NITRIC OXIDE-RELEASING ANTIBACTERIAL COMPOUNDS, FORMULATIONS, AND METHODS PERTAINING THERETO

Methods of sanitizing, disinfecting, decontaminating, and/or sterilizing surfaces with small molecule NO releasing compounds are disclosed. In some embodiments, the compounds are covalently modified to store and release nitric oxide. The compounds may be tailored to release nitric oxide in a controlled manner and can be useful, for example, for preventing the spread of microbial infections, such as nocosomial infections.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

The present patent application claims the benefit and priority of U.S. Provisional Patent Application No. 62/971,624, filed on Feb. 7, 2020, titled “NITRIC OXIDE-RELEASING ANTIBACTERIAL COMPOUNDS, FORMULATIONS, AND METHODS PERTAINING THERETO,” the contents of which is hereby incorporated by reference in its entirety.

FIELD

The present disclosure relates generally to nitric oxide-releasing compounds, their synthesis, and their use in sanitizing, disinfecting, decontaminating, and/or sterilizing various surfaces. Disinfectant formulations comprising these compounds and their methods of use are also disclosed.

BACKGROUND

Respiratory viruses include rhinoviruses and enteroviruses (Picornaviridae), influenza viruses (Orthomyxoviridae), parainfluenza, metapneumovirus and respiratory syncytial viruses (Paramyxoviridae), coronaviruses (Coronaviridae), and several adenoviruses, and these also pose a great challenge to human health. It is believed that individuals can be infected by contacting contaminated surfaces, so it is important to have compositions that can safely disinfect, and, ideally, decontaminate such surfaces.

Bacterial infections also pose a great challenge to human health in community and hospital settings, and antibacterial resistance is leading to multi-drug resistant bacteria that are becoming increasingly difficult to treat. It is increasingly important to be able to decontaminate most, if not all, surfaces in a hospital setting, including the floors, walls, ceilings, medical equipment, and the like.

Conventional treatments for disinfecting inorganic surfaces have included quaternary ammonium salts, such as those found in Lysol®, bleach, ozone, UV light, and the like.

It would be advantageous to have alternative treatments for disinfecting/decontaminating these surfaces.

Nitric oxide is known for having such an orthogonal antimicrobial mechanism of action. See, e.g., U.S. Patent Application Publication No. 2019/0322770. While the precise mechanisms by which nitric oxide (NO) kills or inhibits the replication of a variety of intracellular pathogens is not completely understood, reactivity towards iron centers involved in cellular metabolism, the imposition of nitrosative stress, and activation of host immunity are likely implicated. Nitric oxide is also understood to target cysteine proteases (Saura et al., Immunity, Volume 10, Issue 1, 1 Jan. 1999, Pages 21-28). NO S-nitrosylates the cysteine residue in the active site of certain viral proteases, inhibiting protease activity and interrupting the viral life cycle. Since cysteine proteases are critical for virulence or replication of many viruses, bacteria, and parasites, NO can be used to treat microbial infections. While NO may have useful antimicrobial properties, the application of NO gas to surfaces is difficult, and at relatively high concentrations, NO gas can be harmful to human health.

NO-releasing compounds (i.e., NO donors) have been proposed as therapeutics, often in the form of polymers with side-chains that include nitric oxide-releasing moieties, such as nitroso-thiols and diazeniumdiolates. Nitric oxide-releasing polymers have hetetofore been underused as therapeutics, based at least in part on limited NO payloads, NO release rates that are more rapid than desired, and the lack of targeted NO delivery. Further, if applied to surfaces, the residual polymer, following release of nitric oxide, might be undesirable from a cleanliness perspective.

It would be advantageous to have disinfecting/decontaminating NO-releasing compounds that can deliver antimicrobial concentrations of NO to a surface to be treated, compositions including these compounds, and methods of disinfecting and/or decontaminating a surface using the compositions. The present invention provides such compounds, compositions, and methods.

SUMMARY

Nitric oxide, an endogenously produced diatomic free radical, is associated with numerous biological processes. Application of compounds that release NO (NO donor compounds) can be an effective strategy for disinfecting and/or decontaminating microbial contamination on surfaces, such as inorganic surfaces. Nitric oxide-releasing compounds (also referred to as nitric oxide donors or NO donors), compositions containing such compounds, and methods of disinfecting and/or decontaminating surfaces using the compounds and compositions, are disclosed.

As disclosed herein, a variety of items or surfaces may require processing in order to reduce the bioburden and decrease risk of infections. For example, critical items (such as surgical instruments, which contact sterile tissue), semi-critical items (such as endoscopes, which contact mucous membranes), and noncritical items (such as stethoscopes, which contact only intact skin) require various types of treatment, for example sterilization, high-level disinfection, and low-level disinfection, respectively. The present disclosure provides for various formulations and methods for disinfecting/decontaminating/sterilizing various items (e.g., medical devices or electronics) and surfaces (e.g., workspaces, and patient rooms).

Some embodiments pertain to a method of reducing or preventing microbial load on a surface. The compounds described herein, or formulations comprising them, can be applied to a surface contaminated with a plurality of microbes. The compounds or compositions generate nitric oxide and induce oxidative and/or nitrosative damage to microbial DNA and membrane structures, thereby preventing growth of or reducing microbial load.

In several embodiments, the plurality of microbes comprises one, two, or more of the following: gram-positive bacteria, gram-negative bacteria, fungi, yeast, and viruses.

In one embodiment, the surface is an inorganic surface, such as the external or internal surface of a medical device, such as an endoscope, and in one aspect of this embodiment, the compound generates an anti-microbial coating on the external or internal surface of the medical device.

In several embodiments, the surface is a solid surface in need of disinfection.

In some embodiments, the surface is associated with an animal or its surroundings, such as the surface of an livestock animal and its pens, cages, or holding areas, such as chicken coops.

In some embodiments, the surface is a hard surface, such as any hard surface found in the home or an industrial, institutional or medical setting. In another embodiment, the hard surface is a floor, wall, countertop, appliance, or fixture.

In one embodiment, a method of preventing and/or decreasing microbial contamination on a surface is disclosed. In some embodiments, the method comprises contacting a surface contaminated with a plurality of microbes (or that a surface that could be exposed to microbes) with one or more of the NO-donor compounds or formulations discussed herein. In some embodiments, a NO-donor compound or formulation including the compound generates NO and induces damage to the membrane and/or DNA of the microbes, thereby reducing the number of viable microbes and/or preventing the colonization or infection of an area with microbes. In several embodiments, the surface comprises an organic surface. In some embodiments, the surface is an inorganic surface, such as that of a device, for example, an external or internal surface of a medical device, such as a medical, dental, or optometric device. In some embodiments, the surface is a hard surface. In some embodiments, the surface is chicken coop. In some embodiments, the plurality of microbes comprises one or more of viruses, gram positive bacteria, gram negative bacteria, drug resistant bacteria, molds, yeasts, fungi, and combinations thereof.

Various formulations and methods are disclosed herein in order to sanitize, disinfect, decontaminate and/or sterilize one or more items, surfaces etc.

The present disclosure also provides various embodiments of compounds, formulations and methods for reducing or removing the build-up of mold, bacteria, biofilm, and other pathogens that may arise in appliances such as dishwashers, dryers, and/or washing machines, particularly front loading washing machines and in a fruit and vegetable containment compartments of refrigerators.

The present disclosure also provides various embodiments of consumer product applications for sterilization, disinfection, sanitization, and/or decontamination. Examples of such use can include cosmetics (e.g., make up applicators), eyewear, dental products, toothbrushes, home use products for a medical condition (e.g., CPAP masks), infant care products, and pet care products. In general, the present disclosure applies to various industries that include but are not limited to, health care, sports medicine, veterinary care, dental care, agriculture, food processing, research, packaging, pharmaceuticals, home health, day care, senior care, private and public services, and military/emergency field care.

The process involves cleaning, disinfecting, and/or decontaminating food processing equipment that has been disassembled for cleaning. Representative equipment, and parts thereof, include, but are not limited to, fittings, clamps, product handling utensils, tank vents, pump rotors, impellers, casings, and hoses. In some embodiments, the process involves cleaning the interior surfaces of food process equipment, such as tanks, pipes, and pumps. Representative food processing/food contact surfaces include, but are not limited to, fillers, mixers, conveyors, equipment, pipelines, tanks, vats, evaporators, and pasteurizers. Additional food processing non-food contact surfaces include, but are not limited to, floors, walls, tables, chairs, benches, drains, troughs, and drip pans.

In some embodiments the hard, non-porous, outside surface of air-tight sealed packages containing food or other products can be sanitized using the compounds and formulations disclosed herein.

Additional items in the orthopedic medicine area that can be sterilized, disinfected, sanitized, and/or decontaminated include orthopedic fixtures, orthotics, ultrasound machines, and surgical implant parts.

In some embodiments, the sterilizing, disinfecting, sanitizing, and/or decontaminating effects can be achieved in a chamber or other closed container, into which items to be sterilized, disinfected, sanitized, and/or decontaminated are placed. In one aspect of this embodiment, the chamber is in the form of a movable chamber (e.g., a rotating tumbler), and items like surgical masks, fabrics, medical waste, and the like can be contacted with the formulations. The compounds in the formulations can then degrade and release nitric oxide and nitroxyl (NHO) groups, and treat the items within the chamber. If desired, to accelerate the process, the chamber can be heated above room temperature, for example, to a temperature of up to around 40° C. In another aspect of this embodiment, the chamber is a stationary chamber, which can be preferred for more solid items than fabrics.

In some embodiments, the chamber can comprise a flexible bag or other compliant container that can encompass items of irregular shapes.

In some embodiments, the chamber can comprise an entire room or a whole commercial or residential building. In some embodiments, inside the chamber there is a container of custom size and shape based on the device or devices to be placed inside the container for sterilization, disinfection, sanitation, and/or decontamination.

In some embodiments, the chamber can be operated at room temperature so that heat sensitive materials (e.g., plastics, food, and/or live tissue) can be sterilized, disinfected, sanitized, and/or decontaminated. In other embodiments, a modest temperature increase, for example, up to 40° C., is affected, but with temperatures remaining low enough to avoid damage to the items to be sterilized, disinfected, sanitized, and/or decontaminated.

In one embodiment, the compounds disclosed herein have the following formula:

wherein:

    • X is selected from the group consisting of H, D, R, and RC(O)—,
    • R is C1-12 alkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, optionally substituted with one or more substituents as defined herein,
    • and M+ is a pharmaceutically-acceptable cation.

In some embodiments, M+ is a cation with a valence other than one, for example, +2 or +3 in which case the ratio of the compound of Formula I to the cation is such that the total positive charge equals the total negative charge. So, for a compound with a total charge of negative three, and a cation with a total charge of positive two, there would be two compounds and three cations.

Representative positively charged cations include sodium, potassium, lithium, calcium, magnesium, and quaternary ammonium salts.

Methods for making these compounds are also disclosed. In one embodiment, compounds with an R(CO)— moiety that does not include acidic a C—H (i.e., alpha to the carbonyl), such as aryl, heteraryl, and branched alkyl groups, like t-butyl groups, can be prepared by reacting all acidic a C—H on the methyl group of a compound with the formula R(CO)CH3 with nitric oxide in basic methanol to give trisdiazeniumdiolates. A representative reaction is shown below:

By way of example, the reaction product of acetophenone with nitric oxide in KOH/methanol is:

In another embodiment, compounds where X is H or D can be prepared by reacting acetone with nitric oxide in basic methanol or deuterated methanol to give tris-diazeniumdiolates.

In another embodiment, the NO-releasing compound has the structure of Formula II:

where M+ is as defined above with respect to Formula I. In some embodiments, the cation is sodium, lithium, potassium, or a quaternary ammonium salt.

The compounds of Formula II can be prepared, for example, by reacting acetone, acetonitrile, or ethanol with NO, optionally at high pressures (i.e., pressures above atmospheric pressure, ideally above about 2 ATM of pressure, and preferably above about 10 ATM of pressure) in the presence of a base, such as a methoxide/methanol solution, to form one or more diazeniumdiolate-containing species. In several embodiments, high purity compounds (greater than about 80%, greater than about 90%, greater than about 95%, or greater than about 98%) can be produced according to the methods disclosed herein.

In still another embodiment, the NO-releasing compound has the structure of Formula III:

In some embodiments, the compounds of any of Formulas I, II or III have a purity in excess of 96%, in excess of 97%, in excess of 98%, in excess of 99%, or in excess of 99.5%. The present disclosure also relates to compounds having this purity level.

In one embodiment, the nitric oxide-releasing compound has a NO-release half-life, at room temperature, of between 0.1 and 24 hours. In another embodiment, the NO-release half-life is at least 15 minutes. In some embodiments, the compound has a total releasable NO storage in a range of 2-10 μmol of NO per mg of NO donor compound. In several embodiments, the compound has a total duration of NO release in the range of 1-60 hours. In several embodiments, the total NO release after 4 hours is in the range between 0.1-1.0 μmol of NO per mg of compound.

The compounds can be formulated in a variety of disinfectant/decontaminant compositions, for delivery to a surface to be treated, for example, wipes, sprays, aerosols, dips, and the like.

The surface can be treated using any means known to those of ordinary skill in the art including, but not limited to, dipping, soaking, brushing, spraying, mopping, washing, and the like. The length of treatment required will vary according to treatment conditions, the selection of which is known to those skilled in the art.

In one embodiment, the disinfectant/decontaminant compositions comprise one or more nitric oxide-releasing compounds described herein and an aqueous or alcoholic solution, where the aqueous solution may optionally be buffered to a pH that does not allow for rapid degradation of the compounds. In one aspect of this embodiment, the nitric oxide releasing compound has an aqueous solubility of at least about 25 mg/ml in the aqueous solution, at a physiologically compatible pH.

The compositions can further include one or more additional active agents, depending on the type of microbial surface to be decontaminated. For example, where the surface is contaminated with a bacteria, virus, or fungi, one or more antibacterial, antiviral, or antifungal compounds, such as quaternary ammonium salts, can be present.

Methods for sanitizing, disinfecting, decontaminating, and/or sterilizing surfaces also disclosed. In some embodiments, the methods involve administering one or more of the compounds disclosed herein to a surface in an effective amount to bring about the desired antimicrobial effect, namely, sanitizing, disinfecting, decontaminating, and/or sterilizing the surfaces.

Representative microbes include viruses, Gram-positive bacteria, Gram-negative bacteria, drug-resistant bacteria, molds, yeasts, fungi, and combinations thereof. In some embodiments, the microbes include one, two, or more of the following: gram-positive bacteria, gram-negative bacteria, drug-resistant bacteria, fungi, yeast, and viruses.

While not wishing to be bound by a particular theory, it is believed that the compounds generate nitric oxide and induce oxidative and/or nitrosative damage to microbial DNA and membrane structures, thereby killing the microbes present on the surfaces. In some embodiments, a NO-donor of the compound or composition generates NO and induces damage to the membrane and/or DNA of the microbes.

Representative microbes include bacteria, fungi, and viruses infections, specifically including those which result in gastrointestinal disorders, respiratory disorders, and sexually transmitted diseases.

Representative viruses that can be killed include those associated with one or more of human immunodeficiency virus, herpes simplex virus, papilloma virus, parainfluenza virus, influenza, hepatitis, Coxsackie Virus, herpes zoster, measles, mumps, rubella, rabies, pneumonia, hemorrhagic viral fevers, H1N1, SARS, MERS, and SARS-CoV2. Representative fungi that can be killed include those associated with mold, including black mold, Candida albicans, and Aspergillus niger. Representative bacteria that can be killed include Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus, Group A streptococci, S. pneumoniae, Mycobacterium tuberculosis, Campylobacter jejuni, Salmonella, Shigella, carbapenem-resistant Enterobacteriaceae Methicillin-resistant Staphylococcus aureus, and Burkholderia cepacia. In several embodiments, the microbial load comprises Methicillin-resistant Staphylococcus aureus. In several embodiments, the microbe comprises carbapenem-resistant Enterobacteriaceae. In several embodiments, the microbe comprises Staphylococcus aureus. In several embodiments, the microbial comprises Pseudomonas aeruginosa. In several embodiments, the microbial comprises Burkholderia cepacia. In some embodiments, the microbe is a parasite.

In several embodiments, the microbe being killed is present on an organic surface, such as a crop locus, tree, plant, fruit or vegetable.

The embodiments discussed above will be better understood with reference to the following detailed description.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows the FTIR spectrum of the compound of Formula III.

FIG. 2 shows an ion exchange chromatogram of a composition including the compound of Formula III with a retention time of 11 minutes (λ=252 nm). The UV absorbance spectrum of the analyte retained for approximately 6 minutes, the UV absorbance spectrum of the analyte retained for approximately 10 minutes, and the UV absorbance spectrum of the analyte retained for approximately 11 minutes are shown in the insets.

FIG. 3 shows the 1H NMR spectrum of the compound of Formula III in D2O.

FIG. 4 shows the 13C NMR spectrum of the compound of Formula III

FIG. 5 shows the 2D NMR of the compound of Formula III.

FIG. 6 shows A) HPLC chromatograms (IEX-UV) with the top chromatogram showing the separation of components prior to acid degradation of the compound of Formula III (referenced as MD3), the middle chromatogram showing the separation of components after 5 h of acid degradation, and the bottom chromatogram showing the separation of components after 24 h of acid degradation, B) the 1H NMR spectrum of the acid degraded components, C) a table of NOA Totals for compound of Formula III before and after acid degradation.

FIG. 7 is a 1H NMR spectrum of a the reaction products when compound of Formula III was neutralized (pH 7) at room temperature.

FIG. 8 is a 13C NMR spectrum of the reaction products when compound of Formula III was neutralized (pH 7) at room temperature.

FIG. 9 shows a graphical representation of the nitric oxide analysis (NOA) release profile for compound of Formula III at pH 7.4, as measured by chemiluminescence, showing 6.7 μmol NO/mg of material being released with a T1/2 of approximately 3.75 h, suggesting release of only a single diazeniumdiolate group, which has a theoretical load of 7.6 μmol NO/mg.

FIG. 10 compares the MBC results for 21 strains of P. aeruginosa when grown under aerobic and anaerobic conditions.

FIGS. 11A and B are graphs showing the dose dependent time kill results of P. aeruginosa, in terms of (CFU/ml) versus time (hours), following exposure to the compound of Formula III.

FIG. 12 shows the effect of pH on the time kill results of P. aeruginosa following exposure to the compound of Formula III (0.125 mg/ml).

FIG. 13 shows the effect of pH on the time kill results of P. aeruginosa following exposure to the compound of Formula III (0.0625 mg/ml).

FIG. 14 shows the effect of pH on the time kill results of P. aeruginosa following exposure to the compound of Formula III (0.03125 mg/ml).

FIG. 15 compares the chromatograms (impurity profiles) of different lots of the compound of Formula III that were manufactured with starting reactants acetonitrile (top), ethanol (middle), and acetone (bottom).

DETAILED DESCRIPTION

Formulations and methods for delivering NO-releasing compounds to a surface to be treated are disclosed. In some embodiments, the compounds are present in disinfectant/decontaminating compositions with desired physical properties, such as viscosity and gelation.

As discussed in greater detail herein, formulations and methods for sterilizing, disinfecting, sanitizing, and/or decontaminating a variety of items, ranging from surgical equipment or other medical devices to electronic equipment, as well as services, rooms, and other items including, but not limited to soft goods, foods, and related manufacturing equipment.

A general overview will be provided, with additional detail related to each of the components of such systems and devices provided below.

The formulations disclosed herein can be formulated into any form that enables application of a composition disclosed herein in a manner that achieves a desired beneficial effect. In some embodiments, the formulation is a powder, an aqueous or alcoholic solution, an aerosol, a spray, a foam, a soap, a suspension, an emulsion, a gel, or a wipe. In one embodiment, aqueous solutions are buffered, for example, to a pH between about 10 and about 12, for long-term storage, and, if intended to be used immediately, between about 6 and about 8.

The formulations disclosed herein can be applied, for example, by rubbing, pouring, sprinkling, or spraying on, or otherwise applying the formulations to a surface to be disinfected. The formulations can be applied by introducing them into or onto a solid support, such as a wipe, a towelette, a towel, a mitt, a glove, or a mop, and then applying the formulation to a surface to be disinfected. The formulation can be applied using a delivery device, such as an aerosol dispenser, a pump spray, a trigger spray, or a squeeze bottle.

The formulation can be allowed to remain on the treated surface for a specified amount of time. In one embodiment, a specified amount of time is a time sufficient to clean a medical device, an inorganic surface, or a tree, plant, fruit or vegetable, and the like.

In one embodiment, a specified amount of time is a time sufficient to disinfect a medical device, an inorganic surface, or a tree, plant, fruit or vegetable. In yet another embodiment, a specified amount of time is a time sufficient to sterilize a medical device, an inorganic surface, or a tree, plant, fruit or vegetable.

In aspects of this embodiment, a composition disclosed herein is applied for about 1 minute to about 5 minutes, about 1 minute to about 10 minutes, about 1 minute to about 15 minutes, about 1 minute to about 20 minutes, about 1 minute to about 25 minutes, about 1 minute to about 30 minutes, about 1 minute to about 35 minutes, about 1 minute to about 40 minutes, about 1 minute to about 45 minutes, about 1 minute to about 50 minutes, about 1 minute to about 55 minutes, about 1 minute to about 60 minutes, about 5 minutes to about 10 minutes, about 5 minutes to about 15 minutes, about 5 minutes to about 20 minutes, about 5 minutes to about 25 minutes, about 5 minutes to about 30 minutes, about 5 minutes to about 35 minutes, about 5 minutes to about 40 minutes, about 5 minutes to about 45 minutes, about 5 minutes to about 50 minutes, about 5 minutes to about 55 minutes, about 5 minutes to about 60 minutes, about 5 minutes to about 70 minutes, about 5 minutes to about 80 minutes, about 5 minutes to about 90 minutes, about 5 minutes to about 100 minutes, about 5 minutes to about 110 minutes, about 5 minutes to about 120 minutes.

In some embodiments, the methods for cleaning disinfecting, decontaminating, and/or sterilizing further comprise a rinsing step using a rinse solution, which can be, for example, water. Typically, the rinse solution is used to rinse a cleaned, disinfected and/or sterilized surface.

The compounds are small molecules, i.e., have a molecular weight below around 500 g/mol, and, in some embodiments, around 200 g/mol, not including the associated cation. One of the advantages of using small molecules over polymers is that the compounds can be prepared with relatively lower impurity levels than polymeric compounds. Further, relative to polymeric compounds, the NO load can be higher, because the percent composition ratio between NO to the scaffold can be maximized, as described herein.

Small molecule precursor compounds, which can be converted to the NO-releasing compounds described herein, can be selected with a relatively low number of reactive groups, for example, a hydrogens adjacent a carbonyl group, reducing the possibility that many different species will result from a nitrosation reaction. As a result, nitrosylation of the NO-precursor can proceed with little or no partial reaction products, which provides the potential for relatively pure products.

Knowing the structure of the small molecule allows for more predictable release kinetics than that obtainable with polymers. Additionally, and desirably, the cation associated with the negatively charged diazenium ions can be selected such that it also has desirable properties. For example, quaternary ammonium salts also have antimicrobial properties.

Before the subject disclosure is further described, it is to be understood that the disclosure is not limited to the particular embodiments of the disclosure described below, as variations of the particular embodiments may be made and still fall within the scope of the appended claims. It is also to be understood that the terminology employed is for the purpose of describing particular embodiments, and is not intended to be limiting. Instead, the scope of the present disclosure will be established by the appended claims.

In this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs.

The present invention will be better understood with reference to the following definitions.

Definitions

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this subject matter belongs. The terminology used in the description of the subject matter herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the subject matter.

The term “effective amount,” as used herein, refers broadly to that amount of a recited compound effective to treat, prevent, or reduce the microbial load on a surface to be treated.

Sterilization, disinfection, sanitization, and decontamination methods are used in a broad range of applications, and have used an equally broad range of sterilization, disinfection, sanitization, and decontamination agents. The term “sterilization” generally refers to the inactivation of bio-contamination, especially on inanimate objects. The term “disinfection” generally refers to the inactivation of organisms considered pathogenic. Although the term “sterilization” may be used in describing certain embodiments herein, it would be appreciated that, unless otherwise indicated, such embodiments can also be used for disinfection (e.g., high-level disinfection, low-level disinfection, etc.), sanitization, and/or other types of decontamination, e.g., as provided with their regulatory definitions.

As mentioned above, the term “sterilization” shall be appreciated to not only encompass the removal of all or substantially all microorganisms and or other pathogens from an object or surface but shall also encompass (unless otherwise specified) disinfection, sanitizing, and decontamination.

The terms “disrupting” and “eradicating” refer to the ability of the presently disclosed structures to combat biofilms. The biofilms may be partially eradicated or disrupted, meaning that the cells no longer attach to one another or to a surface. The biofilm may be completely eradicated, meaning that the biofilm is no longer an interconnected, cohesive, or continuous network of cells to a substantial degree.

“Treat” or “treating” or “treatment” refers broadly to any type of action that imparts a desired antimicrobial effect on a surface, including sanitizing, disinfecting, decontaminating, or sterilizing the surface.

The terms “disrupting” and “eradicating” refer broadly to the ability of the presently disclosed structures to combat biofilms. The biofilms may be partially eradicated or disrupted, meaning that the cells no longer attach to one another or to a surface. The biofilm may be completely eradicated, meaning that the biofilm is no longer an interconnected, cohesive, or continuous network of cells to a substantial degree.

The terms “nitric oxide donor” or “NO donor” refer broadly to species and/or molecules that donate, release and/or directly or indirectly transfer a nitric oxide species, and/or stimulate the endogenous production of nitric oxide in vivo and/or elevate endogenous levels of nitric oxide in vivo, such that the biological activity of the nitric oxide species is expressed at the intended site of action.

The terms “nitric oxide releasing” or “nitric oxide donating” refer to species that donate, release and/or directly or indirectly transfer any one (or two or more) of the three redox forms of nitrogen monoxide (NO+, NO−, NO (e.g., NO)) and/or methods of donating, releasing and/or directly or indirectly transferring any one (or two or more) of the three redox forms of nitrogen monoxide (NO+, NO−, NO). In some embodiments, the nitric oxide releasing is accomplished such that the biological activity of the nitrogen monoxide species is expressed at the intended site of action.

The term “microbial infection” as used herein refers broadly to bacterial, fungal, viral, yeast infections, as well other microorganisms, and combinations thereof.

For the general chemical formulas provided herein, if no substituent is indicated, a person of ordinary skill in the art will appreciate that the substituent is hydrogen. A bond that is not connected to an atom, but is shown, indicates that the position of such substituent is variable. A jagged line, wavy line, two wavy lines drawn through a bond or at the end of a bond indicates that some additional structure is bonded to that position. For a great number of the additional monomers disclosed herein, but not explicitly shown in structures, it is understood by those in the art of polymers, that these monomers can be added to change the physical properties of the resultant polymeric materials even where the elemental analysis would not indicate such a distinction could be expected. Such physical properties include solubility, charge, stability, cross-linking, secondary and tertiary structure, and the like. Moreover, if no stereochemistry is indicated for compounds having one or more chiral centers, all enantiomers and diasteromers are included. Similarly, for a recitation of aliphatic or alkyl groups, all structural isomers thereof also are included. Unless otherwise stated, groups shown as A1 through An and referred to herein as an alkyl group, in the general formulas provided herein are independently selected from alkyl or aliphatic groups, particularly alkyl having 20 or fewer carbon atoms, and even more typically lower alkyl having 10 or fewer atoms, such as methyl, ethyl, propyl, isopropyl, and butyl. The alkyl may be optionally substituted (e.g., substituted or not substituted, as disclosed elsewhere herein). The alkyl may be a substituted alkyl group, such as alkyl halide (e.g. —CX3 where X is a halide, and combinations thereof, either in the chain or bonded thereto), alcohols (e.g. aliphatic or alkyl hydroxyl, particularly lower alkyl hydroxyl) or other similarly substituted moieties such as amino-, amino acid-, aryl-, alkyl aryl-, alkyl ester-, ether-, keto-, nitro-, sulfhydryl-, sulfonyl-, sulfoxide modified-alkyl groups.

The term “amino” and “amine” refer to nitrogen-containing groups such as NR3, NH3, NHR2, and NH2R, wherein R can be as described elsewhere herein. Thus, “amino” as used herein can refer to a primary amine, a secondary amine, or a tertiary amine. In some embodiments, one R of an amino group can be a diazeniumdiolate (e.g., NONO).

Whenever a group is described as being “optionally substituted” that group may be unsubstituted or substituted with one or more of the indicated substituents. Likewise, when a group is described as being “unsubstituted or substituted” (or “substituted or unsubstituted”) if substituted, the substituent(s) may be selected from one or more of the indicated substituents. If no substituents are indicated, it is meant that the indicated “optionally substituted” or “substituted” group may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), cycloalkyl(alkyl), heteroaryl(alkyl), heterocyclyl(alkyl), hydroxy, alkoxy, acyl, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, nitro, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, an amino, a mono-substituted amine group, a di-substituted amine group, a mono-substituted amine(alkyl), a di-substituted amine(alkyl), a diamino-group, a polyamino, a diether-group, and a polyether-group.

As used herein, “Ca to Cb” in which “a” and “b” are integers refer to the number of carbon atoms in a group. The indicated group can contain from “a” to “b”, inclusive, carbon atoms. Thus, for example, a “C1 to C4 alkyl” or “C1-C4 alkyl” group refers broadly to all alkyl groups having from 1 to 4 carbons, that is, CH3—, CH3CH2—, CH3CH2CH2—, (CH3)2CH—, CH3CH2CH2CH2—, CH3CH2CH(CH3)— and (CH3)3C—. If no “a” and “b” are designated, the broadest range described in these definitions is to be assumed.

As used herein, the term “alkyl” refers broadly to a fully saturated aliphatic hydrocarbon group. The alkyl moiety may be branched or straight chain. Examples of branched alkyl groups include, but are not limited to, iso-propyl, sec-butyl, t-butyl and the like. Examples of straight chain alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl and the like. The alkyl group may have 1 to 30 carbon atoms (whenever it appears herein, a numerical range such as “1 to 30” refers broadly to each integer in the given range; e.g., “1 to 30 carbon atoms” means that the alkyl group may consist of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The “alkyl” group may also be a medium size alkyl having 1 to 12 carbon atoms. The “alkyl” group could also be a lower alkyl having 1 to 6 carbon atoms. An alkyl group may be substituted or unsubstituted. By way of example only, “C1-C5 alkyl” indicates that there are one to five carbon atoms in the alkyl chain, e.g., the alkyl chain is selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), etc. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl and hexyl.

As used herein, the term “alkylene” refers broadly to a bivalent fully saturated straight chain aliphatic hydrocarbon group. Examples of alkylene groups include, but are not limited to, methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene and octylene. An alkylene group may be represented by , followed by the number of carbon atoms, followed by a “*”. For example,

to represent ethylene. The alkylene group may have 1 to 30 carbon atoms (whenever it appears herein, a numerical range such as “1 to 30” refers broadly to each integer in the given range; e.g., “1 to 30 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 30 carbon atoms, although the present definition also covers the occurrence of the term “alkylene” where no numerical range is designated). The alkylene group may also be a medium size alkyl having 1 to 12 carbon atoms. The alkylene group could also be a lower alkyl having 1 to 6 carbon atoms. An alkylene group may be substituted or unsubstituted. For example, a lower alkylene group can be substituted by replacing one or more hydrogens of the lower alkylene group and/or by substituting both hydrogens on the same carbon with a C36 monocyclic cycloalkyl group

The term “alkenyl” used herein refers broadly to a monovalent straight or branched chain radical of from two to twenty carbon atoms containing a carbon double bond(s) including, but not limited to, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl and the like. An alkenyl group may be unsubstituted or substituted.

The term “alkynyl” used herein refers broadly to a monovalent straight or branched chain radical of from two to twenty carbon atoms containing a carbon triple bond(s) including, but not limited to, 1-propynyl, 1-butynyl, 2-butynyl and the like. An alkynyl group may be unsubstituted or substituted.

As used herein, “cycloalkyl” refers broadly to a completely saturated (no double or triple bonds) mono- or multi-cyclic (such as bicyclic) hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro fashion. As used herein, the term “fused” refers broadly to two rings which have two atoms and one bond in common. As used herein, the term “bridged cycloalkyl” refers broadly to compounds wherein the cycloalkyl contains a linkage of one or more atoms connecting non-adjacent atoms. As used herein, the term “spiro” refers broadly to two rings which have one atom in common and the two rings are not linked by a bridge. Cycloalkyl groups can contain 3 to 30 atoms in the ring(s), 3 to 20 atoms in the ring(s), 3 to 10 atoms in the ring(s), 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s). A cycloalkyl group may be unsubstituted or substituted. Examples of mono-cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. Examples of fused cycloalkyl groups are decahydronaphthalenyl, dodecahydro-1H-phenalenyl and tetradecahydroanthracenyl; examples of bridged cycloalkyl groups are bicyclo[1.1.1]pentyl, adamantanyl and norbornanyl; and examples of spiro cycloalkyl groups include spiro[3.3]heptane and spiro[4.5]decane.

As used herein, “cycloalkenyl” refers broadly to a mono- or multi-cyclic (such as bicyclic) hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fully delocalized pi-electron system throughout all the rings (otherwise the group would be “aryl,” as defined herein). Cycloalkenyl groups can contain 3 to 10 atoms in the ring(s), 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s). When composed of two or more rings, the rings may be connected together in a fused, bridged, or spiro fashion. A cycloalkenyl group may be unsubstituted or substituted.

As used herein, “aryl” refers broadly to a carbocyclic (all carbon) monocyclic or multicyclic (such as bicyclic) aromatic ring system (including fused ring systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings. The number of carbon atoms in an aryl group can vary. For example, the aryl group can be a C6-C14 aryl group, a C6-C10 aryl group or a C6 aryl group. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group may be substituted or unsubstituted. As used herein, “heteroaryl” refers to a monocyclic or multicyclic (such as bicyclic) aromatic ring system (a ring system with fully delocalized pi-electron system) that contain(s) one or more heteroatoms (for example, 1, 2 or 3 heteroatoms), that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. The number of atoms in the ring(s) of a heteroaryl group can vary. For example, the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s), such as nine carbon atoms and one heteroatom; eight carbon atoms and two heteroatoms; seven carbon atoms and three heteroatoms; eight carbon atoms and one heteroatom; seven carbon atoms and two heteroatoms; six carbon atoms and three heteroatoms; five carbon atoms and four heteroatoms; five carbon atoms and one heteroatom; four carbon atoms and two heteroatoms; three carbon atoms and three heteroatoms; four carbon atoms and one heteroatom; three carbon atoms and two heteroatoms; or two carbon atoms and three heteroatoms. Furthermore, the term “heteroaryl” includes fused ring systems where two rings, such as at least one aryl ring and at least one heteroaryl ring or at least two heteroaryl rings, share at least one chemical bond. Examples of heteroaryl rings include, but are not limited to, furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, thiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, purine, pteridine, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline and triazine. A heteroaryl group may be substituted or unsubstituted.

As used herein, “heterocyclyl” or “heteroalicyclyl” refers broadly to three-, four-, five-, six-, seven-, eight-, nine-, ten-, up to 18-membered monocyclic, bicyclic and tricyclic ring system wherein carbon atoms together with from 1 to 5 heteroatoms constitute said ring system. A heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout all the rings. The heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur and nitrogen. A heterocycle may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro fashion. As used herein, the term “fused” refers to two rings which have two atoms and one bond in common. As used herein, the term “bridged heterocyclyl” or “bridged heteroalicyclyl” refers to compounds wherein the heterocyclyl or heteroalicyclyl contains a linkage of one or more atoms connecting non-adjacent atoms. As used herein, the term “spiro” refers to two rings which have one atom in common and the two rings are not linked by a bridge. Heterocyclyl and heteroalicyclyl groups can contain 3 to 30 atoms in the ring(s), 3 to 20 atoms in the ring(s), 3 to 10 atoms in the ring(s), 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s). For example, five carbon atoms and one heteroatom; four carbon atoms and two heteroatoms; three carbon atoms and three heteroatoms; four carbon atoms and one heteroatom; three carbon atoms and two heteroatoms; two carbon atoms and three heteroatoms; one carbon atom and four heteroatoms; three carbon atoms and one heteroatom; or two carbon atoms and one heteroatom. Additionally, any nitrogens in a heteroalicyclic may be quaternized. Heterocyclyl or heteroalicyclic groups may be unsubstituted or substituted. Examples of such “heterocyclyl” or “heteroalicyclyl” groups include but are not limited to, 1,3-dioxin, 1,3-dioxane, 1,4-dioxane, 1,2-dioxolane, 1,3-dioxolane, 1,4-dioxolane, 1,3-oxathiane, 1,4-oxathiin, 1,3-oxathiolane, 1,3-dithiole, 1,3-dithiolane, 1,4-oxathiane, tetrahydro-1,4-thiazine, 2H-1,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, trioxane, hexahydro-1,3,5-triazine, imidazoline, imidazolidine, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, morpholine, oxirane, piperidine N-Oxide, piperidine, piperazine, pyrrolidine, azepane, pyrrolidone, pyrrolidione, 4-piperidone, pyrazoline, pyrazolidine, 2-oxopyrrolidine, tetrahydropyran, 4H-pyran, tetrahydrothiopyran, thiamorpholine, thiamorpholine sulfoxide, thiamorpholine sulfone and their benzo-fused analogs (e.g., benzimidazolidinone, tetrahydroquinoline and/or 3,4-methylenedioxyphenyl). Examples of spiro heterocyclyl groups include 2-azaspiro[3.3]heptane, 2-oxaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2,6-diazaspiro[3.3]heptane, 2-oxaspiro[3.4]octane and 2-azaspiro[3.4]octane.

As used herein, “aralkyl” and “aryl(alkyl)” refer broadly to an aryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2-phenylalkyl, 3-phenylalkyl and naphthylalkyl.

As used herein, “cycloalkyl(alkyl)” refer broadly to an cycloalkyl group connected, as a substituent, via a lower alkylene group. The lower alkylene and cycloalkyl group of a cycloalkyl(alkyl) may be substituted or unsubstituted.

As used herein, “heteroaralkyl” and “heteroaryl(alkyl)” refer broadly to a heteroaryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thienylalkyl, 3-thienylalkyl, furylalkyl, thienylalkyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl and imidazolylalkyl and their benzo-fused analogs.

A “heteroalicyclyl(alkyl)” and “heterocyclyl(alkyl)” refer broadly to a heterocyclic or a heteroalicyclic group connected, as a substituent, via a lower alkylene group. The lower alkylene and heterocyclyl of a (heteroalicyclyl)alkyl may be substituted or unsubstituted. Examples include but are not limited tetrahydro-2H-pyran-4-yl(methyl), piperidin-4-yl(ethyl), piperidin-4-yl(propyl), tetrahydro-2H-thiopyran-4-yl(methyl) and 1,3-thiazinan-4-yl(methyl).

As used herein, the term “hydroxy” refers broadly to a —OH group.

As used herein, “alkoxy” refers broadly to the Formula —OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) is defined herein. A non-limiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, phenoxy and benzoxy. An alkoxy may be substituted or unsubstituted.

As used herein, “acyl” refers broadly to a hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) and heterocyclyl(alkyl) connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl and acryl. An acyl may be substituted or unsubstituted.

As used herein, a “cyano” group refers broadly to a “—CN” group.

The term “halogen atom” or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, such as, fluorine, chlorine, bromine and iodine.

A “thiocarbonyl” group refers broadly to a “—C(═S)R” group in which R can be the same as defined with respect to O-carboxy. A thiocarbonyl may be substituted or unsubstituted. An “O-carbamyl” group refers to a “—OC(═O)N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An O-carbamyl may be substituted or unsubstituted.

An “N-carbamyl” group refers broadly to an “ROC(═O)N(RA)—” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-carbamyl may be substituted or unsubstituted.

An “O-thiocarbamyl” group refers broadly to a “—OC(═S)—N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An O-thiocarbamyl may be substituted or unsubstituted.

An “N-thiocarbamyl” group refers broadly to an “ROC(═S)N(RA)—” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-thiocarbamyl may be substituted or unsubstituted.

A “C-amido” group refers broadly to a “—C(═O)N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A C-amido may be substituted or unsubstituted.

An “N-amido” group refers broadly to a “RC(═O)N(RA)—” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-amido may be substituted or unsubstituted.

An “S-sulfonamido” group refers broadly to a “—SO2N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An S-sulfonamido may be substituted or unsubstituted.

An “N-sulfonamido” group refers broadly to a “RSO2N(RA)—” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-sulfonamido may be substituted or unsubstituted.

An “O-carboxy” group refers broadly to a “RC(═O)O—” group in which R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein. An O-carboxy may be substituted or unsubstituted.

The terms “ester” and “C-carboxy” refer broadly to a “—C(═O)OR” group in which R can be the same as defined with respect to O-carboxy. An ester and C-carboxy may be substituted or unsubstituted.

A “nitro” group refers broadly to an “—NO2” group.

A “sulfenyl” group refers broadly to an “—SR” group in which R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A sulfenyl may be substituted or unsubstituted.

A “sulfinyl” group refers broadly to an “—S(═O)—R” group in which R can be the same as defined with respect to sulfenyl. A sulfinyl may be substituted or unsubstituted.

A “sulfonyl” group refers broadly to an “SO2R” group in which R can be the same as defined with respect to sulfenyl. A sulfonyl may be substituted or unsubstituted.

As used herein, “haloalkyl” refers broadly to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl, tri-haloalkyl and polyhaloalkyl). Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1-chloro-2-fluoromethyl, 2-fluoroisobutyl and pentafluoroethyl. A haloalkyl may be substituted or unsubstituted.

As used herein, “haloalkoxy” refers broadly to an alkoxy group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di-haloalkoxy and tri-haloalkoxy). Such groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, 1-chloro-2-fluoromethoxy and 2-fluoroisobutoxy. A haloalkoxy may be substituted or unsubstituted.

The terms “amino” and “unsubstituted amino” as used herein refer broadly to a —NH2 group.

A “mono-substituted amine” group refers broadly to a “—NHRA” group in which RA can be an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein. The RA may be substituted or unsubstituted. A mono-substituted amine group can include, for example, a mono-alkylamine group, a mono-C1-C6 alkylamine group, a mono-arylamine group, a mono-C6-C1 arylamine group and the like. Examples of mono-substituted amine groups include, but are not limited to, —NH(methyl), —NH(phenyl) and the like.

A “di-substituted amine” group refers broadly to a “—NRARB” group in which RA and RB can be independently an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein. RA and RB can independently be substituted or unsubstituted. A di-substituted amine group can include, for example, a di-alkylamine group, a di-C1-C6 alkylamine group, a di-arylamine group, a di-C6-C10 arylamine group and the like. Examples of di-substituted amine groups include, but are not limited to, —N(methyl)2, —N(phenyl)(methyl), —N(ethyl)(methyl) and the like.s used herein, “mono-substituted amine(alkyl)” group refers broadly to a mono-substituted amine as provided herein connected, as a substituent, via a lower alkylene group. A mono-substituted amine(alkyl) may be substituted or unsubstituted. A mono-substituted amine(alkyl) group can include, for example, a mono-alkylamine(alkyl) group, a mono-C1-C6 alkylamine(C1-C6 alkyl) group, a mono-arylamine(alkyl group), a mono-C6-C10 arylamine(C1-C6 alkyl) group and the like. Examples of mono-substituted amine(alkyl) groups include, but are not limited to, —CH2NH(methyl), —CH2NH(phenyl), —CH2CH2NH(methyl), —CH2CH2NH(phenyl) and the like.

As used herein, “di-substituted amine(alkyl)” group refers broadly to a di-substituted amine as provided herein connected, as a substituent, via a lower alkylene group. A di-substituted amine(alkyl) may be substituted or unsubstituted. A di-substituted amine(alkyl) group can include, for example, a dialkylamine(alkyl) group, a di-C1-C6 alkylamine(C1-C6 alkyl) group, a di-arylamine(alkyl) group, a di-C6-C10 arylamine(C1-C6 alkyl) group and the like. Examples of di-substituted amine(alkyl)groups include, but are not limited to, —CH2N(methyl)2, —CH2N(phenyl)(methyl), —CH2N(ethyl)(methyl), —CH2CH2N(methyl)2, —CH2CH2N(phenyl)(methyl), —NCH2CH2(ethyl)(methyl) and the like.

As used herein, the term “diamino-” denotes a “—N(RA)RB—N(RC)(RD)” group in which RA, RC, and RD can be independently a hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein, and wherein RB connects the two “N” groups and can be (independently of RA, RC, and RD) a substituted or unsubstituted alkylene group. RA, RB, RC, and RD can independently further be substituted or unsubstituted.

As used herein, the term “polyamino” denotes a “—(N(RA)RB—)n—N(RC)(RD)”. For illustration, the term polyamino can comprise —N(RA)alkyl-N(RA)alkyl-N(RA)alkyl-N(RA)alkyl-H. In some embodiments, the alkyl of the polyamino is as disclosed elsewhere herein. While this example has only 4 repeat units, the term “polyamino” may consist of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 repeat units. RA, RC, and RD can be independently a hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein, and wherein RB connects the two “N” groups and can be (independently of RA, RC, and RD) a substituted or unsubstituted alkylene group. RA, RC, and RD can independently further be substituted or unsubstituted. As noted here, the polyamino comprises amine groups with intervening alkyl groups (where alkyl is as defined elsewhere herein).

As used herein, the term “diether-” denotes an “—ORBO—RA” group in which RA can be a hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein, and wherein RB connects the two “O” groups and can be a substituted or unsubstituted alkylene group. RA can independently further be substituted or unsubstituted.

As used herein, the term “polyether” denotes a repeating —(ORB—)nORA group. For illustration, the term polyether can comprise —Oalkyl-Oalkyl-Oalkyl-Oalkyl-ORA. In some embodiments, the alkyl of the polyether is as disclosed elsewhere herein. While this example has only 4 repeat units, the term “polyether” may consist of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 repeat units. RA can be a hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein. RB can be a substituted or unsubstituted alkylene group. RA can independently further be substituted or unsubstituted. As noted here, the polyether comprises ether groups with intervening alkyl groups (where alkyl is as defined elsewhere herein and can be optionally substituted).

Where the number of substituents is not specified (e.g. haloalkyl), there may be one or more substituents present. For example, “haloalkyl” may include one or more of the same or different halogens. As another example, “C1-C3 alkoxyphenyl” may include one or more of the same or different alkoxy groups containing one, two or three atoms. As used herein, a radical indicates species with a single, unpaired electron such that the species containing the radical can be covalently bonded to another species. Hence, in this context, a radical is not necessarily a free radical. Rather, a radical indicates a specific portion of a larger molecule. The term “radical” can be used interchangeably with the term “group.”

When a range of integers is given, the range includes any number falling within the range and the numbers defining ends of the range. For example, when the terms “integer from 1 to 20” is used, the integers included in the range are 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, etc., up to and including 20.

When an amount, concentration, or other value or parameter is given as either a range, preferred range, or a list of upper preferable values and lower preferable values, this is to be understood as specifically disclosing all ranges formed from any pair of any upper range limit or preferred value and any lower range limit or preferred value, regardless of whether ranges are separately disclosed. Where a range of numerical values is recited herein, unless otherwise stated, the range is intended to include the endpoints thereof, and all integers and fractions within the range. It is not intended that the scope of the invention be limited to the specific values recited when defining a range.

The ranges disclosed herein also encompass any and all overlap, sub-ranges, and combinations thereof. Language such as “up to,” “at least,” “greater than,” “less than,” “between,” and the like includes the number recited. Numbers preceded by a term such as “about” or “approximately” include the recited numbers. For example, “about 10 one millipascal-second” includes “10 one millipascal-second.”

Also as used herein, “and/or” refers broadly to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (“or”).

Furthermore, the term “about,” as used herein when referring to a measurable value such as an amount of a compound or agent of this invention, dose, time, temperature, and the like, is meant to encompass variations of 20%, ±10%, +5%, +1%, ±0.5%, or even 0.1% of the specified amount. The term “consists essentially of” (and grammatical variants), shall be given its ordinary meaning and shall also mean that the composition or method referred to can contain additional components as long as the additional components do not materially alter the composition or method. The term “consists of” (and grammatical variants), shall be given its ordinary meaning and shall also mean that the composition or method referred to is closed to additional components. The term “comprising” (and grammatical variants), shall be given its ordinary meaning and shall also mean that the composition or method referred to is open to contain additional components.

The presently disclosed subject matter will now be described more fully hereinafter. However, many modifications and other embodiments of the presently disclosed subject matter set forth herein will come to mind to one skilled in the art to which the presently disclosed subject matter pertains having the benefit of the teachings presented in the foregoing descriptions. Therefore, it is to be understood that the presently disclosed subject matter is not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. In other words, the subject matter described herein covers all alternatives, modifications, and equivalents. In the event that one or more of the incorporated literature, patents, and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in this field. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.

I. Compounds

As disclosed elsewhere herein, some embodiments disclosed herein pertain to small molecules capable of delivering NO to achieve microbicidal activity. In some embodiments, the cations present in the small molecules have antimicrobial or other desired physiological properties. In some embodiments, the compounds are water-soluble.

In one embodiment, provided herein is a NO releasing compound which exhibits potent antimicrobial characteristics, comprising the structure of Formula I:

wherein:

    • X is selected from the group consisting of H, D, R, and RC(O)—,
    • R is C1-12 alkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, optionally substituted with one or more substituents,
    • wherein the substituents are independently selected from the group consisting of —OH, —NH2, —OCH3, —C(O)OH, —CH2OH, —CH2OCH3, —CH2OCH2CH2OH, —OCH2C(O)OH, —CH2OCH2C(O)OH, —CH2C(O)OH, —NHC(O)—CH3, —C(O)O((CH2)aO)b—H, —C(O)O((CH2)aO)b—(CH2)cH, —C(O)O(C1-5alkyl), —C(O)—NH—((CH2)dNH)e—H, —C(O)—NH—((CH2)dNH)e—(CH2)fH, —O—((CH2)aO)b—H, —O—((CH2)aO)b—(CH2)cH, —O—(C1-5alkyl), —NH—((CH2)dNH)e—H, and —NH—((CH2)dNH)e—(CH2)fH,
    • each instance of a, b, c, d, e, f, g, h, i, j, k, and 1 is independently selected from an integer of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10,
    • and M+ is a pharmaceutically-acceptable cation.

In some embodiments, M+ is a cation with a valence other than one, for example, +2 or +3 in which case the ratio of the compound of Formula I to the cation is such that the total positive charge equals the total negative charge. So, for a compound with a total charge of negative three, and a cation with a total charge of positive two, there would be two compounds and three cations.

Representative positively charged cations include sodium, potassium, lithium, calcium, magnesium, and quaternary ammonium salts.

In another embodiment, the compound has the following structure:

wherein M+ refers to a pharmaceutically-acceptable cation. The cation can be any pharmaceutically acceptable, non-toxic cation known to those skilled in the art, including but not limited to sodium, potassium, lithium, calcium, magnesium, ammonium, or substituted ammonium. It will be appreciated by those skilled in the art that when the cation (M) has a valency greater than one, the ratio of negative charge in the methyl trisdiazenium diolate moiety to the positive charge in the cation will balance out. For example, if the cation (M) has a charge of +2, then there is a ratio of 2 methyl trisdiazenium diolate moieties to three M+2 ions, and if the cation (M) has a charge of +3, then there is a 1/1 ratio of cation to methyl trisdiazenium diolate.

One representative compound is shown below:

Formula II is also described as a methane trisdiazeniumdiolate (MTDD), and Formula III as methane trisdiazeniumdiolate sodium salt.

Although various NO donors (e.g., diazeniumdiolates, S-nitrosothiols, metal nitrosyls, organic nitrates) are known to provide for controlled exogenous NO delivery, the diazeniumdiolate moieties in the compounds disclosed herein are attractive because of their good stability and facile storage, and because they spontaneously undergo proton-triggered dissociation under physiological conditions to regenerate nitric oxide, including NO radicals.

The C-diazeniumdiolates described herein are pH-triggered NO-release donors. Reacting with protons under physiological conditions (e.g., 37° C., pH 7.4), 1 mole of Formula III generates two moles of NO radicals and 2 to 3 moles of nitroxyl compounds.

Several embodiments disclosed herein have one or more of the following advantages: efficient and unique synthesis routes and resultant chemical composition of small molecules.

In several embodiments, the NO-releasing compounds are stable at a variety of temperatures 20° C. (e.g., 40° C., 45° C., 55° C., 60° C., 80° C., etc.) and are stable for prolonged storage periods (e.g., 10 hours, 20 hours, 22 hours, 25 hours, 30 hours, etc., days such as 1 day, 3 days, 5 days, 6 days, 7 days, 15 days, 30 days, 45 days, etc., weeks such as 1 week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, etc., months such as 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, etc., or even years (1 year or greater)).

In some embodiments, the compounds have NO storage capacities (in μmol NO/mg of the compounds) of greater than or equal to about: 0.25, 0.4, 0.5, 1.0, 1.5, 2.0, 3.0, or ranges including and/or spanning the aforementioned values. In some embodiments, within 2 h of being added to a PBS buffer solution, the compounds release greater than or equal to about: 25%, 50%, 75%, 85%, 90%, 95%, 100%, or ranges including and/or spanning the aforementioned values, their total wt % of bound NO. In several embodiments, NO release in use for reducing or eliminating a biofilm occurs in similar amounts, e.g., about 20-25%, about 30-50%, about 60-75%, at least 80%, at least 85%, at least 90%, at least 95%, ranges including and/or spanning the aforementioned values, of the total wt % of bound NO.

In some embodiments, the NO release may occur over a period of about 0.01 hours, 0.1 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 10 hours, 15 hours, 20 hours, 24 hours, 36 hours, 48 hours, 60 hours, or ranges including and/or spanning the aforementioned values. In several embodiments, the NO release half-life is equal to or at least about: 0.01 hours, 0.1 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours or ranges including and/or spanning the aforementioned values. In some embodiments, the NO release occurs in less than or equal to about: 0.01 hours, 0.1 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 10 hours, 15 hours, 20 hours, 24 hours or ranges including and/or spanning the aforementioned values.

In some embodiments, the compounds have a degradation rate per hour in an amylase enzyme exposure assay of less than or equal to about: 0.2%, 0.5%, 1.0%, 1.5%, 2.5%, 5.0%, 10%, or ranges including and/or spanning the aforementioned values.

In some embodiments, the compounds have antimicrobial activity. In some embodiments, the compounds can efficiently eradicate or reduce the viability of microbes (e.g., prokaryotic cells, bacteria, viruses, protozoa, fungi, algae, amoebas, slime molds, etc., including drug-resistant microbes) with low toxicity to native tissue and patient cells (e.g., eukaryotic cells, mammalian cells, human cells, etc.).

In some embodiments, the compounds provide greater than or equal to 90% bacterial reduction in a bacterial viability assay performed under static conditions over 2 hours against one or more of P. aeruginosa, S. aureus P. gingivalis, A. actinomycetemcomitans, A. viscosus, and/or S. mutans at a concentration of equal to or less than about: 4 mg/ml, 2 mg/ml, 1 mg/ml, 0.5 mg/ml, 0.1 mg/ml, 0.05 mg/ml or ranges including and/or spanning the aforementioned values. In some embodiments, the disclosed functionalized NO-releasing compounds provide greater than or equal to 99% bacterial reduction and/or a 2 to 3 log reduction in a bacterial viability assay performed under static conditions over 2 hours against a gram positive bacteria at a concentration of equal to or less than about: 4 mg/ml, 2 mg/ml, 1 mg/ml, 0.5 mg/ml, 0.1 mg/ml, 0.05 mg/ml or ranges including and/or spanning the aforementioned values. In some embodiments, the disclosed functionalized NO-releasing polymers provide greater than or equal to 99% bacterial reduction and/or a 2 to 3 log reduction in a bacterial viability assay performed under static conditions over 2 hours against a gram negative bacteria at a r concentration of equal to or less than about: 4 mg/ml, 2 mg/ml, 1 mg/ml, 0.5 mg/ml, 0.1 mg/ml, 0.05 mg/ml or ranges including and/or spanning the aforementioned values. In several embodiments, bacterial reduction is greater than 95%, greater than 98%, or greater than 99%.

II. Compound Synthesis

Several embodiments disclosed herein provide the synthesis and characterization of the diazeniumdiolate NO donor-modified compounds described herein. The synthesis of compounds capable of controlled NO storage and release is important for taking advantage of NO's role in physiology and for developing NO-based therapeutics.

Several embodiments disclosed herein have one or more of the following advantages: efficient and unique synthesis routes and resultant chemical composition of constructs. Certain compounds described herein have been previously disclosed, but the present disclosure describes their synthesis and utility in pharmaceutical compositions and methods of treating or preventing microbial infections, or reducing microbial loads.

There are a number of ways to make the compounds of Formula I. In one embodiment, compounds with an R(CO)— moiety that does not include acidic a C—H (i.e., alpha to the carbonyl), such as aryl, heteraryl, and branched alkyl groups, like t-butyl groups, can be prepared by reacting all acidic a C—H on the methyl group of a compound with the formula R(CO)CH3 with nitric oxide in basic methanol to give trisdiazeniumdiolates. A representative reaction is shown below:

By way of example, the reaction product of acetophenone with nitric oxide in KOH/methanol is:

In another embodiment, compounds where X is H or D can be prepared by reacting acetone with nitric oxide in basic methanol or deuterated methanol to give tris-diazeniumdiolates.

Formula II can be prepared via a number of different approaches, including the reaction of ethanol, acetonitrile, or acetone with nitric oxide gas, in the presence of a basic methanol solution. Ideally, the nitric oxide gas is present at a pressure greater than atmospheric pressure, more ideally, greater than two atmospheres of pressure, and, preferably, greater than ten atmospheres of pressure. Higher pressures help ensure complete reaction. Where there is an incomplete reaction, one of the by-products of the reaction is methane bis-diazeniumdiolate:

Methane bis-diazeniumdiolate does not release NO or NHO under physiological conditions.

The reaction of acetone with nitric oxide, at relatively high pressures, in the presence of basic methanol solutions such as sodium or potassium hydroxide in methanol, tends to provide the purest compound. The 13C NMR for methane tris-diazenium diolate prepared using this approach are shown in FIG. 8, and as can be seen in the figure, there is one product peak.

The proposed reaction mechanism behind this reaction, using sodium hydroxide in methanol to provide the compound where M+ is Na+, is provided below:

The following proposed mechanism relates to the degradation of this compound to form nitric oxide:

1H and 13C NMR spectra for the degradation products are shown in FIGS. 7 and 8, respectively. Although not explicitly shown, it is anticipated that N2O is not produced directly but forms when two moles of nitroxyl (HNO) are released which then dimerize and react to form N2O and H2O as shown. Nitroxyl groups are reactive nitrogen molecules which could play a role in their own right in enhancing the antimicrobial activity of the compound of Formula III.

The degradation pathway is of particular interest, because it provides clarity in how NO is being produced from a carbon bound diazeniumdiolate. Typically, carbon bound diazeniumdiolates do not generate NO. Instead, they produce HNO, which dimerizes to form nitrous oxide, N2O. In this case, the initial decay of Formula III follows the expected HNO pathway, which in turn leads to the formation of an intermediate alcohol. Upon rearrangement of the alcohol intermediate, 2 moles of NO gas can be released. This matches the experimental results, which show that one mole of Formula III releases approximately 2 moles of NO. The NMR of the degraded Formula III by-product (shown in FIGS. 9 and 10) matches the proposed structure for the fully degraded molecule.

III. Sanitizing/Disinfectant/Decontaminating/Sterilizing Formulations

Sanitizing, disinfecting, decontaminating and/or sterilizing formulations comprising one or more compounds of Formulas I, II or III, along with a suitable carrier or excipient, are also disclosed.

According to several embodiments, the compounds described herein can be present in aqueous or alcoholic solutions comprising concentrations equal to or at least about 100 μg/mL, and can be higher, e.g. about 1 mg/ml, about 5 mg/ml, about 10 mg/ml, about 20/ml, or about 40 mg/ml or higher. The solubility of the compounds, in some embodiments, allows for concentrations of up to 125 mg/ml or higher.

In some embodiments, the compositions disclosed herein provide NO-releasing compounds discussed herein having NO storage capacities (in μmol NO/mg powder) of greater than or equal to about: 2.0, 4.0, 6.0, 8.0, or 10.0 or ranges including and/or spanning the aforementioned values. In some embodiments, within 2 h of being added to a PBS buffer solution as described in the Examples, the NO-releasing compounds, release greater than or equal to about: 25%, 50%, 75%, 85%, 90%, 95%, 100%, or ranges including and/or spanning the aforementioned values, their total wt % of bound NO.

In some embodiments, the compositions are in the form of a liquid, a dry powder, a gel, a spray, or an aerosol. The compositions may be provided in the form of a formulation loaded into a wipe, such as a disinfectant wipe.

In some embodiments, the composition includes a concentration of less than or equal to about: 1 mg/ml, 10 mg/ml, 20 mg/ml, 50 mg/ml, 100 mg/ml, or 250 mg/ml of the compounds described herein, or ranges including and/or spanning the aforementioned values.

Powdered Formulations

The powdered formulations described herein include one or more of the compounds described herein, and optionally include one or more of a quaternary ammonium salt, a buffer, a desiccant, an anti-caking additive, and a suspending agent.

Aqueous or Alcoholic Solutions

In several embodiments, the compounds disclosed herein are administered as aqueous or alcoholic solutions, for delivery, for example, via spray, foam, aerosol, drip, and the like. The concentration of the compounds in the solvent is typically between about 0.1 and about 20% by weight, more typically between about 0.5 and about 10% by weight, most typically, between about 1 and about 5% by weight of the solution.

Disinfecting Wipes

One particularly useful application means is to impregnate the compounds disclosed herein and/or compositions comprising the compounds, into a wipe substrate. In one aspect of this embodiment, the wipe is a single use wipe that is impregnated with the disinfecting composition and is stored in a container that will dispense the wipe to a user.

Disinfecting wipes, also known as wet wipes, wet towels, moist towelettes, or baby wipes in specific circumstances, are small to medium-sized moistened pieces of plastic or cloth that often come folded and individually wrapped for convenience, or, in the case of dispensers, as a large roll with individual wipes that can be torn off Wet wipes are used for cleaning purposes, including personal hygiene and surface cleaning.

In one embodiment, the wipes are produced from nonwoven or woven fabrics made of polyester or polypropylene, though in other embodiments, can be prepared from cotton or other natural fibers.

In addition to wipes, the compounds can also be used to impregnate cleansing pads. The impregnated cleansing pads are typically fiber sponges which have been soaked with one or more of the compounds described herein, in an aqueous or alcoholic solvent. Cleansing pads can be stored in sterile packages.

Impregnated industrial-strength cleaning wipes can also be used. These wipes are often cotton rags.

In any form, i.e., wipes or sponges, they are typically moistened with water or other liquids (e.g., ethyl or isopropyl alcohol) depending on the applications. The material may be treated with softeners, lotions, or perfume to adjust the tactile and olfactory properties. Preservatives such as methylisothiazolinone may be added, though the compounds described herein can similarly prevent bacterial or fungal growth in the package. The finished wet wipes can be folded and put in pocket size packages or box dispensers.

In use, the wipes can serve a number of purposes, including cleaning floors, toilet seats, and other surfaces around the home, or surfaces in hospital or private medical care settings, or in other businesses.

In one embodiment, the formulations are absorbed by disinfectant wipes and stored in a sealed container. The compounds described herein are generally stable in alcoholic solutions, but can degrade over time when stored in aqueous solutions. Preferred alcohols are ethanol and isopropyl alcohol, which themselves are antimicrobial.

If the wipes are intended to include aqueous solutions, it can be preferred to either use them within the first several hours of their preparation, or to use a buffered aqueous solution, where the buffer is between a pH of between about 8.4 and about 10, for example, between about 9 and about 9.5.

Whether stored in alcoholic solutions or aqueous solutions, the concentration of the one or more compounds described herein in the solution is typically between about 1 and 250 mg/ml, more typically between about 25 and 125 mg/ml.

In addition to the compounds described herein, the wipes can also be impregnated with additional antimicrobial agents that are not incompatible with the compounds described herein. Examples include quaternary ammonium salts.

In some embodiments, the wipes and solutions including the compositions are stored separately until the wipes are used. For example, batches of disinfectant wipes in a sealed container can be prepared on an as-needed basis. A batch of disinfectant wipes impregnated with the compounds described herein can be quickly prepared, and, ideally, have a shelf-life of up to around eight hours.

For example, the compounds can be present in solution in a pump dispenser, and dispensed as a fluid, preferably as a foam, and absorbed or impregnated in at least one fabric member to form a wet wipe. The wipes can then optionally be stored, for example, in a sealed container, or directly used to clean one or more surfaces.

Optionally, using an approach similar to that disclosed in JP 2005/255657, one can use a single-use device for preparing sterilizing wipes that includes an internal bag which contains a formulation including the compounds described herein, and an external bag which includes sheets that are to be impregnated with the formulation. The external bag can be pressed, or subjected to impact, to break the internal bag and allow the formulation to flow into the sheets. This results in the sheets being impregnated with the compounds described herein. The user can then open the external bag and remove the disinfectant sheets impregnated with the compounds described herein.

Alternatively, using the technology disclosed in U.S. Publication No. 20190023476, wipes, and a burstable sachet including one or more of the compounds described herein, in solution or suspension in a suitable solvent, can be placed in the opening of a package through which the wipes and a burstable sachet may be later removed. In normal use, the opening can be covered, for example, by an adhesive-backed label which seals the inside of the container from the external environment. Ideally, the adhesive label is removable and resealable, and optionally includes a non-adhesive tab portion to aid peeling back of the label.

In use, to prepare a batch of disinfectant wipes, the user presses the container so as to burst the sachet and release the formulation including the one or more compounds within the sealed container. To disperse the formulation evenly between the wipes, the user can, for example, shake the container. Instructions for use may conveniently be provided on the label.

Then, a user can peel back the label and removes the empty sachet from the container, and reseals the container. The disinfectant wipes are ready for use. To dispense an activated disinfectant wipe, a user peels back the label and pulls a wipe out through the opening before replacing the label over the opening.

Aerosols/Sprays

An aerosol spray is a type of dispensing system that creates aerosol mist of liquid particles. It is used with a can or bottle that contains a payload and propellant under pressure. When the container's valve is opened, the payload is forced out of a small opening and emerges as an aerosol or mist. Aerosol spray cans have three major parts: the can, the valve and the actuator or button.

The propellant is typically a gas, and is usually the vapor of a liquid with a boiling point slightly lower than room temperature. Inside the pressurized can, the vapor can exist in equilibrium with its bulk liquid at a pressure that is higher than atmospheric pressure, (and able to expel the payload), but not dangerously high. As gas escapes, it is immediately replaced by evaporating liquid. Since the propellant exists in liquid form in the can, it should be miscible with the payload or dissolved in the payload.

Representative gases include chlorofluorocarbons, hydrofluoroalkanes, hydrofluoroolefins, volatile hydrocarbons, typically propane, n-butane and isobutane, dimethyl ether (DME) and methyl ethyl ether. Nitrous oxide and carbon dioxide can also be used.

Manual pump sprays can be used as an alternative to a stored propellant.

In one embodiment, the formulation comprises about 0.1% to about 10% of a compound described herein, about 60% to about 90% of an alcohol or a buffered aqueous solution, and, optionally, about 0.1% to about 10% of a quaternary ammonium salt, such as benzalkonium chloride.

Optional Additional Components

In addition to one or more of the compounds described herein, any of the disinfectant/decontaminating formulations can optionally include one or more additional components. For example, the formulations can include a solvent, such as water, which may be buffered at a pH in which the compound does not rapidly decompose, and a further antimicrobial agent.

Representative further antimicrobial agents that can be present include, but are not limited to, quaternary ammonium salts, and anionic, cationic, non-ionic and zwitterionic detergents. Sodium dodecyl sulfate (SDS) is a preferred anionic detergent. Preferred non-ionic detergents include Tween, Triton and Brij. CHAPS is a preferred zwitterionic detergent. Such detergents are preferably used at a concentration of 0.1-5% w/v.

Quaternary ammonium salts, also known as quats, are positively charged polyatomic ions of the structure NR4+, with R being an alkyl group or an aryl group. Unlike ammonium ion (NH4+), and primary, secondary, or tertiary ammonium cations, the quaternary ammonium cations are permanently charged, independent of the pH of their solution.

Representative examples are benzalkonium chloride, benzethonium chloride, methylbenzethonium chloride, cetalkonium chloride, cetylpyridinium chloride, cetrimonium, cetrimide, dofanium chloride, tetraethylammonium bromide, didecyldimethylammonium chloride and domiphen bromide. These compounds are active against fungi, amoebas, and enveloped viruses, and are believed to act by disrupting the cell membrane or viral envelope. By themselves, these compounds are not particularly active against endospores, Mycobacterium tuberculosis and non-enveloped viruses, but on information and belief, can be active when the nitric oxide disrupts the cell walls of these microbes.

IV. Sterilization Chambers

When treating relatively large surfaces, particularly where the surfaces are on objects that are physically attached to a ceiling, wall or floor, the formulation is applied directly to the surfaces. However, in some embodiments, where there is an interest in simultaneously sanitizing a relatively large number of relatively small objects, for example, clothing, masks, such as surgical masks, medical waste, and the like, it can be preferred to contact the items with a formulation as described herein, and place the items in a sterilization chambers to allow the compounds to produce nitric oxide, and to allow the nitric oxide to contact the items, over time, and sterilize the items.

In some embodiments, the sterilization chamber is configured to receive the formulations and the items to be sterilized. Depending on the embodiment, the sterilization chamber can be stationary or movable. Whether stationary or movable, chambers can optionally be encased in a housing. In some aspects of this embodiment, the chamber comprises a tumbler-type chamber, which, in operation, is rotated around an axis. For example, in several embodiments, the chamber is rotated about a longitudinal axis, while in additional embodiments, it is rotated around a lateral axis or a vertical axis. In other aspects of this embodiment, the chamber may be movable, but need not rotate in any particular manner, for example, the chamber may simply oscillate, vibrate, shake, or otherwise move in a pattern of predetermined or random motions such the contents inside the sterilization chamber are likewise moved. In still other embodiments, the chamber is a fixed chamber (e.g., is not configured to move the contents placed inside the chamber).

The dimensions of the sterilization chamber are readily adjustable for any particular application or method of sterilizing. For example, the size and shape of the chamber can be adjusted for such embodiments wherein small medical devices are sterilized, while in other embodiments the chamber (or chambers) can be scaled up in size in order to sterilize larger items, large quantities of items, or a plurality of items to be sterilized simultaneously.

In some embodiments, the chamber may be a room to be decontaminated.

Thus, the sterilization chamber provided for herein can be any geometric shape and can vary in dimension depending on the intended use of the sterilization system. In one embodiment, the sterilization chambers have a volume ranging from about 10 L to about 10,000 L.

In some embodiments, the chamber further contains an internal container of custom size and shape based on the device or devices to be sterilized, disinfected, sanitized, and/or decontaminated inside the container. The chamber may include one or more inlet ports to allow a user to deliver the formulations to the chamber, and one or more outlet ports to vent the nitric oxide after the sterilization has been performed.

The chamber can be, for example, a countertop unit, an under-counter, under-cabinet, or wall mounted unit, or a freestanding system, which, for example, can be dimensioned to fit next to an existing countertop or cabinet, or to be portable (e.g. small enough to be moved from one site to another, or provided on a rolling cart or other mobile accessory).

In several embodiments, a plurality of items are sterilized or disinfected simultaneously. While in some embodiments, this involves simply placing the plurality of items within the chamber, in additional embodiments, specialized apparatuses are used. For example, in several embodiments, there is provided a specialized apparatus for disinfecting or sterilizing the exterior of a plurality of devices while simultaneously disinfecting or sterilizing a lumen of each of the devices.

In some embodiments, the chambers include racks or manifold to place endoscopes and similar devices, including other lumen containing devices. For example, an endoscope rack can be used with scopes related to the following fields, gastroenterology, endoscopic ultrasound scopes, pulmonology, ENT (ear, nose, and throat), speech, and urology. Additionally, in some embodiments scopes with working channels such as for biopsy or suction can be used with the endoscope rack. Advantageously, in several embodiments, the endoscope unit (a term that encompasses units to sterilize/disinfect other lumen containing devices) allows the endoscopes or other devices to remain within the unit (either in a bulk section or in individual sections, and maintain sterility while inside the unit. Moreover, the chamber may also serve as a storage area. In some embodiments, the endoscope or other device is placed within a separate compartment, optionally flexible, that creates a barrier between the device and the environment, such that the sterility/disinfected state will be maintained even after being removed from the chamber. Such embodiments advantageously allow the sterilized/disinfected devices to be stored and/or transported to a site of next use while maintaining the sterility/disinfected state.

The material that makes up the inner wall of the chamber can vary depending on the embodiment, but is ideally a non-reactive material, such that the inner wall of the chamber does not react with the nitric oxide that is produced. Suitable materials include, but are not limited to, glass, plastics, polymers, metals, stainless steel (e.g., 304 or 316 stainless), ABS plastic, aluminum, bronze, carbon graphite, cast iron, ceramic (AL203), ceramic magnet, CPVC, EPDM, epoxy, Hastelloy-C®, Kel-F®, LDPE, natural rubber, NORYL®, nylon, polycarbonate, polypropylene, PPS (Ryton®), PTFE (Teflon®), PVC, PVDF (Kynar®), silicone, Titanium, Tygon®, Viton® or combinations thereof. Moreover, in several embodiments, the inner wall of the chamber may be made of a first material while other layers, including insulating or other layers may be other materials.

V. Types of Microbes that can be Treated

The following are non-limiting examples of microbes, including bacteria, viruses, and fungi, that can be treated using the compounds described herein.

Types of Bacterial Infections that can be Treated

In one embodiment, the compounds are used to treat bacterial infections in the respiratory tract. Examples of pathogens that can be treated include Haemophilus influenzae, Pseudomonas aeruginosa, Staphylococcus aureus, Staphylococcus warneri, Staphylococcus lugdunensis, Staphylococcus epidermidis, Streptococcus milleri/anginous, Streptococcus pyogenes, non-tuberculosis Mycobacterium, Mycobacterium tuberculosis, Burkholderia spp., Achromobacter xylosoxidans, Pandoraeasputorum, Stenotrophomonas maltophilia, Alcaligenes xylosoxidans, Haemophilus pittmaniae, Serratia marcescens, Candida albicans, drug resistant Candida albicans, Candida glabrata, Candida krusei, Candida guilliermondii, Candida auris, Candida tropicalis, Aspergillus niger, Aspergillus terreus, Aspergillus fumigatus, Aspergillus flavus, Morganella morganii, Inquilinus limosus, Ralstonia mannitolilytica, Pandoraea apista, Pandoraea pnomenusa, Pandoraea sputorum, Bdellovibrio bacteriovorus, Bordetella bronchiseptica, Vampirovibrio chlorellavorus, Actinobacter baumanni, Cupriadidus metallidurans, Cupriavidus pauculus, Cupriavidus respiraculi, Delftia acidivordans, Exophilia dermatitidis, Herbaspirillum frisingense, Herbaspirillum seropedicae, Klebsiella pneumoniae, Pandoraea norimbergensis, Pandoraea pulmonicola, Pseudomonasmendocina, Pseudomonas pseudoalcaligenes, Pseudomonas putida, Pseudomonas stutzeri, Ralstonia insidiosa, Ralstonia pickettii, Neisseriagonorrhoeae, NDM-1 positive E. coli, Enterobacter cloaca, Vancomycin-resistant E. faecium, Vancomycin-resistant E. faecalis, E. faecium, E. faecalis, Clindamycin-resistant S. agalactiae, S. agalactiae, Bacteroides fragilis, Clostridium difficile, Streptococcus pneumonia, Moraxella catarrhalis, Haemophilus haemolyticus, Haemophilus parainfluenzae, Chlamydophilia pneumoniae, Mycoplasma pneumoniae, Atopobium, Sphingomonas, Saccharibacteria, Leptotrichia, Capnocytophaga, Oribacterium, Aquabacterium, Lachnoanaerobaculum, Campylobacter, Acinetobacter, Agrobacterium; Bordetella; Brevundimonas; Chryseobacterium; Delftia; Enterobacter; Klebsiella; Pandoraea; Pseudomonas; Ralstonia, and Prevotella.

Representative non-tuberculosis Mycobacterium include Mycobacterium abscessus, Mycobacterium avium, Mycobacterium intracellulare, Mycobacterium fortuitum, Mycobacterium gordonae, Mycobacterium kansasii, Mycobacterium avium complex, Mycobacterium marinum, Mycobacterium terrae and Mycobacterium cheloni.

Representative Burkholderia spp. Include Burkholderia cepacia, Burkholderia cepacia complex, Burkholderia multivorans, Burkholderia cenocepacia, Burkholderia stabilis, Burkholderia vietnamiensis, Burkholderia dolosa, Burkholderia ambifaria, Burkholderia anthina, Burkholderia pyrrocinia, Burkholderia gladioli, Burkholderia ubonensis, Burkholderia arboris, Burkholderia latens, Burkholderia lata, Burkholderia metallica, Burkholderia seminalis, Burkholderia contaminans, and Burkholderia diffusa.

In some embodiments, the bacteria are drug resistant, and in some aspects of these embodiments, the bacteria are multi-drug resistant. For example, the bacteria may be resistant to antibiotics such as amikacin, aztreonam, methicillin, vancomycin, nafcillin, gentamicin, ampicillin, chloramphenicol, doxycycline, colistin, delamanid, pretomanid, clofazimine, bedaquiline, and/or tobramycin.

The bacteria may develop resistance to these drugs, but cannot easily develop resistance to the approaches described herein.

Types of Viral Infections that can be Treated

RNA and DNA viruses that can be treated are summarized below.

RNA Viruses

Currently, there are 5 recognized orders and 47 families of RNA viruses, and there are also many unassigned species and genera. Related to but distinct from the RNA viruses are the viroids and the RNA satellite viruses.

There are several main taxa: levivirus and related viruses, picornaviruses, alphaviruses, flaviviruses, dsRNA viruses, and the -ve strand viruses (Wolf et al., “Origins and Evolution of the Global RNA Virome,” mBio, 9(6) (November 2018)).

Positive strand RNA viruses are the single largest group of RNA viruses, with 30 families. Of these, there are three recognized groups. The picorna group (Picornavirata) includes bymoviruses, comoviruses, nepoviruses, nodaviruses, picornaviruses, potyviruses, obemoviruses and a subset of luteoviruses (beet western yellows virus and potato leafroll virus). The flavi-like group (Flavivirata) includes carmoviruses, dianthoviruses, flaviviruses, pestiviruses, statoviruses, tombusviruses, single-stranded RNA bacteriophages, hepatitis C virus and a subset of luteoviruses (barley yellow dwarf virus). The alpha-like group (Rubivirata) includes alphaviruses, carlaviruses, furoviruses, hordeiviruses, potexviruses, rubiviruses, tobraviruses, tricornaviruses, tymoviruses, apple chlorotic leaf spot virus, beet yellows virus and hepatitis E virus.

A division of the alpha-like (Sindbis-like) supergroup has been proposed, with two proposed groups. The ‘altovirus’ group includes alphaviruses, furoviruses, hepatitis E virus, hordeiviruses, tobamoviruses, tobraviruses, tricornaviruses and rubiviruses, and the ‘typovirus’ group includes apple chlorotic leaf spot virus, carlaviruses, potexviruses and tymoviruses.

There are five groups of positive-stranded RNA viruses containing four, three, three, three, and one order(s), respectively. These fourteen orders contain 31 virus families (including 17 families of plant viruses) and 48 genera (including 30 genera of plant viruses). Alphaviruses and flaviviruses can be separated into two families, the Togaviridae and Flaviridae.

This analysis also suggests that the dsRNA viruses are not closely related to each other but instead belong to four additional classes, Birnaviridae, Cystoviridae, Partitiviridae, and Reoviridae, and one additional order (Totiviridae) of one of the classes of positive ssRNA viruses in the same subphylum as the positive-strand RNA viruses.

There are two large clades: One includes the families Caliciviridae, Flaviviridae, and Picornaviridae and a second that includes the families Alphatetraviridae, Birnaviridae, Cystoviridae, Nodaviridae, and Permutotretraviridae.

Satellite viruses include Albetovirus, Aumaivirus, Papanivirus, Virtovirus, and Sarthroviridae, which includes the genus Macronovirus.

Double-stranded RNA viruses (dsRNA viruses) include twelve families and a number of unassigned genera and species recognized in this group. The families include Amalgaviridae, Birnaviridae, Chrysoviridae, Cystoviridae, Endornaviridae, Hypoviridae, Megabirnaviridae, Partitiviridae, Picobirnaviridae, Reoviridae, which includes Rotavirus, Totiviridae, Quadriviridae. Botybirnavirus is one genus, and unassigned species include Botrytis porri RNA virus 1, Circulifer tenellus virus 1, Colletotrichum camelliae filamentous virus 1, Cucurbit yellows associated virus, Sclerotinia sclerotiorum debilitation-associated virus, and Spissistilus festinus virus 1.

Positive-sense ssRNA viruses (Positive-sense single-stranded RNA viruses) include three orders and 34 families, as well as a number of unclassified species and genera. The order Nidovirales includes the families Arteriviridae, Coronaviridae, which includes Coronaviruses, such as SARS-CoV and SARS-CoV-2, Mesoniviridae and Roniviridae. The order Picornavirales includes families Dicistroviridae, Iflaviridae, Marnaviridae, Picornaviridae, which includes Poliovirus, Rhinovirus (a common cold virus), and Hepatitis A virus, Secoviridae, which includes the subfamily Comovirinae, as well as the genus Bacillariornavirus and the species Kelp fly virus. The order Tymovirales includes the families Alphaflexiviridae, Betaflexiviridae, Gammaflexiviridae, and Tymoviridae. A number of families are not assigned to any of these orders, and these include Alphatetraviridae, Alvernaviridae, Astroviridae, Barnaviridae, Benyviridae, Botourmiaviridae, Bromoviridae, Caliciviridae, which includes the Norwalk virus (i.e., norovirus), Carmotetraviridae, Closteroviridae, Flaviviridae, which includes Yellow fever virus, West Nile virus, Hepatitis C virus, Dengue fever virus, and Zika virus, Fusariviridae, Hepeviridae, Hypoviridae, Leviviridae, Luteoviridae, which includes Barley yellow dwarf virus, Polycipiviridae, Narnaviridae, Nodaviridae, Permutotetraviridae, Potyviridae, Sarthroviridae, Statovirus, Togaviridae, which includes Rubella virus, Ross River virus, Sindbis virus, and Chikungunya virus, Tombusviridae, and Virgaviridae. Unassigned genuses include Blunervirus, Cilevirus, Higrevirus, Idaeovirus, Negevirus, Ourmiavirus, Polemovirus, Sinaivirus, and Sobemovirus. Unassigned species include Acyrthosiphon pisum virus, Bastrovirus, Blackford virus, Blueberry necrotic ring blotch virus, Cadicistrovirus, Chara australis virus, Extra small virus, Goji berry chlorosis virus, Harmonia axyridis virus 1, Hepelivirus, Jingmen tick virus, Le Blanc virus, Nedicistrovirus, Nesidiocoris tenuis virus 1, Niflavirus, Nylanderia fulva virus 1, Orsay virus, Osedax japonicus RNA virus 1, Picalivirus, Planarian secretory cell nidovirus, Plasmopara halstedii virus, Rosellinia necatrix fusarivirus 1, Santeuil virus. Secalivirus, Solenopsis invicta virus 3, and Wuhan large pig roundworm virus.

Satellite viruses include the family Sarthroviridae and the genuses Albetovirus, Aumaivirus, Papanivirus, Virtovirus, and the Chronic bee paralysis virus. Six classes, seven orders and twenty four families are currently recognised in this group. A number of unassigned species and genera are yet to be classified

Negative-sense ssRNA viruses (Negative-sense single-stranded RNA viruses) are, with the exception of the Hepatitis D virus, within a single phylum, Negarnaviricota, with two subphyla, Haploviricotina and Polyploviricotina, with four classes, Chunqiuviricetes, Milneviricetes, Monjiviricetes and Yunchangviricetes. The subphylum Polyploviricotina has two classes, Ellioviricetes and Insthoviricetes.

There are also a number of unassigned species and genera. The Phylum Negarnaviricota includes Subphylum Haploviricotina, Class Chunqiuviricetes, Order Muvirales, Family Qinviridae. The Class Milneviricetes includes Order Serpentovirales and Family Aspiviridae. The Class Monjiviricetes includes Order Jingchuvirales and Family Chuviridae. The order

Mononegavirales includes familes Bornaviridae, which includes the Borna disease virus, Filoviridae, which includes the Ebola virus and the Marburg virus, Mymonaviridae, Nyamiviridae, Paramyxoviridae, which includes Measles, Mumps, Nipah, Hendra, and NDV, Pneumoviridae, which RSV and Metapneumovirus, Rhabdoviridae, which Rabies, and Sunviridae, as well as genuses Anphevirus, Arlivirus, Chengtivirus, Crustavirus, and Wastrivirus. Class Yunchangviricetes includes order Goujianvirales and family Yueviridae.

Subphylum Polyploviricotina includes class Ellioviricetes, order Bunyavirales, and the families Arenaviridae, which includes Lassa virus, Cruliviridae, Feraviridae, Fimoviridae, Hantaviridae, Jonviridae, Nairoviridae, Peribunyaviridae, Phasmaviridae, Phenuiviridae, Tospoviridae, as well as genus Tilapineviridae.

Class Insthoviricetes includes order Articulavirales and family Amnoonviridae, which includes the Taastrup virus, and family Orthomyxoviridae, which includes Influenza viruses.

The genus Deltavirus includes the Hepatitis D virus.

Specific viruses include those associated with infection of mucosal surfaces of the respiratory tract, including Betacoronavirus (SARS-COV-2 and MERS-COV), rhinoviruses, influenza virus (including influenza A and B), parainfluenza). Generally, orthomyxoviruses and paramyxoviruses can be treated.

DNA Viruses

A DNA virus is a virus that has DNA as its genetic material and replicates using a DNA-dependent DNA polymerase. The nucleic acid is usually double-stranded DNA (dsDNA) but may also be single-stranded DNA (ssDNA). DNA viruses belong to either Group I or Group II of the Baltimore classification system for viruses. Single-stranded DNA is usually expanded to double-stranded in infected cells. Although Group VII viruses such as hepatitis B contain a DNA genome, they are not considered DNA viruses according to the Baltimore classification, but rather reverse transcribing viruses because they replicate through an RNA intermediate. Notable diseases like smallpox, herpes, and the chickenpox are caused by such DNA viruses.

Some have circular genomes (Baculoviridae, Papovaviridae and Polydnaviridae) while others have linear genomes (Adenoviridae, Herpesviridae and some phages). Some families have circularly permuted linear genomes (phage T4 and some Iridoviridae). Others have linear genomes with covalently closed ends (Poxviridae and Phycodnaviridae).

Fifteen families are enveloped, including all three families in the order Herpesvirales and the following families: Ascoviridae, Ampullaviridae, Asfarviridae, Baculoviridae, Fuselloviridae, Globuloviridae, Guttaviridae, Hytrosaviridae, Iridoviridae, Lipothrixviridae, Nimaviridae and Poxviridae.

Of these, species of the order Herpesvirales, which includes the familes Alloherpesviridae, Herpesviridae, which includes human herpesviruses and the Varicella Zoster, and the families Adenoviridae, which includes viruses which cause human adenovirus infection, and Malacoherpesviridae, infect vertebrates.

Asfarviridae, which includes African swine fever virus, Iridoviridae, Papillomaviridae, Polyomaviridae, which includes Simian virus 40, JC virus, and BK virus, and Poxviridae, which includes Cowpox virus and smallpox, infect vertebrates. Anelloviridae and Circoviridae also infect animals (mammals and birds respectively).

The family Smacoviridae includes a number of single-stranded DNA viruses isolated from the feces of various mammals, and there are 43 species in this family, which includes six genera, namely, Bovismacovirus, Cosmacovirus, Dragsmacovirus, Drosmacovirus, Huchismacovirus and Porprismacovirus. Circo-like virus Brazil hs1 and hs2 have also been isolated from human feces. An unrelated group of ssDNA viruses includes the species bovine stool associated circular virus and chimpanzee stool associated circular virus.

Animal viruses include parvovirus-like viruses, which have linear single-stranded DNA genomes, but unlike the parvoviruses, the genome is bipartate. This group includes Hepatopancreatic parvo-like virus and Lymphoidal parvo-like virus. Parvoviruses have frequently invaded the germ lines of diverse animal species including mammals.

The human respiratory-associated PSCV-5-like virus has been isolated from the respiratory tract.

Representative viruses associated with lung infections that can be treated using the methods described herein include Coronavirus, Picornavirus, influenza virus (including influenza A and B), common cold, respiratory syncytial virus (RSV), adenovirus, parainfluenza, rhinoviruses, and SARS). Generally, orthomyxoviruses and paramyxoviruses can be treated.

In addition to viruses associated with respiratory infections, causing bronchitis, sinusitis, and/or pneumonia, the human papilloma virus (HPV) is associated with certain throat cancers.

Types of Fungal Infections that can be Treated

Representative fungal infections that can be treated include Candida albicans, drug resistant Candida albicans, Candida glabrata, Candida krusei, Candida guilliermondii, Candida auris, Candida tropicalis, Aspergillus niger, Aspergillus terreus, Aspergillus fumigatus, and/or Aspergillus flavus.

VI. Methods of Treating Surfaces

The compounds disclosed herein can be used in sanitizing/disinfecting/decontaminating/sterilizing applications. Depending on the surface to be treated, the compounds and/or formulations discussed herein can be administered by spray, wipe, aerosol, foam, drip, mop and the like.

In some embodiments, methods for cleaning, disinfecting, decontaminating and/or sterilizing a substrate comprise applying a compound as described herein and/or formulations including the compound to the substrate. Suitable substrates include, but are not limited to, chicken coops, farming equipment and containment areas, industrial food preparation equipment, hard surfaces susceptible to the formation of fungal or biofilms, such as industrial equipment, any hard surface found in the home, or an industrial, medical or institutional setting. In another embodiment, the hard surface is a floor, wall, countertop, appliance, or fixture. In still a further embodiment, the substrates include, but are not limited to, those located in dairies, homes, health care facilities, swimming pools, canneries, food processing plants, restaurants, hospitals, institutions, and industry, including secondary oil recovery. Specific areas targeted for application include chicken coops or hard surfaces in the home such as kitchen countertops, cabinets, appliances, waste cans, laundry areas, garbage pails, bathroom fixtures, toilets, water tanks, faucets, mirrors, vanities, tubs, and showers.

The compositions can also be used to sanitize floors, walls, furniture, mirrors, toilet fixtures, windows, and wood surfaces, such as fence rails, porch rails, decks, roofing, siding, window frames, and door frames. Compositions containing compounds according to the present disclosure may be combined with other known antimicrobial or disinfectant compounds, such as quaternary ammonium salts. Disinfecting compositions as disclosed herein are particularly well suited for application on indirect food contact surfaces, such as cutting boards, utensils, containers, dishes, wash basins, appliances, and countertops. The compositions can be used to sanitize diary plant equipment, milking machines, milk pails, tank trucks, and the like. Areas in hospitals would include beds, gurneys, tables, canisters, toilets, waste cans, stands, cabinets, shower stalls, floors, walls or any other non-porous surface. The amount of the compound disclosed herein and/or formulations including the compound used to treat a substrate is a biocidal effective amount, i.e. that amount to sanitize or disinfect the substrate. The biocidal effective amount will depend upon the use intended and can be determined by one of ordinary skill in the art in light of the present detailed disclosure.

Typically, the cleaning/disinfectant/decontaminating/sterilizing compositions can either be supplied in a dilutable concentrated form or in a ready-to-use form. In one embodiment, the composition is provided as a powder which is dissolved in water or similar solution prior to use.

Treatment of Crop Loci, Trees, Plants, and Individual Fruits and Vegetables

Fresh fruits are prone to fungal contamination in the field, during harvest, transport, marketing, and with the consumer, and the compounds and formulations described herein can be used to treat, prevent, or reduce the concentration of this fungal contamination.

It is important to decontaminate fungal contaminants in fresh fruits, because some molds can grow and produce mycotoxins on these commodities, while certain yeasts and molds can cause infections or allergies.

A variety of fruits are or potentially are affected, including grapes, strawberries, blueberries, raspberries, blackberries, and various citrus fruits. Where the surface of these fruits is treated, the fruits can last longer before becoming spoiled.

Tobacco, hemp and marijuana are also crops that are prone to fungal infection, particularly during the drying process.

Grains in silos are often infected with bacteria and/or fungi. The grain can be treated, for example, by applying one or more of the formulations described herein to the grain in the silo, and allowing the formulations to permeate the grain, release NO, and sanitize, disinfect, decontaminate and/or sterilize the grain.

Certain crops are known to be affected by bacterial infection. For example, peppers are known to suffer from peppery leaf spot, caused by Pseudomonas syringae pv. maculicola (brassicas), lettuce is known to suffer from varnish spot, caused by Pseudomonas spp., and corky root, caused by Rhizomonas suberifaciens (lettuce). Cucurbits are known to suffer from angular leaf spot, caused by P. syringae pv. Lachrymans, and tomatoes are known to suffer from bacterial pith necrosis, caused by Pseudomonas corrugata and other bacteria. The bacterial infections can be treated, prevented, and/or their severity reduced by applying one or more of the formulations described herein to the crops.

In the field, the formulations can be generally applied to a crop locus, or to an individual tree or plant in need of treatment thereof, in an effective amount to effect such treatment. The compositions can be applied by conventional application techniques. These techniques include, but are not limited to, root application, leaf application, crop dusting, or spray application.

The most common molds affecting these commodities are Botrytis cinerea, Rhizopus (in strawberries), Alternaria, Penicillium, Cladosporium and Fusarium followed by yeasts, Trichoderma and Aureobasidium. The most common fungi spoiling grapes are Alternaria, B. cinerea and Cladosporium. The most common fungi in citrus fruits are Alternaria, Cladosporium, Penicillium, Fusarium and yeasts. Less common are Trichoderma, Geotrichum and Rhizopus. All of these can be treated using the compositions described herein.

In one embodiment, the formulations include at least one permeabilizing agent, to assist with having the compound(s) penetrate the skin of the fruit or vegetable being treated. The formulations can be supplied as a solid or a liquid, including thixotropic droplets. In one embodiment, the solid is a granule or a powder. In one embodiment, the liquid can be a solution, dispersion or suspension in water or other carrier. In one embodiment, these products are generally diluted into water before being sprayed onto the field from either an airplane or ground application equipment. In one embodiment, solid formulations can be combined with wetting agents or surfactants for better deposition or application on the plants surface or substitute and better uptake by the plant. In a preferred embodiment, solid formulations are used due to higher loading levels.

In one embodiment, the at least one permeabilizing agent functions by improving the permeability of the cell walls of the plant or plants to which it is applied. By improving the permeability of the cell walls, the compounds described herein have better penetration into the plant. Also, in the case of bacteria, fungi, viruses and acaricides, the at least one permeabilizing agent can improve the permeability of the cell walls of these compounds and improve the passage of the active compounds into these biological entities, thus improving the efficacy of these compounds. Increased efficacy can translate into lower effective dosages, which is a tremendous benefit, particularly with governmental regulations constantly lowering the permitted dosages of compounds applied to crops. Further, as the active compound penetrates the plant cell wall, the activity of the formulations is more persistent than formulations applied topically to plants that do not effectively penetrate the plant cell wall.

In one embodiment, the at least one permeabilizing agent is at least one chelating agent. A chelate, sometimes referred to as a sequestrant, a complex ion, or a coordination compound, can be an organic compound that combines with a metal ion to form a complex in which the donor atoms are connected to each other as well as to the metal in one embodiment. Thus, in one embodiment, the metal becomes part of a heterocyclic ring. In one embodiment, donor atoms in the chelate complex may be tied together with additional chelate rings so that each chelating agent may contain two, three, four, five, six or even more donor groups. In a preferred embodiment, the chelating agent is ethylenediaminetetraacetic acid (EDTA), diethylenetriaminepentaacetic acid (DTPA), (hydroxyethyl)ethylenediaminetriacetic acid (HEDTA), nitrilotriacetic acid (NTA), oxalic acid, citric acid, or a combination thereof. A particularly preferred chelate is EDTA, which has two amine donor groups and four carboxyl donor groups. In one embodiment, EDTA supplies the complete requirements for the coordination sphere of many metals with a single molecule where it might take three molecules of ethylenediamine to meet the same-requirements. A chelating agent that supplies two donor electrons to the metal is said to be bidentate. Similarly ter-, quadri, quinqui-, and sexadentate donors, bind the metal in 3, 4, 5, and 6 positions, respectively. Hence, EDTA is sexadentate and ethylenediamine is bidentate, for example.

In one embodiment, the adjuvant composition comprises EDTA tetrasodium in an amount of between 0.1% and 40% by weight. In a particularly preferred embodiment, the adjuvant composition comprises EDTA tetrasodium in an amount of about 20%.

Additional non-limiting examples of these chelating compounds include, but are not limited to the sugars, acids and salts of maleic acid, malonic acid, tartaric acid, citric acid, glycine, lactic acid, malic acid, succinic acid, oxalic acid, dextrose, ethylenediaminetetraacetic acid (EDTA), tris(hydroxymethyl)aminomethane, lactose, mannitol, glutaric acid, malic acid, succinic acid, glycerol, humic acid, fulvic acid, sorbic acid, sorbose, ethylene diamine, 1,2 diaminocyclohexane, trimethylenediamine, tetramethylenediamine, 1,2 diaminopropane, diethylenetriamine, triethylenetetramine, triaminodiethylamine, N-hydroxyethylethylenediamine, sodium polyphosphate, potassium polyphosphate, ammonium polyphosphate, sodium hexametaphosphate and mixtures thereof. In one embodiment, the at least one chelating agent used in the present compositions can be 100% of any particular chelator, or a combination of chelator in any ratio. In one embodiment, a combination or mixture of chelating compounds may dissolve faster than a single compound. In a preferred embodiment, 100% oxalic acid, 100% citric acid, 100% EDTA, and combinations of these are used. In a particularly preferred embodiment, the adjuvant composition comprises between 0.1% and 20% of citric acid based on total formulation weight.

In several embodiments, the compounds allow the efficient reduction in viability and/or eradication of microbes (e.g., prokaryotic cells, bacteria, protozoa, fungi, algae, amoebas, slime molds, etc.) on the treated surfaces. In particular, such compounds are effective against such microbes that have developed at least some degree of drug resistance to traditional antimicrobial or antiviral therapies alone or in combination with other known antimicrobial or antiviral therapies.

Unlike conventional antimicrobial treatments, NO, an endogenously produced free radical, eradicates bacteria using a variety of mechanisms, including, but not limited to, lipid peroxidation, nitrosation of membrane proteins, and DNA damage via reactive oxygen/nitrogen species (e.g., peroxynitrite, dinitrogen trioxide). Multiple biocidal mechanisms allow NO to significantly diminish the risk of fostering microbial resistance.

NO is also a potent antibacterial agent that acts on bacteria via nitrosative and/or oxidative stress. NO is a broad-spectrum antibacterial agent and in some embodiments, the compounds described herein deliver NO, and are therefore capable of eradicating both bacteria and biofilms, potentially through the formation of reactive NO byproducts (e.g., peroxynitrite and dinitrogen trioxide) that cause oxidative and nitrosative damage to microbial DNA and/or membrane structures. Advantageously, the wide range of mechanisms by which NO exerts its antibacterial effects reduces the risk that bacteria will develop resistance. Thus, NO-releasing compounds described herein are useful for disinfecting/decontaminating surfaces contaminated with bacteria. The antibacterial efficacy of NO-releasing materials is dependent on both NO payloads and associated release kinetics. In some instances, high NO total is an important parameter to effectively evaluate storage capability of suitable compounds. However, NO release that is too fast, and high NO storage, can result in undesired toxicity to mammalian cells. Therefore, challenges exist in preparing biocompatible NO-releasing materials with high NO storage and low cytotoxicity, and such challenges, among others, are addressed according to several embodiments disclosed herein.

Treatment of Surfaces Contaminated with Drug-Resistant Bacteria

In several embodiments, the surface has a microbial load to be reduced and/or eliminated that comprises drug-resistant bacteria. In several embodiments, the drug-resistant bacteria comprise carbapenem-resistant Enterobacteriaceae. In several embodiments, the drug-resistant bacteria comprise Methicillin-resistant Staphylococcus aureus. In several embodiments, the microbe comprises human immunodeficiency virus, herpes simplex virus, papilloma virus, parainfluenza virus, influenza, hepatitis, Coxsackie Virus, herpes zoster, measles, mumps, rubella, rabies, pneumonia, (hemorrhagic viral fevers, H1N1, and the like), prions, parasites, fungi, mold, yeast and bacteria (both gram-positive and gram-negative) including, among others, Candida albicans, Aspergillus niger, Escherichia coli (E. coli), Pseudomonas aeruginosa (P. aeruginosa), and Staphylococcus aureus (S. aureus), Group A streptococci, S. pneumoniae, Mycobacterium tuberculosis, Campylobacter jejuni, Salmonella, Shigella, P. gingivalis, A. actinomycetemcomitans, A. viscosus, and/or S. mutans and a variety of drug resistant bacteria. The terms microorganism and microbe shall be used interchangeably. Microbes can include wild-type, genetically-engineered or modified organisms.

Application of the Compounds Described Herein to Treat or Prevent Coronavirus Infection

The compounds described herein can be used to treat surfaces contaminated with SARS-CoV2, the causative agent for Covid-19. The nitric oxide generated as the compounds degrade can be effective at disinfecting and/or decontamining the surface from this virus.

Treatment of Hospitals and Medical/Dental/Chiropractic/Optometric Offices

Hospitals and medical, dental, chiropractic, and optometric offices are frequented by patients, many of whom have infectious diseases, such as a common cold, a flu, and the like, and to other nocosomial infections, such as norovirus. It is important to decontaminate surfaces to minimize the threat of transmission of these diseases. In addition to treating the surfaces of medical devices, as discussed elsewhere herein, countertops, floors, walls, and ceilings, and other exposed surfaces in hospitals and offices where medical treatments are performed can minimize the threat of transmission. Accordingly, in one embodiment, one or more compounds disclosed herein, and formulations including the one or more compounds, are applied to surfaces in hospitals and offices where medical treatments are performed, in a concentration sufficient to sanitize, disinfect, decontaminate and/or sterilize the surfaces from any microbe that might be present. In one embodiment, combinations of the compounds described herein and a quaternary ammonium salt are used.

Treatment of Athletic Facilities

Ringworm is one of the most pervasive infections in athletic facilities, particularly on wrestling mats, and in showers and floors in the locker rooms. Application of the formulations described herein to surfaces that have been exposed, may have been exposed, or may be exposed to ringworm or other microbes common to athletes, can sanitize, disinfect, decontaminate and/or sterilize the surfaces, thereby reducing the risk of infection.

Treatment of Residential Areas and Office Buildings

Residential areas and office buildings are frequented by individuals, some of whom have communicable diseases. These areas/buildings include a variety of surfaces in which people come into regular contact, including floors, countertops, walls, appliances, computers, and the like. These surfaces can be sanitized, disinfected, decontaminated, and/or sterilized using the formulations and methods disclosed herein.

The present invention is further defined in the following Examples. It should be understood that these Examples, while indicating preferred embodiments of the invention, are given by way of illustration only.

EXAMPLES Example 1: Synthesis and Characterization of the Compound of Formula III

This example pertains to the synthesis and identification of one embodiment of the compound of Formula III. This embodiment has the following features, advantages, and/or uses.

In several embodiments, this molecule has antibacterial properties with the NO-releasing material acting as an antibacterial agent. Compounds such as those disclosed were found to be the product of certain high-pressure nitric oxide synthetic strategies. In this context, the compounds can form in trace to major components of the reaction, depending on reaction conditions, such as NO pressure, base content, temperature and, reactant content.

The methane trisdiazeniumdiolate, sodium salt, was prepared according to the following procedure in Table 1:

TABLE 1 Reaction parameters used to synthesize the compound of Formula III Substance Mass/vol mol Eq. NaOH 6 g 0.15 4 MeOH 150 mL (40 mg/mL) Acetone   2.78 mL 0.0325 1
    • NaOH was dissolved in MeOH
    • Acetone was added to the stirring solution.
    • Mixture was transferred to a 400 mL Parr reactor, which was equipped with stir bar.
    • Parr reactor was sealed.
    • 3×N3 Purge with 100 Psi of N2 while stirring.
    • 2.5 bar of NO charged into Parr vessel and allowed to stir at room temperature for 4 days.
      The solution was cloudy with an off-white to slightly yellow precipitate.

The reaction solution was filtered via vacuum filtration using 110 mm GF/F filter paper.

    • A 15 mL aliquot of the filtrate was recovered and saved for testing if needed.

The solids collected during filtration were washed with 150 ml of MeOH.

The solids were dried overnight under vacuum at room temperature.

    • Recovery: 5.0 g+4.2 g=9.2 g off-white powder
    • NCT-19-168; 1HNMR=7.4, 5.8.

The synthesized compound was isolated and tested to confirm identity. FTIR, HPLC, UV-Vis spectroscopy, 1H NMR and 13C NMR analysis were used to support the conclusions that the synthesis yielded the compound of Formula III.

Referring now to FIG. 1, the FTIR spectrum is shown of the compound of Formula III according to the present disclosure. Referring now to FIG. 2, an ion exchange chromatogram is shown of the compound of Formula III with a retention time of 11 minutes (λ=252 nm). The UV absorbance spectrum of the analyte retained for approximately 6 minutes, the UV absorbance spectrum of the analyte retained for approximately 10 minutes, and the UV absorbance spectrum of the analyte retained for approximately 11 minutes are shown in the insets.

FIG. 3 shows the 1H NMR of the compound of Formula III. The peak at 7.5 ppm is assigned to the single proton on the compound of Formula III. FIG. 4 shows the 13C NMR of the compound of Formula III. The peak at 100 ppm is assigned to the single carbon on the compound of Formula III. FIG. 5 shows 2D NMR of the compound of Formula III. FIG. 6 shows A) HPLC chromatograms (IEX-UV) with the top chromatogram (red) showing the separation of components prior to acid degradation of the compound of Formula III (referenced as MD3), the middle chromatogram (blue) showing the separation of components after 5 h of acid degradation, and the bottom chromatogram (black) showing the separation of components after 24 h of acid degradation, B) the 1H NMR spectrum of the acid degraded components, C) a table of NOA Totals for the compound of Formula III before and after acid degradation. FIG. 7 is a 1H NMR spectrum and FIG. 8 is 13C NMR spectrum of a composition comprising the products of the compound of Formula III after degraded at neutral pH at room temperature.

Analytical results for the compound of Formula III are summarized in Table 2.

TABLE 2 Analytical test results compiled for the compound of Formula I. Test Results UV λmax = 264 nm IR O—H and/or N—H (3436 cm−1), Sharp C—H (3025 cm−1), C═N (1649 cm−1), N—O (1354, 1232 cm−1) H NMR Singlet δ = 7.6 ppm, Singlet δ = 5.9 ppm C13, 2D NMR 1 peak at δ = 100 ppm, correlated to 7.6 ppm C, N, H Analysis C, 4.14%; N, 27.85%; H, 1.42% XPS C, O, N, Na confirmed

Antimicrobial Activity

The NO release of the compound of Formula III at pH 7.4 is shown in FIG. 9. The release profile was measured by chemiluminescence, showing 6.7 μmol NO/mg material releasing with a T1/2 of approximately 3.75 h. In keeping with the described degradation pathway, 2 moles of NO are released per 1 mole of Formula III which converts to a theoretical yield of 7.6 μmol NO/mg.

The antibacterial efficacy of the compound of Formula III derived according to the synthesis example against various Pseudomonas bacteria strains are set forth in Tables 3-5. The results from testing varying amounts of the compound mixed with a representative excipient β-cyclodextrin) is set forth in Table 6.

TABLE 3 Antimicrobial data for coumpound of Formula III. MIC MBC Batch Species Strain (mg/ml) (mg/ml) NCT-19-096 Pseudomonas ATCC 0.049 0.049 aeruginosa 19143 NCT-19-096 Pseudomonas ATCC 0.049 0.098 re-slurry aeruginosa 19143 NCT-19-168 Pseudomonas ATCC 0.0625 0.0625 aeruginosa 19143 NCT-19-168 Pseudomonas ATCC 0.5 1 (acid degraded) aeruginosa 19143

TABLE 4 Antimicrobial data for coumpound of Formula III. MBEC- aerobic Batch Species Strain (mg/ml) NCT-19-096 Pseudomonas PAK 0.391 re-slurry aeruginosa

TABLE 5 Antimicrobial data for compound of Formula III against P. aeruginosa. MIC MBC MBEC Batch (mg/ml) (mg/ml) (mg/mL) Reaction Precipitate 0.049 0.049 0.391 Reaction Precipitate 0.391 0.781 1.56 BIOC11 mixture

TABLE 6 The relationship between increasing excipient concentrations on the antimicrobial efficacy of the combined composition of β-cyclodextrin and compound of Formula III (referenced in table as MTDD). MIC against % MTDD NO-loading P. aeruginosa (NCT-19-096) % β-cyclodextrin (μmol/mg) (mg/ml) 100 0 6.7 0.049 90 10 6.0 0.049 80 20 5.4 0.098 60 40 4.0 0.098 40 60 2.7 0.195 20 80 1.3 0.391 10 90 0.7 0.781 0 100 0.0 >6.25

Table 6. The relationship between increasing excipient concentrations on the antimicrobial efficacy of the combined composition of p-cyclodextrin and compound of Formula III (referenced in table as MTDD).

Example 2: In Vitro Antimicrobial Activity of the Compound of Formula III Against P. aeruginosa

Minimum Inhibitory Concentration/Minimum Bactericidal Concentration (MIC/MBC) assays were performed using the CLSI method to evaluate efficacy of the compound of Formula III against several lab and clinical isolates of P. aeruginosa.

As used herein, the Minimum Bactericidal Concentration (MBC) is defined as the lowest concentration of antibiotic that kills 99.9% of the bacteria, and the Minimum Inhibitory Concentration (MIC) is defined as the lowest concentration of an antimicrobial ingredient or agent that is bacteriostatic (prevents the visible growth of bacteria).

MICs are used to evaluate the antimicrobial efficacy of various compounds by measuring the effect of decreasing concentrations of antibiotic/antiseptic over a defined period in terms of inhibition of microbial population growth. These evaluations can be quite useful during the R&D phase of a product to determine appropriate concentrations required in the final product.

Various concentrations of the compounds are inoculated with cultured bacteria, and the results are measured using agar dilution or broth dilution (macro or micro) to determine at what level the MIC endpoint is established.

The Minimum Bactericidal Concentration (MBC) can be determined from the broth dilution of MIC tests by subculturing to agar plates that do not contain the test agent and incubating for 24 hours. The MBC is identified by determining the lowest concentration of antibacterial agent that reduces the viability of the initial bacterial inoculum by a pre-determined reduction such as >99.9%. The MBC is complementary to the MIC; whereas the MIC test demonstrates the lowest level of antimicrobial agent that greatly inhibits growth, the MBC demonstrates the lowest level of antimicrobial agent resulting in microbial death. In other words, if a MIC shows inhibition, plating the bacteria onto agar might still result in organism proliferation because the antimicrobial did not cause death. Antibacterial agents are usually regarded as bactericidal if the MBC is no more than four times the MIC.

The Clinical and Laboratory Standards Institute (CLSI) has established protocols and standards for establishing MIC and MBC in products. A common methodology utilized for MIC is CLSI M07-A9, Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically. CLSI also has developed methods specific for the yeasts, filamentous fungi, and anaerobic bacteria. For MBC determination, CLSI M26-A, Methods for Determining Bactericidal Activity of Antimicrobial Agents, is an accepted industry standard.

As shown in Table 7, the compound of Formula III (referenced as MID3 in the table) was effective against all 9 strains tested, with 0.125 mg/ml resulting in a 3-log reduction of bacteria (MBC).

TABLE 7 MIC/MBC results for the compound of Formula III against strains of P. aeruginosa MD3 (mg/ml) VT-20-110 P. aeruginosa MIC, MIC, MBC, MBC, Phenotype rep 1 rep 2 rep 1 rep 2 PAK WT 0.125 0.0625 0.125 0.125 MDR, CarbR, 0.063 0.125 0.125 0.125 TobraR. ATCC, obligate 0.063 0.063 0.125 0.125 aerobe Not MDR, CarbS, 0.063 0.063 0.125 0.125 TobraS. Mucoid XDR, CarbR, 0.063 0.063 0.125 0.125 TobraI. XDR, CarbR, 0.063 0.063 0.125 0.125 TobraR. XDR, TobraR, 0.063 0.063 0.125 0.125 CarbR. XDR, TobraR, 0.063 0.063 0.125 0.125 CarbR. XDR, TobraR, 0.125 0.063 0.125 0.125 CarbR.

MD3 is therefore effective against P. aeruginosa, a major pathogen affecting cystic fibrosis patients.

Example 3: Efficacy of the Compound of Formula III In Vitro Against Non-Tuberculosis Mycobacterium, Mycobacterium abscessus

MIC/MBC assays were performed using the CLSI method described above to evaluate the efficacy of the compound of Formula III against several lab and clinical isolates of Mycobacterium abscessus, a common NTM species. As shown below in Table 8, the compound of Formula III was effective against all 9 strains tested, with 1 mg/ml resulting in a 3-log reduction of bacterial survival (MBC).

TABLE 8 Formula III is bactericidal against NTM M. abscessus N0010; clinical N0016; clinical N0017; clinical N0018; clinical N0019; clinical N0020; clinical isolate isolate isolate isolate isolate isolate N0046; Ordway Replicate Batch MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC MIC MB MIC MBC 1 VT-20-110 0.25 1 0.25 1 0.5 2 0.25 1 0.25 1 0.25 1 0.5 1 2 VT-20-110 0.25 1 0.25 1 0.25 1 0.25 1 0.25 1 0.25 1 0.25 1

The data show that the compound has in vitro efficacy beyond just P. aeruginosa.

Example 4: Formula III is a Broad-Spectrum Antimicrobial

MIC/MBC assays were performed using the CLSI method to evaluate efficacy of the compound of Formula III against additional clinical isolates of a number of pathogens. The results are shown below in Table 9. The compound of Formula III is referenced as MD3 in the table.

TABLE 9 MIC/MBC results for the compound of Formula III for a range of gram-negative and gram-positive bacteria. MD3 (mg/ml) VT-20-110 MIC, MIC, MBC, MBC, Type Species Strain ID Phenotype rep 1 rep 2 rep 1 rep 2 Gram- Acinetobacter baumannii N0075 Carb-R, XDR 0.063 0.063 0.25 0.25 negative Burkholderia conocepacia N0034 Clinical isolate 0.063 0.063 0.125 0.125 Escherichia coli N0077 Carb-R; 3rd-gen ceph-R; ESBL+, MDR 0.125 0.125 0.25 0.25 Haemophilus influenzae N0097 Chloramphenicol-R, Tet-R, and Amp-R 0.125 0.125 0.125 0.25 Mycobacterium abscessus N0019 Clinical isolate 0.25 0.25 1 1 Pseudomonas aeruginosa N0049 PAK WT 0.125 0.0625 0.125 0.125 Gram- Staphylococcus aureus(MRSA) N0040 Clinical isolate; MDR, MRSA, Vanco-S 0.125 0.125 0.25 0.25 positive Staphylococcus aureus(MSSA) N0007 Wichita; ATCC 29213 0.125 0.125 0.125 0.125 Streptococcus pyogenes N0098 Erythromycin-resistant 0.03125 0.03125 0.0625 0.125

The data show that Formula III has broad-spectrum anti-microbial activity.

Example 5: Efficacy In Vitro Against a P. aeruginosa Biofilm

P. aeruginosa biofilms grown on peg lids of a 96-well plate were exposed to the compound of Formula III for 18-24 h, then biofilms were isolated and surviving bacteria enumerated. The data showed that the compound of Formula III eradicated P. aeruginosa biofilms. A concentration of 0.391 mg/ml MD3 was sufficient to eradicate bacteria biofilms (>3-log reduction).

Example 6: Efficacy In Vitro Against P. aeruginosa Phenotypes Under Aerobic and Anaerobic Growth Conditions

As an extension to the work performed in Example 2, the anti-microbial activity of the compound of Formula III was compared for 21 strains of P. aeruginosa under aerobic and anaerobic growth conditions. The results are presented in FIG. 10. Out of the 21 strains tested, the activity of the compound of Formula III was identical for 18 of the strains. Two of the strains could not be compared because those two bacteria strains could not be grown under anaerobic conditions. Only one strain showed a difference in susceptibility but the difference was minimal. Therefore the compound of Formula III shows very consistent anti-microbial activity to P. aeruginosa under both aerobic and anaerobic growth conditions.

Example 7: Efficacy In Vitro—Time Kill Assay

The bactericidal activity of the compound of Formula III was evaluated over time for P. aeruginosa. P. aeruginosa cultures were grown at 37° C. in either PBS or cation-adjusted Mueller Hinton broth (CAMHB) with varying concentrations of the compound of Formula III, and bacteria were quantified at various timepoints during growth. In PBS, bacteria can survive but have no nutrients to replicate. In CAMHB, bacteria have the nutrients needed to grow and do replicate to high titers with time.

As shown in FIGS. 11A-B, in both PBS and CAMHB, the compound of Formula III (referenced as MD3 in the graphs) eradicated bacteria in a dose- and time-dependent manner. Within the PBS buffer system, the bacteria titers decrease to below the limit of detection for all concentrations of MD3 used but the time required to kill the bacteria increased with decreasing dose. The same trend is observed in the CAMHB system, but because bacteria grow in CAMHB, the lowest two doses of MD3 that were evaluated were insufficient to kill all the bacteria. The bacteria that survived out to 8 hours rapidly reproduced to establish titers close to the untreated control group at 24 hours. All doses greater than or equal to 0.125 mg/ml were effective enough to kill all bacteria within 8 hour; however, the smaller the dose the longer it took to fully kill the bacteria.

Example 8: Nitric Oxide is Key to the Antimicrobial Activity of the Compound of Formula III

The activity of the compound of Formula III against P. aeruginosa and S. aureus was compared with that of its degradation products (referenced as MD3 NO-lib in the table) using the CSLI methods described above. The main degradation product of the compound of Formula III is N-hydroxyl formamide under physiological conditions. In P. aeruginosa, the MBC of the compound of Formula III was 64× lower than the MBC of the MD3 NO-lib, and in S. aureus, the MBC of MD3 was 16× lower than the MBC of the MD3 OG-lib. The data are shown below in Table 10. The compound of Formula III is referenced as MD3 in the table.

TABLE 10 Comparison of MIC/MBC results for the compound of Formula III and its degraded by-products. MD3 MD3 (NO-liberated) VT-20-110 VT-20-110 MIC MIC MBC MBC MIC MIC MBC MBC Species Strain rep 1 rep 2 rep 1 rep 2 rep 1 rep 2 rep 1 rep 2 P. aeruginosa N0049, PAK, lab strain 0.125 0.0625 0.125 0.125 2 2 8 8 S. aureus N0040, MRSA, clinical isolate 0.125 0.125 0.25 0.25 2 2 4 4

Since the compound of Formula III functions by releasing NO, and NO-lib does not, it is reasonable to conclude that NO is a significant driver of Formula III's bactericidal activity in vitro.

In support of the key role that NO plays in the anti-microbial activity of the compound of Formula III, a time-kill study was conducted at three different pH conditions: 6.4, 7.6, and 8.4. The rate of NO release is dependent on pH. The lower the pH, the faster the compound of Formula III degrades and releases NO. In turn, the more NO that is released the faster the bacteria are killed. FIG. 12 shows a graph of the time kill assay results which show that bacterial kill rate it dependent on pH. The untreated results are an average of the untreated samples ran at each pH and show the differences in kill rates is not due to pH itself but due to the differing amounts of NO that are released at each of the time points at the corresponding pH conditions.

Example 9: Animal Toxicology Studies with the Compounds of Formula III

Animal studies were performed using severe, combined, immunodeficient (SCID) mice, using the study design outline in Table 11. No adverse effects were observed even for maximum dose group 6 (100 mg/Kg). All mice remained bright, alert, and responsive throughout the duration of the study.

TABLE 11 Maximum tolerated dose study design carried out in mice. Experiment Maximum tolerated dose (MTD) Test system 6- to 8-week-old female SCID mice (6 total) Dosing Intratracheal, once daily for 3 consecutive days Experimental arms 1. Buffer only 2. 1 mg/kg MD3 3. 3.16 mg/kg MD3 4. 10 mg/kg MD3 5. 31.6 mg/kg MD3 6 100 mg/kg MD3 Readouts Daily clinical scores, adverse effects, animal weights, survival

Example 12: Comparison of the Efficacy of MD3 and MD2

In some cases, the synthesis of MD3 also generated an impurity, referred to herein as MD2, or methane bis-diazeniumdiolate, which has the following formula:

The activity of mixtures of these compounds was evaluated against P. aeruginosa (PAK), with the goal of determining how the percentage of the compound of Formula III (referenced as MD3 in the table) relative to MD2 in different samples affected the activity of the mixture against P. aeruginosa.

TABLE 12 MIC/MBC results for the compound of Formula III of varying purity. MIC MBC TA: Strain: (mg/ml) (mg/ml) Run #1, vial 7 (97% MD3) N0049 0.0625 0.125 Run #3, vial 7 (50% MD3) N0049 0.125 0.25 Run #3, vial 4 (5% MD3) N0049 0.25 0.5

The results, shown in Table 12, demonstrate that the higher the percentage of the compound of Formula III in the sample relative to the percentage of MD2, the better the activity is against PAK. It should be noted that when samples have degraded under simulated physiological conditions, that MD2 does not degrade to any significant degree. Nor does it grow in as a degradation product of the compound of Formula III. Therefore, MD2 does not release NO or HNO when subjected to physiological temperatures and pHs. In the table, where a percentage of MD3 is shown, the balance, adding up to 100%, is predominantly MD2, with minor amounts of other impurities.

Example 13: pH vs Efficacy: MD3

The efficacy of the compound of Formula III against PAK was evaluated at various pHs, 6.4, 7.6, and 8.4, all of which are physiological pHs, though at different places in the human body. For example, tuminal pH in the proximal small bowel ranges from 5.5 to 7.0 and gradually rises to 6.5-7.5 in the distal ileum. There is a decrease in luminal pH from the terminal ileum to the caecum (range 5.5-7.5). The pH in the colon can range from 7.9 to 8.5. A normal blood pH level is 7.40, and this is approximately the pH in the lung. The pH of saliva is ranges from 6.5 to 7.5.

As shown below in FIG. 12, the compound of Formula III at a dosage of 0.125 mg/ml and a pH of 6.4, was sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 102 in two hours, and the concentration stayed at this level for up to 25 hours. In contrast, the compound of Formula III at a dosage of 0.125 mg/ml and a pH of 8.4 was only sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 105 in six hours, and the concentration stayed at this level for up to 25 hours. The compound of Formula III at a dosage of 0.125 mg/ml and a pH of 7.4 was sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 102 in four hours, and the concentration stayed at this level for up to 25 hours. PAK concentrations in untreated control remained at 106 for the entire experiment.

As shown below in FIG. 13, when the experiment was repeated, using the compound of Formula III at a dosage of 0.0625 mg/ml and a pH of 6.4, this was sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 102 in two hours, and the concentration stayed at this level for up to 25 hours. In contrast, the compound of Formula III at a dosage of 0.125 mg/ml and a pH of 8.4 was only sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 105 in eight hours, and the concentration returned to 106 at 25 hours. The compound of Formula III at a dosage of 0.125 mg/ml and a pH of 7.4 was sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 102 in six hours, and the concentration stayed at this level for up to 25 hours. PAK concentrations in untreated control remained at 106 for the entire experiment.

As shown below in FIG. 14, when the experiment was repeated, using the compound of Formula III at a dosage of 0.03125 mg/ml and a pH of 6.4, this was sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 102 in three hours, and the concentration stayed at this level for up to 25 hours. In contrast, the compound of Formula III at a dosage of 0.03125 mg/ml and a pH of 8.4 was insufficient to significantly reduce the concentration of PAK (CFU/ml). MD3 at a dosage of 0.03125 mg/ml and a pH of 7.4 was sufficient to reduce the concentration of PAK (CFU/ml) from 106 to 102 in eight hours, and the concentration stayed at this level for up to 25 hours. PAK concentrations in untreated control remained at 106 for the entire experiment.

Based on the data, a conclusion can be reached that the efficacy of the compound of Formula III is pH-dependent due to the role pH plays on affecting the NO release rate from the compound of Formula III.

Example 14: Comparison of Various Reactants on Purity and Synthetic Process Optimization

The compound of Formula III can be prepared using acetone, ethanol or acetonitrile as a starting material as well as other compound with similar functional groups, though when prepared from ethanol or acetonitrile (or any other compound), the impurity profiles for each will be unique. A set of chromatograms are shown of samples of the compound of Formula III prepared from acetone, ethanol, and acetonitrile. In each case a different set of impurities were observed. While the material prepared from ethanol was comparable in purity to the material prepared from acetone (>90% area), the material prepared from acetonitrile was significantly less pure (<40% area).

The process for synthesizing the compound of Formula III starting from acetone was optimized. A series of experiments were run to evaluate the effects of starting acetone concentration (14 vs 30 mg/mL), base equivalents (4 vs 6), NO pressure (2.5 vs 20 bar), and temperature (10 vs 20° C.) on the yield and purity of product produced. Table 13 show the results of the experiments run at an acetone concentration of 14 mg/ml and a temperature of 20° C. for 4 days. Each condition was run in duplicate.

TABLE 13 Compound of Formula III results associated with synthesis conditions. Pressure Base Eq MD2 MD3 NO Total Sample (bar) (NaOH) (Area %) (Area %) (umol/mg) 1 2.5 4 5.0 93.3 5.5 2 3.1 95.7 5.7 3 2.5 6 20.4 79.0 5.7 4 16.7 82.6 5.2 5 20 4 0.6 97.5 6.3 6 1.0 96.2 6.1 7 20 6 2.8 94.4 5.9 8 3.2 94.2 5.9

The best overall conditions identified from these experiments were an acetone concentration of 14 mg/mL, an NO pressure of 20 bar, a base equivalent of 4. Temperature had no effect between 10 and 20° C. when run at these conditions. At these conditions, an average purity of ˜97% area was achieved with a yield>95%.

Example 15: Comparison of the Antimicrobial Activity of the Compound of Formula III with Other NO Releasing Compounds

Other NO donating compounds have been studied for their potential anti-microbial activity. However it can be anticipated that different NO donors will range in their anti-microbial activity due to their varying chemical properties: NO load capacity, NO release rate, water solubility, pKa, molecular weight, etc. Therefore the antimicrobial activity of the compound of Formula III was compared to two other NO donating compounds that were synthesized and characterized in our laboratories: the first was a hepta-substituted ethanolamine β-cyclodextrin compound where all seven secondary amines were functionalized with diazeniumdiolate groups; the second was 2,6-cis-dimethylpiperidine functioned with a group. The results of their antimicrobial activities are shown in Table 14.

TABLE 14 The MIC/MBCs of two NO donors compared to the MIC/MBCs compound of Formula III for various strains of M. abscessus. M. abscessus N0010; N0016; N0017; N0018; N0019; N0020; clinical clinical clinical clinical clinical clinical N0046 Compound NO Load MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC β-CD/NO 4.2 0.25 hr 4 16 1 16 1 16 4 16 2 8 4 8 8 16 cis-DMP/NO 6.9 1 hr 1 4 2 4 2 4 1 4 2 4 1 2 2 4 MD3 6.7 3 hr 0.25 1 0.25 1 0.25 1 0.25 1 0.25 1 0.25 1 0.25 1

From the compiled data, it is clear the antimicrobial activity of the compound of Formula III is greater than the antimicrobial activity for either of the other two NO donor molecules. A portion of the increased activity can be attributed to its high NO loading capability, but it is no higher than cis-DMP/NO and yet it four times as effective at killing bacteria. This suggests that the extended NO half-life of the compound of Formula III in comparison to other NO donor compounds plays a critical role as well. The compound of Formula III also releases nitroxyl (HNO) in addition to NO upon degradation at neutral pH conditions. HNO is another reactive nitrogen compound. On its own, nitroxyl will form a dimer with itself which then rearranges to yield one mole of N2O and one mole of water.

It is contemplated that various combinations or subcombinations of the specific features and aspects of the embodiments disclosed above may be made and still fall within one or more of the inventions. Further, the disclosure herein of any particular feature, aspect, method, property, characteristic, quality, attribute, element, or the like in connection with an embodiment can be used in all other embodiments set forth herein.

Accordingly, it should be understood that various features and aspects of the disclosed embodiments can be combined with or substituted for one another in order to form varying modes of the disclosed inventions. Thus, it is intended that the scope of the present inventions herein disclosed should not be limited by the particular disclosed embodiments described above.

Moreover, while the invention is susceptible to various modifications, and alternative forms, specific examples thereof have been shown in the drawings and are herein described in detail. It should be understood, however, that the invention is not to be limited to the particular forms or methods disclosed, but to the contrary, the invention is to cover all modifications, equivalents, and alternatives falling within the spirit and scope of the various embodiments described and the appended claims. Any methods disclosed herein need not be performed in the order recited.

The methods disclosed herein include certain actions taken by a practitioner; however, they can also include any third-party instruction of those actions, either expressly or by implication. For example, actions such as “administering an NO-donating composition” include “instructing the administration of an NO-donating composition.” In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.

The contents of all documents referred to herein are hereby incorporated by reference for all purposes.

Claims

1. An antimicrobial formulation for sanitizing, disinfecting, decontaminating and/or sterilizing one or more surfaces, comprising a NO-releasing compound of Formula I and a carrier or excipient, wherein Formula 1 has the following structure: wherein: of —OH, —NH2, —OCH3, —C(O)OH, —CH2OH, —CH2OCH3, —CH2OCH2CH2OH, —OCH2C(O)OH, —CH2OCH2C(O)OH, —CH2C(O)OH, —NHC(O)—CH3, —C(O)O((CH2)aO)b—H, —C(O)O((CH2)aO)b—(CH2)cH, —C(O)O(C1-5alkyl), —C(O)—NH—((CH2)dNH)e—H, —C(O)—NH—((CH2)dNH)e—(CH2)fH, —O—((CH2)aO)b—H, —O—((CH2)aO)b—(CH2)cH, —O—(C1-5alkyl), —NH—((CH2)dNH)e—H, and —NH—((CH2)dNH)e—(CH2)fH,

X is selected from the group consisting of H, D, R, and RC(O)—,
R is C1-12 alkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, optionally substituted with one or more substituents, wherein the substituents are independently selected from the group consisting
each instance of a, b, c, d, e, f, g, h, i, j, k, and 1 is independently selected from an integer of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10,
and M+ is a pharmaceutically-acceptable cation, wherein, where M+ is a cation with a valence other than one, the ratio of the compound of Formula I to the cation is such that the total positive charge equals the total negative charge selected from the group consisting of sodium, potassium, lithium, and quaternary ammonium.

2. (canceled)

3. The formulation of claim 1, wherein the compound has the following structure:

wherein M+ refers to a pharmaceutically-acceptable cation selected from the group consisting of sodium, potassium, lithium, and quaternary ammonium.

4. The formulation of claim 1, wherein the compound has the following structure:

5-6. (canceled)

7. The formulation of claim 1, in the form of an aqueous solution, an alcoholic solution, a dispersion, an aerosol, a spray, a gel, a foam, or a wet wipe.

8. The formulation of claim 1, wherein the compound has a total releasable NO storage in a range of 0.1-8.0 μmol of NO per mg of said compound, a NO half-life in the range of 0.1-24 hours, a total duration of NO release in a range of 1-60 hours, or a total NO release after 4 hours in a range from 0.1-7.0 μmol of NO per mg of said compound.

9-11. (canceled)

12. The formulation of claim 1, further comprising a quaternary ammonium salt.

13. A method of sanitizing, disinfecting, decontaminating, or sterilizing a surface, comprising contacting the surface with an effective, antimicrobial amount of the formulation of claim 1.

14. The method of claim 13, wherein the formulation further comprises a quaternary ammonium salt.

15-16. (canceled)

17. The method of claim 13, wherein the microbe associated with the microbial infection comprises two or more of the following: gram-positive bacteria, gram-negative bacteria, fungi, yeast, and viruses.

18. The method of claim 13, wherein the microbe is present in a hospital setting, a doctor's office, a dentist's office, a chiropractor's office, or an optometrist's office.

19. (canceled)

20. The method of claim 13, wherein the surface is a solid surface.

21. The method of claim 13, wherein the surface is associated with an animal or its surroundings, such as the surface of a livestock animal and its pens, cages, or holding areas, such as chicken coops.

22. (canceled)

24. The method of claim 13, wherein the surface is selected from the group consisting of floors, walls, countertops, appliances, and fixtures.

25. The method of claim 13, wherein the method is used to reduce or remove the build-up of mold, bacteria, biofilm, and other pathogens that are present, may be present, or may have been present, in an appliance.

26-33. (canceled)

34. The method of claim 13, wherein the sterilizing, disinfecting, sanitizing, and/or decontaminating step is performed in a chamber or other closed container, into which items to be sterilized, disinfected, sanitized, and/or decontaminated are placed after being contacted with the formulation.

35. The method of claim 13, wherein the microbe is present in a crop locus, a tree, a plant, a fruit or a vegetable.

36. (canceled)

37. The method of claim 13, wherein the microbial load comprises drug-resistant bacteria.

38. The method of claim 13, wherein the microbe is selected from the group consisting of human immunodeficiency virus, herpes simplex virus, papilloma virus, parainfluenza virus, influenza, hepatitis, Coxsackie Virus, herpes zoster, measles, mumps, rubella, rabies, pneumonia, hemorrhagic viral fevers, H1N1, prions, parasites, fungi, mold, Candida albicans, Aspergillus niger, Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus, Group A streptococci, S. pneumoniae, Mycobacterium tuberculosis, Campylobacter jejuni, Salmonella, Shigella, carbapenem-resistant Enterobacteriaceae Methicillin-resistant Staphylococcus aureus, and Burkholderia cepacia.

39. The method of claim 13, wherein the microbe is a bacteria selected from the group consisting of Haemophilus influenzae, Pseudomonas aeruginosa, Staphylococcus aureus, Staphylococcus wameri, Staphylococcus lugdunensis, Staphylococcus epidermidis, Streptococcus milleri/anginous, Streptococcus pyogenes, non-tuberculosis Mycobacterium, Mycobacterium tuberculosis, Burkholderia spp., Achromobacter xylosoxidans, Pandoraeasputorum, Stenotrophomonas maltophilia, Alcaligenes xylosoxidans, Haemophilus pittmaniae, Serratia marcescens, Candida albicans, drug resistant Candida albicans, Candida glabrata, Candida krusei, Candida guilliermondii, Candida auris, Candida tropicalis, Aspergillus niger, Aspergillus terreus, Aspergillus fumigatus, Aspergillus flavus, Morganella morganii, Inquilinus limosus, Ralstonia mannitolilytica, Pandoraea apista, Pandoraea pnomenusa, Pandoraea sputorum, Bdellovibrio bacteriovorus, Bordetella bronchiseptica, Vampirovibrio chlorellavorus, Actinobacter baumanni, Cupriadidus metallidurans, Cupriavidus pauculus, Cupriavidus respiraculi, Delftia acidivordans, Exophilia dermatitidis, Herbaspirillum frisingense, Herbaspirillum seropedicae, Klebsiella pneumoniae, Pandoraea norimbergensis, Pandoraea pulmonicola, Pseudomonasmendocina, Pseudomonas pseudoalcaligenes, Pseudomonas putida, Pseudomonas stutzeri, Ralstonia insidiosa, Ralstonia pickettii, Neisseriagonorrhoeae, NDM-1 positive E. coli, Enterobacter cloaca, Vancomycin-resistant E. faecium, Vancomycin-resistant E. faecalis, E. faecium, E. faecalis, Clindamycin-resistant S. agalactiae, S. agalactiae, Bacteroides fragilis, Clostridium difficile, Streptococcus pneumonia, Moraxella catarrhalis, Haemophilus haemolyticus, Haemophilus parainfluenzae, Chlamydophilia pneumoniae, Mycoplasma pneumoniae, Atopobium, Sphingomonas, Saccharibacteria, Leptotrichia, Capnocytophaga, Oribacterium, Aquabacterium, Lachnoanaerobaculum, Campylobacter, Acinetobacter, Agrobacterium; Bordetella; Brevundimonas; Chryseobacterium; Delftia; Enterobacter; Klebsiella; Pandoraea; Pseudomonas; Ralstonia, and Prevotella.

40. The method according to claim 13, wherein the microbe comprises one or more of Methicillin-resistant Staphylococcus aureus, carbapenem-resistant Enterobacteriaceae. Staphylococcus aureus, Pseudomonas aeruginosa or Burkholderia cepacia.

41-45. (canceled)

45. The method according to claim 13, wherein the microbe is a virus selected from the group consisting of Coronavirus, Picomavirus, orthomyxoviruses, paramyxoviruses, cytomegalovirus, and adenovirus.

46. The method of claim 45, wherein the virus is SARS, MERS, or SARS-CoV2.

47. The method of claim 12, wherein the microbe is a fungus selected from the group consisting of Histoplasma capsulatum, Blastomyces dermatitidis, Sporothrix schenckii, Coccidioides immitis, Coccidioides posadasii. Aspergilli, Cryptococcosis, Candida, Mucormycosis, and Pneumocystis jirovecii.

48. The method of claim 13, wherein the sanitizing, disinfecting, decontaminating and/or sterilizing effect is associated with the release of NO and HNO from the compound as it degrades.

49. (canceled)

50. A wet wipe impregnated with the formulation of claim 1.

Patent History
Publication number: 20230337678
Type: Application
Filed: Feb 5, 2021
Publication Date: Oct 26, 2023
Inventors: Drew Folk (Durham, NC), Ryan Gerald Anderson (Durham, NC), John Kelly Simons (Durham, NC), Mona Jasmine Rosales Ahonen (Durham, NC), Rebecca Anthouard McDonald (Durham, NC)
Application Number: 17/797,899
Classifications
International Classification: A01N 51/00 (20060101); A01P 1/00 (20060101); A01N 33/12 (20060101);