [38] Such polymers have been shown to be effective in killing bacteria in a clinical environment. [39] These self-disinfecting materials could, therefore, be used to coat surfaces in order to reduce the spread of disease-causing microbes in clinical environments as well as in food-processing and food-handling premises.

Advances in medicine and surgery have led to increasing reliance on a variety of medical devices of which the catheter is the most widely used. Unfortunately, the non-shedding surfaces of catheters can be colonized by microbes resulting in biofilm formation and, consequently, lead to an infection. Such catheter-related infections are a major cause of morbidity and mortality. Photosensitizers such as methylene blue and toluidine blue have been incorporated into silicone, the main polymer used in the manufacture of catheters, and the resulting composites have been shown to exert an antimicrobial effect when exposed to light of a suitable wavelength. [40] [41] [42] Suitable irradiation of such materials has been shown to be able to significantly reduce biofilm accumulation on their surfaces. [43] This approach has potential for reducing the morbidity and mortality associated with catheter-associated infections.

Microbial resistance to aPDT

The generation of reactive oxygen species (ROS) in neutrophils, macrophages, and eosinophils is one of the primary means by which the human immune system combats infecting microbes. [44] Highly adaptable microbes have evolved some level of protection strategies against these reactive molecules by upregulating antioxidant enzymes when exposed to ROS, suggesting one method by which microbes could develop increased resistance to aPDT. [45] However, these biochemical responses are limited when compared to the magnitude of oxidative stress placed on the microbe by aPDT. [4] Numerous investigations involving the repeated exposure of microorganisms to sublethal doses of antimicrobial photodynamic therapy (aPDT) and the subsequent analysis of the resilience of the cultured cells that survive, consistently reveal no significant indication of the development of resistance in these microorganisms. [46] [47] [48] [49] [50] [51] In fact, a study using methylene blue as a photosensitizer (PS) against MRSA, a series of aPDT exposure followed by re-cultivation tests conducted over multiple years showed that the microorganism's sensitivity to aPDT remained unchanged. In contrast, significant resistance to oxacillin emerged in fewer than twelve cycles. [47]

Virulence inhibition by aPDT

Pathogenic microbes cause harm to their hosts and evade host defense mechanisms through a range of virulence factors, which include elements like exotoxins, endotoxins, capsules, adhesins, invasins, and proteases. [52] [53] While antibiotics can inactivate microbes and thereby prevent further production of host-damaging virulence factors, few have any effect on pre-existing virulence factors or those which are released during the bactericidal process. These factors can continue to produce damaging effects even after the offending microbial cells have been inactivated. [54]

Unlike most antimicrobial drugs, antimicrobial photodynamic therapy (aPDT) is typically capable of neutralizing or diminishing the effectiveness of microbial virulence factors, or it can reduce their expression. [55] [56] The ability to inhibit microbial virulence is of particular interest because it could be related to accelerated infection site healing when compared to standard antimicrobial chemotherapy that only relies on bacteriostatic or bactericidal effects. [57] [58] Secreted virulence factors normally contain peptides, and it is well known that some amino acids (e.g. histidine, cysteine, tyrosine, tryptophan and methionine) are highly vulnerable to oxidation. [59] [60] Photodynamic reactions have demonstrated significant effectiveness in diminishing the harmful activity of lipopolysaccharides (LPS), proteases, and various other microbial toxins. [46] [55] [61] The capability to not only eliminate the microbes causing an infection but also to inhibit expression of various molecules that lead to host tissue damage offers a significant benefit over traditional antimicrobial drugs. [5]

Nasal decolonization

Nasal decolonization is recognized as a primary preventive intervention in the development of hospital-acquired infections (HAIs), especially surgical site infections (SSIs). [62] [63] HAIs represent a serious public health concern worldwide, with approximately 2.5 million HAIs annually in the United States leading to high morbidity and mortality (e.g. 30,000 deaths per year directly attributable to HAIs). HAIs affect one in every 31 hospitalized patients in the USA. [64] Staphylococcus aureus , a gram-positive bacterium, is the most common cause of nosocomial pneumonia and surgical site infections and the second-most common cause of bloodstream, cardiovascular, and eye, ear, nose, and throat infections. [65] S. aureus is by far the leading cause of skin and soft tissue HAIs, which can lead to potentially lethal bacteremia. [66] SSIs are among the most common healthcare-associated infections with substantial morbidity and mortality. An analysis of the 2005 Nationwide Inpatient Sample Database showed that S. aureus infections in inpatients tripled the duration of hospital stay, increasing length of stay by an average of 7.5 days for surgical site infections. [66] The anterior nares have been classified as the most consistent site of S. aureus colonization. [67] Asymptomatic S. aureus nasal carriage in healthy individuals has been reported at 20-55%, [68] causing increased risk of surgical-site infection by almost 4-fold. [69] Critically,a growing proportion of these bacterial populations exhibit antibiotic resistance. [70] [71]

Nasal decolonization of S. aureus to reduce the incidence of SSIs is expanding into current standard of care in both intensive care units (ICU) and presurgical settings. [72] [73] Various decolonization strategies have been used in hospitals in an effort to reduce transmission of bacteria and decrease overall infection rate. Decolonization effects are both directly and indirectly related via reduction of the overall bioburden when broadly administered within an acute care setting. There is the added benefit of effects that go beyond the treated patients extending to healthcare workers and other patients. [62]

Several clinical studies performed using the current standard of care – intranasal mupirocin 2% antibiotic ointment – in surgical patients, concluded that this treatment significantly decreased the rate of hospital-acquired infections. [74] [75] [76] One study found a 44% reduction in bloodstream infection rates when universal decolonization was used (e.g. intranasal mupirocin ointment and chlorhexidine body wash) in a trial involving 73,256 hospital patients. [72] In addition, researchers have demonstrated that eradicating S. aureus from the anterior nares also utilizing intranasal mupirocin ointment reduced surgical site infection rates up to 58% in hospitalized patients who were nasal carriers. [77] However, widespread use of mupirocin is associated with development of mupirocin-resistant strains of MRSA, with one hospital in Canada experiencing an increase from 2.7% to 65% resistant strains in three years. [78] A targeted – as opposed to universal – decolonization approach is sometimes recommended because of increasing levels of mupirocin resistance. [79] Currently, only universal decolonization with mupirocin has been demonstrated to be an effective control measure and therefore selective administration of mupirocin is contraindicated. [72]

Nasal aPDT addresses the issues of antibiotic-induced resistance in multiple ways. As a site-specific therapy, it does not interfere with the overall microbiome because it is not systemically administered. Moreover, phenothiazinium photosensitizers can target negatively charged bacterial cells leaving zwitterionic host tissues unharmed. [80] Treatment of the nose specifically targets the respiratory outlet, which is a key source of microbial colonization and dissemination through touch or normal respiration. Yet, the unspecific mechanisms of action effectively prevent development of resistance.

The first large-scale study involving aPDT for nasal decolonization, initially conducted exclusively on specific surgery types, the study demonstrated a significant 42% reduction in surgical site infections. [81] The most significant reduction in SSI rates were in orthopedic and spinal surgeries. Currently, the use of nasal photodisinfection has been expanded to encompass a wide range of surgeries, resulting in an increased effect size with an approximate efficacy of 80%. [81] The technique has been deployed in multiple Canadian hospitals since that time, and is undergoing clinical trials in the US for the same purpose.

Specialty-specific studies have also been carried out, especially in high-risk surgery of the spine. One large Canadian study found that the spine-surgery SSI rate decreased 5.6% (from 7.2% to 1.6%) because of nasal aPDT combined with chlorhexidine bathing, saving on average $45–55 CAD per treated patient ($4.24 million CAD annually). This study concluded that "CSD/nPDT is both efficacious and cost-effective in preventing surgical site infections". No adverse events were reported. [82]

Skin infections

There are three main types of skin infections in humans that have been treated with aPDT: 1) Fungal infections, 2) Mycobacterial infections and 3) Cutaneous Leishmaniasis. The most clinically used photosensitizers are methylene blue and curcumin, as well as the protoporphyrin IX precursors, aminolevulinic acid (ALA) and methyl-ALA.

Fungal infections treated with aPDT have included both Dermatophytosis and Sporotrichosis. Infections with filamentous fungi such as Trichophyton spp. which express keratinase enzymes usually affect the toenails (onychomycosis), but can also affect the skin (tinea). In onychomycosis (tinea unguium), efforts are often made to increase the penetration of photosensitizers into the toenail matrix before the application of light. [83] Cutaneous tinea infections affecting the foot, scalp or crotch have been treated with ALA-aPDT. [84] Sporotrichosis is a zoonosis caused by the dimorphic fungus Sporothrix spp often transmitted by animal bites or scratches. It has been treated with aPDT mediated by ALA or methylene blue. [85]

Skin infections can be caused by non-tuberculous mycobacteria, including rapidly growing species such as Mycobacterium marinum (swimmers' granuloma) and Mycobacterium avium complex. Some of these infections have been treated with aPDT using ALA in combination with conventional antibiotics. [86]

Leishmaniasis is caused by an intracellular parasitic infection caused by single-celled protozoa of the genus Leishmania . It is transmitted by the bites of infected sand flies found in both the Old World (Southern Europe and Middle East) and the New World (Central and South America). Each year there are up to 2 million new cases and 70,000 deaths worldwide. Leishmaniasis infections can be either cutaneous, mucosal, or visceral, with the latter type being the deadliest. Cutaneous leishmaniasis has been treated with aPDT mediated by either ALA [87] or methylene blue, [88] because the standard treatment using systemic amphotericin B or topical pentavalent antimonial preparations have several drawbacks.

Chronic wounds

Chronic wounds are those that do not heal within months of treatment. They are classified into three main types, i.e. venous, diabetic, and pressure ulcers and are frequently sites of microbial infection that become a major deterrent to for patient recovery. aPDT offers a treatment option for chronic wounds, because of its lethal action against drug-resistant microorganisms. [89] [2]

Diabetic Foot ulcers (DFU) affect 10 to 25% of diabetic patients during their lives, requiring long and intensive hospitalization. The economic impact of DFU to worldwide health care systems is significant. [90] [91] DFU are frequently infected with a combination of fungi and bacteria including the genera Serratia , Morganella , Proteus , Haemophilus , Acinetobacter , Enterococcus , and Staphylococcus . In addition, there is an increased likelihood of contracting resistant strains of these and other microorganisms from hospital settings. DFU patients commonly respond poorly to antibiotic therapy. Consequently, amputation becomes indicated to prevent other complications, such as osteonecrosis, thrombosis and more disseminated types of bacteremia. [92]

aPDT has been successfully used to treat the diabetic foot, reducing the incidence of amputation in DFU patients. [93] DFU patients treated with aPDT were associated with only a 2.9% chance of amputation, compared to 100% in the control group (classical antibiotic therapy, without aPDT). Using an initial cohort study of 62 patients [94] and subsequently of 218 patients, [95] Tardivo and colleagues developed the Tardivo algorithm as a prognostic score to determine the risk of amputation and to predict the ideal therapeutic options for the treatment of DFU by aPDT. The score is based on three factors: Wagner's classification, signs of PAD, and location of foot ulcers. [94] Values for the independent parameters are multiplied together and, for patients with scores below 16, treatment with aPDT is associated with approximately 85% (95% CI) chance of recovery. [95]

Oral infections

In the early 90s, Emeritus Professor Michael Wilson from University College London (UCL), initiated scientific investigations on the potential of aPDT to combat bacteria of interest in dentistry. [96] Since then, aPDT has been explored for various oral conditions, such as periodontal disease (gum disease), dental caries (cavities), endodontic treatment (root canal treatment), oral herpes and oral candidiasis. [97] Research and clinical studies have shown promising results in reducing microbial load and treating infections. However, the efficacy of aPDT can vary based on factors like the type and concentration of photosensitizer used, light parameters, and the specific infection being treated. [98]

While aPDT can be considered as an adjunctive treatment to standard of care, it is not currently intended to replace conventional therapies. This may change in the future, as drug-resistance patterns in the oral microbiome develop over time, making aPDT monotherapy increasingly necessary. [99]

Some advantages of aPDT in oral infections include broad-spectrum action since aPDT can target a wide range of microorganisms (e.g. bacteria, fungi, and virus), including antibiotic-resistant strains, and oral biofilm is composed of wide variety of microorganisms. Another advantage is the localized treatment that can be used to target specific infected areas, minimizing damage to healthy tissues, and maintaining the normal microbiota without significant damage. To date, no significant adverse events associated with intraoral aPDT have been reported. [100]

aPDT offers the dental practitioner an intraoral decontamination therapy that its minimally invasive nature, broad-spectrum action, rapid microbicidal effect, reduced antibiotic use, patient comfort factor, high compliance rate, treatment of resistant strains and minimization of microbial resistance selection.

Disinfection of blood-products

During the 1980s, the realization of the presence of the human immunodeficiency virus (HIV) in the global supply of donated blood led to the development of both thorough hemovigilance and of methods for the safe disinfection of microbial species in donated blood and blood products. [101]

Blood is a mixture of cells and proteins and is routinely separated into its constituent parts for use in various therapies, e.g. platelets, red cells and plasma might be used in specific replacement, and proteins (typically clotting factors) derived from the plasma fraction are provided for the treatment of hemophilia, for example. Viruses, such as HIV, might be associated with the cellular components or suspended extracellularly, thus representing a threat of recipient infection whichever of these fractions is used. However, treatments aimed at viral inactivation/destruction must preserve cell/protein function, and this represents a barrier, particularly to cellular disinfection.

In terms of the use of photosensitizers, both methylene blue and riboflavin are employed for the photodisinfection of plasma, using visible or long-wave ultraviolet illumination respectively, while riboflavin is also used for disinfection of platelets. [102] [103] [104] However, neither approach is employed for red blood cell concentrates. Among related approaches, the psoralen derivative Amotosalen, activated by long-wavelength UV light, is used in Europe for disinfection of plasma and platelets. [105] However, this represents a photochemical reaction between the psoralen nucleus and viral nucleic acids, rather than a purely photodynamic effect.

Veterinary applications

In small animal practice, aPDT has been investigated for the treatment of different dermatological diseases with positive results. Although there are limited scientific data in this field, successful applications include otitis externa caused by multidrug-resistant Pseudomonas aeruginosa , [106] dermatophytosis caused by Microsporum canis , [107] and in association with itraconazole for sporotrichosis. [108]

aPDT can also be used as a non-antibiotic platform for the treatment of infectious diseases in food-producing animals. Indeed, overuse of antimicrobials in these animals may lead to contamination of meat and milk by antibiotic-resistant bacteria or antibiotic residues. In this regard, aPDT has proven effective in the treatment of caseous lymphadenitis [109] and streptococcal abscesses in sheep, [110] and is demonstrably more effective than oxytetracycline (gold standard treatment) for bovine digital dermatitis. [111] Other applications of aPDT include the treatment of mastitis in dairy cattle and sheep, [112] [113] [114] and sole ulcers and surgical wound healing in cattle. [111] [115]

Exotic, zoo, and wildlife medicine is challenging and stands out as another field of possibility for aPDT. In this regard, aPDT has been successfully used to treat penguins suffering from pododermatitis [116] [117] and snakes with infectious stomatitis caused by gram-negative bacteria. [118] Additionally, aPDT has been deployed as an adjuvant endodontic treatment for a traumatic tusk fracture in an elephant. [119]

Food decontamination

The ever-increasing demand for food decontamination technologies has resulted in several studies focusing on the evaluation of the antimicrobial efficacy of aPDT in food and its effect on the organoleptic properties of the food products. [120]

aPDT has shown antimicrobial efficacy against microbes on fruits, [121] [122] [123] vegetables, [121] [124] seafood, [125] [126] [127] and meat. [124] The efficacy of aPDT used in this way is dependent on several factors including wavelength of light, temperature, and food-related factors such as acidity, surface properties and water activity. [120] Endogenous porphyrins that are light-absorbing compounds located within certain bacteria produce photosensitized reactions in the presence of light in the blue region of the spectrum (400-500 nm), [128] showing better antimicrobial efficacy than other wavelengths in the visible spectrum (e.g. green and red, 500-700 nm) in the absence of an exogenous photosensitizer. [129] [130]

Acidity of the food being disinfected plays an important role, as gram-positive bacteria have been found to be more sensitive to aPDT in acidic conditions while gram-negative bacteria are more sensitive to aPDT at alkaline conditions. [131] Since aPDT is a surface decontamination technology, the surface characteristics of the tested material play an important role. The irregular surfaces of products like pet food pellets can lead to a shadowing effect, where microorganisms can hide in food crevices and be shielded from the light treatment. [120] Flat surfaces can show better efficacy of aPDT as compared to the spherical or irregular surfaces. [132] Moreover, high water activity conditions contribute to the success of aPDT compared to low water activity conditions, due to limited penetration of light in more desiccated foods. [133] Other factors like irradiance, treatment time (or dose), microbial strain, and distance of the product from the light source also play a major role in the microbicidal efficacy of food-based aPDT. [120] [132] [133]

A recent study demonstrated that appropriate concentrations of a photosensitizer potentially useful for food-based disinfection combined with appropriate peak absorption wavelength light resulted in upwards of 99.999% (5 log10) reduction in MRSA and complete kill in Salmonella cell counts. In addition to bacteria, aPDT has shown efficacy against fungal species. [134] [135] Optimization of the factors influencing antimicrobial efficacy and scalability of aPDT are required for successful application in the food industry.

Related Research Articles

<i>Staphylococcus aureus</i> Species of Gram-positive bacterium

Staphylococcus aureus is a gram-positive spherically shaped bacterium, a member of the Bacillota, and is a usual member of the microbiota of the body, frequently found in the upper respiratory tract and on the skin. It is often positive for catalase and nitrate reduction and is a facultative anaerobe that can grow without the need for oxygen. Although S. aureus usually acts as a commensal of the human microbiota, it can also become an opportunistic pathogen, being a common cause of skin infections including abscesses, respiratory infections such as sinusitis, and food poisoning. Pathogenic strains often promote infections by producing virulence factors such as potent protein toxins, and the expression of a cell-surface protein that binds and inactivates antibodies. S. aureus is one of the leading pathogens for deaths associated with antimicrobial resistance and the emergence of antibiotic-resistant strains, such as methicillin-resistant S. aureus (MRSA), is a worldwide problem in clinical medicine. Despite much research and development, no vaccine for S. aureus has been approved.

Methicillin-resistant <i>Staphylococcus aureus</i> Bacterium responsible for difficult-to-treat infections in humans

Methicillin-resistant Staphylococcus aureus (MRSA) is a group of gram-positive bacteria that are genetically distinct from other strains of Staphylococcus aureus. MRSA is responsible for several difficult-to-treat infections in humans. It caused more than 100,000 deaths worldwide attributable to antimicrobial resistance in 2019.

Bloodstream infections (BSIs) are infections of blood caused by blood-borne pathogens. The detection of microbes in the blood is always abnormal. A bloodstream infection is different from sepsis, which is characterized by severe inflammatory or immune responses of the host organism to pathogens.

<span class="mw-page-title-main">Photodynamic therapy</span> Form of phototherapy

Photodynamic therapy (PDT) is a form of phototherapy involving light and a photosensitizing chemical substance used in conjunction with molecular oxygen to elicit cell death (phototoxicity).

<span class="mw-page-title-main">Light therapy</span> Therapy involving intentional exposure to sunlight

Light therapy, also called phototherapy or bright light therapy is the exposure to direct sunlight or artificial light at controlled wavelengths in order to treat a variety of medical disorders, including seasonal affective disorder (SAD), circadian rhythm sleep-wake disorders, cancers, and skin wound infections. Treating skin conditions such as neurodermatitis, psoriasis, acne vulgaris, and eczema with ultraviolet light is called ultraviolet light therapy.

<span class="mw-page-title-main">Fusidic acid</span> Antibiotic

Fusidic acid, sold under the brand name Fucidin among others, is a steroid antibiotic that is often used topically in creams or ointments and eyedrops but may also be given systemically as tablets or injections. As of October 2008, the global problem of advancing antimicrobial resistance has led to a renewed interest in its use.

<span class="mw-page-title-main">Photosensitizer</span> Type of molecule reacting to light

Photosensitizers are light absorbers that alter the course of a photochemical reaction. They usually are catalysts. They can function by many mechanisms, sometimes they donate an electron to the substrate, sometimes they abstract a hydrogen atom from the substrate. At the end of this process, the photosensitizer returns to its ground state, where it remains chemically intact, poised to absorb more light. One branch of chemistry which frequently utilizes photosensitizers is polymer chemistry, using photosensitizers in reactions such as photopolymerization, photocrosslinking, and photodegradation. Photosensitizers are also used to generate prolonged excited electronic states in organic molecules with uses in photocatalysis, photon upconversion and photodynamic therapy. Generally, photosensitizers absorb electromagnetic radiation consisting of infrared radiation, visible light radiation, and ultraviolet radiation and transfer absorbed energy into neighboring molecules. This absorption of light is made possible by photosensitizers' large de-localized π-systems, which lowers the energy of HOMO and LUMO orbitals to promote photoexcitation. While many photosensitizers are organic or organometallic compounds, there are also examples of using semiconductor quantum dots as photosensitizers.

<span class="mw-page-title-main">Methyl aminolevulinate</span> Chemical compound

Methyl aminolevulinate (MAL) is a drug used as a sensitizer in photodynamic therapy. It is a prodrug that is metabolized to protoporphyrin IX. It is marketed as Metvix.

<span class="mw-page-title-main">Oritavancin</span> Pharmaceutical drug

Oritavancin, sold under the brand name Orbactiv among others, is a semisynthetic glycopeptide antibiotic medication for the treatment of serious Gram-positive bacterial infections. Its chemical structure as a lipoglycopeptide is similar to vancomycin.

Lysostaphin is a Staphylococcus simulans metalloendopeptidase. It can function as a bacteriocin (antimicrobial) against Staphylococcus aureus.

<span class="mw-page-title-main">Temoporfin</span> Chemical compound

Temoporfin (INN) is a photosensitizer used in photodynamic therapy for the treatment of squamous cell carcinoma of the head and neck . It is marketed in the European Union under the brand name Foscan. The U.S. Food and Drug Administration (FDA) declined to approve Foscan in 2000. The EU approved its use in June 2001.

Blood irradiation therapy is an alternative medical procedure in which the blood is exposed to low-level light for therapeutic reasons. The practice was originally developed in the United States, but most recent research on it has been conducted in Germany and in Russia. Low-level laser therapy has been tested for a wide range of conditions, but rigorous double-blinded studies have not yet been performed. Furthermore, it has been claimed that ultraviolet irradiation of blood kills bacteria by DNA damage and also activation of the immune system. Blood irradiation therapy is highly controversial, and has fallen from mainstream use since its heyday in the 1940s and 1950s.

<span class="mw-page-title-main">Pheophorbide</span> Chemical compound

Pheophorbide or phaeophorbide is a product of chlorophyll breakdown and a derivative of pheophytin where both the central magnesium has been removed and the phytol tail has been hydrolyzed. It is used as a photosensitizer in photodynamic therapy.

<span class="mw-page-title-main">Enzybiotics</span> Experimental antibacterial therapy

Enzybiotics are an experimental antibacterial therapy. The term is derived from a combination of the words “enzyme” and “antibiotics.” Enzymes have been extensively utilized for their antibacterial and antimicrobial properties. Proteolytic enzymes called endolysins have demonstrated particular effectiveness in combating a range of bacteria and are the basis for enzybiotic research. Endolysins are derived from bacteriophages and are highly efficient at lysing bacterial cells. Enzybiotics are being researched largely to address the issue of antibiotic resistance, which has allowed for the proliferation of drug-resistant pathogens posing great risk to animal and human health across the globe.

Staphylococcus schleiferi is a Gram-positive, cocci-shaped bacterium of the family Staphylococcaceae. It is facultatively anaerobic, coagulase-variable, and can be readily cultured on blood agar where the bacterium tends to form opaque, non-pigmented colonies and beta (β) hemolysis. There exists two subspecies under the species S. schleiferi: Staphylococcus schleiferi subsp. schleiferi and Staphylococcus schleiferi subsp. coagulans.

Staphylococcus pseudintermedius is a gram positive coccus bacteria of the genus Staphylococcus found worldwide. It is primarily a pathogen for domestic animals, but has been known to affect humans as well. S. pseudintermedius is an opportunistic pathogen that secretes immune modulating virulence factors, has many adhesion factors, and the potential to create biofilms, all of which help to determine the pathogenicity of the bacterium. Diagnoses of Staphylococcus pseudintermedius have traditionally been made using cytology, plating, and biochemical tests. More recently, molecular technologies like MALDI-TOF, DNA hybridization and PCR have become preferred over biochemical tests for their more rapid and accurate identifications. This includes the identification and diagnosis of antibiotic resistant strains.

Decolonization, also bacterial decolonization, is a medical intervention that attempts to rid a patient of an antimicrobial resistant pathogen, such as methicillin-resistant Staphylococcus aureus (MRSA) or antifungal-resistant Candida.

Kerry L. LaPlante is an American pharmacist, academic and researcher. She is the Dean at the University of Rhode Island College of Pharmacy. She is a Professor of Pharmacy and former department Chair of the Department of Pharmacy Practice at the University of Rhode Island, an Adjunct Professor of Medicine at Brown University, an Infectious Diseases Pharmacotherapy Specialist, and the Director of the Rhode Island Infectious Diseases Fellowship and Research Programs at the Veterans Affairs Medical Center in Providence, Rhode Island.

Photodynamic/photothermal combination therapy involves the usage of a chemical compound or nanomaterial that, when irradiated at a certain wavelength, converts light energy into reactive oxygen species (ROS) and heat. This has shown to be highly effective in the treatment of skin infections, showing increased wound healing rates and a lower impact on human cell viability than photodynamic (PD) or photothermal (PT) therapies. The compounds involved often employ additional mechanisms of action or side effect reduction mechanisms, further increasing their efficacy.

<span class="mw-page-title-main">Light-emitting diode therapy</span>

Light-emitting diode therapy (LEDT) is a clinical approach that applies different wavelengths of light to cure diseases or conditions with skin-safe lights. Following NASA's innovation in the 1990s with Light Emitting Diodes (LEDs) that emit a specific narrow light spectrum, LED Therapy (LEDT) showed significant potential. The high precision of narrow-band LED therapy enabled its first use in clinical practices. The commonly used lights in LEDT are blue, red, green, yellow, and infrared (IR).

References

  1. 1 2 Hamblin, Michael R.; Hasan, Tayyaba (2004). "Photodynamic therapy: a new antimicrobial approach to infectious disease?". Photochemical & Photobiological Sciences. 3 (5): 436–450. doi:10.1039/b311900a. ISSN   1474-905X. PMC   3071049 . PMID   15122361.
  2. 1 2 Dai, Tianhong; Huang, Ying-Ying; Hamblin, Michael R. (2009). "Photodynamic therapy for localized infections—State of the art". Photodiagnosis and Photodynamic Therapy. 6 (3–4): 170–188. doi:10.1016/j.pdpdt.2009.10.008. PMC   2811240 . PMID   19932449.
  3. St. Denis, Tyler G; Dai, Tianhong; Izikson, Leonid; Astrakas, Christos; Anderson, Richard Rox; Hamblin, Michael R; Tegos, George P (2011). "All you need is light: Antimicrobial photoinactivation as an evolving and emerging discovery strategy against infectious disease". Virulence. 2 (6): 509–520. doi:10.4161/viru.2.6.17889. ISSN   2150-5594. PMC   3260545 . PMID   21971183.
  4. 1 2 3 Maisch, Tim (2015). "Resistance in antimicrobial photodynamic inactivation of bacteria". Photochemical & Photobiological Sciences. 14 (8): 1518–1526. doi: 10.1039/c5pp00037h . ISSN   1474-905X. PMID   26098395. S2CID   5034232.
  5. 1 2 3 4 5 Wainwright, Mark; Maisch, Tim; Nonell, Santi; Plaetzer, Kristjan; Almeida, Adelaide; Tegos, George P; Hamblin, Michael R (2017). "Photoantimicrobials—are we afraid of the light?". The Lancet Infectious Diseases. 17 (2): e49–e55. doi:10.1016/S1473-3099(16)30268-7. PMC   5280084 . PMID   27884621.
  6. Finsen, Niels (1901). Phototherapy (1st ed.). London, UK: Edward Arnold.
  7. 1 2 3 4 Ackroyd, Roger; Kelty, Clive; Brown, Nicola; Reed, Malcolm (2007-05-01). "The History of Photodetection and Photodynamic Therapy¶". Photochemistry and Photobiology. 74 (5): 656–669. doi:10.1562/0031-8655(2001)0740656thopap2.0.co2. ISSN   0031-8655. PMID   11723793. S2CID   222102425.
  8. Nobel Prize Website
  9. 1 2 Kessel, David (2019-10-02). "Photodynamic Therapy: A Brief History". Journal of Clinical Medicine. 8 (10): 1581. doi: 10.3390/jcm8101581 . ISSN   2077-0383. PMC   6832404 . PMID   31581613.
  10. 1 2 3 4 Baptista, Maurício S.; Cadet, Jean; Di Mascio, Paolo; Ghogare, Ashwini A.; Greer, Alexander; Hamblin, Michael R.; Lorente, Carolina; Nunez, Silvia Cristina; Ribeiro, Martha Simões; Thomas, Andrés H.; Vignoni, Mariana; Yoshimura, Tania Mateus (2017). "Type I and Type II Photosensitized Oxidation Reactions: Guidelines and Mechanistic Pathways". Photochemistry and Photobiology. 93 (4): 912–919. doi:10.1111/php.12716. ISSN   0031-8655. PMC   5500392 . PMID   28084040.
  11. 1 2 Bacellar, Isabel O. L.; Baptista, Mauricio S. (2019-12-24). "Mechanisms of Photosensitized Lipid Oxidation and Membrane Permeabilization". ACS Omega. 4 (26): 21636–21646. doi:10.1021/acsomega.9b03244. ISSN   2470-1343. PMC   6933592 . PMID   31891041.
  12. Bacellar, Isabel O. L.; Oliveira, Maria Cecilia; Dantas, Lucas S.; Costa, Elierge B.; Junqueira, Helena C.; Martins, Waleska K.; Durantini, Andrés M.; Cosa, Gonzalo; Di Mascio, Paolo; Wainwright, Mark; Miotto, Ronei; Cordeiro, Rodrigo M.; Miyamoto, Sayuri; Baptista, Mauricio S. (2018-08-01). "Photosensitized Membrane Permeabilization Requires Contact-Dependent Reactions between Photosensitizer and Lipids". Journal of the American Chemical Society. 140 (30): 9606–9615. doi:10.1021/jacs.8b05014. ISSN   0002-7863. PMID   29989809. S2CID   207191699.
  13. 1 2 Sabino, Caetano P.; Ribeiro, Martha S.; Wainwright, Mark; dos Anjos, Carolina; Sellera, Fábio P.; Dropa, Milena; Nunes, Nathalia B.; Brancini, Guilherme T. P.; Braga, Gilberto U. L.; Arana-Chavez, Victor E.; Freitas, Raul O.; Lincopan, Nilton; Baptista, Maurício S. (2023). "The Biochemical Mechanisms of Antimicrobial Photodynamic Therapy †". Photochemistry and Photobiology. 99 (2): 742–750. doi:10.1111/php.13685. ISSN   0031-8655. PMID   35913428. S2CID   251222413.
  14. Boyce, John M.; Pittet, Didier (2002). "Guideline for Hand Hygiene in Health-Care Settings: Recommendations of the Healthcare Infection Control Practices Advisory Committee and the HICPAC/SHEA/APIC/IDSA Hand Hygiene Task Force". Infection Control & Hospital Epidemiology. 23 (S12): S3–S40. doi:10.1086/503164. ISSN   0899-823X. PMID   12515399. S2CID   20265540.
  15. Kiesslich, Tobias; Gollmer, Anita; Maisch, Tim; Berneburg, Mark; Plaetzer, Kristjan (2013). "A Comprehensive Tutorial on In Vitro Characterization of New Photosensitizers for Photodynamic Antitumor Therapy and Photodynamic Inactivation of Microorganisms". BioMed Research International. 2013: 1–17. doi: 10.1155/2013/840417 . ISSN   2314-6133. PMC   3671303 . PMID   23762860.
  16. Stewart, Philip S; William Costerton, J (2001). "Antibiotic resistance of bacteria in biofilms". The Lancet. 358 (9276): 135–138. doi:10.1016/S0140-6736(01)05321-1. PMID   11463434. S2CID   46125592.
  17. Mah, Thien-Fah C; O'Toole, George A (2001). "Mechanisms of biofilm resistance to antimicrobial agents". Trends in Microbiology. 9 (1): 34–39. doi: 10.1016/S0966-842X(00)01913-2 . PMID   11166241.
  18. Mulani, Mansura S.; Kamble, Ekta E.; Kumkar, Shital N.; Tawre, Madhumita S.; Pardesi, Karishma R. (2019-04-01). "Emerging Strategies to Combat ESKAPE Pathogens in the Era of Antimicrobial Resistance: A Review". Frontiers in Microbiology. 10: 539. doi: 10.3389/fmicb.2019.00539 . ISSN   1664-302X. PMC   6452778 . PMID   30988669.
  19. Nakonieczna, Joanna; Wozniak, Agata; Pieranski, Michal; Rapacka-Zdonczyk, Aleksandra; Ogonowska, Patrycja; Grinholc, Mariusz (2019). "Photoinactivation of ESKAPE pathogens: overview of novel therapeutic strategy". Future Medicinal Chemistry. 11 (5): 443–461. doi: 10.4155/fmc-2018-0329 . ISSN   1756-8919. PMID   30901231. S2CID   85456591.
  20. 1 2 Sabino, Caetano Padial; Wainwright, Mark; Ribeiro, Martha Simões; Sellera, Fábio Parra; dos Anjos, Carolina; Baptista, Mauricio da Silva; Lincopan, Nilton (2020). "Global priority multidrug-resistant pathogens do not resist photodynamic therapy". Journal of Photochemistry and Photobiology B: Biology. 208: 111893. doi:10.1016/j.jphotobiol.2020.111893. PMID   32446039. S2CID   218864416.
  21. Maillard, J.-Y. (2002). "Bacterial target sites for biocide action". Journal of Applied Microbiology. 92: 16S–27S. doi:10.1046/j.1365-2672.92.5s1.3.x. ISSN   1364-5072. PMID   12000609. S2CID   21058126.
  22. McDonnell, Gerald; Russell, A. Denver (1999). "Antiseptics and Disinfectants: Activity, Action, and Resistance". Clinical Microbiology Reviews. 12 (1): 147–179. doi:10.1128/CMR.12.1.147. ISSN   0893-8512. PMC   88911 . PMID   9880479.
  23. Grizante Barião, Patrícia Helena; Tonani, Ludmilla; Brancini, Guilherme Thomaz Pereira; Nascimento, Erika; Braga, Gilberto Úbida Leite; Wainwright, Mark; von Zeska Kress, Marcia Regina (2022-07-11). "In vitro and in vivo photodynamic efficacies of novel and conventional phenothiazinium photosensitizers against multidrug-resistant Candida auris". Photochemical & Photobiological Sciences. 21 (10): 1807–1818. doi: 10.1007/s43630-022-00258-4 . ISSN   1474-9092. PMID   35816272. S2CID   250423362.
  24. Piksa, Marta; Lian, Cheng; Samuel, Imogen C.; Pawlik, Krzysztof J.; Samuel, Ifor D. W.; Matczyszyn, Katarzyna (2023). "The role of the light source in antimicrobial photodynamic therapy". Chemical Society Reviews. 52 (5): 1697–1722. doi: 10.1039/D0CS01051K . hdl: 10023/26974 . ISSN   0306-0012. PMID   36779328. S2CID   256826056.
  25. Sliney, David H. (2007-02-27). "Radiometric Quantities and Units Used in Photobiology and Photochemistry: Recommendations of the Commission Internationale de l'Eclairage (International Commission on Illumination): Photochemistry and Photobiology, 2007, 83". Photochemistry and Photobiology. 83 (2): 425–432. doi:10.1562/2006-11-14-RA-1081. PMID   17115802. S2CID   33854252.
  26. 1 2 Wainwright, M (1998-07-01). "Photodynamic antimicrobial chemotherapy (PACT)". Journal of Antimicrobial Chemotherapy. 42 (1): 13–28. doi: 10.1093/jac/42.1.13 . ISSN   1460-2091. PMID   9700525.
  27. Wainwright, Mark (2018). "Synthetic, small-molecule photoantimicrobials–a realistic approach". Photochemical & Photobiological Sciences. 17 (11): 1767–1779. doi: 10.1039/c8pp00145f . ISSN   1474-905X. PMID   29905338. S2CID   49209175.
  28. Ginimuge, PrashantR; Jyothi, SD (2010). "Methylene blue: Revisited". Journal of Anaesthesiology Clinical Pharmacology. 26 (4): 517–520. doi: 10.4103/0970-9185.74599 . ISSN   0970-9185. PMC   3087269 . PMID   21547182.
  29. Vecchio, Daniela; Gupta, Asheesh; Huang, Liyi; Landi, Giacomo; Avci, Pinar; Rodas, Andrea; Hamblin, Michael R. (2015). "Bacterial Photodynamic Inactivation Mediated by Methylene Blue and Red Light Is Enhanced by Synergistic Effect of Potassium Iodide". Antimicrobial Agents and Chemotherapy. 59 (9): 5203–5212. doi:10.1128/AAC.00019-15. ISSN   0066-4804. PMC   4538466 . PMID   26077247.
  30. Hamblin, Michael R; Abrahamse, Heidi (2020-01-31). "Oxygen-Independent Antimicrobial Photoinactivation: Type III Photochemical Mechanism?". Antibiotics. 9 (2): 53. doi: 10.3390/antibiotics9020053 . ISSN   2079-6382. PMC   7168166 . PMID   32023978.
  31. Hamblin, Michael R (2017-11-02). "Potentiation of antimicrobial photodynamic inactivation by inorganic salts". Expert Review of Anti-infective Therapy. 15 (11): 1059–1069. doi:10.1080/14787210.2017.1397512. ISSN   1478-7210. PMC   5706449 . PMID   29084463.
  32. Wen, Xiang; Zhang, Xiaoshen; Szewczyk, Grzegorz; El-Hussein, Ahmed; Huang, Ying-Ying; Sarna, Tadeusz; Hamblin, Michael R. (2017). "Potassium Iodide Potentiates Antimicrobial Photodynamic Inactivation Mediated by Rose Bengal in In Vitro and In Vivo Studies". Antimicrobial Agents and Chemotherapy. 61 (7). doi:10.1128/AAC.00467-17. ISSN   0066-4804. PMC   5487662 . PMID   28438946.
  33. Du, Meixia; Xuan, Weijun; Zhen, Xiumei; He, Lixia; Lan, Lina; Yang, Shanlin; Wu, Nianning; Qin, Jinmei; zhao, Rui; Qin, Jianglong; Lan, Jian; Lu, Huan; Liang, Cuijin; Li, Yanjun; R Hamblin, Michael (2021). "Antimicrobial photodynamic therapy for oral Candida infection in adult AIDS patients: A pilot clinical trial". Photodiagnosis and Photodynamic Therapy. 34: 102310. doi:10.1016/j.pdpdt.2021.102310. PMID   33901690. S2CID   233409814.
  34. Hamblin, Michael R.; Abrahamse, Heidi (2018-12-03). "Inorganic Salts and Antimicrobial Photodynamic Therapy: Mechanistic Conundrums?". Molecules. 23 (12): 3190. doi: 10.3390/molecules23123190 . ISSN   1420-3049. PMC   6321187 . PMID   30514001.
  35. Narband, Naima; Tubby, Sarah; Parkin, Ivan; Gil-Tomas, Jesus; Ready, Derren; Nair, Sean; Wilson, Michael (2008-11-01). "Gold Nanoparticles Enhance the Toluidine Blue-Induced Lethal Photosensitisation of Staphylococcus aureus". Current Nanoscience. 4 (4): 409–414. Bibcode:2008CNan....4..409N. doi:10.2174/157341308786306134. ISSN   1573-4137.
  36. Gil-Tomás, Jesús; Tubby, Sarah; Parkin, Ivan P.; Narband, Naima; Dekker, Linda; Nair, Sean P.; Wilson, Michael; Street, Cale (2007). "Lethal photosensitisation of Staphylococcus aureus using a toluidine blue O–tiopronin–gold nanoparticle conjugate". Journal of Materials Chemistry. 17 (35): 3739. doi:10.1039/b706615e. ISSN   0959-9428.
  37. Decraene, Valérie; Pratten, Jonathan; Wilson, Michael (2006). "Cellulose Acetate Containing Toluidine Blue and Rose Bengal Is an Effective Antimicrobial Coating when Exposed to White Light". Applied and Environmental Microbiology. 72 (6): 4436–4439. Bibcode:2006ApEnM..72.4436D. doi:10.1128/aem.02945-05. ISSN   0099-2240. PMC   1489612 . PMID   16751564.
  38. Decraene, Valérie; Pratten, Jonathan; Wilson, Michael (2008-06-28). "Novel Light-Activated Antimicrobial Coatings Are Effective Against Surface-Deposited Staphylococcus aureus". Current Microbiology. 57 (4): 269–273. doi:10.1007/s00284-008-9188-7. ISSN   0343-8651. PMID   18587617. S2CID   6690954.
  39. Decraene, Valérie; Pratten, Jonathan; Wilson, Michael (2008). "Assessment of the Activity of a Novel Light-Activated Antimicrobial Coating in a Clinical Environment". Infection Control & Hospital Epidemiology. 29 (12): 1181–1184. doi:10.1086/592413. ISSN   0899-823X. PMID   18950278. S2CID   6008564.
  40. Perni, Stefano; Piccirillo, Clara; Pratten, Jonathan; Prokopovich, Polina; Chrzanowski, Wojciech; Parkin, Ivan P.; Wilson, Michael (2009). "The antimicrobial properties of light-activated polymers containing methylene blue and gold nanoparticles". Biomaterials. 30 (1): 89–93. doi:10.1016/j.biomaterials.2008.09.020. ISSN   0142-9612. PMID   18838166.
  41. Piccirillo, C.; Perni, S.; Gil-Thomas, J.; Prokopovich, P.; Wilson, M.; Pratten, J.; Parkin, I. P. (2009). "Antimicrobial activity of methylene blue and toluidine blue O covalently bound to a modified silicone polymer surface". Journal of Materials Chemistry. 19 (34): 6167. doi:10.1039/b905495b. ISSN   0959-9428.
  42. Perni, Stefano; Prokopovich, Polina; Piccirillo, Clara; Pratten, Jonathan; Parkin, Ivan P.; Wilson, Michael (2009). "Toluidine blue-containing polymers exhibit potent bactericidal activity when irradiated with red laser light". Journal of Materials Chemistry. 19 (18): 2715. doi:10.1039/b820561b. ISSN   0959-9428.
  43. Perni, Stefano; Prokopovich, P.; Parkin, Ivan P.; Wilson, Michael; Pratten, Jonathan (2010). "Prevention of biofilm accumulation on a light-activated antimicrobial catheter material". Journal of Materials Chemistry. 20 (39): 8668. doi:10.1039/c0jm01891k. ISSN   0959-9428.
  44. Thomas, David C. (2017). "The phagocyte respiratory burst: Historical perspectives and recent advances". Immunology Letters. 192: 88–96. doi:10.1016/j.imlet.2017.08.016. ISSN   0165-2478. PMID   28864335.
  45. Ezraty, Benjamin; Gennaris, Alexandra; Barras, Frédéric; Collet, Jean-François (2017). "Oxidative stress, protein damage and repair in bacteria". Nature Reviews Microbiology. 15 (7): 385–396. doi:10.1038/nrmicro.2017.26. ISSN   1740-1534. PMID   28420885. S2CID   205497253.
  46. 1 2 Bartolomeu, Maria; Rocha, Sónia; Cunha, Ângela; Neves, M. G. P. M. S.; Faustino, Maria A. F.; Almeida, Adelaide (2016-03-07). "Effect of Photodynamic Therapy on the Virulence Factors of Staphylococcus aureus". Frontiers in Microbiology. 7: 267. doi: 10.3389/fmicb.2016.00267 . ISSN   1664-302X. PMC   4780358 . PMID   27014198.
  47. 1 2 Pedigo, Lisa A.; Gibbs, Aaron J.; Scott, Robert J.; Street, Cale N. (2009-06-29). "Absence of bacterial resistance following repeat exposure to photodynamic therapy". In Kessel, David H. (ed.). Photodynamic Therapy: Back to the Future. Vol. 7380. pp. 520–526. doi:10.1117/12.822834. S2CID   84999117.
  48. Tavares, Anabela; Carvalho, Carla M. B.; Faustino, Maria A.; Neves, Maria G. P. M. S.; Tomé, João P. C.; Tomé, Augusto C.; Cavaleiro, José A. S.; Cunha, Ângela; Gomes, Newton C. M.; Alves, Eliana; Almeida, Adelaide (2010-01-20). "Antimicrobial Photodynamic Therapy: Study of Bacterial Recovery Viability and Potential Development of Resistance after Treatment". Marine Drugs. 8 (1): 91–105. doi: 10.3390/md8010091 . ISSN   1660-3397. PMC   2817925 . PMID   20161973.
  49. Cassidy, Corona M.; Donnelly, Ryan F.; Tunney, Michael M. (2010). "Effect of sub-lethal challenge with Photodynamic Antimicrobial Chemotherapy (PACT) on the antibiotic susceptibility of clinical bacterial isolates". Journal of Photochemistry and Photobiology B: Biology. 99 (1): 62–66. doi:10.1016/j.jphotobiol.2010.02.004. PMID   20207552.
  50. Giuliani, Francesco; Martinelli, Manuele; Cocchi, Annalisa; Arbia, Debora; Fantetti, Lia; Roncucci, Gabrio (2010). "In Vitro Resistance Selection Studies of RLP068/Cl, a New Zn(II) Phthalocyanine Suitable for Antimicrobial Photodynamic Therapy". Antimicrobial Agents and Chemotherapy. 54 (2): 637–642. doi:10.1128/AAC.00603-09. ISSN   0066-4804. PMC   2812146 . PMID   20008782.
  51. Al-Mutairi, Rawan; Tovmasyan, Artak; Batinic-Haberle, Ines; Benov, Ludmil (2018-07-31). "Sublethal Photodynamic Treatment Does Not Lead to Development of Resistance". Frontiers in Microbiology. 9: 1699. doi: 10.3389/fmicb.2018.01699 . ISSN   1664-302X. PMC   6079231 . PMID   30108561.
  52. Casadevall, Arturo; Pirofski, Liise-anne (2001). "Host-Pathogen Interactions: The Attributes of Virulence". The Journal of Infectious Diseases. 184 (3): 337–344. doi: 10.1086/322044 . ISSN   0022-1899. PMID   11443560.
  53. Leitão, Jorge H. (2020-07-27). "Microbial Virulence Factors". International Journal of Molecular Sciences. 21 (15): 5320. doi: 10.3390/ijms21155320 . ISSN   1422-0067. PMC   7432612 . PMID   32727013.
  54. Lepper, P.; Held, T.; Schneider, E.; Bölke, E.; Gerlach, H.; Trautmann, M. (2002). "Clinical implications of antibiotic-induced endotoxin release in septic shock". Intensive Care Medicine. 28 (7): 824–833. doi:10.1007/s00134-002-1330-6. ISSN   0342-4642. PMID   12122518. S2CID   27591769.
  55. 1 2 Kömerik, Nurgül; Wilson, Michael; Poole, Steve (2007-05-01). "The Effect of Photodynamic Action on Two Virulence Factors of Gram-negative Bacteria¶". Photochemistry and Photobiology. 72 (5): 676–680. doi:10.1562/0031-8655(2000)0720676teopao2.0.co2. ISSN   0031-8655. PMID   11107854. S2CID   24959473.
  56. Kato, Ilka Tiemy; Prates, Renato Araujo; Sabino, Caetano Padial; Fuchs, Beth Burgwyn; Tegos, George P.; Mylonakis, Eleftherios; Hamblin, Michael R.; Ribeiro, Martha Simões (2013). "Antimicrobial Photodynamic Inactivation Inhibits Candida albicans Virulence Factors and Reduces In Vivo Pathogenicity". Antimicrobial Agents and Chemotherapy. 57 (1): 445–451. doi:10.1128/AAC.01451-12. ISSN   0066-4804. PMC   3535901 . PMID   23129051.
  57. Hamblin, Michael; Zahra, Touqir; Contag, Christopher; McManus, Albert; Hasan, Tayyaba (2003). "Optical Monitoring and Treatment of Potentially Lethal Wound Infections In Vivo". The Journal of Infectious Diseases. 187 (11): 1717–1726. doi:10.1086/375244. ISSN   0022-1899. PMC   3441051 . PMID   12751029.
  58. Pérez, Montserrat; Robres, Pilar; Moreno, Bernardino; Bolea, Rosa; Verde, Maria T.; Pérez-Laguna, Vanesa; Aspiroz, Carmen; Gilaberte, Yolanda; Rezusta, Antonio (2021-05-25). "Comparison of Antibacterial Activity and Wound Healing in a Superficial Abrasion Mouse Model of Staphylococcus aureus Skin Infection Using Photodynamic Therapy Based on Methylene Blue or Mupirocin or Both". Frontiers in Medicine. 8. doi: 10.3389/fmed.2021.673408 . ISSN   2296-858X. PMC   8185160 . PMID   34113639.
  59. Pattison, David I.; Rahmanto, Aldwin Suryo; Davies, Michael J. (2012). "Photo-oxidation of proteins". Photochemical & Photobiological Sciences. 11 (1): 38–53. doi:10.1039/c1pp05164d. ISSN   1474-905X. PMID   21858349. S2CID   21787970.
  60. Davies, Michael (2016). "Protein oxidation and peroxidation". Biochemical Journal. 473 (7): 805–825. doi:10.1042/BJ20151227. ISSN   0264-6021. PMC   4819570 . PMID   27026395.
  61. Tubby, Sarah; Wilson, Michael; Nair, Sean P (2009). "Inactivation of staphylococcal virulence factors using a light-activated antimicrobial agent". BMC Microbiology. 9 (1): 211. doi: 10.1186/1471-2180-9-211 . ISSN   1471-2180. PMC   2762988 . PMID   19804627.
  62. 1 2 Seidelman, Jessica L.; Mantyh, Christopher R.; Anderson, Deverick J. (2023-01-17). "Surgical Site Infection Prevention: A Review". JAMA. 329 (3): 244–252. doi:10.1001/jama.2022.24075. ISSN   0098-7484. PMID   36648463. S2CID   255939830.
  63. Popovich, Kyle J.; Aureden, Kathy; Ham, D. Cal; Harris, Anthony D.; Hessels, Amanda J.; Huang, Susan S.; Maragakis, Lisa L.; Milstone, Aaron M.; Moody, Julia; Yokoe, Deborah; Calfee, David P. (2023). "SHEA/IDSA/APIC Practice Recommendation: Strategies to prevent methicillin-resistant Staphylococcus aureus transmission and infection in acute-care hospitals: 2022 Update". Infection Control & Hospital Epidemiology. 44 (7): 1039–1067. doi:10.1017/ice.2023.102. ISSN   0899-823X. PMC   10369222 . PMID   37381690.
  64. Magill, Shelley S.; O'Leary, Erin; Janelle, Sarah J.; Thompson, Deborah L.; Dumyati, Ghinwa; Nadle, Joelle; Wilson, Lucy E.; Kainer, Marion A.; Lynfield, Ruth; Greissman, Samantha; Ray, Susan M.; Beldavs, Zintars; Gross, Cindy; Bamberg, Wendy; Sievers, Marla (2018). "Changes in Prevalence of Health Care–Associated Infections in U.S. Hospitals". New England Journal of Medicine. 379 (18): 1732–1744. doi:10.1056/NEJMoa1801550. ISSN   0028-4793. PMC   7978499 . PMID   30380384.
  65. Mangram, A. J.; Horan, T. C.; Pearson, M. L.; Silver, L. C.; Jarvis, W. R. (1999). "Guideline for Prevention of Surgical Site Infection, 1999. Centers for Disease Control and Prevention (CDC) Hospital Infection Control Practices Advisory Committee". American Journal of Infection Control. 27 (2): 97–132, quiz 133–134, discussion 96. doi:10.1016/S0196-6553(99)70088-X. ISSN   0196-6553. PMID   10196487.
  66. 1 2 Klevens, R. Monina (2007-10-17). "Invasive Methicillin-Resistant Staphylococcus aureus Infections in the United States". JAMA. 298 (15): 1763–1771. doi:10.1001/jama.298.15.1763. ISSN   0098-7484. PMID   17940231.
  67. Cho, Ilseung; Blaser, Martin J. (2012). "The human microbiome: at the interface of health and disease". Nature Reviews Genetics. 13 (4): 260–270. doi:10.1038/nrg3182. ISSN   1471-0056. PMC   3418802 . PMID   22411464.
  68. Nouwen, J (2001). "Determinants of Staphylococcus aureus nasal carriage". The Netherlands Journal of Medicine. 59 (3): 126–133. doi:10.1016/S0300-2977(01)00150-4. PMID   11583828.
  69. von Eiff, Christof; Becker, Karsten; Machka, Konstanze; Stammer, Holger; Peters, Georg (2001-01-04). "Nasal Carriage as a Source of Staphylococcus aureus Bacteremia". New England Journal of Medicine. 344 (1): 11–16. doi: 10.1056/NEJM200101043440102 . ISSN   0028-4793. PMID   11136954.
  70. Willems, Rob J.L.; Hanage, William P.; Bessen, Debra E.; Feil, Edward J. (2011). "Population biology of Gram-positive pathogens: high-risk clones for dissemination of antibiotic resistance". FEMS Microbiology Reviews. 35 (5): 872–900. doi:10.1111/j.1574-6976.2011.00284.x. ISSN   1574-6976. PMC   3242168 . PMID   21658083.
  71. Mlynarczyk-Bonikowska, Beata; Kowalewski, Cezary; Krolak-Ulinska, Aneta; Marusza, Wojciech (2022-07-22). "Molecular Mechanisms of Drug Resistance in Staphylococcus aureus". International Journal of Molecular Sciences. 23 (15): 8088. doi: 10.3390/ijms23158088 . ISSN   1422-0067. PMC   9332259 . PMID   35897667.
  72. 1 2 3 Huang, Susan S.; Septimus, Edward; Kleinman, Ken; Moody, Julia; Hickok, Jason; Avery, Taliser R.; Lankiewicz, Julie; Gombosev, Adrijana; Terpstra, Leah; Hartford, Fallon; Hayden, Mary K.; Jernigan, John A.; Weinstein, Robert A.; Fraser, Victoria J.; Haffenreffer, Katherine (2013-06-13). "Targeted versus Universal Decolonization to Prevent ICU Infection". New England Journal of Medicine. 368 (24): 2255–2265. doi: 10.1056/NEJMoa1207290 . ISSN   0028-4793. PMC   10853913 . PMID   23718152. S2CID   16081350.
  73. Huang, Susan S.; Septimus, Edward; Avery, Taliser R.; Lee, Grace M.; Hickok, Jason; Weinstein, Robert A.; Moody, Julia; Hayden, Mary K.; Perlin, Jonathan B.; Platt, Richard; Ray, G. Thomas (2014). "Cost Savings of Universal Decolonization to Prevent Intensive Care Unit Infection: Implications of the REDUCE MRSA Trial". Infection Control & Hospital Epidemiology. 35 (S3): S23–S31. doi: 10.1086/677819 . hdl: 1969.1/181819 . ISSN   0899-823X. PMC   10920056 . PMID   25222894. S2CID   25417940.
  74. Coates, T.; Bax, R.; Coates, A. (2009-07-01). "Nasal decolonization of Staphylococcus aureus with mupirocin: strengths, weaknesses and future prospects". Journal of Antimicrobial Chemotherapy. 64 (1): 9–15. doi:10.1093/jac/dkp159. ISSN   0305-7453. PMC   2692503 . PMID   19451132.
  75. van Rijen, Miranda; Bonten, Marc; Wenzel, Richard; Kluytmans, Jan (2008-10-08). Cochrane Wounds Group (ed.). "Mupirocin ointment for preventing Staphylococcus aureus infections in nasal carriers". Cochrane Database of Systematic Reviews. 2011 (2): CD006216. doi:10.1002/14651858.CD006216.pub2. PMC   8988859 . PMID   18843708.
  76. van Rijen, M. M. L.; Bonten, M.; Wenzel, R. P.; Kluytmans, J. A. J. W. (2007-12-19). "Intranasal mupirocin for reduction of Staphylococcus aureus infections in surgical patients with nasal carriage: a systematic review". Journal of Antimicrobial Chemotherapy. 61 (2): 254–261. doi:10.1093/jac/dkm480. ISSN   0305-7453. PMID   18174201.
  77. Bode, Lonneke G.M.; Kluytmans, Jan A.J.W.; Wertheim, Heiman F.L.; Bogaers, Diana; Vandenbroucke-Grauls, Christina M.J.E.; Roosendaal, Robert; Troelstra, Annet; Box, Adrienne T.A.; Voss, Andreas; van der Tweel, Ingeborg; van Belkum, Alex; Verbrugh, Henri A.; Vos, Margreet C. (2010-01-07). "Preventing Surgical-Site Infections in Nasal Carriers of Staphylococcus aureus". New England Journal of Medicine. 362 (1): 9–17. doi: 10.1056/NEJMoa0808939 . hdl: 2066/124346 . ISSN   0028-4793. PMID   20054045. S2CID   1009598.
  78. Conly, John M.; Vas, Stephen (2002). "Increasing Mupirocin Resistance ofStaphylococcus Aureusin CAPD — Should it Continue to be Used as Prophylaxis?". Peritoneal Dialysis International: Journal of the International Society for Peritoneal Dialysis. 22 (6): 649–652. doi: 10.1177/089686080202200601 . ISSN   0896-8608. PMID   12556065. S2CID   30628844.
  79. Humphreys, H.; Grundmann, H.; Skov, R.; Lucet, J.-C.; Cauda, R. (2009). "Prevention and control of methicillin-resistant Staphylococcus aureus". Clinical Microbiology and Infection. 15 (2): 120–124. doi: 10.1111/j.1469-0691.2009.02699.x . PMID   19291143.
  80. Tanaka, Masamitsu; Kinoshita, Manabu; Yoshihara, Yasuo; Shinomiya, Nariyoshi; Seki, Shuhji; Nemoto, Koichi; Hirayama, Takahiro; Dai, Tianhong; Huang, Liyi; Hamblin, Michael R.; Morimoto, Yuji (2012). "Optimal Photosensitizers for Photodynamic Therapy of Infections Should Kill Bacteria but Spare Neutrophils". Photochemistry and Photobiology. 88 (1): 227–232. doi:10.1111/j.1751-1097.2011.01005.x. ISSN   0031-8655. PMC   3253242 . PMID   21950417.
  81. 1 2 Bryce, E.; Wong, T.; Forrester, L.; Masri, B.; Jeske, D.; Barr, K.; Errico, S.; Roscoe, D. (2014). "Nasal photodisinfection and chlorhexidine wipes decrease surgical site infections: a historical control study and propensity analysis". Journal of Hospital Infection. 88 (2): 89–95. doi:10.1016/j.jhin.2014.06.017. PMID   25171975.
  82. Banaszek, Daniel; Inglis, Tom; Ailon, Tamir; Charest-Morin, Raphaële; Dea, Nicolas; Fisher, Charles G.; Kwon, Brian K.; Paquette, Scott J.; Street, John (2019). "283. The efficacy and cost-effectiveness of photodynamic therapy in prevention of surgical site infection". The Spine Journal. 19 (9): S138. doi:10.1016/j.spinee.2019.05.299. S2CID   201962834.
  83. Bhatta, Anil Kumar; Keyal, Uma; Wang, Xiu Li (2016). "Photodynamic therapy for onychomycosis: A systematic review". Photodiagnosis and Photodynamic Therapy. 15: 228–235. doi:10.1016/j.pdpdt.2016.07.010. PMID   27477248.
  84. Qiao, Jianjun; Li, Ruoyu; Ding, Yingguo; Fang, Hong (2010). "Photodynamic Therapy in the Treatment of Superficial Mycoses: An Evidence-based Evaluation". Mycopathologia. 170 (5): 339–343. doi:10.1007/s11046-010-9325-2. ISSN   0301-486X. PMID   20526681. S2CID   21330005.
  85. Legabão, Barbara Cipulo; Fernandes, Juliana Aparecida; de Oliveira Barbosa, Gabriela Franco; Bonfim-Mendonça, Patrícia S.; Svidzinski, Terezinha I.E. (2022). "The zoonosis sporotrichosis can be successfully treated by photodynamic therapy: A scoping review". Acta Tropica. 228: 106341. doi: 10.1016/j.actatropica.2022.106341 . PMID   35131203. S2CID   246577798.
  86. Sun, Kedai; Li, Jie; Li, Lingfei; Li, Guangyao; Wang, Liqun; Chen, Jinyi; Wu, Xingru; Luo, Jiefu; Liu, Heyong; Wang, Xiaoyu; Lu, Weiping; Li, Min; Lei, Xia (2022). "A new approach to the treatment of nontuberculous mycobacterium skin infections caused by iatrogenic manipulation: Photodynamic therapy combined with antibiotics: A pilot study". Photodiagnosis and Photodynamic Therapy. 37: 102695. doi:10.1016/j.pdpdt.2021.102695. PMID   34923157. S2CID   245291311.
  87. Enk, Claes D. (2003-04-01). "Treatment of Cutaneous Leishmaniasis With Photodynamic Therapy". Archives of Dermatology. 139 (4): 432–434. doi:10.1001/archderm.139.4.432. ISSN   0003-987X. PMID   12707088. S2CID   4817539.
  88. Rody, K Kandri (2020-03-09). "Effective Treatment of Cutaneous Leishmaniasis with Photodynamic Therapy of Methylene Blue: A Case Report". Biomedical Journal of Scientific & Technical Research. 26 (3). doi: 10.26717/bjstr.2020.26.004351 . ISSN   2574-1241. S2CID   226015449.
  89. Grandi, Vieri; Corsi, Alessandro; Pimpinelli, Nicola; Bacci, Stefano (2022-07-07). "Cellular Mechanisms in Acute and Chronic Wounds after PDT Therapy: An Update". Biomedicines. 10 (7): 1624. doi: 10.3390/biomedicines10071624 . ISSN   2227-9059. PMC   9313247 . PMID   35884929.
  90. Boulton, Andrew JM; Vileikyte, Loretta; Ragnarson-Tennvall, Gunnel; Apelqvist, Jan (2005). "The global burden of diabetic foot disease". The Lancet. 366 (9498): 1719–1724. doi:10.1016/S0140-6736(05)67698-2. PMID   16291066. S2CID   32137879.
  91. Driver, V. R.; Fabbi, M.; Lavery, L. A.; Gibbons, G. (2010). "The Costs of Diabetic Foot: The Economic Case for the Limb Salvage Team". Journal of the American Podiatric Medical Association. 100 (5): 335–341. doi:10.7547/1000335. ISSN   8750-7315. PMID   20847346.
  92. Armstrong, David G.; Boulton, Andrew J.M.; Bus, Sicco A. (2017-06-15). Ingelfinger, Julie R. (ed.). "Diabetic Foot Ulcers and Their Recurrence". New England Journal of Medicine. 376 (24): 2367–2375. doi:10.1056/NEJMra1615439. ISSN   0028-4793. PMID   28614678. S2CID   205117844.
  93. Tardivo, João Paulo; Adami, Fernando; Correa, João Antonio; Pinhal, Maria Aparecida S.; Baptista, Mauricio S. (2014). "A clinical trial testing the efficacy of PDT in preventing amputation in diabetic patients". Photodiagnosis and Photodynamic Therapy. 11 (3): 342–350. doi:10.1016/j.pdpdt.2014.04.007. PMID   24814697.
  94. 1 2 Tardivo, João Paulo; Baptista, Maurício S.; Correa, João Antonio; Adami, Fernando; Pinhal, Maria Aparecida Silva (2015-08-17). Santanelli, di Pompeo d'Illasi, Fabio (ed.). "Development of the Tardivo Algorithm to Predict Amputation Risk of Diabetic Foot". PLOS ONE. 10 (8): e0135707. Bibcode:2015PLoSO..1035707T. doi: 10.1371/journal.pone.0135707 . ISSN   1932-6203. PMC   4539188 . PMID   26281044.
  95. 1 2 MS, Baptista (2022-06-05). "A Cohort Study to Evaluate the Predictions of the Tardivo Algorithm and the Efficacy of Antibacterial Photodynamic Therapy in the Management of the Diabetic". EJMRC. 4 (1). doi:10.17303/ejmrc.2022.4.102. S2CID   264572546.
  96. Wilson, Michael; Dobson, John; Harvey, Wilson (1992). "Sensitization of oral bacteria to killing by low-power laser radiation". Current Microbiology. 25 (2): 77–81. doi:10.1007/BF01570963. ISSN   0343-8651. PMID   1369193. S2CID   37764169.
  97. Gholami, Leila; Shahabi, Shiva; Jazaeri, Marzieh; Hadilou, Mahdi; Fekrazad, Reza (2023-01-05). "Clinical applications of antimicrobial photodynamic therapy in dentistry". Frontiers in Microbiology. 13. doi: 10.3389/fmicb.2022.1020995 . ISSN   1664-302X. PMC   9850114 . PMID   36687594.
  98. Liu, Y.; Qin, R.; Zaat, S. A.; Breukink, E.; Heger, M. (2015). "Antibacterial photodynamic therapy: overview of a promising approach to fight antibiotic-resistant bacterial infections". Journal of Clinical and Translational Research. 1 (3): 140–167. doi: 10.18053/jctres.201503.002 . ISSN   2424-810X. PMC   6410618 . PMID   30873451. S2CID   51685625.
  99. Jao, Ying; Ding, Shinn-Jyh; Chen, Chun-Cheng (2023). "Antimicrobial photodynamic therapy for the treatment of oral infections: A systematic review". Journal of Dental Sciences. 18 (4): 1453–1466. doi:10.1016/j.jds.2023.07.002. PMC   10548011 . PMID   37799910.
  100. Carrera, E T; Dias, H B; Corbi, S C T; Marcantonio, R A C; Bernardi, A C A; Bagnato, V S; Hamblin, M R; Rastelli, A N S (2016-12-01). "The application of antimicrobial photodynamic therapy (aPDT) in dentistry: a critical review". Laser Physics. 26 (12): 123001. Bibcode:2016LaPhy..26l3001C. doi:10.1088/1054-660X/26/12/123001. ISSN   1054-660X. PMC   5687295 . PMID   29151775.
  101. Vuk, Tomislav; Politis, Constantina; Laspina, Stefan; Lozano, Miquel; Haddad, Antoine; de Angelis, Vincenzo; Garraud, Olivier (2023). "Thirty years of hemovigilance – Achievements and future perspectives". Transfusion Clinique et Biologique. 30 (1): 166–172. doi:10.1016/j.tracli.2022.09.070. PMID   36216308. S2CID   252772919.
  102. Wainwright, Mark (2000). "Methylene blue derivatives — suitable photoantimicrobials for blood product disinfection?". International Journal of Antimicrobial Agents. 16 (4): 381–394. doi:10.1016/S0924-8579(00)00207-7. PMID   11118846.
  103. Wainwright, Mark (2002). "Pathogen Inactivation in Blood Products". Current Medicinal Chemistry. 9 (1): 127–143. doi:10.2174/0929867023371355. PMID   11860353.
  104. Wainwright, Mark (2002-03-18). "The emerging chemistry of blood product disinfection". Chemical Society Reviews. 31 (2): 128–136. doi:10.1039/b101905h. PMID   12109206.
  105. Liu, Hong; Wang, Xun (2021). "Pathogen reduction technology for blood component: A promising solution for prevention of emerging infectious disease and bacterial contamination in blood transfusion services". Journal of Photochemistry and Photobiology. 8: 100079. doi: 10.1016/j.jpap.2021.100079 . S2CID   239940064.
  106. Sellera, Fábio P.; Fernandes, Miriam R.; Sabino, Caetano P.; de Freitas, Laura M.; da Silva, Luciano C.B.A.; Pogliani, Fabio C.; Ribeiro, Martha S.; Hamblin, Michael R.; Lincopan, Nilton (2019). "Effective treatment and decolonization of a dog infected with carbapenemase ( VIM -2)-producing Pseudomonas aeruginosa using probiotic and photodynamic therapies". Veterinary Dermatology. 30 (2): 170. doi:10.1111/vde.12714. ISSN   0959-4493. PMC   6610805 . PMID   30604463.
  107. Cabral, Fernanda V.; Sellera, Fábio P.; Ribeiro, Martha S. (2021). "Methylene blue-mediated antimicrobial photodynamic therapy for canine dermatophytosis caused by Microsporum canis: A successful case report with 6 months follow-up". Photodiagnosis and Photodynamic Therapy. 36: 102602. doi:10.1016/j.pdpdt.2021.102602. PMID   34706277. S2CID   239937543.
  108. Cabral, Fernanda V.; Sellera, Fábio P.; Ribeiro, Martha S. (2022). "Feline sporotrichosis successfully treated with methylene blue-mediated antimicrobial photodynamic therapy and low doses of itraconazole". Photodiagnosis and Photodynamic Therapy. 40: 103154. doi:10.1016/j.pdpdt.2022.103154. PMID   36272192. S2CID   253035140.
  109. Sellera, Fábio Parra; Gargano, Ronaldo Gomes; Libera, Alice Maria Melville Paiva Della; Benesi, Fernando José; Azedo, Milton Ricardo; de Sá, Lilian Rose Marques; Ribeiro, Martha Simões; da Silva Baptista, Maurício; Pogliani, Fabio Celidonio (2016). "Antimicrobial photodynamic therapy for caseous lymphadenitis abscesses in sheep: Report of ten cases". Photodiagnosis and Photodynamic Therapy. 13: 120–122. doi:10.1016/j.pdpdt.2015.12.006. PMID   26732393.
  110. Sellera, Fp; Barbosa, Bs; Gargano, Rg; Sabino, Cp; Ribeiro, Ms; Azedo, Mr; Benesi, Fj; Pogliani, Fc (2015-04-13). "Cutaneous streptococcal abscess treated by photodynamic therapy". African Journal of Traditional, Complementary and Alternative Medicines. 12 (2): 65. doi: 10.4314/ajtcam.v12i2.12 . ISSN   0189-6016.
  111. 1 2 Sellera, Fábio P.; Barbosa, Bruna S.; Gargano, Ronaldo G.; Ríspoli, Vívian F.P.; Sabino, Caetano P.; Ollhoff, Rudiger D.; Baptista, Maurício S.; Ribeiro, Martha S.; de Sá, Lilian R.M.; Pogliani, Fabio C. (2021). "Methylene blue-mediated antimicrobial photodynamic therapy can be a novel non-antibiotic platform for bovine digital dermatitis". Photodiagnosis and Photodynamic Therapy. 34: 102274. doi:10.1016/j.pdpdt.2021.102274. PMID   33812078. S2CID   233011040.
  112. Sellera, Fábio Parra; Sabino, Caetano Padial; Ribeiro, Martha Simões; Gargano, Ronaldo Gomes; Benites, Nilson Roberti; Melville, Priscilla Anne; Pogliani, Fabio Celidonio (2016). "In vitro photoinactivation of bovine mastitis related pathogens". Photodiagnosis and Photodynamic Therapy. 13: 276–281. doi:10.1016/j.pdpdt.2015.08.007. PMID   26315923.
  113. Silva, Lara Oliveira; da Silva Souza, Kedma Lorena; de Jesus Beloti, Larissa; Neto, Waldemar Mota Ramos; Núñez, Silvia Cristina; Frias, Danila Fernanda Rodrigues (2022). "Use of photodynamic therapy and photobiomodulation as alternatives for microbial control on clinical and subclinical mastitis in sheep". Lasers in Medical Science. 37 (4): 2305–2310. doi:10.1007/s10103-022-03506-2. ISSN   1435-604X. PMID   35031932. S2CID   245934881.
  114. Moreira, Lívia Helena; de Souza, José Carlos Pereira; de Lima, Carlos José; Salgado, Miguel Angel Castillo; Fernandes, Adriana Barrinha; Andreani, Dora Inés Kozusny; Villaverde, Antonio Balbin; Zângaro, Renato Amaro (2018). "Use of photodynamic therapy in the treatment of bovine subclinical mastitis". Photodiagnosis and Photodynamic Therapy. 21: 246–251. doi:10.1016/j.pdpdt.2017.12.009. hdl: 11449/179502 . PMID   29258951.
  115. Valandro, Patrícia; Massuda, Mayara B.; Rusch, Elidiane; Birgel, Daniela B.; Pereira, Philipe P.L.; Sellera, Fábio P.; Ribeiro, Martha S.; Pogliani, Fabio C.; Birgel Junior, Eduardo H. (2021). "Antimicrobial photodynamic therapy can be an effective adjuvant for surgical wound healing in cattle". Photodiagnosis and Photodynamic Therapy. 33: 102168. doi:10.1016/j.pdpdt.2020.102168. PMID   33497814. S2CID   231770338.
  116. Nascimento, Cristiane Lassálvia; Ribeiro, Martha Simões; Sellera, Fábio Parra; Dutra, Gustavo Henrique Pereira; Simões, Alyne; Teixeira, Carlos Roberto (2015). "Comparative study between photodynamic and antibiotic therapies for treatment of footpad dermatitis (bumblefoot) in Magellanic penguins (Spheniscus magellanicus)". Photodiagnosis and Photodynamic Therapy. 12 (1): 36–44. doi: 10.1016/j.pdpdt.2014.12.012 . PMID   25573284.
  117. Sellera, Fábio Parra; Sabino, Caetano Padial; Ribeiro, Martha Simões; Fernandes, Loriê Tukamoto; Pogliani, Fabio Celidonio; Teixeira, Carlos Roberto; Dutra, Gustavo Henrique Pereira; Nascimento, Cristiane Lassálvia (2014). "Photodynamic therapy for pododermatitis in penguins". Zoo Biology. 33 (4): 353–356. doi:10.1002/zoo.21135. ISSN   0733-3188. PMID   24888264.
  118. Grego, Kathleen Fernandes; Carvalho, Marcelo Pires Nogueira de; Cunha, Marcos Paulo Vieira; Knöbl, Terezinha; Pogliani, Fabio Celidonio; Catão-Dias, José Luiz; Sant'Anna, Sávio Stefanini; Ribeiro, Martha Simões; Sellera, Fábio Parra (2017). "Antimicrobial photodynamic therapy for infectious stomatitis in snakes: Clinical views and microbiological findings". Photodiagnosis and Photodynamic Therapy. 20: 196–200. doi:10.1016/j.pdpdt.2017.10.004. PMID   29037910.
  119. Seewald, Matthias; Gohl, Christine; Egerbacher, Monika; Handschuh, Stephan; Witter, Kirsti (2021). "Endodontic Treatment of a Traumatic Tusk Fracture With Exposed Pulp in an Asian Elephant ( Elephas maximus )". Journal of Veterinary Dentistry. 38 (3): 139–151. doi:10.1177/08987564211054590. ISSN   0898-7564. PMID   34873958. S2CID   244922577.
  120. 1 2 3 4 Ghate, Vinayak S.; Zhou, Weibiao; Yuk, Hyun-Gyun (2019). "Perspectives and Trends in the Application of Photodynamic Inactivation for Microbiological Food Safety". Comprehensive Reviews in Food Science and Food Safety. 18 (2): 402–424. doi: 10.1111/1541-4337.12418 . ISSN   1541-4337. PMID   33336937. S2CID   91271527.
  121. 1 2 Glueck, Michael; Schamberger, Barbara; Eckl, Peter; Plaetzer, Kristjan (2017). "New horizons in microbiological food safety: Photodynamic Decontamination based on a curcumin derivative". Photochemical & Photobiological Sciences. 16 (12): 1784–1791. doi: 10.1039/c7pp00165g . ISSN   1474-905X. PMID   29105723. S2CID   6811115.
  122. Kingsley, D.H.; Perez-Perez, R.E.; Boyd, G.; Sites, J.; Niemira, B.A. (2018). "Evaluation of 405-nm monochromatic light for inactivation of Tulane virus on blueberry surfaces". Journal of Applied Microbiology. 124 (4): 1017–1022. doi: 10.1111/jam.13638 . ISSN   1364-5072. PMID   29144595. S2CID   3857817.
  123. Luksiene, Zivile; Paskeviciute, Egle (2011). "Microbial control of food-related surfaces: Na-Chlorophyllin-based photosensitization". Journal of Photochemistry and Photobiology B: Biology. 105 (1): 69–74. doi:10.1016/j.jphotobiol.2011.06.011. PMID   21807530.
  124. 1 2 Tortik, Nicole; Spaeth, Andreas; Plaetzer, Kristjan (2014). "Photodynamic decontamination of foodstuff from Staphylococcus aureus based on novel formulations of curcumin". Photochemical & Photobiological Sciences. 13 (10): 1402–1409. doi:10.1039/c4pp00123k. ISSN   1474-905X. PMID   24957403. S2CID   19612644.
  125. Josewin, Sherrill; Ghate, Vinayak; Kim, Min-Jeong; Yuk, Hyun-Gyun (2018). "Antibacterial effect of 460 nm light emitting diode in combination with riboflavin against Listeria monocytogenes on smoked salmon". Food Control. 84: 354–361. doi:10.1016/j.foodcont.2017.08.017. ISSN   0956-7135.
  126. Liu, Fang; Li, Zhaojie; Cao, Binbin; Wu, Juan; Wang, Yuming; Xue, Yong; Xu, Jie; Xue, Changhu; Tang, Qing Juan (2016). "The effect of a novel photodynamic activation method mediated by curcumin on oyster shelf life and quality". Food Research International. 87: 204–210. doi:10.1016/j.foodres.2016.07.012. PMID   29606243.
  127. Roh, Heyong Jin; Kang, Gyoung Sik; Kim, Ahran; Kim, Nam Eun; Nguyen, Thanh Luan; Kim, Do-Hyung (2018). "Blue light-emitting diode photoinactivation inhibits edwardsiellosis in fancy carp ( Cyprinus carpio )". Aquaculture. 483: 1–7. Bibcode:2018Aquac.483....1R. doi:10.1016/j.aquaculture.2017.09.046. ISSN   0044-8486.
  128. Maclean, Michelle; MacGregor, Scott J.; Anderson, John G.; Woolsey, Gerry (2009). "Inactivation of Bacterial Pathogens following Exposure to Light from a 405-Nanometer Light-Emitting Diode Array". Applied and Environmental Microbiology. 75 (7): 1932–1937. Bibcode:2009ApEnM..75.1932M. doi:10.1128/AEM.01892-08. ISSN   0099-2240. PMC   2663198 . PMID   19201962.
  129. Ghate, Vinayak S.; Ng, Kheng Siang; Zhou, Weibiao; Yang, Hyunsoo; Khoo, Gek Hoon; Yoon, Won-Byong; Yuk, Hyun-Gyun (2013). "Antibacterial effect of light emitting diodes of visible wavelengths on selected foodborne pathogens at different illumination temperatures". International Journal of Food Microbiology. 166 (3): 399–406. doi:10.1016/j.ijfoodmicro.2013.07.018. PMID   24026011.
  130. Kumar, A.; Ghate, V.; Kim, M.J.; Zhou, W.; Khoo, G.H.; Yuk, H.G. (2016). "Antibacterial efficacy of 405, 460 and 520 nm light emitting diodes on Lactobacillus plantarum , Staphylococcus aureus and Vibrio parahaemolyticus". Journal of Applied Microbiology. 120 (1): 49–56. doi: 10.1111/jam.12975 . PMID   26481103. S2CID   6134117.
  131. Ghate, Vinayak; Kumar, Amit; Zhou, Weibiao; Yuk, Hyun-Gyun (2015). "Effect of organic acids on the photodynamic inactivation of selected foodborne pathogens using 461 nm LEDs". Food Control. 57: 333–340. doi:10.1016/j.foodcont.2015.04.029. ISSN   0956-7135.
  132. 1 2 Prasad, Amritha; Gänzle, Michael; Roopesh, M.S. (2021). "Antimicrobial activity and drying potential of high intensity blue light pulses (455 nm) emitted from LEDs". Food Research International. 148: 110601. doi:10.1016/j.foodres.2021.110601. PMID   34507746.
  133. 1 2 Prasad, Amritha; Gänzle, Michael; Roopesh, M. S. (2019-12-13). "Inactivation of Escherichia Coli and Salmonella Using 365 and 395 nm High Intensity Pulsed Light Emitting Diodes". Foods. 8 (12): 679. doi: 10.3390/foods8120679 . ISSN   2304-8158. PMC   6963940 . PMID   31847186.
  134. de Menezes, Henrique D.; Rodrigues, Gabriela B.; Teixeira, Simone de Pádua; Massola, Nelson S.; Bachmann, Luciano; Wainwright, Mark; Braga, Gilberto U. L. (2014). "In Vitro Photodynamic Inactivation of Plant-Pathogenic Fungi Colletotrichum acutatum and Colletotrichum gloeosporioides with Novel Phenothiazinium Photosensitizers". Applied and Environmental Microbiology. 80 (5): 1623–1632. Bibcode:2014ApEnM..80.1623D. doi:10.1128/AEM.02788-13. ISSN   0099-2240. PMC   3957600 . PMID   24362436.
  135. Al-Asmari, Fahad; Mereddy, Ram; Sultanbawa, Yasmina (2017). "A novel photosensitization treatment for the inactivation of fungal spores and cells mediated by curcumin". Journal of Photochemistry and Photobiology B: Biology. 173: 301–306. doi:10.1016/j.jphotobiol.2017.06.009. PMID   28623822.
Academic journals focused on photodynamic science and technology
Professional associations promoting research on photodynamic therapy